Opinion statement
Since the first demonstrations of the differentiation of pluripotent stem cells to produce functional human cellular models such as cardiomyocytes, the scientific community has been captivated [1, 2••, 3]. In the time since that seminal work, the field has been catapulted forward by the demonstration that adult somatic cells can be reprogrammed to an induced state of pluripotency [4••], and more recently by the development of efficient and sophisticated genome editing tools [5••, 6••, 7], which together afford a theoretically unlimited supply of relevant genetic disease models. In particular, many of the early successes with induced pluripotent stem cell technology have been realized with cardiac arrhythmia syndromes [8••, 9–15]. There is interest in applying stem cell models in large-scale screens to discover novel therapeutics or drug toxicities. This manuscript aims to discuss the potential role of hPSC-derived cardiomyocyte models in therapeutic arrhythmia screens and review recent advances in the field that bring us closer to this reality.
Keywords: Pluripotent, Stem cell, Cardiomyocytes, Arrhythmia, Screening, Electrophysiology, Cardiac electrophysiology
Introduction
A major advance in disease modeling has been the demonstration that human induced pluripotent stem cell (hiPSC) technology can faithfully recapitulate many human diseases, including cardiac arrhythmia syndromes [10, 16, 14]. Most of the initial cardiac disorders to be modeled with hiPSC-derived cardiomyocytes (hiPSC-CM) have been Mendelian arrhythmia syndromes [8••, 9–15], though increasingly, other cardiac diseases have been modeled, such as familial hypertrophic and dilated cardiomyopathy [17, 18]. Importantly, several of these diseases lack targeted therapeutics that directly address their respective physiological defects. In addition to these unmet needs, stem-cell–derived models may hold promise in evaluation of drug-induced QT prolongation, one of the most common causes of post-market drug withdrawal, which remains difficult to predict in the pre-clinical setting [19]. Enthusiasm has been significant for applying stem-cell models to large-scale screens for both novel therapeutics and cardiotoxicity evaluations [20••, 21, 22]. However, most of the early studies in the stem cell field were limited in scope, despite being conceptually innovative. In recent years, the techniques for human pluripotent stem cell culture and cardiac differentiation have dramatically improved [23••, 24, 25]. This review will focus on the role that stem cell models can play in cardiac arrhythmia-related drug screens, and discuss the necessary steps to realize their potential.
Characteristics and relevance of stem-cell–derived cardiomyocytes
The contribution of any particular model is critically dependent on how faithfully it represents the native in vivo condition—in this case, a mature adult human cardiomyocyte (CM). As adult ventricular CMs are obtained only invasively and thereby in short supply, most studies characterizing the properties of human pluripotent stem cell derived cardiomyocytes (hPSC-CM) to date have compared parameters to previously published values [26].
Morphologically, most studies have reported that hPSC-CMs, both hiPSC-CMs and human embryonic stem cell-derived cardiomyocytes (hESC-CMs), are markedly smaller than adult CMs and lack organized sarcomeres and T-tubules, with a gene expression profile more closely resembling that of fetal CMs [27•]. These features of immaturity are similar to the immature electrical parameters recorded by patch clamp electrophysiology [27•, 28, 29•]. In contrast to adult human CMs, hPSC-CMs bear relatively depolarized diastolic potentials, slower action potential upstroke velocities, and spontaneous electrical activity [30].
In terms of action potential shape, most investigators have noted the appearance of three distinct hPSC-CM action potential subtypes, classified as ventricular-like, atrial-like, and nodal-like [28]. However, it has been acknowledged that there is a great deal of heterogeneity of AP characteristics reported between cell lines [31] and different laboratories [27•], and the relative cellular subtype proportions critically depend upon the criteria utilized to distinguish them [32•]. Despite this variability, the most frequently reported subtype population is ventricular-like [8••, 9, 29•, 33], characterized by a prominent plateau phase and longer action potential duration (APD), the length of which, while variable between studies, is comparable to reported values for native ventricular CMs [27•].
Individual currents have also been extensively studied in hPSC-CMs using voltage clamp electrophysiology, demonstrating the presence of the major currents INa, IKr, IKs, ICa,L, and Ito [2••, 34•, 35, 36]. Unlike mature atrial and ventricular adult CMs, hPSC-CMs also universally feature a prominent funny current, If, and an absent or minimal inward rectifier, IK1 [37]. Furthermore, a rather large proportion of the Ca2+ release during an hPSC-CM action potential is IP3-sensitive [38]. A comprehensive analysis of the cellular electrophysiology of CMs derived from a single induced pluripotent stem cell (iPSC) line was recently reported [34•].
The electrophysiological responses of hPSC-CMs to various drug compounds have been explored by several investigators. hPSC-CM sensitivity has been demonstrated to adrenergic and cholinergic compounds [39], cardiac glycosides [40], IKr inhibitors [40, 34, 8••, 9], ICa,L inhibitors [9, 38], IK,ATP activators [9], IKs inhibitors [28, 36], and various inhibitors of Na currents [35, 41]. Despite many subtle and some more substantive differences between hPSC-CMs and adult CMs, the action potentials and component currents do bear remarkable similarity, prompting hope that hPSC-CMs can recapitulate the adult CM intricacies more accurately than common animal models or heterologous expression systems.
Arrhythmia syndromes modeled in hPSC-CMs
Screens utilizing hPSC-CM models may aim to discover novel therapeutics, but could also be used to detect cardiotoxicity in either healthy or diseased states. In considering the value of therapeutic and toxicological screens using arrhythmic disease models, it is helpful to briefly review progress thus far (Table 1).
Table 1.
Disease | Mutation | Derivation | Control comparison |
Method of differentiation |
Cardiomyocyte characterization |
Novel Findings | Reference |
---|---|---|---|---|---|---|---|
LQT1 | KCNQ1 R190Q |
Retroviral (OSKM) |
Healthy volunteer hiPSC |
Embryoid body | Patch clamp, Immunostaining for cardiac markers |
Prolonged APD in LQT1 subjects |
Moretti A et al. [8•] |
LQT1 | KCNQ1 P631fs/33 |
Retroviral (OSKM) |
Healthy volunteer hiPSC |
Embryoid body | Patch clamp, MEA, Immunostaining |
Prolonged MEA FPD, dominant negative physiology only evident in heterologous system |
Egashira T et al. [42] |
LQT2 | KCNH2 A614V |
Retroviral (OSK) |
Healthy volunteer hiPSC |
Embryoid body | Patch clamp, MEA, Immunostaining |
Prolonged APD, pharmacologic shortening effect |
Itzhaki et al. [9] |
LQT2 | KCNH2 G1681A |
Lentiviral (OLSN) |
Unaffected carrier mother, Healthy volunteer hiPSC and hESC H7 line |
Forced aggregation of defined cell number into embryoid bodies in V-96 plates |
Patch clamp, MEA, Immunostaining |
Prolonged APD and FPD, patch clamp but not MEA revealed prolonged APD in unaffected carrier mother |
Matsa et al. [10] |
LQT2 | KCNH2 R176W |
Retroviral (OSKM) |
Healthy volunteer hiPSC and hESC H7 line |
END-2 visceral endoderm cell co- culture |
Perforated patch clamp, MEA, Immunostaining |
Prolonged APD, noted that patch clamp LQT2 recordings were prolonged by 66 %, whereas MEA FPD was only prolonged by 10– 20 % |
Lahti A et al. [43] |
LQT2 | KCNH2 N996I |
Retroviral (OSKM) Targeted genetic mutation by HR in NKX2.5eGFP/w hESCs |
Targeted genetic correction by HR in mutant hiPSC. Targeted hESCs compared to untargeted hESCs |
END-2 visceral endoderm cell co- culture |
Patch clamp, MEA, Immunostaining |
KCNH2 N996I confers a prolonged APD and MEA FPD in both hiPSC and hESC lines studied Between line differences were great, as hiPSC corrected APD measurements were even longer than WT hESC |
Bellin M et al. [31] |
LQT2 | KCNH2 A561V |
Episomal plasmid transfection |
Healthy volunteer hiPSC |
Embryoid body | Patch clamp, MEA, Immunostaining |
LQT2 CMs exhibited prolonged APD and FPD, lower E4031-sensitive currents. Mutant hERG was located peri- nuclear. Treatment with ALLN restored membrane localization and shortened APD |
Mehta A et al. [99] |
LQT3 | SCN5A V1763M |
Synthetic mRNA (OSKML) |
Unaffected sibling control hiPSC |
Guided embryoid body differentiation with small molecules SB203580 and IWP-2 |
Patch clamp, Immunostaining |
Increased late Na+ current and significantly prolonged APD in mutant iPSC- CMs. |
Ma D et al. [44] |
LQT3 | Two patients: SCN5A R535Q & SCN5A V240M |
Retroviral (OSKM) |
Unaffected unrelated iPSC control and hESC (H1) control lines |
END-2 visceral endoderm cell co- culture |
Patch clamp, Immunostaining |
Non-significant trend towards APD prolongation. Significant variability in AP parameters between individual iPSC- CMs |
Fatima A et al. [45] |
LQT3 | SCN5A F1473C With KCNH2 polymorphism K897T |
Polycistronic lentiviral vector: hSTEMCCA -loxP lentiviral (OSKM) |
Mother with KCNH2 polymorphism K897T and SCN5A WT, Father WT for each |
Embryoid body formation, with growth factor and small molecule application: BMP4, Activin A, DKK1, SB- 431542, BMP |
Patch clamp | APD not compared, authors noting the minimal contribution of late INa with the depolarized resting membrane potential of hPSC- CM models KCNH2 polymorphism without influence on anti-arrhythmic efficacy Increased stimulation frequency reduced late INa late, correlating with clinical effect of increasing pacemaker rate |
Terrenoire C et al. [46] |
LQT3/ Brugada |
SCN5A 1798insD |
Retroviral or lentiviral (OSKM) |
hiPSC WT for SCN5A |
END-2 visceral endoderm cell co- culture |
Patch clamp, Immunostaining |
Increased INa late, in mutant hiPSC. Small degree of APD prolongation vs WT (217.2±14.9 vs 173.5±12.2 msec) |
Davis RP et al. [12] |
LQT8 “Timothy syndrome” |
CACNA1C G406R |
Retroviral (OSKM) |
Five diseased and five healthy volunteer controls iPSC lines. |
Embryoid body | Patch clamp, single cell RT-PCR for MLC2V to isolate ventricular cells, Live calcium imaging with Fluo-4 |
Prolonged APD by ~three fold. Higher amplitude and irregular Ca2+ transients in mutants |
Yazawa M et al. [11] |
CPVT1 | RYR2 M4109R |
Retroviral (OSK) plus valproate |
Healthy volunteer hiPSC |
Embryoid body | Patch clamp, MEA, Live calcium imaging with Fluo-4 |
Increased frequency of delayed afterdepolarizations in CPVT iPSC- CMs, which increased with adrenergic stimulation and were abolished by flecainide, Increased Ca transients and increased store- overload-induced Ca release at high extracellular Ca2+ |
Itzhaki I et al. [13] |
CPVT1 | RYR2 S406L |
Retroviral (OSKM) |
Healthy volunteer hiPSC |
Embryoid body | Patch clamp, Immunostaining, Live calcium imaging with Fluo-4 |
CPVT iPSC-CMs exhibited increased Ca2+ cycling abnormalities including alternans, transient fusion, and irregular oscillations. Upon exposure to isoproterenol, diastolic Ca2+ levels elevated in CPVT cells. Dantrolene rescued |
Jung CB et al. [48] |
CPVT1 | RYR2 P2328S |
Retroviral (OSKM) |
Healthy volunteer hiPSC |
END-2 visceral endoderm cell co- culture |
Patch clamp, Immunostaining, Live calcium imaging with Fura-2 AM |
CPVT iPSC-CMs showed higher Ca2+ cycling abnormalities (14 % vs 8 %), which augmented with adrenergic stimulation. CPVT cells had higher diastolic calcium levels and lower SR load. |
Kujala KP et al. [49] |
CPVT1 | RYR2 F2483I |
Retroviral (OSKM) |
Healthy foreskin fibroblast hiPSC clone hiPSC1, hESC lines H1, H9, HES- 2 |
END-2 visceral endoderm cell co- culture |
Patch clamp, MEA recording, Live calcium imaging with Fluo-4, Caffeine releasable SR-Ca2+ stores |
Non-significant excess of arrhythmic activity on MEA in CPVT CMs, excess of DADs in CPVT CMs Slow decay of Ca transients in mutants, Increased Ca- induced Ca- release gain in mutants upon adrenergic stimulation, yet lower SR Ca2+- load |
Zhang XH et al. [50] and Fatima et al. . [100] |
CPVT2 | CASQ2 | Polycistronic lentiviral vector: hSTEMCCA -loxP lentiviral (OSKM) |
Three healthy volunteer hiPSCs |
Embryoid body | Patch clamp, Immunostaining, Live calcium imaging with Fura-2 AM, Transmission electron microscopy |
Isoproterenol induced after- contractions and DADs in CPVT2- CMs and elevated diastolic [Ca2+]i levels. **APD was longer in CPVT2- CMs |
Novak A et al. [51] |
ARVC |
PKP2 2484C>T causing cryptic splicing in exon 12 |
Retroviral (OSKM) |
Healthy volunteer hiPSCs, and H9 hESCs |
Embryoid body, Lipogenic conditions (5F protocol: PPARγ and PPARα agonists, with insulin, dexamethasone, and IBMX) |
TUNEL staining for apoptosis, Patch clamp, Energetics, Immunostaining |
ARVC-CMs had excess apoptosis upon lipogenic induction, depressed energy production and increased glycolytic reliance, mutant cells had prolonged relaxation time Slightly higher mean diastolic potential and prolonged APD |
Kim et al. [14] |
ARVC | PKP2 L614P |
Retroviral (OSKM) |
Healthy volunteer hiPSC, H9 hESCs |
Embryoid body, In some renditions, Lipogenic conditions (indomethacin, insulin, dexamethasone, and IBMX) |
Patch clamp, Immunostaining and Oil Red O staining, Live calcium imaging with Fluo-4, Transmission electron microscopy |
ARVC-CMs feature reduced PKP2 and plakoglobin desmosomal proteins at cell periphery, Increased Oil Red O staining in mutant cells after exposure to lipogenic conditions |
Ma D et al. [15] |
ARVC | Two patients; PKP2 c972InsT/ N leading to premature stop codon PKP2 c148- 151delAC AG/N causing premature stop codon |
Retroviral (OSK) with valproate |
Healthy volunteer hiPSC |
Embryoid body, Lipogenic conditions (insulin, dexamethasone, IBMX, 20 % fetal bovine serum, and StemPro LipoMax) |
Patch clamp, Immunostaining, MEA Transmission electron microscopy |
Significant reduction in PKP2 immunosignal in ARVC-CMs, Prolonged field potential rise time by MEA 33 % of ARVC- CMs featured lipid droplets on EM compared to none in healthy hiPSC- CMs, Lipogenic conditions caused an extensive amount of lipid accumulation in ARVC-CMs ARVC-CMs featured hazy, dissymmetric desmosomes and widened desmosomal gaps ARVC-CMs featured higher percentage of apoptotic cells (3.8 % vs. 1.0 %) |
Caspi O et al. [53] |
OSKM: Oct-4, Sox-2, Klf-4, c-Myc
OSK: Oct-4, Sox-2, Klf-4
OLSN: Oct-4, Lin28, Sox-2, Nanog
OSKML: Oct-4, Sox-2, Klf-4, c-Myc, Lin28
criticized as non-representative of the clinical disorder
LQTS
The Long-QT Syndrome is characterized by a prolongation in the QT interval, the cellular correlate of which is the AP duration. In 2010, seminal work featured action potential recordings from iPSC-CMs derived from two patients harboring mutations in KCNQ1 (LQT1), encoding the alpha subunit of the channel mediating the IKs current [8••]. Other iPSC-derived LQT models have since been reported for LQT1 [42], LQT2 (IKr deficiency) [9, 10, 43], LQT3 (persistence of late INa) and LQT8/Timothy syndrome [11]. The quintessential feature of these has been demonstration of prolonged AP duration, as well as arrhythmogenic elements such as EADs. Importantly, the responses to drugs such as isoproterenol-induced EADs in LQT1 [8••], faithfully recapitulate expected clinical features of the respective disorders. Models of LQT3 syndrome, caused by a gain of function in INa,late that prolongs inward current over the plateau phase have had mixed results. Ma et al. reported significant APD prolongation in iPSC-CMs derived from a patient with a V1763M mutation in SCN5A, which was reversible by mexilitine treatment [44], and Davis et al. reported similar results in cells derived from a patient with a clinical overlap syndrome with Brugada [12]. Another study of LQT3 hiPSC-CMs demonstrated variable APD prolongation not reaching statistical significance [45]. Finally, other investigators have chosen not to report AP comparisons from LQT3 hiPSC-CMs, reasoning that the relatively depolarized membrane potential in hPSC-CMs, compared to their adult counterparts, inactivates a large proportion of the Na+ channel, which is thereby unable to contribute to the AP shape [46]. Supporting this notion, the INa,late modulator ATX has been reported to have no apparent effect on AP duration [47].
Catecholaminergic polymorphic ventricular tachycardia (CPVT)
Several groups have published hiPSC-CM models derived from patients with CPVT, caused by gain of function mutations in RYR2 [13, 48, 49, 50] or loss of function in CASQ2 [51], and characterized by susceptibility of the sarcoplasmic reticulum to premature calcium release when stimulated by catecholamines. In the CPVT1 models, as in other studies of Ca2+ handling in hPSC-CMs [38, 52], transients were evident upon application of caffeine and abolished by ryanodine, suggesting the presence of functional SR and ryanodine receptors (RyR). However, as hPSC-CM models are deficient in t-tubules, and Ca2+ wavefronts have been noted to be slow, suggesting that coupling between L-type Ca2+ channels and SR mediated Ca2+ release may be impaired. Thus, despite increased Ca2+ transients in CPVT hiPSC-CM models, the physiologic details of calcium handling may be less faithful to the native disease condition [27•]. Furthermore, the CASQ2 mutant hiPSC-CMs reported by Novak et al. feature action potential prolongation that differs substantially from the clinical features of the condition [51].
Arrhythmogenic right ventrcular cardiomyopathy (ARVC)
Arrhythmogenic right ventricular cardiomyopathy (ARVC) is marked by defects in desmosomal proteins leading to fibrofatty replacement of the right ventricular myocardium with age and increased incidence of ventricular arrhythmias. Three studies have produced hiPSC-CM models from patients with mutations in PKP2, the most frequently responsible gene for ARVC [53, 15, 14]. These reports revealed accumulation of intracellular lipid in response to lipogenic stimuli. Electrophysiological differences were subtle, with slightly higher mean diastolic potential and prolonged APD noted in one study [14], and prolonged field potential rise time by multi-electrode array in another [53], possibly reflecting slowed conduction and impaired coupling.
Other cardiac diseases
hPSC-CM models of familial forms of hypertrophic cardiomyopathy [18], dilated cardiomyopathy [17], LEOPARD syndrome, Pompe’s disease [54], and other disease have been reported elsewhere. While these clinical syndromes may be accompanied by arrhythmias, we considered them to be outside of the scope of this review.
Standardization is a necessary step
Original methods of cardiac differentiation from hPSCs relied on spontaneous differentiation of aggregates formed from hPSCs known as embryoid bodies, a process with low efficiency and highly variability between cell lines [55]. In contrast, current techniques aim to increase efficiency by recapitulating key developmental cues in embryonic cardiogenesis [56]. In general, there are three frequently reported methods: embryoid body differentiation combined with sequential addition of key growth factors [57], co-culture with visceral endoderm-like cells [29], and monolayer differentiation guided with recombinant growth factors [58, 23••, 59] or small molecules [24]. These techniques can be augmented to shift cell subtype lineage (atrial vs. ventricular) [33], or to improve differentiation, such as by inclusion of ascorbate [60], DMSO [61], or in the case of the monolayer technique, an extracellular matrix sandwich [23••], a method yielding highly efficient preparations of cardiomyocytes. However, efficiency varies even with these protocols, which in turn must be additionally optimized for different cell lines. In parallel, efforts have focused on enriching existing populations of differentiated mixtures of cells derived from hPSCs for cardiomyocytes [62], such as by Percoll gradient [3], genetic selection by fluorescence or antibiotic resistance [63, 64, 65•], selection by oxidative metabolic capacity [66], and sorting by mitochondrial content [67] or cardiomyocyte cell surface markers such as SIRPA [68] or VCAM-1 [69].
Facilitating the maturation of hPSC-CMs has also been an important area of investigation, as prolongation of time in culture alone is insufficient. Each method of differentiation above involves culturing cells on a fixed protein substrate on a two-dimensional culture platform within an incubator controlling physiological temperature and CO2 levels. While this embodiment aligns well with the infrastructure of most academic research laboratories, the conditions differ markedly from the in vivo environment, with the loss of three-dimensional ultrastructure and associated cellular contacts and mechanical stresses. One strategy to assist maturation encompasses growth on engineered substrates such as “biowires” to induce structural alignment and mechanical load [70]. Other strategies have involved genetic introduction of deficient rectifying currents accompanied by electrical field pacing [71], manipulation of bioenergetics [72••, 14], and other techniques. None of these has been adopted on a widespread level or has been shown to scale up for generation of the large supplies of cardiomyocytes necessary for high-throughput applications. An additional variable to consider is the time point after differentiation for cellular recordings, which may in turn depend on the differentiation protocols and cell line.
Once standardized protocols for cardiomyocytes differentiation are developed, in what state should hPSC-CMs be assayed? From a pure physiologist’s vantage point, it is attractive to study the singularized hPSC-CM. Isolated and electromechanically decoupled from neighboring cells, this state mimics traditional physiological experiments with isolated adult CMs. However, experiments with isolated CMs are often performed early after isolation, before the conditions of culture exert their confounding influences. Adult CMs de-differentiate when maintained in prolonged culture, exhibiting a higher diastolic potential, spontaneous activity, loss of T-tubular and sarcomeric organizational structure, and variable action potential phenotypes [73, 74]. Meanwhile, hPSC-CMs are kept in continual culture and are generally phenotyped following a recovery period after dissociation, and it is conceivable that dissociation to a single cell level may substantially alter cellular phenotype. Moreover, there is evidence that removal of the influence of non-myocytes from hPSC-CMs in embryoid bodies by genetic selection results in a functional stalling of maturation [75]. Protocols for cellular dissociation vary, with some resembling traditional cell culture techniques [23••], and others bearing resemblance to animal CM isolation [46]. Unlike freshly isolated adult CMs where the source is relatively homogeneous and the principle quality metric is Ca2+ tolerance, heterogeneous starting populations of hPSC-CM may be differentially affected by dissociation protocols, introducing survival biases. Finally, the appropriate cell plating density and optimal time window to assay hPSC-CMs following dissociation [76] requires exploration.
Streamlining phenotyping
Conventional electrophysiology
How should hPSC-CMs be phenotyped in high-throughput screens? Since its inception by Neher and Sakmann [77], conventional patch clamp electrophysiology has proven to be a rigorous method of evaluation and has previously been used to demonstrate the predictive power of hPSC-CM models for cardiotoxicity [20••]. Perhaps its biggest downside is that manual patch clamping is also tedious and slow. In contrast, commercial 96-well and 384-well automatic patch clamp devices based on glass pipets or planar electrodes are available, the latter having been utilized on commercial hPSC-CMs with an average success rate of 46 % and mean seal resistance of 1.4 GΩ with mean stability of ~18 min [78, 79]. However, these automated systems have traditionally been employed for voltage-clamp recordings of individual currents in heterologous cell lines and may be less applicable for differentiated cells derived from hPSCs [80]. One possible advantage of patch clamp electrophysiology lies in the ability to directly augment the hPSC-CM physiology, such as by “electronically injecting” rectifier current IK1 using a dynamic clamp to model a more mature state [37]. The dynamic clamp may be an improvement over other studies that have employed negative current injection throughout the cardiac cycle to maintain diastolic potentials at more “adult” values between −80 and −100 mV [63, 35]. Finally, the successful throughput of all patch electrophysiology is critically dependent on membrane quality, which varies between cellular preparations. Particularly disadvantageous for drug studies, patch clamp electrophysiology tends to be limited in the capacity for prolonged or serial recordings.
Multi-electrode arrays
Another measure of the electrical activity which has grown in popularity is use of multi-electrode arrays (MEA) [81–83, 21]. MEAs consist of planar arrangements of electrodes that capture local extracellular field potentials from multicellular CM aggregates. As these systems permit simultaneous recordings from multiple electrodes, they are suited for high-throughput measurements, while their non-invasive nature facilitates serial or prolonged recordings. However, compared to traditional patch-clamp electrophysiology, MEAs do not yield as much direct information about the morphology of cellular action potentials, and recording is restricted to multicellular aggregates large enough to generate sufficient electrical signals.
Contractile motion/impedance analysis
Contractile motion and beat rate variability [84] can be measured non-invasively using impedance recordings, and systems for high-throughput have been commercially developed. Guo and colleagues [40] developed an assay utilizing confluent monolayers of hPSC-CMs. These authors assayed a panel of 83 drugs for concentrations that elicited arrhythmic beating and/or slowing of beat rate, noting that the resulting concentration correlated with clinical arrhythmia and QT-prolongation levels. Similar to MEA-based recording, impedance-based assays are non-invasive and amenable to high-throughput. However, while the contractile properties of cardiomyocytes correlate with their electrophysiology, the electromechanical coupling of hPSC-CMs is not as mature as their adult counterparts. Furthermore, the effects of some compounds that harbor independent or differential contractile vs. electrical toxicity may produce readouts that confound measurements of “true” arrhythmogenicity.
Fluorescent optical mapping
Fluorescent Optical “mapping” with voltage-sensitive and calcium-sensitive indicators is a well-established method of imaging electrical activity [85•]. AP morphology and wavefront propagation in whole hearts has been performed with the zwitterionic membrane dyes, di-4-ANEPPS and di-8-ANEPPS for years [85•]. These probes were recently combined with Ca2+ imaging dyes and used to simultaneously image electrical and calcium dynamics in hPSC-CM monolayers [72••]. However, the ANEPPS dyes feature prominent phototoxicity and photobleaching that limit cumulative illumination and degrades signal quality [86]. Furthermore, these dyes have had limited application to the study of isolated cells [87]. While the newer red-shifted probe di-4-ANBDQBS features a higher signal quality, its adequacy has yet to be evaluated for use with hPSC-CMs. In contrast, Ca2+-based dyes such as fluo-4 and rhod-2 have much lower toxicity and higher fluorescence [88]. Some hPSC-CM screens have used high-throughput imaging systems with fluo4-based Ca2+ transients as a surrogate reporter for electrical activity [89, 90]. In addition to small-molecule based membrane dyes, the field of genetically encoded voltage [91–94] and calcium sensors [95] has been evolving rapidly. One of these voltage indicators, A242-ArcLight, was recently demonstrated to robustly report electrical activity in hPSC-CMs, with a signal to noise ratio of ~22 dB when measured in single isolated cells with standard fluorescence microscopy [96]. As genetically encoded probes continue to offer improved signal-to-noise quality, photostability, and reduced toxicity, they may find a place in the non-invasive characterization of electrophysiology in hPSC-CM models.
Screening in practice
Once a suitable method of phenotypic evaluation is chosen, how would a high-throughput drug screen be conceived in practice? Numerous chemical libraries are commercially available for the purpose of drug screening, from small collections of characterized compounds to larger libraries of up to ~1 million compounds with uncharacterized function. As hPSC-CM screening technology develops, initial efforts may be geared towards smaller libraries of characterized molecules where “hits” are more easily clinically translatable, such as from collections of US Food and Drug Administration (FDA) approved drugs. To minimize the confounding influence of cellular heterogeneity and experimental variation, multiple measurements could be taken for each condition or drug concentration. Data may also be recorded pre-drug and post-drug application, with each cell or cell aggregate being its own control. In the case of fluorescence evaluations and MEA recordings, cells may be arrayed multi-well configurations, with up to hundreds of cells or cell aggregates per well. For measures such as action potential duration having inherent rate dependence, rate control, such as by electric field stimulation, could be considered.
Furthermore, to ensure reproducibility, the relative purity of the cell type of interest should be considered. hPSC-CMs destined for screening purposes might be either freed of contaminating non-cardiomyocyte cell types prior to plating, or marked by fluorescent reporters that define and validate their identity as specific cell types. For example, reporters for the ventricular myosin light chain-2-isoform (MLC-2v), which has been demonstrated to be a marker for hPSCs committed to the ventricular lineage, could be utilized to obtain a ventricular lineage [63]. Similarly, as βMHC is the dominant isoform of the myosin heavy chain in the adult heart and detectable in EBs at around 90 days or later [65•], it may be suitable as a marker of relative maturity. In addition to purity, future screens should optimize and report information on cell quality. It is foreseeable that the field may advance beyond small panels of expression markers and instead rely on multiple parameters, including incorporating functional ones towards an overall assessment of cell quality. Recently, Sheehy and colleagues [97] devised a 64-parameter quality assessment on commercially available murine pluripotent stem cell-derived cardiomyocytes based on structural (i.e., sarcomere alignment), electrophysiological (i.e., AP duration), and contractile performance, in comparison to freshly isolated neonatal ventricular myocytes.
Going forward
Current cardiotoxicity evaluations with animal cells and IKr/hERG-expressing heterologous systems have limitations in their prediction of clinical toxicology. Compared to heterologous cell lines, hPSC-CM technology recapitulates at least some of the native milieu and intricacies of human cardiomyocytes, and thus holds promise to be a valuable tool drug discovery and disease modeling. Notably, the FDA and the Health and Environmental Sciences Institute (HESI) recently laid out a new paradigm for preclinical cardiotoxicity testing, which includes tests on stem-cell–derived cardiomyocytes [98]. However, efforts to date have been limited both by the variability, immaturity, and heterogeneity of cardiomyocytes produced by current protocols and by the labor-intensive nature of conventional phenotyping. In particular, the immature nature of the present iteration of hPSC-CM technology is a major barrier to clinical translatability. As techniques for the production of hPSC-CMs continue to advance and standardize, and we gain a clearer understanding of the reproducibility and predictive power of hPSC-CM assays, we can begin to use hPSC-CM screens in a widespread fashion. This will represent a significant advance, moving cardiovascular medicine closer to personalized therapies and targeted therapeutics.
Acknowledgments
The authors thank Drs. Nathan Tucker, Robert Mills, and Patrick Ellinor for critical reading of this manuscript and helpful comments. This works was supported by the Corrigan Minehan Foundation (DJM), Harvard Stem Cell Institute (DJM), and NIH grants R01HL109004 (DJM) and T32HL007208 (JLM).
Footnotes
Conflict of interest
Dr. Jordan S. Leyton-Mange and Dr. David J. Milan each declare no potential conflicts of interest.
Human and Animal Rights and Informed Consent
This article does not contain any studies with human or animal subjects performed by any of the authors.
References
* Of importance
** Of major importance
- 1.Maltsev VA, Wobus AM, Rohwedel J, Bader M, Hescheler J. Cardiomyocytes differentiated in vitro from embryonic stem cells developmentally express cardiac-specific genes and ionic currents. Circ Res. 1994;75(2):233–244. doi: 10.1161/01.res.75.2.233. [DOI] [PubMed] [Google Scholar]
- 2. Kehat I, Kenyagin-Karsenti D, Snir M, Segev H, Amit M, Gepstein A, et al. Human embryonic stem cells can differentiate into myocytes with structural and functional properties of cardiomyocytes. J Clin Investig. 2001;108(3):407–414. doi: 10.1172/JCI12131. Initial demonstration of human embryonic stem cells differentiated by the embryonic body method can form functional cardiomyocytes.
- 3.Xu C, Police S, Rao N, Carpenter MK. Characterization and enrichment of cardiomyocytes derived from human embryonic stem cells. Circ Res. 2002;91(6):501–508. doi: 10.1161/01.res.0000035254.80718.91. [DOI] [PubMed] [Google Scholar]
- 4. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126(4):663–676. doi: 10.1016/j.cell.2006.07.024. Nobel-prize winning work from the Yamanaka lab demonstrating that mature somatic cells can be reprogrammed to a pluripotent state.
- 5. Cong L, Ran FA, Cox D, Lin S, Barretto R, Habib N, et al. Multiplex genome engineering using CRISPR/Cas systems. Science. 2013;339(6121):819–823. doi: 10.1126/science.1231143. Independently, members of the Church and Zhang labs (see reference #6 below) demonstrated use of the CRISPR/Cas9 system in targeted mutagenesis in mammalian cells, marking a new age in genome editing technology.
- 6.Mali P, Yang L, Esvelt KM, Aach J, Guell M, DiCarlo JE, et al. RNA-guided human genome engineering via Cas9. Science. 2013;339(6121):823–826. doi: 10.1126/science.1232033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Musunuru K. Genome editing of human pluripotent stem cells to generate human cellular disease models. Disease models & mechanisms. 2013;6(4):896–904. doi: 10.1242/dmm.012054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Moretti A, Bellin M, Welling A, Jung CB, Lam JT, Bott-Flugel L, et al. Patient-specific induced pluripotent stem-cell models for long-QT syndrome. New Engl J Med. 2010;363(15):1397–1409. doi: 10.1056/NEJMoa0908679. Initial demonstration of an arrhythmogenic disease, in this case LQTI, modeled in human iPSC-derived cardiomyocytes.
- 9.Itzhaki I, Maizels L, Huber I, Zwi-Dantsis L, Caspi O, Winterstern A, et al. Modelling the long QT syndrome with induced pluripotent stem cells. Nature. 2011;471(7337):225–229. doi: 10.1038/nature09747. [DOI] [PubMed] [Google Scholar]
- 10.Matsa E, Rajamohan D, Dick E, Young L, Mellor I, Staniforth A, 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(8):952–962. doi: 10.1093/eurheartj/ehr073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Yazawa M, Hsueh B, Jia X, Pasca AM, Bernstein JA, Hallmayer J, et al. Using induced pluripotent stem cells to investigate cardiac phenotypes in Timothy syndrome. Nature. 2011;471(7337):230–234. doi: 10.1038/nature09855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Davis RP, Casini S, van den Berg CW, Hoekstra M, Remme CA, Dambrot C, et al. Cardiomyocytes derived from pluripotent stem cells recapitulate electrophysiological characteristics of an overlap syndrome of cardiac sodium channel disease. Circulation. 2012;125(25):3079–3091. doi: 10.1161/CIRCULATIONAHA.111.066092. [DOI] [PubMed] [Google Scholar]
- 13.Itzhaki I, Maizels L, Huber I, Gepstein A, Arbel G, Caspi O, et al. Modeling of catecholaminergic polymorphic ventricular tachycardia with patient-specific human-induced pluripotent stem cells. J Am Coll Cardiol. 2012;60(11):990–1000. doi: 10.1016/j.jacc.2012.02.066. [DOI] [PubMed] [Google Scholar]
- 14.Kim C, Wong J, Wen J, Wang S, Wang C, Spiering S, et al. Studying arrhythmogenic right ventricular dysplasia with patient-specific iPSCs. Nature. 2013;494(7435):105–110. doi: 10.1038/nature11799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Ma D, Wei H, Lu J, Ho S, Zhang G, Sun X, 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(15):1122–1133. doi: 10.1093/eurheartj/ehs226. [DOI] [PubMed] [Google Scholar]
- 16.Roden DM, Hong CC. Stem cell-derived cardiomyocytes as a tool for studying proarrhythmia: a better canary in the coal mine? Circulation. 2013;127(16):1641–1643. doi: 10.1161/CIRCULATIONAHA.113.002127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Sun N, Yazawa M, Liu J, Han L, Sanchez-Freire V, Abilez OJ, et al. Patient-specific induced pluripotent stem cells as a model for familial dilated cardiomyopathy. Science Translational Medicine. 2012;4(130):130ra47. doi: 10.1126/scitranslmed.3003552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Lan F, Lee AS, Liang P, Sanchez-Freire V, Nguyen PK, Wang L, et al. Abnormal calcium handling properties underlie familial hypertrophic cardiomyopathy pathology in patient-specific induced pluripotent stem cells. Cell Stem Cell. 2013;12(1):101–113. doi: 10.1016/j.stem.2012.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Roden DM. Drug-induced prolongation of the QT interval. New Engl J Med. 2004;350(10):1013–1022. doi: 10.1056/NEJMra032426. [DOI] [PubMed] [Google Scholar]
- 20. Liang P, Lan F, Lee AS, Gong T, Sanchez-Freire V, Wang Y, et al. Drug screening using a library of human induced pluripotent stem cell-derived cardiomyocytes reveals disease-specific patterns of cardiotoxicity. Circulation. 2013;127(16):1677–1691. doi: 10.1161/CIRCULATIONAHA.113.001883. Using patch clamp electrophysiology, the Wu laboratory at Stanford compared multiple human iPSC-CMs: those derived from patients with the LQT1 syndrome, hypertrophic cardiomyopathy (β-MHC mutation), and dilated cardiomyopathy (TNNT2 mutation), and found drug toxicity effects (i.e., EADs in LQT1 cells) in increased frequency in diseased lines compared to lines created from sibling controls or ES cells.
- 21.Khan JM, Lyon AR, Harding SE. The case for induced pluripotent stem cell-derived cardiomyocytes in pharmacological screening. Br J Pharmacol. 2013;169(2):304–317. doi: 10.1111/j.1476-5381.2012.02118.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Mercola M, Colas A, Willems E. Induced pluripotent stem cells in cardiovascular drug discovery. Circ Res. 2013;112(3):534–548. doi: 10.1161/CIRCRESAHA.111.250266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Zhang J, Klos M, Wilson GF, Herman AM, Lian X, Raval KK, et al. Extracellular matrix promotes highly efficient cardiac differentiation of human pluripotent stem cells: the matrix sandwich method. Circ Res. 2012;111(9):1125–1136. doi: 10.1161/CIRCRESAHA.112.273144. The "matrix-sandwich" method is a highly preferred method of inducing cardiac differentaition in a monolayer format in hPSCs, with efficiencies as high as >80 %.
- 24.Lian X, Hsiao C, Wilson G, Zhu K, Hazeltine LB, Azarin SM, et al. Robust cardiomyocyte differentiation from human pluripotent stem cells via temporal modulation of canonical Wnt signaling. Proc Natl Acad Sci. 2012;109(27):E1848–E1857. doi: 10.1073/pnas.1200250109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Chen G, Gulbranson DR, Hou Z, Bolin JM, Ruotti V, Probasco MD, et al. Chemically defined conditions for human iPSC derivation and culture. Nature Methods. 2011;8(5):424–429. doi: 10.1038/nmeth.1593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Magyar J, Iost N, Kortvely A, Banyasz T, Virag L, Szigligeti P, et al. Effects of endothelin-1 on calcium and potassium currents in undiseased human ventricular myocytes. Pflugers Archiv. 2000;441(1):144–149. doi: 10.1007/s004240000400. [DOI] [PubMed] [Google Scholar]
- 27. Hoekstra M, Mummery CL, Wilde AAM, Bezzina CR, Verkerk AO. Induced pluripotent stem-cell–derived cardiomyocytes as models for cardiac arrhythmias. Front Physiol. 2012;3 doi: 10.3389/fphys.2012.00346. One of the most comprehensive reviews of utilizing hPSC-CMs as models for arrhythmia. The authors review, individually, the present body of work of iPSC models for various LQT syndromes and CPVT. Authors also review evidence for the major cardiac currents and their respective properties in hPSC-CMs.
- 28.Peng S, Lacerda AE, Kirsch GE, Brown AM, Bruening-Wright A. The action potential and comparative pharmacology of stem cell-derived human cardiomyocytes. J Pharmacol Toxicol Meth. 2010;61(3):277–286. doi: 10.1016/j.vascn.2010.01.014. [DOI] [PubMed] [Google Scholar]
- 29. Mummery C, Ward-van Oostwaard D, Doevendans P, Spijker R, van den Brink S, Hassink R, et al. Differentiation of human embryonic stem cells to cardiomyocytes: role of coculture with visceral endoderm-like cells. Circulation. 2003;107(21):2733–2740. doi: 10.1161/01.CIR.0000068356.38592.68. The visceral endoderm coculture method of cardiomyocyte differentation from hPSCs.
- 30.Robertson C, Tran DD, George SC. Concise review: maturation phases of human pluripotent stem cell-derived cardiomyocytes. Stem Cells. 2013;31(5):829–837. doi: 10.1002/stem.1331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Bellin M, Casini S, Davis RP, D'Aniello C, Haas J, Ward-van Oostwaard D, et al. Isogenic human pluripotent stem cell pairs reveal the role of a KCNH2 mutation in long-QT syndrome. EMBO J. 2013;32(24):3161–3175. doi: 10.1038/emboj.2013.240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Knollmann BC. Induced pluripotent stem cell-derived cardiomyocytes: boutique science or valuable arrhythmia model? Circ Res. 2013;112(6):969–976. doi: 10.1161/CIRCRESAHA.112.300567. discussion 76. It has increasingly been recognized that genetic differences between iPSC and ESC lines may underlie variability in their respective phenotypes and drug responses. These authors generated isogenic control lines via genome editing by correcting a KCNH2 mutation in a LQT2 hiPSC line and introducing it into a WT hESC control line. By making comparisons within the same genetic background, such isogenic control strategies may emerge as a preferred method to compare phenotypic effects of mutations.
- 33.Zhang Q, Jiang J, Han P, Yuan Q, Zhang J, Zhang X, et al. Direct differentiation of atrial and ventricular myocytes from human embryonic stem cells by alternating retinoid signals. Cell Res. 2011;21(4):579–587. doi: 10.1038/cr.2010.163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Ma J, Guo L, Fiene SJ, Anson BD, Thomson JA, Kamp TJ, 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(5):H2006–H2017. doi: 10.1152/ajpheart.00694.2011. The January group rigorously evaluated various cardiac currents in cells from one single iPSC line from Cellular Dynamics, Madison, WI.
- 35.Satin J, Kehat I, Caspi O, Huber I, Arbel G, Itzhaki I, et al. Mechanism of spontaneous excitability in human embryonic stem-cell–derived cardiomyocytes. J Physiol. 2004;559(Pt 2):479–496. doi: 10.1113/jphysiol.2004.068213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Wang K, Terrenoire C, Sampson KJ, Iyer V, Osteen JD, Lu J, et al. Biophysical properties of slow potassium channels in human embryonic stem-cell–derived cardiomyocytes implicate subunit stoichiometry. J Physiol. 2011;589(Pt 24):6093–6104. doi: 10.1113/jphysiol.2011.220863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Bett GC, Kaplan AD, Lis A, Cimato TR, Tzanakakis ES, Zhou Q, et al. Electronic "expression" of the inward rectifier in cardiocytes derived from human-induced pluripotent stem cells. Heart Rhythm. 2013;10(12):1903–1910. doi: 10.1016/j.hrthm.2013.09.061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Itzhaki I, Rapoport S, Huber I, Mizrahi I, Zwi-Dantsis L, Arbel G, et al. Calcium handling in human induced pluripotent stem-cell–derived cardiomyocytes. PloS one. 2011;6(4):e18037. doi: 10.1371/journal.pone.0018037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Norstrom A, Akesson K, Hardarson T, Hamberger L, Bjorquist P, Sartipy P. Molecular and pharmacological properties of human embryonic stem cell-derived cardiomyocytes. Exp Biol Med (Maywood) 2006;231(11):1753–1762. doi: 10.1177/153537020623101113. [DOI] [PubMed] [Google Scholar]
- 40.Guo L, Coyle L, Abrams RM, Kemper R, Chiao ET, Kolaja KL. Refining the Human iPSC-Cardiomyocyte Arrhythmic Risk Assessment Model. Toxicol Sci. 2013;136(2):581–594. doi: 10.1093/toxsci/kft205. [DOI] [PubMed] [Google Scholar]
- 41.Sheng X, Reppel M, Nguemo F, Mohammad FI, Kuzmenkin A, Hescheler J, et al. Human pluripotent stem cell-derived cardiomyocytes: response to TTX and lidocain reveals strong cell to cell variability. PloS one. 2012;7(9):e45963. doi: 10.1371/journal.pone.0045963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Egashira T, Yuasa S, Suzuki T, Aizawa Y, Yamakawa H, Matsuhashi T, et al. Disease characterization using LQTS-specific induced pluripotent stem cells. Cardiovasc Res. 2012;95(4):419–429. doi: 10.1093/cvr/cvs206. [DOI] [PubMed] [Google Scholar]
- 43.Lahti AL, Kujala VJ, Chapman H, Koivisto AP, Pekkanen-Mattila M, Kerkela E, et al. Model for long QT syndrome type 2 using human iPS cells demonstrates arrhythmogenic characteristics in cell culture. Disease models & mechanisms. 2012;5(2):220–230. doi: 10.1242/dmm.008409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Ma D, Wei H, Zhao Y, Lu J, Li G, Sahib NB, et al. Modeling type 3 long QT syndrome with cardiomyocytes derived from patient-specific induced pluripotent stem cells. Int J Cardiol. 2013;168(6):5277–5286. doi: 10.1016/j.ijcard.2013.08.015. [DOI] [PubMed] [Google Scholar]
- 45.Fatima A, Kaifeng S, Dittmann S, Xu G, Gupta MK, Linke M, 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(12):e83005. doi: 10.1371/journal.pone.0083005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Terrenoire C, Wang K, Tung KW, Chung WK, Pass RH, Lu JT, et al. Induced pluripotent stem cells used to reveal drug actions in a long QT syndrome family with complex genetics. J Gen Physiol. 2013;141(1):61–72. doi: 10.1085/jgp.201210899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Jonsson MK, Vos MA, Mirams GR, Duker G, Sartipy P, de Boer TP, et al. Application of human stem cell-derived cardiomyocytes in safety pharmacology requires caution beyond hERG. J Mol Cell Cardiol. 2012;52(5):998–1008. doi: 10.1016/j.yjmcc.2012.02.002. [DOI] [PubMed] [Google Scholar]
- 48.Jung CB, Moretti A, Mederos y Schnitzler M, Iop L, Storch U, Bellin 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(3):180–191. doi: 10.1002/emmm.201100194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Kujala K, Paavola J, Lahti A, Larsson K, Pekkanen-Mattila M, Viitasalo M, et al. Cell model of catecholaminergic polymorphic ventricular tachycardia reveals early and delayed afterdepolarizations. PloS one. 2012;7(9):e44660. doi: 10.1371/journal.pone.0044660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Zhang XH, Haviland S, Wei H, Saric T, Fatima A, Hescheler J, 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(2):57–70. doi: 10.1016/j.ceca.2013.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Novak A, Barad L, Zeevi-Levin N, Shick R, Shtrichman R, Lorber A, et al. Cardiomyocytes generated from CPVTD307H patients are arrhythmogenic in response to beta-adrenergic stimulation. J Cell Mol Med. 2012;16(3):468–482. doi: 10.1111/j.1582-4934.2011.01476.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Satin J, Itzhaki I, Rapoport S, Schroder EA, Izu L, Arbel G, et al. Calcium handling in human embryonic stem cell-derived cardiomyocytes. Stem Cells. 2008;26(8):1961–1972. doi: 10.1634/stemcells.2007-0591. [DOI] [PubMed] [Google Scholar]
- 53.Caspi O, Huber I, Gepstein A, Arbel G, Maizels L, Boulos M, et al. Modeling of arrhythmogenic right ventricular cardiomyopathy with human induced pluripotent stem cells. Circulation Cardiovascular Genetics. 2013;6(6):557–568. doi: 10.1161/CIRCGENETICS.113.000188. [DOI] [PubMed] [Google Scholar]
- 54.Huang HP, Chen PH, Hwu WL, Chuang CY, Chien YH, Stone L, et al. Human Pompe disease-induced pluripotent stem cells for pathogenesis modeling, drug testing and disease marker identification. Hum Mol Genet. 2011;20(24):4851–4864. doi: 10.1093/hmg/ddr424. [DOI] [PubMed] [Google Scholar]
- 55.Zwi L, Caspi O, Arbel G, Huber I, Gepstein A, Park IH, et al. Cardiomyocyte differentiation of human induced pluripotent stem cells. Circulation. 2009;120(15):1513–1523. doi: 10.1161/CIRCULATIONAHA.109.868885. [DOI] [PubMed] [Google Scholar]
- 56.Mummery CL, Zhang J, Ng ES, Elliott DA, Elefanty AG, Kamp TJ. Differentiation of human embryonic stem cells and induced pluripotent stem cells to cardiomyocytes: a methods overview. Circ Res. 2012;111(3):344–358. doi: 10.1161/CIRCRESAHA.110.227512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Burridge PW, Thompson S, Millrod MA, Weinberg S, Yuan X, Peters A, et al. A universal system for highly efficient cardiac differentiation of human induced pluripotent stem cells that eliminates interline variability. PloS one. 2011;6(4):e18293. doi: 10.1371/journal.pone.0018293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Xu C, Police S, Hassanipour M, Li Y, Chen Y, Priest C, et al. Efficient generation and cryopreservation of cardiomyocytes derived from human embryonic stem cells. Regenerative Medicine. 2011;6(1):53–66. doi: 10.2217/rme.10.91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Paige SL, Osugi T, Afanasiev OK, Pabon L, Reinecke H, Murry CE. Endogenous Wnt/beta-catenin signaling is required for cardiac differentiation in human embryonic stem cells. PloS one. 2010;5(6):e11134. doi: 10.1371/journal.pone.0011134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Cao N, Liu Z, Chen Z, Wang J, Chen T, Zhao X, et al. Ascorbic acid enhances the cardiac differentiation of induced pluripotent stem cells through promoting the proliferation of cardiac progenitor cells. Cell Res. 2012;22(1):219–236. doi: 10.1038/cr.2011.195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Chetty S, Pagliuca FW, Honore C, Kweudjeu A, Rezania A, Melton DA. A simple tool to improve pluripotent stem cell differentiation. Nature Methods. 2013;10(6):553–556. doi: 10.1038/nmeth.2442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Xu C. Differentiation and enrichment of cardiomyocytes from human pluripotent stem cells. J Mol Cell Cardiol. 2012;52(6):1203–1212. doi: 10.1016/j.yjmcc.2012.03.012. [DOI] [PubMed] [Google Scholar]
- 63.Bizy A, Guerrero-Serna G, Hu B, Ponce-Balbuena D, Willis BC, Zarzoso M, et al. Myosin light chain 2-based selection of human iPSC-derived early ventricular cardiac myocytes. Stem Cell Res. 2013;11(3):1335–1347. doi: 10.1016/j.scr.2013.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Elliott DA, Braam SR, Koutsis K, Ng ES, Jenny R, Lagerqvist EL, et al. NKX2–5(eGFP/w) hESCs for isolation of human cardiac progenitors and cardiomyocytes. Nature Methods. 2011;8(12):1037–1040. doi: 10.1038/nmeth.1740. [DOI] [PubMed] [Google Scholar]
- 65. Kita-Matsuo H, Barcova M, Prigozhina N, Salomonis N, Wei K, Jacot JG, et al. Lentiviral vectors and protocols for creation of stable hESC lines for fluorescent tracking and drug resistance selection of cardiomyocytes. PloS one. 2009;4(4):e5046. doi: 10.1371/journal.pone.0005046. Enrichment of cardiomyocyte populations from heterogeneous differentiated preparations of hPSCs can be done by various methods. Transgenic a-MHC promoter driven selectional markers is a common preferred method across many groups.
- 66.Tohyama S, Hattori F, Sano M, Hishiki T, Nagahata Y, Matsuura T, et al. Distinct metabolic flow enables large-scale purification of mouse and human pluripotent stem cell-derived cardiomyocytes. Cell Stem Cell. 2013;12(1):127–137. doi: 10.1016/j.stem.2012.09.013. [DOI] [PubMed] [Google Scholar]
- 67.Hattori F, Chen H, Yamashita H, Tohyama S, Satoh YS, Yuasa S, et al. Nongenetic method for purifying stem cell-derived cardiomyocytes. Nature Methods. 2010;7(1):61–66. doi: 10.1038/nmeth.1403. [DOI] [PubMed] [Google Scholar]
- 68.Dubois NC, Craft AM, Sharma P, Elliott DA, Stanley EG, Elefanty AG, et al. SIRPA is a specific cell-surface marker for isolating cardiomyocytes derived from human pluripotent stem cells. Nat Biotechnol. 2011;29(11):1011–1018. doi: 10.1038/nbt.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Uosaki H, Fukushima H, Takeuchi A, Matsuoka S, Nakatsuji N, Yamanaka S, et al. Efficient and scalable purification of cardiomyocytes from human embryonic and induced pluripotent stem cells by VCAM1 surface expression. PloS one. 2011;6(8):e23657. doi: 10.1371/journal.pone.0023657. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Nunes SS, Miklas JW, Liu J, Aschar-Sobbi R, Xiao Y, Zhang B, et al. Biowire: a platform for maturation of human pluripotent stem cell-derived cardiomyocytes. Nature Methods. 2013;10(8):781–787. doi: 10.1038/nmeth.2524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Lieu DK, Fu JD, Chiamvimonvat N, Tung KC, McNerney GP, Huser T, et al. Mechanism-based facilitated maturation of human pluripotent stem cell-derived cardiomyocytes. Circulation Arrhythmia and Electrophysiology. 2013;6(1):191–201. doi: 10.1161/CIRCEP.111.973420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Lee P, Klos M, Bollensdorff C, Hou L, Ewart P, Kamp TJ, et al. Simultaneous voltage and calcium mapping of genetically purified human induced pluripotent stem cell-derived cardiac myocyte monolayers. Circ Res. 2012;110(12):1556–1563. doi: 10.1161/CIRCRESAHA.111.262535. Fluorescent indicators of voltage and calcium dynamics may emerge as a preferred, non-invasive method for hPSC-CM phenotyping, as undertaken in cell monolayers in this study by Lee and colleagues.
- 73.Mitcheson JS, Hancox JC, Levi AJ. Cultured adult cardiac myocytes: future applications, culture methods, morphological and electrophysiological properties. Cardiovasc Res. 1998;39(2):280–300. doi: 10.1016/s0008-6363(98)00128-x. [DOI] [PubMed] [Google Scholar]
- 74.Louch WE, Sheehan KA, Wolska BM. Methods in cardiomyocyte isolation, culture, and gene transfer. J Mol Cell Cardiol. 2011;51(3):288–298. doi: 10.1016/j.yjmcc.2011.06.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Kim C, Majdi M, Xia P, Wei KA, Talantova M, Spiering S, et al. Non-cardiomyocytes influence the electrophysiological maturation of human embryonic stem cell-derived cardiomyocytes during differentiation. Stem Cell Dev. 2010;19(6):783–795. doi: 10.1089/scd.2009.0349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Uesugi M, Ojima A, Taniguchi T, Miyamoto N, Sawada K. Low-density plating is sufficient to induce cardiac hypertrophy and electrical remodeling in highly purified human iPS cell-derived cardiomyocytes. J Pharmacol Toxicol Meth. 2013 doi: 10.1016/j.vascn.2013.11.002. [DOI] [PubMed] [Google Scholar]
- 77.Neher E, Sakmann B. Single-channel currents recorded from membrane of denervated frog muscle fibres. Nature. 1976;260(5554):799–802. doi: 10.1038/260799a0. [DOI] [PubMed] [Google Scholar]
- 78.Milligan CJ, Moller C. Automated planar patch-clamp. Methods Mol Biol. 2013;998:171–187. doi: 10.1007/978-1-62703-351-0_13. [DOI] [PubMed] [Google Scholar]
- 79.Stoelzle S, Obergrussberger A, Bruggemann A, Haarmann C, George M, Kettenhofen R, et al. State-of-the-Art Automated Patch Clamp Devices: Heat Activation, Action Potentials, and High Throughput in Ion Channel Screening. Front Pharmacol. 2011;2:76. doi: 10.3389/fphar.2011.00076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Yajuan X, Xin L, Zhiyuan L. A comparison of the performance and application differences between manual and automated patch-clamp techniques. Curr Chem Genom. 2012;6:87–92. doi: 10.2174/1875397301206010087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Harris K, Aylott M, Cui Y, Louttit JB, McMahon NC, Sridhar A. Comparison of electrophysiological data from human-induced pluripotent stem cell-derived cardiomyocytes to functional preclinical safety assays. Toxicol Sci. 2013;134(2):412–426. doi: 10.1093/toxsci/kft113. [DOI] [PubMed] [Google Scholar]
- 82.Igelmund P, Fleischmann BK, Fischer IR, Soest J, Gryshchenko O, Bohm-Pinger MM, et al. Action potential propagation failures in long-term recordings from embryonic stem cell-derived cardiomyocytes in tissue culture. Pflugers Archiv. 1999;437(5):669–679. doi: 10.1007/s004240050831. [DOI] [PubMed] [Google Scholar]
- 83.Caspi O, Itzhaki I, Kehat I, Gepstein A, Arbel G, Huber I, et al. In vitro electrophysiological drug testing using human embryonic stem-cell–derived cardiomyocytes. Stem Cell Dev. 2009;18(1):161–172. doi: 10.1089/scd.2007.0280. [DOI] [PubMed] [Google Scholar]
- 84.Mandel Y, Weissman A, Schick R, Barad L, Novak A, Meiry G, et al. Human embryonic and induced pluripotent stem cell-derived cardiomyocytes exhibit beat rate variability and power-law behavior. Circulation. 2012;125(7):883–893. doi: 10.1161/CIRCULATIONAHA.111.045146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Herron TJ, Lee P, Jalife J. Optical imaging of voltage and calcium in cardiac cells & tissues. Circ Res. 2012;110(4):609–623. doi: 10.1161/CIRCRESAHA.111.247494. A comprehensive and up-to-date review of optical mapping of voltage and calcium signals using small molecule dyes in cardiac tissue and cells.
- 86.Schaffer P, Ahammer H, Muller W, Koidl B, Windisch H. Di-4-ANEPPS causes photodynamic damage to isolated cardiomyocytes. Pflugers Archiv. 1994;426(6):548–551. doi: 10.1007/BF00378533. [DOI] [PubMed] [Google Scholar]
- 87.Hardy ME, Lawrence CL, Standen NB, Rodrigo GC. Can optical recordings of membrane potential be used to screen for drug-induced action potential prolongation in single cardiac myocytes? J Pharmacol Toxicol Meth. 2006;54(2):173–182. doi: 10.1016/j.vascn.2006.02.013. [DOI] [PubMed] [Google Scholar]
- 88.Liau B, Zhang D, Bursac N. Functional cardiac tissue engineering. Regenerative Medicine. 2012;7(2):187–206. doi: 10.2217/rme.11.122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Sirenko O, Crittenden C, Callamaras N, Hesley J, Chen YW, Funes C, et al. Multiparameter in vitro assessment of compound effects on cardiomyocyte physiology using iPSC cells. J Biomol Screen. 2013;18(1):39–53. doi: 10.1177/1087057112457590. [DOI] [PubMed] [Google Scholar]
- 90.Cerignoli F, Charlot D, Whittaker R, Ingermanson R, Gehalot P, Savchenko A, et al. High throughput measurement of Ca(2)(+) dynamics for drug risk assessment in human stem cell-derived cardiomyocytes by kinetic image cytometry. J Pharmacol Toxicol Meth. 2012;66(3):246–256. doi: 10.1016/j.vascn.2012.08.167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Jin L, Han Z, Platisa J, Wooltorton JR, Cohen LB, Pieribone VA. Single action potentials and subthreshold electrical events imaged in neurons with a fluorescent protein voltage probe. Neuron. 2012;75(5):779–785. doi: 10.1016/j.neuron.2012.06.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Cao G, Platisa J, Pieribone VA, Raccuglia D, Kunst M, Nitabach MN. Genetically targeted optical electrophysiology in intact neural circuits. Cell. 2013;154(4):904–913. doi: 10.1016/j.cell.2013.07.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Kralj JM, Douglass AD, Hochbaum DR, Maclaurin D, Cohen AE. Optical recording of action potentials in mammalian neurons using a microbial rhodopsin. Nature Methods. 2012;9(1):90–95. doi: 10.1038/nmeth.1782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Tsutsui H, Jinno Y, Tomita A, Niino Y, Yamada Y, Mikoshiba K, et al. Improved detection of electrical activity with a voltage probe based on a voltage-sensing phosphatase. J Physiol. 2013;591(Pt 18):4427–4437. doi: 10.1113/jphysiol.2013.257048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Zhao Y, Araki S, Wu J, Teramoto T, Chang YF, Nakano M, et al. An expanded palette of genetically encoded Ca(2)(+) indicators. Science. 2011;333(6051):1888–1891. doi: 10.1126/science.1208592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Leyton-Mange JS, Mills RW, Macri VS, Jang MY, Butte FN, Ellinor PT, et al. Rapid Cellular Phenotyping of Human Pluripotent Stem Cell-Derived Cardiomyocytes using a Genetically Encoded Fluorescent Voltage Sensor. Stem Cell Reports. 2014;2(2):163–170. doi: 10.1016/j.stemcr.2014.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Sheehy SP, Pasqualini F, Grosberg A, Park SJ, Aratyn-Schaus Y, Parker KK. Quality metrics for stem cell-derived cardiac myocytes. Stem Cell Reports. 2014;2(3):282–294. doi: 10.1016/j.stemcr.2014.01.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Chi KR. Revolution dawning in cardiotoxicity testing. Nat Rev Drug Discov. 2013;12(8):565–567. doi: 10.1038/nrd4083. [DOI] [PubMed] [Google Scholar]
- 99.Mehta A, Sequiera GL, Ramachandra CJ, Sudibyo Y, Chung Y, Jingwei S, et al. Re-trafficking of HERG Reverses Long QT Syndrome 2 Phenotype in Human iPS-derived Cardiomyocytes. Cardiovasc Res. 2014 doi: 10.1093/cvr/cvu060. [DOI] [PubMed] [Google Scholar]
- 100.Fatima A, Xu G, Shao K, Papadopoulos S, Lehmann M, Arnaiz-Cot JJ, et al. In vitro modeling of ryanodine receptor 2 dysfunction using human induced pluripotent stem cells. Cell Physiol Biochem: international journal of experimental cellular physiology, biochemistry, and pharmacology. 2011;28(4):579–592. doi: 10.1159/000335753. [DOI] [PMC free article] [PubMed] [Google Scholar]