Abstract
Life threatening ventricular arrhythmias leading to sudden cardiac death (SCD) are a major cause of morbidity and mortality. In the absence of structural heart disease, these arrhythmias, especially in the younger population are often an outcome of genetic defects in specialized membrane proteins called ‘ion channels’. In the heart, exceptionally well-orchestrated activity of a diversity of ion channels mediates the cardiac action potential. Alterations in either the function or expression of these channels can disrupt the configuration of the action potential, leading to abnormal electrical activity of the heart that can sometimes initiate an arrhythmia.
Understanding the pathophysiology of inherited arrhythmias can be challenging due to the complexity of the disorder and lack of appropriate cellular and in vivo models. Recent advances in human induced pluripotent stem cell (hiPSC) technology have provided remarkable progress in comprehending the underlying mechanisms of ion channel disorders or ‘channelopathies’ by modeling these complex arrhythmia syndromes in vitro in a dish. To fully realize the potential of iPSCs in elucidating the mechanistic basis and complex pathophysiology of channelopathies, it is crucial to have a basic knowledge of cardiac myocyte electrophysiology. In this review, we will discuss the role of the various ion channels in cardiac electrophysiology and the molecular and cellular mechanisms of arrhythmias, highlighting the promise of hiPSC-CMs as a model for investigating inherited arrhythmia syndromes and testing antiarrhythmic strategies. Overall, this review aims to provide a basic understanding of the electrical activity of the heart and related channelopathies, especially to clinicians or research scientists in the cardiovascular field with limited electrophysiology background.
Keywords: Action potential, ionic currents, channelopathies, electrophysiology, arrhythmia, induced pluripotent stem cells
Introduction
In a subset of individuals that are relatively young and otherwise healthy, life threatening ventricular arrhythmia can develop as a result of mutations in genes encoding ion channels, transporters, and accessory subunits that lead to altered cardiac electrophysiology1, 2.
Over the past two decades, significant progress has been made in the identification of genetic defects underlying many recognized types of inherited arrhythmia syndromes (IAS). However, for most of these inherited disorders, clinical management is still hindered by insufficient knowledge of the functional consequences of the candidate mutation. This is largely due to lack of suitable model systems that appropriately mimic human cardiac electrophysiology. For example, widely used transgenic mice have a heart rate of about eight times faster than that of humans (500 vs. 60 beats/min), and the action potential duration (APD) in mouse CMs is relatively shorter secondary to differences in K+ currents3. Additionally, variable disease expressivity and sensitivity to therapeutic interventions between mutations and/or patients necessitate more personalized treatment. The recent emergence of induced pluripotent stem cell (iPSC) technology4, 5 has provided an unprecedented opportunity for generating and studying iPSC-derived cardiomyocytes from diseased patients. iPSC based studies allow investigation of patient- and/or mutation-specific disease mechanisms as well as identification and assessment of novel and/or custom tailored therapeutic strategies for arrhythmia syndromes.
This review provides a basic understanding of the fundamental electrophysiology of cardiac myocytes and the underlying mechanisms of IAS to help physicians and research scientists without expertise in this field to contribute towards the design of effective treatment strategies. The review will highlight the recent advances and opportunities from hiPSC-CMs models for investigating IAS and testing antiarrhythmic strategies.
Basis of Cardiomyocyte Excitability - Origin of the Membrane Potential
The heart is an electromechanical organ that pumps blood throughout the body undergoing roughly three billion cycles in an average lifetime6. To perform this function in the most efficient way, the heart contracts and relaxes in a highly coordinated way (both spatially and temporally) during each heartbeat referred to as a cardiac cycle. Each cardiac cycle starts with an electrical impulse, originating from a group of specialized pacemaker cells called the sinoatrial node (SAN), which subsequently propagates throughout the heart. This electrical excitation of the heart is made possible by an electrochemical gradient that exists across the membrane of each heart muscle cell or cardiomyocyte (CM)6.
The lipid bilayer membrane of a CM is embedded with integral proteins called ion channels that facilitate the passive diffusion of ions across the hydrophobic cell membrane. Ion channels are generally characterized by two important properties: 1) gating - ion channels open and close in response to a specific stimulus either binding of a ligand (e.g. neurotransmitters) or changes in the transmembrane (TM) potential; 2) selectivity - ion channels allow a high rate of selective passage of only a particular species of ions such as K+, Na+, Cl− or Ca2+. The flux rate of channels is quite high, equivalent to 107 ions for a single monovalent channel current magnitude of 1 pA. These properties of ion channels coupled with the presence of large negatively charged molecules (mainly proteins) inside the cell and energy-requiring activity of membrane pumps (primarily the Na+/K+-ATPase and Ca2+-ATPase) results in an unequal distribution of charged ions across the membrane (Figure 1A) generating an electrochemical gradient which sets the membrane potential (MP)7.
Figure 1. Electrophysiological Basis of the Electrical Activity in the Heart.

(A) Schematic for ion channel proteins embedded in a lipid bilayer and ionic basis for resting membrane potential in a cardiac myocyte. Based on the depicted concentration of cations, the calculated equilibrium potential (E = (RT/zF) * ln [X]out/[X]inside) for K+, Na+ and Ca2+ would be approx. −90 mV, +50 mV and +130 mV at 25oC, respectively. (B) Schematic depicting different phases (0–4) of a typical ventricular action potential (top) with various depolarizing (arrows down) and repolarizing (arrows up) ionic currents. Bottom panel shows the relative amount of different currents between an adult ventricular myocyte (gray) and a ventricular-like hiPSC-CM (red). The associated ion channel abnormalities are also shown. Purple circles represent loss-of-function mutations and blue circles represent gain-of-function mutations. (C) Excitation-contraction coupling in a cardiac myocyte. Ca2+ entering via plasma membrane channels activates ryanodine receptors (ryanodine receptor 2 [RYR2]) and initiates Ca2+-induced Ca2+-release mechanism (CICR). CASQ2 is the cardiac isoform of the high-capacity Ca2+-sequestering protein, calsequestrin present inside SR. Phospholamban (PLN, brown oval) is shown on top of Sarco/endoplasmic reticulum Ca2+-ATPase type-2a (SERCA 2a). Mutations in proteins associated with SR Ca2+ release or uptake are associated with CPVT.
When ion channels open, they bias the MP of the cell towards the equilibrium potential of the permeant ion. The equilibrium potential for each ion, which can be calculated by the Nernst equation, is the MP where the chemical and the electrical forces are in balance such that there is no net flow of ions through the open channel. For example, under resting conditions, an atrial or ventricular myocyte is permeable to K+ because a specific type of K+ channel (Kir2.1) is constitutively in an open state, whereas other types of ion channels are closed unless activated by a change in MP. For this reason, the resting membrane potential (RMP) of a resting (i.e. between beats) CM is steered towards the K+ equilibrium potential of −90 mV. By convention, MP is measured as the voltage of the intracellular side of the membrane with respect to a ground electrode located outside the cell7.
The direction of ion currents conducted by an ion channel (into the cell [inward] or out of the cell [outward]) is determined by the electrochemical gradient of the permeant ions. For example, outward flow of K+ ions through K+ channels drive the myocyte towards −90 mV while inward flow of Na+ ions through opening of voltage gated Na+ channels forces the MP towards a positive value (up to +40 mV).
Action Potential of a Human Cardiac Myocyte
An action potential (AP) is generated by the orchestrated opening and closing of multiple ion channels present in the plasma membrane of an individual CM that conduct depolarizing, inward (Na+ and Ca2+) and repolarizing, outward (K+) currents8. The AP waveform in different regions of the heart is distinct owing to differences in the expression and/or the properties of the underlying ion channels. These differences and the unique architecture of the cardiac conduction system (AV node and His-Purkinje system) contribute to the normal unidirectional propagation of excitation through the atria to the ventricles and to the generation of normal cardiac rhythms in the heart9, 10.
Figure 1B illustrates the five phases (0 – 4) of a ventricular AP. Phase 4 represents the resting state in a normal working CM wherein the RMP is ~ −90 mV. During phase 0, a depolarizing stimulus from a neighboring cell elevates the RMP to the threshold at which voltage-gated Na+ (NaV) channels open, allowing Na+ ions to rapidly diffuse into the cell down their electrochemical gradient. The rapid phase 0 depolarization shifts the MP towards the Na+ equilibrium potential of +40 mV and is responsible for the rapid propagation of the cardiac impulse. Within a few ms after activation, Na+ channels rapidly inactivate and thus are unable to conduct Na+. The depolarized MP results in a brief activation of the transient outward potassium current (Ito), that in turn leads to a brief, and only partial repolarization (phase 1). Phase 2 is the characteristic prolonged “plateau phase” of the CM AP, where inward and outward currents are nearly equal (hence only a minor change in MP). During phase 2, activation of voltage-gated L-type Ca2+ channels (LTCC) leads to the entry of Ca2+ into the cell, which in turn activates nearby intracellular ryanodine receptors (RyR2) present in the membrane of the sarcoplasmic reticulum (SR) and release of SR-stored Ca2+ into the cytosol. During phase 2, the inward currents mediated by LTCC (ICaL) and the sodium calcium exchanger (INCX) are electrically balanced by outward currents (IKr and IKs) mediated by two different voltage-gated delayed rectifier K+ channels. As LTCC inactivate late in the plateau phase, IKs and IKr return the MP towards the K+ equilibrium potential causing the “repolarization phase” or phase 3 of AP. Inwardly rectifying K+ channels (Kir2.1, that mediate IK1) contribute to late phase 3 repolarization and to the maintenance of the resting MP in phase 411–13.
The inward and outward conductances during the plateau phase are delicately balanced and modest changes during this phase can lead to differing AP morphologies such as a spike-and-dome shape in the ventricular and a triangular shape in the atrial CMs. Atrial AP also displays a less negative RMP, attributable to the reduced expression of Kir2.1. Ito is more prominent in human atrial than in ventricular myocytes14. Moreover, atrial cells selectively express two additional K+ currents; the ultra-rapidly activating delayed outward rectifying current (IKur) and the acetylcholine-activated K+ current (IKACh) that are absent in ventricular myocytes. IKACh is small under normal conditions but it becomes prominent during increased vagal tone15. Activation of IKur and IKACh during phase 3 explains the lower plateau potential and shorter APD of atria.
The AP morphology of pacemaker cells in the SAN and the atrioventricular node (AVN) is significantly different from that of ventricular and atrial cells. A hallmark feature of pacemaker cells is their ‘automaticity’, i.e. spontaneous beating in the absence of an external stimulus. The automaticity is driven by both voltage and Ca2+-dependent mechanisms and involves the funny current (If) carried by the hyperpolarization-activated cyclic nucleotide-gated (HCN) channels16. In pacemaker cells, the RMP at the onset of phase 4 is more depolarized (−50 to −65 mV) compared to ventricular cells. Due to this depolarized RMP, the Na+ channels are mostly inactivated. As a result, the phase 0 in pacemaker cells is mediated by the relatively slowly activating ICaL, leading to very low upstroke velocity. Phase 1 is absent in pacemaker cells due to an absence of Ito. Phase 2 repolarization again represents the balance between ICaL, IKr and IKs and the final phase 3 repolarization results from inactivation of ICaL and the recovery from inactivation of IKr. If activates at the end of phase 3 repolarization and contributes to the initiation of phase 4 diastolic depolarization17.
Excitation Contraction Coupling
The synchronous beating of the heart depends on the coupling between the electrical activity (excitation) and the mechanical pumping (contraction), hence the name “excitation-contraction (EC) coupling” (Figure 1C)18, 19. EC coupling involves finely controlled changes in the concentration of Ca2+ ions inside the myocyte, raising the cytoplasmic [Ca2+] from ~60–100 nM to 600–1000 nM and activation of the contractile apparatus20. During phase 2 of the AP, Ca2+ ions enter the cell via LTCCs and activate nearby (within 15 nm) clusters of specialized SR Ca2+ release channels (RYR2, type 2 ryanodine receptors)21. Activation of RYR2 induces massive release of Ca2+ from the SR, the principal intracellular Ca2+ storage site, a process referred to as “calcium-induced calcium release” (CICR)22. The Ca2+ released into the cytoplasm from the SR then binds to and activates cardiac troponin C (TnC), the Ca2+-sensing protein of the contractile apparatus and begins myofilament contraction. During diastole, relaxation of the myocardium occurs by the pumping of cytosolic Ca2+ back into the SR by Ca2+-ATPase type-2a (SERCA2a) or into the extracellular space by Na+/Ca2+-exchanger type-1 (NCX). The NCX is electrogenic in nature and imports three Na+ ions in exchange for a single Ca2+ ion extruded, creating a depolarizing transient inward current (INCX). The sequential rapid release of Ca2+ from the SR into the cytosol followed by rapid reuptake by SR or extrusion from the cell creates a Ca2+ transient. The amplitude of these transients largely depends upon the amount of Ca2+ release from the SR by RYR2 and determines the strength of the systolic contraction23.
Characteristics of Human iPSC-Derived Cardiomyocytes
Native human CMs are ideal to study the molecular mechanisms associated with cardiac ion channel abnormalities. However, it is not only highly invasive and cumbersome to obtain cardiac tissue from patients but also not currently possible to maintain these cells in long-term culture. Consequently, the majority of functional studies on specific mutations associated with IAS have relied on overexpressing the mutated channels in heterologous expression systems, including Xenopus oocytes, human embryonic kidney (HEK) cells, and Chinese Hamster Ovary (CHO) cells. An important shortcoming of these models is that they lack crucial constituents of cardiac ion channel macromolecular complexes that might be required to reproduce the exact molecular and electrophysiological phenotype linked with the mutation24. Transgenic mouse models have been very useful in this regard (Table 1), but substantial differences in gene expression profile and physiology between species severely limit the validity of extrapolating data from rodents to humans. For example, the heart rate in mice is about eight times faster than that of humans and cardiac repolarization in mouse CMs relies primarily on Ito, IK,slow1, IK,slow2, and ISS ion currents, whereas in humans, repolarization is mostly dependent on IKs and IKr. In addition, Ca2+ handling and myofilament proteins differ in expression and pathophysiology in mouse versus human hearts3.
Table 1.
Cardiac Ion Channels: Genetics, Properties and Role
| Channel or current |
Genes (encoding Major pore- forming α-subunit) |
Minimal β-subunit/s* |
Expression | Current properties and contribution to AP |
Pathophysiological significance |
Functional state in hiPSCs (compared to adult CMs) |
Knockout mouse phenotype |
Ref. |
|---|---|---|---|---|---|---|---|---|
| Voltage-gated Sodium Channels (INa): Characterized by a prominent transient (INaT) and a minor late component (INaL) | ||||||||
| Nav1.5 | SCN5A |
SCN1B– SCN4B |
All cardiac cell types (very low expression in nodal cells) |
Tetrodotoxin (TTX) resistant (IC50 ≥ 1 µM), inward current responsible for the rapid upstroke of the AP (Phase 0) and for proper cardiac excitability and impulse propagation |
BrS (LOF), sick sinus syndrome and LQTS3 (GOF leading to enhanced INaL) |
Slightly lower functional expression**. A lower upstroke velocity in hiPSC-CMs does not necessarily denote low INa density because at the depolarized MDP typical for hiPSC-CMs, a fraction of Na+ channels remain inactivated and thus unable to open during the upstroke |
SCN5A−/− mice are Embryonically lethal; SCN5A−/+ display conduction related abnormalities |
24, 123 |
| Nav1.8 | SCN10A | Very low expression if at all in cardiac myocytes, predominantly in intracardiac neurons |
TTX-resistant, affects cardiac conduction indirectly via neutrally mediated effect or modulation of Nav1.5 expression |
BrS | No functional evidence |
SCN10A−/− mice have shorter PR intervals |
124, 125 |
|
| Voltage-gated Calcium Channels (ICa) | ||||||||
| L-type (long-lasting type) (ICaL) Cav1.2 |
CACNA1C |
CACNB2, CACNG |
All cardiac cell types (nodal and some atrial cells also express limited amounts of Cav1.3) |
Inward current during the plateau (phase 2) of the AP, involved in EC coupling, conduction and automaticity |
LQTS8 (Timothy syndrome, GOF) and BrS (LOF) |
Robust expression of ICaL with similar characteristics |
CACNA1C−/− leads to embryonically lethality. Heart- specific graded reduction in gene expression leads to hypertrophy while severe reduction causes early adulthood lethality |
123, 126– 128 |
| T-type (transient-type) (ICaT) Cav3.1 |
CACNA1G | Principally in pacemaker, atrial, and Purkinje cells |
Activation at more Hyperpolarized potentials than LTCC and highly sensitive to Ni. Together with ICaL, plays a role in conduction and diastolic pacemaker current |
ICaT expression has recently been reported in SAN-like pacemaker cells derived from hPSCs |
31 | |||
|
Potassium Channels: Mediate AP repolarization. The human genome includes ~80 genes that encode for K+ channel subunits, but only a subset of these genes are highly expressed in the heart. Each K+ channel subunit is composed of either two (e.g. Kir channels) or six (e.g. voltage-gated K+ (Kv) channels) transmembrane (TM) segments | ||||||||
| Transient Outward K+ Channels (Ito) | ||||||||
|
Ito,f (fast) Kv4.2, Kv4.3 |
KCND2, KCND3 |
KCNE2–3, KCNIP1–2, DPP6 |
Greater Ito,f density is present in the atria and Purkinje fibers than in the ventricular myocytes |
Displays faster gating properties resulting in brief K+ efflux and a short-lasting repolarization (the phase 1 notch in the AP) |
BrS (GOF), reduced current densities have been found in failing heart and chronic AFib |
Slightly lower functional expression**. Slow recovery of Ito from inactivation and different subunit stoichiometry (i.e. relatively high Kv1.4 and low KCNIP2 mRNA expression) in hiPSC- CMs might lead to smaller contribution to AP. At depolarized MDP of hiPSC-CMs, a large proportion of the channels are unavailable |
In Kv4.2 −/− mouse, Ito,s is upregulated and compensates for the loss |
123, 129– 132 |
|
Ito,s (Slow) Kv1.4 |
KCNA4 | KCNB1–4 | Ventricular myocytes |
Recovers slowly from inactivation in comparison to Ito,f. Contributes to early repolarization during phase 1 |
||||
|
Delayed Outward Rectifier K+
Channels: So named because of delayed onset in their activation, which starts during the plateau phase of AP. These are voltage-gated K+ (Kv) channels. They play a prominent role in phase 3 repolarization. LOF or GOF mutations in these channels disrupt normal cardiac repolarization and result in various cardiac rhythm disorders, including congenital LQTS, SQTS, and familial AFib (Figures 1B and4). | ||||||||
|
IKur (ultra-rapid) Kv1.5 |
KCNA5 |
KCNAB1–2 (affect membrane trafficking and properties of activation and inactivation) |
Atria-specific | Activates rapidly upon Membrane depolarization and undergoes very slow inactivation. Contributes to both early and late repolarization of the AP |
Both LOF and GOF mutations in KCNA5 have been linked to familial AFib (likely explained by different underlying mechanisms of arrhythmia generation; i.e., AP prolongation leading to EADs vs AP shortening leading to reentry) |
Lower proportion of atrial-like cells in hiPSC- CM cultures make an exact comparison difficult. RA-guided differentiation results in increased atrial-like phenotype and higher expression of Kv1.5 mRNA and IKur density in hPSC-atrial CMs |
Precise role of Kv1.5 loss in transgenic mouse models is uncertain due to the compensatory mechanisms and effects of the inserted transgene |
30, 133– 135 |
|
IKr (rapid) Kv11.1 |
KCNH2 (hERG1) |
All cardiac cell types |
IKr is characterized by fast inactivation, fast recovery and slow deactivation. It plays a major role in phase 3 repolarization |
LQTS2 (LOF) and acquired LQTS due to high susceptibility of hERG to drug block, SQTS1 (GOF) |
Comparable current densities as that of adult CMs |
KCNH2−/− result in embryonic lethality while KCNH2 −/+ result in mild APD prolongation |
59, 86, 123, 136 |
|
|
IKs (slow) Kv7.1 |
KCNQ1 |
KCNE1 (imparts unique slow activation- deactivation kinetics and physiological voltage range of activation to Kv7.1) |
All cardiac cell types |
Activates slowly during the depolarized plateau (phase 2 of the AP) |
LQTS1 (LOF), SQTS2 (GOF) |
Comparable current densities as that of adult CMs |
KCNQ1−/− loss does not lead to a robust cardiac phenotype in mouse |
58, 137 |
| Inward Rectifier K+ Channel (IK1) | ||||||||
| Kir2.1 (Kir2.2/2.3) |
KCNJ2 (KCNJ12/ KCNJ4) |
Kir2.1 is the Predominant isoform. Expression is higher in the ventricles than in the atria. Virtually absent in nodal tissue |
Plays a major role in setting the RMP and terminal repolarization. Protects the ventricular cell from abnormal pacemaker activity |
LQTS7 (Andersen-Tawil syndrome, LOF), SQTS and familial AFib (GOF) |
IK1 density is considerably smaller in hiPSC-CMs, which (in combination with presence of If) contributes to the spontaneous activity and relatively depolarized RMP |
Kir2.1−/− leads to perinatal lethality due to cleft palate. Kir2.1−/− myocytes showed broader APD and frequent spontaneous APs |
32, 123, 138 |
|
| Acetylcholine-activated K+ Channel (IKACh) | ||||||||
| Kir3.1 (GIRK1) and Kir3.4 (GIRK4) |
KCNJ3 and KCNJ5 |
GIRK1 Requires GIRK4 for Membrane targeting |
Abundantly expressed in atria, SAN and AVN |
Mediates Parasympathetic regulation of the heart rate. Activation of IKACh hyperpolarizes the MP and shortens the APD |
Has been associated with AFib. Studies focusing on IKACh in hiPSC-CMs may be useful in modeling atrial arrhythmias |
Atrial-like CMs were found to have substantially higher IKACh (activated by carbachol) density that also hyperpolarized RMP compared to ventricular- like CMs |
KCNJ5−/− results in diminished vagal control of the heart rate |
133, 139, 140 |
| Hyperpolarization-activated, intracellular Cyclic Nucleotide-gated (HCN) Pacemaker Channel (If) | ||||||||
| Pacemaker channels |
HCN1, HCN4 |
HCN4 is the most abundant isoform in the nodal tissue. HCN2 has been shown to express in ventricles at very low levels |
A non-selective inward current that is directly regulated by cAMP levels (affected by sympathetic stimulation). Contributes to diastolic depolarization in pacemaker cells (phase 4) |
SA node dysfunction such as sick sinus syndrome |
hiPSC-CMs express substantial If contributing to their spontaneous activity. In this regard, they resemble immature neonatal myocytes |
Constitutive HCN4−/− results in Embryonic lethality while inducible cardiac- specific KO has bradycardia. Constitutive HCN2−/− results in sinus dysrhythmia |
17, 32, 123 |
|
AFib, atrial fibrillation; AP, action potential; APD, action potential duration; AVN, atrio-ventricular node; EAD, early afterdepolarization ;EC, electro-mechanical coupling; GOF, gain-of-function; hPSC, human pluripotent stem cells; KO, knockout; LOF, loss-of-function; LQTS, long QT syndrome; MDP, maximum diastolic potential; MP, membrane potential; Ni, Nickel; RA, retinoic acid; RMP, resting membrane potential; SAN, sino-atrial node; SQTS, short QT syndrome.
An array of accessory subunits is required in most cases to reconstitute a native current phenotype in a heterologous expression system. Many of these accessory subunits have not been completely characterized, have regional or species variability10 and in some cases led to conflicting results depending on the expression system used. It is not possible to provide an exhaustive list of all the accessory subunits for each channel here; only the important ones are listed here.
An exact comparison of current density (e.g. for INa and Ito) is difficult due to different experimental conditions (e.g. temperature, different ionic concentrations and holding potential), regional variability in expression in an intact heart (e.g. epi- vs endocardium) and heterogeneous population of atrial-, ventricular- and nodal-like hiPSC-CMs in cultures.
The recent discovery of somatic cell reprogramming to generate iPSCs4, 5 has created great excitement because of the ability to produce for the first time, patient- and disease-specific human iPSC-derived cardiomyocytes (hiPSC-CMs). To generate hiPSCs, somatic cells can be obtained from patient’s hair, blood, skin, fat, urine, or oral mucosa24, 25. These cells are then reprogrammed to a pluripotent state by introducing pluripotency-associated genes. The resulting iPSCs are then differentiated into CMs by using a number of strategies (for review, see [26,27]). After approximately 8–12 days, clusters of beating cells typically appear which upon further maturation in culture can be enzymatically dissociated into single CMs for molecular and functional analysis.
The patch clamp technique is the gold standard to measure various AP parameters from hiPSC-CMs that correspond to specific ionic current properties. These parameters include RMP or maximal diastolic potential (MDP), cycle length (CL), AP amplitude (APA), maximum upstroke velocity (dv/dtmax), and APD at various percentage levels of repolarization (i.e., APD20, APD50, and APD90). For example, MDP/RMP reflects activity of steady state K+ currents, primarily IK1. The upstroke velocity reflects the magnitude of INa, while the AP durations (APDs) denote different phases of repolarization. APD20 is importantly regulated by ICaL and Ito1, while APD90 is most dependent on IKs and IKr25.
The common hiPSC differentiation protocols predominantly generate ventricular-like hiPSC-CMs28, 29, although protocols aimed at specifically generating atrial-like30 and nodal-like31 hiPSC-CMs have also been recently described. The distinction between different hiPSC-CMs is often made based on the AP morphology. For example, ventricular-like cells are characterized by a more negative MDP, a rapid AP upstroke, and a long plateau phase (see Figure 1B). The absence of a prominent plateau phase is defined as a characteristic of atrial-like cells, resulting in shorter APD compared to ventricular-like APs. Nodal-like APs display more depolarized MDP, a slower AP upstroke, and a prominent phase 4 depolarization24. Recordings of subtype specific currents may also be used to distinguish atrial and nodal CMs within the hiPSC-CM population. For example, IKur and IKACh are specific to atrial cells30, while If and T-type Ca2+ current (ICaT) are specific for nodal cells31 (Table 1).
In general, hiPSC-CMs are electrophysiologically immature compared to isolated adult ventricular CMs (AVCMs) as reflected by the lower upstroke velocity (secondary to less negative MDP), and less prominent notch (phase 1)24, 32. One of the most remarkable differences between hiPSC-CMs and native adult human primary CMs relates to the fact that hiPSC-CMs (including ventricular-like and atrial-like cells) display innate automaticity resulting in spontaneous beating, like in fetal human CMs24. This is due to substantially reduced density of IK1 and expression of pacemaker current (If) in hiPSC-CMs that is virtually absent in AVCMs. Recent studies have aimed to overcome these limitations by artificially enhancing the IK1 density through either viral over-expression of Kir2.133, 34, or by in-silico injection of IK1 with kinetics of Kir2.135, 36. Over-expression of Kir2.1 in human embryonic stem cell-derived cardiomyocytes (hESC-CMs) abolished cell automaticity resulting in similar AP characteristics to those of adult CMs but the Ca2+ handling properties remained immature33. However, in a more recent study, enhanced Kir2.1 expression in hiPSC-CMs improved both AP and Ca2+ handling properties34. Similarly, enhanced IK1 by in-silico injection in hiPSC-CMs, induced a more physiological and stable MDP, a ventricular-like AP morphology, and increased AP upstroke velocity comparable to that from freshly isolated human myocytes35, 36. Additional approaches including three-dimensional (3D) tissue engineering, mechanical loading, modulation of substrate stiffness, phasic electrical stimulation, hormonal treatment (e.g., thyroid hormone), and long-term culture have been utilized to enhance the maturity of hiPSC-CMs37–44.
Mechanisms of Arrhythmia
Although most hearts beat with remarkable fidelity and resilience, under certain circumstances the rhythm of the heart can fail. Accurate identification of specific mechanisms underlying arrhythmia may at times be challenging for the clinician and may require invasive EP study. Differentiating and understanding these complex mechanisms is critical to the development of appropriate diagnosis and treatment strategy. The fundamental mechanisms of arrhythmia can be classified into two major categories: 1) enhanced or abnormal impulse generation (focal activity) and 2) improper impulse conduction (reentry). Focal activity includes automaticity and triggered activity13, 45.
Automaticity refers to the ability of CMs to initiate spontaneous AP. Normally, the SAN has the highest intrinsic rate and sets the heart rate. Abnormal patterns of automaticity of the SAN can result in either sinus tachycardia or bradycardia. If the SAN is unable to generate impulses or if these impulses fail to propagate, latent pacemakers in other regions of the heart can take over initiating excitation. Atrial and ventricular myocytes, which in the normal heart typically do not exhibit spontaneous activity, may exhibit automaticity properties (e.g. under β-adrenergic stimulation or reduced extracellular K+). The spontaneity in SAN arises as a result of net inward current during the phase 4 of AP causing diastolic depolarization46 (Figure 2A).
Figure 2. Key Electrophysiological Mechanisms of Cardiac Arrhythmia.

Ectopic impulse generation by (A) Enhanced automaticity. (B) APD prolongation (in blue) leading to the development of Phase 2 and Phase 3 EADs (red). (C) Development of delayed after depolarizations (DADs) that occur due to Ca2+ overload. (D) Reentry requires a vulnerable substrate, which can be caused by APD shortening or dispersion of refractoriness. Schematic shows a depolarizing wavefront around an anatomical obstacle. Under normal conditions, the depolarizing waves around the obstacle cancel each other out. Certain conditions e.g. ischemia might generate areas of unidirectional block, or sufficiently slow conduction to enable recovery of excitability in time for re-excitation by the depolarizing wavefront.
Triggered activity arises from premature activation of cardiac tissues by two types of afterdepolarizations47. Early afterdepolarizations or EADs occur during phase 2 or 3 of AP repolarization, delayed afterdepolarizations or DADs occur after repolarization is complete. When the amplitude of an afterdepolarization is large enough, a spontaneous AP can be triggered48. EADs are often but not always associated with prolonged APD which occur when the depolarizing currents (e.g., INaL, ICaL or INCX) predominate the repolarizing currents (e.g., IKr, IKs and IK1). Two mechanisms have been proposed for EADs. First, the prolonged repolarization of the APD allows recovery and reactivation of ICaL49 resulting in enhanced inward ICaL that further depolarizes the membrane. This sets up a positive feedback loop, triggering a premature AP13. Secondly, at MPs negative to the threshold of ICaL activation (but before full repolarization), spontaneous Ca2+ release from the SR can activate INCX resulting in membrane depolarization13 (Figure 2B). DADs arise as a result of abnormal spontaneous Ca2+ release from the SR into the cytoplasm. The resulting diastolic increase in [Ca2+]i induces a transient increase in inward INCX, generating DADs46 (Figure 2C). When the depolarization induced by the DADs is sufficiently large, INa can be activated resulting in triggered activity that also underlies arrhythmogenicity observed in Catecholaminergic Polymorphic Ventricular Tachycardia (CPVT)13.
Under normal conditions, the myocardium has a unidirectional propagation of depolarization wavefront that cancel each other out when the two meet from the opposite sides. Abnormal AP propagation can occur when a depolarization wavefront encounters a region that lags in recovery (i.e. longer effective refractory period (ERP)) from a previous depolarization, and thus the wavefront is blocked unidirectionally. However, the same region might be outside its ERP when the wave reaches to the other end of this region (e.g. via propagating around the periphery of the region). This will lead to retrograde excitation of this region and the wave front loops back and due to slow conduction might re-excite the adjacent regions that have recovered excitability. This circulating wave results in a circuitous and repetitive mode of depolarization, termed re-entry50 (Figure 2D).
The pathogenesis of arrhythmias in the whole heart is a complex phenomenon51, 52 involving different layers of the myocardium, 3-D cellular coupling and different regions with unique structural and electrical features that can remodel with time. Although abnormal electrical behavior can be modeled in single cells, some events (e.g., re-entry) can be demonstrated only in multicellular preparations such as monolayers of hiPSC-CMs53, 54. Caution must be exercised in extrapolating the findings from cellular studies to the whole heart.
Platforms for Functional Analysis of Human iPSC-CMs
iPSC technology has great potential to advance precision medicine. However, to be able to better utilize this human platform, appropriate methods for functional analysis of patient-specific iPSC-CMs are essential for understanding the pathogenic mechanisms of cardiac disorders as well as performing preclinical safety studies for novel drug candidates. A concise overview of the commonly used methodology for iPSC-CM functional analysis is described below.
Patch Clamp
High fidelity intracellular electrical recordings from CMs provide the most detailed and precise evaluation of the electrophysiological properties of CMs. Sharp electrodes penetrating the cell membrane can accurately record MP and thus characterize APs in detail. The patch clamp technique55, provides the opportunity to measure both membrane potential (or AP) and underlying ionic currents. The technique involves gently pressing a fine-tip glass pipette, containing a solution with an ionic composition similar to the cytosol, against the cell membrane using a precision micromanipulator under guidance from an inverted microscope, followed by application of a mild suction to obtain a high resistance seal between the glass pipette and the cell membrane. Once a giga-ohm resistance seal is formed, the small patch of membrane plugging the pipette tip can be ruptured to provide electrical access to the inside of the cell, a recording configuration known as the whole cell patch clamp technique (Figure 3A) that allows the measurement of APs in current-clamp mode or membrane currents in voltage-clamp mode56.
Figure 3. Platforms for Functional Analysis of Human iPSC-CMs.

Graphical illustration summarizing different methods currently used for hiPSC-CM functionality analyses. (A) Patch Clamp (B) MEA (C) Fluorescence Imaging (D) Impedance. Representative MEA64, 121 and Fluorescence Imaging70, 122 traces reprinted with permission of the publisher.
Many seminal studies of channelopathies have employed the patch clamp method for electrophysiological analysis of hiPSC-CMs57–60. The biggest disadvantage of the patch clamp method remains its low throughput. The recordings are laborious, technically challenging and requires skilled personnel. Moreover, the invasive and terminal nature of the technique allows recordings to be performed only for a short period of time from single cells. However, it provides very high time (<1ms) and voltage- or current-amplitude resolution (unmatched by any other technique) while allowing control of the intracellular and extracellular ion composition7, 55. Further, using specific blockers and recording protocols that can control the MP, activity of an individual ion channel (or whole cell current) can be separated from other ion currents with high fidelity.
Multielectrode Array (MEA)
MEA is a non-invasive and non-terminal method that allows long-term measurements of extracellular field potential signals from clusters and monolayers of hiPSC-CMs61. CMs are plated on the wells of the MEA plate containing electrodes on the bottom, and field potential is measured. The field potential duration (FPD) correlates roughly to the QT interval in the in vivo EKG and the APD in vitro. The field potential signal is composed of an initial rapid spike which corresponds to the AP upstroke, a slow wave/plateau phase, followed by a repolarizing wave that corresponds to repolarization56, 62, 63 (Figure 3B). Over the past few years MEAs have been increasingly used for predicting cardiotoxicity (manifested as prolonged FPDs and development of triggered activity) of hiPSC-CMs and can be scaled up for moderate throughput. The MEA platform is widely accepted and a relatively easy to understand assay64–66. MEA allows generation of detailed activation maps and conduction velocity measurements; however, the conduction velocity in hiPSC-CMs monolayers is relatively slow (10–20 cm/s compared to 60 cm/s in adult human left ventricle) likely due to the immaturity of hiPSC-CMs67. The disadvantages of the MEA method are its low resolution and inability to analyze ionic currents.
Fluorescence Imaging
Fluorescence microscopy methods that utilize Ca2+- or voltage-sensitive dyes provide a non-invasive method for measuring intracellular ion fluctuations and voltage changes. However, such indicator dyes can bind to intracellular molecules likely interfering with the normal functioning of the cell and most Ca2+ indicator dyes, including Fura-2 and Fluo-4, are cytotoxic. Despite novel developments in software tools used for data analysis, Ca2+ imaging remains a rather slow method, both for performing the assay itself and for data analysis68. Voltage-sensitive dyes, such as di-4-ANEPPS, respond to changes in MP by changing their fluorescence emission. They are commonly used for studying whole hearts but have their limitations in single cell and monolayer imaging due to significant cytotoxicity69. To overcome these challenges, genetically encoded fluorescent indicators (GECI for intracellular calcium ions and GEVI for voltage) were recently introduced to improve the efficiency of cellular phenotyping in hiPSC-CMs. These genetically encoded indicators consist of a sensing element (e.g. CaM for Ca2+), often fused to an autofluorescent protein (like circularly permuted enhanced GFP; cpEGFP) that alters its fluorescent intensity based on conformational changes in the sensing element70 (Figure 3C).
In a recent study, Shinnawi et al.70 demonstrated successful expression of genetically encoded voltage (ArcLight) and calcium (GCaMP5G) fluorescent indicators in hiPSC-CMs derived from patients with different IAS70. The use of these reporters in comparison to traditional voltage- and Ca2+-sensitive dyes offers several advantages including significant photostability, superior signal-to-noise ratio and minimal cytotoxicity. Most fluorescent dyes (except Fura-2, which is ratiometric) and all genetic indicators allow monitoring of only relative changes in membrane voltage and intracellular Ca2+ levels and do not provide absolute values70.
Impedance
Functional assays based on cellular impedance (an indirect measure of CM contractility) offer a non-invasive, label free and high throughput analysis method71. Cells are seeded onto a multi-well cell culture plate that contains gold film electrodes embedded onto the bottom of the wells. The mechanical displacement of the cells during CM contraction is measured as variations in impedance (equivalent to resistance in a DC circuit) that directly correlates with the beating frequency (Figure 3D). These measurements utilize weak alternating current (AC) between the electrodes with tissue culture medium as the electrolyte. The electronic hardware monitors the voltage across the electrodes, and the impedance is calculated using the AC version of Ohm’s law where impedance (Z) rather than resistance (R) is calculated as Z = V/I. Monitoring of the impedance signal does not alter cellular physiology either in an excitatory, suppressive, or cytotoxic fashion71. Clearly, this method has great potential in ascertaining the beat rate/arrhythmogenicity of hiPSC-CMs, however caution should be taken to avoid over-interpreting the data. It is important to realize here that the changes in the beating pattern of the cells are recognized only due to a measurement of a minute change in resistance and not due to their direct electrical activity72. Impedance measurements have been validated for mouse ESCs73, hiPSC-CMs73, rat neonatal primary CMs74, 75 and further cell lines, e.g. cardiac muscle cells (HL-1) and 3D cell clusters (hESC-CM™)76. The recently introduced Cardio ECR platform (ACEA Biosciences) combines MEA with contractility measurements for a simultaneous and more comprehensive analysis of the excitation-contraction coupling77.
Inherited Arrhythmia Syndromes (IAS) and hiPSC Modeling
Molecular and biophysical studies over the past two decades have linked genetic mutations in ion channels or ion channel regulatory proteins (genotype), to IAS (phenotype). These disorders include LQTS, SQTS, Brugada syndrome, (BrS), CPVT, and familial atrial fibrillation (AFib)46. Table 2 summarizes the genotype-phenotype correlation of various channelopathies. As illustrated in Figure 4, mutations in different ion channels can lead to a very similar pathological phenotype and different mutations in the same ion channel gene can cause different channelopathies. For example, mutations in genes encoding K+, Na+, and Ca2+ channels can all lead to AP prolongation and manifest as LQTS, while different mutations in K+ can cause both LQTS and SQTS.
Table 2.
hiPSC-CM Models of Inherited Arrhythmia Syndromes
| Syndro me |
Causal gene | Effect on ion channel |
Experimental approach |
Cellular phenotype | Ref |
|---|---|---|---|---|---|
| LQTS1 |
KCNQ1 (R190Q) |
Trafficking defect, altered channel activation and deactivation properties |
Patch clamp | Reduced IKs, APD prolongation, increased susceptibility to catecholamine-induced tachyarrhythmia, attenuation of this phenotype with beta blockade |
58 |
|
KCNQ1 exon7 deletion |
Possible Haploinsufficiency and trafficking defect of KCNQ1 |
Patch clamp | Reduced IKs, APD prolongation, reduced wild type KCNQ1 mRNA and protein, small molecule ML277 partially restored APD and reversed the decreased IKs. |
141 |
|
|
KCNQ1 (R594Q R190Q) |
Trafficking defect | Patch clamp, MEA, |
Prolonged APD, reduced IKs activation that was reversed by hERG allosteric modulator LUF7346 |
142 | |
| LQTS2 |
KCNH2 (A614V) |
Trafficking defect | Patch clamp, MEA |
Reduced IKr, APD prolongation, induction of EADs and triggered activity, potential improvement with pinacidil |
86 |
|
KCNH2 (G1681A) |
Trafficking defect | Patch clamp | APD prolongation and EADs | 57 | |
|
KCNH2 (R176W) |
Trafficking defect | Patch clamp, MEA |
Reduced IKr, APD prolongation, mild incidence of EADs |
59 | |
|
KCNH2 (N006I) |
Trafficking defect | Patch clamp, MEA |
Prolonged APD, reduced IKr activation that was reversed by hERG allosteric modulator LUF7346 |
142 | |
| LQTS3 |
SCN5A (V1763M) |
Gating defect- inactivation of sodium channel |
Patch-clamp | Enhanced INaL, APD prolongation, phenotype reversed by Nav1.5 blockade by mexiletine |
91 |
|
SCN5A (F1473C) |
Gating defect- deficiency in sodium channel inactivation |
Patch clamp | Delayed repolarization, arrhythmia, prolonged QT interval with increase in pacing improving the phenotype |
92 | |
|
SCN5A (V240M R535Q) |
Gating defect- deficiency in sodium channel inactivation |
Patch clamp | Insignificant increase in APD, delayed time to peak INa inactivation |
90 | |
|
SCN5A (R1644H) |
Gating defect- deficiency in sodium channel inactivation |
Patch clamp, MEA |
Prolonged APD, high EADs, and accelerated recovery from inactivation of Na+ currents. Rescue of abnormal phenotypeby mexiletine and ranolazine |
143 | |
| LQTS7 |
KCNJ2 (R218W), (R67W), (R218Q) |
Trafficking defect | MEA, calcium imaging |
Strong arrhythmic events, higher incidence of irregular Ca2+ release. Flecainide, but not pilsicainide, suppressed irregular Ca2+ release and arrhythmic events |
97 |
| LQTS8 |
CACNA1C (G1216A) |
Gating defect- loss of voltage- dependent channel inactivation |
Patch clamp, calcium imaging |
APD prolongation and DADs, abnormal calcium handling, irregular and slow contraction. Roscovitine rescued abnormal cellular phenotype |
60 |
| LQTS14 |
CALM1 (F142L) |
Gating defect- impaired Ca2+- dependent inactivation |
Patch clamp, MEA, calcium imaging |
Prolonged APD, defective ICaL inactivation, altered rate-dependency and response to isoproterenol. Repolarization abnormalities reversed by verapamil |
144 |
| LQTS15 | CALM2 (N98S) | Gating defect- suppression of L- type Ca2+ channel (LTCC) inactivation |
Patch clamp | Lower beating rate, prolonged APD, and impaired ICaL inactivation, correction of the mutant allele rescued abnormal phenotype |
145 |
|
CALM2 (D130G) |
Gating defect- disruption of Ca2+/CaM- Dependent inactivation of LTCC |
Patch clamp, Fluorescence imaging |
Prolonged APD, disrupted Ca2+ cycling properties, and diminished Ca2+/CaM- dependent inactivation of ICaL. Suppressing the mutant gene rescued abnormal phenotype |
146 | |
| BrS |
SCN5A (R620H), (R811H |
Trafficking defect | Patch clamp, calcium imaging |
Reduced INa and maximal upstroke velocity of AP, abnormal Ca2+ transients, and variation in beating interval |
81 |
|
PKP2 (c.2484C>T) |
Trafficking defect | Patch clamp, MEA, calcium imaging |
Reduced INa, deficit restored by transfection of WT gene |
147 | |
| CPVT1 |
RYR2 (M4109R) |
Patch clamp, calcium imaging, MEA |
DADs, isoproterenol enhanced DADs and developed triggered activity. Flecainide and thapsigargin eliminated DADs. Ca2+ transient irregularities that worsened with adrenergic stimulation and Ca2+ overload and improved with β-blockers |
106 | |
| RYR2 (F2483I) | Patch clamp, calcium imaging |
Longer duration of Ca2+ release from SR long after repolarization, arrhythmias and DADs with adrenergic stimulation |
107 | ||
| RYR2 (S406L) | Patch clamp, Calcium imaging |
Elevated diastolic Ca2+ concentrations, reduced SR Ca2+ content, and arrhythmias. Dantrolene ameliorated arrhythmia and restored normal Ca2+ |
108 | ||
| RYR2 (P2328S) | Patch clamp, Calcium imaging |
Increased non-alternating variability of Ca2+ transients in response to isoproterenol and β-agonists decreased AP upslope velocity |
108 | ||
| RYR2 (R420Q) | Calcium imaging |
Less developed ultrastructure, isoproterenol either ineffective, caused arrhythmias, or markedly increased diastolic Ca2 |
148 | ||
| RYR2 (L3741P) | Calcium imaging, MEA |
Altered intracellular Ca2+ homeostasis, β- adrenergic stimulation potentiated spontaneous Ca2+ waves and prolonged Ca2+ sparks. Flecainide ameliorated disease phenotype |
149 | ||
| RYR2 (I4587V) | Patch clamp, Calcium imaging |
Increased diastolic Ca2+ waves and DADs with pacing, while S107 suppressed the DADs |
150 | ||
| CPVT2 |
CASQ2 (D307H) |
Patch clamp, Calcium imaging |
β-adrenergic agonist caused DADs, oscillatory arrhythmic pre-potentials, and diastolic [Ca2+]i rise |
108, 148, 151 |
|
|
CASQ2 (D307H) |
Patch clamp, Calcium imaging |
Ca2+-transient irregularities, EADs, and reduced threshold for store overload- induced Ca2+-release, β-blockers prevented arrhythmia |
152 |
APD, action potential duration; EAD, early after depolarization; DAD, delayed after depolarization; CALM1, Calmodulin 1; CALM2, Calmodulin 2; PKP2, Plakophilin-2.
Figure 4.

Diagram showing overlap between the genes associated with Brugada Syndrome (BrS), Short QT Syndrome (SQTS), Long QT syndrome (LQTS), Catecholaminergic Polymorphic Ventricular Tachycardia (CPVT) and Atrial Fibrillation (AFib).
Brugada syndrome
BrS is an inherited disorder characterized by ventricular arrhythmias and SCD occurring in otherwise healthy individuals at a relatively young age (< ~40 years), typically during conditions of high vagal tone (i.e., during sleep). On EKG analysis, a typical pattern is observed comprising ST-segment elevation in the right-precordial leads V1–V378, 79. In high-risk patients, with (recurrent) ventricular tachyarrhythmias, implantable cardioverter defibrillator (ICD) is considered, as pharmacological options are limited.
While several genetic mutations have been associated with BrS, all lead to a general imbalance of currents during the early phases of the AP tipping in favor of repolarization. The most common and best studied of these is the LOF mutation of the SCN5A gene (encoding Nav1.5)50 leading to reduced Na+ channel availability, either through decreased surface channel expression or through altered channel gating properties78, 79. Although much of our current understanding regarding the molecular mechanisms of BrS was gleamed from heterologous expression studies, very recently, patient-specific iPSC-CMs harboring LOF genetic mutations in SCN5A were shown to recapitulate the pro-arrhythmic phenotype associated with either BrS80, 81 or overlapped LQT3/BrS82.
Long QT syndrome
LQTS is a potentially lethal cardiac disorder, characterized by QT interval prolongation and increased risk of ventricular arrhythmias, particularly torsade de pointes (TdP) that can lead to SCD. Increased arrhythmic risk associated with a prolonged QT interval can be congenital or acquired. Congenital LQTS is associated with mutations in ion channels and/or accessory proteins, whereas acquired QT prolongation is most commonly associated with block of the hERG K+ channel by some antiarrhythmic agents or as an unwanted side effect of many other medications. Additionally it can be associated with electrolyte imbalance (hypokalemia, hypocalcaemia, and hypomagnesaemia). The mutations associated with LQTS result in a favoring of depolarizing currents over repolarizing ones during phase 2 and 3 of the AP50. At the single cell level, LQTS results from prolongation of the APD that can initiate EADs and TdP arrhythmia that sometimes degenerates into potentially lethal ventricular fibrillation.
LQTS1
LQTS1 is the most common type of LQT disorders that accounts for ~40% of total cases, often triggered by adrenergic drives, e.g., emotional stress, physical exertion, diving and swimming, etc. LQTS1 results from LOF mutations in the KCNQ1 gene that encodes the α-subunit of the channel conducting IKs. LQTS1 patients respond well to β-blockers while some are less responsive or even resistant to this medication83.
Moretti et al58 were the first to model LQTS1 using hiPSC-CMs derived from a patient carrying the R190Q KCNQ1 mutation. This missense mutation caused a trafficking defect that reduced KCNQ1 protein surface expression, leading to a 70% reduction in IKs. Consequently, the diseased cells recapitulated the hallmark LQTS phenotype characterized as APD prolongation and an increase in arrhythmia with β‑adrenergic agonists. Propranolol pre-treatment reduced isoproterenol-induced arrhythmias in spontaneously beating LQTS1 hiPSC-CMs58. In a later study, Ma et al84 derived iPSC-CMs from a LQTS1 patient harboring a different KCNQ1 mutation (exon 7 del) and demonstrated that patient-specific iPSC-CMs faithfully recapitulated the clinical phenotype, and responded to a small molecule ML277, an investigational agent under development to treat arrhythmia. Egashira et al85 generated iPSC-CMs from a sporadic LQTS patient with 1893delC mutation in the KCNQ1 gene with clinical suspicion of LQTS1. MEA recordings revealed markedly prolonged FPD, a characteristic of LQTS phenotype. Further analysis identified a trafficking defect in KCNQ1 as the cause of the patient’s clinical phenotype.
Taken together, these findings clearly demonstrate the potential of patient-specific hiPSC-CMs as robust models for IAS despite their relatively immature electrophysiological profile.
LQTS2
LQTS2 accounts for 30% of all LQTS cases and is caused by LOF mutations in the KCNH2 (hERG1) gene that encodes the α-subunit of the channels that conduct IKr. An auditory stimulus is often a trigger for arrhythmic events in LQTS2 patients. The first reports of iPSC-based models of LQTS2 were reported by Itzhaki et al86 and Matsa et al87. It was demonstrated86 that hiPSC-CMs derived from a patient with missense mutation A614V in KCNH2 exhibited significant prolongation of the AP, smaller IKr, EADs, and triggered arrhythmias. Importantly, the authors evaluated the potency of several pharmacological agents that worsened or mitigated the cellular LQTS2 phenotype. In a later study87, hiPSC-CMs were derived from a patient with the A561T mutation in KCNH2. The authors87 used allele-specific RNA interference (RNAi) to selectively degrade mutant mRNA while leaving wild-type mRNA intact. This approach normalized the APD, increased IKr, and decreased the frequency of EADs. In a recent study, Mehta et al.88 provide solid evidence in favor of hiPSCs having promising drug repurposing applications with regard to the development of precision medicine treatment of LQTS. The authors demonstrated that Lumacaftor, a drug already in clinical use for cystic fibrosis due to its ability to foster trafficking of mutant CFTR channels to the cell surface, can also rescue the pathological phenotype of class 2 (trafficking deficient) LQTS2 hiPSC-CMs derived from patients not protected by β-blockers.
LQTS3
LQTS3 is caused by GOF mutations in the SCN5A (Nav1.5) gene that accounts for ~10% of all LQTS cases. Most of these mutations result in enhanced late Na+ current (INaL) during the plateau and repolarization phase of the AP due to defective inactivation of the Nav1.5 channel. The net increase in the depolarizing current ultimately causes APD prolongation and proarrhythmia. In contrast to the triggers of LQTS1 and LQTS2, which are often increased heart rate and stressful auditory stimuli, respectively, LQTS3 patients exhibit QT prolongation at lower heart rates and, consequently, have an increased risk for cardiac events during rest or sleep89. LQTS3 models of patient-specific hiPSC-CMs have been shown to recapitulate key electrophysiological disease features such as increased INaL and prolonged APD at the single-cell level64, 90. Ma et al91 modeled LQTS3 in hiPSC-CMs derived from a patient with the V1763M mutation and demonstrated enhanced INaL and prolonged APD. Interestingly, mexiletine, a Na+ channel blocker, preferentially inhibited INaL and rescued the abnormal electrophysiology in diseased cells.
In an elegant study, Terrenoire et al92 investigated the underlying cause of pathogenicity in a patient with variants in both SCN5A (F1473C) and KCNH2 (K897T). The authors demonstrated by patch clamp recordings that the observed clinical phenotype of severe QT prolongation and arrhythmias was primarily due to late Na+ channel defect and not influenced by the KCNH2 polymorphism. Interestingly, specific to the patient’s hiPSC-CMs, INaL displayed very pronounced rate-dependence such that enhancing the pacing rate markedly reduced INaL and increased its inhibition by mexiletine. This type of analysis clearly demonstrates the promise of patient-specific hiPSC-CM platform in improving management of IAS and for developing patient-specific clinical regimens.
LQTS7
LQTS7, also known as Andersen-Tawil syndrome (ATS), is a rare autosomal dominant genetic disorder typified by mild QT interval prolongation and appearance of significant U waves, frequent ventricular ectopy, bidirectional ventricular tachycardia (VT), and more rarely syncope, recurrent polymorphic VT, and cardiac arrest. Exercise is an important trigger of ventricular tachyarrhythmia and syncope in some patients with ATS, therefore patients are commonly treated with β-blockers. ATS type-1 (ATS1) has been linked to mutations in KCNJ293 and accounts for about 70% of all ATS cases. The genetic cause of the remaining 30% of cases (ATS2) remains unknown94. KCNJ2 encodes for the Kir2.1 potassium channel that contributes to IK1. Most KCNJ2 mutations in ATS1 cause LOF and dominant-negative suppression of Kir2.1 channel function, leading to reduced IK195, 96. Recently Kuroda et al97, modeled ATS using the hiPSC platform. A significant challenge in modeling LQTS7 is that since IK1 density is already compromised in hiPSC-CMs, it is difficult to obtain a clear difference between healthy control and diseased hiPSC-CMs using voltage clamp techniques. But interestingly, in this study, the authors employed MEA and Ca2+ imaging platforms to uncover the pathogenesis of ATS in patient-derived hiPSC-CMs. hiPSC-CMs from three ATS patients carrying either one of the KCNJ2 mutations (R218W, R218Q or R67W) clearly exhibited arrhythmic events and abnormal Ca2+ release that was significantly suppressed by flecainide and a reverse-mode Na+/Ca2+ exchanger (NCX) inhibitor.
More recently, a new role of hiPSC-CMs has emerged in deciphering the pathogenicity of variants of uncertain significance (VUS). Recent advances in sequencing technologies have made it feasible to sequence the entire exome of patients suspected of having LQTS to identify causal genes. Such testing can identify hundreds of novel nonsynonymous coding variants in any given individual98 and determining which one of them is pathogenic or benign is currently the biggest challenge in the management of IAS, especially when the variant is of uncertain significance. Using the iPSC platform, Gélinas et al99 demonstrated the pathogenicity of a novel variant Gly52Val in the KCNJ2 gene identified by whole-exome sequencing in an LQTS8 patient. Patient-specific hiPSC-CMs displayed significant APD prolongation and incidence of arrhythmia. Immunohistochemistry and whole-cell current recordings in HEK cells heterologously expressing the WT or the mutant Kir2.1 channel further confirmed a surface trafficking defect in the mutant channels.
In future studies, the creation of isogenic hiPSC lines with single variant changes using clustered regularly interspaced short palindromic repeats (CRISPR) genome editing will allow direct comparison of phenotypes at a cellular level. A unique advantage of genome editing technology in disease modeling is the ability to study variants in an isogenic background to rule out phenotypic variability from epigenetic differences or unknown genetic modifiers100. This novel approach of combining iPSCs and genome editing technology to decipher VUS pathogenicity in LQTS could aid in tailoring drug treatment and ICD therapy, and could potentially be applied to other cardiac disorders to accelerate progress toward realizing the promise of precision medicine in IAS.
We recently validated this approach in our lab101 by generating hiPSC-CMs from a VUS carrier of LQTS2 (T983I KCNH2) and demonstrated their aberrant electrophysiological phenotype (reduced IKr and prolonged APD). We further delineated the VUS pathogenicity using CRISPR genome-editing to selectively correct the variant which normalized the abnormality and introduced the homozygous KCNH2 variant in an otherwise healthy individual which exacerbated the disease phenotype. To our knowledge, this is the first report of combining human iPSC and genome editing platforms to decipher pathogenicity of uncertain variant.
LQTS8
LQTS8 (Timothy Syndrome), results from rare single amino acid substitutions in CACNA1C gene that encodes the LTCC. It is the most severe form of LQTS, characterized by multi-organ dysfunction and high mortality. Besides marked QT interval prolongation and severe ventricular arrhythmia, LQTS8 patients can present with congenital heart defects, AV block, syndactyly, autism, malignant hypoglycemia, and an abnormal immune system102, 103. The G406R and G402S mutations in CACNA1C reduce the rate of channel inactivation and thereby prolong the plateau phase of the cardiac AP102, 103. In 2011, Yazawa and colleagues60 modeled LQTS8 using patient-specific hiPSC-CMs. The diseased cells recapitulated electrical abnormalities such as prolonged APD, DADs, and altered Ca2+ transients. Roscovitine accelerated ICaL inactivation, reduced APD, restored the irregular Ca2+ transient, and decreased the frequency of abnormal depolarizations in LQTS8 hiPSC-CMs60. Although roscovitine rescued the electrophysiological abnormality in LQTS8 hiPSC-CMs, the exact mechanism by which it restored cardiac function was not clear. In a separate study104, it was shown that roscovitine exhibits its therapeutic effects in part by inhibiting CDK5, a key mediator involved in the regulation of CaV1.2 channels in CMs. These studies provide mechanistic insights into the regulation of CaV1.2 and the development of future therapeutics for Timothy syndrome patients104.
Catecholaminergic Polymorphic Ventricular Tachycardia
CPVT is an inherited disease characterized by abnormal intracellular Ca2+ handling and signaling in CMs. The underlying mechanism of CPVT involves abnormal Ca2+ release from the SR, which is often due either to GOF mutations in RYR2 (CPVT1)105 or LOF mutations in calsequestrin (CPVT2), a SR calcium buffering protein. Such abnormal Ca2+ release may cause DADs, similar to digitalis toxicity, leading to triggered activity and ventricular tachyarrhythmias. Clinically, CPVT is precipitated by increased levels of catecholamines (e.g. epinephrine) under conditions of emotional stress and physical exertion. β-blockers are the drugs of choice for CPVT patients, but they often fail to prevent fatal arrhythmias.
To date, several groups have successfully modeled CPVT using the hiPSC platform106–108. CPVT1 patient-specific hiPSC-CMs carrying the F2483I mutation in the RYR2107 recapitulated DADs after catecholaminergic stimulation while Ca2+ imaging studies revealed higher amplitudes and longer durations of spontaneous Ca2+ release at basal state compared to control hiPSC-CMs. In addition, the CICR events continued after repolarization that were abolished by forskolin. In yet another CPVT1 model of hiPSC-CMs harboring the RYR2-M4109R mutation106, hiPSC-CMs showed increased DAD incidence and Ca2+ transient irregularities as compared to control hiPSC-CMs, which were further exacerbated by β-adrenergic stimulation106. Flecainide eliminated DADs in this CPVT1 hiPSC-CM model106. These data indicate that hiPSC-CMs can recapitulate the cardinal features of CPVT, making them useful models to investigate IAS mechanisms and to expedite personalized therapy.
Short QT Syndrome
SQTS is a rare inheritable, autosomal dominant disorder characterized by accelerated cardiac repolarization, abnormally short QT intervals and an increased propensity to develop atrial and ventricular tachyarrhythmia. Cardiac arrest seems to be the most frequent symptom (up to 40%)109. SQTS has been associated with mutations in six genes (KCNQ1, KCNH2, KCNJ2, CACNA1C, CACNB2, and CACNA2D1) with GOF mutations in KCNH2 (SQTS1) being the most prevalent subtype110, 111. Since it is a repolarization abnormality, the causative ion channel genes overlap with those of BrS and LQTS (Figure 4). Additionally, Afib or atrial flutter are also observed in SQTS patients (particularly SQT1)112. Besides ICD, several pharmacological approaches have been applied to treat SQTS, all aiming to restore the duration of ventricular repolarization to physiological values. Gaita et al.113 tested several antiarrhythmic drugs (flecainide, sotalol, ibutilide, and hydroquinidine) on two SQTS families carrying the N588K mutation in KCNH2113. Hydroquinidine was found to be the only agent that effectively prolonged the QT interval and abolished the inducibility of ventricular arrhythmias during programmed electrical stimulation.
To date, there has been only one report of modeling SQTS using the hiPSC platform. El-Battrawy and colleagues114 demonstrated that hiPSC-CMs derived from a patient carrying a N588K mutation in the KCNH2 recapitulated single-cell phenotype of SQTS. In contrast to the single cell phenotype of LQTS2, SQTS patient-specific hiPSC-CMs displayed markedly enhanced IKr density and shortened APD compared to healthy control. These cells further demonstrated abnormal Ca2+ transients and arrhythmic activities. Carbachol increased the arrhythmic events in SQTS while quinidine, but not sotalol or metoprolol, prolonged the APD and abolished arrhythmic activity induced by carbachol. These studies provide novel opportunities to further elucidate the pathophysiological mechanisms of SQTS and development of effective treatment strategies.
Atrial Fibrillation
AFib represents the most common type of supraventricular arrhythmia especially in the elderly population, oftentimes associated with underlying ischemic or structural heart disease, hypertension, diabetes, autonomic imbalance, as well as electrical remodeling of the atria. An underlying genetic component (Figure 4) has been associated with AFib only in a small number of cases of ‘lone AFib’ (AFib in the absence of predisposing factors), however inheritance is complex and mostly ‘non-Mendelian’115. The primary arrhythmia mechanisms contributing to AFib are focal ectopic firing and reentrant activity (Figure 2D). Ablation techniques have improved outcomes over the last two decades, but are associated with a risk of complications, and are only successful in a limited number of patients with permanent AFib. Pharmacologic therapy remains as the first-line treatment, and an important adjuvant to ablation therapy, however it is limited by a lack of efficacy and serious side effects such as ventricular proarrhythmia.
One of the major barriers to an improved mechanistic understanding of AFib, and thus drug development, has been a lack of appropriate models53. Until recently, hiPSC-based models of AFib were lacking because most hiPSC-CM differentiation protocols tend to generate a predominant ventricular-like phenotype. This methodological hurdle has been recently solved by manipulation of retinoic acid (RA) signaling, a physiological pathway that also drives atrial specification in vivo30, 116. Recently, Laksman et al.53 used human ESCs to generate an atrial-specific tissue model of AFib for pharmacologic testing. These atrial-like CM sheets showed uniform AP propagation, and rapid reentrant rotor patterns as seen in AFib could be induced. It was demonstrated53 that anti-arrhythmic drugs such as flecainide and dofetilide modulated reentrant arrhythmogenic rotor activation patterns in a manner that helps explain their efficacy in treating and preventing AFib.
Limitations of hiPSC-CMs
hiPSC-CMs have emerged as a promising experimental tool for translational cardiovascular research and drug development. However, their applicability as a human adult CM model is still hindered by several limitations. We have already mentioned on several occasions in this review, one of the major limitations of hiPSC technology — the immaturity of hiPSC-CMs. Compared to adult CMs, immature hiPSC-CMs have several differences, including a lack of T-tubules, abnormal morphology, altered gene expression, and reduced sarcomere organization117, 118. Genetic modifications during reprogramming, incomplete reprogramming resulting in residual epigenetic memory, induced alterations during passaging in culture, and differentiation-dependent variability and heterogeneity of iPSC-CMs are additional problems119. With regard to the challenges specific to disease modeling, it is not yet possible to model all human diseases using the hiPSC platform, for example diseases based on complex interactions of multiple genetic and environmental factors or on long incubation time, such as atherosclerosis and congenital heart diseases120. Moreover, differences in the individual genetic background present a particularly vexing problem to the ‘disease-in-a-dish’ approach due to the uncontrolled impact of genetic modifier loci. Thanks to the recent advances in genome editing tools such as zinc finger nucleases (ZFN), transcription activator–like effector nuclease (TALEN), and CRISPR genome editing, this limitation can now be addressed using genetically manipulated isogenic controls119. The robust combination of hiPSCs and genome editing will provide a unique opportunity to systematically and faithfully recapitulate human diseases in vitro, revealing new insights into the pathophysiology of monogenic and complex disorders119. However, in order to use hiPSCs for precision medicine, the development of improved differentiation protocols and 3D bioengineering approaches to create mature, adult-like CMs will be of paramount importance.
Conclusions and Future Perspectives
IAS represents a group of potentially lethal disorders that remain a diagnostic and therapeutic challenge due to complexity of the disorder and variable disease penetrance. Studies based on in vitro expression of mutations and transgenic animal models have been instrumental in offering the practitioner tools for deciphering these rare disorders but their potential remains limited. At present, there are still many unanswered questions such as, why mutations in the same gene can cause different types of arrhythmias, or why patients carrying the same mutation may exhibit completely different phenotypes ranging from being completely asymptomatic throughout life to experiencing SCD at an early age. In this regard, the role of the genetic modifiers that modulate the outcome in a mutation-and patient-specific manner remains unknown. Recent advances in the derivation of hiPSCs directly from diseased patients now provide a great opportunity to unravel these diverse mechanisms unique to a mutation/patient. Combined with the rapidly advancing genome editing approaches, iPSC technology has already emerged as a powerful platform for studying genotype–phenotype association and for predicting patient response to individualized therapy50. Cleary, iPSCs hold enormous potential in advancing the practice of precision medicine. In the future, a more thorough knowledge of the disease processes of these syndromes may enable us to use targeted therapy alone and minimize the need for ICDs.
Acknowledgments
Sources of Funding
We gratefully acknowledge funding support from National Institutes of Health (NIH) T32 EB009035 (PG), NIH R01 HL129798 (TJK), NIH U01 HL134764 (TJK) NIH R01 HL113006 (JCW), NIH R01 HL128170 (JCW), NIH R24 HL117756 (JCW) and American Heart Association 17SDG33660926 (VG).
Nonstandard Abbreviations and Acronyms
- AFib
Atrial fibrillation
- AP
Action potential
- APD
Action potential duration
- AVCM
Adult ventricular cardiomyocytes
- BrS
Brugada syndrome
- CALM
Calmodulin
- CICR
Calcium-induced calcium release
- CM
Cardiomyocytes
- CPVT
Catecholaminergic polymorphic ventricular tachycardia
- DAD
Delayed afterdepolarization
- DPP6
Dipeptidyl Peptidase Like 6
- EAD
Early afterdepolarization
- EKG
Electrocardiogram
- GOF
Gain-of-function
- HCN
Hyperpolarization-activated cyclic nucleotide-gated
- hERG
Human ether-a-go-go related gene
- hESC
Human embryonic stem cells
- ICD
Implantable cardioverter defibrillator
- iPSC
Induced pluripotent stem cells
- iPSC-CM
Induced pluripotent stem cell-derived cardiomyocytes
- KCHIP2
Kv channel-interacting protein 2
- Kir
Inward rectifier K+ channels
- Kv
Voltage-gated K+ channels
- LOF
Loss-of-function
- LQTS
Long QT syndrome
- LTCC
L-type Ca2+ channels
- MEA
Multielectrode array
- MiRP1
MinK-related peptide 1
- MP
Membrane potential
- NCX
Na+/Ca2+- exchanger type-1
- PKP2
Plakophilin-2
- RA
Retinoic acid
- RMP
Resting membrane potential
- RYR2
Type 2 ryanodine receptors
- SAN
Sinoatrial node
- SCD
Sudden cardiac death
- SERCA2a
Sarco/endoplasmic reticulum Ca2+-ATPase type-2a
- SQTS
Short QT syndrome
- TM
Transmembrane
Footnotes
Disclosures
None
References
- 1.Bezzina CR, Lahrouchi N and Priori SG. Genetics of sudden cardiac death. Circ Res 2015;116:1919–36. [DOI] [PubMed] [Google Scholar]
- 2.Abriel H, Rougier JS and Jalife J. Ion channel macromolecular complexes in cardiomyocytes: Roles in sudden cardiac death. Circ Res 2015;116:1971–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Rajamohan D, Matsa E, Kalra S, Crutchley J, Patel A, George V and Denning C. Current status of drug screening and disease modelling in human pluripotent stem cells. Bioessays 2013;35:281–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K and Yamanaka S. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 2007;131:861–72. [DOI] [PubMed] [Google Scholar]
- 5.Takahashi K and Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006;126:663–76. [DOI] [PubMed] [Google Scholar]
- 6.Marban E. Cardiac channelopathies. Nature 2002;415:213–8. [DOI] [PubMed] [Google Scholar]
- 7.Hille B. Ionic channels of excitable membranes Sunderland, Mass.: Sinaur Associates; 2001. [Google Scholar]
- 8.Kanno S and Saffitz JE. The role of myocardial gap junctions in electrical conduction and arrhythmogenesis. Cardiovasc Pathol 2001;10:169–77. [DOI] [PubMed] [Google Scholar]
- 9.Nerbonne JM and Guo W. Heterogeneous expression of voltage-gated potassium channels in the heart: Roles in normal excitation and arrhythmias. J Cardiovasc Electrophysiol 2002;13:406–9. [DOI] [PubMed] [Google Scholar]
- 10.Nerbonne JM and Kass RS. Molecular physiology of cardiac repolarization. Physiol Rev 2005;85:1205–53. [DOI] [PubMed] [Google Scholar]
- 11.Grant AO. Cardiac ion channels. Circ Arrhythm Electrophysiol 2009;2:185–94. [DOI] [PubMed] [Google Scholar]
- 12.Fermini B and Fossa AA. The impact of drug-induced QT interval prolongation on drug discovery and development. Nat Rev Drug Discov 2003;2:439–47. [DOI] [PubMed] [Google Scholar]
- 13.Tse G. Mechanisms of cardiac arrhythmias. J Arrhythm 2016;32:75–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Shibata EF, Drury T, Refsum H, Aldrete V and Giles W. Contributions of a transient outward current to repolarization in human atrium. Am J Physiol 1989;257:H1773–81. [DOI] [PubMed] [Google Scholar]
- 15.Wickman K, Krapivinsky G, Corey S, Kennedy M, Nemec J, Medina I and Clapham DE. Structure, G-protein activation, and functional relevance of the cardiac G protein-gated K+ channel, IKACh. Ann N Y Acad Sci 1999;868:386–98. [DOI] [PubMed] [Google Scholar]
- 16.Lakatta EG, Vinogradova T, Lyashkov A, Sirenko S, Zhu W, Ruknudin A and Maltsev VA. The integration of spontaneous intracellular Ca2+ cycling and surface membrane ion channel activation entrains normal automaticity in cells of the heart’s pacemaker. Ann N Y Acad Sci 2006;1080:178–206. [DOI] [PubMed] [Google Scholar]
- 17.Biel M, Wahl-Schott C, Michalakis S and Zong X. Hyperpolarization-activated cation channels: from genes to function. Physiol Rev 2009;89:847–85. [DOI] [PubMed] [Google Scholar]
- 18.Sandow A. Excitation-contraction coupling in muscular response. Yale J Biol Med 1952;25:176–201. [PMC free article] [PubMed] [Google Scholar]
- 19.Bers DM. Cardiac excitation-contraction coupling. Nature 2002;415:198–205. [DOI] [PubMed] [Google Scholar]
- 20.Cannell MB, Berlin JR and Lederer WJ. Intracellular calcium in cardiac myocytes: calcium transients measured using fluorescence imaging. Soc Gen Physiol Ser 1987;42:201–14. [PubMed] [Google Scholar]
- 21.Cheng H, Lederer WJ and Cannell MB. Calcium sparks: elementary events underlying excitation-contraction coupling in heart muscle. Science 1993;262:740–4. [DOI] [PubMed] [Google Scholar]
- 22.Endo M. Calcium release from the sarcoplasmic reticulum. Physiol Rev 1977;57:71–108. [DOI] [PubMed] [Google Scholar]
- 23.Landstrom AP, Dobrev D and Wehrens XHT. Calcium signaling and cardiac arrhythmias. Circ Res 2017;120:1969–1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Hoekstra M, Mummery CL, Wilde AA, Bezzina CR and Verkerk AO. Induced pluripotent stem cell derived cardiomyocytes as models for cardiac arrhythmias. Front Physiol 2012;3:346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Casini S, Verkerk AO and Remme CA. Human iPSC-derived cardiomyocytes for investigation of disease mechanisms and therapeutic strategies in inherited arrhythmia syndromes: Strengths and limitations. Cardiovasc Drugs Ther 2017;31:325–344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Burridge PW, Keller G, Gold JD and Wu JC. Production of de novo cardiomyocytes: human pluripotent stem cell differentiation and direct reprogramming. Cell Stem Cell 2012;10:16–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Smith AS, Macadangdang J, Leung W, Laflamme MA and Kim DH. Human iPSC-derived cardiomyocytes and tissue engineering strategies for disease modeling and drug screening. Biotechnol Adv 2017;35:77–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Lian X, Hsiao C, Wilson G, Zhu K, Hazeltine LB, Azarin SM, Raval KK, Zhang J, Kamp TJ and Palecek SP. Robust cardiomyocyte differentiation from human pluripotent stem cells via temporal modulation of canonical Wnt signaling. Proc Natl Acad Sci U S A 2012;109:E1848–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Burridge PW, Matsa E, Shukla P, Lin ZC, Churko JM, Ebert AD, Lan F, Diecke S, Huber B, Mordwinkin NM, Plews JR, Abilez OJ, Cui B, Gold JD and Wu JC. Chemically defined generation of human cardiomyocytes. Nat Methods 2014;11:855–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Devalla HD, Schwach V, Ford JW, Milnes JT, El-Haou S, Jackson C, Gkatzis K, Elliott DA, Chuva de Sousa Lopes SM, Mummery CL, Verkerk AO and Passier R. Atrial-like cardiomyocytes from human pluripotent stem cells are a robust preclinical model for assessing atrial-selective pharmacology. EMBO Mol Med 2015;7:394–410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Protze SI, Liu J, Nussinovitch U, Ohana L, Backx PH, Gepstein L and Keller GM. Sinoatrial node cardiomyocytes derived from human pluripotent cells function as a biological pacemaker. Nat Biotechnol 2017;35:56–68. [DOI] [PubMed] [Google Scholar]
- 32.Satin J, Kehat I, Caspi O, Huber I, Arbel G, Itzhaki I, Magyar J, Schroder EA, Perlman I and Gepstein L. Mechanism of spontaneous excitability in human embryonic stem cell derived cardiomyocytes. J Physiol 2004;559:479–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Lieu DK, Fu JD, Chiamvimonvat N, Tung KC, McNerney GP, Huser T, Keller G, Kong CW and Li RA. Mechanism-based facilitated maturation of human pluripotent stem cell-derived cardiomyocytes. Circ Arrhythm Electrophysiol 2013;6:191–201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Vaidyanathan R, Markandeya YS, Kamp TJ, Makielski JC, January CT and Eckhardt LL. IK1-enhanced human-induced pluripotent stem cell-derived cardiomyocytes: An improved cardiomyocyte model to investigate inherited arrhythmia syndromes. American Journal of Physiology - Heart and Circulatory Physiology 2016;310:H1611–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Bett GC, Kaplan AD, Lis A, Cimato TR, Tzanakakis ES, Zhou Q, Morales MJ and Rasmusson RL. Electronic “expression” of the inward rectifier in cardiocytes derived from human-induced pluripotent stem cells. Heart rhythm : the official journal of the Heart Rhythm Society 2013;10:1903–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Meijer van Putten RM, Mengarelli I, Guan K, Zegers JG, van Ginneken AC, Verkerk AO and Wilders R. Ion channelopathies in human induced pluripotent stem cell derived cardiomyocytes: A dynamic clamp study with virtual IK1. Front Physiol 2015;6:7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Tulloch NL, Muskheli V, Razumova MV, Korte FS, Regnier M, Hauch KD, Pabon L, Reinecke H and Murry CE. Growth of engineered human myocardium with mechanical loading and vascular coculture. Circ Res 2011;109:47–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Jacot JG, McCulloch AD and Omens JH. Substrate stiffness affects the functional maturation of neonatal rat ventricular myocytes. Biophysical Journal 2008;95:3479–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Zhang D, Shadrin IY, Lam J, Xian HQ, Snodgrass HR and Bursac N. Tissue-engineered cardiac patch for advanced functional maturation of human ESC-derived cardiomyocytes. Biomaterials 2013;34:5813–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Nunes SS, Miklas JW, Liu J, Aschar-Sobbi R, Xiao Y, Zhang B, Jiang J, Masse S, Gagliardi M, Hsieh A, Thavandiran N, Laflamme MA, Nanthakumar K, Gross GJ, Backx PH, Keller G and Radisic M. Biowire: A platform for maturation of human pluripotent stem cell-derived cardiomyocytes. Nat Methods 2013;10:781–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Mihic A, Li J, Miyagi Y, Gagliardi M, Li SH, Zu J, Weisel RD, Keller G and Li RK. The effect of cyclic stretch on maturation and 3D tissue formation of human embryonic stem cell-derived cardiomyocytes. Biomaterials 2014;35:2798–808. [DOI] [PubMed] [Google Scholar]
- 42.Chattergoon NN, Giraud GD, Louey S, Stork P, Fowden AL and Thornburg KL. Thyroid hormone drives fetal cardiomyocyte maturation. The FASEB Journal 2012;26:397–408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Lundy SD, Zhu WZ, Regnier M and Laflamme MA. Structural and functional maturation of cardiomyocytes derived from human pluripotent stem cells. Stem Cells and Development 2013;22:1991–2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Ruan JL, Tulloch NL, Razumova MV, Saiget M, Muskheli V, Pabon L, Reinecke H, Regnier M and Murry CE. Mechanical stress conditioning and electrical stimulation promote contractility and force maturation of induced pluripotent stem cell-derived human cardiac tissue. Circulation 2016;134:1557–1567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Antzelevitch C and Burashnikov A. Overview of basic mechanisms of cardiac arrhythmia. Card Electrophysiol Clin 2011;3:23–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Bartos DC, Grandi E and Ripplinger CM. Ion channels in the heart. Compr Physiol 2015;5:1423–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.January CT, Chau V and Makielski JC. Triggered activity in the heart: Cellular mechanisms of Early after-depolarizations. Eur Heart J 1991;12 Suppl F:4–9. [DOI] [PubMed] [Google Scholar]
- 48.Wit AL and Boyden PA. Triggered activity and atrial fibrillation. Heart rhythm : the official journal of the Heart Rhythm Society 2007;4:S17–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.January CT and Riddle JM. Early afterdepolarizations: Mechanism of induction and block. A role for L-type Ca2+ current. Circ Res 1989;64:977–90. [DOI] [PubMed] [Google Scholar]
- 50.Martin CA, Matthews GD and Huang CL. Sudden cardiac death and inherited channelopathy: The basic electrophysiology of the myocyte and myocardium in ion channel disease. Heart 2012;98:536–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Holzem KM, Madden EJ and Efimov IR. Human cardiac systems electrophysiology and arrhythmogenesis: Iteration of experiment and computation. Europace 2014;16 Suppl 4:iv77–iv85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Noble D. Modeling the heart--from genes to cells to the whole organ. Science 2002;295:1678–82. [DOI] [PubMed] [Google Scholar]
- 53.Laksman Z, Wauchop M, Lin E, Protze S, Lee J, Yang W, Izaddoustdar F, Shafaattalab S, Gepstein L, Tibbits GF, Keller G and Backx PH. Modeling Atrial Fibrillation using human embryonic stem cell-derived atrial tissue. Sci Rep 2017;7:5268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Kadota S, Minami I, Morone N, Heuser JE, Agladze K and Nakatsuji N. Development of a reentrant arrhythmia model in human pluripotent stem cell-derived cardiac cell sheets. Eur Heart J 2013;34:1147–56. [DOI] [PubMed] [Google Scholar]
- 55.Neher E and Sakmann B. Single-channel currents recorded from membrane of denervated frog muscle fibres. Nature 1976;260:799–802. [DOI] [PubMed] [Google Scholar]
- 56.Laurila E, Ahola A, Hyttinen J and Aalto-Setala K. Methods for in vitro functional analysis of iPSC derived cardiomyocytes - Special focus on analyzing the mechanical beating behavior. Biochim Biophys Acta 2016;1863:1864–72. [DOI] [PubMed] [Google Scholar]
- 57.Matsa E, Dixon JE, Medway C, Georgiou O, Patel MJ, Morgan K, Kemp PJ, Staniforth A, Mellor I and Denning C. Allele-specific RNA interference rescues the Long-QT syndrome phenotype in human-induced pluripotency stem cell cardiomyocytes. Eur Heart J 2014;35:1078–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Moretti A, Bellin M, Welling A, Jung CB, Lam JT, Bott-Flugel L, Dorn T, Goedel A, Hohnke C, Hofmann F, Seyfarth M, Sinnecker D, Schomig A and Laugwitz KL. Patient-specific induced pluripotent stem-cell models for Long-QT syndrome. N Engl J Med 2010;363:1397–409. [DOI] [PubMed] [Google Scholar]
- 59.Lahti AL, Kujala VJ, Chapman H, Koivisto AP, Pekkanen-Mattila M, Kerkela E, Hyttinen J, Kontula K, Swan H, Conklin BR, Yamanaka S, Silvennoinen O and Aalto-Setala K. Model for Long QT syndrome type 2 using human iPS cells demonstrates arrhythmogenic characteristics in cell culture. Disease Models & Mechanisms 2012;5:220–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Yazawa M and Dolmetsch RE. Modeling Timothy syndrome with iPS cells. J Cardiovasc Transl Res 2013;6:1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Zhu H, Scharnhorst KS, Stieg AZ, Gimzewski JK, Minami I, Nakatsuji N, Nakano H and Nakano A. Two dimensional electrophysiological characterization of human pluripotent stem cell-derived cardiomyocyte system. Sci Rep 2017;7:43210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Halbach M, Egert U, Hescheler J and Banach K. Estimation of action potential changes from field potential recordings in multicellular mouse cardiac myocyte cultures. Cell Physiol Biochem 2003;13:271–84. [DOI] [PubMed] [Google Scholar]
- 63.Guo L, Qian JY, Abrams R, Tang HM, Weiser T, Sanders MJ and Kolaja KL. The electrophysiological effects of cardiac glycosides in human iPSC-derived cardiomyocytes and in guinea pig isolated hearts. Cell Physiol Biochem 2011;27:453–62. [DOI] [PubMed] [Google Scholar]
- 64.Navarrete EG, Liang P, Lan F, Sanchez-Freire V, Simmons C, Gong T, Sharma A, Burridge PW, Patlolla B, Lee AS, Wu H, Beygui RE, Wu SM, Robbins RC, Bers DM and Wu JC. Screening drug-induced arrhythmia [corrected] using human induced pluripotent stem cell-derived cardiomyocytes and low-impedance microelectrode arrays. Circulation 2013;128:S3–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Shinozawa T, Nakamura K, Shoji M, Morita M, Kimura M, Furukawa H, Ueda H, Shiramoto M, Matsuguma K, Kaji Y, Ikushima I, Yono M, Liou SY, Nagai H, Nakanishi A, Yamamoto K and Izumo S. Recapitulation of clinical individual susceptibility to drug-induced QT prolongation in healthy subjects using iPSC-derived cardiomyocytes. Stem Cell Reports 2017;8:226–234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Stillitano F, Hansen J, Kong CW, Karakikes I, Funck-Brentano C, Geng L, Scott S, Reynier S, Wu M, Valogne Y, Desseaux C, Salem JE, Jeziorowska D, Zahr N, Li R, Iyengar R, Hajjar RJ and Hulot JS. Modeling susceptibility to drug-induced Long QT with a panel of subject-specific induced pluripotent stem cells. eLife 2017;6. [DOI] [PMC free article] [PubMed]
- 67.Veerman CC, Kosmidis G, Mummery CL, Casini S, Verkerk AO and Bellin M. Immaturity of human stem-cell-derived cardiomyocytes in culture: Fatal flaw or soluble problem? Stem Cells and Development 2015;24:1035–52. [DOI] [PubMed] [Google Scholar]
- 68.Penttinen K, Siirtola H, Avalos-Salguero J, Vainio T, Juhola M and Aalto-Setala K. Novel analysis software for detecting and classifying Ca2+ transient abnormalities in stem cell-derived cardiomyocytes. PLoS ONE 2015;10:e0135806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Herron TJ, Lee P and Jalife J. Optical imaging of voltage and calcium in cardiac cells & tissues. Circ Res 2012;110:609–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Shinnawi R, Huber I, Maizels L, Shaheen N, Gepstein A, Arbel G, Tijsen AJ and Gepstein L. Monitoring human-induced pluripotent stem cell-derived cardiomyocytes with genetically encoded calcium and voltage fluorescent reporters. Stem Cell Reports 2015;5:582–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Peters MF, Lamore SD, Guo L, Scott CW and Kolaja KL. Human stem cell-derived cardiomyocytes in cellular impedance assays: Bringing cardiotoxicity screening to the front line. Cardiovasc Toxicol 2015;15:127–39. [DOI] [PubMed] [Google Scholar]
- 72.Koci B, Luerman G, Duenbostell A, Kettenhofen R, Bohlen H, Coyle L, Knight B, Ku W, Volberg W, Woska JR Jr. and Brown MP. An impedance-based approach using human iPSC-derived cardiomyocytes significantly improves in vitro prediction of in vivo cardiotox liabilities. Toxicol Appl Pharmacol 2017;329:121–127. [DOI] [PubMed] [Google Scholar]
- 73.Doerr L, Thomas U, Guinot DR, Bot CT, Stoelzle-Feix S, Beckler M, George M and Fertig N. New easy-to-use hybrid system for extracellular potential and impedance recordings. J Lab Autom 2015;20:175–88. [DOI] [PubMed] [Google Scholar]
- 74.Xi B, Wang T, Li N, Ouyang W, Zhang W, Wu J, Xu X, Wang X and Abassi YA. Functional cardiotoxicity profiling and screening using the xCELLigence RTCA Cardio System. J Lab Autom 2011;16:415–21. [DOI] [PubMed] [Google Scholar]
- 75.Abassi YA, Xi B, Li N, Ouyang W, Seiler A, Watzele M, Kettenhofen R, Bohlen H, Ehlich A, Kolossov E, Wang X and Xu X. Dynamic monitoring of beating periodicity of stem cell-derived cardiomyocytes as a predictive tool for preclinical safety assessment. British Journal of Pharmacology 2012;165:1424–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Himmel HM, Wettwer E, Li Q and Ravens U. Four different components contribute to outward current in rat ventricular myocytes. Am J Physiol 1999;277:H107–18. [DOI] [PubMed] [Google Scholar]
- 77.Guo L, Eldridge S, Furniss M, Mussio J and Davis M. Use of human induced pluripotent stem cell–derived cardiomyocytes (hiPSC-CMs) to monitor compound effects on cardiac myocyte signaling pathways Current Protocols in Chemical Biology: John Wiley & Sons, Inc.; 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Remme CA. Cardiac sodium channelopathy associated with SCN5A mutations: Electrophysiological, molecular and genetic aspects. J Physiol 2013;591:4099–116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Remme CA and Bezzina CR. Sodium channel (dys)function and cardiac arrhythmias. Cardiovasc Ther 2010;28:287–94. [DOI] [PubMed] [Google Scholar]
- 80.Selga E, Sendfeld F, Martinez-Moreno R, Medine CN, Tura-Ceide O, Wilmut SI, Perez GJ, Scornik FS, Brugada R and Mills NL. Sodium channel current loss of function in induced pluripotent stem cell-derived cardiomyocytes from a Brugada Syndrome patient. J Mol Cell Cardiol 2018;114:10–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Liang P, Sallam K, Wu H, Li Y, Itzhaki I, Garg P, Zhang Y, Vermglinchan V, Lan F, Gu M, Gong T, Zhuge Y, He C, Ebert AD, Sanchez-Freire V, Churko J, Hu S, Sharma A, Lam CK, Scheinman MM, Bers DM and Wu JC. Patient-specific and genome-edited induced pluripotent stem cell-derived aardiomyocytes elucidate single-cell phenotype of Brugada Syndrome. J Am Coll Cardiol 2016;68:2086–2096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Davis RP, Casini S, van den Berg CW, Hoekstra M, Remme CA, Dambrot C, Salvatori D, Oostwaard DW, Wilde AA, Bezzina CR, Verkerk AO, Freund C and Mummery CL. Cardiomyocytes derived from pluripotent stem cells recapitulate electrophysiological characteristics of an overlap syndrome of cardiac sodium channel disease. Circulation 2012;125:3079–91. [DOI] [PubMed] [Google Scholar]
- 83.Bhuiyan ZA, Al-Shahrani S, Al-Aama J, Wilde AA and Momenah TS. Congenital Long QT Syndrome: An update and present perspective in Saudi Arabia. Front Pediatr 2013;1:39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Ma D, Wei H, Lu J, Huang D, Liu Z, Loh LJ, Islam O, Liew R, Shim W and Cook SA. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Egashira T, Yuasa S, Suzuki T, Aizawa Y, Yamakawa H, Matsuhashi T, Ohno Y, Tohyama S, Okata S, Seki T, Kuroda Y, Yae K, Hashimoto H, Tanaka T, Hattori F, Sato T, Miyoshi S, Takatsuki S, Murata M, Kurokawa J, Furukawa T, Makita N, Aiba T, Shimizu W, Horie M, Kamiya K, Kodama I, Ogawa S and Fukuda K. Disease characterization using LQTS-specific induced pluripotent stem cells. Cardiovasc Res 2012;95:419–29. [DOI] [PubMed] [Google Scholar]
- 86.Itzhaki I, Maizels L, Huber I, Zwi-Dantsis L, Caspi O, Winterstern A, Feldman O, Gepstein A, Arbel G, Hammerman H, Boulos M and Gepstein L. Modelling the Long QT syndrome with induced pluripotent stem cells. Nature 2011;471:225–9. [DOI] [PubMed] [Google Scholar]
- 87.Matsa E, Ahrens JH and Wu JC. Human induced pluripotent stem cells as a platform for personalized and precision cardiovascular medicine. Physiol Rev 2016;96:1093–126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Mehta A, Ramachandra CJA, Singh P, Chitre A, Lua CH, Mura M, Crotti L, Wong P, Schwartz PJ, Gnecchi M and Shim W. Identification of a targeted and testable antiarrhythmic therapy for Long-QT syndrome type 2 using a patient-specific cellular model. Eur Heart J 2018;39:1446–1455. [DOI] [PubMed] [Google Scholar]
- 89.Schwartz PJ, Priori SG, Spazzolini C, Moss AJ, Vincent GM, Napolitano C, Denjoy I, Guicheney P, Breithardt G, Keating MT, Towbin JA, Beggs AH, Brink P, Wilde AA, Toivonen L, Zareba W, Robinson JL, Timothy KW, Corfield V, Wattanasirichaigoon D, Corbett C, Haverkamp W, Schulze-Bahr E, Lehmann MH, Schwartz K, Coumel P and Bloise R. Genotype-phenotype correlation in the Long-QT syndrome: Gene-specific triggers for life-threatening arrhythmias. Circulation 2001;103:89–95. [DOI] [PubMed] [Google Scholar]
- 90.Fatima A, Kaifeng S, Dittmann S, Xu G, Gupta MK, Linke M, Zechner U, Nguemo F, Milting H, Farr M, Hescheler J and Saric T. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Ma D, Wei H, Zhao Y, Lu J, Li G, Sahib NB, Tan TH, Wong KY, Shim W, Wong P, Cook SA and Liew R. Modeling type 3 Long QT syndrome with cardiomyocytes derived from patient-specific induced pluripotent stem cells. Int J Cardiol 2013;168:5277–86. [DOI] [PubMed] [Google Scholar]
- 92.Terrenoire C, Wang K, Tung KW, Chung WK, Pass RH, Lu JT, Jean JC, Omari A, Sampson KJ, Kotton DN, Keller G and Kass RS. Induced pluripotent stem cells used to reveal drug actions in a Long QT syndrome family with complex genetics. J Gen Physiol 2013;141:61–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Plaster NM, Tawil R, Tristani-Firouzi M, Canun S, Bendahhou S, Tsunoda A, Donaldson MR, Iannaccone ST, Brunt E, Barohn R, Clark J, Deymeer F, George AL Jr., Fish FA, Hahn, Nitu A, Ozdemir C, Serdaroglu P, Subramony SH, Wolfe G, YH Fu and Ptacek LJ. Mutations in Kir2.1 cause the developmental and episodic electrical phenotypes of Andersen’s syndrome. Cell 2001;105:511–9. [DOI] [PubMed] [Google Scholar]
- 94.Nguyen H-L, Pieper GH and Wilders R. Andersen–Tawil syndrome: Clinical and molecular aspects. International Journal of Cardiology 2013;170:1–16. [DOI] [PubMed] [Google Scholar]
- 95.Preisig-Muller R, Schlichthorl G, Goerge T, Heinen S, Bruggemann A, Rajan S, Derst C, Veh RW and Daut J. Heteromerization of Kir2.x potassium channels contributes to the phenotype of Andersen’s syndrome. Proc Natl Acad Sci U S A 2002;99:7774–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Ai T, Fujiwara Y, Tsuji K, Otani H, Nakano S, Kubo Y and Horie M. Novel KCNJ2 mutation in familial periodic paralysis with ventricular dysrhythmia. Circulation 2002;105:2592–4. [DOI] [PubMed] [Google Scholar]
- 97.Kuroda Y, Yuasa S, Watanabe Y, Ito S, Egashira T, Seki T, Hattori T, Ohno S, Kodaira M, Suzuki T, Hashimoto H, Okata S, Tanaka A, Aizawa Y, Murata M, Aiba T, Makita N, Furukawa T, Shimizu W, Kodama I, Ogawa S, Kokubun N, Horigome H, Horie M, Kamiya K and Fukuda K. Flecainide ameliorates arrhythmogenicity through NCX flux in Andersen-Tawil syndrome-iPS cell-derived cardiomyocytes. Biochem Biophys Rep 2017;9:245–256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Xue Y, Chen Y, Ayub Q, Huang N, Ball EV, Mort M, Phillips AD, Shaw K, Stenson PD, Cooper DN, Tyler-Smith C and Genomes Project C. Deleterious- and disease-allele prevalence in healthy individuals: Insights from current predictions, mutation databases, and population-scale resequencing. Am J Hum Genet 2012;91:1022–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Gelinas R, El Khoury N, Chaix MA, Beauchamp C, Alikashani A, Ethier N, Boucher G, Villeneuve L, Robb L, Latour F, Mondesert B, Rivard L, Goyette P, Talajic M, Fiset C and Rioux JD. Characterization of a human induced pluripotent stem cell-derived cardiomyocyte model for the study of variant pathogenicity: Validation of a KCNJ2 mutation. Circ Cardiovasc Genet 2017;10. [DOI] [PubMed]
- 100.Bassett AR. Editing the genome of hiPSC with CRISPR/Cas9: disease models. Mamm Genome 2017;28:348–364. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Garg P, Oikonomopoulos A, Chen H, Li Y, Lam CK, Sallam K, Perez M, Lux RL, Sanguinetti MC and WU JC. Genome editing of induced pluripotent stem cells to decipher cardiac channelopathy variant. J Am Coll Cardiol 2018. (in press). [DOI] [PMC free article] [PubMed]
- 102.Splawski I, Timothy KW, Sharpe LM, Decher N, Kumar P, Bloise R, Napolitano C, Schwartz PJ, Joseph RM, Condouris K, Tager-Flusberg H, Priori SG, Sanguinetti MC and Keating MT. CaV1.2 calcium channel dysfunction causes a multisystem disorder including arrhythmia and autism. Cell 2004;119:19–31. [DOI] [PubMed] [Google Scholar]
- 103.Splawski I, Timothy KW, Decher N, Kumar P, Sachse FB, Beggs AH, Sanguinetti MC and Keating MT. Severe arrhythmia disorder caused by cardiac L-type calcium channel mutations. Proc Natl Acad Sci U S A 2005;102:8089–96; discussion 8086–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Song L, Park SE, Isseroff Y, Morikawa K and Yazawa M. Inhibition of CDK5 alleviates the cardiac phenotypes in Timothy Syndrome. Stem Cell Reports 2017;9:50–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Marks AR, Priori S, Memmi M, Kontula K and Laitinen PJ. Involvement of the cardiac ryanodine receptor/calcium release channel in Catecholaminergic Polymorphic Ventricular Tachycardia. J Cell Physiol 2002;190:1–6. [DOI] [PubMed] [Google Scholar]
- 106.Itzhaki I, Maizels L, Huber I, Gepstein A, Arbel G, Caspi O, Miller L, Belhassen B, Nof E, Glikson M and Gepstein L. Modeling of Catecholaminergic Polymorphic Ventricular Tachycardia with patient-specific human-induced pluripotent stem cells. J Am Coll Cardiol 2012;60:990–1000. [DOI] [PubMed] [Google Scholar]
- 107.Fatima A, Xu G, Shao K, Papadopoulos S, Lehmann M, Arnaiz-Cot JJ, Rosa AO, Nguemo F, Matzkies M, Dittmann S, Stone SL, Linke M, Zechner U, Beyer V, Hennies HC, Rosenkranz S, Klauke B, Parwani AS, Haverkamp W, Pfitzer G, Farr M, Cleemann L, Morad M, Milting H, Hescheler J and Saric T. In vitro modeling of ryanodine receptor 2 dysfunction using human induced pluripotent stem cells. Cell Physiol Biochem 2011;28:579–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Jung CB, Moretti A, Mederos y Schnitzler M, Iop L, Storch U, Bellin M, Dorn T, Ruppenthal S, Pfeiffer S, Goedel A, Dirschinger RJ, Seyfarth M, Lam JT, Sinnecker D, Gudermann T, Lipp P and Laugwitz KL. Dantrolene rescues arrhythmogenic RYR2 defect in a patient-specific stem cell model of Catecholaminergic Polymorphic Ventricular Tachycardia. EMBO Mol Med 2012;4:180–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Mazzanti A, O’Rourke S, Ng K, Miceli C, Borio G, Curcio A, Esposito F, Napolitano C and Priori SG. The usual suspects in sudden cardiac death of the young: A focus on inherited arrhythmogenic diseases. Expert Rev Cardiovasc Ther 2014;12:499–519. [DOI] [PubMed] [Google Scholar]
- 110.Fernandez-Falgueras A, Sarquella-Brugada G, Brugada J, Brugada R and Campuzano O. Cardiac channelopathies and sudden death: Recent clinical and genetic advances. Biology (Basel) 2017;6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Mazzanti A, Underwood K, Nevelev D, Kofman S and Priori SG. The new kids on the block of arrhythmogenic disorders: Short QT syndrome and Early repolarization. J Cardiovasc Electrophysiol 2017;28:1226–1236. [DOI] [PubMed] [Google Scholar]
- 112.Giustetto C, Schimpf R, Mazzanti A, Scrocco C, Maury P, Anttonen O, Probst V, Blanc JJ, Sbragia P, Dalmasso P, Borggrefe M and Gaita F. Long-term follow-up of patients with Short QT syndrome. J Am Coll Cardiol 2011;58:587–95. [DOI] [PubMed] [Google Scholar]
- 113.Gaita F, Giustetto C, Bianchi F, Schimpf R, Haissaguerre M, Calo L, Brugada R, Antzelevitch C, Borggrefe M and Wolpert C. Short QT syndrome: pharmacological treatment. J Am Coll Cardiol 2004;43:1494–9. [DOI] [PubMed] [Google Scholar]
- 114.El-Battrawy I, Lan H, Cyganek L, Zhao Z, Li X, Buljubasic F, Lang S, Yucel G, Sattler K, Zimmermann WH, Utikal J, Wieland T, Ravens U, Borggrefe M, Zhou XB and Akin I. Modeling Short QT syndrome using human-induced pluripotent stem cell-derived cardiomyocytes. J Am Heart Assoc 2018;7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Olesen MS, Nielsen MW, Haunso S and Svendsen JH. Atrial fibrillation: The role of common and rare genetic variants. Eur J Hum Genet 2014;22:297–306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Zaffran S, Robrini N and Bertrand N. Retinoids and cardiac development. Journal of Developmental Biology 2014;2:50. [Google Scholar]
- 117.Yang X, Pabon L and Murry CE. Engineering adolescence: Maturation of human pluripotent stem cell-derived cardiomyocytes. Circ Res 2014;114:511–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Sayed N, Liu C and Wu JC. Translation of human-induced pluripotent ctem cells: From clinical trial in a dish to precision medicine. J Am Coll Cardiol 2016;67:2161–2176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Soldner F and Jaenisch R. Medicine. iPSC disease modeling. Science 2012;338:1155–6. [DOI] [PubMed] [Google Scholar]
- 120.Youssef AA, Ross EG, Bolli R, Pepine CJ, Leeper NJ and Yang PC. The promise and challenge of induced pluripotent stem cells for cardiovascular applications. JACC Basic Transl Sci 2016;1:510–523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Zwi L, Caspi O, Arbel G, Huber I, Gepstein A, Park IH and Gepstein L. Cardiomyocyte differentiation of human induced pluripotent stem cells. Circulation 2009;120:1513–23. [DOI] [PubMed] [Google Scholar]
- 122.Karakikes I, Termglinchan V, Cepeda DA, Lee J, Diecke S, Hendel A, Itzhaki I, Ameen M, Shrestha R, Wu H, Ma N, Shao NY, Seeger T, Woo N, Wilson KD, Matsa E, Porteus MH, Sebastiano V and Wu JC. A comprehensive TALEN-based knockout library for generating human-induced pluripotent stem cell-based models for cardiovascular diseases. Circ Res 2017;120:1561–1571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Ma J, Guo L, Fiene SJ, Anson BD, Thomson JA, Kamp TJ, Kolaja KL, Swanson BJ and January CT. High purity human-induced pluripotent stem cell-derived cardiomyocytes: electrophysiological properties of action potentials and ionic currents. American Journal of Physiology - Heart and Circulatory Physiology 2011;301:H2006–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.van den Boogaard M, Smemo S, Burnicka-Turek O, Arnolds DE, van de Werken HJ, Klous P, McKean D, Muehlschlegel JD, Moosmann J, Toka O, Yang XH, Koopmann TT, Adriaens ME, Bezzina CR, de Laat W, Seidman C, Seidman JG, Christoffels VM, Nobrega MA, Barnett P and Moskowitz IP. A common genetic variant within SCN10A modulates cardiac SCN5A expression. J Clin Invest 2014;124:1844–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Chambers JC, Zhao J, Terracciano CM, Bezzina CR, Zhang W, Kaba R, Navaratnarajah M, Lotlikar A, Sehmi JS, Kooner MK, Deng G, Siedlecka U, Parasramka S, El-Hamamsy I, Wass MN, Dekker LR, de Jong JS, Sternberg MJ, McKenna W, Severs NJ, de Silva R, Wilde AA, Anand P, Yacoub M, Scott J, Elliott P, Wood JN and Kooner JS. Genetic variation in SCN10A influences cardiac conduction. Nat Genet 2010;42:149–52. [DOI] [PubMed] [Google Scholar]
- 126.Hofmann F, Flockerzi V, Kahl S and Wegener JW. L-type CaV1.2 calcium channels: From in vitro findings to in vivo function. Physiol Rev 2014;94:303–26. [DOI] [PubMed] [Google Scholar]
- 127.Goonasekera SA, Hammer K, Auger-Messier M, Bodi I, Chen X, Zhang H, Reiken S, Elrod JW, Correll RN, York AJ, Sargent MA, Hofmann F, Moosmang S, Marks AR, Houser SR, Bers DM and Molkentin JD. Decreased cardiac L-type Ca2+ channel activity induces hypertrophy and heart failure in mice. J Clin Invest 2012;122:280–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Seisenberger C, Specht V, Welling A, Platzer J, Pfeifer A, Kuhbandner S, Striessnig J, Klugbauer N, Feil R and Hofmann F. Functional embryonic cardiomyocytes after disruption of the L-type alpha1C (Cav1.2) calcium channel gene in the mouse. J Biol Chem 2000;275:39193–9. [DOI] [PubMed] [Google Scholar]
- 129.Niwa N and Nerbonne JM. Molecular determinants of cardiac transient outward potassium current (Ito) expression and regulation. J Mol Cell Cardiol 2010;48:12–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Guo W, Jung WE, Marionneau C, Aimond F, Xu H, Yamada KA, Schwarz TL, Demolombe S and Nerbonne JM. Targeted deletion of Kv4.2 eliminates Ito,f and results in electrical and molecular remodeling, with no evidence of ventricular hypertrophy or myocardial dysfunction. Circ Res 2005;97:1342–50. [DOI] [PubMed] [Google Scholar]
- 131.Guo W, Xu H, London B and Nerbonne JM. Molecular basis of transient outward K+ current diversity in mouse ventricular myocytes. J Physiol 1999;521 Pt 3:587–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Cordeiro JM, Nesterenko VV, Sicouri S, Goodrow RJ Jr., Treat JA, Desai M, Wu Y, Doss MX, Antzelevitch C and Di Diego JM. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Argenziano M, Lambers E, Hong L, Sridhar A, Zhang M, Chalazan B, Menon A, Savio-Galimberti E, Wu JC, Rehman J and Darbar D. Electrophysiologic characterization of calcium handling in human induced pluripotent stem cell-derived atrial cardiomyocytes. Stem Cell Reports 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Marczenke M, Piccini I, Mengarelli I, Fell J, Ropke A, Seebohm G, Verkerk AO and Greber B. Cardiac subtype-specific modeling of Kv1.5 ion channel deficiency using human pluripotent stem cells. Front Physiol 2017;8:469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.London B, Guo W, Pan X, Lee JS, Shusterman V, Rocco CJ, Logothetis DA, Nerbonne JM and Hill JA. Targeted replacement of Kv1.5 in the mouse leads to loss of the 4-aminopyridine-sensitive component of IK,slow and resistance to drug-induced QT prolongation. Circ Res 2001;88:940–6. [DOI] [PubMed] [Google Scholar]
- 136.Salama G and London B. Mouse models of Long QT syndrome. J Physiol 2007;578:43–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Lee MP, Ravenel JD, Hu RJ, Lustig LR, Tomaselli G, Berger RD, Brandenburg SA, Litzi TJ, Bunton TE, Limb C, Francis H, Gorelikow M, Gu H, Washington K, Argani P, Goldenring JR, Coffey RJ and Feinberg AP. Targeted disruption of the KvLQT1 gene causes deafness and gastric hyperplasia in mice. J Clin Invest 2000;106:1447–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Zaritsky JJ, Redell JB, Tempel BL and Schwarz TL. The consequences of disrupting cardiac inwardly rectifying K+ current (IK1) as revealed by the targeted deletion of the murine Kir2.1 and Kir2.2 genes. J Physiol 2001;533:697–710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Wickman K, Nemec J, Gendler SJ and Clapham DE. Abnormal heart rate regulation in GIRK4 knockout mice. Neuron 1998;20:103–14. [DOI] [PubMed] [Google Scholar]
- 140.Krapivinsky G, Gordon EA, Wickman K, Velimirovic B, Krapivinsky L and Clapham DE. The G-protein-gated atrial K+ channel IKACh is a heteromultimer of two inwardly rectifying K+-channel proteins. Nature 1995;374:135–41. [DOI] [PubMed] [Google Scholar]
- 141.Ma D, Wei H, Lu J, Huang D, Liu Z, Loh LJ, Islam O, Liew R, Shim W and Cook SA. 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 Research & Therapy 2015;6:39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Sala L, Yu Z, Ward-van Oostwaard D, van Veldhoven JP, Moretti A, Laugwitz KL, Mummery CL, AP IJ and Bellin M. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Malan D, Zhang M, Stallmeyer B, Muller J, Fleischmann BK, Schulze-Bahr E, Sasse P and Greber B. Human iPS cell model of type 3 Long QT syndrome recapitulates drug-based phenotype correction. Basic Res Cardiol 2016;111:14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Rocchetti M, Sala L, Dreizehnter L, Crotti L, Sinnecker D, Mura M, Pane LS, Altomare C, Torre E, Mostacciuolo G, Severi S, Porta A, De Ferrari GM, George AL Jr., Schwartz PJ, Gnecchi M, Moretti A and Zaza A. Elucidating arrhythmogenic mechanisms of Long-QT syndrome CALM1-F142L mutation in patient-specific induced pluripotent stem cell-derived cardiomyocytes. Cardiovasc Res 2017;113:531–541. [DOI] [PubMed] [Google Scholar]
- 145.Yamamoto Y, Makiyama T, Harita T, Sasaki K, Wuriyanghai Y, Hayano M, Nishiuchi S, Kohjitani H, Hirose S, Chen J, Yokoi F, Ishikawa T, Ohno S, Chonabayashi K, Motomura H, Yoshida Y, Horie M, Makita N and Kimura T. Allele-specific ablation rescues electrophysiological abnormalities in a human iPS cell model of Long-QT syndrome with a CALM2 mutation. Hum Mol Genet 2017;26:1670–1677. [DOI] [PubMed] [Google Scholar]
- 146.Limpitikul WB, Dick IE, Tester DJ, Boczek NJ, Limphong P, Yang W, Choi MH, Babich J, DiSilvestre D, Kanter RJ, Tomaselli GF, Ackerman MJ and Yue DT. A precision medicine approach to the rescue of function on malignant calmodulinopathic Long-QT syndrome. Circ Res 2017;120:39–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Cerrone M, Lin X, Zhang M, Agullo-Pascual E, Pfenniger A, Chkourko Gusky H, Novelli V, Kim C, Tirasawadichai T, Judge DP, Rothenberg E, Chen HS, Napolitano C, Priori SG and Delmar M. Missense mutations in plakophilin-2 cause sodium current deficit and associate with a Brugada Syndrome phenotype. Circulation 2014;129:1092–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Novak A, Barad L, Lorber A, Gherghiceanu M, Reiter I, Eisen B, Eldor L, Itskovitz-Eldor J, Eldar M, Arad M and Binah O. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Preininger MK, Jha R, Maxwell JT, Wu Q, Singh M, Wang B, Dalal A, McEachin ZT, Rossoll W, Hales CM, Fischbach PS, Wagner MB and Xu C. A human pluripotent stem cell model of Catecholaminergic Polymorphic Ventricular Tachycardia recapitulates patient-specific drug responses. Disease Models & Mechanisms 2016;9:927–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Sasaki K, Makiyama T, Yoshida Y, Wuriyanghai Y, Kamakura T, Nishiuchi S, Hayano M, Harita T, Yamamoto Y, Kohjitani H, Hirose S, Chen J, Kawamura M, Ohno S, Itoh H, Takeuchi A, Matsuoka S, Miura M, Sumitomo N, Horie M, Yamanaka S and Kimura T. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Novak A, Barad L, Zeevi-Levin N, Shick R, Shtrichman R, Lorber A, Itskovitz-Eldor J and Binah O. Cardiomyocytes generated from CPVTD307H patients are arrhythmogenic in response to β-adrenergic stimulation. J Cell Mol Med 2012;16:468–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Maizels L, Huber I, Arbel G, Tijsen AJ, Gepstein A, Khoury A and Gepstein L. Patient-specific drug screening using a human induced pluripotent stem cell model of Catecholaminergic Polymorphic Ventricular Tachycardia type 2. Circ Arrhythm Electrophysiol 2017;10. [DOI] [PubMed] [Google Scholar]
