133
Views
7
CrossRef citations to date
0
Altmetric
Review

Gene mutations in cardiac arrhythmias: a review of recent evidence in ion channelopathies

, , , , &
Pages 1-13 | Published online: 18 Jan 2013

Abstract

Over the past 15 years, molecular genetic studies have linked gene mutations to many inherited arrhythmogenic disorders, in particular, “ion channelopathies”, in which mutations in genes encode functional units of ion channels and/or their transporter-associated proteins in patients without primary cardiac structural abnormalities. These disorders are exemplified by congenital long QT syndrome (LQTS), short QT syndrome, Brugada syndrome (BrS) and catecholaminergic polymorphic ventricular tachycardia (CPVT). Functional and pathophysiological studies have led to better understanding of the clinical spectrum, ion channel structures and cellular electrophysiology involving dynamics of intracellular calcium cycling in many subtypes of these disorders and more importantly, development of potentially more effective pharmacological agents and even curative gene therapy. In this review, we have summarized (1) the significance of unveiling mutations in genes encoding transporter-associated proteins as the cause of congenital LQTS, (2) the technique of catheter ablation applied at the right ventricular outflow tract may be curative for severely symptomatic BrS, (3) mutations with channel function modulated by protein Kinase A-dependent phosphorylation can be the culprit of CPVT mimicry in Andersen-Tawil syndrome (LQT7), (4) ablation of the ion channel anchoring protein may prevent arrhythmogenesis in Timothy syndrome (LQT8), (5) altered intracellular Ca2+ cycling can be the basis of effective targeted pharmacotherapy in CPVT, and (6) the technology of induced pluripotent stem cells is a promising diagnostic and research tool as it has become a new paradigm for pathophysiological study of patient- and disease-specific cells aimed at screening new drugs and eventual clinical application of gene therapy. Lastly, we have discussed (7) genotype-phenotype correlation in relation to risk stratification of patients with congenital LQTS in clinical practice.

Introduction

Applying the technology of DNA sequencing,Citation1 Curran et alCitation2 and Wang et alCitation3,Citation4 noted that mutations in the KCNQ1, KCNH2, and SCN5A genes, encoding the α-subunit of ion channels that conduct potassium delayed-rectifier currents (IKs and IKr) and the sodium current (INa), respectively, could be responsible for three subtypes (LQT1-3) of congenital long-QT syndrome (LQTS). These seminal works have inspired many investigators and prompted extensive basic research, leading to subsequent identification of various gene mutations causing cardiac arrhythmias referred to as “inherited arrhythmogenic disorders” (). These arrhythmogenic disorders with mutations in genes encoding developmental components of cardiac structures produce diseases associated with a structurally abnormal heart, exemplified by hypertrophic cardiomyopathy, arrhythmogenic right ventricular dysplasia (cardiomyopathy), and dilated cardiomyopathy, whereas those with mutations in genes encoding functional units of ion channels and/or their transporter-associated proteins produce diseases associated with a structurally normal heart, such as congenital LQTS, short-QT syndrome, Brugada syndrome (BrS), and catecholaminergic polymorphic ventricular tachycardia (CPVT), known as “ion channelopathies” (). All these arrhythmogenic disorders are usually genetically heterogeneous, and their clinical courses are underscored by variable clinical expressivity ranging from being asymptomatic to episodic syncope, abortive cardiac arrest, and sudden cardiac death (SCD).

Table 1 Inherited arrhythmogenic disorders

Table 2 Congenital long-QT syndrome (LQTS)

Table 3 Brugada syndrome (BrS)

Table 4 Catecholaminergic polymorphic ventricular tachycardia (CPVT)

Table 5 Short-QT syndrome (SQTS)

To confirm that a specific gene mutation is linked to cardiac arrhythmias, functional studies to illustrate consequences of the mutation are required. These functional studies usually use heterologous expression systems, primarily Xenopus oocytes, human embryonic kidney (HEK) cells, and Chinese hamster ovary cells.Citation5 Electrophysiological effects of the mutant ion channel are then compared to those of the wild-type counterpart. However, in order to include important constituents of the macromolecular complex of an ion channel in the complex living environment so as to reproduce the exact molecular and electrophysiological phenotype, it is often necessary to generate a transgenic mouse model carrying the specific gene mutation.Citation6

Through collaborative endeavors between clinical and basic science researchers over the past 15 years, we now have better understanding of the clinical spectrum, molecular genetics, ion-channel structures, and cellular electrophysiology relating to these inherited arrhythmogenic disorders. Taking ion channelopathies as an example, the discovery of genetic defects involving the L-type Ca2+ channel (Cav1.2), ryanodine receptor (RyR2), and calsequestrin has provided incontrovertible evidence linking cardiac arrhythmias to abnormal intracellular Ca2+ cycling. As such, research and development of targeted pharmacotherapy and/or gene therapy can be contemplated. In this review, we focus on unique findings that have been recently described; in particular those pertaining to the underlying pathophysiology for better diagnosis and development of new targeted treatment modalities in certain ion channelopathies.

Mutations in genes encoding transporter-associated proteins as the cause of congenital LQTS

Congenital LQTS is an inherited disorder of delayed cardiac repolarization, electrocardiographically reflected as QT-interval prolongation, with a propensity to lethal ventricular tachyarrhythmias causing recurrent syncope, seizures, and SCD.Citation7 Molecular genetic studies have demonstrated that three subtypes of congenital LQTS – LQT1, LQT2, and LQT3 – constitute 65%–75%, while each in the remaining susceptibility genes accounts for <1%, leaving approximately 20%–25% of patients with a yet-to-be-determined genotype.Citation8

While congenital LQTS is predominantly caused by mutations in genes that encode ion channels, the discovery that transporter-associated proteins could also be the culprit is of diagnostic and therapeutic importance.Citation8 Following identification of mutations in ankyrin-B (ankyrin 2) as the cause for LQT4,Citation9 those in caveolin-3 (CAV3), yotiao (AKAP9), and α-1-syntrophin have been found to be responsible for LQT9, LQT11, and LQT12, respectively.Citation8

LQT4

Ankyrin-B, a membrane adapter, binds to multiple proteins that can directly or indirectly contribute to cardiac electrical activity.Citation9 These multiple proteins include Na+ channels (Nav1.5), Cl/HCO3 exchanger, Na+/K+ adenosine triphosphatase (ATPase), Kir6.2, and Ca2+ release channels such as those mediated by RyR2 or inositol 1,4,5-trisphosphate (IP3) receptors. LQT4 is caused by mutations of ankyrin-B that result in loss of function.Citation9 As has been shown in a heterozygous mouse model of null ankyrin-B, loss of ankyrin-B function disrupts the cellular organization of these ankyrin-B-binding proteins, leading to intracellular Ca2+ overload and store-overload-induced Ca2+ release (SOICR) from the sarcoplasmic reticulum (SR), triggering afterdepolarizations in response to catecholaminergic stimulation.Citation9 Clinically, LQT4 patients manifest mild QT prolongation, catecholamine-sensitive polymorphic ventricular tachycardia (VT), and SCD, and may also present with sinus bradycardia, atrial fibrillation and atrioventricular conduction defects.Citation8

LQT9

Caveolae are flask-shaped invaginations of the plasma membrane involved in vesicular trafficking and signal-transduction pathways, in which caveolins are the principal proteins. Cardiac ion channels that are specifically localized to caveolae include Nav1.5, the voltage-dependent K+ channel (Kv1.5), Na+/Ca2+ exchanger, and Cav1.2; in addition, a variety of other signaling molecules, including β2-adrenergic receptors and associated proteins of the G-protein/adenylyl cyclase/protein kinase A (PKA) pathway, have been found in caveolae. LQT9 is caused by mutations of CAV3, with resultant interference of signaling between caveolae-enriched ion channels and a network of integrated signaling molecules.Citation8,Citation10 Of interest, in CAV3 knockout mice, progressive cardiac hypertrophy/cardiomyopathy is observed,Citation11 and in HEK293 cell lines mutant CAV3 exhibits a two- to threefold increase in late Na+ current similar to that seen in LQT3-associated SCN5A mutations.Citation10 Moreover, CAV3 mutations are also linked to sudden infant death syndrome.Citation12 However, because of multiple ion-channel involvement, the exact mechanisms by which CAV3 mutations exert arrhythmogenesis remain to be determined.Citation8

LQT11

Yotiao (AKAP9) is a member of a family of PKA-anchoring proteins. LQT11 is due to mutations of AKAP9 that is required to be assembled with the α-subunit of the IKs channel (KvLQT1) during β2-adrenergic activation (BAS); AKAP9-associated PKA activity is also connected with activities of RyR2 and Cav1.2. Hence, mutations of this PKA-anchoring protein interfere with PKA-dependent phosphorylation, thereby markedly inhibiting responses of at least three ion channels, ie, KvLQT1, RyR2, and Cav1.2, to cyclic adenosine monophosphate (cAMP).Citation13 Clinically, LQT11 is likened to LQT1 and LQT5.Citation8

LQT12

α-1-Syntrophin belongs to a family of cytoplasmic adapter proteins, linking many membrane proteins and signaling molecules, including plasma membrane calcium ATPase and neuronal nitric oxide synthase (nNOS); nNOS can directly affect persistent (or late) INa, and variants in the nNOS regulatory protein NOS1AP (CAPON) can regulate and moderate the QT interval.Citation14,Citation15 LQT12 is related to mutations of α-1-syntrophin, which cause a decrease in its binding activity for plasma membrane calcium ATPase and nNOS along with an increase in Nav1.5 nitrosylation; the resultant persistent INa creates a clinical phenotype akin to LQT3.Citation8

Mutations with channel function modulated by PKA-dependent phosphorylation as the cause of CPVT mimicry in LQT7 (Andersen–Tawil syndrome)

Andersen–Tawil syndrome (ATS) (LQT7) is an autosomal dominant disorder clinically characterized by ventricular arrhythmias associated with long QT interval, periodic paralysis, and multiple skeletal dysmorphic features.Citation16Citation18 More than 20 mutations have been identified in the KCNJ2 gene, which encodes the α-subunit of the inward-rectifier potassium channel (Kir2.1).Citation16Citation18

Electrophysiologically, the corresponding Kir2.1 current (IK1) participates in the terminal phase of action potential (AP) repolarization responsible for setting the resting membrane potential in mammalian heart cells.Citation19 Of the mutations identified in distinct regions of KCNJ2 in ATS patients, a dominant negative effect of the mutant allele can be found in both trafficking and non-trafficking defective channels, contributing to loss of Kir2.1 function.Citation20

Although patients with ATS have a high prevalence (approximately 64%) of ventricular arrhythmias,Citation16,Citation17 these arrhythmias are usually not related to exercise and/or emotional distress. The relatively benign clinical course is attributed to lack of significant enhancement of transmural dispersion of repolarization produced by IK1 down-regulation.Citation21 However, a subset of ATS patients, especially females, are prone to exercise- and/or stress-induced bidirectional VT and/or polymorphic VT, mimicking CPVT,Citation17,Citation20,Citation22,Citation23 which is a malignant form of inherited arrhythmogenic disorder, affecting the young with otherwise a structurally normal heart and QT interval.Citation24 However, mutations of CPVT are primarily (approximately 60% of patients) located in the gene encoding RyR2 (CPVT1, autosomal dominant) or calsequestrin (CPVT2, autosomal recessive) in SR.

Molecular genetic studies have identified several mutations in the KCNJ2 gene, such as G144D, R67W, R82W, R260P, T305S, and V227F,Citation20,Citation22,Citation23 that may predispose ATS patients to catecholamine-sensitive ventricular tachyarrhythmias, regarded as CPVT mimicry or CPVT3.Citation17,Citation20,Citation22,Citation23 Of interest, among these mutations, heterozygous Kir2.1-V227F can function normally like wild-type channels.Citation23 However, when stimulated by cAMP-dependent PKA, the Kir2.1-V227F mutant can significantly downregulate IK1. This unique biophysical property of latent loss of function modulated by PKA-dependent phosphorylation of the IK1 channel provides a rather specific mechanism by which a subset of ATS patients may manifest ventricular arrhythmias under high adrenergic tone, simulating the phenotype of CPVT.

Ablation of the ion channel-anchoring protein may prevent arrhythmogenesis in LQT8 (Timothy syndrome)

Timothy syndrome (TS), a subtype (LQT8) of congenital LQTS, clinically manifests multisystem involvement (eg, syndactyly, dysmorphic facial features, autism, etc).Citation25 In association with marked QT-interval prolongation, cardiac arrhythmias are the most serious aspect of its clinical features. These include bradycardia, atrioventricular block, polymorphic VT, and ventricular fibrillation (VF). Because of high prevalence (71%) and severity of VT/VF, the majority of TS patients seldom survive beyond 3 years of age.Citation25,Citation26 Notably, the mode of induction of cardiac events (eg, syncope, cyanotic spell, and SCD) is usually triggered by an increase in sympathetic tone.

Molecular genetic studies have revealed that TS patients have mutations with a Gly to Arg substitution at position 406 (G406R) of Cav1.2 in the CACNA1C gene, which results in “near-complete” elimination of voltage-dependent inactivation (VDI) of Cav1.2 (gain of function).Citation25,Citation26 Electrophysiological studies have illustrated that G406R-mutated Cav1.2 is inactivated at a slow rateCitation25 alongside a high probability of undergoing coordinated openings and closings (coupled gating).Citation27

It is suggested that G436R-mutated Cav1.2 (corresponding to G406R in humans) in rabbit ventricular myocytes creates a new phosphorylation site (Ser-439) for Ca2+/calmodulin-dependent protein kinase II (CaMKII), and phosphorylation of this site is correlated with an increased open probability of gating of the mutant cardiac Cav1.2.Citation28 Unfortunately, others have demonstrated that phosphorylation by CaMKII is not necessary for the slower rate of Cav1.2 inactivation.Citation27,Citation29 Moreover, in exogenously expressed (HEK293 cells) rabbit cardiac Cav1.2, it is shown that the G436R mutation displays multiple altered gating mechanisms of Cav1.2.Citation29 Besides affecting the open-state VDI, it significantly slows the channel activation at voltages less than 10 mV and disturbs its deactivation across, the latter of which also favors a high open-probability state of Cav1.2 (also arrhythmogenic).Citation30 Surprisingly, the closed state of VDI remains intact, suggesting that two different gating mechanisms are involved in the open and closed states of VDI. Collectively, these findings imply that impaired open state of VDI and slowed deactivation of Cav1.2 can synergistically increase AP duration and cause early and delayed afterdepolarizations (EADs and DADs) and triggered activity (TA) due to Ca2+ overload.Citation25,Citation31 Experimentally, roscovitine, an open-state VDI enhancer, can revert functional defects of the mutated Cav1.2.Citation29

Recently, in ventricular myocytes obtained from a transgenic mouse model that expresses Cav1.2-LQT8, the mutated Cav1.2 is shown to be abnormally coupled to AKAP150, a specific protein kinase that phosphatases to regions near Cav1.2.Citation32 With the formation of this aberrant ion channel-anchoring protein complex, AKAP150 functionally serves like a channel subunit that stabilizes the open conformation and augments the probability of coordinated openings, enhancing Cav1.2-LQT8 currents. Of note, ablation of AKAP150 restores normal gating (inactivation) of Cav1.2-LQT8, thereby preventing EADs, DADs, and torsade des pointes. Hence, it appears that AKAP150 is required for increased Cav1.2 activity and coupled gating seen in LQT8 ventricular myocytes. Since AKAP150 also binds to the carboxyl tail of Cav1.2 via leucine-zipper motifs, it is postulated that AKAP150 binds to the C-terminal tail of Cav1.2-LQT8, thereby facilitating longer channel openings and interaction between multiple Cav1.2-LQT8 channels to increase the frequency of “coupled gating”; consequently, greater Ca2+ influx leads to intracellular Ca2+ overload. This unique AKAP150-dependent change in Cav1.2-LQT8 gating may represent an important mechanism of arrhythmogenesis in LQT8.

Altered intracellular Ca2+ cycling and targeted pharmacotherapy for CPVT

CPVT is an inherited arrhythmogenic disorder characterized by episodic syncope occurring during physical activity or acute emotional distress in individuals with otherwise normal hearts and electrocardiograms (ECGs). Notably, episodes of syncope usually coincide with documented bidirectional or polymorphic VT that may degenerate into VF. By the age of 40 years, cardiac events occur in 80% of patients, and the mortality rate can be as high as 30%–50%.Citation24 In 60% of CPVT patients, molecular genetic studies have revealed that mutations are mostly located in the gene encoding RyR2 (autosomal dominant) and rarely in the calsequestrin gene (CASQ2, autosomal recessive).

Although more than 70 RyR2 mutations have been identified in CPVT, mechanisms by which RyR2 mutations alter the properties of RyR2 receptors, thereby generating ventricular tachyarrhythmias, remain highly debatable. The general consensus is that RyR2 mutations lead to abnormally augmented Ca2+ release, resulting in DADs and DAD-mediated TA.Citation24 In an RyRR4496C knock-in mouse model (equivalent to human R4497C mutation), it has been demonstrated that (1) RyR2+/RyRR4496C mice are prone to bidirectional VT after catecholaminergic stimulation and that VT can degenerate into VF;Citation33 (2) DADs and DAD-mediated TA induced by isoproterenol can be abolished by ryanodine but not by K201 (an agent that enhances binding of FKBP12.6 to RyR2);Citation34 (3) there is an increase in Ca2+sensitivity of RyR2 conjoined with a lowered SOICR threshold;Citation35 (4) there is elevation of SR Ca2+ load in the absence of BAS; ouabain, an Na+/K+-ATPase antagonist, can enhance the propensity to DADs and TA, and that JTV-519, a RyR2 stabilizer, can effectively reduce these arrhythmias;Citation36 and (5) Purkinje cells of RyR2+/RyRR4496C mice are more vulnerable than the ventricular myocyte to altered intracellular Ca2+ cycling leading to EADs, DADs, and TA, which can be greatly exacerbated by BAS.Citation37 Taken together, these latter findings suggest that medications like digitalis are contraindicated and that Purkinje cells are critical contributors to arrhythmogenesis in CPVT similar to many other clinical situations, eg, after myocardial infarction, in the presence of dilated cardiomyopathy or idiopathic VF, and with other inherited ion channelopathies, including BrS and LQTS.Citation38

Missense mutations of CASQ2 have been shown to alter the Ca2+-binding capacity and/or the Ca2+-dependent polymerization of calsequestrin. It seems that mechanistically different defects in CASQ2 all lead to increased diastolic SR Ca2+ release and manifest a similar CPVT phenotype. It is illustrated that expression of truncated CASQ2DEL (G112 + 15X) and CASQ2R33Q mutations can alter myocyte Ca2+ signaling through two distinctly different mechanisms:Citation39 CASQ2DEL mutation disrupts the polymerization of calsequestrin required for high-capacity Ca2+ binding, whereas CASQ2R33Q mutation compromises the ability of calsequestrin to control the RyR2 channel activity. Consequently, with CASQ2DEL mutation, local Ca2+ release terminates at the same free luminal Ca2+ concentrations as those of the wild type. In contrast, with CASQ2R33Q mutation, compromised interactions between RyR2 and calsequestrin markedly lower the threshold of luminal Ca2+ concentrations for the termination of SR Ca2+ release. These findings suggest that calsequestrin possesses dual function by providing a local source of releasable Ca2+ and by exerting effects on luminal Ca2+-dependent RyR2 gating (ie, via protein–protein interactions or serving as a luminal Ca2+ sensor).Citation40 It should be noted, however, calsequestrin-null mice are viable and display normal SR Ca2+ release and contractile function under basal conditions while still maintaining functional SR Ca2+ storage. Despite lack of calsequestrin, calsequestrin-null mice exhibit an increase in diastolic SR Ca2+ leak without apparent upregulation of other Ca2+-binding proteins and are susceptible to CPVT-related ventricular arrhythmias, especially under BAS. Thus, although calsequestrin modulates SR Ca2+ release, it is not essential for luminal Ca2+ regulation of RyR2, nor is it required for SOICR termination in cardiac myocytes. Currently, the consensus is that the luminal Ca2+ sensor lies within the RyR2 complex, likely to be regulated by a number of accessory proteins, such as calsequestrin, triadin, junctin, FKBP12.6, calmodulin, kinases, and others associated with the RyR2 complex.Citation41

It has been shown that heterozygous CASQK206N missense mutation can generate an additional N-glycosylation site, which can result in the recombinant protein having a higher molecular weight with reduced Ca2+-binding capacity exhibiting an altered aggregation state; consequently, despite the interaction of CASQK206N with triadin and the protein level of RyR2 remaining unchanged, the response of the CASQK206N myocyte to caffeine is impaired, as there is a lower SR Ca2+ load alongside a higher opening state of RyR2. Therefore, the CASQK206N myocyte displays a higher rate of spontaneous SR Ca2+ release under basal conditions and under BAS compared to the wild-type counterpart.Citation42

Clinical observations have shown that β-blockers provide only incomplete protection, and other antiarrhythmic agents, such as quinidine, verapamil, and amiodarone, are either ineffective or only partially efficacious in CPVT.Citation43 Some patients need to resort to implantable cardioverter/defibrillators, left cardiac sympathetic denervation, and even to cardiac transplantation.Citation24 Based on better understanding of the pathophysiology of CVPT, targeted pharmacotherapy appears to be very promising.Citation43,Citation44 Recently, flecainide therapy has been advocated to be efficacious in suppressing ventricular arrhythmias in CPVT as well as certain ATS patients.Citation20,Citation45Citation47 Flecainide directly inhibits RyR2 by perturbing RyR2-mediated premature Ca2+ release, thereby suppressing arrhythmogenic Ca2+ waves and effectively suppressing ventricular arrhythmias in CASQ2 knockout mice.Citation45,Citation47 However, flecainide is a potent Na+-channel inhibitor that also blocks IKr, and Ca2+-independent transient outward K+ current.Citation48 Being a potent Na+-channel inhibitor, it decreases the probability for DADs to reach the threshold for AP generation.Citation49 Moreover, it selectively increases the density of functional Kir2.1 via reducing Kir2.1 polyamine blockade in the human ventricular myocyte.Citation50 Although the exact mechanism whereby flecainide exerts its antiarrhythmic action is complex, the therapeutic efficacy of flecainide in CPVT patients has been clinically confirmed.Citation46

For targeted pharmacotherapy, simulated electropharmacological testing suggests that combination therapy with a Ca2+-channel blocker and an Iup (SR Ca2+-ATPase current) inhibitor is more efficacious than a Ca2+-channel blocker alone in suppressing CPVT-associated arrhythmias.Citation31 Furthermore, in RyRR4496C+/− knock-in mice, CaMKII overexpression enhances RyR2 phosphorylation and increases the propensity to TA; KN93, a CaMKII inhibitor, prevents TA via counteracting effects of BAS.Citation51 Thus, tackling a specific site in intracellular Ca2+ cycling appears logical and promising for suppressing cardiac arrhythmias associated with CPVT.

Catheter ablation of the right ventricular outflow tract may be curative for severely symptomatic BrS

BrS is an inherited arrhythmogenic disorder that exhibits ECG ST-segment elevation with a negative T-wave in the right precordial leads (V1–V3), predisposing to VF and SCD in otherwise healthy individuals.Citation52 BrS is recognized as an important cause of SCD in young men, especially in Southeast Asia.Citation53 Notably, the Brugada ECG pattern is dynamic and functional; it is more pronounced at night or at rest and may be enhanced by an increase in vagal tone, such as at night or after a big meal, and may become apparent during a febrile illness. Of note, sodium-channel blockers (class 1A and 1C) and acetylcholine may enhance or provoke the Brugada ECG pattern, facilitating the diagnosis of the syndrome.Citation52

Genetically, mutations in eleven genes have been identified in patients with BrS. Most of these mutations are located in SCN5A, encoding the α-subunit of Nav1.5,Citation54 in SCN1BCitation5 and SCN3B,Citation55 encoding the β-subunits of the cardiac sodium channel, or in GPD1LCitation56 and MOG1,Citation57 involving the trafficking of Nav1.5 to the cell membrane; all of these mutations result in loss of function of Nav1.5, thereby reducing INa. Other mutations are located in CACNA1C, CACNB2bCitation58 and CACNA2D1,Citation59 which encode α1-, β2b-, and α2δ1-subunits of Cav1.2, respectively, with resultant reduction of the L-type calcium current (ICa,L) and in KCNE3, encoding MiRP2, α-, and β-subunits of several potassium channels,Citation60 and in KCNJ8, encoding the ATP-sensitive potassium channel.Citation61 Overall, mutations in SCN5A account for approximately 20% and those in other genes 10%, leaving no definitive genetic defects in 70% of BrS patients.Citation52 Although the heart appears grossly normal, right ventricular structural abnormalities, such as fibrofatty degenerative changes, lymphocytic myocarditis, and atypical cardiomyopathic alterations in the ventricular outflow tract (RVOT) are not infrequently observed. Clinical features overlapping with the right ventricular dysplasia/cardiomyopathy, another form of inherited arrhythmogenic disorder with distinctly abnormal right ventricle (), have been reported. Differential diagnosis between the two may at times present as a clinical dilemma.Citation52

Despite the discovery of various genetic mutations, the pathophysiology underpinning BrS remains elusive. There has been much debate about whether it is a depolarization disorder (ie, conduction delay or failure) or repolarization disorder (ie, transmural repolarization gradient through the right ventricular wall).Citation52 More significantly, the theory of loss of function of Nav1.5 or other ion channels as the basis has been questioned. This is because (1) there is often lack of a hereditary (familial) pattern in most index BrS patients, (2) sodium-channel blockers do not usually provoke the Brugada ECG pattern in normal subjects, and (3) the ECG pattern can also be seen or provoked in patients with structural heart diseases. Hence, it has been suggested that BrS is a multifactorial disorder with various causes of right ventricular conduction disturbances, of which “current-to-load mismatch” is the common denominator; different etiologies may require different approaches to management of cardiac arrhythmias in patients with BrS.Citation52

Of therapeutic relevance are two recent reports. In nine BrS patients treated with implantation of cardioverter/defibrillators because of recurrent VF, Nademanee et alCitation62 noted that all these nine patients had typical Brugada ECG pattern and electrically inducible VT/VF. Electrophysiological mapping revealed the presence of low-voltage, fractionated late potentials spanning beyond the electrocardiographic QRS complex, clustering exclusively in the RVOT anterior aspect during sinus rhythm. Because the magnitude of these electrogram abnormalities over the epicardium was much greater than those recorded in the endocardium, the researchers applied radiofrequency catheter ablation at these sites; as a result, the Brugada ECG pattern became normalized in eight patients (89%) and VT/VF rendered electrically noninducible in seven patients (78%). All patients were off medication except for one (amiodarone therapy continued), and there was no recurrence of VT/VF observed in any patients during a follow-up period of 20 ± 6 months. The study has demonstrated that the BrS ECG pattern can be accounted for by delayed depolarization over the anterior aspect of RVOT. Moreover, in ten BrS patients, Sunsaneewitayakul et alCitation63 noted a late activation zone recorded as electrical activity occurring from the J point to +60 (J+60) millisecond interval of V1 or V2 ECG lead in the RVOT endocardium during an isopotential mapping. Radiofrequency endocardial catheter ablation of the late-activation zone modified the Brugada ECG pattern in three (75%) of four patients with VF storm. During a follow-up period of 12–30 months, there was no recurrence of VF in any of these four patients (100%). It seems that the endocardium of RVOT can be considered as an alternative site for radiofrequency catheter ablation in the treatment of recurrent VF in BrS.

Technology of induced pluripotent stem cells as a promising diagnostic and research tool in inherited arrhythmogenic disorders

Electrophysiological characteristics of the mouse and human hearts are distinctly different in the distribution of ion channels, AP morphology, and heart rate.Citation64 A mouse model may not always demonstrate the same phenotype as humans. Therefore, the use of induced pluripotent stem cell (iPSC) technology to study inherited arrhythmogenic disorders has become a new paradigm, as it can provide direct evidence linking genetic mutations to cardiac arrhythmias in humans.Citation65,Citation66 The iPSC technology is to reprogram somatic cells into pluripotent stem cells via transduction of a key set of transcriptional factors (eg, c-Myc, Oct3/4, Sox2, and Klf4 or Oct3/4, Sox2, Nanog, and Lin28). These iPSCs can then differentiate into the cell types of the three germ layers, and when generated from human somatic cells, the human iPSCs resemble those of human embryonic stem cells (ESCs) in morphology, gene expression, and epigenetic status of pluripotent cell-specific genes.Citation67 Human iPSCs can differentiate to functional cardiac myocytes (CMs),Citation68 and similar to human ESCs these human iPSCs possess a capacity of differentiating into nodal-, atrial-, and ventricular-like cells, each with specific AP characteristics; both iPSC- and ESC-derived CMs exhibit typical responsiveness to BAS. Consequently, the possibility of creating patient- and disease-specific human iPSC-derived CMs aimed at individualized diagnostic and pharmacological testing has become a reality. To date, iPSC-derived CMs have been assessed in a limited number of patients with ion channelopathies.Citation65,Citation66

LQT1

In iPSC-derived CMs produced from two patients with KCNQ1R190Q mutation,Citation69 functional study revealed a dominant negative trafficking defect associated with a 70%–80% reduction in IKs alongside altered activation and deactivation properties of KvLQT1. Compared to control, these iPSC-derived CMs with KCNQ1R190Q showed increased propensity to EADs in response to isoproterenol, which could be attenuated by propranolol.Citation69 These findings are in accord with clinical observations that cardiac events are usually triggered by enhanced sympathetic tone and that β-blockers are beneficial in LQT1.Citation70

LQT2

In iPSC-derived CMs produced from a patient with A614V missense in KCNH2,Citation71 there was prolongation of AP duration, which could be ascribed to significant reduction of IKr. These iPSC-derived CMs with KCNH2A614 also exhibited marked arrhythmogenicity characterized by EADs and EAD-mediated TA. K+-channel blockers could aggravate, whereas Ca2+-channel blockers, IK(ATP)-channel openers, and late Na+-channel blockers could ameliorate the disease phenotype.Citation71 In another patient with KCNH2G1681A, iPSC-derived CMsCitation72 also showed prolonged AP duration. Exposure to E4031 (an IKr blocker) provoked EADs. Moreover, isoprenaline facilitated EADs; this latter effect could be reversed by β-blockers.Citation72

In an asymptomatic patient with KCNH2R176W whose sister and father had died suddenly at an early age, iPSC-derived CMs with such a mutation showed prolonged AP duration and significantly reduced IKr density.Citation73 These iPSC-derived CMs with KCNH2R176W were sensitive to potentially arrhythmogenic drugs such as sotalol, and exhibited a pronounced inverse correlation between the beating rate and repolarization time. Hence, iPSC-derived CMs may be used to provide a direct way for identifying asymptomatic patients at risk of proarrhythmia.

LQT3

iPSC-derived CMs were produced from a mouse model with a human LQT3 mutation of Nav1.5.Citation74 These iPSC-derived CMs showed biophysical effects of the mutation on Nav1.5, ie, faster recovery from inactivation and larger late currents than those of the wild-type control, prolonged AP duration, and development of EADs at low pacing rates, consistent with classic features of LQT3.

In a mouse model, iPSC-derived CMs carrying SCN5A1798insD/+ mutationCitation75 showed a decrease in INa density with a large persistent INa and reduced AP upstroke velocity coupled with prolonged AP duration. These electrophysiological characteristics were subsequently observed in iPSC-derived CMs generated from a patient with equivalent SCN5A1795insD/+ mutation. Of note, this latter mutation clinically gave rise to a phenotype of LQT3 and BrS with conduction defects due to both gain- and loss-of-function effects on Nav1.5, respectively.Citation76

LQT8

Human iPSC-derived CMs generated from two patients with TS (LQT8) showed irregular contractions, excessive Ca2+ influx, and markedly prolonged AP duration (three times longer than that of the wild-type control),Citation77 irregular electrical activity and abnormal intracellular Ca2+ transients associated with delayed inactivation of Cav1.2. Roscovitine, a compound that increases VDI of Cav1.2, was able to revert delayed inactivation of Cav1.2 and restored irregular intracellular Ca2+ transients.

CPVT

iPSC-derived CMs created from a patient with RyR2F2483I heterozygous mutation showed high amplitude and long duration of spontaneous Ca2+ release at basal state.Citation78 In response to catecholaminergic stimulation, these iPSC-derived CMs were prone to DADs, which paradoxically could be abolished by increasing the cytosolic cAMP level with forskolin. iPSC-derived CMs were also produced from a CPVT patient with RyR2M4109R heterozygous mutation. In the presence of isoproterenol or forskolin, these CPVT iPSC-derived CMs readily developed DADs and DAD-mediated TA related to SOICR, which could be improved with β-blockers and could be eliminated by both flecainide and thapsigargin (an Iup inhibitor);Citation71 the threshold of SOICR was significantly reduced in these CPVT human iPSC-derived CMs.

Lastly, in iPSC-derived CMs produced from a CPVT patient carrying RyR2S406L mutation,Citation79 catecholamines led to an increase in diastolic intracellular Ca2+ transients and a reduced SR Ca2+ content coupled with an increased susceptibility to DADs due to increased frequency and duration of Ca2+ sparks. Dantrolene, a drug effective for treating malignant hyperthermia (presumably via inhibiting the ryanodine receptor) was able to rescue arrhythmogenic properties of CPVT iPSC-derived CMs.

Genotype–phenotype correlation and risk stratification in congenital LQTS

Clinically relevant genotype–phenotype relationships have been investigated in the ECG ST-T waves, responses to epinephrine, types of arrhythmogenic trigger, responsiveness to antiarrhythmic agents, and risk stratification.Citation80Citation82 Characteristically, an arrhythmic event is frequently exertion-triggered in LQT1 and auditory-triggered in LQT2, and by contrast it usually occurs at rest in LQT3. β-blocker therapy is most effective in LQT1 followed by LQT2 and least effective in LQT3; in fact, it should be considered contraindicated in LQT3.Citation70 Because IKs, IKr, INaK, IK1, and ICa,L are catecholamine-sensitive, the trigger of arrhythmic events is often related to enhanced sympathetic tone in certain congenital LQTS, such as LQT1 (KCNQ1-IKs), LQT2 (KCNH2-IKs), LQT4 (ANK2-INaK, INaCa, IP), LQT5 (KCNE-IKs), LQT7 (KCNJ2-IK1), LQT8 (CACNA1C-ICa,L), and LQT11 (AKAP9-IKs). An arrhythmic event triggered by swimming is relatively specific for LQT1,Citation70 but it also has been reported in cases of CPVT and LQT7.Citation17

LQT1, LQT2 and LQT3 constitute the majority (approximately 65%–75%) of congenital LQTS.Citation83 Among these three subtypes, it has been suggested that the prognostic value might be assigned according to the location of a particular mutation. In LQT2, Moss et alCitation84 noticed that patients with mutations in the pore region of the KCNH2-encoded K+ channel (Kv 11.1) did worse than those who harbored mutations in the C-terminus. Similarly, Migdalovich et alCitation85 found that pore-loop mutations displayed a significant higher risk in men (>twofold) than in women, but overall women were more vulnerable than men to life-threatening cardiac events (26% vs 14%). Furthermore, Kim et alCitation86 noted that the type of trigger, in addition to sex, location of mutations, and QTc interval (≥500 m), was an important risk factor for arrhythmic events in LQT2; risk factors for arousal-triggered cardiac events included sex (female: male > 13 years: hazard ratio [HR] = 9.10, P < 0.001) and the presence of pore-loop mutations (HR = 2.19, P = 0.009), and β-blocker therapy was beneficial in patients with exercise-triggered events (HR = 0.29, P < 0.01), but not in those with arousal and nonexercise/nonarousal events.

Similarly, in LQT1 patients, Shimizu et alCitation87 reported that patients with mutations involving the transmembrane-spanning domains and/or pore regions of Kv LQT1 had a poorer outcome compared to those harboring mutations in the C-terminus. Crotti et alCitation88 noted that LQT1 mutation KCNQ1A341V was associated with a high clinical severity (SCD rate 14%) despite β-blocker therapy. These KCNQ1A341V patients were more likely to have a longer QTc interval (485 ± 43 vs 465 ± 38 milliseconds), cardiac events (75% vs 24%), and were younger at first event (6 vs 11 years) compared to non-A341V patients. Moreover, this KCNQ1A341V “hot spot” was predictive of high clinical severity independent of ethnic origin.

Nevertheless, it is well appreciated that variable expressivity and “genotype–phenotype violation” are not infrequently encountered in “inheritable arrhythmogenic disorders,” presumably due to incomplete penetrance, epigenetic factors, and gene-to-gene interaction (eg, compound mutations, modifier genes, polymorphisms, etc).Citation80,Citation89Citation91 Consequently, the concept of a malignant domain should not be generally applied.Citation90 In genotype-positive and phenotype-negative patients, genotype (including the site of mutation), age, sex, family history, and the QTc interval all need to be taken into consideration while contemplating preventive strategy with lifestyle adjustment, pharmacotherapy, and/or prophylactic implantation of an implantable cardioverter/defibrillator.Citation80,Citation90,Citation92

Conclusion

Our current understanding of the pathophysiology of inherited arrhythmogenic disorders has come mostly from intensive and extensive investigations of ion channelopathies, especially congenital LQTS and others like CPVT and BrS over the past 15 years. The accumulated knowledge of the functionality of each ion channel in relation to gene mutations has facilitated research and development of more effective targeted pharmacotherapy and nonpharmacological treatment modalities, such as gene therapy and catheter ablation.

Technology with the ability to generate human iPSCsCitation67,Citation68 has become a new paradigm for pathophysiological study of patient- and disease-specific cells and for screening of new drugs, and clinical application of gene therapy. However, because this technology is relatively new, the validity and reproducibility of the findings need to be carefully verified. Human iPSC-derived CMs are usually immature, more akin to fetal CMs, as yet to recapitulate all the characteristics of adult CMs. Moreover, better purification of human iPSC-derived CMs is needed, as they often represent a mixture of cell types – atrial, nodal, and ventricular – that display various electrophysiological properties; as a result, CMs so derived might not fully exhibit disease-specific phenotypic features. Further refinement of the technology (eg, the method used for cell isolation and cultivation, selection of CM-specific promoters, etc) is currently under way.Citation65,Citation66 It is anticipated that continued efforts and collaboration between clinical and basic science researchers are required to overcome difficulties concerning the use of iPSC technology in the diagnosis and management of arrhythmic disorders in clinical practice.

Acknowledgments

This work was in part supported by grants from the Ministry of Education (Development Plan for World Class Universities and Research Centers of Excellence – National Central University).

Disclosure

The authors report no conflicts of interest in this work.

References

  • SangerFCoulsonARA rapid method for determining sequences in DNA by primed synthesis with DNA polymeraseJ Mol Biol19759434414481100841
  • CurranMESplawskiITimothyKWVincentGMGreenEDKeatingMTA molecular basis for cardiac arrhythmia: HERG mutations cause long QT syndromeCell19958057958037889573
  • WangQCurranMESplawskiIPositional cloning of a novel potassium channel gene: KVLQT1 mutations cause cardiac arrhythmiasNat Genet199612117238528244
  • WangQShenJSplawskiISCN5A mutations associated with an inherited cardiac arrhythmia, long QT syndromeCell19958058058117889574
  • WatanabeHKoopmannTTLe ScouarnecSSodium channel beta1 subunit mutations associated with Brugada syndrome and cardiac conduction disease in humansJ Clin Invest200811862260226818464934
  • SabirINKilleenMJGraceAAHuangCLVentricular arrhythmogenesis: insights from murine modelsProg Biophys Mol Biol2008982–320821819041335
  • SchwartzPJPrioriSGSpazzoliniCGenotype-phenotype correlation in the long-QT syndrome: gene-specific triggers for life-threatening arrhythmiasCirculation20011031899511136691
  • AckermanMJMohlerPJDefining a new paradigm for human arrhythmia syndromes: phenotypic manifestations of gene mutations in ion channel- and transporter-associated proteinsCirc Res2010107445746520724725
  • MohlerPJSchottJJGramoliniAOAnkyrin-B mutation causes type 4 long-QT cardiac arrhythmia and sudden cardiac deathNature2003421692363463912571597
  • VattaMAckermanMJYeBMutant caveolin-3 induces persistent late sodium current and is associated with long-QT syndromeCirculation2006114202104211217060380
  • WoodmanSEParkDSCohenAWCaveolin-3 knockout mice develop a progressive cardiomyopathy and show hyperactivation of the p42/44 MAPK cascadeJ Biol Chem200227741389883899712138167
  • CronkLBYeBKakuTNovel mechanism for sudden infant death syndrome: persistent late sodium current secondary to mutations in caveolin-3Heart Rhythm20074216116617275750
  • ChenLMarquardtMLTesterDJSampsonKJAckermanMJKassRSMutation of an A-kinase-anchoring protein causes long-QT syndromeProc Natl Acad Sci USA200710452209902099518093912
  • AarnoudseAJNewton-ChehCde BakkerPICommon NOS1AP variants are associated with a prolonged QTc interval in the Rotterdam StudyCirculation20071161101617576865
  • ArkingDEPfeuferAPostWA common genetic variant in the NOS1 regulator NOS1AP modulates cardiac repolarizationNat Genet200638664465116648850
  • PlasterNMTawilRTristani-FirouziMMutations in Kir2.1 cause the developmental and episodic electrical phenotypes of Andersen’s syndromeCell2001105451151911371347
  • Tristani-FirouziMEtheridgeSPKir 2.1 channelopathies: the Andersen-Tawil syndromePflugers Arch2010460228929420306271
  • ZhangLBensonDWTristani-FirouziMElectrocardiographic features in Andersen-Tawil syndrome patients with KCNJ2 mutations: characteristic T-U-wave patterns predict the KCNJ2 genotypeCirculation2005111212720272615911703
  • LopatinANNicholsCGInward rectifiers in the heart: an update on I(K1)J Mol Cell Cardiol200133462563811273717
  • Barajas-MartinezHHuDOntiverosGBiophysical and molecular characterization of a novel de novo KCNJ2 mutation associated with Andersen-Tawil syndrome and catecholaminergic polymorphic ventricular tachycardia mimicryCirc Cardiovasc Genet201141515721148745
  • TsuboiMAntzelevitchCCellular basis for electrocardiographic and arrhythmic manifestations of Andersen-Tawil syndrome (LQT7)Heart Rhythm20063332833516500306
  • TesterDJAryaPWillMGenotypic heterogeneity and phenotypic mimicry among unrelated patients referred for catecholaminergic polymorphic ventricular tachycardia genetic testingHeart Rhythm20063780080516818210
  • VegaALTesterDJAckermanMJMakielskiJCProtein kinase A-dependent biophysical phenotype for V227F-KCNJ2 mutation in catecholaminergic polymorphic ventricular tachycardiaCirc Arrhythm Electrophysiol20092554054719843922
  • LiuNPrioriSGDisruption of calcium homeostasis and arrhythmogenesis induced by mutations in the cardiac ryanodine receptor and calsequestrinCardiovasc Res200877229330118006488
  • SplawskiITimothyKWSharpeLMCa(V)1.2 calcium channel dysfunction causes a multisystem disorder including arrhythmia and autismCell20041191193115454078
  • SplawskiITimothyKWDecherNSevere arrhythmia disorder caused by cardiac L-type calcium channel mutationsProc Natl Acad Sci USA20051022380898096 discussion8086808815863612
  • NavedoMFChengEPYuanCIncreased coupled gating of L-type Ca2+ channels during hypertension and Timothy syndromeCirc Res2010106474875620110531
  • ErxlebenCLiaoYGentileSCyclosporin and Timothy syndrome increase mode 2 gating of CaV1.2 calcium channels through aberrant phosphorylation of S6 helicesProc Natl Acad Sci USA2006103103932393716537462
  • YarotskyyVGaoGPetersonBZElmslieKSThe Timothy syndrome mutation of cardiac CaV1.2 (L-type) channels: multiple altered gating mechanisms and pharmacological restoration of inactivationJ Physiol2009587Pt 355156519074970
  • SicouriSTimothyKWZygmuntACCellular basis for the electrocardiographic and arrhythmic manifestations of Timothy syndrome: effects of ranolazineHeart Rhythm20074563864717467634
  • SungRJWuYHLaiNHBeta-adrenergic modulation of arrhythmogenesis and identification of targeted sites of antiarrhythmic therapy in Timothy (LQT8) syndrome: a theoretical studyAm J Physiol Heart Circ Physiol20102981H33H4419855067
  • ChengEPYuanCNavedoMFRestoration of normal L-type Ca2+ channel function during Timothy syndrome by ablation of an anchoring proteinCirc Res2011109325526121700933
  • CerroneMColombiBSantoroMBidirectional ventricular tachycardia and fibrillation elicited in a knock-in mouse model carrier of a mutation in the cardiac ryanodine receptorCirc Res20059610e77e8215890976
  • LiuNColombiBMemmiMArrhythmogenesis in catecholaminergic polymorphic ventricular tachycardia: insights from a RyR2 R4496C knock-in mouse modelCirc Res200699329229816825580
  • Fernandez-VelascoMRuedaARizziNIncreased Ca2+ sensitivity of the ryanodine receptor mutant RyR2R4496C underlies catecholaminergic polymorphic ventricular tachycardiaCirc Res20091042201209 12p following 20919096022
  • SedejSHeinzelFRWaltherSNa+-dependent SR Ca2+ overload induces arrhythmogenic events in mouse cardiomyocytes with a human CPVT mutationCardiovasc Res2010871505920080988
  • KangGGiovannoneSFLiuNPurkinje cells from RyR2 mutant mice are highly arrhythmogenic but responsive to targeted therapyCirc Res2010107451251920595652
  • SinhaAMSchmidtMMarschangHRole of left ventricular scar and Purkinje-like potentials during mapping and ablation of ventricular fibrillation in dilated cardiomyopathyPacing Clin Electrophysiol200932328629019272055
  • TerentyevDKubalovaZValleGModulation of SR Ca release by luminal Ca and calsequestrin in cardiac myocytes: effects of CASQ2 mutations linked to sudden cardiac deathBiophys J20089542037204818469084
  • GyorkeIHesterNJonesLRGyorkeSThe role of calsequestrin, triadin, and junctin in conferring cardiac ryanodine receptor responsiveness to luminal calciumBiophys J20048642121212815041652
  • KnollmannBCChopraNHlaingTCasq2 deletion causes sarcoplasmic reticulum volume increase, premature Ca2+ release, and catecholaminergic polymorphic ventricular tachycardiaJ Clin Invest200611692510252016932808
  • KirchheferUWehrmeisterDPostmaAVThe human CASQ2 mutation K206N is associated with hyperglycosylation and altered cellular calcium handlingJ Mol Cell Cardiol20104919510520302875
  • PrioriSGChenSRInherited dysfunction of sarcoplasmic reticulum Ca2+ handling and arrhythmogenesisCirc Res2011108787188321454795
  • SungRJLoCPHsiaoPYTienHCTargeting intracellular calcium cycling in catecholaminergic polymorphic ventricular tachycardia: a theoretical investigationAm J Physiol Heart Circ Physiol20113014H1625H163821742998
  • HilliardFASteeleDSLaverDFlecainide inhibits arrhythmogenic Ca2+ waves by open state block of ryanodine receptor Ca2+ release channels and reduction of Ca2+ spark massJ Mol Cell Cardiol201048229330119835880
  • van der WerfCKannankerilPJSacherFFlecainide therapy reduces exercise-induced ventricular arrhythmias in patients with catecholaminergic polymorphic ventricular tachycardiaJ Am Coll Cardiol201157222244225421616285
  • WatanabeHChopraNLaverDFlecainide prevents catecholaminergic polymorphic ventricular tachycardia in mice and humansNat Med200915438038319330009
  • KatritsisDRowlandEO’NunainSShakespeareCFPolonieckiJCammAJEffect of flecainide on atrial and ventricular refractoriness and conduction in patients with normal left ventricle. Implications for possible antiarrhythmic and proarrhythmic mechanismsEur Heart J19951612193019358682029
  • LiuNDenegriMRuanYFSodium channel blockers prevent triggered activity but not abnormal Ca2+ release in a knock-in mouse model with ryanodine receptor mutation R4496CCirculation201012221A13707
  • CaballeroRDolz-GaitonPGomezRFlecainide increases Kir2.1 currents by interacting with cysteine 311, decreasing the polyamine-induced rectificationProc Natl Acad Sci USA201010735156311563620713726
  • LiuNRuanYDenegriMCalmodulin kinase II inhibition prevents arrhythmias in RyR2(R4496C+/−) mice with catecholaminergic polymorphic ventricular tachycardiaJ Mol Cell Cardiol201150121422220937285
  • HoogendijkMGDiagnostic dilemmas: overlapping features of Brugada syndrome and arrhythmogenic right ventricular cardiomyopathyFront Physiol2012314422654761
  • NademaneeKVeerakulGNimmannitSArrhythmogenic marker for the sudden unexplained death syndrome in Thai menCirculation1997968259526009355899
  • ChenQKirschGEZhangDGenetic basis and molecular mechanism for idiopathic ventricular fibrillationNature199839266732932969521325
  • HuDBarajas-MartinezHBurashnikovEA mutation in the beta 3 subunit of the cardiac sodium channel associated with Brugada ECG phenotypeCirc-Cardiovasc Gene200923270278
  • LondonBMichalecMMehdiHMutation in glycerol-3-phosphate dehydrogenase 1 like gene (GPD1-L) decreases cardiac Na+ current and causes inherited arrhythmiasCirculation2007116202260226817967977
  • KattygnarathDMaugenreSNeyroudNMOG1: a new susceptibility gene for Brugada syndromeCirc Cardiovasc Genet20114326126821447824
  • AntzelevitchCPollevickGDCordeiroJMLoss-of-function mutations in the cardiac calcium channel underlie a new clinical entity characterized by ST-segment elevation, short QT intervals, and sudden cardiac deathCirculation2007115444244917224476
  • BurashnikovEPfeifferRBarajas-MartinezHMutations in the cardiac L-type calcium channel associated with inherited J-wave syndromes and sudden cardiac deathHeart Rhythm20107121872188220817017
  • DelpónECordeiroJMNúñezLFunctional effects of KCNE3 mutation and its role in the development of Brugada syndromeCirc Arrhythm Electrophysiol20081320921819122847
  • Barajas-MartínezHHuDFerrerTMolecular genetic and functional association of Brugada and early repolarization syndromes with S422L missense mutation in KCNJ8Heart Rhythm20129454855522056721
  • NademaneeKVeerakulGChandanamatthaPPrevention of ventricular fibrillation episodes in Brugada syndrome by catheter ablation over the anterior right ventricular outflow tract epicardiumCirculation2011123121270127921403098
  • SunsaneewitayakulBYaoYThamareeSZhangSEndocardial mapping and catheter ablation for ventricular fibrillation prevention in Brugada syndromeJ Cardiovasc Electrophysiol201223Suppl 1s10s1622988965
  • WatanabeHYangTStroudDMStriking in vivo phenotype of a disease-associated human SCN5A mutation producing minimal changes in vitroCirculation201112491001101121824921
  • HoekstraMMummeryCLWildeAABezzinaCRVerkerkAOInduced pluripotent stem cell derived cardiomyocytes as models for cardiac arrhythmiasFront Physiol2012334623015789
  • YoshidaYYamanakaSRecent stem cell advances: induced pluripotent stem cells for disease modeling and stem cell-based regenerationCirculation20101221808720606130
  • TakahashiKTanabeKOhnukiMInduction of pluripotent stem cells from adult human fibroblasts by defined factorsCell2007131586187218035408
  • ZhangJWilsonGFSoerensAGFunctional cardiomyocytes derived from human induced pluripotent stem cellsCirc Res20091044e30e4119213953
  • MorettiABellinMWellingAPatient-specific induced pluripotent stem-cell models for long-QT syndromeN Engl J Med2010363151397140920660394
  • MossAJKassRSLong QT syndrome: from channels to cardiac arrhythmiasJ Clin Invest200511582018202416075042
  • ItzhakiIMaizelsLHuberIModelling the long QT syndrome with induced pluripotent stem cellsNature2011471733722522921240260
  • MatsaERajamohanDDickEDrug evaluation in cardiomyocytes derived from human induced pluripotent stem cells carrying a long QT syndrome type 2 mutationEur Heart J201132895296221367833
  • LahtiALKujalaVJChapmanHModel for long QT syndrome type 2 using human iPS cells demonstrates arrhythmogenic characteristics in cell cultureDis Model Mech20125222023022052944
  • MalanDFriedrichsSFleischmannBKSassePCardiomyocytes obtained from induced pluripotent stem cells with long-QT syndrome 3 recapitulate typical disease-specific features in vitroCirc Res2011109884184721799153
  • DavisRPCasiniSvan den BergCWCardiomyocytes derived from pluripotent stem cells recapitulate electrophysiological characteristics of an overlap syndrome of cardiac sodium channel diseaseCirculation2012125253079309122647976
  • RemmeCAVerkerkAONuyensDOverlap syndrome of cardiac sodium channel disease in mice carrying the equivalent mutation of human SCN5A-1795insDCirculation2006114242584259417145985
  • YazawaMHsuehBJiaXUsing induced pluripotent stem cells to investigate cardiac phenotypes in Timothy syndromeNature2011471733723023421307850
  • FatimaAXuGShaoKIn vitro modeling of ryanodine receptor 2 dysfunction using human induced pluripotent stem cellsCell Physiol Biochem201128457959222178870
  • JungCBMorettiAMederos y SchnitzlerMDantrolene rescues arrhythmogenic RYR2 defect in a patient-specific stem cell model of catecholaminergic polymorphic ventricular tachycardiaEMBO Mol Med20124318019122174035
  • AckermanMJGenetic testing for risk stratification in hypertrophic cardiomyopathy and long QT syndrome: fact or fiction?Curr Opin Cardiol200520317518115861004
  • GoldenbergIBradleyJMossABeta-blocker efficacy in high-risk patients with the congenital long-QT syndrome types 1 and 2: implications for patient managementJ Cardiovasc Electrophysiol201021889390120233272
  • LiuJFJonsCMossAJRisk factors for recurrent syncope and subsequent fatal or near-fatal events in children and adolescents with long QT syndromeJ Am Coll Cardiol201157894195021329841
  • TesterDJWillMLHaglundCMAckermanMJCompendium of cardiac channel mutations in 541 consecutive unrelated patients referred for long QT syndrome genetic testingHeart Rhythm20052550751715840476
  • MossAJZarebaWKaufmanESIncreased risk of arrhythmic events in long-QT syndrome with mutations in the pore region of the human ether-a-go-go-related gene potassium channelCirculation2002105779479911854117
  • MigdalovichDMossAJLopesCMMutation and gender-specific risk in type 2 long QT syndrome: implications for risk stratification for life-threatening cardiac events in patients with long QT syndromeHeart Rhythm20118101537154321440677
  • KimJALopesCMMossAJTrigger-specific risk factors and response to therapy in long QT syndrome type 2Heart Rhythm20107121797180520850565
  • ShimizuWHorieMOhnoSMutation site-specific differences in arrhythmic risk and sensitivity to sympathetic stimulation in the LQT1 form of congenital long QT syndrome: multicenter study in JapanJ Am Coll Cardiol200444111712515234419
  • CrottiLSpazzoliniCSchwartzPJThe common long-QT syndrome mutation KCNQ1/A341V causes unusually severe clinical manifestations in patients with different ethnic backgrounds: toward a mutation-specific risk stratificationCirculation2007116212366237517984373
  • CrottiLMontiMCInsoliaRNOS1AP is a genetic modifier of the long-QT syndromeCirculation2009120171657166319822806
  • GiudicessiJRAckermanMJDeterminants of incomplete penetrance and variable expressivity in heritable cardiac arrhythmia syndromesTransl Res Epub9172012
  • KoboriASaraiNShimizuWAdditional gene variants reduce effectiveness of beta-blockers in the LQT1 form of long QT syndromeJ Cardiovasc Electrophysiol200415219019915028050
  • PrioriSGSchwartzPJNapolitanoCRisk stratification in the long-QT syndromeN Engl J Med2003348191866187412736279
  • SplawskiITristani-FirouziMLehmannMHSanguinettiMCKeatingMTMutations in the hminK gene cause long QT syndrome and suppress IKs functionNat Genet19971733383409354802
  • AbbottGWSestiFSplawskiIMiRP1 forms IKr potassium channels with HERG and is associated with cardiac arrhythmiaCell199997217518710219239
  • AiTFujiwaraYTsujiKNovel KCNJ2 mutation in familial periodic paralysis with ventricular dysrhythmiaCirculation2002105222592259412045162
  • YeBTesterDJVattaMMakielskiJCAckermanMJAB1-1: Molecular and functional characterization of novel cav3-encoded caveolin-3 mutations in congenital long QT syndromeHeart Rhythm200635S116688893
  • DomingoAMKakuTTesterDJAB16-6: sodium channel β4 subunit mutation causes congenital long QT syndromeHeart Rhythm200635S34
  • Medeiros-DomingoAKakuTTesterDJSCN4B-encoded sodium channel beta4 subunit in congenital long-QT syndromeCirculation2007116213414217592081
  • UedaKValdiviaCMedeiros-DomingoASyntrophin mutation associated with long QT syndrome through activation of the nNOS-SCN5A macromolecular complexProc Natl Acad Sci USA2008105279355936018591664
  • WuGAiTKimJJAlpha-1-syntrophin mutation and the long-QT syndrome: a disease of sodium channel disruptionCirc Arrhythm Electrophysiol20081319320119684871
  • YangYLiangBLiuJIdentification of a Kir3.4 mutation in congenital long QT syndromeAm J Hum Genet201086687288020560207
  • KapplingerJWildeAAntzelevitchCA worldwide compendium of putative Brugada syndrome associated mutations in the SCN5A encoded cardiac sodium channelHeart Rhythm20096S392
  • Schulze-BahrEEckardtLBreithardtGSodium channel gene (SCN5A) mutations in 44 index patients with Brugada syndrome: different incidences in familial and sporadic diseaseHum Mutat200321665165214961552
  • Medeiros-DomingoATanBHCrottiLGain-of-function mutation S422L in the KCNJ8-encoded cardiac K(ATP) channel Kir6.1 as a pathogenic substrate for J-wave syndromesHeart Rhythm20107101466147120558321
  • GiudicessiJRYeDTesterDJTransient outward current (I(to)) gain-of-function mutations in the KCND3-encoded Kv4.3 potassium channel and Brugada syndromeHeart Rhythm2011871024103221349352
  • LaitinenPJBrownKMPiippoKMutations of the cardiac ryanodine receptor (RyR2) gene in familial polymorphic ventricular tachycardiaCirculation2001103448549011157710
  • PrioriSGNapolitanoCTisoNMutations in the cardiac ryanodine receptor gene (hRyR2) underlie catecholaminergic polymorphic ventricular tachycardiaCirculation2001103219620011208676
  • LahatHPrasEOlenderTA missense mutation in a highly conserved region of CASQ2 is associated with autosomal recessive catecholamine-induced polymorphic ventricular tachycardia in Bedouin families from IsraelAm J Hum Genet20016961378138411704930
  • PostmaAVDenjoyIHoorntjeTMAbsence of calsequestrin 2 causes severe forms of catecholaminergic polymorphic ventricular tachycardiaCirc Res2002918e21e2612386154
  • PostmaAVBhuiyanZAShkolnikovaMInvolvement of the Kir2 gene family in catecholaminergic polymorphic ventricular tachycardia; analysis for mutations and identification of numerous pseudogenesEur Heart J20042566
  • MohlerPJSplawskiINapolitanoCA cardiac arrhythmia syndrome caused by loss of ankyrin-B functionProc Natl Acad Sci USA2004101249137914215178757
  • BrugadaRHongKDumaineRSudden death associated with short-QT syndrome linked to mutations in HERGCirculation20041091303514676148
  • BellocqCvan GinnekenACBezzinaCRMutation in the KCNQ1 gene leading to the short QT-interval syndromeCirculation2004109202394239715159330
  • PrioriSGPanditSVRivoltaIA novel form of short QT syndrome (SQT3) is caused by a mutation in the KCNJ2 geneCirc Res200596780080715761194
  • TemplinCGhadriJRRougierJSIdentification of a novel loss-of-function calcium channel gene mutation in short QT syndrome (SQTS6)Eur Heart J20113291077108821383000