You are on page 1of 46

Physiol Rev 97: 89 –134, 2017

Published November 2, 2016; doi:10.1152/physrev.00008.2016

MOLECULAR PATHOPHYSIOLOGY OF CONGENITAL


LONG QT SYNDROME
M. S. Bohnen, G. Peng, S. H. Robey, C. Terrenoire, V. Iyer, K. J. Sampson, and R. S. Kass

Department of Pharmacology, Columbia University Medical Center, New York, New York; and The New York
Stem Cell Foundation Research Institute, New York, New York

Bohnen MS, Peng G, Robey SH, Terrenoire C, Iyer V, Sampson KJ, Kass RS.

L
Molecular Pathophysiology of Congenital Long QT Syndrome. Physiol Rev 97: 89 –134,
2017. Published November 2, 2016; doi:10.1152/physrev.00008.2016.—Ion
channels represent the molecular entities that give rise to the cardiac action potential,
the fundamental cellular electrical event in the heart. The concerted function of these

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


channels leads to normal cyclical excitation and resultant contraction of cardiac muscle. Research
into cardiac ion channel regulation and mutations that underlie disease pathogenesis has greatly
enhanced our knowledge of the causes and clinical management of cardiac arrhythmia. Here we
review the molecular determinants, pathogenesis, and pharmacology of congenital Long QT Syn-
drome. We examine mechanisms of dysfunction associated with three critical cardiac currents
that comprise the majority of congenital Long QT Syndrome cases: 1) IKs, the slow delayed
rectifier current; 2) IKr, the rapid delayed rectifier current; and 3) INa, the voltage-dependent
sodium current. Less common subtypes of congenital Long QT Syndrome affect other cardiac
ionic currents that contribute to the dynamic nature of cardiac electrophysiology. Through the
study of mutations that cause congenital Long QT Syndrome, the scientific community has ad-
vanced understanding of ion channel structure-function relationships, physiology, and pharmaco-
logical response to clinically employed and experimental pharmacological agents. Our understand-
ing of congenital Long QT Syndrome continues to evolve rapidly and with great benefits: genotype-
driven clinical management of the disease has improved patient care as precision medicine
becomes even more a reality.

I. INTRODUCTION 89 yielded much insight into the pathophysiology of congenital


II. IKs DYSFUNCTION IN CONGENITAL... 91 LQTS.
III. IKr DYSFUNCTION IN CONGENITAL... 99
IV. INa DYSFUNCTION IN CONGENITAL... 106 Clinically, congenital LQTS patients often first present after
V. OTHER SUBTYPES OF CONGENITAL LQTS 114 episodes of syncope and/or seizure, and the ECG reveals a
VI. GENOTYPE-DRIVEN CLINICAL... 116 prolonged QT interval. The ECG measures electrical activ-
VII. CONCLUSIONS 120 ity of the heart over time, at the patient’s body surface. The
primary electrical signals observed include the P wave,
which signifies atrial depolarization; the QRS complex,
I. INTRODUCTION
which arises from ventricular depolarization; and the T
wave, due to ventricular repolarization (FIGURE 1A). The
A genetic disorder disrupting electrical activity in the heart,
QT interval, therefore, reflects the time elapsed from the
congenital Long QT Syndrome (LQTS) can lead to life-
initiation of ventricular depolarization to the end of ventric-
threatening arrhythmias and sudden cardiac death. In the
ular repolarization. The QT interval shortens with increas-
first cases of congenital LQTS, described in 1957, several
children in one family presented with prolongation of the ing heart rate thus requiring a normalization, or “correc-
QT interval on the electrocardiogram (ECG) and congenital tion,” for heart rate. For a diagnosis of LQTS, this rate-
deafness (189). This came to be known as Jervell and corrected QT (QTc) interval prolongation on a 12-lead
Lange-Nielsen syndrome (JLNS), the autosomal recessive ECG generally is referenced as ⬎470 ms for males and
form of LQTS. The more common, autosomal dominant ⬎480 ms for females. QTc also varies with age, and thus, an
form of congenital LQTS that presents without deafness age-appropriate prolonged QTc interval in a patient aids
was first described in 1963 and 1964 in two separate cases the diagnosis of LQTS (209). However, diagnosis of LQTS
and became known as Romano-Ward syndrome (346, based on absolute QT interval cutoffs can be challenging,
459). Since these initial patient descriptions, advances in since there is considerable overlap in the QTc distribution
our understanding of the mechanisms of cardiac electrical of affected patients and otherwise healthy individuals
excitability at the tissue, cellular, and molecular level have (193). Asymptomatic patients can have intervals beyond

0031-9333/17 Copyright © 2017 the American Physiological Society 89


BOHNEN ET AL.

A The ventricular cellular action potential results from the


summation of a large number of ion channel currents and
QRS
electrogenic pumps that control the cellular membrane po-
tential (see Nerbonne and Kass review, Ref. 299), but in this
ECG
review we will focus on three key ion channels that are
P-Wave T-Wave
well-established to be linked to LQTS and that are illus-
trated in FIGURE 1B. The cellular resting membrane poten-
QT Interval tial is approximately ⫺85 mV, determined largely by in-
wardly rectifying K⫹ channels. Inward rectification is a
B property that hinders the outward flow of K⫹ as membrane
potentials become positive, but passes K⫹ more efficiently
at potentials negative to the K⫹ equilibrium potential where
the flow of K⫹ would be inward. Hence, the term “inward
Ventricular AP rectification” is used to describe these channels (304). Ac-
tivation of Nav1.5, the primary voltage-gated sodium chan-

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


nel in the heart, leads to sodium influx (INa) and membrane
depolarization. As the cell reaches approximately ⫺40 mV,
voltage-gated L-type calcium channels begin to open, lead-
ing to calcium influx (ICa) (299). Concurrently during the
IKs
upstroke of the action potential, potassium channels, in-
cluding those carrying the IKs and IKr delayed rectifier cur-
IKr
rents, begin to activate slowly. As the cell reaches ⫹30 mV,
INa
INa inactivates almost completely. At this time, a brief and
small repolarization of the membrane potential occurs via
fast activation of the transient outward K⫹ current, Ito. In
FIGURE 1. ECG to cellular ionic currents. A: membrane depolar-
the plateau phase that follows, influx of Ca2⫹ through volt-
ization and the rapid upstroke of the ventricular action potential give age-gated calcium channels is balanced mainly by IKs and
rise to the QRS complex. The duration of the QT-interval corre- IKr. The plateau phase ceases as calcium channels inactivate
sponds to the time to ventricular repolarization. The relatively stable and outward potassium efflux persists, leading to a net out-
membrane potential during the plateau phase of the action potential ward membrane current, and cell repolarization back to the
gives rise to a brief isoelectric period. Ventricular repolarization
gives rise to the T-wave. B: time course of several ionic currents that
cellular resting potential. During this process, the Na⫹-K⫹-
underlie ventricular action potential morphology (currents not to ATPase helps maintain intracellular concentrations of these
scale). The rapidly activating and inactivating INa drives membrane key ions (304).
depolarization. Two K⫹ currents, IKs and IKr, contribute most to the
repolarizing current necessary to drive membrane potential back to In LQTS patients, the QT is prolonged presumably due to
rest.
prolongation of underlying action potential durations

these cutoff and develop no arrhythmias; similarly, QTc


intervals below this cutoff can be seen in patients with es- K+ Na+
tablished LQTS (with clinical arrhythmias and positive ge-
netic testing) (16, 17, 338). Clinical scoring systems (368),
Extracellular
as well as genetic testing, can be helpful to assist with the
diagnosis of congenital LQTS (193), particularly when QT
intervals are on the borderline (within 20 ms of these cut-
offs) or when clinical history is equivocal. This review will
focus primarily on the various forms of congenital LQTS.
Cytosol
Ion channels are the molecular entities underlying most
ionic currents in the heart, allowing passive diffusion of
ions across the cell membrane’s electrochemical gradients K+ Na+
(FIGURE 2). A selectivity filter in the channel pore, deter-
mined by distinct atomic components (152, 315), endows FIGURE 2. Schematic of a generic K⫹ and Na⫹ ion channel. Ion
selective permeation of ions, such as Na⫹, K⫹, and Ca2⫹ channels allow for selective permeation of ions through the plasma
membrane down their electrochemical gradient. The classic K⫹
(246). Some ion channels exhibit voltage-dependent gating, channel consists of four identical pore-forming subunits, whereas
where voltage-sensing domains respond to changes in mem- each Na⫹ channel is formed by a single polypeptide with four homol-
brane potential to cause channel opening or closing (247). ogous domains.

90 Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org


CONGENITAL LONG QT SYNDROME

(APDs), most often caused by decreased repolarizing IKs or encodes hERG), respectively, while LQT3 is associated
IKr activity, or persistent sodium influx that extends with mutations in SCN5A, the gene coding for the Nav1.5
through the plateau phase. A loss of IKs or IKr function, or a sodium channel alpha subunit. Disease association for vari-
gain of INa function, predisposes ventricular myocytes to ants in these three proteins is supported by genome-wide
early afterdepolarizations (EADs), and in some cases to de- association studies (300) and functional electrophysiologi-
layed afterdepolarizations (DADs) which may underlie de- cal characterization of mutant channels. In addition, LQTS-
generation into a characteristic sinusoidal wave pattern on associated mutations exist less frequently in other ion chan-
the ECG, referred to as torsades de pointes, which may nels, modulatory channel subunits, and signaling- or cyto-
further regress into ventricular fibrillation and sudden car- skeleton-associated proteins. Understanding the molecular
diac death. EADs are driven in large part by calcium entry mechanisms that cause LQTS allows for optimization of
via L-type calcium channels during prolonged action poten- genotype-specific treatments. In this review, we discuss the
tial plateau phases, whereas DADs, which occur over the molecular physiology, biology, and pathophysiology un-
diastolic range of potentials after action potential repolar- derlying congenital LQTS, and the cellular and molecular
ization, are caused by intracellular calcium overload, also a underpinnings of genotype-driven clinical management of
consequence of action potential prolongation (126). Addi- LQTS.

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


tionally, arrhythmic activity may result from altered refrac-
toriness and impulse block, also putative consequences of
II. IKs DYSFUNCTION IN CONGENITAL
prior APD prolongation. Thus treatment in LQTS aims to
LQTS
prevent malignant ventricular arrhythmia by shortening the
QTc interval to minimize cardiac event rates.
A. Introduction
Ion channels may interact with a variety of molecular enti-
ties that contribute to their trafficking, stabilization, signal- Of the different subtypes of inherited LQTS, subtypes 1, 5,
ing, and function (299). Coordination among different ion and 11 are associated with mutations in proteins that par-
channel types facilitates the ionic balance necessary for the ticipate in the macromolecular complex which generates
generation of an action potential and normal electrical and modulates the slow delayed rectifier potassium current
propagation through the heart. LQTS mutations may cause (IKs), which plays a critical role in the repolarization of the
an increase or decrease in ion channel function, disrupting cardiac action potential. Among all LQTS subtypes, LQT1
normal ionic balance leading to pathological electrical ac- is the most common, representing 30-35% of all congenital
tivity in the heart. LQTS (8). Upregulation of IKs during ␤-adrenergic stimu-
lation is critical to normal physiology by shortening ventric-
There are 15 subtypes of congenital LQTS, each associated ular APD and allowing for adequate diastolic filling in the
with mutations on a different gene (15) (TABLE 1). The most context of an elevated heart rate (354). Cardiac events in
common subtypes, LQT1, LQT2, and LQT3, account for patients with IKs-associated LQTS are often triggered by
the vast majority of congenital LQTS. LQT1 and LQT2 are stress and exercise, consistent with the role of adrenergic
associated with mutations in KCNQ1 and KCNH2 (which stimulation in the regulation of IKs (350, 371). Insight into

Table 1. Subtypes of congenital LQTS and their associated genes, proteins, and effects on cardiac currents
LQT Subtype Gene Protein Current

LQT1 KCNQ1 KCNQ1 (Kv7.1) 2IKs


LQT2 KCNH2 hERG (Kv11.1) 2IKr
LQT3 SCN5A Nav1.5 1INa
LQT4 (ankyrin-B syndrome) ANK2 Ankyrin-B Multichannel interactions
LQT5 KCNE1 KCNE1 (minK) 2IKs
LQT6 KCNE2 KCNE2 (MiRP1) 2IKr
LQT7 (Andersen-Tawil syndrome type 1) KCNJ2 Kir2.1 2IK1
LQT8 (Timothy syndrome) CACNA1C Cav1.2 1ICa
LQT9 CAV3 Caveolin 3 1INa
LQT10 SCN4B Nav1.5 ␤4 1INa
LQT11 AKAP9 AKAP-9 (yotiao) 2IKs
LQT12 SNTA1 ␣1-Syntrophin 1INa
LQT13 KCNJ5 Kir3.4 (GIRK4) 2IKACh
LQT14 CALM1 Calmodulin Multichannel interactions
LQT15 CALM2 Calmodulin Multichannel interactions

Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org 91


BOHNEN ET AL.

the molecular mechanisms of disease mutations have (minK) generates IKs (36, 358). While KCNE1 had previ-
greatly improved our understanding of LQTS pathophysi- ously been thought to be an independent potassium channel
ology and helped provide a first step to the future develop- (167, 412), it was confirmed that KCNQ1 is actually the ␣-
ment of targeted therapies. or pore-forming subunit of IKs while KCNE1 is a critical ␤-
or modulatory subunit. Coexpression of KCNQ1 and
KCNE1 generates the hallmark IKs current with slow acti-
B. Physiology vation. KCNQ1 and KCNE1 have been shown to be ex-
pressed in all four chambers in the heart (45), as well as the
IKs is an outward potassium current with unique kinetics inner ear (301, 351), where IKs is thought to play a role in
and voltage dependence and plays a key role in the repolar- K⫹ secretion into the endolymph (68). This explains the
ization of the cardiac action potential (299). In 1969, the observation that congenital deafness is a key feature of
delayed rectifier potassium current in sheep Purkinje fibers JLNS. In addition, KCNQ1 and KCNE1 are expressed else-
was studied and shown to comprise two kinetically distinct where in the body, including the pancreas, the kidneys, and
components (305). These currents were later subjected to the brain (1).
pharmacological dissection in guinea pig ventricular myo-

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


cytes and identified as IKs and IKr (360). 1. KCNQ1, the pore-forming subunit

IKs is slowly activating and most prominent during the pla-


KCNQ1, like most voltage-gated potassium channels,
teau and repolarizing phases of the cardiac action potential,
consists of six transmembrane helices (451) (FIGURE 3A).
where it contributes to counterbalancing calcium influx and
Four subunits of KCNQ1 come together to form a chan-
repolarization (FIGURE 1B). Expression of IKs has been dem-
nel that is capable of voltage-dependent gating (FIGURE
onstrated in both human atrial and ventricular myocytes
3B). On each subunit, the helices S1–S4 comprise the
(198, 235, 457). In addition, it has also been measured in
voltage-sensing domain, where the S4 helix, with its pos-
cardiomyocytes from a variety of non-human mammalian
itively charged arginine residues, senses changes in mem-
species including dogs (241, 406, 437, 442, 493) and rab-
brane potential (310). Following the voltage-sensing do-
bits (352). On the other hand, IKs is expressed at very low
main is the pore domain, which comprises the pore-loop,
levels or absent in mouse hearts (478), most likely because
an extracellular segment containing the selectivity filter,
at very high heart rates in mouse heart (⬃500 beats/min)
and helices that line the pore, S5 and S6. Furthermore,
this channel would have little or no time to activate and not
the cytoplasmic loop between S4 and S5 plays important
affect cardiac electrophysiology in the mouse.
roles in the voltage sensor-to-pore coupling and in volt-
age-dependent gating (82, 229, 498), which has been
Importantly, IKs is subject to upregulation by ␤-adrenergic
demonstrated in other voltage-gated channels as well
stimulation to control APD in the face of sympathetic nerve
(75, 122, 355). The cytoplasmic loop between S2 and S3
activity (210, 444). During sympathetic activation, adren-
also plays a role in channel gating (498). The COOH-
ergic stimulation increases the outward IKs current, which
terminal domain (CTD) of KCNQ1 is large and contains
counterbalances the concomitant increase in inward cal-
four intracellular ␣-helices referred to as A-D. A wide
cium current, prevents prolongation of the cardiac APD,
range of functions has been attributed to the CTD includ-
and allows for adequate diastolic filling times between heart
ing calmodulin binding, interaction with ␤-subunits and
beats (386). However, insufficient IKs activation such as
scaffolding proteins, as well as channel assembly and
that seen in LQT1 results in failure to counterbalance the
trafficking (159, 469).
calcium influx, prolonging the action potential and increas-
ing susceptibility to arrhythmia. This is consistent with ex-
ercise being a key trigger of cardiac events seen in LQT1 2. KCNE1, the ␤-subunit
patients (371).
Coexpression of KCNE1 with KCNQ1 leads to a drastic
change in channel function to generate IKs. Most promi-
C. Molecular Biology nently, assembly with KCNE1 leads to a delay in the onset
of activation, an increase in channel amplitude (FIGURE 3C),
The first known subtypes of inherited LQTS, LQT1-3, were as well as a depolarizing shift in the current-voltage rela-
distinguished by mapping to distinct chromosomes, with tionship (not illustrated) (36, 358). This results in a channel
LQT1 mapping to chromosome 11 (191, 212, 213). Even- that, compared with most other voltage-gated potassium
tually it was found that the KCNQ1 (KvLQT1) gene is channels, activates at more positive voltages and with
responsible for LQT1 and encodes a potassium channel slower kinetics. KCNE1 is a 129-amino acid protein that
(451). The current conducted by this channel was rapidly consists of a single transmembrane helix, with an extracel-
activating and minimally inactivating, unlike any previ- lular NH2 terminus and intracellular COOH terminus
ously known current in the heart, but soon it was shown (412). It is thought to have extensive contact with KCNQ1
that KCNQ1 together with the accessory protein KCNE1 including the voltage-sensing domain (37, 84, 203, 309,

92 Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org


CONGENITAL LONG QT SYNDROME

A KCNE1 KCNQ1

Extracellular

S1 S2 S3 S4 S5 S6
+
+
+
FIGURE 3. Molecular biology of IKs and
Cytosol regulation by PKA-mediated signaling.
A: the IKs macromolecular complex, in-
cAMP cluding KCNQ1, KCNE1, and associated

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


+ + scaffolding and signaling proteins. B and
C + C: single pulse voltage-clamp recordings
S27 P PKA AC9 PDE4D3 of KCNQ1 expressed alone or coex-
pressed with KCNE1 in Xenopus oocytes.
– AKAP-9 D: dialysis with 200 ␮M cAMP and 0.2
␮M okadaic acid (OA) increases IKs am-
PP1 plitude and slows deactivation when het-
N C
erologously expressed in CHO cells.
[From Chen et al. (78).]
B C D 60mV
–40mV
KCNQ1+KCNE1
–80mV
+cAMP/OA
Control

KCNQ1
40pA/pF
10µA

1s 2s 1s

378, 479), the pore domain (84, 262, 311), as well as the 1988 it was shown that stimulation of PKA activity by a
CTD (161, 510). Previous crosslinking studies suggest that cAMP analog can upregulate the delayed rectifier current
KCNE1 is located in a cleft between the voltage-sensing (444). Later it was shown that the scaffolding protein
domain and pore domain of different KCNQ1 subunits (84, A-kinase anchoring protein 9 (AKAP-9), also known as
479), underlying its ability to modulate KCNQ1 gating. yotiao, plays a central role in adrenergic regulation of IKs
With respect to the stoichiometry of KCNE1 to KCNQ1, by compartmentalizing key elements of the PKA signal-
some studies suggest a fixed 2:4 ratio (278, 326), while ing pathway, allowing for spatiotemporal control.
others suggest a flexibility in stoichiometry (293, 456) that
AKAP-9 binds to the CTD of KCNQ1 and recruits sig-
allows for modulation of kinetics of assembled channels
naling proteins including protein kinase A (PKA), protein
to provide another level of flexibility in channel function.
phosphatase 1 (PP1) (255), adenylyl cyclase 9 (AC9)
Although three other members of the KCNE family,
KCNE2-KCNE4, also are expressed in the heart (45) and (238), and the phosphodiesterase PDE4D3 (419) (FIGURE
are capable of modulating KCNQ1 activity (45, 154, 3A). Together these proteins form the IKs macromolecu-
367, 422), whether they associate with KCNQ1 in vivo lar complex that can tightly control the phosphorylation
to contribute to potassium currents in the heart remains state of the channel in response to adrenergic stimula-
to be explored. tion. PKA phosphorylates KCNQ1 at the S27 residue,
adding a phosphate group and hence a change in charge
3. Molecular components of adrenergic stimulation to this residue, which leads to increased channel activa-
tion and slower deactivation (226, 255) (FIGURE 3D). In
There have been considerable efforts to elucidate the mo- addition, phosphorylation of AKAP-9 itself contributes
lecular pathway for the ␤-adrenergic regulation of IKs. In to the PKA-mediated upregulation of IKs (78).

Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org 93


BOHNEN ET AL.

4. Role of PIP2 and calmodulin in IKs function D. Molecular Pathophysiology

The lipid molecule phosphatidylinositol 4,5-bisphosphate There are more than 530 disease-causing mutations associ-
(PIP2) is critical for KCNQ1 function. PIP2 is found in the ated with the IKs complex (15). While most of these are
inner leaflet of plasma membranes (260) and can regulate a missense mutations, they also include nonsense mutations,
variety of ion channels (51, 83, 172, 179, 250, 475). PIP2 splice site mutations, frameshifts, as well as deletions. The
mainly binds to KCNQ1 at its cytoplasmic loops and the vast majority are mutations in KCNQ1, which cause LQT1,
COOH-terminal region near S6 (80, 208, 498), which are but a number of LQTS-causing mutations have also been
thought to form the interface between the voltage-sensing found in KCNE1 and AKAP-9, which are classified as
domain and the pore domain (82, 497). PIP2 binding is LQT5 and LQT11, respectively. JLNS, an autosomal reces-
sive form of LQTS with severe bilateral sensorineural deaf-
mediated by electrostatic interactions between the an-
ness, has so far only been associated with mutations in
ionic lipid headgroup and positively charged channel res-
KCNQ1 or KCNE1 (423). Nonetheless, the majority of
idues (418). Rundown of PIP2 in the membrane leads to a
LQTS arising from mutations in IKs are autosomal domi-
drastically reduced IKs amplitude and accelerated deacti-
nant in a form known as Romano-Ward syndrome, which is
vation, suggesting that PIP2 stabilizes the open state of

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


not associated with deafness.
IKs (250). Furthermore, PIP2 rundown reduces the cur-
rent amplitude of KCNQ1 in the absence of KCNE1
Congenital LQTS patients with dysfunction in IKs present
(239). In addition, PIP2 plays a critical role in the cou- with prolongation of the QT interval on ECG. An ECG
pling between voltage sensor movement and pore open- characteristic associated with LQT1 patients more specifi-
ing/closing (498). Interestingly, one modulatory effect of cally is a broad based T wave (FIGURE 4A). Dysfunction of
KCNE1 on KCNQ1 is a large (100-fold) increase in chan- IKs reduces repolarizing current during the plateau phase,
nel sensitivity for PIP2, which contributes to augmenta- thereby leading to APD prolongation (FIGURE 4B). With a
tion of KCNQ1 current amplitude (239). Whether PIP2 prolonged APD, the cardiomyocyte is vulnerable to EADs
level is under control by physiological mechanisms to (441, 500), where a second depolarization occurs prior to
modulate IKs in cardiomyocytes remains to be demon- the complete repolarization of the first due to recovery of
strated. voltage-gated Na⫹ or Ca2⫹ channels from inactivation,
triggering life-threatening arrhythmia such as torsades de
Calmodulin is a calcium-binding protein that serves in myr- pointes (26, 464). As previously mentioned, during adren-
iad calcium signaling pathways, some of which regulate ion ergic stimulation IKs plays an especially important role in
channels (399). Calmodulin has been linked to IKs function controlling the cardiac APD. This manifests clinically in
(141, 379). Multiple studies have shown that calmodulin LQT1 patients as a susceptibility to arrhythmia during ex-
binding to the CTD of KCNQ1 is important for normal ercise (371).
channel trafficking and folding (141, 379).
1. LQT1
5. Channel trafficking
Missense mutations are responsible for the majority of
LQT1 cases and can cause channel loss of function through
A variety of molecular entities are involved in the traf- a variety of molecular mechanisms, including defects in ion
ficking of KCNQ1KCNE1. The small GTPase-RAB11 permeation (altering the pathway through which ions flow
plays an important role in the exocytosis of KCNQ1- through open channels), channel gating (mechanisms that
KCNE1 to the plasma membrane, while RAB5 mediates regulate the opening and/or closing or channels), traffick-
endocytosis of KCNQ1-KCNE1 from the plasma mem- ing, KCNQ1-KCNE1 interaction, PKA-mediated signaling
brane into endosomes (374). In a physiological stress pathway, PIP2 binding, and calmodulin binding (FIGURE 4,
response, serum and glucocorticoid-regulated kinase 1 C AND D) (TABLE 2). Non-missense mutations can also cause
(SGK1) upregulates KCNQ1-KCNE1 expression by in- LQT1. Mutations belonging to certain groups may bear
creasing RAB-11-dependent channel exocytosis (374). implications on patient phenotype, severity of arrhythmia,
There is also evidence to suggest that KCNQ1-KCNE1 as well as response to therapy.
can be internalized from the plasma membrane via clath-
rin-mediated endocytosis, a process facilitated by A) PERMEATION DEFECT. A number of LQT1 mutations that
KCNE1 (480). Furthermore, KCNQ1 expression is reg- lead to defects in permeation are found in the pore region of
ulated by ubiquitination, which can label membrane pro- KCNQ1. Pore mutations are thought in general to carry
teins for internalization and degradation. For example, greater risk of cardiac events than other mutations. One
the ubiquitin ligase Nedd42 can ubiquitinate KCNQ1 study shows that mutations in highly conserved residues,
(190), while ubiquitin-specific protease 2 (USP2) can pre- many of which are located in the pore-loop, are associated
vent channel ubiquitination (221). Both processes allow with higher risk of cardiac events (61, 197). Three LQT1
for control of KCNQ1 expression. mutations near the selectivity filter, T322M, T322A, and

94 Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org


CONGENITAL LONG QT SYNDROME

A LQT1 ECG C
40mV
–40mV

–80mV

KCNQ1 + KCNE1

IKs
LQTS IKs
B WT
LQT1 FIGURE 4. IKs dysfunction leading to con-
genital LQTS. A: ECG from a LQT1 patient
demonstrates a characteristic broad-
based T wave (unpublished data). B: simu-
AP lated action potential (top) and IKs (bottom)

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


in WT (black) and heterozygous LQT1
(gray) conditions, demonstrating the effect
5μA of 50% reduction in IKs. C: single pulse
voltage-clamp IKs recording in Xenopus
IKs oocytes demonstrating loss of channel
2s
function in an IKs-associated LQTS muta-
tion. D: topology of KCNQ1 and KCNE1 in
D KCNE1 KCNQ1 the plasma membrane. Several examples
of LQT1- and LQT5-associated mutations
N are highlighted in blue and teal, respec-
S225L tively. These mutations represent a variety
of mechanisms of loss-of-function including
Extracellular
disruption of permeation (yellow-filled
square), gating (yellow-filled circle), traffick-
ing (yellow-filled triangle), PKA-mediated
L51H + signaling (yellow-filled star), KCNQ1-
+ G314S KCNE1 interaction (white-filled square),
D220H + PIP2 affinity (white-filled circle), and cal-
modulin affinity (white-filled triangle).
A341V
A344V
Cytosol D76N
L114P S373P
G189R V254M
P127T R539W
C S546L
T587M

N C

G325R, have been shown to abolish channel conductance B) GATING DEFECT. LQT1 mutations that cause defects in
(13, 61). They exert dominant negative effects on wild-type KCNQ1 gating can be found in the pore domain (S5–S6).
(WT) IKs current in heterologous expression systems (61), Similar to other voltage-gated potassium channels, the
suggesting that mutant subunits coassemble with WT sub- COOH-terminal region of the S6 helix plays a key role in
units to form disrupted channels. Molecular dynamic sim- KCNQ1 gating (55, 157, 247). Scanning mutagenesis and
ulations suggest that these mutations disrupt the conforma- heterologous expression studies show that a number of
tion of the selectivity filter, leading to diminished K⫹ per- LQT1-linked residues in this region, such as F351 and
meation. A pair of adjacent LQT1 pore mutations, G314S L353, control KCNQ1 gating properties. For example,
and Y315C, located in the selectivity filter, dramatically F351A leads to a drastic slowing of channel activation and a
reduce IKs current amplitude and exert dominant negative depolarizing shift in voltage dependence of activation, while
effects on WT currents (52, 237). Immunofluorescence L353K leads to a constitutively open channel (55). To further
studies show that Y315C traffics to the membrane nor- elucidate the mechanism of F351A, a technique known as
mally, suggesting that the mutation results in trafficking of voltage clamp fluorometry (VCF), which utilizes fluorophore
non-conducting channels. labeling to allow simultaneous measurement of voltage sensor

Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org 95


BOHNEN ET AL.

is made constitutively active (41). Structural and muta-


Table 2. Representative LQT1-associated mutations classified tional studies suggest that R231 forms a salt-bridge in-
by mechanism teraction with E160 in S2 that stabilizes the channel in its
Reference closed state (340, 393, 472). Mutating R231 may disrupt
Mechanism Mutations Nos. this interaction and cause a defect in channel gating,
resulting in constitutive activation. One tool that can be
K⫹ permeation G314S, Y315C 52, 237
used to elucidate the effects of these S4 mutations on
T322A, T322M, G325R 13, 61
voltage sensor movement is VCF. The technique is capa-
Gating D202H 113, 114
ble of providing insights on IKs channel gating not possi-
S225L 52, 170
ble with current measurement alone.
R231C 41
A344V 391
LQT1 gating mutations are also present in other transmem-
Trafficking Y111C, L114P, P117L 98, 375
T587M, R591H, R594Q 205
brane regions (S1–S3) of the voltage-sensing domain. For
PKA-mediated G189R, R190Q, R243C, 39
example, the mutation D202H leads to biophysical defects
in IKs, shifting the current-voltage relationship to more pos-

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


signaling V254M
A341V 169 itive potentials, slowing activation, and accelerating deac-
G589D 255 tivation, all leading to reduced channel opening (114). Sin-
KCNQ1-KCNE1 gle-channel recordings of IKs utilized as a tool to study
interaction S546L, K557E 110 effects of select mutations (113) shows that D202H causes
PIP2 affinity R539W, R555C 312
a decrease in channel open probability, a decrease in open
S546L, K557E 110
states dwell time, and an increase in closed states dwell
Calmodulin affinity S373P, W392R 379
time, while maintaining single-channel conductance.
Large-scale defects
Nonsense R518X, Q530X 181
C) TRAFFICKING DEFECT. In addition to altering channel gating,
Deletion-insertion,
frameshift ⌬544 81 LQT1 mutations can also disrupt channel trafficking. Sev-
Splice site 1032G⬎A 286 eral LQT1 mutations in helix D of the CTD, including
T587M, R591H, and R594Q, have been found to diminish
channel trafficking to the membrane (205). These muta-
movement and channel current, has been used (37, 292, 308, tions are thought to disrupt a coiled-coil motif in helix D
309, 348, 498). It was shown using VCF that F351A alters the that plays an important role in channel trafficking (205).
coupling between voltage sensor and the pore (309), resulting Yet trafficking mutations are not limited to the CTD of
in a slowly activating channel that partially resembles IKs. In KCNQ1. LQT1 mutations in the NH2-terminal region of
addition, a LQT1 mutation in on the S6 helix, A344V, also KCNQ1, including Y111C, L114P, and P117L, have also
affects channel gating by shifting the current-voltage relation- been found to reduce surface expression of KCNQ1 and
ship of IKs in the depolarizing direction by 30 mV, thereby increase retention in the endoplasmic reticulum (98). This
destabilizing channel opening (391). region appears to be a conserved trafficking determinant
across the KCNQ family. In particular, Y111C and L114P
In addition to those in the pore domain, a number of LQT1 disrupt SKG1’s ability to increase channel trafficking to the
mutations that alter channel gating are found on the volt-
plasma membrane, suggesting that the NH2 terminus may
age-sensing S4 helix of KCNQ1. For example, the mutation
be important in RAB-mediated exocytosis of channels
S225L exerts dominant negative suppression of WT IKs
(375).
current (52). This mutation alters channel gating, shift-
ing the current-voltage relationship of IKs toward more
D) DEFECT IN PKA-MEDIATED SIGNALING. Given the critical im-
depolarized membrane potentials (170). The effect of this
mutation on voltage sensor movement remains to be de- portance of adrenergic stimulation and PKA-mediated sig-
termined, but given its location it is possible that it dis- naling in the regulation of IKs and repolarization of the
rupts the movement of the S4 helix in response to changes cardiac action potential during stress, disruption of this
in voltage. Another LQT1-associated mutation on S4, stimulation is expected to prolong the QT interval. For
R231C, decreases peak IKs amplitude, but it also leads to example, the mutation G589D is thought to disrupt a leu-
constitutive activation at the same time (41). Interest- cine zipper motif to which AKAP-9 binds, resulting in fail-
ingly, in one family, this mutation causes familial atrial ure of IKs to be stimulated by cAMP (255). A mutation on
fibrillation, which is more consistent with action poten- S6, A341V, exhibits dominant suppression of IKs that fails
tial shortening than prolongation (41, 296). To explain to respond to adrenergic stimulation with cAMP (169).
this finding, the study has used a computational model to This effect is mediated by a reduction in the phosphoryla-
show that the atrial action potential is more susceptible tion S27 on KCNQ1 and is not due to disruption in AKAP-
to shortening than ventricular action potential when IKs 9’s interaction with KCNQ1. This result implicates a role of

96 Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org


CONGENITAL LONG QT SYNDROME

S6 in the regulation of the phosphorylation state of brane. These results are consistent with the role calmodulin
KCNQ1. plays in channel assembly and trafficking (141, 379).

Mutations that lead to defective adrenergic stimulation of H) NON-MISSENSE MUTATIONS. Non-missense mutations can
IKs in LQT1 patients are suggested to be associated with also cause LQT1. For example, nonsense LQT1 mutations
higher risk of cardiac events during exercise and greater such as R518X and Q530X introduce a stop codon, leading
response to ␤-blocker therapy. One study has identified to early termination of channel transcription and loss of IKs
missense mutations in the cytoplasmic loops between S2/S3 function (181). These mutations are mostly associated with
and S4/S5 of KCNQ1 to be associated with an elevated risk autosomal recessive LQTS, although autosomal dominant
of aborted cardiac arrest and sudden cardiac death (39). cases have also been reported for R518X. One study shows
Four mutations in these cytoplasmic loops, G189R, that these mutant channels only mildly affect WT IKs cur-
R190Q, R243C, and V254M, all diminish IKs upregulation rent, consistent with their mostly recessive mode of inheri-
in response to forskolin, an activator of the PKA signaling tance (349). It is thought that the nonsense transcripts are
pathway, suggesting that patients with these mutations are selectively degraded and do not interfere with WT channel
expected to be especially susceptible to arrhythmic events production. Interestingly, the same study suggests that

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


during stress. This study bears implications on better risk- LQT1 patients with nonsense mutations have reduced risk
stratification for LQT1 patients and predicting response to of cardiac events compared with patients with missense
␤-blocker therapy. In addition, it suggests that the cytoplas- noncytoplasmic loop mutations, although the explanation
mic loops are involved in adrenergic stimulation of remains to be determined.
KCNQ1.
Another non-missense LQT1 mutation is the deletion-inser-
E) DISRUPTED KCNQ1-KCNE1 INTERACTION. A number of LQT1 tion at residue 544, denoted ⌬544, which leads to a frame-
mutations disrupt the KCNQ1-KCNE1 interaction. Two shift that alters subsequent 107 amino acids and introduces
such mutations, S546L and K557E, are located in the helix an early stop codon (81). It is an autosomal recessive mu-
C of the CTD of KCNQ1 and disrupt its interaction with tation occurring in the CTD. The mutation has been shown
the COOH terminus of KCNE1, as demonstrated by GST to disrupt channel assembly in vitro (364), underscoring the
pulldown assays (110). The same mutations also decrease role of the CTD of KCNQ1 in channel assembly.
channel affinity for PIP2, leading to decreased current am-
plitude and a depolarizing shift in the current-voltage rela- LQT1 mutations can also disrupt transcript splicing. A base
tionship. The decreased PIP2 affinity may be due to disrup- substitution at a consensus splice donor site the end of exon
tion of a potential PIP2 binding site on helix C. 7, 1032G⬎A, can lead to a dropped exon 7 or exons 7 and
8 (286). In terms of channel regions affected, exon 7 en-
F) DECREASED PIP2 AFFINITY. Some LQT1 mutations have been
codes parts of the pore-loop and S6, while exon 8 encodes
shown to decrease channel PIP2 affinity. In addition to the the rest of S6 and part of the CTD. Transcripts lacking one
mutations described above, two LQT1 mutations on helix or both of these exons therefore are not expected to produce
C of the CTD, R539W and R555C, both lead to decreased functioning channels. A study shows that this splicing mu-
PIP2 affinity, decreased IKs amplitude, and a depolarizing tation exerts a dominant-negative suppression of WT IKs,
shift in the current-voltage relationship, underscoring the likely through a direct interaction between mutant and WT
importance of the CTD in PIP2 binding (312). The cytoplas- channels preventing trafficking to the membrane (428). In
mic loops of KCNQ1 between S2/S3 and S4/S5 also play addition to splice donor sites, splice acceptor sites can also
important roles in PIP2 binding and channel gating (208, be mutated in LQT1, such as the base substitution 922-1
498). Several LQT1-linked residues in these cytoplasmic
G¡C at the end of intron 6, which leads to the loss of exons
loops, including residues 195, 258, and 259, are thought to
7 and 8 (286).
form a binding site for PIP2. Disease mutations in this bind-
ing site may result in decreased PIP2 affinity, leading to
altered channel function (80, 498). 2. LQT5

G) DECREASED CALMODULIN AFFINITY. A number of LQT1 mu- Coassembly of KCNQ1 with the KCNE1 subunit is key to
tations have been found to weaken calmodulin binding to the generation of the IKs current. Thus it follows that
KCNQ1 as the underlying mechanism of disease. For ex- KCNE1 mutations can alter the physiologically critical cur-
ample, the mutations S373P and W392R, located in the rent of the assembled channel and lead to LQT5. Similar to
CTD, reduce calmodulin binding both in the absence and LQT1, the mode of inheritance for LQT5 can be autosomal
presence of KCNE1 (379). Both of these mutations cause dominant (RW) or recessive (JLNS) (107, 430). Mutations
decrease in surface channel expression and dramatic reduc- in KCNE1 can lead to defects in gating, trafficking,
tion in IKs amplitude. Overexpression of calmodulin is able KCNQ1-KCNE1 interaction, as well as adrenergic stimu-
to increase S373P mutant channel expression in the mem- lation (FIGURE 4D) (TABLE 3).

Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org 97


BOHNEN ET AL.

A) GATING DEFECT. One LQT5 mutation that affects channel interaction (166). Furthermore, the mutation P127T, lo-
gating is D76N. It is an autosomal dominant mutation in cated in the COOH-terminal region of KCNE1, appears to
the COOH terminus of KCNE1 that has been shown to disrupt the interaction of KCNE1 with helix C in the CTD
drastically suppress IKs amplitude, accelerate deactivation, of KCNQ1 (110). Interestingly, the mutation was also
and cause a depolarizing shift in the voltage dependence of found to diminish PKA-stimulated upregulation of IKs by
IKs activation when expressed in Xenopus oocytes and decreasing phosphorylation at the S27 residue. Since PKA
CHO cells (76, 405). Overexpression of the mutant KCNE1 phosphorylation at this site was previously shown to be
in guinea pig ventricular myocytes leads to APD prolonga- independent of KCNE1 (226), it is possible that the disrup-
tion and early afterdepolarizations (176). The mutant ap- tion in adrenergic stimulation by P127T is independent of
pears to traffic normally to the cell surface (53) and does not the mutation’s disruption of KCNQ1-KCNE1 interaction
disrupt KCNE1 binding to the COOH terminus of KCNQ1 (110).
(510). However, it reduces IKs upregulation secondary to
stimulation of the PKA signaling pathway, suggesting a role 3. LQT11
for the COOH terminus of KCNE1 in adrenergic stimula-
tion of IKs (226). AKAP-9 is a scaffolding protein and part of the IKs macro-

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


molecular complex that plays a critical role in the compart-
B) TRAFFICKING DEFECT. In addition to channel gating, LQT5 mentalization of adrenergic-stimulated PKA signaling path-
mutations can lead to defects in channel trafficking. The way leading to IKs upregulation. That a mutation in
JLNS mutation L51H, located in the transmembrane helix AKAP-9, S1570L, can cause LQTS is a testament to the
of KCNE1, results in diminished KCNE1 trafficking to the critical role of PKA signaling in the regulation of IKs mac-
cell surface (53). Furthermore, when coexpressed with romolecular complex (79). This mutation is located near
KCNQ1 in HEK cells, the mutant KCNE1 decreases the COOH-terminal binding domain of AKAP-9, disrupt-
KCNQ1 trafficking to the surface (53, 220). In addition, ing its interaction with KCNQ1, reducing cAMP-stimu-
coexpression of the mutant KCNE1 with KCNQ1 in CHO lated phosphorylation of KCNQ1, and abolishing IKs up-
cells leads to a diminished current amplitude and channel regulation in response to cAMP-mediated stimulation.
biophysical properties that resemble KCNQ1 alone rather Computational modeling suggests that a disruption in the
than IKs, consistent with a drastic reduction of functional basal phosphorylation state of IKs alone can alter IKs func-
KCNE1 in the membrane. These results together suggest tion sufficiently to prolong APD (79).
that the mutant KCNE1 interacts with KCNQ1 to disrupt
the trafficking of both proteins to the membrane surface,
leading to a reduction in current. The transmembrane re- E. Molecular Pharmacology
gion of KCNE1 may therefore be important to channel
trafficking. 1. ␤-Blockers

C) DISRUPTED KCNQ1-KCNE1 INTERACTION. LQT5 mutations ␤-Blockers have been demonstrated as a particularly effec-
can disrupt the interaction between KCNE1 and KCNQ1 tive form of therapy for LQT1 patients, who are more sen-
required to generate IKs. For example, the double mutation sitive to stress- and exercise-induced arrhythmia than other
T58P/L59P, located in the transmembrane region of LQT subtypes (282, 371). Insufficient upregulation of IKs
KCNE1, results in near-complete loss of IKs amplitude but by adrenergic stimulation to counterbalance concomitant
has minimal effect when coexpressed with WT IKs in Xeno- rise in inward calcium current is thought to underlie APD
pus oocytes (181). The mutation leads to a diminished abil- prolongation in LQT1 during sympathetic activation (386).
ity for KCNE1 to associate with KCNQ1 in coimmunopre- ␤-Blockers antagonize adrenergic receptors and helps pre-
cipitation studies, suggesting that the transmembrane re- vent this imbalance between potassium and calcium cur-
gion of KCNE1 is important for the KCNQ1-KCNE1 rents, decreasing predisposition for cardiac arrhythmic
events.

2. Channel activators
Table 3. Representative LQT5-associated mutations classified
by mechanism
While IKs plays a critical role in cardiac repolarization, it is
Reference not a direct target of drugs currently used to treat LQTS.
Mechanism Mutations Nos.
However, conceptually, IKs activators could allow for more
precise rescue of disease phenotype. In this section we
Gating D76N 76, 405
briefly review IKs activators and refer readers to other stud-
Trafficking L51H 53
ies in which activators of the ATP-sensitive K⫹ channel,
KCNQ1-KCNE1 interaction T58P/L59P 166, 180
such as nicorandil, have been used in LQTS patients and
P127T 110
model systems (14, 388). Currently there are no IKs activa-
PKA-mediated signaling D76N, P127T 110, 226
tors being used in clinical trials or therapy, but a number of

98 Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org


CONGENITAL LONG QT SYNDROME

small molecules that activate IKs have been identified at the 1), underlies congenital Long QT (LQT) syndromes type 2
benchside and may guide future development of therapeutic and 6, which arise from mutations in the KCNH2 and
agents. For example, the compounds DIDS and mefenamic KCNE2 genes, respectively. LQT2 is the second most com-
acid both increase IKs current amplitude (5). In addition, mon cause of congenital LQTS. KCNH2 mutations lead to
DIDS has been shown to drastically slow IKs deactivation. defective hERG protein, resulting in a decrease in IKr. Mu-
Interestingly, the effects of these drugs appear to be de- tations in KCNE2 cause defects in the KCNE2 (or MiRP1)
pendent on the presence of KCNE1. Compared with protein leading to LQT6, which also results in a decrease in
KCNQ1 alone, current augmentation by these com- IKr. Irrespective of the underlying cause, a decrease in IKr
pounds is much greater in the presence of KCNE1, sug- delays repolarization of the cardiac action potential pro-
gesting their effects are mediated by the ␤-subunit. In- longs the QT interval on the ECG, and predisposes patients
deed, deletion of the residues 39 – 43 of KCNE1 leads to to lethal arrhythmia. This section reviews IKr dysfunction
a diminished response to these compounds. To demon- leading to congenital and IKr-mediated drug-induced
strate the potential for activators as a class of therapeutic LQTS. [For a detailed summary of hERG channel structure,
agents for LQTS in in vitro studies, DIDS and mefenamic molecular biology, and basic electrophysiology, see Van-
acid have been shown to rescue IKs function in a LQT5 denberg et al. (436).]

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


mutation, D76N. Future studies will be required to better
understand their mechanisms of action and to develop
IKs-specific activators that can be effective and used B. Physiology
safely in patients.
Heterologous expression of hERG reveals a strong, near
Another KCNQ1 activator, ML277, has been shown to identical resemblance to IKr in cardiomyocytes (359, 427).
augment current for KCNQ1 alone more effectively than IKr is distinguished based on its relatively slow activation
KCNQ1 with KCNE1 (492). In fact, its activating effect and deactivation kinetics, combined with rapid inactivation
diminishes with progressive increase in KCNE1:KCNQ1 and recovery from inactivation (FIGURE 5). Inactivation re-
stoichiometry, suggesting that the ␤-subunit may act to pre- fers to a conformation of the channel protein in which the
clude the drug from binding to the channel (491). While a channels cannot conduct ions even if the activating machin-
drug with such properties is expected to have minimal ef- ery is in a conformation that would promote conduction of
fectiveness in human cardiomyocytes, surprisingly the same ions. Channels will not conduct ions when in an inacti-
study showed that the drug can augment IKs and shorten vated state, but will conduct ions after the channels re-
action potential in human iPSC-derived cardiomyocytes cover from the inactivated state, a recovery that takes
from a healthy control. The mechanism underlying this ef- place at negative voltages (174). Deactivation refers to
fect requires further elucidation. the transition to a conformation in which channels return
to a closed, nonconducting state, a transition that also
3. Channel blockers occurs at negative (diastolic) voltages. hERG channels
undergo voltage-dependent and C-type inactivation. Be-
cause channel activation is slow relative to the rapidly
IKs-specific blockers are not useful as therapeutic agents for
occurring inactivation process, the hERG I–V curve takes
LQT1, but serve as useful tools for research by allowing for
on a bell-shaped relationship, as shown in FIGURE 5C.
pharmacological dissection of IKs currents. Chromanol
Upon repolarization of the membrane, channels recover
293B is the first known IKs-specific blocker, with an IC50 of
from inactivation much faster than they deactivate. This
6.9 ␮M in Xenopus oocytes (62, 139). A more effective
crucial channel property results in a marked outward K⫹
blocker, HMR1556, with an IC50 of 0.12 ␮M, was de-
veloped using Chromanol 293B as the lead compound current during the repolarization phase of the cardiomy-
(139). These drugs inhibit KCNQ1 with KCNE1 more ocyte action potential. By this time, a large percentage of
effectively than KCNQ1 alone. Blockers of IKs are spec- hERG channels have recovered from inactivation, such
ulated to serve useful roles in the treatment of disease that outward potassium efflux helps return the cell to its
conditions resulting from IKs gain of function, such as resting potential, despite the fact that the electrochemical
familial atrial fibrillation. gradient for potassium efflux decreases as repolarization
progresses (FIGURE 1B).

III. IKr DYSFUNCTION IN CONGENITAL Since hERG inactivation and recovery from inactivation
LQTS proceed more rapidly than activation or deactivation of
hERG (365, 394, 401), IKr contributes to prolongation of
the plateau phase duration and thus cardiomyocyte con-
A. Introduction traction, in addition to cardiac repolarization (361, 394).
As hERG recovers from inactivation during cell repolariza-
Alterations in IKr, the rapid component of the delayed rec- tion, the repolarization itself promotes greater hERG recov-
tifier current in the cardiomyocyte action potential (FIGURE ery from inactivation due to the voltage dependence of

Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org 99


BOHNEN ET AL.

A KCNE2 hERG

Extracellular

S1 S2 S3 S4 S5 S6
+
+
+
+
FIGURE 5. hERG structure and electro-
Cytosol physiology. A: schematic of the IKr channel
complex. Four hERG1 subunits tetramer-
ize to comprise the pore-forming alpha

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


subunit of IKr. hERG1 contains a voltage-
cNBD sensing domain (purple), including the S4
helix which contains positively charged gat-
PASD
C ing residues, and a pore domain (gray).
KCNE2, an accessory subunit of the IKr
channel complex, consists of a single
N C transmembrane helix (blue). B: voltage-
clamp protocol (top panel) and heterolo-
gously expressed hERG1 ionic currents
B +50 mV C (bottom panel) recorded from a Xenopus
–70 mV oocyte. Currents were recorded at poten-
–80
tials that ranged from ⫺70 to ⫹50 mV;
deactivating (“tail”) currents were mea-
1.0
sured at ⫺70 mV. C: current-voltage (I-V)
relationship for hERG1 currents measured
0.8
at the end of test pulses, as indicated by
Relative current

red circle in B. D: voltage dependence of


0.6 hERG1 current activation. The peak of tail
currents measured at ⫺70 mV (indicated
0.4 by blue square in B) were normalized to the
largest value and plotted as a function of
0.5 μA 0.2 the test potential. E: voltage dependence
of hERG1 inactivation. Channel availability
0.0 is decreased at positive potentials, result-
–80 –60 –40 –20 0 20 40 ing in a decreased magnitude of peak out-
1 sec mV ward currents and the bell-shaped I-V rela-
tionship depicted in C. [B–E from Sangui-
D E netti (356), with permission of Springer.]
1.0 1.0

0.8 0.8
Channel availability
Relative current

0.6 0.6

0.4 0.4

0.2 0.2

0.0 0.0
–80 –60 –40 –20 0 20 40 –120 –80 –40 0 40
mV mV

hERG inactivation gating (359, 394, 427). As the cell con- URE 5E) acts to oppose cell depolarization (242, 394),
tinues to repolarize and return to its resting membrane po- which helps prevent premature heart beats from leading to
tential, the slower process of channel deactivation pro- an early action potential and a tachyarrhythmia. Loss of
gresses, leading to closure of hERG (436). The fraction of hERG function thus predisposes to arrhythmia in the set-
channels remaining open near the resting potential (see FIG- ting of premature beats(46).

100 Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org


CONGENITAL LONG QT SYNDROME

C. Molecular Biology hERG subunit (see below) (94). Furthermore, the PAS do-
main and cNBD appear to bind, working in concert to
The KCNH2 gene, located on chromosome 7q35-36, en- modulate hERG gating by positioning the NH2-terminal
coding the human ether a go-go-related K⫹ channel pro- residues in close proximity to the cytosolic side of S6 (23,
tein (hERG), was first discovered in 1994 (460). Muta- 100, 160, 287).
tions in KCNH2 associated with congenital LQTS were
discovered a year later in 1995 (97), and soon after, it hERG can coassemble with two different ␤-subunits in het-
was determined that hERG represents the ␣-subunits of erologous systems: KCNE1, encoded for by the KCNE1
the K⫹ channel responsible for IKr (359, 427). hERG may gene; and KCNE2 (or the MiRP1 protein), encoded for by
be referred to as “hERG1,” since other hERG proteins the KCNE2 gene (FIGURE 5A) (2, 259). KCNE1 and
(hERG2 and hERG3) have since been discovered (385). KCNE2 are single-pass transmembrane subunits that can
hERG1a, the main hERG isoform present in cardiomyo- interact with the hERG channel (2, 3, 20, 259). While the
cytes, is 1,159 amino acids in length, with a predicted molec- precise physiological role of the KCNEs in regulating hERG
ular weight of 127 kDa. As with most voltage-gated K⫹ chan- and IKr remains unclear (21, 257, 462), mutations in
nels, functional hERG channels are composed of four hERG KCNE1 and KCNE2 lead to LQT5 and LQT6, respectively,

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


␣-subunits, forming either a hERG1a homomeric channel, or and mutation of either KCNE1 or KCNE2 can predispose
a hERG1a/hERG1b heteromeric channel that contributes patients to drug-induced LQT syndrome, possibly via mod-
to native cardiac IKr activity. The heteromeric assembly of ulation of hERG activity (2, 9, 53, 295, 376, 402).
hERG isoforms 1a and 1b has distinctively altered kinetics
compared with hERG1a monomeric channels, including KCNE1 and hERG associate in heterologous systems and
faster activation and deactivation. hERG1b is expressed in may contribute to regulation of IKr; however, KCNE1 is
smaller amounts at the mRNA level in the heart compared better characterized physiologically as the ␤-subunit of the
with hERG1a, but nevertheless adds to the complexity of KCNQ1 complex that produces IKs (36, 53, 259, 358).
hERG channel regulation and potential therapeutic modal- KCNE2 was reported to alter gating properties and drug
ities (195, 196, 231, 323, 353, 426). responses of hERG channels(2), and mutations in KCNE2
leading to LQT6 likely result from changes in hERG and
Each of the four hERG ␣-subunits consists of six transmem- thus IKr current activity, producing a pro-arrhythmic
brane helices, S1–S6, as shown in FIGURE 5A. The voltage- state, further supported by the KCNE2 T10M mutation
sensing domain spans from S1 to S4; the S4 transmembrane that confers an arrhythmogenic substrate to auditory
segment of hERG contains the primary positively charged stimuli, a known trigger of LQT2-associated arrhythmia
amino acids required for voltage-sensing and opening of the (see FIGURE 7) (2, 151, 182, 251). It is possible that
activation gate (325, 408, 505). The pore domain of the physiologically relevant KCNE2 expression exists only in
channel, spanning from S5–S6, harbors the K⫹ permeation the Purkinje fibers and pacemaker cells of the human
pathway necessary for K⫹ conduction (FIGURE 5A) (105). heart (297, 330), which further confounds the exact role
The long cytoplasmic NH2 terminus contains the PAS do- of KCNE2 as a ␤-subunit of hERG in vivo. Moreover,
main with a PAS-cap, and together these sequences make up KCNE2 coassembles with KCNQ1, which decreases IKs
the “EAG” domain that is conserved among EAG-related current(192), adding to the complexity and diversity of
voltage-gated potassium channels(274). The long cytoplas- IKs and IKr channel subunit interactions.
mic COOH terminus of the channel contains the cyclic
nucleotide-binding domain (cNBD) (56), as well as a more
distal RXR endoplasmic reticulum retention signal (224), D. Molecular Pathophysiology
and a coiled-coil domain (188).
A decrease in IKr results in LQT2 via mutations in the
The NH2-terminal PAS domain accelerates deactivation of KCNH2 gene that encodes hERG, and LQT6 via mutations
hERG and plays a role in channel trafficking (77, 274, 436). in the KCNE2 gene that encodes KCNE2 (or MiRP1) pro-
Slow deactivation of hERG may involve interaction of the tein. The mechanisms underlying disease pathogenesis are
PAS domain in the NH2 terminus of the channel, with the described below.
S4 –S5 linker (450), which couples movement of the trans-
membrane voltage sensor to activation gate movement 1. LQT2
(247). Experimental hERG mutants with a deleted PAS do-
main have faster deactivation kinetics (274, 401). Proper Patients with congenital LQT2 often present clinically after
folding of the PAS domain also leads to trafficking of hERG syncope or seizures not explained by a known medical con-
from the endoplasmic reticulum to the plasma membrane dition. Prolongation of the QT interval on the ECG sup-
(314). The cNBD contributes to channel trafficking and ports the diagnosis, and in LQT2 in particular, the T-wave
gating as well, while cAMP binding to the cNBD domain may appear as a classic notched, or “bifid” T-wave (FIGURE
results in changes in the gating kinetics of the channel, 6A) (503). Syncope occurs following a common pathologi-
which are altered in the presence of KCNE2, an accessory cal process in the LQT syndromes, wherein delayed ventric-

Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org 101


BOHNEN ET AL.

A COOH terminus, including 8% in the c-NBD (207). In


LQT2 ECG terms of molecular mechanism, a small proportion of LQT2
mutations result in nonsense-mediated mRNA decay (150).
More commonly, decreased protein trafficking to the cell
surface leads to eventual endoplasmic reticulum-associated
degradation (ERAD) of mutant hERG (22, 149, 443, 512).

Several important general characteristics of KCNH2 mu-


tations associated with congenital LQT2 were recently
B highlighted in a large-scale functional study of 167 dif-
WT ferent LQT2-associated missense mutations (23). Func-
LQT2 tional analysis was performed by voltage clamp after
coexpression of WT and mutant subunits in HEK293
AP cells. Anderson et al. (23) found that 1) hERG trafficking
defects comprised the most common (88%) mechanism

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


of loss of function overall, derived from mutations in the
PAS domain, pore domain, and C-linker/cyclic nucleo-
tide-binding domain. Only the distal COOH-terminal re-
gion did not yield trafficking defects as the primary mech-
anism of loss-of-function. 2) Greater than 70% of pore
mutations led to dominant negative suppression of
IKr hERG, whereas the other intracellular domain mutation
locations did not yield dominant negative suppression of
current. 3) All mutant channels, regardless of mutation
location within the channel, were rescued pharmacolog-
ically by E4031, a hERG pore blocker that stabilizes
FIGURE 6. IKr dysfunction leading to congenital LQTS. A: ECG from channel structure and folding, thus promoting channel
a LQT2 patient demonstrates a characteristic “notched,” or bifid, T retention at the cell surface (148). Coexpression of WT
wave with QTc prolongation (unpublished data). B: simulated action with pore mutant channel rendered heteromeric pore mu-
potential (top) and IKr (bottom) in WT (black) and heterozygous LQT2 tant-WT channels especially responsive to pharmacolog-
(gray) conditions, demonstrating the effect of 50% reduction in IKr.
ical correction compared with coexpression of WT with
non-pore mutant channels. Overall, these data suggest
ular repolarization (and resultant QT interval prolongation that pharmacological recovery of hERG channel function
on the ECG) precipitates an EAD, in some cases DADs, and is feasible for a large proportion of LQT2 muta-
ventricular tachycardia. EADs trigger the torsades de tions(345) and raise the possibility of this approach as a
pointes sinusoidal waveform on the ECG, which may prog- therapeutic strategy for LQT-2 patients, but to our
ress to ventricular fibrillation and sudden cardiac death knowledge this has not been implemented in the clinic.
(356).
A) MUTATION SEVERITY: HAPLOTYPE INSUFFICIENCY VS. DOMINANT-

Homozygous mutations in KCNH2 leading to LQT2 are NEGATIVE LOSS OF FUNCTION. Heterozygous mutation result-
extremely rare in humans, resulting in either death in utero, ing in defective KCNH2 gene expression or hERG protein
or very severe prolongation of the QT interval upon birth that does not impact normal functioning of the WT
(175, 194). Heterozygous mutation leading to one defective hERG protein that remains yields haplotype insuffi-
hERG copy is far more common and may result in signifi- ciency. Only the gene product from the mutant KCNH2
cant prolongation of the cardiac action potential duration allele is negatively affected, while WT hERG subunits
(see FIGURE 6B). hERG mutations leading to LQT2 occur still homomerize to form functional channels. In con-
by a variety of genetic mechanisms. To date, ⬃500 muta- trast, some hERG mutations have increased pathogenic-
tions in KCNH2 have been identified in association with ity by exerting a dominant-negative loss of channel func-
LQT2 (23). A study analyzing 226 different LQT mutations tion, wherein the mutant and defective KCNH2 gene
in genotype-confirmed LQT2 patients reported that 62% of product reduces function of the WT hERG protein en-
mutations were missense, 24% were frameshift, while 14% coded in the patient’s genome via heteromerization of
were a combination of nonsense mutations, inframe inser- mutant with WT channel, rendering the healthy hERG
tions/deletions, or splice site mutants in the KCNH2 gene. subunits nonfunctional when combined with mutant
Of the combined 226 mutations, 32% resided in transmem- hERG, either by decreasing forward trafficking of mutant
brane and pore-pore domains; 29% in the NH2 terminus, WT channels, or decreasing function of the heteromers at
including 8% in the PAS/PAC domains; and 31% in the the cell surface. In a study of LQT2 patients with 44 unique

102 Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org


CONGENITAL LONG QT SYNDROME

KCNE2 hERG
G626A/D/V/S
N Y611H
G584S
Extracellular T10M

S1 S2 S3 S4 S5 S6
+ FIGURE 7. Topology of hERG and KCNE1
+ G628S in the plasma membrane, representative
+ LQT2- and LQT6-associated mutations
highlighted. Different mechanisms of loss
V65M + of function in hERG or KCNE2, including
gating (yellow-filled circle), K⫹ permeation
(black-filled square), trafficking (white-filled
Cytosol
triangle), or combined defects (green-filled
star) are categorized.

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


V822M
cNBD
PASD M124R R835W
C
D16A
N C

mutations in different regions of the hERG channel, it was tions, categorized by mechanism of loss of function (see
discovered that those patients harboring mutations in the TABLE 4). Representative LQT-associated hERG mutations
pore region of the channel were more susceptible to cardiac are divided into four general classes: I) decreased hERG
events than patients with non-pore region hERG mutations, synthesis, II) trafficking defect, III) gating defect, and IV)
likely due to a greater dominant-negative suppression of IKr
decreased K⫹ permeability (436).
current exerted by pore vs. non-pore mutations (187, 283).

Perhaps counterintuitively, more harmful mutations in one I) Decreased hERG synthesis. Defective biogenesis of
KCNH2 allele, resulting in a premature stop codon for hERG may occur via mRNA processing abnormalities or
instance, prevents formation of a hERG protein product mRNA instability (150, 504). The hERG R1014X mu-
from the mutant KCNH2 allele, leading to haplotype insuf- tant mRNA transcript is degraded by “nonsense-medi-
ficiency and the possibility of a less severe phenotype com- ated mRNA decay,” a cellular damage-control mecha-
pared with some dominant-negative mutations, as WT nism that destroys mRNA transcripts harboring non-
hERG remains unaffected(356). sense mutations (premature stop codons), which prevents
translation of a shortened hERG peptide (150, 356).
Studies of specific hERG mutants have greatly enhanced our Other nonsense-mediated decay mutants include
understanding of the underlying mechanisms of loss of W1001X (150) and Y652X (409). These mutations would
function leading to LQT2. FIGURE 7 provides a schematic of result in haplotype insufficiency, as there would be 50%
representative LQT-associated hERG and KCNE2 muta- loss of function while the remaining WT hERG channels

Table 4. Representative LQT2- and LQT6-associated mutations classified by mechanism


Mechanism Mutations Reference Nos.

hERG LQT2 mutations


K⫹ permeation G628S 59, 116, 357
Gating D16A, G584S, T613A, I711V, R835W 23, 329, 509
Trafficking Y611H, V822M 512
Decreased hERG synthesis R1014X, W1001X, Y652X 50, 150, 356, 409
Multiple mechanisms (e.g., gating and trafficking) F29L, M124R 23, 206, 390
KCNE2 LQT6 mutations
Gating T10M, V65M 151, 182

Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org 103


BOHNEN ET AL.

function normally. Nevertheless, a 50% reduction in IKr with a milder loss-of-function phenotype directly and solely
can lead to clinically significant LQTS. attributed to its effect on inactivation. This finding adds
further to the complexity of hERG mutant phenotypes as
II) hERG trafficking defects. Defective hERG trafficking is most other pore mutations cause increased arrhythmic
the most common mechanism of loss of hERG function (22, events compared with non-pore hERG mutations (283).
23, 436), and most KCNH2 missense mutations cause traf- Recently, the hERG T613A mutation in the outer region of
ficking defects in hERG (22). Trafficking mutants may re- the pore helix, a regulatory site for C-type inactivation, was
sult in either dominant negative loss of hERG function (22, found to cause greater than 80% inhibition of maximal
201) or haploinsufficiency (131). If WT hERG associates hERG current via a hyperpolarizing shift in channel inacti-
with trafficking-defective mutants to form heteromeric vation when expressed in Xenopus oocytes alone; coexpres-
channels in the endoplasmic reticulum (ER) or Golgi appa- sion of WT and mutant channel resulted in an intermediate
ratus, dominant negative suppression of IKr results: 93% loss of channel function, without dominant-negative sup-
reduction in IKr was observed by this mechanism (123). pression of current (329).

Defects in trafficking may be further subdivided by the un- Gating defects can arise from mutations across a wide range

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


derlying mechanism, protein trafficking, or protein misfold- of locations in hERG, and predispose individuals to LQT2
ing (22, 124, 512); hERG protein becomes core glycosy- of varying severity dependent on the functional impact of
lated in the ER, with modifications made in the Golgi ap- the mutation.
paratus. When the mature hERG protein traffics to the
plasma membrane, it weighs 155 kDa, versus 135 kDa for IV) hERG K⫹ permeation defect. The potassium selectivity
the core-glycosylated-only hERG protein. This difference in filter in the pore of the hERG channel allows for potassium
molecular weight aids in determining whether hERG pos- permeation with great specificity. Conserved amino acid
sesses a trafficking and/or protein folding defect (512). The residues comprise the selectivity filter, and mutation of any
hERG Y611H and hERG V822M LQT2 associated muta- of these residues or nearby amino acids may greatly alter
tions were found to have a molecular weight of 135 kDa, potassium permeation, even if only one of four hERG
and thus had decreased trafficking to the plasma membrane ␣-subunits within the mature channel bears the mutation
due to protein misfolding. These mutant hERG channels (i.e., dominant negative suppression of IKr). The hERG
were retained in the ER, and subsequently ubiquitinated G628S mutation, for instance, traffics to the cell surface and
and degraded in proteasomes (512). gates normally, which was confirmed by voltage-clamp flu-
orometry experimentation, but the mutant hERG channels
III) hERG gating defects. Alteration in activation, deacti- do not conduct potassium under physiological conditions
vation, and/or inactivation kinetics can result in loss of due to blockade of potassium permeation by intracellular
function of hERG and a decrease in IKr at physiologically potassium (116). The G628S mutation was also found to be
relevant membrane potentials. Any hERG mutation that, cause lethal arrhythmia in transgenic rabbits with the mu-
for instance, enhances the speed of inactivation, or causes a tation, as these rabbits possessed greater than 50% inci-
depolarizing shift in channel activation, ultimately leads to dence of sudden cardiac death after one year (59).
loss of hERG function at voltages ranging from the resting
potential to the plateau phase of the action potential, de- B) COMBINED MECHANISMS OF LOSS-OF-FUNCTION IN LQT2. Many
pending on the nature of the gating defect (48, 77, 290, 357, mutations leading to LQT2 confer a loss-of-function hERG
363, 509). Several hERG gating defect mutations have been phenotype by multiple molecular mechanisms. Kanters
described in mammalian cell lines (48, 363, 509). et al. (206) recently reported the mechanistic basis of chan-
nel dysfunction of the hERG F29L mutation in a founder
Both the I711V and R835W mutations represent examples population of Danish families, which results in a malignant
of c-NBD region LQT2 mutations in the COOH terminus form of LQT2. The mutation causes clinically significant
of the channel, with altered channel gating. These hERG prolonged QT, with a penetrance of 73%. The F29L muta-
mutants deactivate faster at ⫺50 mV compared with WT tion was found to 1) reduce trafficking of hERG to the cell
hERG. In addition, the R835W mutation confers a right- surface, posited to occur based on decreased glycosylation
ward, depolarizing shift (⫹16 mV) in the V0.5 of activation, of the hERG F29L channel; and 2) reduce steady-state in-
while inactivation measured at 0 mV remains unchanged activation current density, positively shift the voltage de-
(23). Within the NH2 terminus of hERG, the LQT2 hERG pendence of inactivation, and increase the rate of deactiva-
D16A mutation exhibits a ⫹13 mV rightward shift in the tion. Similarly, the M124R mutation in the NH2 terminus
V0.5 of activation, with a minor slowing of inactivation at 0 residing close to the hydrophobic surface of hERG, results
mV and a slowing of deactivation at ⫺50 mV (23, 302). in faster channel deactivation, in addition to reduced traf-
ficking to the cell surface (23, 390). Furthermore, the exact
The hERG G584S pore mutation increases the rate of chan- amino acid substitution can impact the mechanism of loss
nel inactivation (509). G584S represents a pore mutation of function of the hERG channel: the LQT2-associated

104 Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org


CONGENITAL LONG QT SYNDROME

G626A mutation in the conserved K⫹ selectivity filter in the E. Molecular Pharmacology


channel pore traffics to the cell surface normally, while the
LQT2-associated G626D, G626V, and G626S mutations hERG channel modulation by pharmacological agents has
had trafficking defects. The exact amino acid substitution become an increasingly studied research area because it is
has potential therapeutic implications: of the three G626 now well established that a wide variety of clinically used
trafficking mutants, only hERG G626S had its trafficking drugs block hERG and thus inhibit IKr, leading to drug-
defect corrected by E4031, an experimental pharmacologi- induced LQTS. The section below details mechanisms
cal agent that increases forward trafficking of hERG chan- and the biomedical relevance of drug-induced LQTS, fol-
nels in addition to its pore blocking effects (23). lowed by a discussion of various pharmacological tools
that hold promise as novel agents for the treatment of
2. LQT6 LQT2 (and LQT6) in the future.

KCNE2 (or MiRP1) protein, encoded for by the KCNE2 1. Drug-induced LQTS
gene, modulates hERG activity as an accessory ␤-subunit

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


(FIGURE 5A). Mutations in KCNE2 cause LQT6 due to a Most drugs that block IKr conduction bind to the open state
decrease in IKr (see FIGURE 7 AND TABLE 4) (2). The V65M of the hERG channel, binding the inner pore thereby pre-
mutation in KCNE2, for example, induces faster inactiva- venting permeation of potassium (67, 215, 267, 400, 483,
tion of colocalized hERG/mutant KCNE2 channel com- 513). The anti-arrhythmic pharmacological agent MK-499
plexes (182). The KCNE2 T10M was found linked to pa- interacts mostly with polar and/or aromatic residues within
tients with cardiac arrhythmia and LQTS, with particular the channel pore, including Thr623, Tyr652, and Phe656,
susceptibility to arrhythmia onset in the setting of auditory to induce hERG block (266). Different hERG-blocking
stimuli and hypokalemia, both common triggers of arrhyth- drugs may interact with different combinations of these
four residues (266, 319). For instance, the anti-arrhythmic
mia in LQT2 patients as well. Coexpression of KCNE2
agent dofetilide interacts with Phe656 to mediate hERG
T10M with hERG decreases tail currents and causes a hy-
binding (233). While the aromatic and polar residues above
perpolarizing shift in steady-state inactivation, along with
are integral to hERG’s supreme sensitivity to block by phar-
slowed recovery from inactivation compared with WT
macological agents, C-type inactivation may increase affin-
channels in CHO cells. The impact of the KCNE2 T10M
ity of drugs to hERG’s pore residues and enhance drug
mutation in the setting of known LQT2 arrhythmia triggers block (125, 171).
supports the hypothesis that KCNE2 associates with hERG
in the human heart to regulate IKr (151). As KCNE2 is Some drugs, including pentamidine (90, 228) and probucol
known to interact with other K⫹ ␣-subunits in addition to (156), cause a reduction in IKr by reducing trafficking of
hERG, defective KCNE2 may lead to varying phenotypes. hERG to the plasma membrane, leading to prolongation of
Indeed, the KCNE2 M54T mutation leads clinically to the QT interval. The widely clinically employed drugs flu-
LQT6 in addition to sinus bradycardia, the latter clinical oxetine and ketoconazole block hERG directly and de-
finding mediated by reduction of the HCN4 pacemaker crease hERG channel density at the cell surface (337, 411).
channel activity (297). The complexity of interactions of Several other pharmacological agents precipitate drug-in-
KCNE2 with a variety of potassium channels may inform duced LQTS in susceptible patients, via hERG blockade or
differential phenotypes observed clinically in those harbor- modulation of other important ionic currents (e.g., INa, IKs)
ing a KCNE2 mutation. (439, 482).

3. Autoimmune-associated LQT syndrome KCR1, a 12-pass transmembrane segment regulatory pro-


tein, interacts with hERG and mitigates the proarrhythmic
effects of hERG-blocking agents (178, 223). In heterolo-
Recently, Yue et al. (494) discovered that anti-Ro antibod- gous conditions, KCR1 decreases the sensitivity of hERG to
ies block the hERG channel, resulting in an autoimmune- a variety of well-established hERG channel blockers, such
associated LQTS. The anti-Ro antibodies, present in the as sotalol, quinidine, and dofetilide (223). Loss of function
serum of patients with a variety of common autoimmune of KCR1 is associated with greater risk of arrhythmia: the
illnesses, were found to inhibit IKr in HEK293 cells, and a KCR1 E33D mutant found in a patient with ventricular
guinea pig autoimmune model with circulating anti-Ro an- fibrillation and QT-interval prolongation conferred re-
tibodies had a prolonged QTc interval, caused by inhibition duced protection against drug-induced hERG blockade
of native IKr. Anti-Ro likely blocks hERG by binding within (168). The KCR1 I447V variant is associated with lower
the pore region of the channel. Patients with anti-Ro-posi- frequency of drug-induced arrhythmia, and reduced sensi-
tive autoimmune illness may be well advised to take partic- tivity hERG block by dofetilide when coexpressed heterolo-
ular caution to avoid medications that prolong the QT in- gously (322). These results suggest that functional KCR1
terval (494). provides protection against a drug-induced decrease in IKr,

Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org 105


BOHNEN ET AL.

with a mechanism of action thought to entail modulation of have been found, each having specific binding residues that
KCR1 ␣-1,2-glucosyltransferase activity (291). mediate their mechanism of hERG activation within the
“type 2” paradigm.
2. Drug screening for hERG toxicity
Other compounds have predominating mechanisms of
hERG activation that do not fall into the “type 1” or
Given the propensity for clinically relevant pharmacologi-
“type 2” classes. For instance, mallotoxin and KB130015
cal agents to block hERG, leading to drug-induced arrhyth-
primarily enhance hERG activation while shifting the
mia, it is now a required practice for drugs early in devel-
voltage dependence of activation to more negative mem-
opment to undergo hERG toxicity screens. High-through-
brane voltages, likely by binding within the channel pore
put screening techniques, including radioligand binding,
(140, 499). Some compounds, such as A935142 (407),
ion flux and fluorescence assays, and hERG-Lite which
have both “type 1” and “type 2” characteristics. As ac-
measures cell surface density of hERG, detect candidate
tivators bind hERG at various locations within the chan-
drugs with strong hERG-blocking and/or traffic-altering
nel, efforts to correlate binding location and channel stoi-
properties (436). The development of automated patch-
chiometry with mechanism of hERG activation by small
clamp electrophysiology enhances the throughput of data

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


molecule compounds will aid the design of more selective
acquisition tremendously, allowing for numerous hERG re-
and potent hERG channel activators by a variety of
cordings from different cells, applying different drugs and
mechanisms (476).
at different concentrations, to be collected simultaneously.
Automated patch-clamp technology is the best initial high-
throughput hERG toxicity drug screening method to date IV. INa DYSFUNCTION IN CONGENITAL
(163). A focus on hERG toxicity screening early in the drug LQTS
development process may save time, and prevent or mini-
mize adverse clinical outcomes due to inadvertent IKr block.
A. Introduction
3. Pharmacological treatment in LQT2
LQT3, LQT9, LQT10, and LQT12 account for 5–10% of
congenital LQTS (7) and are associated with gain-of-func-
␤-Blocker treatment represents first-line pharmacological tion mutations in the cardiac voltage-gated Na⫹ channel
therapy in LQT2 patients. For a more detailed discussion of complex of proteins. As such, patients diagnosed with Na⫹
␤-blocker treatment in congenital LQTS, refer to section VI channel-associated LQT often, though not exclusively,
of this review. have a unique risk profile for arrhythmia triggers and re-
quire different pharmacological approaches to disease man-
A growing interest in hERG activators has developed. Such agement than patients with LQT1 or LQT2 (370). Electro-
agents may be especially useful in the setting of congenital physiological experiments have elucidated the connection
LQT2 and/or drug-induced LQT syndromes. hERG activa- between aberrant function of single ion channels and the
tors may be subdivided into type 1 activators type 2 activa- unique clinical presentation of this set of diseases. In each
tors, based on mechanism of hERG activation (436). case, congenital mutations result in a failure of the cardiac
Na⫹ current (INa) to properly inactivate. However, even
Type 1 hERG activators include RPR260243, an experi- within the subset of Na⫹ channel-associated LQT, differ-
mental small molecule compound that slows hERG deacti- ences in mechanisms of pathological INa generation and risk
vation in a temperature- and voltage-dependent fashion as stratification in some patients makes LQT3 an important
its primary means of channel activation (204, 321). case study in the value of patient-specific medicine. Below
RPR260243 may slow deactivation via binding to the we will discuss the role of INa in the conduction of electrical
S4 –S5 linker and the cytoplasmic regions of the S5 and S6 impulses in the heart, mechanisms by which congenital mu-
hERG domains (321). tations may alter these currents, and advances in the discov-
ery of potent and selective pharmacological agents to treat
Type 2 hERG activators act primarily by slowing channel disease.
inactivation, and/or shifting the voltage dependence of in-
activation to more positive membrane voltages (69, 138,
153, 164). Type 2 activators may also have other relatively B. Physiology
minor effects on deactivation kinetics and voltage depen-
dence of activation, as well as single-channel open proba- In the heart, the majority of voltage-gated Na⫹ current is
bility (69, 138, 164, 165, 407, 511). Several type 2 hERG carried by the cardiac isoform of the voltage-gated Na⫹
activators have been described, including experimental channel, Nav1.5 (137). Upon membrane depolarization to a
compounds PD118057 (320), ICA105574 (28, 134, 263), threshold around ⫺50 mV, channels activate and allow the
and NS1643 (153, 155, 318), and key insights into their influx of Na⫹ down its electrochemical gradient. The rapid
mechanism of action alongside their hERG binding location influx of positive charge across the cell membrane drives the

106 Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org


CONGENITAL LONG QT SYNDROME

A B C
0nA 1.0
C

Normalized Current
O 0.8

0.6

30pA
0.4
2pA

40ms –10 50ms


10ms 0.2 500ms

1nA
–100 –100
0
10ms –140 –120 –100 –80 –60 –40 –20 0 20 40
Membrane Potential (mV)

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


D SCN4B SCN5A
N I II III IV
Extracellular
+ + + +
+ + + +
+ + + +
+ + + +

Cytosol

C IFM motif
Syntrophin-α PDZ binding
N domain
C

Dystrophin

FIGURE 8. Physiology and molecular biology of INa. A: consecutive recordings of single Na⫹ channels
recorded under cell-attached conditions in HEK293 cells transfected with Nav1.5 cDNA (unpublished data).
B: WT Na⫹ currents recorded under whole cell patch-clamp conditions and obtained by depolarizations from
⫺120 to ⫺30 mV (unpublished data). At high gain (inset), whole cell currents return nearly to 0 nA. C: voltage
dependence of activation (red-filled square) and inactivation (blue-filled circle) (unpublished data). D: topology of
Nav1.5 in the plasma membrane with accessory proteins implicated in LQTS.

membrane potential from its resting voltage to a peak of highly voltage dependent. The voltage dependence of inac-
⫹50 mV, near the Na⫹ equilibrium potential (FIGURE 1). tivation is independent of the voltage dependence of chan-
The rapid activation of Nav1.5 is therefore critical to main- nel activation, as inactivation may occur from either closed
taining the speed of cardiac conduction. This rapid depo- or opened states (66). Closed-state inactivation can be mea-
larization coordinates the stimulation of voltage-gated sured with subactivation threshold depolarizing pulses.
Ca2⫹ and K⫹ channels necessary for excitation-contraction When these pulses are sufficiently long to approach equilib-
coupling and, eventually, repolarization (299). rium, these experiments are referred to as “steady-state in-
activation” experiments. As the holding potential becomes
In a healthy human heart, the Na⫹ current is rapidly atten- more depolarized, channels enter a closed-inactivated state
uated in milliseconds as channels inactivate, a process that and are unavailable to open upon supra-activation thresh-
was first observed and defined in Hodgkin and Huxley’s old depolarization with a midpoint near ⫺70 mV (FIGURE
giant squid axon experiments (173). Inactivation is respon- 8C). Because this midpoint is in the range of the resting
sible for the refractory period in neurons and myocytes membrane potential in a ventricular myocyte (⫺85 mV),
during which the firing of new impulses is not possible. small changes in the V0.5 of steady-state inactivation caused
Similar properties are observed in Nav1.5, with inactivation by mutations or drugs, or small changes in resting mem-
progressing with a time to half peak current on the order of brane potential, can have a large impact on channel avail-
1 ms and a mean open channel duration of roughly 0.5 ms ability, Na⫹ current density, and conduction velocity (86).
(49) (FIGURE 8, A AND B).
The voltage dependence of inactivation from the open state
Na⫹ channel inactivation is a complex multistep process has been more difficult to dissect, as channel fast-inactiva-
that involves multiple channel components and is itself tion is a nonequilibrium process. The rate of decay of Na⫹

Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org 107


BOHNEN ET AL.

currents increases with increasing voltage over the physio- nature of eukaryotic voltage-gated Na⫹ channels allows the
logically relevant range, reaching half-inactivation in 1.5 voltage sensors to modulate different aspects of channel
ms at ⫺40 mV and 0.5 ms at ⫹20 mV (34). Recovery from gating. Mutagenesis (66, 384), cysteine accessibility (380,
inactivation has the opposite voltage dependence of the on- 381, 383), and voltage-clamp fluorometry (71, 72) have
set of inactivation: channels recover more rapidly at hyper- revealed distinct properties of each voltage sensor. Move-
polarized potentials, and very slowly at potentials more ments of the DIV voltage sensor have been shown to corre-
positive than activation threshold (approximately ⫺45 mV) late with the voltage dependence and kinetics of inactiva-
(508). The presence of multiple inactivation states is evident tion (71, 147), and DIV-S4 activation is necessary and suf-
in the time course of recovery from inactivation. Following ficient for inactivation gating (66). The putative docking
an initial depolarizing pulse to inactivate channels, channel site for the inactivation gate resides in the intracellular
recovery during a period at resting membrane potential fol- NH2-terminal loops of S4 in DIII and DIV, potentially
lows a biexponential time course, suggesting the existence bridging our understanding of voltage-sensing domains and
of two distinct populations of channels (508): one in the voltage-dependent gating transitions (268, 415).
fast-inactivated state and one in the slow-inactivated state.
1. Accessory proteins

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


C. Molecular Biology
In the heart, Nav1.5 is associated with several accessory
Nav1.5 is encoded by the gene SCN5A (452). Unlike the proteins that modulate its trafficking and biophysical prop-
canonical voltage-gated K⫹ channels, Nav1.5 is composed erties. Four genes that encode ␤-subunits of the voltage-
of a single polypeptide chain (137). The protein folds into gated Na⫹ channel have been identified, SCN1B through
four homologous asymmetric domains, each containing six 4B, and all are expressed in the heart (253, 342). These
transmembrane helices (S1–S6) (70) (FIGURE 8D). S1 ␤-subunits have an Ig (immunoglobulin)-like NH2-terminal
through S4 form a voltage-sensing unit in each domain. S4 domain, a single transmembrane helix, and a short cyto-
spans the membrane and contains several charged arginine plasmic COOH-terminal domain. While they are similar in
residues capable of “sensing” changes in membrane poten- structure, the ␤-subunits interact with the ␣-subunit
tial. Depolarization drives a conformational change in S4 through different mechanisms. ␤1 and ␤3 interact nonco-
that is transmitted to the pore-forming helices, S5 and S6. valently with the ␣-subunit, and ␤2 and ␤4 interact via
Along with the extracellular reentrant loop that connects covalent disulfide bond (490).
them, S5 and S6 from each domain form the central ion
conduction pathway (70). Immunohistochemical studies suggest that ␤-subunits play an
important role in subcellular localization of different channel
The intracellular segments of Nav1.5 have been shown to be isoforms (253). Nav1.5 predominantly colocalizes with the ␤2
critical to proper channel function. The linker between do- and ␤4 subunits, encoded by the gene SCN2B and SCN4B, in
mains III and IV contains the three amino acid isoleucine, the intercalated disks. ␤4 has no significant effect on Nav1.5
phenylalanine, methionine (“IFM”) motif, shown to be a activation or inactivation gating (490). Small amounts of ␤1
key molecular determinant of fast inactivation (313, 465). subunit were also found to colocalize with Nav1.5 (101, 211,
Upon channel opening, this motif has been proposed to 253). Unlike ␤4, ␤1 displays a dramatic functional effect on
bind to a hydrophobic cluster in the intracellular linkers inactivation of Nav1.5 currents expressed in tsA cells. ␤1, but
between S4 and S5 of domains III and IV (268, 415). Con- not ␤2, caused a significant positive shift in the V0.5 of steady-
sistent with these findings, disruption of the structure of the state inactivation (101, 211, 342).
DIII/DIV linker by proteolytic cleavage (434) or mutation
(47) can impair inactivation, and coexpression of a cytoso- Nav1.5 is also associated with several other structural and
lic peptide containing the IFM motif can potentiate inacti- signaling complexes in the cardiomyocyte. Caveolin and
vation (111). syntrophin are two such proteins that have been implicated
in functional perturbation of INa and Na⫹ channel-associ-
The intracellular COOH-terminal domain has been shown to ated LQT. The six known caveolin proteins are encoded by
play an important role in channel function. The COOH ter- three caveolin genes: CAV1, CAV2, and CAV3 (30). Cav-3
minus has a structured proximal domain and unstructured is the most abundant isoform in cardiac tissue where it is
distal region (91). It has been proposed that interactions be- associated with cytoplasmic signaling factors (396), is re-
tween the COOH terminus and DIII/DIV linker are critical in sponsible for the formation of lipid raft microdomains and
the stabilization of the inactivated state (285), and further- caveolae (218), and potentially plays a role in t-tubule or-
more that this interaction may be facilitated by calmodulin ganization (133). Cav-3 null mice lacked caveolae and had
(435). In fact, several LQT3-causing mutations have been abnormal t-tubule development in skeletal muscle. Bio-
identified in the COOH terminus (34, 343). chemical and microscopic techniques have revealed that
Nav1.5 colocalizes with Cav-3 in lipid rafts, which facilitate
Multiple techniques have been used to study the role of the increase in peak INa density in response to ␤-adrenergic
voltage-sensing units in channel function. The asymmetric stimulation (486).

108 Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org


CONGENITAL LONG QT SYNDROME

Caveolin and Nav1.5 also both associate with the cytoskel- underlie tissue-specific phenotypes (185), as well as ar-
etal dystrophin complex (106). Syntrophin-␣1 is a member rhythmia risk stratification (32). Two mechanisms have
of the dystrophin-associated protein complex and is the been established for the generation of persistent INaL, and
most abundantly expressed syntrophin in the heart. Syntro- an additional set of mechanisms has been proposed for
phins mediate the interaction of dystrophin with the PDZ- aberrant Na⫹ conduction in the absence of INaL. TABLE 5
binding domain on the COOH terminus of Nav1.5 (135). lists a set of Nav1.5 mutations known to perturb channel
Isolated cardiomyocytes from dystrophin knockout mice function by specific mechanisms.
showed markedly decreased INa density, and ECGs showed
significant QRS prolongation, consistent with an interac- A) CHANNEL BURSTING. Channel bursting is best exemplified
tion between the dystrophin-associated protein complex by the ⌬KPQ mutation, a deletion of amino acids 1505–
and ion channels in the heart. 1507 in the DIII/DIV linker near the IFM motif. ⌬KPQ was
first identified in 1995 (453) and characterized that same
year (47). Like WT currents, currents passed by single
D. Molecular Pathophysiology
⌬KPQ channels typically activate and inactivate quickly.
However, during a small fraction of depolarizing pulses,

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


1. LQT3
channels enter a slow gating mode, or “burst” mode, char-
acterized by a persistent noninactivating current.
Like other forms of LQT, the hallmark of LQT3 is a pro-
longation of the QT interval on the ECG arising from a Modal transitions have been shown to be highly dependent
prolongation of the ventricular action potential and delayed on inter-pulse duration and resting membrane potential.
ventricular repolarization. The discovery that mutations in INaL from ⌬KPQ channels in heterologous expression sys-
multiple genes can give rise to LQT has allowed for an
tems (289) is highly rate dependent, decreasing dramati-
improved understanding of the risk stratification for car-
cally with increased pulse frequency. In isolated transgenic
diac events in this set of patients (FIGURE 9, A–C). While
⌬KPQ mouse cardiomyocytes, changes in pacing rate
patients with K⫹ channel-associated LQT are generally at
caused pause-induced spontaneous depolarizations (127).
higher risk or cardiac events during exercise, patients with
This is consistent with a decrease in EAD frequency and
LQT3 have elevated risk at rest (370, 371). However, even
polymorphic VT during increased pacing in intact mouse
within the umbrella of LQT3 there is a range of triggers for
hearts (120), and explains the elevated risk of arrhythmia
arrhythmia that is not easily explained by a “one size fits
and sudden cardiac death at rest in human patients with
all” model, suggesting patient-specific differences in disease
LQT3 (370, 371).
mechanism (244).

LQT3 is a disease of impaired Nav1.5 inactivation. Failure The inverse dependence on interpulse duration suggests
of the channel to inactivate or remain inactivated during the that transitions between background and burst gating
plateau or repolarization phases of the ventricular action modes occur predominantly at rest. At higher resting poten-
potential allows for a depolarizing current sufficient to pro- tials, a larger fraction of channels exist in closed-inactivated
long the action potential, leaving patients susceptible to states, leaving fewer channels available to transition into
asynchronous EADs or DADs (184, 240). In most cases, bursting states. Indeed, INaL is smaller during test pulses
impaired inactivation is manifest as a sustained noninacti- from more depolarized holding potentials (185). Taken to-
vating inward current, or late current (INaL) (FIGURE 9D). gether, Clancy and Rudy (85) were able to develop a com-
Rather than inactivating completely, many disease-associ- putational model of ⌬KPQ function by allowing transitions
ated mutations cause a persistent INaL during prolonged between background and burst modes only when channels
depolarizations. It should be noted that INaL of smaller exist in the closed state. This model was able to reproduce
magnitude has also been detected in tissue isolated from single-channel and whole cell gating properties, as well as
normal hearts (432); thus in pathology some INaL may re- prolongation of the ventricular action potential and the
sult from the failure to control or modulate normal activity. occurrence of EADs.
Nevertheless, while currents through these channels still
provide the rapid influx of Na⫹ necessary for cardiac con- B) LATE REOPENING. Following the identification of modal
duction, at high gain one can observe INaL that amounts to transitions in ⌬KPQ channels, other SCN5a mutations
as little as 1% of the magnitude of the peak. Because the were identified in patients with LQT3 that cause INaL by a
membrane impedance during the plateau phase of the ac- different mechanism. At the whole cell level, the N1325S
tion potential is high, this small depolarizing current has a and R1644H mutations had markedly different inactiva-
substantial effect on AP duration. tion (109) and recovery from inactivation time courses
(448). Single-channel records revealed that these gating de-
The mechanisms by which gain-of-function Na⫹ currents fects were in fact not due to bursting channels, but to dis-
may arise have been studied in depth, and recent computa- persed channel reopening that occurred during prolonged
tional work suggests that these divergent mechanisms may depolarizations (109). Additional studies revealed that

Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org 109


BOHNEN ET AL.

A SCN5A
I II III E1295K IV
Extracellular
+ + + +
+ + + +
+ + + +
+ + + +

Cytosol I1768V
N1325S
Y1795C
ΔKPQ
N S1904L
C

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


B D
LQT3 ECG

WT
F1473C

1%
50ms
V5
2ms

C E
1.0

WT
F1473C
AP
Normalized Current

0.5
INa
–10
5s

–100
WT
0.0
LQT3
–90 –60 –30

Voltage (mV)
FIGURE 9. INa dysfunction leading to congenital LQTS. A: topology of Nav1.5 in the plasma membrane.
Several representative LQT3-associated mutations were selected because there is strong evidence that they
induce INaL by channel bursting (red-filled triangle) or by late reopening (red-filled circle), or prolong the action
potential without INaL (red-filled square). B: ECG from a LQT3 patient (unpublished data). C: simulated action
potential (top) and INa (bottom) in WT (green) and heterozygous LQT3 (purple) conditions. INa peak current was
truncated to enhance view of INaL. D: representative current recordings from WT (purple) and F1473C
(orange) Nav1.5 expressed in HEK293 cells at low and high gain (inset) elicited by depolarization to ⫺10 mV
(35). E: steady-state channel availability for WT (green triangle) and F1473C (purple square). [D and E from
Bankston et al. (35).]

110 Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org


CONGENITAL LONG QT SYNDROME

Changes in steady-state properties of the cardiac sodium


Table 5. Representative LQT3-associated mutations classified current have been proposed as mechanisms for AP prolon-
by mechanism of gain of function gation and LQT, even in the absence of sustained INaL dur-
Reference ing prolonged depolarizing steps (FIGURE 9E) (6, 344, 395,
Mechanism Mutations Nos. 461). These mutations cause depolarizing shifts in channel
availability such that the window current is either larger, or
INaL: bursting ⌬KPQ (1505–1507) 47 exists at more depolarized voltages where rectification of
Y1795C 243 K⫹ currents increases the relative contribution of INa to
INaL: late reopening R1644H 109 total membrane current. These patients are therefore more
N1325S 109 susceptible to drug-induced LQT, most commonly arising
G1631D 447 from block of hERG channels, less commonly from block of
S1904L 34 other K⫹ channels and drugs that potentiate inward cur-
R1623Q 202 rents, including INa (439, 482). Additionally, several dis-
INaL: cAMP-dependent D1790G 416 ease-causing mutations have been shown to induce depolar-
Window current E1295K 6 izing shifts in channel availability in addition to canonical

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


G1631D 447 INaL during prolonged depolarization (35, 448, 463).
F1483del 397
Nonequilibrium gating I1768V 87 D) NONEQUILIBRIUM GATING DEFECTS. In addition to changes in
steady-state parameters of channel activity, time-dependent
changes in gating kinetics have also been posited as a mech-
these late reopenings contribute to a fraction of the INaL anism for AP prolongation. As membrane potential declines
from ⌬KPQ channels, as well (73). into the repolarization phase of the AP, WT channels re-
cover slowly from inactivation. As membrane repolariza-
An additional mutation, S1904L, was later shown to also tion is more rapid than recovery from inactivation, WT
induce late reopenings and INaL (34). Interestingly, this mu- channels never recover at potentials that facilitate channel
tation does not reside in the DIII/DIV linker, but rather activation. Some LQT3 mutant channels, such as the
exists in the distal COOH-terminal domain. Transition I1768V mutation, have enhanced recovery from inactiva-
metal ion FRET showed that this mutation destabilized a tion and are able to recover at voltages above the channel
critical intramolecular interaction within the COOH-termi- activation threshold (87). Importantly, this nonequilibrium
nal domain (144), contributing to our understanding of the reopening is different than window current because it oc-
inactivation gate as a macromolecular complex of different curs at potentials at which activation and steady-state chan-
channel components including the DIII/DIV linker and nel availability do not overlap.
COOH terminus (285).
E) PHOSPHORYLATION-DEPENDENT INaL.There exists one known

A computational model has been constructed to simulate mutation, D1790G, for which in vitro studies found INaL
late reopening currents at the single-channel and whole cell only arises after PKA-dependent phosphorylation (416).
level (34). Function of the S1904L mutation could be sim- D1790G channels showed marked INaL in response to
ulated by reducing the rate of transition into the slow inac- cAMP-dependent PKA stimulation and lacked INaL under
tivated states, allowing channels to reside for a prolonged basal conditions. Furthermore, the D1790E mutation
time in fast inactivated states from which they may reopen. showed no response to cAMP application, suggesting that
An interesting prediction of this model is that the amplitude the negative charge at residue 1790 is important in modu-
of INaL will not display the inverse rate dependence present lating PKA phosphorylation. Interestingly, alanine mu-
in bursting channels, and in fact the patient experienced tagenesis of consensus PKA phosphorylation sites showed
arrhythmic events that were not limited to periods of rest. that S38 and S525 are important in mediating INaL in
D1790G channels, despite residing in disparate parts of the
C) WINDOW CURRENT. In WT channels, the voltage depen- channel. These effects were found despite the fact that most
dence of channel availability (V0.5 ⫽ ⫺71 mV) and channel spontaneous arrhythmic events in LQT3 patients occur
during rest or sleep, suggesting perhaps a role of basal phos-
activation (V0.5 ⫽ ⫺25 mV) overlap only slightly. There-
phorylation of the Nav1.5 channel in D1790G patients
fore, there is a very small range, or “window,” of voltages in
(371).
which channels may activate but not completely inactivate.
This range may provide a small noninactivating current
known as “window current.” Typically, this window cur- 2. LQT associated with mutations to accessory
rent is very small and only exists at voltages at which repo- proteins
larizing currents through delayed rectifier K⫹ channels are
strong, and therefore contribute little to net membrane cur- In addition to mutations in SCN5A, mutations in the acces-
rent. sory proteins important in cardiac INa have been associated

Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org 111


BOHNEN ET AL.

with LQT (11), although there are few documented cases of E. Molecular Pharmacology
such diseases.
The pharmacology of Na⫹ channel-associated LQT has
3. LQT10 been difficult to navigate because the diversity in patient
phenotypes (392, 397) and risk of on- and off-target effects
of standard antiarrhythmic therapies (276) has limited their
Mutations in the ␤4 subunit give rise to LQT10. The L179F
use clinically. As a result, genetically identified LQT3 pa-
mutation was identified in a 21-mo-old girl with bradycar-
tients are the most likely to receive implantable cardio-
dia and severe QT prolongation (261). When coexpressed
verter-defibrillators (ICDs) as a precautionary measure de-
with Nav1.5 in HEK cells, this mutation caused both a
spite previously having been asymptomatic (372). There is
dramatic INaL and a depolarizing shift in the V0.5 of steady-
therefore a large unmet medical need for effective pharma-
state inactivation that increased the window current. Inter-
cological intervention, and our understanding of Na⫹ chan-
estingly, this ␤-subunit mutation induced a larger INaL than
nel blockade and ␤-adrenergic receptor blockade in LQT3
the ⌬KPQ deletion in the ␣-subunit. Mutations in SCN4B
continues to evolve.
have also been associated with Sudden Infant Death Syn-
drome (SIDS) (413). The SIDS-associated S206L caused a

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


1. Local anesthetics
depolarizing shift in stead-state inactivation and a dramatic
increase in INaL. Recently, a mutation was reported in the
The utility of local anesthetics in the treatment of LQT3 is
␤1b subunit, a splice variant of ␤1, that caused a speeding
rooted in their ability to inhibit the voltage-gated Na⫹
of recovery from inactivation, significant INaL, APD prolon-
channel with preference for INaL (FIGURE 10). Soon after the
gation in HL-1 cells, and QT prolongation and stress-in-
discovery of the ⌬KPQ mutation and its role in LQT3, it
duced syncope in the patient (342). This was the first report
was reported that the commonly prescribed local anesthetic
of a mutation in ␤1b associated with LQT.
and class 1b antiarrhythmic agent lidocaine inhibited INaL
more effectively than peak current (19). This phenomenon
4. LQT9 and LQT12 has been observed in block of several mutations by other
clinically relevant local anesthetic-like drugs including
LQT9 is a Na⫹ channel-associated arrhythmia arising from mexiletine (35, 258), flecainide (288), and ranolazine (127),
mutations in Cav3, the gene encoding Cav-3, a cardiac iso- as well as recent experimental drugs GS967 (43) and elecla-
form of caveolin (30). Four Cav-3 mutations were identified zine (128, 200), making them useful in normalizing the
in a sample of 905 LQTS patients (438). Sixty-seven percent QT-interval (130, 281, 284, 370) and preventing arrhyth-
of patients identified with mutations in Cav3 exhibited syn- mic events in some patients (347). This approach has re-
cope or cardiac arrest during periods of rest, consistent with cently been supported by a study of patients with a wide
the risk profile for patients with mutations directly in the range of LQT3 mutations in which a robust improvement in
␣-subunit, Nav1.5. Two mutations, F97C and S141R, were QT interval and a reduction in arrhythmic events was in-
studied further by coexpression in HEK-293 cells with duced by oral mexiletine (258). Because of the efficacy of
Nav1.5. The S141R mutation sped recovery from slow in- these drugs in treating LQT3 patients and because of a
activation, and both mutations caused a significant INaL. common underlying mechanism, preferential inhibition of
Consistent with the functional importance of the Cav-3/ late versus peak Na⫹ channel current, development of more
Nav1.5 interaction, three additional mutations in Cav3 selective and effective blockers of INaL is underway by the
have been shown to induce INaL and are associated with pharmaceutical community. Nevertheless, it is important to
SIDS (92). recognize and understand the mechanisms underlying inhi-
bition of this current.
In mouse skeletal muscle, knockout of Cav-3 caused a dis-
ruption of the dystrophin complex in lipid rafts (133). Per- Local anesthetic-like drugs bind to a conserved phenylala-
haps not surprisingly, mutations in syntrophin-␣, a member nine (127, 336) in the central cavity of the channel in a
of the dystrophin protein complex, are also associated with highly state-specific manner. This specificity is imparted
LQTS and give rise to variant 12 of the disease (473). Two both by state-dependent changes in drug access to the bind-
mutations in SNTA1, the gene encoding syntrophin-␣1 have ing site (243, 336) and by allosteric modulation of channel
been identified. The A257G mutation nearly doubled the gating (162). Clinically relevant channel blockers have been
peak current density, increased window current, and sped shown to bind preferentially either to inactivated states, as
recovery from fast inactivation, although it did not induce is the case for lidocaine, flecainide, and mexiletine, and, in
INaL relative to peak current density in isolated neonatal rat the case of ranolazine, to open channels (275).
cardiomyocytes (473). It is likely, however, that absolute
INaL was greater in A257G myocytes because of the increase The state dependence of drug binding has been assayed by
in channel expression. The A390V mutation does cause a electrophysiological as well as biochemical experiments. Li-
persistent INaL and a depolarizing shift in steady-state inac- docaine induces a hyperpolarizing shift in steady-state inac-
tivation in a nitric oxide synthase-dependent manner (431). tivation(202), slows recovery from inactivation, and stabi-

112 Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org


CONGENITAL LONG QT SYNDROME

A B
F1473C

Mexiletine Ranolazine

1%
50ms

2ms

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


C D
* * *
Flecainide 80

60

Percent Block
40

20

0
Mexiletine Ranolazine Flecainide
FIGURE 10. Representative recordings at low and high (insets) gain of F1473C inhibition by 50 ␮M
mexiletine (A), 50 ␮M ranolazine (B), and 10 ␮M flecainide (C). D: mexiletine, ranolazine, and flecainide inhibit
INa with preference for INaL. [From Bankston et al. (35).]

lizes voltage-sensing units shown to provide the rate-limit- in patients suspected to have loss-of-function Na⫹ chan-
ing step in fast inactivation (27, 66, 382). Lidocaine has also nel mutations in Brugada Syndrome because of its ability
been studied at the single-channel level. In cell-attached to reduce peak INa and unmask latent disease phenotypes
patches, lidocaine reduced the propensity of ⌬KPQ chan- in a clinical setting (58). It has therefore been proposed
nels to burst, increased the number of depolarizing sweeps that an understanding of channel pathology and drug
with no channel openings, and had no effect on the mean properties may help guide a pharmacogenomic approach
open time of background or burst openings (31, 108). to disease management (88).
Taken together, these findings are consistent with stabiliza-
tion or promotion of inactivated channels. Ranolazine, on
2. ␤-Blockers
the other hand, has no effect on steady-state inactivation
and has therefore been interpreted as an open-channel
blocker (275). There remains some debate over the clinical utility of
␤-blockers in treatment of LQT3. A mainstay therapy for
It is therefore not surprising that mutations that alter patients with QT prolongation, clinical data suggest that
channel gating by different mechanisms will produce prophylactic ␤-blocker therapy in patients with LQT3 may
unique allosteric effects on drug binding. For example, be less effective at preventing cardiac events as it is in K⫹
flecainide (243) and mexiletine (347) have been shown to channel-associated LQT (282, 331). Interestingly, however,
more potently inhibit some mutations than others when ␤-blocker therapy did not show evidence of arrhythmia
expressed in heterologous systems. Mutation-specific induction, as might be expected from the heart rate-slowing
variability is therefore a major hurdle in disease manage- effects of ␤-blockade in LQT3 patients with elevated risk of
ment (88), as heightened sensitivity to Na⫹ channel arrhythmia at rest. This has prompted further investigation
blocking drugs can be pro-arrhythmic in certain settings into the mechanism of ␤-blocker activity in models of
(112, 276). In fact, flecainide is a useful diagnostic agent LQT3.

Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org 113


BOHNEN ET AL.

Experiments in animal models of LQT3 (64, 333) showed a ankyrin-B (271). These mutations in general lead to disrup-
protective effect of sympathetic stimulation against APD tion of normal targeting and regulation of transporters as
prolongation and torsades de pointes (387), suggesting a well as ion channels, leading to disrupted calcium handling
deleterious effect of ␤-blockade. However, experiments in that contributes to arrhythmia (272, 273). However, vari-
heterologous expression systems have shown that ␤-block- ation in severity of cardiac phenotypes exists between dif-
ers have a direct effect on Na⫹ channels (33). ␤-Blockers ferent mutations and can be correlated to the degree of loss
exhibit preferential inhibition of INaL and bind to the local of function observed in vitro (271).
anesthetic binding site, and at high doses have a similar
pharmaceutical profile as local anesthetics.
B. LQT7: Andersen-Tawil Syndrome Type 1
Ahrens-Nicklas et al. (12) constructed a computational
Andersen-Tawil syndrome type 1 arises from LQT7 muta-
model of ⌬KPQ-expressing ventricular myocytes stimu-
tions, which are loss-of-function mutations in the KCNJ2
lated by the ␤-agonist isoproterenol and inhibited by the
gene (327). This gene encodes Kir2.1, an inward rectifier
␤-blocker propranolol. Isoproterenol shortened APD and
potassium channel that underlies the cardiac IK1 current
EADs, consistent with the inverse-rate dependence of INaL
(104, 248, 303, 496), which contributes to setting the myo-

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


and animal models of LQT3. At low doses, propranolol
cardial resting potential and aids terminal repolarization of
mitigated this effect and prolonged APD, but at higher
the action potential (264). Because the membrane potential
doses where propranolol inhibits Na⫹ currents, it provided
of cardiomyocytes is more positive than the potassium equi-
a beneficial reduction in APD. Taken together, the experi-
librium potential, the channel actually conducts an outward
mental and computational data suggest a potential benefit
current in physiological conductions. Kir2.1 expression has
of ␤-blockade in treatment of LQT3, but more robust
been demonstrated in both human atria and ventricles
blinded studies must be performed to better understand the
(458). The protein consists of two transmembrane helices, a
clinical relevance.
pore-loop, as well as an NH2- and COOH-terminal region.
Four Kir2.1 subunits may come together to form a func-
V. OTHER SUBTYPES OF CONGENITAL tional channel. It has been shown that PIP2 binding to
LQTS Kir2.1 plays an important role in channel activation (179,
410).
A. LQT4: Ankyrin-B Syndrome
Andersen-Tawil syndrome is an autosomal dominant, mul-
tisystemic disorder characterized by periodic paralysis, ven-
The correct targeting of ion channels and transporters to
tricular arrhythmias, as well as dysmorphic features includ-
their respective subcellular domains is critical for normal
ing low-set ears, hypertelorism, and hypoplastic mandible
physiological function. It was discovered that autosomal
(362, 417). Early reports of patients with Andersen-Tawil
dominant loss-of-function mutations in ANK2, which en-
syndrome described QT prolongation on ECG; however,
codes ankyrin-B, can cause LQTS (273, 366). Ankyrin-B is
most ECGs of patients with this disorder are characterized
an adaptor protein that functions in the cytoskeletal net-
by prominent U waves that merge with the T wave, and a
work to ensure proper localization and stabilization of ion
characteristic prolonged “Q-T-U” interval. The physiolog-
channels and transporters. Although ankyrin-B was origi-
ical basis for the U wave is not well understood, but the
nally characterized as a 220-kDa protein, multiple splice
large U waves in Andersen-Tawil syndrome may be the
variants exist (474). The protein is composed of a mem-
result of delayed after depolarization or tissue-dependent
brane-binding domain, a spectrin-binding domain, and a
differences in repolarization including the mid myocardium
regulatory domain (271). It has been demonstrated to play
and His-Purkinje system (317, 328, 502). Andersen-Tawil
important roles in the normal localization of the Na⫹/Ca2⫹
syndrome type 1 results from KCNJ2 mutations, almost all
exchanger, the Na⫹-K⫹-ATPase, as well as the IP3 receptor
of which are dominant negative (424, 425). Many of these
(IP3R) which mediates intracellular calcium release (95,
mutations, such as R218W and G300D, located in the
269, 270, 273, 429). In addition, ankyrin-B has been shown
COOH-terminal region, either decrease PIP2 affinity or af-
to regulate voltage-gated Na⫹ channels (74), ATP-gated K⫹
fect residues involved in PIP2 binding (103, 249, 398, 501).
channels (236), as well as ryanodine receptors (65) in the
Some disease mutations, such as ⌬312–314, lead to dimin-
heart.
ished channel trafficking to the cell surface (44). Disease-
associated mutations can also be found in the NH2-terminal
While a loss-of-function ankyrin-B mutation was initially
region as well as the channel pore (424).
described as LQT4, it was later discovered that mutations in
ankyrin-B are not always associated with QT prolongation
and may present with a variety of cardiac phenotypes in- C. LQT8: Timothy Syndrome
cluding bradycardia, sinus arrhythmia, as well as atrial fi-
brillation (96, 232, 271). Most of these mutations, such as Timothy syndrome is caused by gain-of-function muta-
E1425G, are located in or near the regulatory domain of tions in the CACNA1C gene, which encodes Cav1.2, the

114 Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org


CONGENITAL LONG QT SYNDROME

␣1C (pore-forming) subunit of a cardiac voltage-gated D. LQT13


calcium channel. The channel generates the L-type cal-
cium current, which allows calcium influx during the Mutations in the KCNJ5 gene can also cause LQTS. KCNJ5
plateau phase of the action potential, sustaining mem- encodes a G protein-coupled inwardly rectifying potassium
brane depolarization and leading to calcium release from channel, Kir3.4 (or GIRK4). Kir3.4 and another GIRK iso-
intracellular stores which triggers excitation-contraction form Kir3.1(GIRK1) may form homo- and heterotetramers
coupling in cardiomyocytes. The channel undergoes both to generate the IKACh current in the heart (99, 219, 222), a
calcium-dependent and voltage-dependent inactivation. potassium current that is stimulated by acetylcholine
Inactivation of calcium channels during the action poten- through a GPCR-mediated pathway which has been dem-
tial causes an imbalance between the inward calcium and onstrated to slow heart rate during parasympathetic stimu-
outward potassium currents, resulting in repolarization. lation (136, 467). Similar to other inward rectifier channels,
The ␣1C subunit consists of four homologous domains of Kir3.4 contains two transmembrane helices with a pore-
six transmembrane helices each (S1–S6), including a volt- loop in-between as well as an NH2- and COOH-terminal
age-sensing domain (S1–S4) and a pore domain (S5–S6), region. Activation of GPCR receptors by acetylcholine
similar to voltage-gated sodium channels. Different causes the G␤␥ subunit to activate Kir3.4, resulting in an

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


splice variants of the ␣1C subunit may be expressed in the outward K⫹ current and hyperpolarization of the mem-
heart (445). brane potential, which in pacemaker cells results in slowing
of heart rate (214, 341, 468).
Timothy syndrome is an extremely rare genetic multisys-
temic disorder that includes QT prolongation, syndactyly, The presence of IKACh has been best demonstrated in sino-
craniofacial abnormalities, congenital heart defects, as well atrial node, atrioventricular node, and atrial cells of the
as autism. All known Timothy syndrome mutations to date heart (225). While it has also been shown to be expressed in
are located at the COOH-terminal end of S6 helices in dif- the ventricles (42, 183, 217, 466, 485), its role there is not
ferent domains of ␣1C (54, 142, 403, 404). Most com- well understood. Identification of LQTS mutations on
monly, Timothy syndrome arises from the de novo missense KCNJ5, however, suggests a role for this current in control-
mutation G406R on exon 8a, which is present in a minor ling ventricular action potential (484). For example, the
cardiac splice variant of CACNA1C (404). This mutation disease mutation G387R, a conserved residue located at the
is located at the COOH-terminal end of the first S6 helix COOH-terminal region of Kir3.4, has been shown to cause
of the ␣1C subunit and dramatically disrupts voltage- autosomal dominant LQTS. The mutation has been shown
dependent inactivation of Cav1.2, leading to increased to reduce trafficking of both Kir3.4 as well as Kir3.1 to the
plasma membrane and decrease current amplitude. Interest-
calcium influx that can result in APD prolongation. Tim-
ingly, a number of these LQTS patients also have atrial
othy syndrome resulting from this mutation came to be
fibrillation, suggesting a role for Kir3.4 in electrical conduc-
known as the “classic” variant. Two patients were later
tion in both atria and ventricles.
found to have the mutations G406R and G402S, respec-
tively, on exon 8, the major splice variant in the heart
(403). Similar to the classic Timothy syndrome mutation, E. LQT14 and LQT15
these two “atypical” Timothy syndrome mutations also
disrupt channel inactivation. Interestingly, neither of the
Calmodulin is a ubiquitous calcium-binding protein that
two atypical Timothy syndrome patients had syndactyly,
critically mediates cellular calcium signaling. Calmodulin is
as opposed to 100% of patients with classic Timothy important not only for the calcium-dependent inactivation
syndrome, suggesting differential expression patterns of of L-type calcium channels and the inactivation of Nav1.5
the ␣1C splice variants. One Timothy syndrome muta- channels (435, 514), but also in the trafficking and assem-
tion, I1166T, shows minimal effect on Cav1.2 inactiva- bly of KCNQ1 (379). Three genes, CALM1-3, all encode
tion, but is thought to cause disease by increasing win- the identical calmodulin protein. For example, a recent
dow current, which occurs in voltage ranges where chan- study suggests that de novo mutations in CALM1 or
nels activate but do not completely inactivate (54). CALM2 are associated with severely prolonged QT inter-
Efforts are underway to develop mechanism-based ther- val, ventricular arrhythmias, as well as atrioventricular
apies. In a landmark study, it was found that roscovitine, block in infants (93). Located in calmodulin EF-motifs that
a cyclin-dependent kinase inhibitor, partially recovers directly bind calcium, these mutations result in a reduction
inactivation of mutant Cav1.2 channels (487, 488) and in calcium affinity that may lead to abnormalities in sodium
reduces APD in ventricular-like cardiomyocytes derived currents, potassium currents, as well as calcium dynamics in
from iPSC reprogrammed from the skin cells of a Timo- the heart. In addition to exhibiting prolonged QT, these
thy syndrome patient (489). The study provides a model patients show neurodevelopmental delay, consistent with
for future development of therapies using human iPSC- widespread calmodulin expression in different tissues
derived cardiomyocytes. (316).

Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org 115


BOHNEN ET AL.

VI. GENOTYPE-DRIVEN CLINICAL domains either directly or indirectly. Thus the location of
MANAGEMENT OF CONGENITAL LQTS the hERG mutation variably impacts susceptibility to com-
mon triggers in LQT2 patients.

A. Introduction In LQT3, rates of cardiac events are high, and risk is likely
related to the degree of channel dysfunction or late current.
Clinical management of congenital LQTS aims to minimize The canonical SCN5A ⌬KPQ mutation results in 2.4-fold
symptomatic arrhythmia and prevent life-threatening car- greater risk of experiencing cardiac events through age 40
diac events. As described in detail in the preceding sections, compared with the SCN5A D1790G mutation (244). Fur-
the three most common congenital Long QT syndromes thermore, while preliminary data suggest similar risk for
(LQT1 due to mutation in KCNQ1; LQT2 due to mutation mutations in the COOH-terminal versus transmembrane
in hERG; and LQT3 due to mutation in Nav1.5) result in region of SCN5A, a 2.5-fold risk of ACA and SCD is con-
lengthening of the APD in cardiomyocytes, leading to a ferred by mutations that cause both sodium late current and
prolonged QT interval on the ECG. window current, compared with mutations causing only
one such gain-of-function defect (38).

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


By consensus guidelines, management strategies in congen-
ital LQTS today are mostly uniform, regardless of geno- 2. Genotype-specific triggers of arrhythmia
type. However, given the remarkable strides made in under-
standing the molecular identity of LQTS subtypes, as well
The first recognition of the relevance of genotype in man-
as the specific impact of a wide range of LQT-associated
agement of LQTS occurred in the context of a seminal study
mutations, the ultimate hope is that understanding a pa-
linking molecular identity in LQTS with clinical features,
tient’s genotype and specific LQT mutation can lead to
establishing genotype-specific triggers of arrhythmic events
rational treatment considerations. This section reviews the
(371). Six hundred seventy symptomatic patients with
features of molecular physiology that may influence geno-
LQT1, LQT2, and LQT3 were followed, and strong differ-
type-specific strategies for clinical management.
ences arose in the trigger of arrhythmic events based on
patient genotype (371). LQT1 patients experienced 62% of
B. Risk Factors for Arrhythmogenesis their cardiac events during exercise, and swimming or div-
ing was found to be a particularly potent trigger of arrhyth-
1. Mutation location, type, and cardiac risk mia (10, 279). In contrast, LQT2 patients experienced the
highest percentage of arrhythmic events in the setting of
emotional arousal (43%), including sudden loud noises and
In LQT1, mutations in the transmembrane regions includ-
startle (e.g., alarm clock awaking the patient from sleep), or
ing cytoplasmic loop (C-loop) domains of KCNQ1 confers
other triggers of emotional stress and fear; 13% of LQT2
a twofold greater risk of cardiac events [defined as syncope,
patient triggers occurred in the setting of exercise. LQT3
aborted cardiac arrest (ACA), or sudden cardiac death
patients experienced the highest percentage of arrhythmic
(SCD)] compared with patients harboring mutations in the
events, 39%, at rest or during sleep (371). LQT1 patients,
COOH-terminal domain of the KCNQ1 protein (280). The
by extension, experience more cardiac events in the setting
C-loops are involved in sympathetic regulation of KCNQ1
of sympathetic nervous system stimulation induced by ex-
(256), and mutations in this region may underlie an in-
ercise than LQT2 or LQT3 patients. More generally, these
creased risk of cardiac events in these patients compared
findings highlight the varying pathophysiology of the LQT
with mutations in other regions of the channel due to the
subtypes, and the specific life-style modifications and treat-
substantial regulation of KCNQ1, and therefore IKs activ-
ment considerations dependent on genotype (described
ity, by sympathetic stimulation (39).
later in this section).
In LQT2, Shimizu et al. (389) reported that pore region
missense mutations, located in the S5-loop-S6 portion of 3. Role of heart rate in LQT variant susceptibility to
the hERG channel, confer greater risk of cardiac events than arrhythmia
non-pore mutations in hERG (389). Specific triggers of car-
diac events were further subcategorized in LQT2 based on The variable triggers of arrhythmia in congenital LQTS can
mutation location and age at the time of the cardiac event. be explained in part by the role of heart rate in arrhythmo-
A study of 634 LQT2 patients revealed that while patients genesis. Heart rate contributes greatly to action potential
with pore mutations in hERG confer a ⬎2-fold risk of car- and QT duration (298), and therefore impacts the most
diac events compared with individuals with non-pore mu- common forms of congenital LQT syndrome in a genotype-
tations overall, non-pore transmembrane domain muta- specific, and even mutation-specific manner.
tions specifically led to a 6.8-fold increase in exercise-in-
duced cardiac events (216), suggesting that adrenergic In LQT1, a higher basal heart rate increases risk of cardiac
pathways interact with hERG non-pore transmembrane events, providing some prognostic value, unlike in LQT2

116 Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org


CONGENITAL LONG QT SYNDROME

patients (40, 57, 373). Functional IKs current is pivotal to Given the diagnostic utility of stress testing, attempts at
normal QT adaptation, or shortening with heart rate. At using this test to guide genotype and mutation-specific risk
increased heart rates (and/or during ␤-adrenergic stimula- stratification have been made. Laksman et al. (230) tested
tion of the heart), IKs facilitates cardiac repolarization and the hypothesis that exercise testing can predict QT interval
shortening of the QT interval: IKs has slow deactivation response and effect of ␤-blockers in LQT1 patients with
kinetics, rendering the channel open for a longer period of mutations in the C-loops (which, as described above, rep-
time. Residual IKs channel activation from previous cardiac resent the site of final sympathetic regulation of KCNQ1).
stimulation can enhance repolarization in the setting of In this retrospective analysis, C-loop KCNQ1 missense mu-
short diastolic intervals (119). Indeed, a hallmark of LQT1 tations were not associated with an increased QTc interval
is the failure of the QT interval to adapt to increases in heart during exercise stress testing or response to ␤-blocker ther-
rate, as often seen during diagnostic exercise stress testing apy (230). Despite the known limitations of retrospective
(29, 230, 470). This feature of IKs molecular physiology analysis, this study highlights the need for caution in the use
may also explain the predilection toward arrhythmias dur- of exercise stress tests for genotype-specific risk stratifica-
ing swimming and diving in LQT1, which can produce a tion.
complex autonomically-driven “diving reflex” with abrupt

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


changes in heart rate. 5. Female gender and pregnancy as risk factors for
arrhythmogenesis
In contrast to LQT1 and LQT2, LQT3 patients experience
greater risk of arrhythmic events at slower heart rates (332, Female gender is a risk factor for developing torsades de
370). Bradycardia can produce prime conditions for pointes in LQTS (186, 234), and the baseline QTc interval
SCN5A channel dysfunction, in a mutation-dependent in healthy females is prolonged compared with males. Fur-
manner. For example, the gain-of-function late sodium cur- thermore, women are at increased risk of developing drug-
rent generated by channel “burst” gating in the canonical induced torsades de pointes as a consequence of hERG-
⌬KPQ deletion mutation has been shown to be strongly blocking medications (254). In men, testosterone levels cor-
attenuated with increases in pacing rate (289). The fraction relate with QTc interval duration (507). In a large study of
of channels available to enter bursting channel states is 1,166 LQT2 syndrome patients, followed through age 40,
larger at slower pacing frequency (371). The observation of ACA and SCD occurred significantly more frequently in
strong association of arrhythmic events with bradycardia females (26%) than in males (14%). With regard to loca-
has led to concerns about the use of ␤-blockers, which slow tion of the mutation, males with pore mutations had a
heart rate, in the LQT3 subtype (33). However, it is impor- greater than twofold risk of a first ACA or SCD compared
tant to note that ␤-blockers have multiple intracellular tar- with males with non-pore mutations. Pore mutations did
gets that may be protective against arrhythmia, and direct not increase risk of these life-threatening cardiac events in
effects of ␤-blockers on sodium channel function have been females (265).
reported (33). Additionally, some LQT3 mutations can ex-
ert their effect via mechanisms other than channel bursting, Women with LQTS are particularly prone to developing
and therefore can have different features of rate dependence arrhythmias in the peripartum period. Seth et al. (377) dem-
(34). For example, the LQT3 SCN5A S1904L mutation onstrated that the postpartum state (defined as 9 mo post-
triggers arrhythmia more often at faster heart rates (185). delivery) confers a 2.7-fold increased risk of a cardiac event
Biophysical characterization alongside paired computa- and a 4.1-fold increased risk of a life-threatening cardiac
tional studies demonstrates increased late channel reopen- event. The majority of these events did not occur during
ings at faster pacing frequencies with the S1904L mutation, labor and delivery. This risk further appears to be genotype
in direct contrast to ⌬KPQ and F1473C mutations (35, specific; the LQT2 genotype, in particular, conferred
185). greater risk of cardiac events postpartum, compared with
females with LQT1 and LQT3 genotypes (377). Changes in
4. Genotype-guided exercise testing sex hormones associated with menopause may also affect
risk of arrhythmia. Within five years leading to the onset of
Since changes in heart rate can affect propensity for devel- menopause, the odds ratio for a syncopal event for LQT2
opment of arrhythmia, exercise testing in congenital LQTS females is 3.4 compared with LQT1 females, and after
has evolved into an essential tool in the diagnostic workup menopause, this increases to 8.1 (60). In LQT2, females
of affected patients. Almost 40% of LQTS patients do not older than 15 years of age have a 3.7-fold increased risk of
have a prolonged QTc interval on an ECG at rest. Exercise suffering cardiac events compared with males, while there is
stress testing can aid in diagnosis and risk stratification of no significant difference in risk of cardiac events between
congenital LQTS, with particular utility in uncovering cases males and females younger than 15 years of age (495). A
of concealed LQTS that have otherwise gone undiagnosed study of 634 LQT2 patients revealed that females ⬎13
(177, 339). Exercise ECG during stress and recovery is a years of age were nine times more susceptible specifically to
particularly helpful tool in uncovering concealed LQT1 “arousal” triggers compared with males ⬎13 years of age
syndrome (177). (216).

Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org 117


BOHNEN ET AL.

These clinical observations suggest that sex hormones may specifically swimming and possibly competitive athletics.
impact propensity for arrhythmia formation in LQTS in a LQT2 patients would be well advised to limit loud star-
genotype-specific manner. Sex hormones are known to play tling noises in their surroundings, including alarm clocks
a role in cardiac ion channel regulation and arrhythmia. and telephones (38).
Estrogen has been shown to decrease IKr activity and pro-
long the QTc interval (227), and ␤-estradiol inhibits hERG 2. ␤-Blockers and targeting of the sympathetic
channel expression in cultured HEK293 cells (24). Proges- nervous system
terone modulates both IKs and L-type calcium channels,
leading to a shortened QTc interval (294). According to consensus guidelines (335), ␤-blocker treat-
ment carries a class I recommendation in the setting of a
Animal models, including canine, mouse, rat, rabbit, and clinical diagnosis of LQTS. ␤-Blockers reduce overall risk of
guinea pig, have been extensively employed to better under- cardiac events in children and adults, and treatment repre-
stand gender differences in cardiac electrophysiology (186), sents a class IIa indication in all individuals confirmed as
and strongly support the role for male sex hormones short- carriers of LQTS mutations (118).
ening the QT interval via enhanced IKr activity (129, 245,

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


477). In a LQT2 rabbit model, estradiol enhances risk of However, the efficacy of ␤-blockers is strongly genotype-
sudden cardiac death, while progesterone is protective specific. ␤-Blockers produce greatest symptom relief and
(307). In canines, females have prolonged APD50, APD70, cardiac event reduction in LQT1 patients, followed by
and APD90 values compared with males. Dofetilide, a LQT2 patients, and are least effective in LQT3 patients
hERG channel blocker, prolonged the APD90 of Purkinje (334, 440). High-risk LQTS patient populations, including
fibers to a greater extent in female canines compared with LQT1 males and LQT2 females, experienced 67 and 71%
males with rate dependence, and the canine studies demon- risk reduction with ␤-blocker treatment, respectively (145).
strate consistency of sex-related differences in cardiac repo- Along with genotypic considerations, mutation-specific
larization between humans and canines (4). Further re- benefits exist based on location of the mutation in the ion
search is needed to elucidate the mechanisms underlying sex channel. For instance, ␤-blocker treatment resulted in 88%
hormone and arrhythmia formation, and to possibly un- risk reduction of life-threatening cardiac events in LQT1
cover therapeutic avenues through sex hormone sensitive patients harboring cytoplasmic-loop missense mutations
pathways. compared with LQT1 patients harboring missense muta-
tions in regions outside of the cytoplasmic loop (39).
␤-Blockers confer trigger-specific benefits as well, reduc-
C. Genotype-Specific Management of ing the risk of exercise-induced cardiac events by 78 and
Congenital LQTS 71% in LQT1 and LQT2 patients, respectively. How-
ever, ␤-blockers were not protective against events trig-
Congenital LQTS represents a potentially lethal condition, gered by arousal or rest/sleep in LQT1 and LQT2 (146,
yet advances in device therapy, medical options, and surgi- 216). These findings are in line with the molecular phys-
cal techniques have aided in the clinical management of the iology of these mutations. In nondiseased conditions, up-
disorder (421). Today, most clinical management of LQTS regulation of repolarizing currents by ␤-adrenergic stim-
is uniform across subtypes and consists of targeting of the ulation can enhance repolarization and favorably adapt
sympathetic nervous system, through ␤-blocker therapy QT interval for the concomitant increase in heart rate.
and left cervicothoracic sympathetic denervation (LCSD) in However, in the setting of increased heart rates and
selected refractory cases. Primary and secondary prevention LQT1 (and in some cases LQT2) disease, delayed rectifier
of cardiac arrest with an implantable cardioverter-defibril- current is not available for cardiac repolarization. One
lator (ICD), alongside antiarrhythmic therapy, is consid- possible mechanism for ␤-blocker effect could be preven-
ered after weighing risks and benefits for an individual pa- tion of sudden and sustained increase in heart rates,
tient. which in itself is associated with arrhythmia formation in
these subtypes.
As more is uncovered regarding the molecular physiology of
these conditions, there is growing potential for tailored In LQT2 events associated with sudden noises and startle,
therapy to a given patient’s genotype and specific LQT- both adrenergic and other neural pathways may be trig-
causing mutation. gered. As such, ␤-adrenergic blockade in LQT2 may not
fully mitigate the risks. Although ␤-blockers remain first-
1. Lifestyle modification line medical treatment in LQT2, a life-threatening cardiac
event rate of 6 –7% while on therapy remains a serious
Given the striking association of cardiac events with geno- concern (334).
type-specific triggers, counseling should be given regard-
ing lifestyle modification. As several examples, LQT1 It is important to note that ␤-adrenergic stimulation affects
patients are counseled about exercise limitations, most multiple intracellular targets, including the pathways for

118 Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org


CONGENITAL LONG QT SYNDROME

intracellular calcium cycling that regulate L-type calcium settings and arrhythmic storm) and mexiletine (for suppres-
current, the ryanodine receptor, and sarcoplasmic reticu- sive therapy) are commonly clinically employed. Mexiletine
lum calcium uptake (18, 506). In LQTS, with impaired administration shortens the QT interval in LQT3 patients,
repolarizing currents (or enhanced depolarizing currents), and not in LQT2 patients (370). Mexiletine blocks mutant
␤-adrenergic stimulation can be pro-arrhythmic, regardless Nav1.5 channel function, diminishing the late sodium cur-
of the genotype producing the impaired repolarization. rent more than the peak sodium current by stabilizing the
␤-Blockers therefore are commonly prescribed to all LQTS inactivated state of the Nav1.5 channel (449). Ranolazine,
patients regardless of genotype. on the other hand, is thought to block Nav1.5 current via
open channel block (275). The utility of local anesthetic-
In selected cases, left cervicothoracic sympathetic denerva- like drugs is weighed against their off-target undesirable
tion (LCSD), which surgically interrupts adrenergic input to side effects. For example, flecainide inhibits peak INa and
the heart, can be considered. In a study of 147 high-risk has been reported to unmask Brugada syndrome-type pat-
LQTS patients, cardiac event rate dropped by 91% per terns on surface ECG (indeed, this class of agents is clini-
patient post-LCSD (369). When carried out at experienced cally utilized as a provocative challenge in the diagnosis of
centers, this treatment modality can be very effective in Brugada syndrome) (58). Various mechanisms of drug

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


refractory cases. block of Nav1.5 underlie, in part, the mutation-specific ef-
ficacy of LQT3 pharmacological agents (see sect. IV).
3. Other pharmacological treatments in LQT2
Pharmacological sodium channel blockade appears on the
⫹ surface to be a specific and targeted strategy against gain-
Treatment of LQT2 syndrome by increasing serum K con-
of-function mutations in the sodium channel, and in indi-
centration has been explored, as hERG conductance is
vidual cases does provide remarkable reduction in arrhyth-
modulated by changes in extracellular K⫹, and LQT2 pa-
mia burden. However, a mortality benefit for these agents in
tients can develop low body and/or serum potassium levels
LQT3 has yet to be established. Anti-arrhythmic therapy in
(436). IKr activity decreases in the setting of hypokalemia
LQT3, as with all LQT subtypes, must be considered care-
(359) and increases in the setting of hyperkalemia, due
fully after thorough review of all patient-specific factors,
mainly to extracellular K⫹ effects on inactivation gating
including mutation or genotype-specific risk, clinical vari-
(446, 454, 455).
ables (e.g., family history, age, gender), and overall treat-
ment plan (including role of ICD placement and/or LCSD)
As such, potassium supplementation and therapy with spi-
(143). Given the complexities of drug effect, these agents
ronolactone, a potassium-sparing diuretic, were tested as
are often employed empirically in LQTS, with close atten-
treatment options to enhance IKr activity, and each treat-
tion to clinical response.
ment significantly shortened the QTc interval (89, 117).
However, there was no evidence of a decrease in cardiac
events. Safety dilemmas have been raised, including hyper- D. Emerging Translational Strategies in
kalemia, with the use of spironolactone in LQT2 treatment Congenital LQTS
(38). Potential adverse effects of spironolactone may be
explained by the fact that its metabolism into canrenoic acid 1. Cellular models of LQTS
was actually shown to induce hERG blockade and reduce
channel activity (63).
The physiological significance of cardiac channelopathies
can be difficult to model at the cellular level because of the
4. Other pharmacological treatments in LQT3 complex interplay between several ionic currents in the car-
diomyocyte. While heterologous expression systems and
The efficacy of local anesthetic drugs in LQT3 treatment is animal models are invaluable tools in our understanding of
due to preferential inhibition of late sodium current (more disease phenotypes, they lack the genetic background and
than peak current). Limitation of the use of late sodium molecular biology of human cells. Induced pluripotent stem
current blockers is due, in part, to off-target drug effects as cell-derived cardiomyocytes (iPSC-CMs) represent a useful
well as to the relative selectivity of block of late versus peak platform for studying human cardiac ion channel function
sodium channel currents. The introduction of newer com- in health and disease in a cell system with human molecular
pounds with significantly higher selectivity for INaL block and physiological components, acquired from unaffected
offers great potential for more effective clinical manage- patients and/or those carrying mutations.
ment of LQT3. The need for improved pharmacotherapy is
supported by a high breakthrough cardiac event rate for In 2010, iPSC-CMs were generated from skin cells taken
LQT3 patients on ␤-blocker treatment, affecting as many as from two LQT1 patients with the autosomal dominant
15% of patients, generating a growing interest in INa-spe- KCNQ1 R190Q mutation (277). These cardiomyocytes
cific blocking agents (143). The use of the local anesthetic were spontaneously beating and classified as ventricular-,
class of sodium channel blockers lidocaine (during acute atrial-, or nodal-like based on cellular electrophysiology

Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org 119


BOHNEN ET AL.

and morphology. Compared with WT controls, LQT1 car- agents in the treatment of congenital LQTS. The experi-
diomyocytes demonstrate decreased IKs amplitude and pro- mental drug GS-967 preferentially inhibits late INa over
longed APD, consistent with LQT1 pathogenesis. More re- peak INa with considerable selectivity, and does so more
cently, Jouni et al. (199) derived iPSC-CMs from a LQT2 potently and efficaciously than flecainide and ranolazine,
patient harboring a hERG A561P mutation, to study the while also reducing arrhythmia burden in experimental rab-
electrophysiological consequences of hERG loss-of-func- bit cardiomyocytes (43). Computational modeling of GS-
tion in a patient-specific genetic background. Ventricular-, 967 suggests therapeutic benefit of late INa block by de-
atrial-, and nodal-like action potentials were recorded, and creasing arrhythmogenesis in the setting of LQTS (481).
a trafficking defect was characterized as the mechanism of Another experimental compound, F15845, selectively
loss of IKr, leading to a prolonged APD. Furthermore, a blocks late INa potently, preventing cardiac angina and ar-
study by Jones et al. (195) reported the use of mRNA silenc- rhythmia in animal models (324, 433). The benefits of any
ing in human iPSC-CMs to show that the hERG1b isoform selective late INa blocker must be weighed against the rela-
is a significant component of cardiac repolarization, and tive effects on peak INa, in addition to off-target effects that
that loss of hERG1b function provides a substrate for ar- include block of peak INa and IKr. Studies to date suggest
rhythmogenesis. Several additional studies have investi- that selective late INa blockers are not proarrhythmic; clin-

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


gated LQT3 in the context of iPSC-CMs (121, 252, 420). ical trials of these and related drugs are currently underway,
and it is expected that, in the near future, these studies will
In the context of drug development, iPSC-derived cardio- inform whether this drug class represents a viable treatment
myocytes provide a means for testing compounds in pa- option in congenital LQTS (25).
tient-specific genetic backgrounds, allowing for a targeted
approach in optimizing therapy. Terrenoire et al. (420) Along with more selective targeting of LQT-associated
studied human iPSC-CMs from a patient harboring a LQTS mutant ion channels, growing interest in both mutation-
mutation in SCN5A and a polymorphism in hERG. Upon specific and tissue-specific treatment strategies may be-
electrophysiological characterization of ionic currents, it come fruitful. For instance, ectopic rhythms leading to
was determined that the SCN5A F1473C mutation was ventricular fibrillation in LQT patients has been shown
responsible for the LQTS clinical phenotype via increased to originate 50% of the time from the Purkinje fiber cells
INaL, while the hERG K897T variant was a benign poly- in the cardiac specialized conduction system (158, 306,
morphism. Experiments in the iPSC-CMs revealed key rate- 414). The Purkinje system is especially prone to arrhyth-
dependent properties of INaL and its inhibition by mexile- mia formation, owing to differences in membrane current
tine; these results provided insight into control of arrhyth- density, APD and morphology, and calcium cycling (102,
mias in the proband. This study offers an example of how 132, 158). In a computational model of the His-Purkinje
iPSC-CMs can elucidate mechanisms of LQTS pathogenesis system, LQT3-associated mutations were shown to con-
to inform patient-specific therapy. fer a more severe phenotype in Purkinje fibers compared
with LQT1 and LQT2 mutations (185), predictions that
Use of iPSC-CMs is not without limitations. Discrepancies were confirmed in isolated murine Purkinje fiber cells
may still exist in the expression profile of cardiac ion chan- expressing ⌬KPQ mutant Na⫹ channels (175). Contin-
nels between iPSC-derived cardiomyocytes and native hu- ued research into the functional implications of muta-
man cardiac cells due in part to differences in cell maturity tions in different cell types may uncover further therapies
and growth environment. Cardiomyocyte differentiation directed at tissues most prone to arrhythmia formation.
can produce a mix of atrial-like and ventricular-like cells of For example, targeting the insertion of these Purkinje
varying morphology. Furthermore, recent publications us- fibers in the ventricular endocardium via catheter abla-
ing iPSC-CMs report a resting membrane potential depo- tion may prove to be an effective strategy in reducing
larized to near ⫺60 mV (252, 420), which can alter prop- arrhythmia burden (471).
erties of important cardiac channels such as Nav1.5 and
affect state-dependent pharmacological inhibition. Efforts
are underway to improve iPSC-CMs by using physiological VII. CONCLUSIONS
systems to promote cellular maturity such that they more
closely resemble native mature cardiomyocytes. As effi- The growing body of mechanistic insights into the patho-
ciency and specificity of cell differentiation improve, iPSC- physiology of channelopathies and other arrhythmia-caus-
CMs hold great promise as a model cell system for the study ing mutations has helped elucidate the molecular underpin-
of LQTS disease and patient-specific pharmacology (115). nings of congenital LQTS and genotype-phenotype correla-
tion in disease. Although insights from benchside to bedside
2. Pharmacological and medical approach to LQTS have facilitated progress toward better therapeutic strate-
gies, there remains a need for tailoring management toward
More specific targeting of the mechanisms underlying ion individuals in a genotype-driven and mechanism-specific
channel defects that predispose to arrhythmia guides the manner to optimize care. Techniques such as iPSC-derived
advancement and rational drug design of pharmacological cardiomyocytes and high-throughput patch clamping that

120 Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org


CONGENITAL LONG QT SYNDROME

allow for screening of compounds in patient-specific con- Schulze-Bahr E, Semsarian C, Towbin JA, Watkins H, Wilde A, Wolpert C, Zipes DP,
Heart Rhythm S, European Heart Rhythm Association. HRS/EHRA expert consensus
texts may help pave the way for future development of statement on the state of genetic testing for the channelopathies and cardiomyopa-
therapeutics. In addition, continued progress toward fun- thies: this document was developed as a partnership between the Heart Rhythm
damental understanding of mechanisms of ion channel Society (HRS) and the European Heart Rhythm Association (EHRA). Europace 13:
function and drug-channel interaction will guide the devel- 1077–1109, 2011.

opment of more effective, mechanism-based molecular 9. Ackerman MJ, Tester DJ, Jones GS, Will ML, Burrow CR, Curran ME. Ethnic differ-
agents in the treatment of LQTS. ences in cardiac potassium channel variants: implications for genetic susceptibility to
sudden cardiac death and genetic testing for congenital long QT syndrome. Mayo Clin
Proc 78: 1479 –1487, 2003.
ACKNOWLEDGMENTS
10. Ackerman MJ, Tester DJ, Porter CJ. Swimming, a gene-specific arrhythmogenic trig-
M. S. Bohnen, G. Peng, and S. H. Robey contributed ger for inherited long QT syndrome. Mayo Clin Proc 74: 1088 –1094, 1999.

equally to this work. 11. Adsit GS, Vaidyanathan R, Galler CM, Kyle JW, Makielski JC. Channelopathies from
mutations in the cardiac sodium channel protein complex. J Mol Cell Cardiol 61:
Present address of C. Terrenoire: The New York Stem Cell 34 – 43, 2013.
Foundation Research Institute, New York, NY 10032. 12. Ahrens-Nicklas RC, Clancy CE, Christini DJ. Re-evaluating the efficacy of ␤-adrener-

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


gic agonists and antagonists in long QT-3 syndrome through computational modelling.
Address for reprint requests and other correspondence: Cardiovasc Res 82: 439 – 447, 2009.
R. S. Kass, Dept. of Pharmacology, Columbia University 13. Aidery P, Kisselbach J, Schweizer PA, Becker R, Katus HA, Thomas D. Impaired ion
Medical Center, 630 W. 168th St., New York, NY 10032 channel function related to a common KCNQ1 mutation: implications for risk strati-
(e-mail: rsk20@cumc.columbia.edu). fication in long QT syndrome 1. Gene 511: 26 –33, 2012.

14. Aizawa Y, Uchiyama H, Yamaura M, Nakayama T, Arita M. Effects of the ATP-


GRANTS sensitive K channel opener nicorandil on the QT interval and the effective refractory
period in patients with congenital long QT syndrome. Investigator Group for K-Chan-
nel Openers and Arrhythmias. J Electrocardiol 31: 117–123, 1998.
This work was supported by the American Heart Associa-
tion and National Heart, Lung, and Blood Institute Grants 15. Alders M, Christiaans I. Long QT syndrome. In: Gene Reviews, edited by Pagon RA,
F30HL129656, 5K08HL116790, and 5R01HL123483. Adam MP, Ardinger HH, Wallace SE, Amemiya A, Bean LJH, Bird TD, Fong CT,
Mefford HC, Smith RJH, Stephens K. Seattle, WA: Univ. of Washington, 1993.

16. Allan WC, Timothy K, Vincent GM, Palomaki GE, Neveux LM, Haddow JE. Long QT
DISCLOSURES syndrome in children: the value of rate corrected QT interval and DNA analysis as
screening tests in the general population. J Med Screen 8: 173–177, 2001.
No conflicts of interest, financial or otherwise, are declared 17. Allan WC, Timothy K, Vincent GM, Palomaki GE, Neveux LM, Haddow JE. Long QT
by the authors. syndrome in children: the value of the rate corrected QT interval in children who
present with fainting. J Med Screen 8: 178 –182, 2001.

18. Amanfu RK, Saucerman JJ. Modeling the effects of beta1-adrenergic receptor blockers
REFERENCES and polymorphisms on cardiac myocyte Ca2⫹ handling. Mol Pharmacol 86: 222–230,
2014.
1. Abbott GW. Biology of the KCNQ1 potassium channel. New J Sci 2014: 1–26, 2014.
19. An RH, Bangalore R, Rosero SZ, Kass RS. Lidocaine block of LQT-3 mutant human
2. Abbott GW, Sesti F, Splawski I, Buck ME, Lehmann MH, Timothy KW, Keating MT, Na⫹ channels. Circ Res 79: 103–108, 1996.
Goldstein SA. MiRP1 forms IKr potassium channels with HERG and is associated with
cardiac arrhythmia. Cell 97: 175–187, 1999. 20. Anantharam A, Abbott GW. Does hERG coassemble with a beta subunit? Evidence for
roles of MinK and MiRP1. Novartis Found Symp 266: 100 –112, 2005.
3. Abbott GW, Xu X, Roepke TK. Impact of ancillary subunits on ventricular repolar-
ization. J Electrocardiol 40: S42– 46, 2007. 21. Anantharam A, Lewis A, Panaghie G, Gordon E, McCrossan ZA, Lerner DJ, Abbott
GW. RNA interference reveals that endogenous Xenopus MinK-related peptides gov-
4. Abi-Gerges N, Small BG, Lawrence CL, Hammond TG, Valentin JP, Pollard CE.
ern mammalian K⫹ channel function in oocyte expression studies. J Biol Chem 278:
Evidence for gender differences in electrophysiological properties of canine Purkinje
11739 –11745, 2003.
fibres. Br J Pharmacol 142: 1255–1264, 2004.
22. Anderson CL, Delisle BP, Anson BD, Kilby JA, Will ML, Tester DJ, Gong Q, Zhou Z,
5. Abitbol I, Peretz A, Lerche C, Busch AE, Attali B. Stilbenes and fenamates rescue the
loss of I(KS) channel function induced by an LQT5 mutation and other IsK mutants. Ackerman MJ, January CT. Most LQT2 mutations reduce Kv11.1 (hERG) current by a
EMBO J 18: 4137– 4148, 1999. class 2 (trafficking-deficient) mechanism. Circulation 113: 365–373, 2006.

6. Abriel H, Cabo C, Wehrens XH, Rivolta I, Motoike HK, Memmi M, Napolitano C, 23. Anderson CL, Kuzmicki CE, Childs RR, Hintz CJ, Delisle BP, January CT. Large-scale
Priori SG, Kass RS. Novel arrhythmogenic mechanism revealed by a long-QT syn- mutational analysis of Kv11.1 reveals molecular insights into type 2 long QT syn-
drome mutation in the cardiac Na⫹ channel. Circ Res 88: 740 –745, 2001. drome. Nat Commun 5: 5535, 2014.

7. Ackerman MJ, Priori SG, Willems S, Berul C, Brugada R, Calkins H, Camm AJ, Ellinor 24. Ando F, Kuruma A, Kawano S. Synergic effects of beta-estradiol and erythromycin on
PT, Gollob M, Hamilton R, Hershberger RE, Judge DP, Le Marec H, McKenna WJ, hERG currents. J Membr Biol 241: 31–38, 2011.
Schulze-Bahr E, Semsarian C, Towbin JA, Watkins H, Wilde A, Wolpert C, Zipes DP.
HRS/EHRA expert consensus statement on the state of genetic testing for the chan- 25. Antzelevitch C, Nesterenko V, Shryock JC, Rajamani S, Song Y, Belardinelli L. The role
nelopathies and cardiomyopathies this document was developed as a partnership of late I Na in development of cardiac arrhythmias. Handb Exp Pharmacol 221: 137–
between the Heart Rhythm Society (HRS) and the European Heart Rhythm Associa- 168, 2014.
tion (EHRA). Heart Rhythm 8: 1308 –1339, 2011.
26. Antzelevitch C, Sicouri S. Clinical relevance of cardiac arrhythmias generated by
8. Ackerman MJ, Priori SG, Willems S, Berul C, Brugada R, Calkins H, Camm AJ, Ellinor afterdepolarizations. Role of M cells in the generation of U waves, triggered activity
PT, Gollob M, Hamilton R, Hershberger RE, Judge DP, Le Marec H, McKenna WJ, and torsade de pointes. J Am Coll Cardiol 23: 259 –277, 1994.

Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org 121


BOHNEN ET AL.

27. Arcisio-Miranda M, Muroi Y, Chowdhury S, Chanda B. Molecular mechanism of 45. Bendahhou S, Marionneau C, Haurogne K, Larroque MM, Derand R, Szuts V, Escande
allosteric modification of voltage-dependent sodium channels by local anesthetics. J D, Demolombe S, Barhanin J. In vitro molecular interactions and distribution of KCNE
Gen Physiol 136: 541–554, 2010. family with KCNQ1 in the human heart. Cardiovasc Res 67: 529 –538, 2005.

28. Asayama M, Kurokawa J, Shirakawa K, Okuyama H, Kagawa T, Okada J, Sugiura S, 46. Benhorin J, Medina A. Images in clinical medicine. Congenital long-QT syndrome. N
Hisada T, Furukawa T. Effects of an hERG activator, ICA-105574, on electrophysio- Engl J Med 336: 1568, 1997.
logical properties of canine hearts. J Pharmacol Sci 121: 1– 8, 2013.
47. Bennett PB, Yazawa K, Makita N, George AL Jr. Molecular mechanism for an inherited
29. Aziz PF, Wieand TS, Ganley J, Henderson J, Patel AR, Iyer VR, Vogel RL, McBride M, cardiac arrhythmia. Nature 376: 683– 685, 1995.
Vetter VL, Shah MJ. Genotype- and mutation site-specific QT adaptation during ex-
ercise, recovery, and postural changes in children with long-QT syndrome. Circ Ar- 48. Berecki G, Zegers JG, Verkerk AO, Bhuiyan ZA, de Jonge B, Veldkamp MW, Wilders
rhythm Electrophysiol 4: 867– 873, 2011. R, van Ginneken AC. HERG channel (dys)function revealed by dynamic action poten-
tial clamp technique. Biophys J 88: 566 –578, 2005.
30. Balijepalli RC, Kamp TJ. Caveolae, ion channels and cardiac arrhythmias. Prog Biophys
Mol Biol 98: 149 –160, 2008. 49. Berman MF, Camardo JS, Robinson RB, Siegelbaum SA. Single sodium channels from
canine ventricular myocytes: voltage dependence and relative rates of activation and
31. Balser JR, Nuss HB, Orias DW, Johns DC, Marban E, Tomaselli GF, Lawrence JH. inactivation. J Physiol 415: 503–531, 1989.
Local anesthetics as effectors of allosteric gating. Lidocaine effects on inactivation-
deficient rat skeletal muscle Na channels. J Clin Invest 98: 2874 –2886, 1996. 50. Bhuiyan ZA, Momenah TS, Gong Q, Amin AS, Ghamdi SA, Carvalho JS, Homfray T,
Mannens MM, Zhou Z, Wilde AA. Recurrent intrauterine fetal loss due to near ab-

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


32. Bankston JR, Kass RS. Fading sodium channels in failing hearts. Circ Res 101: 1073– sence of HERG: clinical and functional characterization of a homozygous nonsense
1074, 2007. HERG Q1070X mutation. Heart Rhythm 5: 553–561, 2008.

33. Bankston JR, Kass RS. Molecular determinants of local anesthetic action of beta- 51. Bian J, Cui J, McDonald T. VHERG K⫹ channel activity is regulated by changes in
blocking drugs: Implications for therapeutic management of long QT syndrome vari- phosphatidyl inositol 4,5-bisphosphate. Circ Res 89: 1168 –1176, 2001.
ant 3. J Mol Cell Cardiol 48: 246 –253, 2010.
52. Bianchi L, Priori SG, Napolitano C, Surewicz KA, Dennis AT, Memmi M, Schwartz PJ,
34. Bankston JR, Sampson KJ, Kateriya S, Glaaser IW, Malito DL, Chung WK, Kass RS. A Brown AM. Mechanisms of I(Ks) suppression in LQT1 mutants. Am J Physiol Heart Circ
novel LQT-3 mutation disrupts an inactivation gate complex with distinct rate-depen- Physiol 279: H3003–H3011, 2000.
dent phenotypic consequences. Channels 1: 273–280, 2007.
53. Bianchi L, Shen Z, Dennis AT, Priori SG, Napolitano C, Ronchetti E, Bryskin R,
35. Bankston JR, Yue M, Chung W, Spyres M, Pass RH, Silver E, Sampson KJ, Kass RS. A Schwartz PJ, Brown AM. Cellular dysfunction of LQT5-minK mutants: abnormalities
novel and lethal de novo LQT-3 mutation in a newborn with distinct molecular of IKs, IKr and trafficking in long QT syndrome. Hum Mol Genet 8: 1499 –1507, 1999.
pharmacology and therapeutic response. PLoS One 2: e1258, 2007.
54. Boczek NJ, Miller EM, Ye D, Nesterenko VV, Tester DJ, Antzelevitch C, Czosek RJ,
36. Barhanin J, Lesage F, Guillemare E, Fink M, Lazdunski M, Romey G. K(V)LQT1 and lsK Ackerman MJ, Ware SM. Novel Timothy syndrome mutation leading to increase in
(minK) proteins associate to form the I(Ks) cardiac potassium current. Nature 384: CACNA1C window current. Heart Rhythm 12: 211–219, 2015.
78 – 80, 1996.
55. Boulet IR, Labro AJ, Raes AL, Snyders DJ. Role of the S6 C-terminus in KCNQ1
37. Barro-Soria R, Rebolledo S, Liin SI, Perez ME, Sampson KJ, Kass RS, Larsson HP. channel gating. J Physiol 585: 325–337, 2007.
KCNE1 divides the voltage sensor movement in KCNQ1/KCNE1 channels into two
steps. Nat Commun 5: 3750, 2014. 56. Brelidze TI, Carlson AE, Sankaran B, Zagotta WN. Structure of the carboxy-terminal
region of a KCNH channel. Nature 481: 530 –533, 2012.
38. Barsheshet A, Dotsenko O, Goldenberg I. Genotype-specific risk stratification and
management of patients with long QT syndrome. Ann Noninvasive Electrocardiol 18: 57. Brink PA, Crotti L, Corfield V, Goosen A, Durrheim G, Hedley P, Heradien M,
499 –509, 2013. Geldenhuys G, Vanoli E, Bacchini S, Spazzolini C, Lundquist AL, Roden DM, George
AL Jr, Schwartz PJ. Phenotypic variability and unusual clinical severity of congenital
39. Barsheshet A, Goldenberg I, JOU, Moss AJ, Jons C, Shimizu W, Wilde AA, McNitt S, long-QT syndrome in a founder population. Circulation 112: 2602–2610, 2005.
Peterson DR, Zareba W, Robinson JL, Ackerman MJ, Cypress M, Gray DA, Hofman
N, Kanters JK, Kaufman ES, Platonov PG, Qi M, Towbin JA, Vincent GM, Lopes CM. 58. Brugada R, Brugada J, Antzelevitch C, Kirsch GE, Potenza D, Towbin JA, Brugada P.
Mutations in cytoplasmic loops of the KCNQ1 channel and the risk of life-threatening Sodium channel blockers identify risk for sudden death in patients with ST-segment
events: implications for mutation-specific response to beta-blocker therapy in type 1 elevation and right bundle branch block but structurally normal hearts. Circulation
long-QT syndrome. Circulation 125: 1988 –1996, 2012. 101: 510 –515, 2000.

40. Barsheshet A, Peterson DR, Moss AJ, Schwartz PJ, Kaufman ES, McNitt S, Polonsky S, 59. Brunner M, Peng X, Liu GX, Ren XQ, Ziv O, Choi BR, Mathur R, Hajjiri M, Odening
Buber J, Zareba W, Robinson JL, Ackerman MJ, Benhorin J, Towbin JA, Vincent GM, KE, Steinberg E, Folco EJ, Pringa E, Centracchio J, Macharzina RR, Donahay T, Scho-
Zhang L, Goldenberg I. Genotype-specific QT correction for heart rate and the risk of field L, Rana N, Kirk M, Mitchell GF, Poppas A, Zehender M, Koren G. Mechanisms of
life threatening cardiac events in adolescents with the congenital long-QT syndrome. cardiac arrhythmias and sudden death in transgenic rabbits with long QT syndrome. J
Heart Rhythm 8: 1207–1213, 2011. Clin Invest 118: 2246 –2259, 2008.

41. Bartos DC, Duchatelet S, Burgess DE, Klug D, Denjoy I, Peat R, Lupoglazoff JM, 60. Buber J, Mathew J, Moss AJ, Hall WJ, Barsheshet A, McNitt S, Robinson JL, Zareba W,
Fressart V, Berthet M, Ackerman MJ, January CT, Guicheney P, Delisle BP. R231C Ackerman MJ, Kaufman ES, Luria D, Eldar M, Towbin JA, Vincent M, Goldenberg I.
mutation in KCNQ1 causes long QT syndrome type 1 and familial atrial fibrillation. Risk of recurrent cardiac events after onset of menopause in women with congenital
Heart Rhythm 8: 48 –55, 2011. long-QT syndrome types 1 and 2. Circulation 123: 2784 –2791, 2011.

42. Beckmann C, Rinne A, Littwitz C, Mintert E, Bosche LI, Kienitz MC, Pott L, Bender K. 61. Burgess DE, Bartos DC, Reloj AR, Campbell KS, Johnson JN, Tester DJ, Ackerman MJ,
G protein-activated (GIRK) current in rat ventricular myocytes is masked by consti- Fressart V, Denjoy I, Guicheney P, Moss AJ, Ohno S, Horie M, Delisle BP. High-risk
tutive inward rectifier current [I(K1)]. Cell Physiol Biochem 21: 259 –268, 2008. long QT syndrome mutations in the Kv7.1 (KCNQ1) pore disrupt the molecular basis for
rapid K⫹ permeation. Biochemistry 51: 9076 –9085, 2012.
43. Belardinelli L, Liu G, Smith-Maxwell C, Wang WQ, El-Bizri N, Hirakawa R, Karpinski
S, Li CH, Hu L, Li XJ, Crumb W, Wu L, Koltun D, Zablocki J, Yao L, Dhalla AK, 62. Busch AE, Suessbrich H, Waldegger S, Sailer E, Greger R, Lang H, Lang F, Gibson KJ,
Rajamani S, Shryock JC. A novel, potent, and selective inhibitor of cardiac late sodium Maylie JG. Inhibition of IKs in guinea pig cardiac myocytes and guinea pig IsK channels
current suppresses experimental arrhythmias. J Pharmacol Exp Ther 344: 23–32, 2013. by the chromanol 293B. Pflügers Arch 432: 1094 –1096, 1996.

44. Bendahhou S, Donaldson MR, Plaster NM, Tristani-Firouzi M, Fu YH, Ptacek LJ. 63. Caballero R, Moreno I, Gonzalez T, Arias C, Valenzuela C, Delpon E, Tamargo J.
Defective potassium channel Kir2.1 trafficking underlies Andersen-Tawil syndrome. J Spironolactone and its main metabolite, canrenoic acid, block human ether-a-go-go-
Biol Chem 278: 51779 –51785, 2003. related gene channels. Circulation 107: 889 – 895, 2003.

122 Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org


CONGENITAL LONG QT SYNDROME

64. Calvillo L, Spazzolini C, Vullo E, Insolia R, Crotti L, Schwartz PJ. Propranolol prevents 85. Clancy CE, Rudy Y. Linking a genetic defect to its cellular phenotype in a cardiac
life-threatening arrhythmias in LQT3 transgenic mice: implications for the clinical arrhythmia. Nature 400: 566 –569, 1999.
management of LQT3 patients. Heart Rhythm 11: 126 –132, 2014.
86. Clancy CE, Rudy Y. Na⫹ channel mutation that causes both Brugada and long-QT
65. Camors E, Mohler PJ, Bers DM, Despa S. Ankyrin-B reduction enhances Ca spark- syndrome phenotypes: a simulation study of mechanism. Circulation 105: 1208 –1213,
mediated SR Ca release promoting cardiac myocyte arrhythmic activity. J Mol Cell 2002.
Cardiol 52: 1240 –1248, 2012.
87. Clancy CE, Tateyama M, Liu H, Wehrens XH, Kass RS. Non-equilibrium gating in
66. Capes DL, Goldschen-Ohm MP, Arcisio-Miranda M, Bezanilla F, Chanda B. Domain cardiac Na⫹ channels: an original mechanism of arrhythmia. Circulation 107: 2233–
IV voltage-sensor movement is both sufficient and rate limiting for fast inactivation in 2237, 2003.
sodium channels. J Gen Physiol 142: 101–112, 2013.
88. Clancy CE, Zhu ZI, Rudy Y. Pharmacogenetics and anti-arrhythmic drug therapy: a
67. Carmeliet E. Voltage- and time-dependent block of the delayed K⫹ current in cardiac theoretical investigation. Am J Physiol Heart Circ Physiol 292: H66 –H75, 2007.
myocytes by dofetilide. J Pharmacol Exp Ther 262: 809 – 817, 1992.
89. Compton SJ, Lux RL, Ramsey MR, Strelich KR, Sanguinetti MC, Green LS, Keating MT,
68. Casimiro MC, Knollmann BC, Ebert SN, Vary JC Jr, Greene AE, Franz MR, Grinberg Mason JW. Genetically defined therapy of inherited long-QT syndrome. Correction of
A, Huang SP, Pfeifer K. Targeted disruption of the Kcnq1 gene produces a mouse abnormal repolarization by potassium. Circulation 94: 1018 –1022, 1996.
model of Jervell and Lange-Nielsen Syndrome. Proc Natl Acad Sci USA 98: 2526 –2531,
2001. 90. Cordes JS, Sun Z, Lloyd DB, Bradley JA, Opsahl AC, Tengowski MW, Chen X, Zhou
J. Pentamidine reduces hERG expression to prolong the QT interval. Br J Pharmacol
69. Casis O, Olesen SP, Sanguinetti MC. Mechanism of action of a novel human ether-a-
145: 15–23, 2005.

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


go-go-related gene channel activator. Mol Pharmacol 69: 658 – 665, 2006.
91. Cormier JW, Rivolta I, Tateyama M, Yang AS, Kass RS. Secondary structure of the
70. Catterall WA. Voltage-gated sodium channels at 60: structure, function, and patho-
human cardiac Na⫹ channel C terminus: evidence for a role of helical structures in
physiology. J Physiol 590: 2577–2589, 2012.
modulation of channel inactivation. J Biol Chem 277: 9233–9241, 2002.
71. Cha A, Ruben PC, George AL Jr, Fujimoto E, Bezanilla F. Voltage sensors in domains
92. Cronk LB, Ye B, Kaku T, Tester DJ, Vatta M, Makielski JC, Ackerman MJ. Novel
III and IV, but not I and II, are immobilized by Na⫹ channel fast inactivation. Neuron 22:
mechanism for sudden infant death syndrome: persistent late sodium current second-
73– 87, 1999.
ary to mutations in caveolin-3. Heart Rhythm 4: 161–166, 2007.
72. Chanda B, Bezanilla F. Tracking voltage-dependent conformational changes in skeletal
93. Crotti L, Johnson CN, Graf E, De Ferrari GM, Cuneo BF, Ovadia M, Papagiannis J,
muscle sodium channel during activation. J Gen Physiol 120: 629 – 645, 2002.
Feldkamp MD, Rathi SG, Kunic JD, Pedrazzini M, Wieland T, Lichtner P, Beckmann
73. Chandra R, Starmer CF, Grant AO. Multiple effects of KPQ deletion mutation on BM, Clark T, Shaffer C, Benson DW, Kaab S, Meitinger T, Strom TM, Chazin WJ,
gating of human cardiac Na⫹ channels expressed in mammalian cells. Am J Physiol Schwartz PJ, George AL Jr. Calmodulin mutations associated with recurrent cardiac
Heart Circ Physiol 274: H1643–H1654, 1998. arrest in infants. Circulation 127: 1009 –1017, 2013.

74. Chauhan VS, Tuvia S, Buhusi M, Bennett V, Grant AO. Abnormal cardiac Na⫹ channel 94. Cui J, Melman Y, Palma E, Fishman GI, McDonald TV. Cyclic AMP regulates the HERG
properties and QT heart rate adaptation in neonatal ankyrin(B) knockout mice. Circ K⫹ channel by dual pathways. Curr Biol 10: 671– 674, 2000.
Res 86: 441– 447, 2000.
95. Cunha SR, Bhasin N, Mohler PJ. Targeting and stability of Na/Ca exchanger 1 in
75. Chen J, Mitcheson JS, Tristani-Firouzi M, Lin M, Sanguinetti MC. The S4 –S5 linker cardiomyocytes requires direct interaction with the membrane adaptor ankyrin-B. J
couples voltage sensing and activation of pacemaker channels. Proc Natl Acad Sci USA Biol Chem 282: 4875– 4883, 2007.
98: 11277–11282, 2001.
96. Cunha SR, Hund TJ, Hashemi S, Voigt N, Li N, Wright P, Koval O, Li J, Gudmundsson
76. Chen J, Zheng R, Melman YF, McDonald TV. Functional interactions between KCNE1 H, Gumina RJ, Karck M, Schott JJ, Probst V, Le Marec H, Anderson ME, Dobrev D,
C-terminus and the KCNQ1 channel. PLoS One 4: e5143, 2009. Wehrens XH, Mohler PJ. Defects in ankyrin-based membrane protein targeting path-
ways underlie atrial fibrillation. Circulation 124: 1212–1222, 2011.
77. Chen J, Zou A, Splawski I, Keating MT, Sanguinetti MC. Long QT syndrome-associ-
ated mutations in the Per-Arnt-Sim (PAS) domain of HERG potassium channels ac- 97. Curran ME, Splawski I, Timothy KW, Vincent GM, Green ED, Keating MT. A molec-
celerate channel deactivation. J Biol Chem 274: 10113–10118, 1999. ular basis for cardiac arrhythmia: HERG mutations cause long QT syndrome. Cell 80:
795– 803, 1995.
78. Chen L, Kurokawa J, Kass RS. Phosphorylation of the A-kinase-anchoring protein
Yotiao contributes to protein kinase A regulation of a heart potassium channel. J Biol 98. Dahimene S, Alcolea S, Naud P, Jourdon P, Escande D, Brasseur R, Thomas A, Baro I,
Chem 280: 31347–31352, 2005. Merot J. The N-terminal juxtamembranous domain of KCNQ1 is critical for channel
surface expression: implications in the Romano-Ward LQT1 syndrome. Circ Res 99:
79. Chen L, Marquardt ML, Tester DJ, Sampson KJ, Ackerman MJ, Kass RS. Mutation of an
1076 –1083, 2006.
A-kinase-anchoring protein causes long-QT syndrome. Proc Natl Acad Sci USA 104:
20990 –20995, 2007. 99. Dascal N, Schreibmayer W, Lim NF, Wang W, Chavkin C, DiMagno L, Labarca C,
80. Chen L, Zhang Q, Qiu Y, Li Z, Chen Z, Jiang H, Li Y, Yang H. Migration of PIP2 lipids Kieffer BL, Gaveriaux-Ruff C, Trollinger D. Atrial G protein-activated K⫹ channel:
on voltage-gated potassium channel surface influences channel deactivation. Scientific expression cloning and molecular properties. Proc Natl Acad Sci USA 90: 10235–
Reports 5: 15079, 2015. 10239, 1993.

81. Chouabe C, Neyroud N, Guicheney P, Lazdunski M, Romey G, Barhanin J. Properties 100. De la Pena P, Machin A, Fernandez-Trillo J, Dominguez P, Barros F. Mapping of
of KvLQT1 K⫹ channel mutations in Romano-Ward and Jervell and Lange-Nielsen interactions between the N- and C-termini and the channel core in HERG K⫹ chan-
inherited cardiac arrhythmias. EMBO J 16: 5472–5479, 1997. nels. Biochem J 451: 463– 474, 2013.

82. Choveau FS, Rodriguez N, Abderemane Ali F, Labro AJ, Rose T, Dahimene S, Boudin 101. Dhar Malhotra J, Chen C, Rivolta I, Abriel H, Malhotra R, Mattei LN, Brosius FC, Kass
H, Le Henaff C, Escande D, Snyders DJ, Charpentier F, Merot J, Baro I, Loussouarn G. RS, Isom LL. Characterization of sodium channel alpha- and beta-subunits in rat and
KCNQ1 channels voltage dependence through a voltage-dependent binding of the mouse cardiac myocytes. Circulation 103: 1303–1310, 2001.
S4 –S5 linker to the pore domain. J Biol Chem 286: 707–716, 2011.
102. Dobrzynski H, Anderson RH, Atkinson A, Borbas Z, D’Souza A, Fraser JF, Inada S,
83. Chuang HH, Prescott ED, Kong H, Shields S, Jordt SE, Basbaum AI, Chao MV, Julius D. Logantha SJ, Monfredi O, Morris GM, Moorman AF, Nikolaidou T, Schneider H, Szuts
Bradykinin and nerve growth factor release the capsaicin receptor from V, Temple IP, Yanni J, Boyett MR. Structure, function and clinical relevance of the
PtdIns(4,5)P2-mediated inhibition. Nature 411: 957–962, 2001. cardiac conduction system, including the atrioventricular ring and outflow tract tis-
sues. Pharmacol Ther 139: 260 –288, 2013.
84. Chung DY, Chan PJ, Bankston JR, Yang L, Liu G, Marx SO, Karlin A, Kass RS. Location
of KCNE1 relative to KCNQ1 in the I(KS) potassium channel by disulfide cross-linking 103. Donaldson MR, Jensen JL, Tristani-Firouzi M, Tawil R, Bendahhou S, Suarez WA,
of substituted cysteines. Proc Natl Acad Sci USA 106: 743–748, 2009. Cobo AM, Poza JJ, Behr E, Wagstaff J, Szepetowski P, Pereira S, Mozaffar T, Escolar

Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org 123


BOHNEN ET AL.

DM, Fu YH, Ptacek LJ. PIP2 binding residues of Kir2.1 are common targets of muta- sion of repolarisation and arrhythmias in DeltaKPQ SCN5A (long QT3) mice. Cardio-
tions causing Andersen syndrome. Neurology 60: 1811–1816, 2003. vasc Res 57: 1085–1093, 2003.

104. Doupnik CA, Davidson N, Lester HA. The inward rectifier potassium channel family. 121. Fatima A, Kaifeng S, Dittmann S, Xu G, Gupta MK, Linke M, Zechner U, Nguemo F,
Curr Opin Neurobiol 5: 268 –277, 1995. Milting H, Farr M, Hescheler J, Saric T. The disease-specific phenotype in cardiomy-
ocytes derived from induced pluripotent stem cells of two long QT syndrome type 3
105. Doyle DA, Morais Cabral J, Pfuetzner RA, Kuo A, Gulbis JM, Cohen SL, Chait BT, patients. PLoS One 8: e83005, 2013.
MacKinnon R. The structure of the potassium channel: molecular basis of K⫹ conduc-
tion and selectivity. Science 280: 69 –77, 1998. 122. Ferrer T, Rupp J, Piper DR, Tristani-Firouzi M. The S4 –S5 linker directly couples
voltage sensor movement to the activation gate in the human ether-a-go-go-related
106. Doyle DD, Goings G, Upshaw-Earley J, Ambler SK, Mondul A, Palfrey HC, Page E.
gene (hERG) K⫹ channel. J Biol Chem 281: 12858 –12864, 2006.
Dystrophin associates with caveolae of rat cardiac myocytes: relationship to dystro-
glycan. Circ Res 87: 480 – 488, 2000. 123. Ficker E, Dennis AT, Obejero-Paz CA, Castaldo P, Taglialatela M, Brown AM. Reten-
tion in the endoplasmic reticulum as a mechanism of dominant-negative current
107. Duggal P, Vesely MR, Wattanasirichaigoon D, Villafane J, Kaushik V, Beggs AH. Muta-
suppression in human long QT syndrome. J Mol Cell Cardiol 32: 2327–2337, 2000.
tion of the gene for IsK associated with both Jervell and Lange-Nielsen and Romano-
Ward forms of Long-QT syndrome. Circ J 97: 142–146, 1998. 124. Ficker E, Dennis AT, Wang L, Brown AM. Role of the cytosolic chaperones Hsp70 and
Hsp90 in maturation of the cardiac potassium channel HERG. Circ Res 92: e87–100,
108. Dumaine R, Kirsch GE. Mechanism of lidocaine block of late current in long Q-T
2003.
mutant Na⫹ channels. Am J Physiol Heart Circ Physiol 274: H477–H487, 1998.

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


125. Ficker E, Jarolimek W, Brown AM. Molecular determinants of inactivation and dofeti-
109. Dumaine R, Wang Q, Keating MT, Hartmann HA, Schwartz PJ, Brown AM, Kirsch GE.
lide block in ether a-go-go (EAG) channels and EAG-related K⫹ channels. Mol Phar-
Multiple mechanisms of Na⫹ channel-linked long-QT syndrome. Circ Res 78: 916 –
macol 60: 1343–1348, 2001.
924, 1996.
126. Fozzard HA. Afterdepolarizations and triggered activity. Basic Res Cardiol 87 Suppl 2:
110. Dvir M, Strulovich R, Sachyani D, Ben-Tal Cohen I, Haitin Y, Dessauer C, Pongs O,
105–113, 1992.
Kass R, Hirsch JA, Attali B. Long QT mutations at the interface between KCNQ1 helix
C and KCNE1 disrupt I(KS) regulation by PKA and PIP2. J Cell Sci 127: 3943–3955, 127. Fredj S, Sampson KJ, Liu H, Kass RS. Molecular basis of ranolazine block of LQT-3
2014. mutant sodium channels: evidence for site of action. Br J Pharmacol 148: 16 –24, 2006.
111. Eaholtz G, Scheuer T, Catterall WA. Restoration of inactivation and block of open 128. Fuller H, Justo F, Nearing BD, Kahlig KM, Rajamani S, Belardinelli L, Verrier RL.
sodium channels by an inactivation gate peptide. Neuron 12: 1041–1048, 1994.
Eleclazine, a new selective cardiac late sodium current inhibitor, confers concurrent
112. Echt DS, Liebson PR, Mitchell LB, Peters RW, Obias-Manno D, Barker AH, Arensberg protection against autonomically induced atrial premature beats, repolarization alter-
D, Baker A, Friedman L, Greene HL. Mortality and morbidity in patients receiving nans and heterogeneity, and atrial fibrillation in an intact porcine model. Heart Rhythm
encainide, flecainide, or placebo The Cardiac Arrhythmia Suppression Trial. N Engl J 13: 1679 –1686, 2016.
Med 324: 781–788, 1991.
129. Fulop L, Banyasz T, Szabo G, Toth IB, Biro T, Lorincz I, Balogh A, Peto K, Miko I,
113. Eldstrom J, Wang Z, Werry D, Wong N, Fedida D. Microscopic mechanisms for long Nanasi PP. Effects of sex hormones on ECG parameters and expression of cardiac ion
QT syndrome type 1 revealed by single-channel analysis of I(Ks) with S3 domain channels in dogs. Acta Physiol 188: 163–171, 2006.
mutations in KCNQ1. Heart Rhythm 12: 386 –394, 2015.
130. Funasako M, Aiba T, Ishibashi K, Nakajima I, Miyamoto K, Inoue Y, Okamura H, Noda
114. Eldstrom J, Xu H, Werry D, Kang C, Loewen ME, Degenhardt A, Sanatani S, Tibbits T, Kamakura S, Anzai T, Noguchi T, Yasuda S, Miyamoto Y, Fukushima Kusano K,
GF, Sanders C, Fedida D. Mechanistic basis for LQT1 caused by S3 mutations in the Ogawa H, Shimizu W. Pronounced shortening of QT interval with mexiletine infusion
KCNQ1 subunit of IKs. J Gen Physiol 135: 433– 448, 2010. test in patients with type 3 congenital long QT syndrome. Circ J 80: 340 –345, 2016.

115. Eng G, Lee BW, Protas L, Gagliardi M, Brown K, Kass RS, Keller G, Robinson RB, 131. Furutani M, Trudeau MC, Hagiwara N, Seki A, Gong Q, Zhou Z, Imamura S, Na-
Vunjak-Novakovic G. Autonomous beating rate adaptation in human stem cell-de- gashima H, Kasanuki H, Takao A, Momma K, January CT, Robertson GA, Matsuoka R.
rived cardiomyocytes. Nat Commun 7: 10312, 2016. Novel mechanism associated with an inherited cardiac arrhythmia: defective protein
trafficking by the mutant HERG (G601S) potassium channel. Circulation 99: 2290 –
116. Es-Salah-Lamoureux Z, Xiong PY, Goodchild SJ, Ahern CA, Fedida D. Blockade of 2294, 1999.
permeation by potassium but normal gating of the G628S nonconducting hERG chan-
nel mutant. Biophys J 101: 662– 670, 2011. 132. Gaborit N, Le Bouter S, Szuts V, Varro A, Escande D, Nattel S, Demolombe S.
Regional and tissue specific transcript signatures of ion channel genes in the non-
117. Etheridge SP, Compton SJ, Tristani-Firouzi M, Mason JW. A new oral therapy for long diseased human heart. J Physiol 582: 675– 693, 2007.
QT syndrome: long-term oral potassium improves repolarization in patients with
HERG mutations. J Am Coll Cardiol 42: 1777–1782, 2003. 133. Galbiati F, Engelman JA, Volonte D, Zhang XL, Minetti C, Li M, Hou H Jr, Kneitz B,
Edelmann W, Lisanti MP. Caveolin-3 null mice show a loss of caveolae, changes in the
118. European. Heart Rhythm Association, Heart Rhythm Society, Zipes DP, Camm AJ, microdomain distribution of the dystrophin-glycoprotein complex, and t-tubule ab-
Borggrefe M, Buxton AE, Chaitman B, Fromer M, Gregoratos G, Klein G, Moss AJ, normalities. J Biol Chem 276: 21425–21433, 2001.
Myerburg RJ, Priori SG, Quinones MA, Roden DM, Silka MJ, Tracy C, Smith SC, Jr,
Jacobs AK, Adams CD, Antman EM, Anderson JL, Hunt SA, Halperin JL, Nishimura R, 134. Garg V, Stary-Weinzinger A, Sanguinetti MC. ICA-105574 interacts with a common
Ornato JP, Page RL, Riegel B, Priori SG, Blanc JJ, Budaj A, Camm AJ, Dean V, Deckers binding site to elicit opposite effects on inactivation gating of EAG and ERG potassium
JW, Despres C, Dickstein K, Lekakis J, McGregor K, Metra M, Morais J, Osterspey A, channels. Mol Pharmacol 83: 805– 813, 2013.
Tamargo JL, Zamorano JL, American College of Cardiology, American Heart Associ-
ation Task Force, European Society of Cardiology Committee for Practice Guidelines. 135. Gavillet B, Rougier JS, Domenighetti AA, Behar R, Boixel C, Ruchat P, Lehr HA,
ACC/AHA/ESC 2006 guidelines for management of patients with ventricular arrhyth- Pedrazzini T, Abriel H. Cardiac sodium channel Nav1.5 is regulated by a multiprotein
mias and the prevention of sudden cardiac death: a report of the American College of complex composed of syntrophins and dystrophin. Circ Res 99: 407– 414, 2006.
Cardiology/American Heart Association Task Force and the European Society of
Cardiology Committee for Practice Guidelines (Writing Committee to Develop 136. Gehrmann J, Meister M, Maguire CT, Martins DC, Hammer PE, Neer EJ, Berul CI,
Guidelines for Management of Patients With Ventricular Arrhythmias and the Preven- Mende U. Impaired parasympathetic heart rate control in mice with a reduction of
tion of Sudden Cardiac Death). J Am Coll Cardiol 48: e247–346, 2006. functional G protein betagamma-subunits. Am J Physiol Heart Circ Physiol 282: H445–
H456, 2002.
119. Faber GM, Rudy Y. Action potential and contractility changes in [Na⫹]I overloaded
cardiac myocytes: a simulation study. Biophys J 78: 2392–2404, 2000. 137. Gellens ME, George AL Jr, Chen LQ, Chahine M, Horn R, Barchi RL, Kallen RG.
Primary structure and functional expression of the human cardiac tetrodotoxin-
120. Fabritz L, Kirchhof P, Franz MR, Nuyens D, Rossenbacker T, Ottenhof A, Haverkamp insensitive voltage-dependent sodium channel. Proc Natl Acad Sci USA 89: 554 –
W, Breithardt G, Carmeliet E, Carmeliet P. Effect of pacing and mexiletine on disper- 558, 1992.

124 Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org


CONGENITAL LONG QT SYNDROME

138. Gerlach AC, Stoehr SJ, Castle NA. Pharmacological removal of human ether-a-go-go- 157. Hackos DH, Chang TH, Swartz KJ. Scanning the intracellular S6 activation gate in the
related gene potassium channel inactivation by 3-nitro-N-(4-phenoxyphenyl) benz- shaker K⫹ channel. J Gen Physiol 119: 521–532, 2002.
amide (ICA-105574). Mol Pharmacol 77: 58 – 68, 2010.
158. Haissaguerre M, Vigmond E, Stuyvers B, Hocini M, Bernus O. Ventricular arrhythmias
139. Gerlach U, Brendel J, Lang HJ, Paulus EF, Weidmann K, Bruggemann A, Busch AE, and the His-Purkinje system. Nat Rev Cardiol 13: 155–166, 2016.
Suessbrich H, Bleich M, Greger R. Synthesis and activity of novel and selective I(Ks)-
channel blockers. J Med Chem 44: 3831–3837, 2001. 159. Haitin Y, Attali B. The C-terminus of Kv7 channels: a multifunctional module. J Physiol
586: 1803–1810, 2008.
140. Gessner G, Macianskiene R, Starkus JG, Schonherr R, Heinemann SH. The amioda-
rone derivative KB130015 activates hERG1 potassium channels via a novel mecha- 160. Haitin Y, Carlson AE, Zagotta WN. The structural mechanism of KCNH-channel
nism. Eur J Pharmacol 632: 52–59, 2010. regulation by the eag domain. Nature 501: 444 – 448, 2013.

141. Ghosh S, Nunziato DA, Pitt GS. KCNQ1 assembly and function is blocked by long-QT 161. Haitin Y, Wiener R, Shaham D, Peretz A, Cohen EB, Shamgar L, Pongs O, Hirsch JA,
syndrome mutations that disrupt interaction with calmodulin. Circ Res 98: 1048 – Attali B. Intracellular domains interactions and gated motions of I(KS) potassium
1054, 2006. channel subunits. EMBO J 28: 1994 –2005, 2009.

142. Gillis J, Burashnikov E, Antzelevitch C, Blaser S, Gross G, Turner L, Babul-Hirji R, 162. Hanck DA, Nikitina E, McNulty MM, Fozzard HA, Lipkind GM, Sheets MF. Using
Chitayat D. Long QT, syndactyly, joint contractures, stroke and novel CACNA1C lidocaine and benzocaine to link sodium channel molecular conformations to state-
mutation: expanding the spectrum of Timothy syndrome. Am J Med Genet 158: 182– dependent antiarrhythmic drug affinity. Circ Res 105: 492– 499, 2009.
187, 2012.
163. Hancox JC, McPate MJ, El Harchi A, Zhang YH. The hERG potassium channel and

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


143. Giudicessi JR, Ackerman MJ. Genotype- and phenotype-guided management of con- hERG screening for drug-induced torsades de pointes. Pharmacol Ther 119: 118 –132,
genital long QT syndrome. Curr Probl Cardiol 38: 417– 455, 2013. 2008.

144. Glaaser IW, Osteen JD, Puckerin A, Sampson KJ, Jin X, Kass RS. Perturbation of 164. Hansen RS, Diness TG, Christ T, Demnitz J, Ravens U, Olesen SP, Grunnet M.
sodium channel structure by an inherited Long QT Syndrome mutation. Nat Commun Activation of human ether-a-go-go-related gene potassium channels by the dipheny-
3: 706, 2012. lurea 1,3-bis-(2-hydroxy-5-trifluoromethyl-phenyl)-urea (NS1643). Mol Pharmacol
69: 266 –277, 2006.
145. Goldenberg I, Bradley J, Moss A, McNitt S, Polonsky S, Robinson JL, Andrews M,
Zareba W. Beta-blocker efficacy in high-risk patients with the congenital long-QT 165. Hansen RS, Diness TG, Christ T, Wettwer E, Ravens U, Olesen SP, Grunnet M.
syndrome types 1 and 2: implications for patient management. J Cardiovasc Electro- Biophysical characterization of the new human ether-a-go-go-related gene channel
physiol 21: 893–901, 2010. opener NS3623 [N-(4-bromo-2-(1H-tetrazol-5-yl)-phenyl)-N’-(3’-trifluoromethyl-
phenyl)urea]. Mol Pharmacol 70: 1319 –1329, 2006.
146. Goldenberg I, Thottathil P, Lopes CM, Moss AJ, McNitt S, JOU, Robinson JL, Zareba
W, Ackerman MJ, Kaufman ES, Towbin JA, Vincent M, Barsheshet A. Trigger-specific 166. Harmer SC, Wilson AJ, Aldridge R, Tinker A. Mechanisms of disease pathogenesis in
ion-channel mechanisms, risk factors, and response to therapy in type 1 long QT long QT syndrome type 5. Am J Physiol Cell Physiol 298: C263–C273, 2010.
syndrome. Heart Rhythm 9: 49 –56, 2012.
167. Hausdorff SF, Goldstein SA, Rushin EE, Miller C. Functional characterization of a
147. Goldschen-Ohm MP, Capes DL, Oelstrom KM, Chanda B. Multiple pore conforma- minimal K⫹ channel expressed from a synthetic gene. Biochemistry 30: 3341–3346,
tions driven by asynchronous movements of voltage sensors in a eukaryotic sodium 1991.
channel. Nat Commun 4: 1350, 2013.
168. Hayashi K, Fujino N, Ino H, Uchiyama K, Sakata K, Konno T, Masuta E, Funada A,
148. Gong Q, Jones MA, Zhou Z. Mechanisms of pharmacological rescue of trafficking- Sakamoto Y, Tsubokawa T, Hodatsu A, Yasuda T, Kanaya H, Kim MY, Kupershmidt S,
defective hERG mutant channels in human long QT syndrome. J Biol Chem 281: Higashida H, Yamagishi M. A KCR1 variant implicated in susceptibility to the long QT
4069 – 4074, 2006. syndrome. J Mol Cell Cardiol 50: 50 –57, 2011.

149. Gong Q, Keeney DR, Molinari M, Zhou Z. Degradation of trafficking-defective long 169. Heijman J, Spatjens RL, Seyen SR, Lentink V, Kuijpers HJ, Boulet IR, de Windt LJ, David
QT syndrome type II mutant channels by the ubiquitin-proteasome pathway. J Biol M, Volders PG. Dominant-negative control of cAMP-dependent IKs upregulation in
Chem 280: 19419 –19425, 2005. human long-QT syndrome type 1. Circ Res 110: 211–219, 2012.

150. Gong Q, Zhang L, Vincent GM, Horne BD, Zhou Z. Nonsense mutations in hERG 170. Henrion U, Strutz-Seebohm N, Duszenko M, Lang F, Seebohm G. Long QT syn-
cause a decrease in mutant mRNA transcripts by nonsense-mediated mRNA decay in drome-associated mutations in the voltage sensor of I(Ks) channels. Cell Physiol
human long-QT syndrome. Circulation 116: 17–24, 2007. Biochem 24: 11–16, 2009.

151. Gordon E, Panaghie G, Deng L, Bee KJ, Roepke TK, Krogh-Madsen T, Christini DJ, 171. Herzberg IM, Trudeau MC, Robertson GA. Transfer of rapid inactivation and sensi-
Ostrer H, Basson CT, Chung W, Abbott GW. A KCNE2 mutation in a patient with tivity to the class III antiarrhythmic drug E-4031 from HERG to M-eag channels. J
cardiac arrhythmia induced by auditory stimuli and serum electrolyte imbalance. Physiol 511: 3–14, 1998.
Cardiovasc Res 77: 98 –106, 2008.
172. Hilgemann DW, Ball R. Regulation of cardiac Na⫹,Ca2⫹ exchange and KATP potas-
152. Gouaux E, Mackinnon R. Principles of selective ion transport in channels and pumps. sium channels by PIP2. Science 273: 956 –959, 1996.
Science 310: 1461–1465, 2005.
173. Hodgkin AL, Huxley AF. Currents carried by sodium and potassium ions through the
153. Grunnet M, Abbruzzese J, Sachse FB, Sanguinetti MC. Molecular determinants of membrane of the giant axon of Loligo. J Physiol 116: 449 – 472, 1952.
human ether-a-go-go-related gene 1 (hERG1) K⫹ channel activation by NS1643. Mol
Pharmacol 79: 1–9, 2011. 174. Hodgkin AL, Huxley AF. A quantitive description of membrane current and its appli-
cation to conduction and excitation in nerve. J Physiol 117: 500 –544, 1952.
154. Grunnet M, Jespersen T, Rasmussen HB, Ljungstrom T, Jorgensen NK, Olesen SP,
Klaerke DA. KCNE4 is an inhibitory subunit to the KCNQ1 channel. J Physiol 542: 175. Hoorntje T, Alders M, van Tintelen P, van der Lip K, Sreeram N, van der Wal A,
119 –130, 2002. Mannens M, Wilde A. Homozygous premature truncation of the HERG protein: the
human HERG knockout. Circulation 100: 1264 –1267, 1999.
155. Guo J, Cheng YM, Lees-Miller JP, Perissinotti LL, Claydon TW, Hull CM, Thouta S,
Roach DE, Durdagi S, Noskov SY, Duff HJ. NS1643 interacts around L529 of hERG to 176. Hoppe UC, Marban E, Johns DC. Distinct gene-specific mechanisms of arrhythmia
alter voltage sensor movement on the path to activation. Biophys J 108: 1400 –1413, revealed by cardiac gene transfer of two long QT disease genes, HERG and KCNE1.
2015. Proc Natl Acad Sci USA 98: 5335–5340, 2001.

156. Guo J, Massaeli H, Li W, Xu J, Luo T, Shaw J, Kirshenbaum LA, Zhang S. Identification 177. Horner JM, Horner MM, Ackerman MJ. The diagnostic utility of recovery phase QTc
of IKr and its trafficking disruption induced by probucol in cultured neonatal rat during treadmill exercise stress testing in the evaluation of long QT syndrome. Heart
cardiomyocytes. J Pharmacol Exp Ther 321: 911–920, 2007. Rhythm 8: 1698 –1704, 2011.

Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org 125


BOHNEN ET AL.

178. Hoshi N, Takahashi H, Shahidullah M, Yokoyama S, Higashida H. KCR1, a membrane 199. Jouni M, Si-Tayeb K, Es-Salah-Lamoureux Z, Latypova X, Champon B, Caillaud A,
protein that facilitates functional expression of non-inactivating K⫹ currents associ- Rungoat A, Charpentier F, Loussouarn G, Baro I, Zibara K, Lemarchand P, Gaborit N.
ates with rat EAG voltage-dependent K⫹ channels. J Biol Chem 273: 23080 –23085, Toward personalized medicine: using cardiomyocytes differentiated from urine-de-
1998. rived pluripotent stem cells to recapitulate electrophysiological characteristics of type
2 long QT syndrome. J Am Heart Assoc 4: e002159, 2015.
179. Huang CL, Feng S, Hilgemann DW. Direct activation of inward rectifier potassium
channels by PIP2 and its stabilization by Gbetagamma. Nature 391: 803– 806, 1998. 200. Justo F, Fuller H, Nearing BD, Rajamani S, Belardinelli L, Verrier RL. Inhibition of the
cardiac late sodium current with eleclazine protects against ischemia-induced vulner-
180. Huang FD, Chen J, Lin M, Keating MT, Sanguinetti MC. Long-QT syndrome-associ- ability to atrial fibrillation and reduces atrial and ventricular repolarization abnormal-
ated missense mutations in the pore helix of the HERG potassium channel. Circulation ities in the absence and presence of concurrent adrenergic stimulation. Heart Rhythm
104: 1071–1075, 2001. 13: 1860 –1867, 2016.

181. Huang L, Bitner-Glindzicz M, Tranebjaerg L, Tinker A. A spectrum of functional 201. Kagan A, Yu Z, Fishman GI, McDonald TV. The dominant negative LQT2 mutation
effects for disease causing mutations in the Jervell and Lange-Nielsen syndrome. A561V reduces wild-type HERG expression. J Biol Chem 275: 11241–11248, 2000.
Cardiovasc Res 51: 670 – 680, 2001.
202. Kambouris NG, Nuss HB, Johns DC, Marban E, Tomaselli GF, Balser JR. A revised
182. Isbrandt D, Friederich P, Solth A, Haverkamp W, Ebneth A, Borggrefe M, Funke H, view of cardiac sodium channel “blockade” in the long-QT syndrome. J Clin Invest 105:
Sauter K, Breithardt G, Pongs O, Schulze-Bahr E. Identification and functional char- 1133–1140, 2000.
acterization of a novel KCNE2 (MiRP1) mutation that alters HERG channel kinetics. J
Mol Med 80: 524 –532, 2002. 203. Kang C, Tian C, Sonnichsen FD, Smith JA, Meiler J, George AL Jr, Vanoye CG, Kim HJ,
Sanders CR. Structure of KCNE1 and implications for how it modulates the KCNQ1

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


183. Ito H, Hosoya Y, Inanobe A, Tomoike H, Endoh M. Acetylcholine and adenosine potassium channel. Biochemistry 47: 7999 – 8006, 2008.
activate the G protein-gated muscarinic K⫹ channel in ferret ventricular myocytes.
Naunyn-Schmiedebergs Arch Pharmacol 351: 610 – 617, 1995. 204. Kang J, Chen XL, Wang H, Ji J, Cheng H, Incardona J, Reynolds W, Viviani F, Tabart M,
Rampe D. Discovery of a small molecule activator of the human ether-a-go-go-related
184. Iyer V, Roman-Campos D, Sampson KJ, Kang G, Fishman GI, Kass RS. Purkinje cells as gene (HERG) cardiac K⫹ channel. Mol Pharmacol 67: 827– 836, 2005.
sources of arrhythmias in long QT syndrome type 3. Scientific Reports 5: 13287, 2015.
205. Kanki H, Kupershmidt S, Yang T, Wells S, Roden DM. A structural requirement for
185. Iyer V, Sampson KJ, Kass RS. Modeling tissue- and mutation-specific electrophysio- processing the cardiac K⫹ channel KCNQ1. J Biol Chem 279: 33976 –33983, 2004.
logical effects in the long QT syndrome: role of the Purkinje fiber. PLoS One 9: e97720,
206. Kanters JK, Skibsbye L, Hedley PL, Dembic M, Liang B, Hagen CM, Eschen O, Grun-
2014.
net M, Christiansen M, Jespersen T. Combined gating and trafficking defect in Kv11.1
186. James AF, Choisy SC, Hancox JC. Recent advances in understanding sex differences in manifests as a malignant long QT syndrome phenotype in a large Danish p F29L
cardiac repolarization. Prog Biophys Mol Biol 94: 265–319, 2007. founder family. Scand J Clin Lab Invest 75: 699 –709, 2015.

187. January CT, Gong Q, Zhou Z. Long QT syndrome: cellular basis and arrhythmia 207. Kapplinger JD, Tester DJ, Salisbury BA, Carr JL, Harris-Kerr C, Pollevick GD, Wilde
mechanism in LQT2. J Cardiovasc Electrophysiol 11: 1413–1418, 2000. AA, Ackerman MJ. Spectrum and prevalence of mutations from the first 2,500 con-
secutive unrelated patients referred for the FAMILION long QT syndrome genetic
188. Jenke M, Sanchez A, Monje F, Stuhmer W, Weseloh RM, Pardo LA. C-terminal test. Heart Rhythm 6: 1297–1303, 2009.
domains implicated in the functional surface expression of potassium channels. EMBO
208. Kasimova MA, Zaydman MA, Cui J, Tarek M. PIP2-dependent coupling is prominent in
J 22: 395– 403, 2003.
Kv7.1 due to weakened interactions between S4 –S5 and S6. Scientific Reports 5: 7474,
189. Jervell A, Lange-Nielsen F. Congenital deaf-mutism, functional heart disease with 2015.
prolongation of the Q-T interval and sudden death. Am Heart J 54: 59 – 68, 1957.
209. Kass RS, Moss AJ. Long QT syndrome: novel insights into the mechanisms of cardiac
190. Jespersen T, Membrez M, Nicolas CS, Pitard B, Staub O, Olesen SP, Baro I, Abriel H. arrhythmias. J Clin Invest 112: 810 – 815, 2003.
The KCNQ1 potassium channel is down-regulated by ubiquitylating enzymes of the
210. Kass RS, Wiegers SE. The ionic basis of concentration-related effects of noradrenaline
Nedd4/Nedd4-like family. Cardiovasc Res 74: 64 –74, 2007.
on the action potential of calf cardiac Purkinje fibres. J Physiol 322: 541–558, 1982.
191. Jiang C, Atkinson D, Towbin JA, Splawski I, Lehmann MH, Li H, Timothy K, Taggart
211. Kazen-Gillespie KA, Ragsdale DS, D’Andrea MR, Mattei LN, Rogers KE, Isom LL.
RT, Schwartz PJ, Vincent GM. Two long QT syndrome loci map to chromosomes 3
Cloning, localization, and functional expression of sodium channel beta1A subunits. J
and 7 with evidence for further heterogeneity. Nat Genet 8: 141–147, 1994.
Biol Chem 275: 1079 –1088, 2000.
192. Jiang M, Xu X, Wang Y, Toyoda F, Liu XS, Zhang M, Robinson RB, Tseng GN. 212. Keating M, Atkinson D, Dunn C, Timothy K, Vincent GM, Leppert M. Linkage of a
Dynamic partnership between KCNQ1 and KCNE1 and influence on cardiac IKs cardiac arrhythmia, the long QT syndrome, and the Harvey ras-1 gene. Science 252:
current amplitude by KCNE2. J Biol Chem 284: 16452–16462, 2009. 704 –706, 1991.
193. Johnson JN, Ackerman MJ. QTc: how long is too long? Br J Sports Med 43: 657– 662, 213. Keating M, Dunn C, Atkinson D, Timothy K, Vincent GM, Leppert M. Consistent
2009. linkage of the long-QT syndrome to the Harvey ras-1 locus on chromosome 11. Am J
Hum Genet 49: 1335–1339, 1991.
194. Johnson WH Jr, Yang P, Yang T, Lau YR, Mostella BA, Wolff DJ, Roden DM, Benson
DW. Clinical, genetic, and biophysical characterization of a homozygous HERG mu- 214. Kennedy ME, Nemec J, Corey S, Wickman K, Clapham DE. GIRK4 confers appropri-
tation causing severe neonatal long QT syndrome. Pediatr Res 53: 744 –748, 2003. ate processing and cell surface localization to G- protein-gated potassium channels. J
Biol Chem 274: 2571–2582, 1999.
195. Jones DK, Liu F, Vaidyanathan R, Eckhardt LL, Trudeau MC, Robertson GA. hERG 1b
is critical for human cardiac repolarization. Proc Natl Acad Sci USA 111: 18073–18077, 215. Kiehn J, Wible B, Lacerda AE, Brown AM. Mapping the block of a cloned human
2014. inward rectifier potassium channel by dofetilide. Mol Pharmacol 50: 380 –387, 1996.

196. Jones EM, Roti Roti EC, Wang J, Delfosse SA, Robertson GA. Cardiac IKr channels 216. Kim JA, Lopes CM, Moss AJ, McNitt S, Barsheshet A, Robinson JL, Zareba W, Acker-
minimally comprise hERG 1a and 1b subunits. J Biol Chem 279: 44690 – 44694, 2004. man MJ, Kaufman ES, Towbin JA, Vincent M, Goldenberg I. Trigger-specific risk
factors and response to therapy in long QT syndrome type 2. Heart Rhythm 7: 1797–
197. Jons C, Moss AJ, Lopes CM, McNitt S, Zareba W, Goldenberg I, Qi M, Wilde AA, 1805, 2010.
Shimizu W, Kanters JK, Towbin JA, Ackerman MJ, Robinson JL. Mutations in con-
served amino acids in the KCNQ1 channel and risk of cardiac events in type-1 217. Koumi S, Sato R, Nagasawa K, Hayakawa H. Activation of inwardly rectifying potas-
long-QT syndrome. J Cardiovasc Electrophysiol 20: 859 – 865, 2009. sium channels by muscarinic receptor-linked G protein in isolated human ventricular
myocytes. J Membr Biol 157: 71– 81, 1997.
198. Jost N, Virag L, Bitay M, Takacs J, Lengyel C, Biliczki P, Nagy Z, Bogats G, Lathrop DA,
Papp JG, Varro A. Restricting excessive cardiac action potential and QT prolongation: 218. Kovtun O, Tillu VA, Ariotti N, Parton RG, Collins BM. Cavin family proteins and the
a vital role for IKs in human ventricular muscle. Circulation 112: 1392–1399, 2005. assembly of caveolae. J Cell Sci 128: 1269 –1278, 2015.

126 Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org


CONGENITAL LONG QT SYNDROME

219. Krapivinsky G, Gordon EA, Wickman K, Velimirovic B, Krapivinsky L, Clapham DE. 240. Lindegger N, Hagen BM, Marks AR, Lederer WJ, Kass RS. Diastolic transient inward
The G-protein-gated atrial K⫹ channel IKACh is a heteromultimer of two inwardly current in long QT syndrome type 3 is caused by Ca2⫹ overload and inhibited by
rectifying K⫹-channel proteins. Nature 374: 135–141, 1995. ranolazine. J Mol Cell Cardiol 47: 326 –334, 2009.

220. Krumerman A, Gao X, Bian JS, Melman YF, Kagan A, McDonald TV. An LQT mutant 241. Liu DW, Antzelevitch C. Characteristics of the delayed rectifier current (IKr and IKs)
minK alters KvLQT1 trafficking. Am J Physiol Cell Physiol 286: C1453–C1463, 2004. in canine ventricular epicardial, midmyocardial, and endocardial myocytes. A weaker
IKs contributes to the longer action potential of the M cell. Circ Res 76: 351–365, 1995.
221. Krzystanek K, Rasmussen HB, Grunnet M, Staub O, Olesen SP, Abriel H, Jespersen T.
Deubiquitylating enzyme USP2 counteracts Nedd4-2-mediated downregulation of 242. Liu GX, Derst C, Schlichthorl G, Heinen S, Seebohm G, Bruggemann A, Kummer W,
KCNQ1 potassium channels. Heart Rhythm 9: 440 – 448, 2012. Veh RW, Daut J, Preisig-Muller R. Comparison of cloned Kir2 channels with native
inward rectifier K⫹ channels from guinea-pig cardiomyocytes. J Physiol 532: 115–126,
222. Kubo Y, Reuveny E, Slesinger PA, Jan YN, Jan LY. Primary structure and functional
2001.
expression of a rat G-protein-coupled muscarinic potassium channel. Nature 364:
802– 806, 1993. 243. Liu H, Tateyama M, Clancy CE, Abriel H, Kass RS. Channel openings are necessary but
not sufficient for use-dependent block of cardiac Na⫹ channels by flecainide: evidence
223. Kupershmidt S, Yang IC, Hayashi K, Wei J, Chanthaphaychith S, Petersen CI, Johns
from the analysis of disease-linked mutations. J Gen Physiol 120: 39 –51, 2002.
DC, George AL Jr, Roden DM, Balser JR. The IKr drug response is modulated by KCR1
in transfected cardiac and noncardiac cell lines. FASEB J 17: 2263–2265, 2003.
244. Liu JF, Moss AJ, Jons C, Benhorin J, Schwartz PJ, Spazzolini C, Crotti L, Ackerman MJ,
224. Kupershmidt S, Yang T, Chanthaphaychith S, Wang Z, Towbin JA, Roden DM. De- McNitt S, Robinson JL, Qi M, Goldenberg I, Zareba W. Mutation-specific risk in two
fective human Ether-a-go-go-related gene trafficking linked to an endoplasmic retic- genetic forms of type 3 long QT syndrome. Am J Cardiol 105: 210 –213, 2010.

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


ulum retention signal in the C terminus. J Biol Chem 277: 27442–27448, 2002.
245. Liu XK, Katchman A, Whitfield BH, Wan G, Janowski EM, Woosley RL, Ebert SN. In
225. Kurachi Y, Tung RT, Ito H, Nakajima T. G protein activation of cardiac muscarinic K⫹ vivo androgen treatment shortens the QT interval and increases the densities of
channels. Prog Neurobiol 39: 229 –246, 1992. inward and delayed rectifier potassium currents in orchiectomized male rabbits. Car-
diovasc Res 57: 28 –36, 2003.
226. Kurokawa J, Chen L, Kass RS. Requirement of subunit expression for cAMP-mediated
regulation of a heart potassium channel. Proc Natl Acad Sci USA 100: 2122–2127, 2003. 246. Long SB, Campbell EB, Mackinnon R. Crystal structure of a mammalian voltage-
dependent Shaker family K⫹ channel. Science 309: 897–903, 2005.
227. Kurokawa J, Tamagawa M, Harada N, Honda S, Bai CX, Nakaya H, Furukawa T.
Acute effects of oestrogen on the guinea pig and human IKr channels and drug- 247. Long SB, Campbell EB, Mackinnon R. Voltage sensor of Kv1.2: structural basis of
induced prolongation of cardiac repolarization. J Physiol 586: 2961–2973, 2008. electromechanical coupling. Science 309: 903–908, 2005.

228. Kuryshev YA, Ficker E, Wang L, Hawryluk P, Dennis AT, Wible BA, Brown AM, Kang 248. Lopatin AN, Nichols CG. Inward rectifiers in the heart: an update on I(K1). J Mol Cell
J, Chen XL, Sawamura K, Reynolds W, Rampe D. Pentamidine-induced long QT Cardiol 33: 625– 638, 2001.
syndrome and block of hERG trafficking. J Pharmacol Exp Ther 312: 316 –323, 2005.
249. Lopes CM, Zhang H, Rohacs T, Jin T, Yang J, Logothetis DE. Alterations in conserved
229. Labro AJ, Boulet IR, Choveau FS, Mayeur E, Bruyns T, Loussouarn G, Raes AL, Kir channel-PIP2 interactions underlie channelopathies. Neuron 34: 933–944, 2002.
Snyders DJ. The S4 –S5 linker of KCNQ1 channels forms a structural scaffold with the
S6 segment controlling gate closure. J Biol Chem 286: 717–725, 2011. 250. Loussouarn G, Park KH, Bellocq C, Baro I, Charpentier F, Escande D. Phosphatidyl-
inositol-4,5-bisphosphate, PIP2, controls KCNQ1/KCNE1 voltage-gated potassium
230. Laksman ZW, Hamilton RM, Chockalingam P, Ballantyne E, Stephenson EA, Gross GJ, channels: a functional homology between voltage-gated and inward rectifier K⫹ chan-
Gula LJ, Klein GJ, Wilde AA, Krahn AD. Mutation location effect on severity of phe- nels. EMBO J 22: 5412–5421, 2003.
notype during exercise testing in type 1 long-QT syndrome: impact of transmem-
brane and C-loop location. J Cardiovasc Electrophysiol 24: 1015–1020, 2013. 251. Lu Y, Mahaut-Smith MP, Huang CL, Vandenberg JI. Mutant MiRP1 subunits modulate
HERG K⫹ channel gating: a mechanism for pro-arrhythmia in long QT syndrome type
231. Larsen AP, Olesen SP, Grunnet M, Jespersen T. Characterization of hERG1a and 6. J Physiol 551: 253–262, 2003.
hERG1b potassium channels-a possible role for hERG1b in the I (Kr) current. Pflügers
Arch 456: 1137–1148, 2008. 252. Ma D, Wei H, Zhao Y, Lu J, Li G, Sahib NB, Tan TH, Wong KY, Shim W, Wong P, Cook
SA, Liew R. Modeling type 3 long QT syndrome with cardiomyocytes derived from
232. Le Scouarnec S, Bhasin N, Vieyres C, Hund TJ, Cunha SR, Koval O, Marionneau C, patient-specific induced pluripotent stem cells. Int J Cardiol 168: 5277–5286, 2013.
Chen B, Wu Y, Demolombe S, Song LS, Le Marec H, Probst V, Schott JJ, Anderson
ME, Mohler PJ. Dysfunction in ankyrin-B-dependent ion channel and transporter 253. Maier SK, Westenbroek RE, McCormick KA, Curtis R, Scheuer T, Catterall WA.
targeting causes human sinus node disease. Proc Natl Acad Sci USA 105: 15617–15622, Distinct subcellular localization of different sodium channel alpha and beta subunits in
2008. single ventricular myocytes from mouse heart. Circulation 109: 1421–1427, 2004.
233. Lees-Miller JP, Duan Y, Teng GQ, Duff HJ. Molecular determinant of high-affinity 254. Makkar RR, Fromm BS, Steinman RT, Meissner MD, Lehmann MH. Female gender as
dofetilide binding to HERG1 expressed in Xenopus oocytes: involvement of S6 sites. a risk factor for torsades de pointes associated with cardiovascular drugs. JAMA 270:
Mol Pharmacol 57: 367–374, 2000. 2590 –2597, 1993.
234. Lehmann MH, Hardy S, Archibald D, quart B, MacNeil DJ. Sex difference in risk of 255. Marx SO, Kurokawa J, Reiken S, Motoike H, D’Armiento J, Marks AR, Kass RS.
torsade de pointes with D,L-sotalol. Circulation 94: 2535–2541, 1996. Requirement of a macromolecular signaling complex for beta adrenergic receptor
235. Li GR, Feng J, Wang Z, Fermini B, Nattel S. Adrenergic modulation of ultrarapid modulation of the KCNQ1-KCNE1 potassium channel. Science 295: 496 – 499, 2002.
delayed rectifier K⫹ current in human atrial myocytes. Circ Res 78: 903–915, 1996.
256. Matavel A, Medei E, Lopes CM. PKA and PKC partially rescue long QT type 1
236. Li J, Kline CF, Hund TJ, Anderson ME, Mohler PJ. Ankyrin-B regulates Kir6.2 mem- phenotype by restoring channel-PIP2 interactions. Channels 4: 3–11, 2010.
brane expression and function in heart. J Biol Chem 285: 28723–28730, 2010.
257. Mazhari R, Greenstein JL, Winslow RL, Marban E, Nuss HB. Molecular interactions
237. Li W, Du R, Wang QF, Tian L, Yang JG, Song ZF. The G314S KCNQ1 mutation exerts between two long-QT syndrome gene products, HERG and KCNE2, rationalized by
a dominant-negative effect on expression of KCNQ1 channels in oocytes. Biochem in vitro and in silico analysis. Circ Res 89: 33–38, 2001.
Biophys Res Commun 383: 206 –209, 2009.
258. Mazzanti A, Maragna R, Faragli A, Monteforte N, Bloise R, Memmi M, Novelli V,
238. Li Y, Chen L, Kass RS, Dessauer CW. The A-kinase anchoring protein Yotiao facilitates Baiardi P, Bagnardi V, Etheridge SP, Napolitano C, Priori SG. Gene-specific therapy
complex formation between adenylyl cyclase type 9 and the IKs potassium channel in with mexiletine reduces arrhythmic events in patients with long QT syndrome type 3.
heart. J Biol Chem 287: 29815–29824, 2012. J Am Coll Cardiol 67: 1053–1058, 2016.

239. Li Y, Zaydman MA, Wu D, Shi J, Guan M, Virgin-Downey B, Cui J. KCNE1 enhances 259. McDonald TV, Yu Z, Ming Z, Palma E, Meyers MB, Wang KW, Goldstein SA, Fishman
phosphatidylinositol 4,5-bisphosphate (PIP2) sensitivity of IKs to modulate channel GI. A minK-HERG complex regulates the cardiac potassium current I(Kr). Nature 388:
activity. Proc Natl Acad Sci USA 108: 9095–9100, 2011. 289 –292, 1997.

Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org 127


BOHNEN ET AL.

260. McLaughlin S, Wang J, Gambhir A, Murray D. PIP2 and proteins: interactions, organi- 279. Moss AJ, Robinson JL, Gessman L, Gillespie R, Zareba W, Schwartz PJ, Vincent GM,
zation, and information flow. Annu Rev Biophys Biomol Struct 31: 151–175, 2002. Benhorin J, Heilbron EL, Towbin JA, Priori SG, Napolitano C, Zhang L, Medina A,
Andrews ML, Timothy K. Comparison of clinical and genetic variables of cardiac
261. Medeiros-Domingo A, Kaku T, Tester DJ, Iturralde-Torres P, Itty A, Ye B, Valdivia C, events associated with loud noise versus swimming among subjects with the long QT
Ueda K, Canizales-Quinteros S, Tusie-Luna MT, Makielski JC, Ackerman MJ. SCN4B- syndrome. Am J Cardiol 84: 876 – 879, 1999.
encoded sodium channel beta4 subunit in congenital long-QT syndrome. Circulation
116: 134 –142, 2007. 280. Moss AJ, Shimizu W, Wilde AA, Towbin JA, Zareba W, Robinson JL, Qi M, Vincent
GM, Ackerman MJ, Kaufman ES, Hofman N, Seth R, Kamakura S, Miyamoto Y, Gold-
262. Melman YF, Um SY, Krumerman A, Kagan A, McDonald TV. KCNE1 binds to the enberg I, Andrews ML, McNitt S. Clinical aspects of type-1 long-QT syndrome by
KCNQ1 pore to regulate potassium channel activity. Neuron 42: 927–937, 2004. location, coding type, and biophysical function of mutations involving the KCNQ1
gene. Circulation 115: 2481–2489, 2007.
263. Meng J, Shi C, Li L, Du Y, Xu Y. Compound ICA-105574 prevents arrhythmias
induced by cardiac delayed repolarization. Eur J Pharmacol 718: 87–97, 2013. 281. Moss AJ, Windle JR, Hall WJ, Zareba W, Robinson JL, McNitt S, Severski P, Rosero S,
Daubert JP, Qi M, Cieciorka M, Manalan AS. Safety and efficacy of flecainide in
264. Miake J, Marban E, Nuss HB. Functional role of inward rectifier current in heart
subjects with Long QT-3 syndrome (DeltaKPQ mutation): a randomized, double-
probed by Kir2.1 overexpression and dominant-negative suppression. J Clin Invest
blind, placebo-controlled clinical trial. Ann Noninvasive Electrocardiol 10: 59 – 66, 2005.
111: 1529 –1536, 2003.
282. Moss AJ, Zareba W, Hall WJ, Schwartz PJ, Crampton RS, Benhorin J, Vincent GM,
265. Migdalovich D, Moss AJ, Lopes CM, Costa J, Ouellet G, Barsheshet A, McNitt S,
Locati EH, Priori SG, Napolitano C, Medina A, Zhang L, Robinson JL, Timothy K,
Polonsky S, Robinson JL, Zareba W, Ackerman MJ, Benhorin J, Kaufman ES, Platonov
Towbin JA, Andrews ML. Effectiveness and limitations of beta-blocker therapy in
PG, Shimizu W, Towbin JA, Vincent GM, Wilde AA, Goldenberg I. Mutation and

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


congenital long-QT syndrome. Circulation 101: 616 – 623, 2000.
gender specific risk in type-2 long QT syndrome: implications for risk stratification for
life-threatening cardiac events in patients with long QT syndrome. Heart Rhythm 8: 283. Moss AJ, Zareba W, Kaufman ES, Gartman E, Peterson DR, Benhorin J, Towbin JA,
1537–1543, 2011. Keating MT, Priori SG, Schwartz PJ, Vincent GM, Robinson JL, Andrews ML, Feng C,
Hall WJ, Medina A, Zhang L, Wang Z. Increased risk of arrhythmic events in long-QT
266. Mitcheson JS, Chen J, Lin M, Culberson C, Sanguinetti MC. A structural basis for syndrome with mutations in the pore region of the human ether-a-go-go-related gene
drug-induced long QT syndrome. Proc Natl Acad Sci USA 97: 12329 –12333, 2000. potassium channel. Circulation 105: 794 –799, 2002.
267. Mitcheson JS, Chen J, Sanguinetti MC. Trapping of a methanesulfonanilide by closure 284. Moss AJ, Zareba W, Schwarz KQ, Rosero S, McNitt S, Robinson JL. Ranolazine short-
of the HERG potassium channel activation gate. J Gen Physiol 115: 229 –240, 2000. ens repolarization in patients with sustained inward sodium current due to type-3
long-QT syndrome. J Cardiovasc Electrophysiol 19: 1289 –1293, 2008.
268. Miyamoto K, Nakagawa T, Kuroda Y. Solution structures of the cytoplasmic linkers
between segments S4 and S5 (S4 –S5) in domains III and IV of human brain sodium 285. Motoike HK, Liu H, Glaaser IW, Yang AS, Tateyama M, Kass RS. The Na⫹ channel
channels in SDS micelles. J Pept Res 58: 193–203, 2001. inactivation gate is a molecular complex: a novel role of the COOH-terminal domain.
J Gen Physiol 123: 155–165, 2004.
269. Mohler PJ, Davis JQ, Bennett V. Ankyrin-B coordinates the Na/K ATPase, Na/Ca
exchanger, and InsP3 receptor in a cardiac T-tubule/SR microdomain. PLoS Biol 3: 286. Murray A, Donger C, Fenske C, Spillman I, Richard P, Dong YB, Neyroud N, Cheva-
e423, 2005. lier P, Denjoy I, Carter N, Syrris P, Afzal AR, Patton MA, Guicheney P, Jeffery S.
Splicing mutations in KCNQ1: a mutation hot spot at codon 344 that produces in
270. Mohler PJ, Davis JQ, Davis LH, Hoffman JA, Michaely P, Bennett V. Inositol 1,4,5-
frame transcripts. Circulation 100: 1077–1084, 1999.
trisphosphate receptor localization and stability in neonatal cardiomyocytes requires
interaction with ankyrin-B. J Biol Chem 279: 12980 –12987, 2004. 287. Muskett FW, Thouta S, Thomson SJ, Bowen A, Stansfeld PJ, Mitcheson JS. Mechanistic
insight into human ether-a-go-go-related gene (hERG) K⫹ channel deactivation gating
271. Mohler PJ, Le Scouarnec S, Denjoy I, Lowe JS, Guicheney P, Caron L, Driskell IM,
from the solution structure of the EAG domain. J Biol Chem 286: 6184 – 6191, 2011.
Schott JJ, Norris K, Leenhardt A, Kim RB, Escande D, Roden DM. Defining the cellular
phenotype of “ankyrin-B syndrome” variants: human ANK2 variants associated with 288. Nagatomo T, January CT, Makielski JC. Preferential block of late sodium current in
clinical phenotypes display a spectrum of activities in cardiomyocytes. Circulation 115: the LQT3 DeltaKPQ mutant by the class I(C) antiarrhythmic flecainide. Mol Pharmacol
432– 441, 2007. 57: 101–107, 2000.
272. Mohler PJ, Rivolta I, Napolitano C, LeMaillet G, Lambert S, Priori SG, Bennett V. 289. Nagatomo T, January CT, Ye B, Abe H, Nakashima Y, Makielski JC. Rate-dependent
Nav1.5 E1053K mutation causing Brugada syndrome blocks binding to ankyrin-G and QT shortening mechanism for the LQT3 deltaKPQ mutant. Cardiovasc Res 54: 624 –
expression of Nav1.5 on the surface of cardiomyocytes. Proc Natl Acad Sci USA 101: 629, 2002.
17533–17538, 2004.
290. Nakajima T, Furukawa T, Tanaka T, Katayama Y, Nagai R, Nakamura Y, Hiraoka M.
273. Mohler PJ, Schott JJ, Gramolini AO, Dilly KW, Guatimosim S, duBell WH, Song LS, Novel mechanism of HERG current suppression in LQT2: shift in voltage dependence
Haurogne K, Kyndt F, Ali ME, Rogers TB, Lederer WJ, Escande D, Le Marec H, of HERG inactivation. Circ Res 83: 415– 422, 1998.
Bennett V. Ankyrin-B mutation causes type 4 long-QT cardiac arrhythmia and sudden
cardiac death. Nature 421: 634 – 639, 2003. 291. Nakajima T, Hayashi K, Viswanathan PC, Kim MY, Anghelescu M, Barksdale KA, Shuai
W, Balser JR, Kupershmidt S. HERG is protected from pharmacological block by
274. Morais Cabral JH, Lee A, Cohen SL, Chait BT, Li M, Mackinnon R. Crystal structure alpha-1,2-glucosyltransferase function. J Biol Chem 282: 5506 –5513, 2007.
and functional analysis of the HERG potassium channel N terminus: a eukaryotic PAS
domain. Cell 95: 649 – 655, 1998. 292. Nakajo K, Kubo Y. Steric hindrance between S4 and S5 of the KCNQ1/KCNE1
channel hampers pore opening. Nat Commun 5: 4100, 2014.
275. Moreno JD, Yang PC, Bankston JR, Grandi E, Bers DM, Kass RS, Clancy CE. Ranola-
zine for congenital and acquired late INa-linked arrhythmias: in silico pharmacological 293. Nakajo K, Ulbrich MH, Kubo Y, Isacoff EY. Stoichiometry of the KCNQ1-KCNE1 ion
screening. Circ Res 113: e50 – 61, 2013. channel complex. Proc Natl Acad Sci USA 107: 18862–18867, 2010.

276. Moreno JD, Zhu ZI, Yang PC, Bankston JR, Jeng MT, Kang C, Wang L, Bayer JD, 294. Nakamura H, Kurokawa J, Bai CX, Asada K, Xu J, Oren RV, Zhu ZI, Clancy CE, Isobe
Christini DJ, Trayanova NA, Ripplinger CM, Kass RS, Clancy CE. A computational M, Furukawa T. Progesterone regulates cardiac repolarization through a nongenomic
model to predict the effects of class I anti-arrhythmic drugs on ventricular rhythms. Sci pathway: an in vitro patch-clamp and computational modeling study. Circulation 116:
Transl Med 3: 98ra83, 2011. 2913–2922, 2007.

277. Moretti A, Bellin M, Welling A, Jung CB, Lam JT, Bott-Flugel L, Dorn T, Goedel A, 295. Napolitano C, Priori SG, Schwartz PJ, Bloise R, Ronchetti E, Nastoli J, Bottelli G,
Hohnke C, Hofmann F, Seyfarth M, Sinnecker D, Schomig A, Laugwitz KL. Patient- Cerrone M, Leonardi S. Genetic testing in the long QT syndrome: development and
specific induced pluripotent stem-cell models for long-QT syndrome. N Engl J Med validation of an efficient approach to genotyping in clinical practice. JAMA 294: 2975–
363: 1397–1409, 2010. 2980, 2005.

278. Morin TJ, Kobertz WR. Counting membrane-embedded KCNE beta-subunits in func- 296. Nattel S, Burstein B, Dobrev D. Atrial remodeling and atrial fibrillation: mechanisms
tioning K⫹ channel complexes. Proc Natl Acad Sci USA 105: 1478 –1482, 2008. and implications. Circ Arrhythm Electrophysiol 1: 62–73, 2008.

128 Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org


CONGENITAL LONG QT SYNDROME

297. Nawathe PA, Kryukova Y, Oren RV, Milanesi R, Clancy CE, Lu JT, Moss AJ, Difran- 318. Perissinotti LL, Guo J, De Biase PM, Clancy CE, Duff HJ, Noskov SY. Kinetic model for
cesco D, Robinson RB. An LQTS6 MiRP1 mutation suppresses pacemaker current NS1643 drug activation of WT and L529I variants of Kv11.1 (hERG1) potassium
and is associated with sinus bradycardia. J Cardiovasc Electrophysiol 24: 1021–1027, channel. Biophys J 108: 1414 –1424, 2015.
2013.
319. Perry M, de Groot MJ, Helliwell R, Leishman D, Tristani-Firouzi M, Sanguinetti MC,
298. Nemec J, Buncova M, Shusterman V, Winter B, Shen WK, Ackerman MJ. QT interval Mitcheson J. Structural determinants of HERG channel block by clofilium and ibutilide.
variability and adaptation to heart rate changes in patients with long QT syndrome. Mol Pharmacol 66: 240 –249, 2004.
Pacing Clin Electrophysiol 32: 72– 81, 2009.
320. Perry M, Sachse FB, Abbruzzese J, Sanguinetti MC. PD-118057 contacts the pore
299. Nerbonne JM, Kass RS. Molecular physiology of cardiac repolarization. Physiol Rev 85: helix of hERG1 channels to attenuate inactivation and enhance K⫹ conductance. Proc
1205–1253, 2005. Natl Acad Sci USA 106: 20075–20080, 2009.
300. Newton-Cheh C, Eijgelsheim M, Rice KM, de Bakker PI, Yin X, Estrada K, Bis JC, 321. Perry M, Sachse FB, Sanguinetti MC. Structural basis of action for a human ether-a-
Marciante K, Rivadeneira F, Noseworthy PA, Sotoodehnia N, Smith NL, Rotter JI,
go-go-related gene 1 potassium channel activator. Proc Natl Acad Sci USA 104: 13827–
Kors JA, Witteman JC, Hofman A, Heckbert SR, O’Donnell CJ, Uitterlinden AG,
13832, 2007.
Psaty BM, Lumley T, Larson MG, Stricker BH. Common variants at ten loci
influence QT interval duration in the QTGEN Study. Nat Genet 41: 399 – 406, 322. Petersen CI, McFarland TR, Stepanovic SZ, Yang P, Reiner DJ, Hayashi K, George AL,
2009. Roden DM, Thomas JH, Balser JR. In vivo identification of genes that modify ether-a-
go-go-related gene activity in Caenorhabditis elegans may also affect human cardiac
301. Neyroud N, Tesson F, Denjoy I, Leibovici M, Donger C, Barhanin J, Faure S, Gary F,
arrhythmia. Proc Natl Acad Sci USA 101: 11773–11778, 2004.
Coumel P, Petit C, Schwartz K, Guicheney P. A novel mutation in the potassium

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


channel gene KVLQT1 causes the Jervell and Lange-Nielsen cardioauditory syn-
323. Phartiyal P, Jones EM, Robertson GA. Heteromeric assembly of human ether-a-go-
drome. Nat Genet 15: 186 –189, 1997.
go-related gene (hERG) 1a/1b channels occurs cotranslationally via N-terminal inter-
302. Ng CA, Hunter MJ, Perry MD, Mobli M, Ke Y, Kuchel PW, King GF, Stock D, Van- actions. J Biol Chem 282: 9874 –9882, 2007.
denberg JI. The N-terminal tail of hERG contains an amphipathic alpha-helix that
324. Pignier C, Rougier JS, Vie B, Culie C, Verscheure Y, Vacher B, Abriel H, Le Grand B.
regulates channel deactivation. PLoS One 6: e16191, 2011.
Selective inhibition of persistent sodium current by F 15845 prevents ischaemia-
303. Nichols CG, Makhina EN, Pearson WL, Sha Q, Lopatin AN. Inward rectification and induced arrhythmias. Br J Pharmacol 161: 79 –91, 2010.
implications for cardiac excitability. Circ Res 78: 1–7, 1996.
325. Piper DR, Sanguinetti MC, Tristani-Firouzi M. Voltage sensor movement in the hERG
304. Noble D. The Initiation of the Heartbeat. Oxford, UK: Clarendon, 1978. K⫹ channel. Novartis Found Symp 266: 46 –52, 2005.

305. Noble D, Tsien RW. Outward membrane currents activated in the plateau range of 326. Plant LD, Xiong D, Dai H, Goldstein SA. Individual IKs channels at the surface of
potentials in cardiac Purkinje fibres. J Physiol 200: 205–231, 1969. mammalian cells contain two KCNE1 accessory subunits. Proc Natl Acad Sci USA 111:
E1438 –1446, 2014.
306. Nogami A, Sugiyasu A, Kubota S, Kato K. Mapping and ablation of idiopathic ventric-
ular fibrillation from the Purkinje system. Heart Rhythm 2: 646 – 649, 2005. 327. Plaster NM, Tawil R, Tristani-Firouzi M, Canun S, Bendahhou S, Tsunoda A, Donald-
son MR, Iannaccone ST, Brunt E, Barohn R, Clark J, Deymeer F, George AL Jr, Fish FA,
307. Odening KE, Choi BR, Liu GX, Hartmann K, Ziv O, Chaves L, Schofield L, Centracchio Hahn A, Nitu A, Ozdemir C, Serdaroglu P, Subramony SH, Wolfe G, Fu YH, Ptacek LJ.
J, Zehender M, Peng X, Brunner M, Koren G. Estradiol promotes sudden cardiac
Mutations in Kir21 cause the developmental and episodic electrical phenotypes of
death in transgenic long QT type 2 rabbits while progesterone is protective. Heart
Andersen’s syndrome. Cell 105: 511–519, 2001.
Rhythm 9: 823– 832, 2012.
328. Postema PG, Ritsema van Eck HJ, Opthof T, van Herpen G, van Dessel PF, Priori SG,
308. Osteen JD, Barro-Soria R, Robey S, Sampson KJ, Kass RS, Larsson HP. Allosteric gating
Wolpert C, Borggrefe M, Kors JA, Wilde AA. IK1 modulates the U-wave: insights in a
mechanism underlies the flexible gating of KCNQ1 potassium channels. Proc Natl Acad
100-year-old enigma. Heart Rhythm 6: 393– 400, 2009.
Sci USA 109: 7103–7108, 2012.
329. Poulsen KL, Hotait M, Calloe K, Klaerke DA, Rebeiz A, Nemer G, Tejada MA,
309. Osteen JD, Gonzalez C, Sampson KJ, Iyer V, Rebolledo S, Larsson HP, Kass RS.
KCNE1 alters the voltage sensor movements necessary to open the KCNQ1 channel Refaat MM. The mutation PT613a in the pore helix of the Kv 11.1 potassium
gate. Proc Natl Acad Sci USA 107: 22710 –22715, 2010. channel is associated with long QT syndrome. Pacing Clin Electrophysiol 38: 1304 –
1309, 2015.
310. Panaghie G, Abbott GW. The role of S4 charges in voltage-dependent and voltage-
independent KCNQ1 potassium channel complexes. J Gen Physiol 129: 121–133, 330. Pourrier M, Zicha S, Ehrlich J, Han W, Nattel S. Canine ventricular KCNE2 expression
2007. resides predominantly in Purkinje fibers. Circ Res 93: 189 –191, 2003.

311. Panaghie G, Tai KK, Abbott GW. Interaction of KCNE subunits with the KCNQ1 K⫹ 331. Priori SG. From trials to guidelines to clinical practice: the need for improvement.
channel pore. J Physiol 570: 455– 467, 2006. Europace 6: 176 –178, 2004.

312. Park KH, Piron J, Dahimene S, Merot J, Baro I, Escande D, Loussouarn G. Impaired 332. Priori SG, Cantu F, Schwartz PJ. the long QT syndrome–new diagnostic and thera-
KCNQ1-KCNE1 and phosphatidylinositol-4,5-bisphosphate interaction underlies the peutic approach in the era of molecular biology. Schweiz Med Wochenschr 126: 1727–
long QT syndrome. Circ Res 96: 730 –739, 2005. 1731, 1996.

313. Patton DE, West JW, Catterall WA, Goldin AL. Amino acid residues required for fast 333. Priori SG, Napolitano C, Cantu F, Brown AM, Schwartz PJ. Differential response to
Na(⫹)-channel inactivation: charge neutralizations and deletions in the III-IV linker. Na⫹ channel blockade, beta-adrenergic stimulation and rapid pacing in a cellular
Proc Natl Acad Sci USA 89: 10905–10909, 1992. model mimicking the SCN5A and HERG defects present in the long-QT syndrome.
Circ Res 78: 1009 –1015, 1996.
314. Paulussen A, Raes A, Matthijs G, Snyders DJ, Cohen N, Aerssens J. A novel mutation
(T65P) in the PAS domain of the human potassium channel HERG results in the long 334. Priori SG, Napolitano C, Schwartz PJ, Grillo M, Bloise R, Ronchetti E, Moncalvo C,
QT syndrome by trafficking deficiency. J Biol Chem 277: 48610 – 48616, 2002. Tulipani C, Veia A, Bottelli G, Nastoli J. Association of long QT syndrome loci and
cardiac events among patients treated with beta-blockers. JAMA 292: 1341–1344,
315. Payandeh J, Scheuer T, Zheng N, Catterall WA. The crystal structure of a voltage-
2004.
gated sodium channel. Nature 475: 353–358, 2011.
335. Priori SG, Wilde AA, Horie M, Cho Y, Behr ER, Berul C, Blom N, Brugada J, Chiang
316. Pearson PJ, Lin PJ, Schaff HV. Production of endothelium-derived contracting factor is
enhanced after coronary reperfusion. Ann Thoracic Surg 51: 788 –793, 1991. CE, Huikuri H, Kannankeril P, Krahn A, Leenhardt A, Moss A, Schwartz PJ, Shimizu W,
Tomaselli G, Tracy C. HRS/EHRA/APHRS expert consensus statement on the diag-
317. Perez Riera AR, Ferreira C, Filho CF, Ferreira M, Meneghini A, Uchida AH, Schapach- nosis and management of patients with inherited primary arrhythmia syndromes:
nik E, Dubner S, Zhang L. The enigmatic sixth wave of the electrocardiogram: the U document endorsed by HRS, EHRA, and APHRS in May 2013 and by ACCF, AHA,
wave. Cardiol J 15: 408 – 421, 2008. PACES, and AEPC in June 2013. Heart Rhythm 10: 1932–1963, 2013.

Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org 129


BOHNEN ET AL.

336. Ragsdale DS, McPhee JC, Scheuer T, Catterall WA. Molecular determinants of state- 355. Sanguinetti MC. Dysfunction of delayed rectifier potassium channels in an inherited
dependent block of Na⫹ channels by local anesthetics. Science 265: 1724 –1728, cardiac arrhythmia. Ann NY Acad Sci 868: 406 – 413, 1999.
1994.
356. Sanguinetti MC. HERG1 channelopathies. Pflügers Arch 460: 265–276, 2010.
337. Rajamani S, Eckhardt LL, Valdivia CR, Klemens CA, Gillman BM, Anderson CL, Hol-
zem KM, Delisle BP, Anson BD, Makielski JC, January CT. Drug-induced long QT 357. Sanguinetti MC, Curran ME, Spector PS, Keating MT. Spectrum of HERG K⫹-channel
syndrome: hERG K⫹ channel block and disruption of protein trafficking by fluoxetine dysfunction in an inherited cardiac arrhythmia. Proc Natl Acad Sci USA 93: 2208 –2212,
and norfluoxetine. Br J Pharmacol 149: 481– 489, 2006. 1996.

338. Ramirez AH, Schildcrout JS, Blakemore DL, Masys DR, Pulley JM, Basford MA, Roden 358. Sanguinetti MC, Curran ME, Zou A, Shen J, Spector PS, Atkinson DL, Keating MT.
DM, Denny JC. Modulators of normal electrocardiographic intervals identified in a Coassembly of K(V)LQT1 and minK (IsK) proteins to form cardiac I(Ks) potassium
large electronic medical record. Heart Rhythm 8: 271–277, 2011. channel. Nature 384: 80 – 83, 1996.

359. Sanguinetti MC, Jiang C, Curran ME, Keating MT. A mechanistic link between an
339. Refaat MM, Hotait M, Tseng ZH. Utility of the exercise electrocardiogram testing in
inherited and an acquired cardiac arrhythmia: HERG encodes the IKr potassium
sudden cardiac death risk stratification. Ann Noninvasive Electrocardiol 19: 311–318,
channel. Cell 81: 299 –307, 1995.
2014.
360. Sanguinetti MC, Jurkiewicz NK. Two components of cardiac delayed rectifier K⫹
340. Restier L, Cheng L, Sanguinetti MC. Mechanisms by which atrial fibrillation-associated
current. Differential sensitivity to block by class III antiarrhythmic agents. J Gen Physiol
mutations in the S1 domain of KCNQ1 slow deactivation of IKs channels. J Physiol 586:
96: 195–215, 1990.
4179 – 4191, 2008.

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


361. Sanguinetti MC, Tristani-Firouzi M. hERG potassium channels and cardiac arrhythmia.
341. Reuveny E, Slesinger PA, Inglese J, Morales JM, Iniguez-Lluhi JA, Lefkowitz RJ, Bourne
Nature 440: 463– 469, 2006.
HR, Jan YN, Jan LY. Activation of the cloned muscarinic potassium channel by G
protein beta gamma subunits. Nature 370: 143–146, 1994. 362. Sansone V, Griggs RC, Meola G, Ptacek LJ, Barohn R, Iannaccone S, Bryan W, Baker N,
Janas SJ, Scott W, Ririe D, Tawil R. Andersen’s syndrome: a distinct periodic paralysis.
342. Riuro H, Campuzano O, Arbelo E, Iglesias A, Batlle M, Perez-Villa F, Brugada J, Perez
Ann Neurol 42: 305–312, 1997.
GJ, Scornik FS, Brugada R. A missense mutation in the sodium channel beta1b subunit
reveals SCN1B as a susceptibility gene underlying long QT syndrome. Heart Rhythm 363. Sasano T, Ueda K, Orikabe M, Hirano Y, Kawano S, Yasunami M, Isobe M, Kimura A,
11: 1202–1209, 2014. Hiraoka M. Novel C-terminus frameshift mutation, 1122fs/147, of HERG in LQT2:
additional amino acids generated by frameshift cause accelerated inactivation. J Mol
343. Rivolta I, Abriel H, Tateyama M, Liu H, Memmi M, Vardas P, Napolitano C, Priori SG,
Cell Cardiol 37: 1205–1211, 2004.
Kass RS. Inherited Brugada and long QT-3 syndrome mutations of a single residue of
the cardiac sodium channel confer distinct channel and clinical phenotypes. J Biol Chem 364. Schmitt N, Schwarz M, Peretz A, Abitbol I, Attali B, Pongs O. A recessive C-terminal
276: 30623–30630, 2001. Jervell and Lange-Nielsen mutation of the KCNQ1 channel impairs subunit assembly.
EMBO J 19: 332–340, 2000.
344. Rivolta I, Clancy CE, Tateyama M, Liu H, Priori SG, Kass RS. A novel SCN5A mutation
associated with long QT-3: altered inactivation kinetics and channel dysfunction. 365. Schonherr R, Heinemann SH. Molecular determinants for activation and inactivation
Physiol Genomics 10: 191–197, 2002. of HERG, a human inward rectifier potassium channel. J Physiol 493: 635– 642, 1996.

345. Robertson GA, January CT. HERG trafficking and pharmacological rescue of LQTS-2 366. Schott JJ, Charpentier F, Peltier S, Foley P, Drouin E, Bouhour JB, Donnelly P, Verg-
mutant channels. Handb Exp Pharmacol 349 –355, 2006. naud G, Bachner L, Moisan JP, et al. Mapping of a gene for long QT syndrome to
chromosome 4q25-27. Am J Hum Genet 57: 1114 –1122, 1995.
346. Romano C, Gemme G, Pongiglione R. Rare cardiac arrhythmias of the pediatric age. I.
Repetitive paroxysmal tachycardia. Minerva Pediatr 15: 1155–1164, 1963. 367. Schroeder BC, Waldegger S, Fehr S, Bleich M, Warth R, Greger R, Jentsch TJ. A
constitutively open potassium channel formed by KCNQ1 and KCNE3. Nature 403:
347. Ruan Y, Liu N, Bloise R, Napolitano C, Priori SG. Gating properties of SCN5A muta- 196 –199, 2000.
tions and the response to mexiletine in long-QT syndrome type 3 patients. Circulation
116: 1137–1144, 2007. 368. Schwartz PJ, Moss AJ, Vincent GM, Crampton RS. Diagnostic criteria for the long QT
syndrome. An update. Circulation 88: 782–784, 1993.
348. Ruscic KJ, Miceli F, Villalba-Galea CA, Dai H, Mishina Y, Bezanilla F, Goldstein SA. IKs
channels open slowly because KCNE1 accessory subunits slow the movement of S4 369. Schwartz PJ, Priori SG, Cerrone M, Spazzolini C, Odero A, Napolitano C, Bloise R, De
voltage sensors in KCNQ1 pore-forming subunits. Proc Natl Acad Sci USA 110: E559 – Ferrari GM, Klersy C, Moss AJ, Zareba W, Robinson JL, Hall WJ, Brink PA, Toivonen
566, 2013. L, Epstein AE, Li C, Hu D. Left cardiac sympathetic denervation in the management of
high-risk patients affected by the long-QT syndrome. Circulation 109: 1826 –1833,
349. Ruwald MH, Xu Parks X, Moss AJ, Zareba W, Baman J, McNitt S, Kanters JK, Shimizu 2004.
W, Wilde AA, Jons C, Lopes CM. Stop-codon and C-terminal nonsense mutations are
associated with a lower risk of cardiac events in patients with long QT syndrome type 370. Schwartz PJ, Priori SG, Locati EH, Napolitano C, Cantu F, Towbin JA, Keating MT,
1. Heart Rhythm 13: 122–131, 2016. Hammoude H, Brown AM, Chen LS. Long QT syndrome patients with mutations of
the SCN5A and HERG genes have differential responses to Na⫹ channel blockade and
350. Ryan JJ, Kalscheur M, Dellefave L, McNally E, Archer SL. A KCNE1 missense variant to increases in heart rate. Implications for gene-specific therapy. Circulation 92: 3381–
(V47I) causing exercise-induced long QT syndrome (Romano Ward). Int J Cardiol 156: 3386, 1995.
e33–35, 2012.
371. Schwartz PJ, Priori SG, Spazzolini C, Moss AJ, Vincent GM, Napolitano C, Denjoy I,
351. Sakagami M, Fukazawa K, Matsunaga T, Fujita H, Mori N, Takumi T, Ohkubo H, Guicheney P, Breithardt G, Keating MT, Towbin JA, Beggs AH, Brink P, Wilde AA,
Nakanishi S. Cellular localization of rat Isk protein in the stria vascularis by immuno- Toivonen L, Zareba W, Robinson JL, Timothy KW, Corfield V, Wattanasirichaigoon D,
histochemical observation. Hear Res 56: 168 –172, 1991. Corbett C, Haverkamp W, Schulze-Bahr E, Lehmann MH, Schwartz K, Coumel P,
Bloise R. Genotype-phenotype correlation in the long-QT syndrome: gene-specific
352. Salata JJ, Jurkiewicz NK, Jow B, Folander K, Guinosso PJ Jr, Raynor B, Swanson R, triggers for life-threatening arrhythmias. Circulation 103: 89 –95, 2001.
Fermini B. IK of rabbit ventricle is composed of two currents: evidence for IKs. Am J
Physiol Heart Circ Physiol 271: H2477–H2489, 1996. 372. Schwartz PJ, Spazzolini C, Priori SG, Crotti L, Vicentini A, Landolina M, Gasparini M,
Wilde AA, Knops RE, Denjoy I, Toivonen L, Monnig G, Al-Fayyadh M, Jordaens L,
353. Sale H, Wang J, O’Hara TJ, Tester DJ, Phartiyal P, He JQ, Rudy Y, Ackerman MJ, Borggrefe M, Holmgren C, Brugada P, De Roy L, Hohnloser SH, Brink PA. Who are
Robertson GA. Physiological properties of hERG 1a/1b heteromeric currents and a the long-QT syndrome patients who receive an implantable cardioverter-defibrillator
hERG 1b-specific mutation associated with Long-QT syndrome. Circ Res 103: e81–95, and what happens to them?: data from the European Long-QT Syndrome Implantable
2008. Cardioverter-Defibrillator (LQTS ICD) Registry. Circulation 122: 1272–1282, 2010.

354. Sampson KJ, Kass RS. Molecular mechanisms of adrenergic stimulation in the heart. 373. Schwartz PJ, Vanoli E, Crotti L, Spazzolini C, Ferrandi C, Goosen A, Hedley P,
Heart Rhythm 7: 1151–1153, 2010. Heradien M, Bacchini S, Turco A, La Rovere MT, Bartoli A, George AL Jr, Brink PA.

130 Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org


CONGENITAL LONG QT SYNDROME

Neural control of heart rate is an arrhythmia risk modifier in long QT syndrome. J Am 392. Silver ES, Liberman L, Chung WK, Spotnitz HM, Chen JM, Ackerman MJ, Moir C,
Coll Cardiol 51: 920 –929, 2008. Hordof AJ, Pass RH. Long QT syndrome due to a novel mutation in SCN5A: treat-
ment with ICD placement at 1 month and left cardiac sympathetic denervation at 3
374. Seebohm G, Strutz-Seebohm N, Birkin R, Dell G, Bucci C, Spinosa MR, Baltaev R, months of age. J Interv Card Electrophysiol 26: 41– 45, 2009.
Mack AF, Korniychuk G, Choudhury A, Marks D, Pagano RE, Attali B, Pfeufer A, Kass
RS, Sanguinetti MC, Tavare JM, Lang F. Regulation of endocytic recycling of KCNQ1/ 393. Smith JA, Vanoye CG, George AL Jr, Meiler J, Sanders CR. Structural models for the
KCNE1 potassium channels. Circ Res 100: 686 – 692, 2007. KCNQ1 voltage-gated potassium channel. Biochemistry 46: 14141–14152, 2007.

375. Seebohm G, Strutz-Seebohm N, Ureche ON, Henrion U, Baltaev R, Mack AF, Korni- 394. Smith PL, Baukrowitz T, Yellen G. The inward rectification mechanism of the HERG
ychuk G, Steinke K, Tapken D, Pfeufer A, Kaab S, Bucci C, Attali B, Merot J, Tavare JM, cardiac potassium channel. Nature 379: 833– 836, 1996.
Hoppe UC, Sanguinetti MC, Lang F. Long QT syndrome-associated mutations in
395. Smits JP, Veldkamp MW, Bezzina CR, Bhuiyan ZA, Wedekind H, Schulze-Bahr E,
KCNQ1 and KCNE1 subunits disrupt normal endosomal recycling of IKs channels.
Wilde AA. Substitution of a conserved alanine in the domain IIIS4 –S5 linker of the
Circ Res 103: 1451–1457, 2008.
cardiac sodium channel causes long QT syndrome. Cardiovasc Res 67: 459 – 466, 2005.
376. Sesti F, Abbott GW, Wei J, Murray KT, Saksena S, Schwartz PJ, Priori SG, Roden DM,
396. Song KS, Scherer PE, Tang Z, Okamoto T, Li S, Chafel M, Chu C, Kohtz DS, Lisanti
George AL Jr, Goldstein SA. A common polymorphism associated with antibiotic-
MP. Expression of caveolin-3 in skeletal, cardiac, and smooth muscle cells. Caveolin-3
induced cardiac arrhythmia. Proc Natl Acad Sci USA 97: 10613–10618, 2000. is a component of the sarcolemma and co-fractionates with dystrophin and dystro-
phin-associated glycoproteins. J Biol Chem 271: 15160 –15165, 1996.
377. Seth R, Moss AJ, McNitt S, Zareba W, Andrews ML, Qi M, Robinson JL, Goldenberg
I, Ackerman MJ, Benhorin J, Kaufman ES, Locati EH, Napolitano C, Priori SG, 397. Song W, Xiao Y, Chen H, Ashpole NM, Piekarz AD, Ma P, Hudmon A, Cummins TR,

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


Schwartz PJ, Towbin JA, Vincent GM, Zhang L. Long QT syndrome and pregnancy. J Shou W. The human Nav1.5 F1486 deletion associated with long QT syndrome leads
Am Coll Cardiol 49: 1092–1098, 2007. to impaired sodium channel inactivation and reduced lidocaine sensitivity. J Physiol
590: 5123–5139, 2012.
378. Shamgar L, Haitin Y, Yisharel I, Malka E, Schottelndreier H, Peretz A, Paas Y, Attali B.
KCNE1 constrains the voltage sensor of Kv7.1 K⫹ channels. PLoS One 3: e1943, 2008. 398. Soom M, Schonherr R, Kubo Y, Kirsch C, Klinger R, Heinemann SH. Multiple PIP2
binding sites in Kir2.1 inwardly rectifying potassium channels. FEBS Lett 490: 49 –53,
379. Shamgar L, Ma L, Schmitt N, Haitin Y, Peretz A, Wiener R, Hirsch J, Pongs O, Attali B.
2001.
Calmodulin Is Essential for Cardiac IKS Channel Gating and Assembly. Impaired Func-
tion in Long-QT Mutations. Circ Res 98: 1055–1063, 2006. 399. Sorensen AB, Sondergaard MT, Overgaard MT. Calmodulin in a heartbeat. FEBS J
280: 5511–5532, 2013.
380. Sheets MF, Chen T, Hanck DA. Outward stabilization of the voltage sensor in domain
II but not domain I speeds inactivation of voltage-gated sodium channels. Am J Physiol 400. Spector PS, Curran ME, Keating MT, Sanguinetti MC. Class III antiarrhythmic drugs
Heart Circ Physiol 305: H1213–H1221, 2013. block HERG, a human cardiac delayed rectifier K⫹ channel. Open-channel block by
methanesulfonanilides. Circ Res 78: 499 –503, 1996.
381. Sheets MF, Hanck DA. Charge immobilization of the voltage sensor in domain IV is
independent of sodium current inactivation. J Physiol 563: 83–93, 2005. 401. Spector PS, Curran ME, Zou A, Keating MT, Sanguinetti MC. Fast inactivation causes
rectification of the IKr channel. J Gen Physiol 107: 611– 619, 1996.
382. Sheets MF, Hanck DA. Molecular action of lidocaine on the voltage sensors of sodium
channels. J Gen Physiol 121: 163–175, 2003. 402. Splawski I, Shen J, Timothy KW, Lehmann MH, Priori S, Robinson JL, Moss AJ,
Schwartz PJ, Towbin JA, Vincent GM, Keating MT. Spectrum of mutations in long-QT
383. Sheets MF, Hanck DA. The outermost lysine in the S4 of domain III contributes little syndrome genes. KVLQT1, HERG, SCN5A, KCNE1, and KCNE2. Circulation 102: 1178 –
to the gating charge in sodium channels. Biophys J 82: 3048 –3055, 2002. 1185, 2000.

384. Sheets MF, Kyle JW, Kallen RG, Hanck DA. The Na channel voltage sensor associated 403. Splawski I, Timothy KW, Decher N, Kumar P, Sachse FB, Beggs AH, Sanguinetti MC,
with inactivation is localized to the external charged residues of domain IV, S4. Biophys Keating MT. Severe arrhythmia disorder caused by cardiac L-type calcium channel
J 77: 747–757, 1999. mutations. Proc Natl Acad Sci USA 102: 8089 – 8096, 2005.

385. Shi W, Wymore RS, Wang HS, Pan Z, Cohen IS, McKinnon D, Dixon JE. Identification 404. Splawski I, Timothy KW, Sharpe LM, Decher N, Kumar P, Bloise R, Napolitano C,
of two nervous system-specific members of the erg potassium channel gene family. J Schwartz PJ, Joseph RM, Condouris K, Tager-Flusberg H, Priori SG, Sanguinetti MC,
Neurosci 17: 9423–9432, 1997. Keating MT. Ca(V)1.2 calcium channel dysfunction causes a multisystem disorder
including arrhythmia and autism. Cell 119: 19 –31, 2004.
386. Shimizu W, Antzelevitch C. Cellular basis for the ECG features of the LQT1 form of
the long-QT syndrome: effects of beta-adrenergic agonists and antagonists and so- 405. Splawski I, Tristani-Firouzi M, Lehmann MH, Sanguinetti MC, Keating MT. Mutations
dium channel blockers on transmural dispersion of repolarization and torsade de in the hminK gene cause long QT syndrome and suppress IKs function. Nat Genet 17:
pointes. Circulation 98: 2314 –2322, 1998. 338 –340, 1997.

387. Shimizu W, Antzelevitch C. Differential effects of beta-adrenergic agonists and antag- 406. Stengl M, Ramakers C, Donker DW, Nabar A, Rybin AV, Spatjens RL, van der Nagel
onists in LQT1, LQT2 and LQT3 models of the long QT syndrome. J Am Coll Cardiol T, Wodzig WK, Sipido KR, Antoons G, Moorman AF, Vos MA, Volders PG. Temporal
35: 778 –786, 2000. patterns of electrical remodeling in canine ventricular hypertrophy: focus on IKs
downregulation and blunted beta-adrenergic activation. Cardiovasc Res 72: 90 –100,
388. Shimizu W, Antzelevitch C. Effects of a K⫹ channel opener to reduce transmural 2006.
dispersion of repolarization and prevent torsade de pointes in LQT1, LQT2, and
407. Su Z, Limberis J, Souers A, Kym P, Mikhail A, Houseman K, Diaz G, Liu X, Martin RL,
LQT3 models of the long-QT syndrome. Circulation 102: 706 –712, 2000.
Cox BF, Gintant GA. Electrophysiologic characterization of a novel hERG channel
389. Shimizu W, Moss AJ, Wilde AA, Towbin JA, Ackerman MJ, January CT, Tester DJ, activator. Biochem Pharmacol 77: 1383–1390, 2009.
Zareba W, Robinson JL, Qi M, Vincent GM, Kaufman ES, Hofman N, Noda T, Kama-
408. Subbiah RN, Clarke CE, Smith DJ, Zhao J, Campbell TJ, Vandenberg JI. Molecular basis
kura S, Miyamoto Y, Shah S, Amin V, Goldenberg I, Andrews ML, McNitt S. Genotype-
of slow activation of the human ether-a-go-go related gene potassium channel. J
phenotype aspects of type 2 long QT syndrome. J Am Coll Cardiol 54: 2052–2062,
Physiol 558: 417– 431, 2004.
2009.
409. Sun Y, Zhang P, Li X, Zhang H, Li J, Liu G, Guo J. A novel nonsense mutation Y652X
390. Shushi L, Kerem B, Goldmit M, Peretz A, Attali B, Medina A, Towbin JA, Kurokawa J,
in the S6/pore region of human ether-go-go gene found in a long QT syndrome family.
Kass RS, Benhorin J. Clinical, genetic, and electrophysiologic characteristics of a new Scand Cardiovasc J 43: 181–186, 2009.
PAS-domain HERG mutation (M124R) causing Long QT syndrome. Ann Noninvasive
Electrocardiol 10: 334 –341, 2005. 410. Takano M, Kuratomi S. Regulation of cardiac inwardly rectifying potassium channels
by membrane lipid metabolism. Prog Biophys Mol Biol 81: 67–79, 2003.
391. Siebrands CC, Binder S, Eckhoff U, Schmitt N, Friederich P. Long QT 1 mutation
KCNQ1A344V increases local anesthetic sensitivity of the slowly activating delayed 411. Takemasa H, Nagatomo T, Abe H, Kawakami K, Igarashi T, Tsurugi T, Kabashima N,
rectifier potassium current. Anesthesiology 105: 511–520, 2006. Tamura M, Okazaki M, Delisle BP, January CT, Otsuji Y. Coexistence of hERG current

Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org 131


BOHNEN ET AL.

block and disruption of protein trafficking in ketoconazole-induced long QT syn- 431. Ueda K, Valdivia C, Medeiros-Domingo A, Tester DJ, Vatta M, Farrugia G, Ackerman
drome. Br J Pharmacol 153: 439 – 447, 2008. MJ, Makielski JC. Syntrophin mutation associated with long QT syndrome through
activation of the nNOS-SCN5A macromolecular complex. Proc Natl Acad Sci USA 105:
412. Takumi T, Ohkubo H, Nakanishi S. Cloning of a membrane protein that induces a 9355–9360, 2008.
slow voltage-gated potassium current. Science 242: 1042–1045, 1988.
432. Undrovinas AI, Maltsev VA, Kyle JW, Silverman N, Sabbah HN. Gating of the late Na⫹
413. Tan BH, Pundi KN, Van Norstrand DW, Valdivia CR, Tester DJ, Medeiros-Domingo channel in normal and failing human myocardium. J Mol Cell Cardiol 34: 1477–1489,
A, Makielski JC, Ackerman MJ. Sudden infant death syndrome-associated mutations in 2002.
the sodium channel beta subunits. Heart Rhythm 7: 771–778, 2010.
433. Vacher B, Pignier C, Letienne R, Verscheure Y, Le Grand B. F 15845 inhibits persistent
414. Tan VH, Yap J, Hsu LF, Liew R. Catheter ablation of ventricular fibrillation triggers and sodium current in the heart and prevents angina in animal models. Br J Pharmacol 156:
electrical storm. Europace 14: 1687–1695, 2012. 214 –225, 2009.

415. Tang L, Kallen RG, Horn R. Role of an S4 –S5 linker in sodium channel inactivation 434. Valenzuela C, Bennett PB Jr. Gating of cardiac Na⫹ channels in excised membrane
probed by mutagenesis and a peptide blocker. J Gen Physiol 108: 89 –104, 1996. patches after modification by alpha-chymotrypsin. Biophys J 67: 161–171, 1994.
416. Tateyama M, Rivolta I, Clancy CE, Kass RS. Modulation of cardiac sodium channel 435. Van Petegem F, Lobo PA, Ahern CA. Seeing the forest through the trees: towards a
gating by protein kinase A can be altered by disease-linked mutation. J Biol Chem 278: unified view on physiological calcium regulation of voltage-gated sodium channels.
46718 – 46726, 2003. Biophys J 103: 2243–2251, 2012.
417. Tawil R, Ptacek LJ, Pavlakis SG, DeVivo DC, Penn AS, Ozdemir C, Griggs RC. Ander- 436. Vandenberg JI, Perry MD, Perrin MJ, Mann SA, Ke Y, Hill AP. hERG K(⫹) channels:

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


sen’s syndrome: potassium-sensitive periodic paralysis, ventricular ectopy, and dys- structure, function, and clinical significance. Physiol Rev 92: 1393–1478, 2012.
morphic features. Ann Neurol 35: 326 –330, 1994.
437. Varro A, Balati B, Iost N, Takacs J, Virag L, Lathrop DA, Csaba L, Talosi L, Papp JG.
418. Telezhkin V, Reilly JM, Thomas AM, Tinker A, Brown DA. Structural requirements of The role of the delayed rectifier component IKs in dog ventricular muscle and Purkinje
membrane phospholipids for M-type potassium channel activation and binding. J Biol fibre repolarization. J Physiol 523: 67– 81, 2000.
Chem 287: 10001–10012, 2012.
438. Vatta M, Ackerman MJ, Ye B, Makielski JC, Ughanze EE, Taylor EW, Tester DJ,
419. Terrenoire C, Houslay MD, Baillie GS, Kass RS. The cardiac IKs potassium channel Balijepalli RC, Foell JD, Li Z, Kamp TJ, Towbin JA. Mutant caveolin-3 induces persis-
macromolecular complex includes the phosphodiesterase PDE4D3. J Biol Chem 284: tent late sodium current and is associated with long-QT syndrome. Circulation 114:
9140 –9146, 2009. 2104 –2112, 2006.
420. Terrenoire C, Wang K, Tung KW, Chung WK, Pass RH, Lu JT, Jean JC, Omari A, 439. Veerman CC, Verkerk AO, Blom MT, Klemens CA, Langendijk PN, van Ginneken
Sampson KJ, Kotton DN, Keller G, Kass RS. Induced pluripotent stem cells used to AC, Wilders R, Tan HL. Slow delayed rectifier potassium current blockade contrib-
reveal drug actions in a long QT syndrome family with complex genetics. J Gen Physiol utes importantly to drug-induced long QT syndrome. Circ Arrhythm Electrophysiol 6:
141: 61–72, 2013. 1002–1009, 2013.
421. Tester DJ, Ackerman MJ. Genetic testing for potentially lethal, highly treatable inher- 440. Vincent GM, Schwartz PJ, Denjoy I, Swan H, Bithell C, Spazzolini C, Crotti L, Piippo
ited cardiomyopathies/channelopathies in clinical practice. Circulation 123: 1021– K, Lupoglazoff JM, Villain E, Priori SG, Napolitano C, Zhang L. High efficacy of beta-
1037, 2011. blockers in long-QT syndrome type 1: contribution of noncompliance and QT-pro-
longing drugs to the occurrence of beta-blocker treatment “failures”. Circulation 119:
422. Tinel N, Diochot S, Borsotto M, Lazdunski M, Barhanin J. KCNE2 confers background
215–221, 2009.
current characteristics to the cardiac KCNQ1 potassium channel. EMBO J 19: 6326 –
6330, 2000. 441. Viswanathan PC, Rudy Y. Pause induced early afterdepolarizations in the long QT
syndrome: a simulation study. Cardiovasc Res 42: 530 –542, 1999.
423. Tranebjaerg L, Samson RA, Green GE. Jervell and Lange-Nielsen Syndrome. In: Gene
Reviews, edited by Pagon RA, Adam MP, Ardinger HH, Wallace SE, Amemiya A, Bean 442. Volders PG, Sipido KR, Carmeliet E, Spatjens RL, Wellens HJ, Vos MA. Repolarizing
LJH, Bird TD, Fong CT, Mefford HC, Smith RJH, Stephens K. Seattle, WA:Univ. of K⫹ currents ITO1 and IKs are larger in right than left canine ventricular midmyocar-
Washington, 1993. dium. Circulation 99: 206 –210, 1999.
424. Tristani-Firouzi M, Etheridge SP. Kir 2.1 channelopathies: the Andersen-Tawil syn- 443. Walker VE, Wong MJ, Atanasiu R, Hantouche C, Young JC, Shrier A. Hsp40 chaper-
drome. Pflügers Arch 460: 289 –294, 2010. ones promote degradation of the HERG potassium channel. J Biol Chem 285: 3319 –
3329, 2010.
425. Tristani-Firouzi M, Jensen JL, Donaldson MR, Sansone V, Meola G, Hahn A, Bendah-
hou S, Kwiecinski H, Fidzianska A, Plaster N, Fu YH, Ptacek LJ, Tawil R. Functional and 444. Walsh KB, Kass RS. Regulation of a heart potassium channel by protein kinase A and C.
clinical characterization of KCNJ2 mutations associated with LQT7 (Andersen syn- Science 242: 67– 69, 1988.
drome). J Clin Invest 110: 381–388, 2002.
445. Wang D, Papp AC, Binkley PF, Johnson JA, Sadee W. Highly variable mRNA expres-
426. Trudeau MC, Leung LM, Roti ER, Robertson GA. hERG1a N-terminal eag domain- sion and splicing of L-type voltage-dependent calcium channel alpha subunit 1C in
containing polypeptides regulate homomeric hERG1b and heteromeric hERG1a/ human heart tissues. Pharmacogenet Genomics 16: 735–745, 2006.
hERG1b channels: a possible mechanism for long QT syndrome. J Gen Physiol 138:
581–592, 2011. 446. Wang DT, Hill AP, Mann SA, Tan PS, Vandenberg JI. Mapping the sequence of con-
formational changes underlying selectivity filter gating in the K(v)11.1 potassium chan-
427. Trudeau MC, Warmke JW, Ganetzky B, Robertson GA. HERG, a human inward nel. Nat Struct Mol Biol 18: 35– 41, 2011.
rectifier in the voltage-gated potassium channel family. Science 269: 92–95, 1995.
447. Wang DW, Crotti L, Shimizu W, Pedrazzini M, Cantu F, De Filippo P, Kishiki K,
428. Tsuji K, Akao M, Ishii TM, Ohno S, Makiyama T, Takenaka K, Doi T, Haruna Y, Miyazaki A, Ikeda T, Schwartz PJ, George AL Jr. Malignant perinatal variant of long-QT
Yoshida H, Nakashima T, Kita T, Horie M. Mechanistic basis for the pathogenesis of syndrome caused by a profoundly dysfunctional cardiac sodium channel. Circ Arrhythm
long QT syndrome associated with a common splicing mutation in KCNQ1 gene. J Electrophysiol 1: 370 –378, 2008.
Mol Cell Cardiol 42: 662– 669, 2007.
448. Wang DW, Yazawa K, George AL Jr, Bennett PB. Characterization of human cardiac
429. Tuvia S, Buhusi M, Davis L, Reedy M, Bennett V. Ankyrin-B is required for intracellular Na⫹ channel mutations in the congenital long QT syndrome. Proc Natl Acad Sci USA
sorting of structurally diverse Ca2⫹ homeostasis proteins. J Cell Biol 147: 995–1008, 93: 13200 –13205, 1996.
1999.
449. Wang HW, Zheng YQ, Yang ZF, Li CZ, Liu YM. Effect of mexiletine on long QT
430. Tyson J, Tranebjaerg L, Bellman S, Wren C, Taylor JF, Bathen J, Aslaksen B, Sorland SJ, syndrome model. Acta Pharmacol Sin 24: 316 –320, 2003.
Lund O, Malcolm S, Pembrey M, Bhattacharya S, Bitner-Glindzicz M. IsK and KvLQT1:
mutation in either of the two subunits of the slow component of the delayed rectifier 450. Wang J, Trudeau MC, Zappia AM, Robertson GA. Regulation of deactivation by an
potassium channel can cause Jervell and Lange-Nielsen syndrome. Hum Mol Genet 6: amino terminal domain in human ether-a-go-go-related gene potassium channels. J
2179 –2185, 1997. Gen Physiol 112: 637– 647, 1998.

132 Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org


CONGENITAL LONG QT SYNDROME

451. Wang Q, Curran ME, Splawski I, Burn TC, Millholland JM, VanRaay TJ, Shen J, Timothy 472. Wu D, Delaloye K, Zaydman MA, Nekouzadeh A, Rudy Y, Cui J. State-dependent
KW, Vincent GM, de Jager T, Schwartz PJ, Toubin JA, Moss AJ, Atkinson DL, Landes electrostatic interactions of S4 arginines with E1 in S2 during Kv7.1 activation. J Gen
GM, Connors TD, Keating MT. Positional cloning of a novel potassium channel gene: Physiol 135: 595– 606, 2010.
KVLQT1 mutations cause cardiac arrhythmias. Nat Genet 12: 17–23, 1996.
473. Wu G, Ai T, Kim JJ, Mohapatra B, Xi Y, Li Z, Abbasi S, Purevjav E, Samani K, Ackerman
452. Wang Q, Li Z, Shen J, Keating MT. Genomic organization of the human SCN5A gene MJ, Qi M, Moss AJ, Shimizu W, Towbin JA, Cheng J, Vatta M. Alpha-1-syntrophin
encoding the cardiac sodium channel. Genomics 34: 9 –16, 1996. mutation and the long-QT syndrome: a disease of sodium channel disruption. Circ
Arrhythm Electrophysiol 1: 193–201, 2008.
453. Wang Q, Shen J, Splawski I, Atkinson D, Li Z, Robinson JL, Moss AJ, Towbin JA,
Keating MT. SCN5A mutations associated with an inherited cardiac arrhythmia, long 474. Wu HC, Yamankurt G, Luo J, Subramaniam J, Hashmi SS, Hu H, Cunha SR. Identifi-
QT syndrome. Cell 80: 805– 811, 1995. cation and characterization of two ankyrin-B isoforms in mammalian heart. Cardiovasc
Res 107: 466 – 477, 2015.
454. Wang S, Liu S, Morales MJ, Strauss HC, Rasmusson RL. A quantitative analysis of the
activation and inactivation kinetics of HERG expressed in Xenopus oocytes. J Physiol 475. Wu L, Bauer CS, Zhen XG, Xie C, Yang J. Dual regulation of voltage-gated calcium
502: 45– 60, 1997. channels by PtdIns(4,5)P2. Nature 419: 947–952, 2002.

455. Wang S, Morales MJ, Liu S, Strauss HC, Rasmusson RL. Time, voltage and ionic 476. Wu W, Sachse FB, Gardner A, Sanguinetti MC. Stoichiometry of altered hERG1
concentration dependence of rectification of h-erg expressed in Xenopus oocytes. channel gating by small molecule activators. J Gen Physiol 143: 499 –512, 2014.
FEBS Lett 389: 167–173, 1996.
477. Wu ZY, Chen K, Haendler B, McDonald TV, Bian JS. Stimulation of N-terminal trun-
cated isoform of androgen receptor stabilizes human ether-a-go-go-related gene-

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


456. Wang W, Xia J, Kass RS. MinK-KvLQT1 fusion proteins, evidence for multiple stoichi-
ometries of the assembled IsK channel. J Biol Chem 273: 34069 –34074, 1998. encoded potassium channel protein via activation of extracellular signal regulated
kinase 1/2. Endocrinology 149: 5061–5069, 2008.
457. Wang Z, Fermini B, Nattel S. Rapid and slow components of delayed rectifier current
in human atrial myocytes. Cardiovasc Res 28: 1540 –1546, 1994. 478. Xu H, Guo W, Nerbonne JM. Four kinetically distinct depolarization-activated K⫹
currents in adult mouse ventricular myocytes. J Gen Physiol 113: 661– 678, 1999.
458. Wang Z, Yue L, White M, Pelletier G, Nattel S. Differential distribution of inward
rectifier potassium channel transcripts in human atrium versus ventricle. Circulation 479. Xu X, Jiang M, Hsu KL, Zhang M, Tseng GN. KCNQ1 and KCNE1 in the IKs channel
98: 2422–2428, 1998. complex make state-dependent contacts in their extracellular domains. J Gen Physiol
131: 589 – 603, 2008.
459. Ward OC. A new familial cardiac syndrome in children. J Ir Med Assoc 54: 103–106,
1964. 480. Xu X, Kanda VA, Choi E, Panaghie G, Roepke TK, Gaeta SA, Christini DJ, Lerner DJ,
Abbott GW. MinK-dependent internalization of the IKs potassium channel. Cardiovasc
460. Warmke JW, Ganetzky B. A family of potassium channel genes related to eag in Res 82: 430 – 438, 2009.
Drosophila and mammals. Proc Natl Acad Sci USA 91: 3438 –3442, 1994.
481. Yang PC, Song Y, Giles WR, Horvath B, Chen-Izu Y, Belardinelli L, Rajamani S, Clancy
461. Wedekind H, Smits JP, Schulze-Bahr E, Arnold R, Veldkamp MW, Bajanowski T, CE. A computational modelling approach combined with cellular electrophysiology
Borggrefe M, Brinkmann B, Warnecke I, Funke H, Bhuiyan ZA, Wilde AA, Breithardt data provides insights into the therapeutic benefit of targeting the late Na⫹ current. J
G, Haverkamp W. De novo mutation in the SCN5A gene associated with early onset Physiol 593: 1429 –1442, 2015.
of sudden infant death. Circulation 104: 1158 –1164, 2001.
482. Yang T, Chun YW, Stroud DM, Mosley JD, Knollmann BC, Hong C, Roden DM.
462. Weerapura M, Nattel S, Chartier D, Caballero R, Hebert TE. A comparison of cur- Screening for acute IKr block is insufficient to detect torsades de pointes liability: role
rents carried by HERG, with and without coexpression of MiRP1, and the native rapid of late sodium current. Circulation 130: 224 –234, 2014.
delayed rectifier current. Is MiRP1 the missing link? J Physiol 540: 15–27, 2002.
483. Yang T, Snyders DJ, Roden DM. Ibutilide, a methanesulfonanilide antiarrhythmic, is a
463. Wehrens XH, Rossenbacker T, Jongbloed RJ, Gewillig M, Heidbuchel H, Doevendans potent blocker of the rapidly activating delayed rectifier K⫹ current (IKr) in AT-1
PA, Vos MA, Wellens HJ, Kass RS. A novel mutation L619F in the cardiac Na⫹ channel cells. Concentration-, time-, voltage-, and use-dependent effects. Circulation 91:
SCN5A associated with long-QT syndrome (LQT3): a role for the I–II linker in inac- 1799 –1806, 1995.
tivation gating. Hum Mutat 21: 552, 2003.
484. Yang Y, Yang Y, Liang B, Liu J, Li J, Grunnet M, Olesen SP, Rasmussen HB, Ellinor PT,
464. Weiss JN, Garfinkel A, Karagueuzian HS, Chen PS, Qu Z. Early afterdepolarizations Gao L, Lin X, Li L, Wang L, Xiao J, Liu Y, Liu Y, Zhang S, Liang D, Peng L, Jespersen T,
and cardiac arrhythmias. Heart Rhythm 7: 1891–1899, 2010. Chen YH. Identification of a Kir3.4 mutation in congenital long QT syndrome. Am J
Hum Genet 86: 872– 880, 2010.
465. West JW, Patton DE, Scheuer T, Wang Y, Goldin AL, Catterall WA. A cluster of
hydrophobic amino acid residues required for fast Na⫹-channel inactivation. Proc Natl 485. Yang ZK, Boyett MR, Janvier NC, McMorn SO, Shui Z, Karim F. Regional differences
Acad Sci USA 89: 10910 –10914, 1992. in the negative inotropic effect of acetylcholine within the canine ventricle. J Physiol
492: 789 – 806, 1996.
466. Wickman K, Clapham DE. Ion channel regulation by G proteins. Physiol Rev 75: 865–
885, 1995. 486. Yarbrough TL, Lu T, Lee HC, Shibata EF. Localization of cardiac sodium channels in
caveolin-rich membrane domains: regulation of sodium current amplitude. Circ Res
467. Wickman K, Nemec J, Gendler SJ, Clapham DE. Abnormal heart rate regulation in 90: 443– 449, 2002.
GIRK4 knockout mice. Neuron 20: 103–114, 1998.
487. Yarotskyy V, Gao G, Du L, Ganapathi SB, Peterson BZ, Elmslie KS. Roscovitine binds
468. Wickman KD, Iniguez-Lluhl JA, Davenport PA, Taussig R, Krapivinsky GB, Linder ME, to novel L-channel (CaV1.2) sites that separately affect activation and inactivation. J
Gilman AG, Clapham DE. Recombinant G-protein beta gamma-subunits activate the Biol Chem 285: 43–53, 2010.
muscarinic-gated atrial potassium channel. Nature 368: 255–257, 1994.
488. Yarotskyy V, Gao G, Peterson BZ, Elmslie KS. The Timothy syndrome mutation of
469. Wiener R, Haitin Y, Shamgar L, Fernandez-Alonso MC, Martos A, Chomsky-Hecht O, cardiac CaV1.2 (L-type) channels: multiple altered gating mechanisms and pharma-
Rivas G, Attali B, Hirsch JA. The KCNQ1 (Kv7.1) COOH terminus, a multitiered cological restoration of inactivation. J Physiol 587: 551–565, 2009.
scaffold for subunit assembly and protein interaction. J Biol Chem 283: 5815–5830,
2008. 489. Yazawa M, Hsueh B, Jia X, Pasca AM, Bernstein JA, Hallmayer J, Dolmetsch RE. Using
induced pluripotent stem cells to investigate cardiac phenotypes in Timothy syn-
470. Wong JA, Gula LJ, Klein GJ, Yee R, Skanes AC, Krahn AD. Utility of treadmill testing in drome. Nature 471: 230 –234, 2011.
identification and genotype prediction in long-QT syndrome. Circ Arrhythm Electro-
physiol 3: 120 –125, 2010. 490. Yu FH, Westenbroek RE, Silos-Santiago I, McCormick KA, Lawson D, Ge P, Ferriera
H, Lilly J, DiStefano PS, Catterall WA, Scheuer T, Curtis R. Sodium channel beta4, a
471. Wright M, Sacher F, Haissaguerre M. Catheter ablation for patients with ventricular new disulfide-linked auxiliary subunit with similarity to beta2. J Neurosci 23: 7577–
fibrillation. Curr Opin Cardiol 24: 56 – 60, 2009. 7585, 2003.

Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org 133


BOHNEN ET AL.

491. Yu H, Lin Z, Mattmann ME, Zou B, Terrenoire C, Zhang H, Wu M, McManus OB, Kass 503. Zhang L, Timothy KW, Vincent GM, Lehmann MH, Fox J, Giuli LC, Shen J, Splawski I,
RS, Lindsley CW, Hopkins CR, Li M. Dynamic subunit stoichiometry confers a pro- Priori SG, Compton SJ, Yanowitz F, Benhorin J, Moss AJ, Schwartz PJ, Robinson JL,
gressive continuum of pharmacological sensitivity by KCNQ potassium channels. Proc Wang Q, Zareba W, Keating MT, Towbin JA, Napolitano C, Medina A. Spectrum of
Natl Acad Sci USA 110: 8732– 8737, 2013. ST-T-wave patterns and repolarization parameters in congenital long-QT syndrome:
ECG findings identify genotypes. Circulation 102: 2849 –2855, 2000.
492. Yu H, Lin Z, Xu K, Huang X, Long S, Wu M, McManus OB, Le Engers J, Mattmann ME,
Engers DW, Le UM, Lindsley CW, Hopkins CR, Li M. Identification of a novel, small 504. Zhang L, Vincent GM, Baralle M, Baralle FE, Anson BD, Benson DW, Whiting B,
molecule activator of KCNQ1 channels. In: Probe Reports from the NIH Molecular Timothy KW, Carlquist J, January CT, Keating MT, Splawski I. An intronic mutation
Libraries Program. Bethesda, MD: NIH, 2010. causes long QT syndrome. J Am Coll Cardiol 44: 1283–1291, 2004.

493. Yue L, Feng J, Li GR, Nattel S. Transient outward and delayed rectifier currents in 505. Zhang M, Liu J, Tseng GN. Gating charges in the activation and inactivation processes
canine atrium: properties and role of isolation methods. Am J Physiol Heart Circ Physiol of the HERG channel. J Gen Physiol 124: 703–718, 2004.
270: H2157–H2168, 1996.
506. Zhang X, Szeto C, Gao E, Tang M, Jin J, Fu Q, Makarewich C, Ai X, Li Y, Tang A, Wang
494. Yue Y, Castrichini M, Srivastava U, Fabris F, Shah K, Li Z, Qu Y, El-Sherif N, Zhou Z, J, Gao H, Wang F, Ge XJ, Kunapuli SP, Zhou L, Zeng C, Xiang KY, Chen X. Cardio-
January C, Hussain MM, Jiang XC, Sobie EA, Wahren-Herlenius M, Chahine M, toxic and cardioprotective features of chronic beta-adrenergic signaling. Circ Res 112:
Capecchi PL, Laghi-Pasini F, Lazzerini PE, Boutjdir M. Pathogenesis of the Novel 498 –509, 2013.
Autoimmune-Associated Long-QT Syndrome. Circulation 132: 230 –240, 2015.
507. Zhang Y, Ouyang P, Post WS, Dalal D, Vaidya D, Blasco-Colmenares E, Soliman EZ,
495. Zareba W, Moss AJ, Locati EH, Lehmann MH, Peterson DR, Hall WJ, Schwartz PJ, Tomaselli GF, Guallar E. Sex-steroid hormones and electrocardiographic QT-interval
Vincent GM, Priori SG, Benhorin J, Towbin JA, Robinson JL, Andrews ML, Napolitano duration: findings from the third National Health and Nutrition Examination Survey

Downloaded from http://physrev.physiology.org/ by 10.220.33.3 on November 5, 2016


C, Timothy K, Zhang L, Medina A. Modulating effects of age and gender on the clinical and the Multi-Ethnic Study of Atherosclerosis. Am J Epidemiol 174: 403– 411, 2011.
course of long QT syndrome by genotype. J Am Coll Cardiol 42: 103–109, 2003.
508. Zhang Z, Zhao Z, Liu Y, Wang W, Wu Y, Ding J. Kinetic model of Nav1.5 channel
496. Zaritsky JJ, Redell JB, Tempel BL, Schwarz TL. The consequences of disrupting cardiac provides a subtle insight into slow inactivation associated excitability in cardiac cells.
inwardly rectifying K⫹ current [I(K1)] as revealed by the targeted deletion of the PLoS One 8: e64286, 2013.
murine Kir2.1 and Kir22 genes. J Physiol 533: 697–710, 2001.
509. Zhao JT, Hill AP, Varghese A, Cooper AA, Swan H, Laitinen-Forsblom PJ, Rees MI,
497. Zaydman MA, Cui J. PIP2 regulation of KCNQ channels: biophysical and molecular Skinner JR, Campbell TJ, Vandenberg JI. Not all hERG pore domain mutations have a
mechanisms for lipid modulation of voltage-dependent gating. Front Physiol 5: 195, severe phenotype: G584S has an inactivation gating defect with mild phenotype com-
2014. pared with G572S, which has a dominant negative trafficking defect and a severe
phenotype. J Cardiovasc Electrophysiol 20: 923–930, 2009.
498. Zaydman MA, Silva JR, Delaloye K, Li Y, Liang H, Larsson HP, Shi J, Cui J. Kv7.1 ion
channels require a lipid to couple voltage sensing to pore opening. Proc Natl Acad Sci 510. Zheng R, Thompson K, Obeng-Gyimah E, Alessi D, Chen J, Cheng H, McDonald TV.
USA 110: 13180 –13185, 2013. Analysis of the interactions between the C-terminal cytoplasmic domains of KCNQ1
and KCNE1 channel subunits. Biochem J 428: 75– 84, 2010.
499. Zeng H, Lozinskaya IM, Lin Z, Willette RN, Brooks DP, Xu X. Mallotoxin is a novel
human ether-a-go-go-related gene (hERG) potassium channel activator. J Pharmacol 511. Zhou J, Augelli-Szafran CE, Bradley JA, Chen X, Koci BJ, Volberg WA, Sun Z, Cordes
Exp Ther 319: 957–962, 2006. JS. Novel potent human ether-a-go-go-related gene (hERG) potassium channel en-
hancers and their in vitro antiarrhythmic activity. Mol Pharmacol 68: 876 – 884, 2005.
500. Zeng J, Rudy Y. Early afterdepolarizations in cardiac myocytes: mechanism and rate
dependence. Biophys J 68: 949 –964, 1995. 512. Zhou Z, Gong Q, Epstein ML, January CT. HERG channel dysfunction in human long
QT syndrome. Intracellular transport and functional defects. J Biol Chem 273: 21061–
501. Zhang H, He C, Yan X, Mirshahi T, Logothetis DE. Activation of inwardly rectifying 21066, 1998.
K⫹ channels by distinct PtdIns(4,5)P2 interactions. Nat Cell Biol 1: 183–188, 1999.
513. Zou A, Curran ME, Keating MT, Sanguinetti MC. Single HERG delayed rectifier K⫹
502. Zhang L, Benson DW, Tristani-Firouzi M, Ptacek LJ, Tawil R, Schwartz PJ, George AL, channels expressed in Xenopus oocytes. Am J Physiol Heart Circ Physiol 272: H1309 –
Horie M, Andelfinger G, Snow GL, Fu YH, Ackerman MJ, Vincent GM. Electrocardio- H1314, 1997.
graphic features in Andersen-Tawil syndrome patients with KCNJ2 mutations: char-
acteristic T-U-wave patterns predict the KCNJ2 genotype. Circulation 111: 2720 – 514. Zuhlke RD, Pitt GS, Deisseroth K, Tsien RW, Reuter H. Calmodulin supports both
2726, 2005. inactivation and facilitation of L-type calcium channels. Nature 399: 159 –162, 1999.

134 Physiol Rev • VOL 97 • JANUARY 2017 • www.prv.org

You might also like