You are on page 1of 11

Potassium Channel

Remodeling in Heart
Disease
Vincent Algalarrondo, MD, PhDa,b,c, Stanley Nattel, MDa,b,c,*

KEYWORDS
 Cardiac repolarization  Heart disease  Potassium  Potassium channel  Arrhythmia
 Atrial Fibrillation

KEY POINTS
 Cardiac repolarization and underlying ionic mechanisms are substantially altered by heart disease.
 In hypertrophy, myocardial infarction, and heart failure, potassium channels and currents are down-
regulated and repolarization is delayed, creating a substrate for ventricular tachyarrhythmia.
 Mechanisms leading to arrhythmia with potassium channel downregulation include early afterdepo-
larizations, reduced cardiomyocyte resting membrane resistance, and increased repolarization
heterogeneity.
 A variety of extracardiac conditions (eg, hypokalemia, class III antiarrhythmic drugs) further reduce
repolarization reserve and increase the arrhythmic risk in patients with heart disease.
 In atrial fibrillation, the combined effects of reduced calcium current and increased potassium cur-
rent reduce action potential duration and refractory period and promote reentry, while hyperpola-
rizing the resting membrane potential, stabilizing reentrant rotors.

INTRODUCTION shorten the cardiomyocyte refractory period to


allow cardiomyocytes to sustain very high activa-
Potassium channels are substantially altered in tion rates. These pathophysiologic processes are
cardiovascular diseases (for an overview, see complex and include redundancy and negative
Table1). Teleologically, the types of potassium feedback. Although apparently designed to main-
channel remodeling could be classified into 2 gen- tain cardiac homeostasis, they also induce elec-
eral secondary categories. The first type of remod- trophysiologic alterations that lead to potentially
eling, occurring in the ventricles with heart failure serious arrhythmias. Therefore, understanding
(HF), aims to prolong the action potential (AP) to in- them is of value for cardiologists in daily practice.
crease calcium entry into the cytoplasm and In addition, K1 channel remodeling can be
compensate cardiomyocyte contraction. The sec- induced by a host of other cardiac and extracar-
ond type of remodeling process, typically occur- diac conditions.
ring in atria during atrial fibrillation (AF), aims to

Disclosures: V. Algalarrondo received scholarship funding from St. Jude Medical, Biotronik, Boston, Medtronic
and Sorin.
Funding Sources: Canadian Institutes of Health Research (6957 and 44365, S. Nattel) and Heart and Stroke
cardiacEP.theclinics.com

Foundation of Canada (S. Nattel). Fédération Française de Cardiologie (V. Algalarrondo).


a
Department of Medicine, Research Center, Montreal Heart Institute, University of Montreal, 5000 Belanger
Street East, Montreal, Quebec H1T 1C8, Canada; b Department of Pharmacology and Therapeutics, McGill Uni-
versity, 3655 Promenade Sir-William-Osler, Montréal, Québec H3G 1Y6, Canada; c Faculty of Medicine, Univer-
sity Duisburg-Essen, Hufelandstr. 55, Essen 45122, Germany
* Corresponding author. Montreal Heart Institute, University of Montreal, 5000 Belanger Street East, Mon-
treal, Quebec H1T 1C8, Canada.
E-mail address: stanley.nattel@icm-mhi.org

Card Electrophysiol Clin - (2016) -–-


http://dx.doi.org/10.1016/j.ccep.2016.01.006
1877-9182/16/$ – see front matter Ó 2016 Elsevier Inc. All rights reserved.
2 Algalarrondo & Nattel

Table 1
Main disease-related modifications in KD currents in pathologic situations

Ventricular Atrial
Hypertrophy Heart Failure Myocardial Infarction Fibrillation
Current Density Exercise Overload Atria Ventricle Border Zone Normal Zone Atria PVa
APD 4 [ — [ [ [ Y Y
Ito 4 Y Y Y Y/4b Y Y Y
IKurc — — — — — — Y(?)d —
IK1 [ Y 4 Y Y Y [ Y
IKr 4 Y 4 4 Y Y 4 [
IKs 4 — Y Y Ye Y 4 [
IKss [ Y — 4 — — — —
Iff — — Y — — — — —
IK(Ach,c) c
— — — — — — [ —
IK(ATP) — — — — — [ ?d —
ISK — — — [ — [ 4 d
[
IK2P c
— — — — — — [ —

When discrepancies were noted across models, results documented on human samples are reported.
Abbreviations: [, current is increased; Y, current is decreased; 4, current is unchanged; APD, action potential duration;
Ito, transient outward K1 current; IK1, inward rectifier K1 current; IK2P, 2-pore-domain K1 current; IKur, ultrarapid rectifier
K current; IKr, delayed rectifier K1 current; IKs, slow rectifier K1 current; IKss, steady-state outward K1 current; IK(ACh),
1

acetylcholine-dependent K1 current; ISK, small calcium-activated K1 current; PV, pulmonary veins.


a
Compared with the left atrium.
b
Ito is reduced initially then return to normal within 2 months.
c
Atrial-specific currents.
d
Conflicting results.
e
ERG, KvLQT1 and minK are down regulated at day 2 after MI then ERG and KvLQT1 normalized at day 5 after MI,
whereas minK remains downregulated.
f
In sinoatrial node.
Data from Refs.7,12,30,73,74

Cardiac repolarization is in itself a complex pro- valvular ischemic or congenital heart diseases).
cess. Current state of the art recognizes more than In hypertensive patients, there is a weak but signif-
10 different types of cardiac K1 currents, and icant correlation between QTc interval and blood
more than 20 types of K1 channels are known to pressure and hypertrophic indices like the Cornell
be expressed in the heart.1,2 Herein, we aim to voltage indices.3 It is also known that hypertensive
present the K1 channel remodeling processes in patients with prolonged QTc intervals are at
4 common pathologic situations: cardiac hypertro- increased risk of cardiovascular morbidity and
phy, HF, myocardial infarction (MI), and AF. Spe- mortality.4 In patients with successful antihyper-
cific regional remodeling profiles such as the tensive therapy, left ventricular mass reduction is
border zone of the MI scar and the pulmonary correlated with a decrease in the QTc interval5;
veins (PVs) are also detailed. for a review, see Ref.6 In experimental models of
cardiac hypertrophy, prolongation of repolariza-
tion has been documented by longer QTc intervals
OVERVIEW OF THE REMODELING PROCESSES
and AP durations (APDs). In patch clamp analysis,
OF POTASSIUM CHANNELS IN PATHOLOGIC
total K1 current amplitudes are similar in cardiac
SITUATIONS
hypertrophy subjects and controls, but because
Remodeling of K1 Currents Associated with
the membrane capacitance is larger in hypertro-
Hypertrophy
phy, the resulting K1 current densities are reduced
Hypertrophy in cardiac hypertrophy.7 This phenomenon has
Cardiac hypertrophy is an adaptive response been documented for transient outward K1 cur-
to volume or pressure overload that occurs in rent (Ito), IK1 and slow rectifier K1 current (IKs):
various pathologic situations (hypertension, compared with control, proteins and transcripts
Potassium Channel Remodeling in Heart Disease 3

are unchanged in cardiac hypertrophy but current and IK1, although there are some discrepancies
densities are lower because the cells are larger (for among studies. These discrepancies could be
Ito: Kv4.2, Kv4.3 and KChIP2; for IK1: Kir2.1 and caused by differences in the HF models or to
Kir2.2).7 For IKs, downregulation is observed at time-course differences in electrophysiologic
the transcript and the protein levels, especially in remodeling after the onset of HF.13 Table 2 sum-
the epicardium. Finally, Iss amplitude (noninacti- marizes the changes in messenger RNA (mRNA)
vating current) is increased in cardiac hypertrophy and protein expression in various models.
but not enough to restore a normal Iss current den- Because the classical K1 currents are downregu-
sity.7 Together, these results suggest downregula- lated, other K1 currents could play an important
tion of repolarizing K1 current densities in cardiac role in the repolarization of remodeled myocytes.
hypertrophy owing to pressure overload. This For instance, inhibition of the small conductance
downregulation prolongs the APD; this could be Ca21-activated K1 current small calcium-
proarrhythmic and may explain, at least in part, activated K1 current (ISK) has been recently re-
the correlation between hypertrophy, QTc interval, ported to prolong APD by 9% in failing human
and cardiovascular events. hearts, suggesting a compensatory role.14 This
report supports previous results in HF rabbits, in
Exercise-induced hypertrophy which increased cytosolic Ca21 in HF seems to
In contrast with hypertrophy owing to chronic activate ISK, thus reducing APD and compensating
pressure overload, exercise-induced hypertrophy for the HF-induced reduction in repolarization
does not modify corrected QTc interval and QTc in- reserve (Fig. 2).14–16
terval dispersion in professional athletes.8 How- Reduced repolarizing currents induce proarrhyth-
ever, T wave morphology is commonly altered in mic consequences in several ways. First, increased
trained athletes, suggesting that repolarization is duration of repolarization may induce early afterde-
remodeled in cardiac hypertrophy owing to exer- polarizations (see Fig. 1A). Early afterdepolariza-
cise. In 2010, Yang and colleagues9 reported an tions are triggered if AP is prolonged in the
extensive study of cardiac repolarization in a specific voltage and time window that allows either
swim training mouse model. Although myocytes the late Ca21 current ICaL or the Na1 current INa to
had a 17% increase in membrane capacitance reactivate and generate a new AP.12 In HF patients,
with exercise training, most of the electrical pa- repolarization reserve is reduced and the addition of
rameters were similar between trained mice and class III antiarrhythmic drugs or hypokalemia may
controls: ECG intervals were similar and APDs trigger early afterdepolarizations and ventricular
were unchanged. Using patch clamp methods, tachyarrhythmias more easily. The downregulation
the authors documented that K1 and Ca21 cur- of inward rectifying IK1 current increases the mem-
rents increased in parallel with each other, result- brane resistance in the phase 4 of the AP, increasing
ing in unchanged AP shapes in exercise. The the risk that a delayed afterdepolarization will
increased current densities were due to increased generate an arrhythmia (see Fig. 1B).
transcription and translation of channel subunits.
The voltage-gated properties were unaltered. In Remodeling of K1 Currents Associated with
contrast with hypertrophy caused by pressure Myocardial Infarction
overload, the repolarization remodeling in exercise
parallels cardiac hypertrophy and returns to MI causes changes in K1 current expression, den-
normal if training is stopped. sity and function. In most cases, the proarrhythmic
substrate in ischemic cardiomyopathy results from
Remodeling of K1 Currents Associated with a heterogeneous scar where islands of living cells,
Congestive Heart Failure called the border zone, are partially surrounded by
fibrotic tissue.
Between one-third and one-half of HF-related
deaths are caused by ventricular tachyarrhyth- Remodeling in the myocardial infarction border
mias, and sudden death prevention is crucial in zone
HF. QTc prolongation occurs in HF10 and is asso- Post MI border zone remodeling is characterized
ciated with an increased sudden death rate in by prolonged repolarization owing to global down-
ischemic cardiomyopathy.11 In experimental HF regulation of K1 currents that promotes early after-
models, the APs are prolonged and early afterde- depolarizations (Fig. 3B; see Table 1). Remodeling
polarization have been reproducibly documented begins within the first days after MI and is
(Figs. 1A and 3A; for a review, see Ref.12). APD observed in both Purkinje cells and cardiomyo-
prolongation is induced by downregulation of the cytes. It tends to recover after the MI episode for
main “classical” cardiac K1 currents: Ito, slow Ito, IKr, and IKs. The a-subunit expression for these
(IKS) and rapid delayed rectifier K1 current (IKr), 3 currents normalizes after the acute episode.
4 Algalarrondo & Nattel

Fig. 1. (A) Compared with normal ac-


tion potential (AP) (1), the reduced
K1 currents in ventricular pathology
prolong the action potential duration
(APD) (2); this may induce early after-
depolarizations (3) that can trigger an
ectopic AP (4). (B) Reduced IK1 (1)
reduces resting membrane conduc-
tance, which increases membrane
resistance R. Since the voltage change
(V) induced by any membrane current
(I) is influenced by the membrane
resistance (V 5 IR), any given depola-
rizing current during a delayed after-
depolarization will induce a stronger
depolarization in pathologic condi-
tion than in normal (2), thus more
easily reaching the threshold poten-
tial of the myocyte to trigger an
ectopic AP (3). (C) Propagation of
ectopic activations according to
timing and APD variability. APDs in
3 cells (from 1 to 3) at 4 different
ectopic beat coupling intervals (from
A to D) are represented. If AP vari-
ability is high as often occurs in dis-
ease states (lower panel), premature
beats with coupling conditions B
and C arrive when some cell(s) are
refractory whereas other(s) are
available for activation, which may
induce unidirectional block and
reentry (asterisk). AERP, atrial effec-
tive refractory period; DAD, de-
layed afterdepolarization; EAD, early
afterdepolarization.

Table 2
Changes reported in mRNA and protein expression of KD currents in heart failure

Reference Subunit mRNA Changes Model/Remarks


Kääb et al,75 1998 Kv4.3 Y; HERG, Kv1.4, Kir2.1 4 Human heart
Wang et al,76 1998 Kir2.1, Kir2.2, Kir2.3 4 Human heart
Borlak and Thum,77 2003 Kv4.3, Kv1.5, K2P 1.1, Kir2.1, Kir3.4, Human heart
Kir6.2 Y; ERG 4; KvLQT1, Mink [
Zicha et al,78 2004 Kv4.3 Y; KChiP2 4 Human and dog hearts, proteins
correspond with mRNA
Akar et al,79 2005 Kv4.3 Y; KChiP2, KvLQT1, Mink, Dog heart; proteins correspond with
Kir2.1 4; Kv1.4, ERG [ mRNA but not to current
Rose et al,80 2005 Kv1.4, Kv4.2, KChIP2, Kir2.1 Y; Kv4.3, Tachypacing in rabbit; proteins:
ERG, KvLQT1, Mink 4 Kv1.4, Kv4.2, Kv4.3, KChIP2, Kir2.1
KvLQT1 4
Tsuji et al,41 2006 Kv1.4, Kv4.3, KvLQT1, minK Y; AVB in rabbit; proteins correspond
ERG 4 with mRNA
Yang et al,81 2015 — Rat with AVF. proteins: SK3 [ SK2 4

Abbreviations: Y, decrease; [, increase; 4, no change; AVB, atrioventricular block; AVF, arteriovenous fistula; mRNA,
messenger RNA.
Data from Refs.41,75–81
Potassium Channel Remodeling in Heart Disease 5

Fig. 2. Consequences of repolariza-


tion reserve on action potentials
(APs) and the associated surface elec-
trocardiograph (ECG). Under normal
conditions (A), individual reductions
of IKr or IKs induce only a minor pro-
longation of AP duration (APD) and
QT interval because of repolarization
reserve. (B) When this compensatory
effect is impaired (eg, in heart disease
with reduced IKr and/or IKs) (C), the
APD prolongation is enhanced, as is
as the risk of early after depolariza-
tions (dashed red line).

Fig. 3. Schematic summarizing the


principal alterations in the action po-
tential (AP) and corresponding K1
currents alterations in 4 situations:
heart failure (HF; A), border zone of
the myocardial infarction (MI; B),
atrial fibrillation (AF; C) and pulmo-
nary veins (D). Blue numbers indicate
AP phases. Red are changes in pathol-
ogy (A–C) or differences in pulmonary
vein versus atrium (D).
6 Algalarrondo & Nattel

However, this recovery is incomplete and the b- patients.22 This increased IK(Ach) is caused by acti-
subunit minK remains downregulated in the longer vation of the basal current (with downregulation of
term, which would accelerate IKs inactivation and the carbachol-induced current); Kv3.1 mRNA and
decrease its amplitude (for a review see Ref.12). protein remain stable. Functional upregulation of
constitutive IK(Ach) seems to be caused by changes
Remodeling in the normal zones of hearts with in regulation by protein kinase C isoforms.23
prior myocardial infarction Two-pore domain K1 channels (K2P) have been
Studies of zones remote from the MI scar zone recently identified as potential contributors to AF
have been performed in small animal models (rab- pathophysiology. Expressed mostly in the atria,
bit and rat). APD prolongation leads to triggered these channels are regulated by various factors,
activity and spatial refractoriness heterogeneity. including lipids, pH, temperature, or membrane
As in the border zone, APD prolongation is caused stretch (but not voltage) and thus they conduct a
by downregulation of the main K1 currents. Math- background current throughout the AP. The elec-
ematical modeling of border and normal zones in trophysiologic role of the cardiac K2P channel
MI ventricles highlights the interplay between the K2P3.1 was first characterized in mice.24 Recently,
2 zones and the crucial role of refractoriness het- Schmidt and colleagues25 documented that
erogeneity in generating a substrate for arrhythmia K2P3.1 transcripts, protein, and the corresponding
(Fig. 1C).17 IK2P current are upregulated in the atria of patients
with chronic AF, contributing to AP abbreviation.
Remodeling of K1 Currents Associated with
The contribution of small conductance calcium-
Atrial Fibrillation
activated potassium channels in AF is still contro-
Remodeling of K1 currents in atrial fibrillation versial. It was first documented that increased ISK
AF, the most common sustained cardiac was associated with arrhythmia and that pharma-
arrhythmia, results from a variety of conditions, cologic inhibition of ISK was antiarrhythmic in
like ectopic activity in the PVs and atrial remodel- experimental models.26,27 In subsequent reports,
ing in HF and hypertension. By itself, AF induces however, ISK inhibition did not modify AP shape
electrical remodeling that shortens atrial refractory and the expression of SK2 and SK3 subunits
periods and promotes AF (“AF begets AF”18). This was reduced in human AF atria.27 Therefore, the
remodeling has been reproduced in experimental precise role of ISK in human AF remains to be
models by performing high-rate atrial pacing. It clarified.
has been shown that K1 current changes play a
key role in the pathogenesis of AF. The remodeling Remodeling of K1 currents in pulmonary veins
process begins within the first day after the onset Following the observations from Haı̈ssaguerre and
of AF and results in complex modifications of the colleagues28 of the crucial role of PVs in the initia-
atrial AP (see Fig. 3C). During the initial phase of tion of AF, basic research characterized further the
repolarization (phase 1), APD is prolonged electrophysiology of the myocardial sleeves in
because of the downregulation of Ito; ultrarapid PVs.29,30 Anatomically, myocardial fibers extend
rectifier K1 current (IKur) was documented as from left atria to PVs for 1 to 3 cm, with a specific
reduced or unchanged.12 Thereafter, the structure that transits from a linear to a circular or-
combining effects of stable IKr/IKs, decreased ICaL ganization. At the cellular level, PV cardio-
and increased IK1/acetylcholine-dependent K1 myocytes have distinct properties, including
current [IK(Ach)]/2-pore-domain K1 current (IK2P) decreased phase 0 upstroke velocity and short
induce a shortening of phases 2 and 3 of the AP. APD, and the fibers have a specific organization
IK1 increases also occur, paralleling upregulation (Fig. 3D).29–32 PV sleeve cardiomyocytes also
of Kir2.1 (KCNJ2) mRNA and protein. This upregu- have small background IK1 that could lead to de-
lation is associated with a decreased expression polarization and spontaneous activity.30,33
of microRNAs 1 and 26 (miR-1 and miR-26) both Furthermore, as compared with the left atrium,
in humans and in experimental models.19,20 Simi- PVs show lower expression of Kir3.1 and Kir3.4
larly, decreases in Ito density parallel the downre- that underlie IK(Ach). This K1 channel subunit profile
gulation of Kv4.3 mRNA and protein. Several is associated with lower resting vagal tone at
reports in experimental models suggest that baseline, greater response to vagal stimulation,
reduced miR-1 and increased miR-301a may and shorter refractory periods in PVs than in left
decrease the expression of KCND2 (Kv4.2), and atrium.34 The intrinsic ion channel and AP proper-
that increased miR-133 may reduce the expres- ties of PVs make them more susceptible to
sion of KChIP2 (for further reviews, see Refs.2,21). reentrant arrhythmias. With remodeling, the short
The upregulation of IK(Ach) has been docu- PV AP becomes even shorter; however,
mented both in experimental models and in AF remodeling-induced increases in IK1 should
Potassium Channel Remodeling in Heart Disease 7

restore the intrinsically smaller resting potential in documented in HF,40,41 consistent with the delete-
PV cells. Overall, AF-related tachycardia remodel- rious effect of the IKr blocker sotalol in patients
ing decreases the AP differences between PV and with HF in the Survival With ORal D-sotalol
left atrial cardiomyocytes and in the absence of (SWORD) trial42 and the increased proarrhythmic
other cardiac pathology the PVs do not play a cen- risk with sotalol in HF.43 The increased predisposi-
tral role in the AF promotion caused by atrial tachy- tion to excess APD prolongation with class III
cardia remodeling.35 agents may occur without any APD prolongation
at baseline.36 Practically, loss of repolarization
KD CHANNEL REMODELING: reserve could be detected by recording ECGs
TRANSLATIONAL INTEREST AND CLINICAL and measuring QTc intervals after the initiation of
IMPORTANCE class III antiarrhythmic drugs, possibly within a
Hypokalemia and K1 Currents in Heart few hours of the first oral dose. Another phenome-
Disease Patients non that should be considered in the adverse
event risk of class III antiarrhythmic drugs is their
Hypokalemia ([K1]<3.6 mmol/L) is a common situ- “reverse use dependency.” This “reverse use de-
ation in daily clinical practice that is associated pendency” means that the APD prolonging effect
with increased morbidity and mortality in patients is greater for low cardiac frequencies, which in-
with heart diseases like hypertension, MI, resusci- creases the risk of EAD-triggered arrhythmia with
tation from out-of-hospital cardiac arrest, and bradycardia (for a review see Ref.44).
HF.36,37 Whereas low [K1] should enhance extra-
cellular/intracellular K1 gradient, thus increasing Specific Targets in Atrial Fibrillation
K1 currents, the main effect of hypokalemia’s is
to prolong the QTc interval and APD. This prolon- The upregulation of K1 currents in AF promotes
gation is caused by hypokalemia-induced reduc- the arrhythmia and may contribute to its increasing
tions in multiple K1 currents including IKr, IK1, resistance to drug therapy over time, but may also
and Ito. Recently, it was demonstrated that down- create opportunities for new pharmacologic ap-
regulation of IKr in hypokalemia is caused, at least proaches. In particular, drugs that inhibit inward-
in part, by accelerated internalization and degra- rectifier K1 currents may be particularly interesting
dation of hERG channels.38 Furthermore, hypoka- in view of their role in AF perpetuation.45 On the
lemia downregulates the Na1–K1 ATPase pump other hand, downregulation of ionic currents like
and increases [Na1]i, which leads to increased IKur may limit the value of targeting them in AF.45
[Ca21]i via activation of the Na1/Ca21 exchanger.
Low [K1]e correspondingly increases the effects PHARMACOLOGIC MODULATION OF KD
of class III antiarrhythmic drugs by reducing repo- CHANNELS IN HEART DISEASE PATIENTS
larization reserve; conversely, high [K1]e protects
against drug-induced torsades de pointes. The Reentry is a crucial arrhythmic mechanism.
combined proarrhythmic effects of electrolyte Because short refractory periods increase the like-
imbalance and/or class III antiarrhythmic agents lihood of reentry, it has long been recognized that
multiply the risk of life-threatening arrhythmia if drugs that prolong APD and refractory periods by
they occur on top of a predisposed substrate blocking K1 channels have antiarrhythmic effects
with reduced repolarization reserve. against reentry. Such drugs were allocated to
class III of the Singh and Vaughan Williams46 anti-
Repolarization Reserve arrhythmic drug classification. After the Cardiac
Arrhythmia Suppression Trial (CAST), class III
The term “repolarization reserve” refers to the abil-
agents were widely studied to treat patients with
ity of cardiomyocytes to compensate for the loss
significant heart diseases. Great successes, but
of a repolarizing current by recruiting another one
also great failures, have been recorded in this field
and thus to minimize the APD prolongation
of research.
induced by this loss.39 Repolarization reserve
may result in limited alterations in the AP with K1
Amiodarone
channel block, and the surface ECG could be un-
changed compared with normal. However, if this Amiodarone blocks IKr and IKs to prolong the APD
compensatory mechanism is lost or overridden (class III action),47 but also blocks Na1 channels to
by additional repolarization abnormalities, the reduce conduction velocities (class I action),48 has
resulting effect on the AP, the ECG, and finally a noncompetitive blocking effect on b receptors
the cardiac arrhythmic risk could be exaggerated (class II effect)49 and inhibits ICaL (class IV
(see Fig. 2). Downregulation of K1 currents and action).50 These multiple blocking effects likely
associated repolarization reserve has been contribute to its clinical efficacy and safety.
8 Algalarrondo & Nattel

Clinically, cardiac side effects like torsades de amiodarone, dronedarone blocks INa, ICaL, IKr, IKs,
pointes or HF worsening are unusual, which has Ito, IK(ACh), and b-adrenergic receptors59,60 Clini-
made amiodarone a first-line antiarrhythmic drug cally, the drug has moderate efficacy to maintain
to treat HF patients, illustrating the notion that mul- sinus rhythm in AF patients and clearly inferior ef-
tiple channel blockers may have increased safety ficacy compared to amiodarone. Clinical studies
in patients with remodeling-related decreases in that included significant numbers of HF patients
repolarization reserve. Clinical efficacy was specif- (European Trial of Dronedarone in Moderate to Se-
ically reported in the AF treatment of HF patients.51 vere Congestive Heart Failure [ANDROMEDA] and
In patients with a high risk of sudden cardiac Permanent Atrial Fibrillation Outcome Study Using
death, amiodarone does not prevent sudden Dronedarone on Top of Standard Therapy
death per se; however, in implantable cardi- [PALLAS]) were interrupted early because of
overter–defibrillator carriers, amiodarone plus b- excess events in the dronedarone group,61 again
blockers prevent shocks with greater efficacy highlighting the risks of reduced repolarization
than sotalol or b-blockers alone. reserve with heart disease. As a consequence,
dronedarone should be avoided for patients with
Other Class III Antiarrhythmic Drugs and Heart significant cardiovascular disease.62
Disease
Vernakalant
Available class III anti arrhythmic agents include Vernakalant is a multicurrent blocker that was
ibutilide, dofetilide, and sotalol. All block IKr, pro- developed as an atrial selective antiarrhythmic
longing effective refractory periods with minimal agent.63 The electrophysiologic profile of vernaka-
effects on conduction velocity, and thereby sup- lant includes inhibition of IKur, Ito, IKr, IK(ACh), and
press reentry.52–56 They also, however, carry a INa.64 Vernakalant modestly increases the QTc in-
risk of torsades de pointes (incidence 2.4% for terval and is contraindicated in patients with a
sotalol, 3.6%–8.3% for ibutilide, and 1%–3% for long QTc.65–67 Current clinical use only involves
dofetilide), which is increased in the presence of intravenous administration to convert AF. The
heart disease.45 Predisposing factors for drug- only clinical report on patients with heart diseases
induced torsades de pointes include female sex, involved patients undergoing cardiac surgery and
high drug doses, HF, recent cardioversion, hypo- vernakalant was safe.68
kalemia, bradycardia, and elevated baseline
serum creatinine level.57 Ranolazine
Sotalol is a mixed class III and class II drug. The Ranolazine was initially described as a late Na1
class II action is advantageous to allow for rate current blocker and was primarily evaluated to
control in addition to the rhythm control effect treat chronic angina. However, experimental and
owing to class III properties. However, the brady- clinical studies showed that ranolazine had an
cardic action of sotalol may increase the risk of interesting electrophysiologic profile and could
torsades de pointes. Sotalol may also be useful be potentially helpful to treat AF (for a review see
to reduce the number of shocks in implantable Ref.69). Ranolazine prolongs atrial APD and refrac-
cardioverter–defibrillator carriers. Ibutilide was tory periods but does not prolong ventricular repo-
constructed by comparing sotalol with other b- larization in experimental models of hypertrophy
blockers and with clofilium phosphate, another and CHF.56 Ranolazine is a multichannel blocker
class III antiarrhythmic agent.58 Intravenous ibuti- that inhibits (peak and late) INa, ICaL, Na1/Ca21
lide is used to cardiovert AF and atrial flutter; the exchanger, IKr, and stabilizes RyR2.70,71 Ranola-
risk of ibutilide-induced long QT syndrome is zine was recently tested in a reduced-dose combi-
enhanced in HF patients.56 Because of the risk of nation with dronedarone; the combination was
torsades de pointes with dofetilide, only certified found to have synergistic efficacy with low toxicity
physicians are authorized to prescribe the drug; in AF patients.72
initiation is usually performed in hospital, which
limits its clinical use. SUMMARY
Heart disease produces substantial remodeling of
Other Multichannel Blockers and Heart
K1 channels that in general promotes arrhythmia
Diseases
occurrence. In the case of ventricular arrhythmias,
Dronedarone K1 channel remodeling contributes to the
Dronedarone was constructed on the basis of the arrhythmic risk and increases vulnerability to
amiodarone structure, designed to retain similar torsades de pointes with K1 channel inhibiting
efficacy while avoiding the leading type of amio- drugs. Atrial K1 channel remodeling caused by
darone side effect (thyroid disorders). Like AF promotes arrhythmia stability and presents
Potassium Channel Remodeling in Heart Disease 9

opportunities for the development of new drugs 15. Chua S-K, Chang P-C, Maruyama M, et al. Small-
targeting atrial inward rectifier K1 currents. conductance calcium-activated potassium channel
and recurrent ventricular fibrillation in failing rabbit
ventricles. Circ Res 2011;108:971–9.
REFERENCES
16. Hsieh Y-C, Chang P-C, Hsueh C-H, et al. Apamin-
1. Nerbonne JM, Kass RS. Molecular physiology of car- sensitive potassium current modulates action poten-
diac repolarization. Physiol Rev 2005;85:1205–53. tial duration restitution and arrhythmogenesis of
2. Schmitt N, Grunnet M, Olesen S-P. Cardiac potas- failing rabbit ventricles. Circ Arrhythm Electrophysiol
sium channel subtypes: new roles in repolarization 2013;6:410–8.
and arrhythmia. Physiol Rev 2014;94:609–53. 17. Decker KF, Rudy Y. Ionic mechanisms of electro-
3. Schillaci G, Pirro M, Ronti T, et al. Prognostic impact physiological heterogeneity and conduction block
of prolonged ventricular repolarization in hyperten- in the infarct border zone. Am J Physiol Heart Circ
sion. Arch Intern Med 2006;166:909–13. Physiol 2010;299:H1588–97.
4. Salles GF, Cardoso CRL, Muxfeldt ES. Prognostic 18. Wijffels MC, Kirchhof CJ, Dorland R, et al. Atrial fibril-
value of ventricular repolarization prolongation in lation begets atrial fibrillation. A study in awake
resistant hypertension: a prospective cohort study. chronically instrumented goats. Circulation 1995;
J Hypertens 2009;27:1094–101. 92:1954–68.
5. Karpanou EA, Vyssoulis GP, Psichogios A, et al. 19. Girmatsion Z, Biliczki P, Bonauer A, et al. Changes
Regression of left ventricular hypertrophy results in in microRNA-1 expression and IK1 up-regulation in
improvement of QT dispersion in patients with hy- human atrial fibrillation. Heart Rhythm 2009;6:
pertension. Am Heart J 1998;136:765–8. 1802–9.
6. Klimas J, Kruzliak P, Rabkin SW. Modulation of the 20. Luo X, Pan Z, Shan H, et al. MicroRNA-26 governs
QT interval duration in hypertension with antihyper- profibrillatory inward-rectifier potassium current
tensive treatment. Hypertens Res 2015;38:447–54. changes in atrial fibrillation. J Clin Invest 2013;123:
7. Marionneau C, Brunet S, Flagg TP, et al. Distinct 1939–51.
cellular and molecular mechanisms underlie func- 21. Luo X, Yang B, Nattel S. MicroRNAs and atrial fibril-
tional remodeling of repolarizing K1 currents with lation: mechanisms and translational potential. Nat
left ventricular hypertrophy. Circ Res 2008;102: Rev Cardiol 2015;12:80–90.
1406–15. 22. Voigt N, Trausch A, Knaut M, et al. Left-to-right atrial
8. Omiya K, Sekizuka H, Kida K, et al. Influence of inward rectifier potassium current gradients in pa-
gender and types of sports training on QT variables tients with paroxysmal versus chronic atrial fibrilla-
in young elite athletes. Eur J Sport Sci 2014; tion. Circ Arrhythm Electrophysiol 2010;3:472–80.
14(Suppl 1):S32–8. 23. Makary S, Voigt N, Maguy A, et al. Differential pro-
9. Yang K-C, Foeger NC, Marionneau C, et al. Homeo- tein kinase C isoform regulation and increased
static regulation of electrical excitability in physio- constitutive activity of acetylcholine-regulated po-
logical cardiac hypertrophy. J Physiol 2010;588: tassium channels in atrial remodeling. Circ Res
5015–32. 2011;109:1031–43.
10. Davey PP, Barlow C, Hart G. Prolongation of the QT 24. Donner BC, Schullenberg M, Geduldig N, et al.
interval in heart failure occurs at low but not at high Functional role of TASK-1 in the heart: studies in
heart rates. Clin Sci (Lond) 2000;98:603–10. TASK-1-deficient mice show prolonged cardiac
11. Karwatowska-Prokopczuk E, Wang W, Cheng ML, repolarization and reduced heart rate variability.
et al. The risk of sudden cardiac death in patients Basic Res Cardiol 2011;106:75–87.
with non-ST elevation acute coronary syndrome 25. Schmidt C, Wiedmann F, Voigt N, et al. Upregulation
and prolonged QTc interval: effect of ranolazine. of K(2P)3.1 K1 Current causes action potential
Europace 2013;15:429–36. shortening in patients with chronic atrial fibrillation.
12. Nattel S, Maguy A, Le Bouter S, et al. Arrhythmo- Circulation 2015;132:82–92.
genic ion-channel remodeling in the heart: heart fail- 26. Ozgen N, Dun W, Sosunov EA, et al. Early electrical
ure, myocardial infarction, and atrial fibrillation. remodeling in rabbit pulmonary vein results from
Physiol Rev 2007;87:425–56. trafficking of intracellular SK2 channels to mem-
13. Long VP, Bonilla IM, Vargas-Pinto P, et al. Heart fail- brane sites. Cardiovasc Res 2007;75:758–69.
ure duration progressively modulates the arrhythmia 27. Skibsbye L, Poulet C, Diness JG, et al. Small-
substrate through structural and electrical remodel- conductance calcium-activated potassium (SK)
ing. Life Sci 2015;123:61–71. channels contribute to action potential repolariza-
14. Yu C-C, Corr C, Shen C, et al. Small conductance tion in human atria. Cardiovasc Res 2014;103:
calcium-activated potassium current is important in 156–67.
transmural repolarization of failing human ventricles. 28. Haı̈ssaguerre M, Jaı̈s P, Shah DC, et al. Sponta-
Circ Arrhythm Electrophysiol 2015;8:667–76. neous initiation of atrial fibrillation by ectopic beats
10 Algalarrondo & Nattel

originating in the pulmonary veins. N Engl J Med 43. Lehmann MH, Hardy S, Archibald D, et al. Sex differ-
1998;339(10):659–66. ence in risk of torsade de pointes with d,l-sotalol.
29. Ehrlich JR, Cha T-J, Zhang L, et al. Characterization Circulation 1996;94:2535–41.
of a hyperpolarization-activated time-dependent po- 44. Dorian P. Mechanisms of action of class III agents
tassium current in canine cardiomyocytes from pul- and their clinical relevance. Europace 2000;
monary vein myocardial sleeves and left atrium. 1(Suppl C):C6–9.
J Physiol 2004;557:583–97. 45. Dobrev D, Carlsson L, Nattel S. Novel molecular tar-
30. Ehrlich JR, Cha T-J, Zhang L, et al. Cellular electro- gets for atrial fibrillation therapy. Nat Rev Drug Dis-
physiology of canine pulmonary vein cardiomyo- cov 2012;11:275–91.
cytes: action potential and ionic current properties. 46. Singh BN, Vaughan Williams EM. A third class of
J Physiol 2003;551:801–13. anti-arrhythmic action. Effects on atrial and ventricu-
31. Jaı̈s P, Hocini M, Macle L, et al. Distinctive electro- lar intracellular potentials, and other pharmacolog-
physiological properties of pulmonary veins in pa- ical actions on cardiac muscle, of MJ 1999 and
tients with atrial fibrillation. Circulation 2002;106: AH 3474. Br J Pharmacol 1970;39:675–87.
2479–85. 47. Kodama I, Kamiya K, Toyama J. Cellular electrophar-
32. Chen Y-C, Pan N-H, Cheng C-C, et al. Heteroge- macology of amiodarone. Cardiovasc Res 1997;35:
neous expression of potassium currents and 13–29.
pacemaker currents potentially regulates arrhyth- 48. Mason JW, Hondeghem LM, Katzung BG. Block of
mogenesis of pulmonary vein cardiomyocytes. inactivated sodium channels and of depolarization-
J Cardiovasc Electrophysiol 2009;20:1039–45. induced automaticity in guinea pig papillary muscle
33. Nattel S. Paroxysmal atrial fibrillation and pulmonary by amiodarone. Circ Res 1984;55:278–85.
veins: relationships between clinical forms and auto- 49. Kadoya M, Konishi T, Tamamura T, et al. Electro-
matic versus re-entrant mechanisms. Can J Cardiol physiological effects of amiodarone on isolated rab-
2013;29:1147–9. bit heart muscles. J Cardiovasc Pharmacol 1985;7:
34. Arora R, Ng J, Ulphani J, et al. Unique autonomic 643–8.
profile of the pulmonary veins and posterior left 50. Nattel S, Talajic M, Quantz M, et al. Frequency-
atrium. J Am Coll Cardiol 2007;49:1340–8. dependent effects of amiodarone on atrioventricular
35. Cha T-J, Ehrlich JR, Zhang L, et al. Atrial ionic re- nodal function and slow-channel action potentials:
modeling induced by atrial tachycardia in the pres- evidence for calcium channel-blocking activity. Cir-
ence of congestive heart failure. Circulation 2004; culation 1987;76:442–9.
110:1520–6. 51. Deedwania PC, Singh BN, Ellenbogen K, et al.
36. Gennari FJ. Hypokalemia. N Engl J Med 1998;339: Spontaneous conversion and maintenance of sinus
451–8. rhythm by amiodarone in patients with heart failure
37. Osadchii OE. Mechanisms of hypokalemia-induced and atrial fibrillation: observations from the veterans
ventricular arrhythmogenicity. Fundam Clin Pharma- affairs congestive heart failure survival trial of antiar-
col 2010;24:547–59. rhythmic therapy (CHF-STAT). The Department of
38. Guo J, Massaeli H, Xu J, et al. Extracellular K1 con- Veterans Affairs CHF-STAT Investigators. Circulation
centration controls cell surface density of IKr in rab- 1998;98:2574–9.
bit hearts and of the HERG channel in human cell 52. Sanguinetti MC, Jurkiewicz NK. Two components of
lines. J Clin Invest 2009;119:2745–57. cardiac delayed rectifier K1 current. Differential
39. Roden DM. Taking the “idio” out of “idiosyncratic”: sensitivity to block by class III antiarrhythmic agents.
predicting torsades de pointes. Pacing Clin Electro- J Gen Physiol 1990;96:195–215.
physiol 1998;21:1029–34. 53. Jurkiewicz NK, Sanguinetti MC. Rate-dependent
40. Han W, Chartier D, Li D, et al. Ionic remodeling of prolongation of cardiac action potentials by a meth-
cardiac Purkinje cells by congestive heart failure. anesulfonanilide class III antiarrhythmic agent. Spe-
Circulation 2001;104:2095–100. cific block of rapidly activating delayed rectifier K1
41. Tsuji Y, Zicha S, Qi X-Y, et al. Potassium channel current by dofetilide. Circ Res 1993;72:75–83.
subunit remodeling in rabbits exposed to long- 54. Yang T, Snyders DJ, Roden DM. Ibutilide, a metha-
term bradycardia or tachycardia: discrete arrhyth- nesulfonanilide antiarrhythmic, is a potent blocker
mogenic consequences related to differential of the rapidly activating delayed rectifier K1 current
delayed-rectifier changes. Circulation 2006;113: (IKr) in AT-1 cells. Concentration-, time-, voltage-,
345–55. and use-dependent effects. Circulation 1995;91:
42. Waldo AL, Camm AJ, deRuyter H, et al. Effect of d- 1799–806.
sotalol on mortality in patients with left ventricular 55. Tai CT, Chen SA, Feng AN, et al. Electropharmaco-
dysfunction after recent and remote myocardial logic effects of class I and class III antiarrhythmia
infarction. The SWORD Investigators. Survival with drugs on typical atrial flutter: insights into the mech-
Oral d-Sotalol. Lancet 1996;348:7–12. anism of termination. Circulation 1998;97:1935–45.
Potassium Channel Remodeling in Heart Disease 11

56. Kowey PR, VanderLugt JT, Luderer JR. Safety and risk/ 69. Gupta T, Khera S, Kolte D, et al. Antiarrhythmic
benefit analysis of ibutilide for acute conversion of properties of ranolazine: a review of the current evi-
atrial fibrillation/flutter. Am J Cardiol 1996;78:46–52. dence. Int J Cardiol 2015;187:66–74.
57. Kannankeril P, Roden DM, Darbar D. Drug-induced 70. Antzelevitch C, Belardinelli L, Zygmunt AC, et al.
long QT syndrome. Pharmacol Rev 2010;62:760–81. Electrophysiological effects of ranolazine, a novel
58. Hester JB, Gibson JK, Cimini MG, et al. N-[(omega- antianginal agent with antiarrhythmic properties. Cir-
amino-1-hydroxyalkyl)phenyl]methanesulfonamide culation 2004;110:904–10.
derivatives with class III antiarrhythmic activity. 71. Parikh A, Mantravadi R, Kozhevnikov D, et al. Rano-
J Med Chem 1991;34:308–15. lazine stabilizes cardiac ryanodine receptors: a
59. Varró A, Takács J, Németh M, et al. Electrophysio- novel mechanism for the suppression of early after-
logical effects of dronedarone (SR 33589), a nonio- depolarization and torsades de pointes in long QT
dinated amiodarone derivative in the canine heart: type 2. Heart Rhythm 2012;9:953–60.
comparison with amiodarone. Br J Pharmacol 72. Reiffel JA, Camm AJ, Belardinelli L, et al, HARMONY
2001;133:625–34. Investigators. The HARMONY trial: combined rano-
60. Gautier P, Guillemare E, Marion A, et al. Electrophys- lazine and dronedarone in the management of
iologic characterization of dronedarone in guinea paroxysmal atrial fibrillation: mechanistic and thera-
pig ventricular cells. J Cardiovasc Pharmacol peutic synergism. Circ Arrhythm Electrophysiol
2003;41:191–202. 2015;8:1048–56.
61. Nattel S. Dronedarone in atrial fibrillation–Jekyll and 73. Volk T, Nguyen TH, Schultz JH, et al. Regional alter-
Hyde? N Engl J Med 2011;365:2321–2. ations of repolarizing K1 currents among the left
62. Camm AJ, Lip GYH, De Caterina R, et al. ESC Com- ventricular free wall of rats with ascending aortic ste-
mittee for Practice Guidelines-CPG, Document Re- nosis. J Physiol 2001;530:443–55.
viewers. 2012 focused update of the ESC 74. Yang K-C, Nerbonne JM. Mechanisms contributing
Guidelines for the management of atrial fibrillation: to myocardial potassium channel diversity, regula-
an update of the 2010 ESC Guidelines for the man- tion and remodeling. Trends Cardiovasc Med
agement of atrial fibrillation–developed with the spe- 2015;15:S1050–738.
cial contribution of the European Heart Rhythm 75. Kääb S, Dixon J, Duc J, et al. Molecular basis of
Association. Europace 2012;14:1385–413. transient outward potassium current downregulation
63. Nattel S, De Blasio E, Beatch GN, et al. RSD1235: a in human heart failure: a decrease in Kv4.3 mRNA
novel antiarrhythmic agent with a unique electro- correlates with a reduction in current density. Circu-
physiological profile that terminates AF in dogs. lation 1998;98:1383–93.
Eur Heart J 2001;22(Suppl):448. 76. Wang Z, Yue L, White M, et al. Differential distribu-
64. Fedida D, Orth PMR, Chen JYC, et al. The mecha- tion of inward rectifier potassium channel transcripts
nism of atrial antiarrhythmic action of RSD1235. in human atrium versus ventricle. Circulation 1998;
J Cardiovasc Electrophysiol 2005;16:1227–38. 98:2422–8.
65. Dorian P, Pinter A, Mangat I, et al. The effect of ver- 77. Borlak J, Thum T. Hallmarks of ion channel gene
nakalant (RSD1235), an investigational antiar- expression in end-stage heart failure. FASEB J
rhythmic agent, on atrial electrophysiology in 2003;17:1592–608.
humans. J Cardiovasc Pharmacol 2007;50:35–40. 78. Zicha S, Xiao L, Stafford S, et al. Transmural expres-
66. Roy D, Pratt CM, Torp-Pedersen C, et al, Atrial sion of transient outward potassium current subunits
Arrhythmia Conversion Trial Investigators. Vernaka- in normal and failing canine and human hearts.
lant hydrochloride for rapid conversion of atrial fibril- J Physiol 2004;561:735–48.
lation: a phase 3, randomized, placebo-controlled 79. Akar FG, Wu RC, Juang GJ, et al. Molecular mech-
trial. Circulation 2008;117:1518–25. anisms underlying K1 current downregulation in
67. Camm AJ, Capucci A, Hohnloser SH, et al. canine tachycardia-induced heart failure. Am J
A randomized active-controlled study comparing Physiol Heart Circ Physiol 2005;288:H2887–96.
the efficacy and safety of vernakalant to amiodarone 80. Rose J, Armoundas AA, Tian Y, et al. Molecular cor-
in recent-onset atrial fibrillation. J Am Coll Cardiol relates of altered expression of potassium currents
2011;57:313–21. in failing rabbit myocardium. Am J Physiol Heart
68. Kowey PR, Dorian P, Mitchell LB, et al, Atrial Circ Physiol 2005;288:H2077–87.
Arrhythmia Conversion Trial Investigators. Vernaka- 81. Yang D, Wang T, Ni Y, et al. Apamin-Sensitive
lant hydrochloride for the rapid conversion of atrial K(1) current upregulation in volume-overload
fibrillation after cardiac surgery: a randomized, heart failure is associated with the decreased
double-blind, placebo-controlled trial. Circ Arrhythm interaction of CK2 with SK2. J Membr Biol 2015;
Electrophysiol 2009;2:652–9. 248:1181–9.

You might also like