You are on page 1of 5

Europace (2008) 10, 1133–1137

doi:10.1093/europace/eun193

REVIEW

Role of potassium currents in cardiac arrhythmias


Ursula Ravens1* and Elisabetta Cerbai2
1
Department of Pharmacology and Toxicology, Medical Faculty, Dresden University of Technology, Dresden, Germany;
and 2Centro Interuniversitario di Medicina Molecolare, University of Florence, Florence, Italy
Received 29 April 2008; accepted after revision 4 July 2008; online publish-ahead-of-print 24 July 2008

KEYWORDS Abnormal excitability of myocardial cells may give rise to ectopic beats and initiate re-entry around an
Potassium channels; anatomical or functional obstacle. As Kþ currents control the repolarization process of the cardiac
Ventricular fibrillation; action potential (AP), the Kþ channel function determines membrane potential and refractoriness of
Torsades de pointes; the myocardium. Both gain and loss of the Kþ channel function can lead to arrhythmia. The former
Atrial fibrillation; because abbreviation of the active potential duration (APD) shortens refractoriness and wave length,
Genetic polymorphism; and thereby facilitates re-entry and the latter because excessive prolongation of APD may lead to
Drugs; torsades de pointes (TdP) arrhythmia and sudden cardiac death. The pro-arrhythmic consequences of
Remodelling malfunctioning Kþ channels in ventricular and atrial tissue are discussed in the light of three pathophy-
siologically relevant aspects: genetic background, drug action, and disease-induced remodelling. In the
ventricles, loss-of-function mutations in the genes encoding for Kþ channels and many drugs (mainly
hERG channel blockers) are related to hereditary and acquired long-QT syndrome, respectively, that
put individuals at high risk for developing TdP arrhythmias and life-threatening ventricular fibrillation.
Similarly, down-regulation of Kþ channels in heart failure also increases the risk for sudden cardiac
death. Mutations and polymorphisms in genes encoding for atrial Kþ channels can be associated with
gain-of-function and shortened, or with loss-of-function and prolonged APs. The block of atrial Kþ chan-
nels becomes a particular therapeutic challenge when trying to ameliorate atrial fibrillation (AF). This
arrhythmia has a strong tendency to cause electrical remodelling, which affects many Kþ channels.
Atrial-selective drugs for the treatment of AF without affecting the ventricles could target structures
such as IKur or constitutively active IK,ACh channels.

Introduction and a plateau phase (phase 2), in which repolarization is


slowed due to the activation of inward Ca2þ current.
Regular excitation is generated in the sino-atrial node and During the final rapid repolarization phase (phase 3),
spreads throughout the heart in an orderly manner, membrane potential returns to the resting level (phase 4).
whereas disorganization of electrical activity is the basis The various potassium (Kþ) currents are repolarizing
of atrial or ventricular fibrillation. Arrhythmias are caused outward currents. Their major contributions to the cardiac
by the perturbation of physiological impulse formation, AP are control of stable resting membrane potential and
impaired impulse conduction, or disturbed electrical termination of the AP.
recovery. Abnormal excitability of myocardial cells may
give rise to ectopic beats and initiate re-entry around an
anatomical or functional obstacle (reviewed in Jalife1).
The long-lasting action potential (AP) of the working Potassium channels
myocardium maintains a refractory state for as long as the Ions traverse the lipid bilayer of the plasmalemma along their
muscle contracts and thus ensures rhythmic pump function. electrochemical gradient via hydrophilic ion channels. These
Typical cardiac APs consist of five distinct phases (Figure 1). ion channels open and close in a voltage- and time-dependent
In phase 0, Naþ influx triggers a rapid depolarization manner (activation, inactivation, or deactivation) and pass
followed by an early fast repolarization phase (phase 1) ions only in the open state. After-repolarization, some chan-
nels must recover from inactivation before they become
available for re-opening, and during this time, the myocardial
* Corresponding author: Institut für Pharmakologie und Toxikologie, cells remain refractory for re-excitation.
Fetscherstr. 74, 01307 Dresden, Germany. Tel: þ49 351 4586300; fax: þ49
351 4586315. Potassium channels form the largest family of ion
E-mail address: ravens@rcs.urz.tu-dresden.de channel proteins. Determining and depending on membrane

Published on behalf of the European Society of Cardiology. All rights reserved. & The Author 2008.
For permissions please email: journals.permissions@oxfordjournals.org.

Downloaded from https://academic.oup.com/europace/article-abstract/10/10/1133/469096


by guest
on 03 February 2018
1134 U. Ravens and E. Cerbai

Figure 1 Inward, depolarizing and outward, repolarizing currents that underlie the atrial and ventricular action potential. Inward currents:
INa sodium current; ICa,L L-type calcium current; Ito transient outward current; IKur ultra rapidly activating delayed rectifier current; IKr and IKs
rapidly and slowly activating delayed rectifier current; IK1 inward rectifier current; IK,ACh acetylcholine-activated potassium current. Note
that IKur is present in atria only. Phase 0, rapid depolarization; phase 1, rapid early repolarization phase; phase 2, slow repolarization
phase (‘plateau’ phase); phase 3, rapid late repolarization phase; phase 4, resting membrane potential. Adapted from The Sicilian gambit.28

potential, Kþ channels can also be activated by ligand Potassium channels—differences between


binding, and hence are divided into voltage- and ligand-gated atrium and ventricle
channels. The ion-conducting pore of a Kþ channel is formed
by four a-subunits that co-assemble as homo- or hetero- Despite general similarity in the mechanisms of AP gener-
tetramers with different biophysical properties. Their gating ation, APs exhibit distinct shapes in atrial and ventricular
characteristics are further modulated by ancillary subunits. myocardium (Figure 1). The most striking differences are
Cardiac Kþ channels are further classified according to that (i) the plateau phase occurs at more negative potentials
their function (cf. Figure 1). (i) The transient outward and (ii) overall APD is shorter in atrial when compared with
current Ito exhibits rapid activation and subsequent ventricular cells. These differences are due to the non-
inactivation during the early repolarization phase. (ii) The uniform distribution of ion channels, including Kþ channels.
delayed rectifier channels conduct at least three different For instance, IKur is detected only in atrial but not in ventri-
currents IKur, IKr, and IKs. All three currents activate at posi- cular myocytes, although the Kv1.5 protein is abundant in
tive potentials with distinct time courses, i.e. ultrarapid, both chambers. Rapid activation of IKur in the positive poten-
rapid, and slow, respectively. Inactivation of IKur and IKs is tial range following the AP upstroke may offset depolarizing
slow, whereas that of IKr is extremely fast. Contribution of ICa,L and hence leads to the less positive plateau phase
each of these currents to repolarization depends on the in atrial than ventricular cells. The acetylcholine-activated
number of open channels and the electrochemical driving inward rectifying current IK,ACh, too, is not detected in
force that declines as the membrane potential returns to ventricle.
its resting level. In the course of repolarization, IKr recovers
rapidly from inactivation into an open state which deacti- Contribution of K1 channel function to
vates very slowly, so that this current exhibits inward
arrhythmias
rectification.2 Therefore, IKr increases again at negative
potentials and strongly accelerates final repolarization. Because of their impact on membrane potential and
(iii) The major classical cardiac inward rectifier K þcurrent refractoriness, Kþ currents play a prominent role in arrhyth-
is IK1. This channel is always open and conducts Kþ better mogenesis. An increase in Kþ currents abbreviates APD and
into than out of the cell. Atrial myocytes also express an thereby facilitates re-entry. Conversely, impaired Kþ current
acetylcholine-dependent channel that conducts IK,ACh amplitudes prolong APD with contrasting outcome, whereas
in response to the stimulation of G-protein-coupled mus- moderate prolongation maintains the myocardium in a refrac-
carinic (M2) and adenosine (A1) receptors. The activation tory state and is actually considered an anti-arrhythmic
of IK,ACh hyperpolarizes the membrane and shortens the mechanism (class III anti-arrhythmic action according to the
active potential duration (APD). The third inward rectifier classification of Vaughan Williams), excessive prolongation
channel in cardiomyocytes is closed under physiological predisposes to early after-depolarizations that may trigger
metabolic conditions and is activated when the cells are torsades de pointes arrhythmias (TdP). This form of
deprived of intra-cellular adenosine triphosphate. Similar arrhythmia may either resolve spontaneously or deteriorate
to IK,ACh, IK,ATP causes profound APD shortening. into ventricular fibrillation causing sudden cardiac death.

Downloaded from https://academic.oup.com/europace/article-abstract/10/10/1133/469096


by guest
on 03 February 2018
Role of potassium currents in cardiac arrhythmias 1135

Therefore, both gain and loss of Kþ channel function can (increase in ‘repolarization reserve’).6,7 Although there
lead to arrhythmia. Malfunction of Kþ channels may be appears to be no strict concentration–response relationship
due to diverse causes such as mutations in the encoding for triggering TdP, drug plasma concentrations should not be
genes for any of the subunits, drug actions, or remodelling allowed to rise above the therapeutic level and interference
in adaptation to heart disease. These three factors will be with drug metabolism or excretion should be avoided.
discussed first for ventricular and, subsequently, for atrial Even in asymptomatic patients, a genetic predisposition
arrhythmias. related to congenital LQTS may exacerbate drug action
that turns the otherwise borderline QT interval into overt
prolongation. Some polymorphisms, for instance, in the
Ventricular arrhythmias gene encoding for a common ancillary subunit (i.e. the
Arrhythmogenic potential of K1 channel mutations KCNE2 gene) have normal function at baseline, but are
susceptible to block by sulfamethoxazole that imposes no
The number of mutations identified in Kþ channel-encoding
prolongation in healthy individuals.8
genes that have been related to arrhythmias is increasing
Arrhythmogenic exacerbation frequently occurs due to
rapidly. The consequences of missense mutations include
pharmacokinetic interactions by co-administered drugs
faulty protein folding and disturbed co-assembly between
that interfere with biotransformation and excretion of a
subunits and therefore early degradation, disruption of
previously tolerated drug resulting in excessive plasma
trafficking or defects in plasmalemmal integration, altered
concentrations. If the parent compound is more effective
voltage dependency, or impaired ion selectivity of the
than the metabolite in producing a pro-arrhythmic event,
channel. These adverse effects reduce repolarizing Kþ
inhibition of drug metabolizing enzymes will enhance
currents and thereby delay the repolarization process. The
arrhythmogenicity. Conversely, if the metabolite is more
resulting long-cardiac APD or long-QT interval in the electro-
effective, induction of enzymes is pro-arrhythmogenic.
cardiogram predisposes a patient to TdP arrhythmias. There-
The antifungal agent, ketoconazole for instance, interferes
fore, patients with the hereditary long-QT syndrome (LQTS)
with biotransformation of the non-sedating antihistaminic
are at increased risk for sudden cardiac death. Mutations in
terfenadine into a metabolite that does not prolong the
the genes for KvLQT1 (IKs) and hERG (IKr) account for 80–90%
AP. Thus, co-medication of the two drugs results in a high
of all hereditary LQTS, although mutations that cause LQTS
plasma concentration of terfenadine leading to acquired
were also discovered in other ion channels, in ion-handling,
LQTS. Such an interaction applies for many drugs that
or associated proteins (for recent review, see Saenen and
inhibit cytochrome P450 enzymes and even grapefruit
Vrints3). As mentioned above, rare gain-of-function mutations
juice may interfere (reviewied in Priori et al. 4).
in hERG and KvLQT1 shorten cardiac APs and also give rise
Intuitively, cardiac drugs and, in particular, anti-
to potentially lethal arrhythmias (short-QT syndrome).
arrhythmics that prolong APD (class III action according to
the Vaughan Williams classification) are expected to produce
Arrhythmogenic potential of K1 channel blockers an increased risk not only because of their mechanism of
Many drugs used in cardiac and non-cardiac diseases prolong action but also because they are given to patients with dis-
APs and give rise to acquired LQTS. Besides underlying heart eased hearts that are per se at a high risk for rhythm disturb-
disease, several factors predispose to drug-induced TdP. ances. However, drugs for treating non-cardiac diseases such
These include female gender, long-QT interval at baseline, as antihistamines, antibiotics, antipsychotic, or prokinetic
bradycardia, low Kþ and Mg2þ plasma levels, old age, and agents also may increase risk. Comprehensive lists of drugs
the increased incidence of heart disease. Similar to con- with reported or suspected risk for TdP are available in the
genital LQTS, the actual incidence of drug-induced TdP is internet (for instance, http://www.qtdrugs.org/).
low and that of proven drug-associated syncope or sudden
cardiac death is even lower.4 The absolute incidence of
cardiotoxicity of any drug must be judged in relation to
Disease-induced remodelling
the severity of the treated disease: a high risk may be Heart failure is associated with a plethora of ventricular
perfectly acceptable when treating a life-threatening ionic changes that predispose to arrhythmias (for recent
condition, whereas even a very low incidence as reported review, see Nass et al. 9). These remodelling processes
for non-sedating antihistaminic drugs is not acceptable as include electrophysiological changes that lead to APD
these drugs are widely prescribed for minor complaints. prolongation, especially in the endocardial region. We and
Nevertheless, the increasing number of drugs recognized others have shown that Ito amplitude is down-regulated,
to cause acquired LQTS has become a concern for patients, but other Kþ currents (IKr, IKs, and IK1) are depressed as
physicians, and safety regulation authorities. well.10,11 Reduced Kþ currents increase the propensity for
Almost all drugs with reported QT-prolongation and early after-depolarizations, dispersion of repolarization,
TdP are blockers of potassium channels, particularly IKr and ventricular arrhythmias, thereby significantly increasing
channels.5 In fact, hERG channels are sensitive to block by the risk of sudden cardiac death in heart failure patients. In
a surprisingly large variety of agents, and block of hERG contrast, delayed after-depolarizations occurring at high
channels is so common that drug safety agencies require intra-cellular Ca2þ load may initiate triggered activity
data on hERG channel block for new drugs when filed for and induce ventricular tachycardia, particularly in non-
registration. hERG screening tests are performed at very ischaemic heart failure patients, and decreased IK1 enhances
early stages in drug development and even promising new the propensity for triggered activity.12 Kþ channel down-
compounds are usually abandoned when testing positive. regulation also contributes to enhanced sensitivity of
This is not always justified, because the block of IKr may in failing myocardium to other triggering factors such as
part be compensated by changes in opposing currents hypokalaemia, ischaemia, and anti-arrhythmic agents with

Downloaded from https://academic.oup.com/europace/article-abstract/10/10/1133/469096


by guest
on 03 February 2018
1136 U. Ravens and E. Cerbai

class III effects. In addition, overexpression of pacemaker induce AF;19 however, unlike ventricular fibrillation, this
channels may predispose ventricular myocytes from failing arrhythmias is not immediately life threatening.
hearts to enhanced automaticity.13
Disease-induced remodelling
Atrial arrhythmias Atrial fibrillation has a strong tendency to become persistent.
Atrial fibrillation (AF) is initiated when a suitable trigger Persistent AF is thought to develop because of ‘remodelling’,
meets an appropriate substrate. The underlying pathophy- which refers to any change in the atrial function that pro-
siological mechanisms include ectopic electrical activity motes AF or occurs as a consequence of the arrhythmia20
and single or multiple re-entry circuits. Re-entry often and involves alterations in myocardial structural and electro-
develops in large fibrotic atria associated with valvular physiological properties. Structural remodelling includes
disease or heart failure, whereas in seemingly healthy interstitial fibrosis and is closely related to the renin–
hearts, rapid local ectopic activity initiating in the pulmon- angiotensin–aldosterone system.21 Electrical remodelling
ary veins can give rise to re-entry circuits. comprises triangularization of the AP shape, shortening of
APD, and lack of adaptation of APD to cycle length and is
Arrhythmogenic potential of K1 channel mutations due to alterations in the ion channel function as well as
changes in Ca2þ handling of the cardiomyocytes.11,22
In contrast to the vast majority of several hundreds of known
Reduced refractoriness supports multiple wavelet re-entry
Kþ channel mutations being associated with LQTS, there
and generation of sustained, high-frequency spiral waves
are, but a few, mutations directly linked to familial AF
(‘rotors’) with fibrillatory conduction, which are thought to
(Table 1). Nevertheless, LQTS patients may also exhibit poly-
contribute to sustained AF.23
morphic atrial tachyarrhythmias.14 Although in the ventri-
Electrical remodelling by AF affects depolarizing currents
cles loss-of-function mutations predominantly predispose
(with reduced ICa,L, unaffected conventional INa, and acti-
to chaotic electrical activity, the Kþ channel mutations
vated sodium/calcium exchanger) as well as repolarizing
associated with AF exhibit—with one exception—gain-of-
Kþ currents with Ito and IKur current densities being
function, leading to abbreviation of the APD and facilitating
decreased. The contribution of IKr and IKs to electrical remo-
re-entry. In the individual studies, gain-of-function was
delling is less clear, because their presence in the human
verified by expressing mutated channels in cell lines and
atrium is difficult to assess experimentally.24 Basal activity
measuring increased Kþ current density when compared
of the inwardly rectifying IK1 current is enhanced, but
with wild-type channels (reviewed in Roberts15). For the
acetylcholine-activated IK,ACh current is reduced. Moreover,
loss-of-function mutation, it was demonstrated that mice
a component of constitutively active IK,ACh that activates in
expressing the mutated channels had long APs with early
the absence of any agonist is detected in persistent AF and
after-depolarizations and were more prone to stress-induced
may contribute to AP shortening.25 The contribution of
arrhythmias.16 Recent evidence from a genome-wide associ-
other ion channels (chloride channels, TASK-1, and so on)
ation study identified three novel single nucleotide poly-
to AF pathophysiology is still unknown.
morphisms that confer predisposition to AF.17 Moreover, a
Atrial fibrillation-induced changes in Kþ current amplitude
systematic candidate gene-based analysis of the gene
need not necessarily be due to corresponding alteration in
encoding for hERG discovered a hitherto unknown variant,
the gene expression, but can be caused by AF-induced
which strongly associated with AF.18
effects on protein kinases and phosphatases that modify
channel phosphorylation status (see, for instance, Christ
Arrhythmogenic potential of drugs acting et al. 6) or by enhanced protein degradation through pro-
on K1 channels teases.21 There is some controversy in the literature con-
Vagal nerve stimulation induces AF because of shortening of cerning the expression of Kþ channels in remodelled atria
the refractory period due to acetylcholine release and sub- (see Dobrev and Ravens,22 for references): mRNA and
sequent activation of IK,ACh. In analogy, drugs that shorten protein expression of Ito channels are always found
APD by activating Kþ currents are expected to predispose depressed, IKur channels are not changed or decreased; IKr
to AF. However, much less is known about the actual inci- channels are not altered or decreased and IKs channels are
dence of drug-induced AF than of TdP in the case of LQTS. decreased or increased; inward rectifier Kþ channels for
As outlined above, prolongation of atrial APD by the block IK1 are enhanced, for IK,ACh and IK,ATP decreased.26,27 More-
of Kþ channels, as, for instance, with cesium, may also over, these discrepant findings do not always go in line

Table 1 Mutations in genes encoding for potassium channel proteins associated with familial atrial fibrillation

Gene (Protein) Current Mutation Amino acid change Change Origin References

KCNA5 (Kv1.5) IKur 1123G!T E375X Loss-of-function Mostly Caucasian Olson et al.16
KCNH2 (Kv11.1) IKr 1764C!G N588K Gain-of-function — Hong et al.29
KCNQ1 (Kv7.1) IKs 418A!G S140G Gain-of-function Chinese Chen et al.30
KCNQ1 (Kv7.1) IKs 491G!A V141M Gain-of-function Caucasian Hong et al.31
KCNQ1 (Kv7.1) IKs 40C!T R14C Gain-of-function (stretch) — Otway et al.32
KCNE2 IKs 79C!T R27C Gain-of-function Chinese Yang et al.33
KCNJ2 (Kir2.1) IK1 277G!A V93I Gain-of-function Chinese Xia et al.34

Downloaded from https://academic.oup.com/europace/article-abstract/10/10/1133/469096


by guest
on 03 February 2018
Role of potassium currents in cardiac arrhythmias 1137

with the actually measured changes in the current ampli- 11. Wettwer E, Amos GJ, Posival H, Ravens U. Transient outward current in
tude, indicating that AF-induced channel dysregulation human ventricular myocytes of subepicardial and subendocardial origin.
Circ Res 1994;75:473–82.
must have occurred. Although ancillary subunits of Kþ 12. Pogwizd SM, Bers DM. Cellular basis of triggered arrhythmias in heart
channels profoundly affect their expression and biophysi- failure. Trends Cardiovasc Med 2004;14:61–6.
cal properties, little is presently known about AF-induced 13. Cerbai E, Mugelli A. I(f) in non-pacemaker cells: role and pharmacological
remodelling in these regulatory proteins. implications. Pharmacol Res 2006;53:416–23.
The Kþ currents, IKur and IK,Ach, have recently attracted 14. Kirchhof P, Eckardt L, Franz MR, Monnig G, Loh P, Wedekind H et al.
Prolonged atrial action potential durations and polymorphic atrial
special attention because they are confined to atrial tachyarrhythmias in patients with long QT syndrome. J Cardiovasc
myocardium and hence their blockade is not expected to Electrophysiol 2003;14:1027–33.
cause pro-arrhythmic effects in the ventricles. In the past 15. Roberts R. Mechanisms of disease: genetic mechanisms of atrial
decade, several new compounds were developed for the fibrillation. Nat Clin Pract Cardiovasc Med 2006;3:276–82.
prolongation of effective refractory period by the block of 16. Olson TM, Alekseev AE, Liu XK, Park S, Zingman LV, Bienengraeber M et al.
Kv1.5 channelopathy due to KCNA5 loss-of-function mutation causes
IKur, however, the efficacy of most of these new drugs human atrial fibrillation. Hum Mol Genet 2006;15:2185–91.
in converting AF back to SR has been rather disappointing, 17. Gudbjartsson DF, Arnar DO, Helgadottir A, Gretarsdottir S, Holm H,
possibly due to low IKur amplitude in AF. With regard to Sigurdsson A et al. Variants conferring risk of atrial fibrillation on
IK,ACh channels, we have recently reported that these chromosome 4q25. Nature 2007;448:353–7.
channels develop constitutive activity during human AF, i.e. 18. Sinner MF, Pfeufer A, Akyol M, Beckmann BM, Hinterseer M, Wacker A
et al. The non-synonymous coding IKr-channel variant KCNH2-K897T is
these channels become activated, despite the absence of
associated with atrial fibrillation: results from a systematic candidate
stimulating acetylcholine.25 Constitutively active IK,ACh can gene-based analysis of KCNH2 (HERG). Eur Heart J 2008;29:907–14.
hyperpolarize the membrane and hence contribute to persist- 19. Satoh T, Zipes DP. Cesium-induced atrial tachycardia degenerating
ence of AF by stabilization of rotors. Therefore, selectively into atrial fibrillation in dogs: atrial torsades de pointes? J Cardiovasc
targeting constitutively active IK,ACh channels only may pre- Electrophysiol 1998;9:970–5.
20. Nattel S. New ideas about atrial fibrillation 50 years on. Nature 2002;
serve physiological stimulation by vagal nerves and could
415:219–26.
serve as a promising remodelling-related drug target. 21. Goette A, Lendeckel U, Klein HU. Signal transduction systems and atrial
fibrillation. Cardiovasc Res 2002;54:247–58.
22. Dobrev D, Ravens U. Remodeling of cardiomyocyte ion channels in human
Acknowledgements atrial fibrillation. Basic Res Cardiol 2003;98:137–48.
The authors gratefully acknowledge helpful comments by Dobromir 23. Pandit SV, Berenfeld O, Anumonwo JM, Zaritski RM, Kneller J, Nattel S
et al. Ionic determinants of functional reentry in a 2-D model of
Dobrev and Niels Voigt.
human atrial cells during simulated chronic atrial fibrillation. Biophys
J 2005;88:3806–21.
Conflict of interest: none declared. 24. Pacifico A, Henry PD. Class I or class III agents for atrial fibrillation: are
asking the right question? Pacing Clin Electrophysiol 2003;26:1613–9.
25. Dobrev D, Friedrich A, Voigt N, Jost N, Wettwer E, Christ T et al. The G
Funding protein-gated potassium current I(K,ACh) is constitutively active in
patients with chronic atrial fibrillation. Circulation 2005;112:3697–706.
This work was supported by a grant from the Fondation 26. Brundel BJ, van Gelder IC, Henning RH, Tuinenburg AE, Wietses M,
Leducq and by a European Union grant (NORMACOR, Grandjean JG et al. Alterations in potassium channel gene expression
LSHM-CT-2006-018676). in atria of patients with persistent and paroxysmal atrial fibrillation:
differential regulation of protein and mRNA levels for Kþ channels.
J Am Coll Cardiol 2001;37:926–32.
References 27. Dobrev D, Graf E, Wettwer E, Himmel HM, Hala O, Doerfel C et al.
Molecular basis of downregulation of G-protein-coupled inward rectifying
1. Jalife J. Ventricular fibrillation: mechanisms of initiation and maintenance.
Kþ current (IK,ACh) in chronic human atrial fibrillation: decrease in GIRK4
Annu Rev Physiol 2000;62:25–50.
mRNA correlates with reduced I(K,ACh) and muscarinic receptor-
2. Yang T, Snyders DJ, Roden DM. Rapid inactivation determines the
mediated shortening of action potentials. Circulation 2001;104:2551–7.
rectification and [Kþ]o dependence of the rapid component of the
28. The Sicilian gambit. A new approach to the classification of anti-
delayed rectifier Kþ current in cardiac cells. Circ Res 1997;80:782–9.
arrhythmic drugs based on their actions on arrhythmogenic mechanisms.
3. Saenen JB, Vrints CJ. Molecular aspects of the congenital and acquired
Task Force of the Working Group on Arrhythmias of the European Society
long QT syndrome: clinical implications. J Mol Cell Cardiol 2008;44:
633–46. of Cardiology. Circulation 1991;84:1831–51.
4. Priori SG, Aliot E, Blomstrom-Lundqvist C, Bossaert L, Breithardt G, 29. Hong K, Bjerregaard P, Gussak I, Brugada R. Short QT syndrome and atrial
Brugada P et al. Task force on sudden cardiac death of the European fibrillation caused by mutation in KCNH2. J Cardiovasc Electrophysiol
Society of Cardiology. Eur Heart J 2001;22:1374–450. 2005;16:394–6.
5. Sanguinetti MC, Tristani-Firouzi M. hERG potassium channels and cardiac 30. Chen YH, Xu SJ, Bendahhou S, Wang XL, Wang Y, Xu WY et al. KCNQ1
arrhythmia. Nature 2006;440:463–9. gain-of-function mutation in familial atrial fibrillation. Science 2003;
6. Christ T, Boknik P, Wohrl S, Wettwer E, Graf EM, Bosch RF et al. L-type 299:251–4.
Ca2þ current downregulation in chronic human atrial fibrillation is 31. Hong K, Piper DR, Diaz-Valdecantos A, Brugada J, Oliva A, Burashnikov E
associated with increased activity of protein phosphatases. Circulation et al. De novo KCNQ1 mutation responsible for atrial fibrillation and short
2004;110:2651–7. QT syndrome in utero. Cardiovasc Res 2005;68:433–40.
7. Roden DM. Taking the ‘idio’ out of ‘idiosyncratic’: predicting torsades de 32. Otway R, Vandenberg JI, Guo G, Varghese A, Castro ML, Liu J et al.
pointes. Pacing Clin Electrophysiol 1998;21:1029–34. Stretch-sensitive KCNQ1 mutation A link between genetic and environ-
8. Sesti F, Abbott GW, Wei J, Murray KT, Saksena S, Schwartz PJ et al. mental factors in the pathogenesis of atrial fibrillation? J Am Coll
A common polymorphism associated with antibiotic-induced cardiac Cardiol 2007;49:578–86.
arrhythmia. Proc Natl Acad Sci U S A 2000;97:10613–8. 33. Yang Y, Xia M, Jin Q, Bendahhou S, Shi J, Chen Y et al. Identification
9. Nass RD, Aiba T, Tomaselli GF, Akar FG. Mechanisms of disease: ion of a KCNE2 gain-of-function mutation in patients with familial atrial
channel remodeling in the failing ventricle. Nat Clin Pract Cardiovasc fibrillation. Am J Hum Genet 2004;75:899–905.
Med 2008;5:196–207. 34. Xia M, Jin Q, Bendahhou S, He Y, Larroque MM, Chen Y et al. A Kir2.1
10. Nabauer M, Kaab S. Potassium channel down-regulation in heart failure. gain-of-function mutation underlies familial atrial fibrillation. Biochem
Cardiovasc Res 1998;37:324–34. Biophys Res Commun 2005;332:1012–9.

Downloaded from https://academic.oup.com/europace/article-abstract/10/10/1133/469096


by guest
on 03 February 2018

You might also like