You are on page 1of 13

Journal of the American College of Cardiology Vol. 47, No.

1, 2006
© 2006 by the American College of Cardiology Foundation ISSN 0735-1097/06/$32.00
Published by Elsevier Inc. doi:10.1016/j.jacc.2005.08.059

CARDIOVASCULAR GENOMIC MEDICINE

Genomics and Cardiac Arrhythmias


Robert Roberts, MD, FACC
Ottawa, Ontario, Canada
Sudden cardiac death in patients younger than 35 years of age is primarily due to genetic
causes. Familial hypertrophic cardiomyopathy accounting for 30% to 40% is associated with
structural heart disease while the Brugada syndrome and the long QT syndrome (LQTS) are
associated with normal cardiac function. This is a review of the genetics of supraventricular
and ventricular arrhythmias. Atrial fibrillation is mapped to nine chromosomal loci and four
genes are identified. Adenosine monophosphate-activated protein kinase is one gene
responsible for Wolff-Parkinson-White syndrome. The LQTS and the Brugada syndromes
are due to defects primarily in cardiac sodium and potassium ion channels. The role of single
nucleotide polymorphisms in predisposing to arrhythmias in acquired disorders such as
hypertrophy is discussed. (J Am Coll Cardiol 2006;47:9 –21) © 2006 by the American
College of Cardiology Foundation

The past decade has been golden for cardiovascular diseases offspring. A major project referred to as the HapMap
with the elucidation of several genes responsible for familial project is ongoing to map the chromosomal location of
cardiomyopathies and the ion channelopathies. However, these SNPs and generate haplotypes involving multiple
these diseases, referred to as single gene disorders, were SNPs that tend to be inherited as blocks or units (7). The
amenable to chromosomal mapping of the gene locus effect of SNPs on a particular phenotype may be through
through conventional genetic linkage analysis. In these haplotypes containing several SNPs rather than a single
disorders, a single gene predominates in the expression of SNP. To identify these SNPs or haplotypes on a genome-
the phenotype. However, even with single gene disorders, wide basis for a particular phenotype such as atrial fibrilla-
there are modifier genes, which have a significant influence tion would require hundreds of thousands of DNA markers,
on the phenotype but would not be detected by this which, until recently, would be prohibited (6). Chips are
conventional technique. There is suggestive evidence that now available with hundreds of thousands of SNPs such as
arrhythmias such as atrial fibrillation occurring in associa- the Affymetrix 500,000 chip (Affymetrix, Santa Clara,
tion with acquired structural heart disease are more likely in California) to perform these studies. Secondly, such high-
those individuals with genetic predisposition (1,2). This is density DNA markers make it possible to utilize mathemat-
particularly pertinent in polygenic diseases such as coronary ical models to identify associations with polymorphisms that
artery disease and hypertension (3,4). Locating and identi- have minimal effect on the phenotype (5,6,8). It is now
fying modifier genes and genes that predispose but do not possible to genotype 100 DNA samples for 500,000 geno-
per se induce the phenotype has not been feasible due to types over a period of a few hours. This has ushered in a new
lack of appropriate statistical models and high throughput era for detecting genetic variants that affect human disease.
genotyping technology. While both of these problems are
still with us, there are currently methods making it possible
and practical to pursue such research goals. GENOMIC IMPLICATIONS FOR DIAGNOSIS AND
The sequencing of the human genome has brought with TREATMENT OF SUDDEN CARDIAC DEATH (SCD)
it a wealth of information as well as enabling the develop-
ment of high throughput technology. It is now recognized Sudden cardiac death remains a worldwide scourge. There
that 99.9% of the three billion bases that comprise the are considerable data to indicate the proximate cause of
human genome are identical (5,6). Thus, about three SCD is electrical due to cardiac conduction defects or
million bases account for all of the differences between dysrhythmias. In the U.S. over 400,000 and in Canada over
human beings including one’s resistance or vulnerability to 50,000 die annually from SCD (9). It is of note that this
disease. The current and future challenge is to identify these number has not changed for the past three decades. Thus,
differences referred to as single nucleotide polymorphisms treatment for SCD has not been as progressive as in other
(SNPs). Currently, it appears there are over 15 million forms of cardiac disease, indeed one may say it has been
SNPs circulating in the population from which three mil- regressive. A major landmark study evaluating drugs in the
lion can be selected to be passed on by each parent to their treatment of arrhythmias was the international clinical trial
referred to as Cardiac Arrhythmia Suppression Trial
(CAST) (10). The major and unexpected finding was that
From the University of Ottawa Heart Institute, Ottawa, Ontario, Canada.
Manuscript received July 20, 2005; revised manuscript received August 4, 2005, most of the drugs used in the trial were shown to be
accepted August 17, 2005. pro-arrhythmic. Quinidine, a drug used for decades in the
10 Roberts JACC Vol. 47, No. 1, 2006
Genomics and Cardiac Arrhythmias January 3, 2006:9–21

structural or electrical cardiac disease. The person without


Abbreviations and Acronyms structural heart disease (Brugada syndrome) is perhaps at a
AMPK⫽ adenosine monophosphate-activated protein greater disadvantage because they are more likely to be
kinase younger and less likely to have a reason to visit the
AP ⫽ action potential physician.
HCM ⫽ hypertrophic cardiomyopathy
ICD ⫽ implantable cardioverter-defibrillator
LQTS ⫽ long QT syndrome THE FREQUENCY AND PREVALENCE
SCD ⫽ sudden cardiac death
SNP ⫽ single nucleotide polymorphism
OF GENETIC-INDUCED ARRHYTHMIAS
SQTS ⫽ short QT syndrome In considering the role of genetics and arrhythmias, one
WPW ⫽ Wolff-Parkinson-White syndrome
must pause as to the frequency and prevalence of the
genetic-induced arrhythmias. Throughout the world, SCD
occurring in the young adult (⬍35 years) whether associated
treatment of supraventricular and ventricular arrhythmias with structural or nonstructural heart disease is predomi-
was shown to prolong the QT interval and induce ventric- nantly due to genetic defects. Familial hypertrophic cardio-
ular arrhythmias and SCD. Therapy had to be developed myopathy (HCM) with a prevalence of 1 in 500 is the most
empirically without knowing the molecular basis for the common cause of SCD in the young (12). While there are
disease or the molecular mechanism of the drug. Current no population studies on the prevalence of the genotype,
knowledge of ion channels facilitates the development of estimates based on the phenotype, which usually underes-
drugs without known adverse effects such as prolongation of timates the genotype, show it is very common for a genetic
the QT. It is expected that genetic defects, many of which disorder. It is estimated over 12 million individuals in the
involve ion channels, will help to identify molecular mech- world carry the genetic defect for HCM. There are over
anisms for arrhythmias, which are also applicable to ar- 600,000 individuals with this genetic defect in the U.S. and
rhythmias caused by acquired disease. There is some en- 60,000 in Canada. Brugada syndrome, another familial
couragement with the overlap between the mechanism cause of SCD in the young, is the most common cause of
responsible for ventricular tachycardia of familial long QT SCD occurring in individuals without structural heart dis-
syndrome (LQTS) and that induced by quinidine in ac- ease (13). Wolff-Parkinson-White syndrome (WPW) is the
quired disorders. second most common cause of supraventricular arrhythmias
Young individuals who die with monogenic disorders in the Western world and the most common cause in China
such as Brugada syndrome or prolonged QT syndrome are (14). The role of WPW in SCD remains controversial. In
usually asymptomatic, and SCD is their first and often last those studies in which the cardiac conduction system was
symptom. In the future, when appropriate therapy becomes evaluated, WPW was a major cause of SCD, particularly in
available to prevent SCD in the asymptomatic individuals, infants (15). Wolff-Parkinson-White syndrome, while al-
genetic screening will be a prerequisite (e.g., Brugada ways a genetic disease, appears to be mostly sporadic rather
syndrome, LQTS, and so on). Gene testing on the basis of than familial. In a recent study, 30% of patients with atrial
family history or an incidental electrocardiogram (ECG) fibrillation with or without structural heart disease had a
finding (e.g., insurance) followed by appropriate therapy is family history of atrial fibrillation (16). Offspring with even
likely to be a future routine to prevent SCD. Most of SCD a single parent having atrial fibrillation will have 85%
over the age of 35 years occurs in association with acquired increased risk of developing atrial fibrillation (16). For
structural heart disease and symptoms. There is, however, single gene disorders, many genes with their corresponding
the concern of who is vulnerable for SCD. For example, in mutations have been identified, and the appropriate tech-
heart failure 50% die of SCD due to arrhythmias (11). The nology could be assembled soon to screen populations at risk
only effective treatment is an indwelling defibrillator. The of having these genetic defects.
dilemma is identifying which half is vulnerable. One hope is
to detect SNPs that predispose to SCD. Identifying molec-
ular defects responsible for arrhythmias should lead to novel
THE ROLE OF GENETICS IN ACQUIRED ARRHYTHMIAS
therapies specifically designed to target the abnormality. In the middle-aged and elderly population, which accounts
Identification of the defect in the cholesterol receptor of for most of SCD, the cause is almost always associated with
familial hypercholesterolemia leads to the development of structural heart disease predominantly due to coronary
drugs (statins) to inhibit cholesterol synthesis. These drugs artery disease. What is the role of genetic defects in these
are now the mainstay of treatment to prevent coronary individuals? While no gene or polymorphism has yet been
artery disease in the general population. Pharmacogenetics definitely identified to induce arrhythmias, there is increas-
and pharmacogenomics will assist in selecting the drug and ing evidence that genetic predisposition contributes signif-
determining the dose on an individual basis to minimize icantly to acquired arrhythmias (17). For example, a recent
adverse side effects. Determining who is at risk for arrhyth- finding shows that atrial fibrillation occurring with struc-
mias and SCD is a problem common to those with tural and nonstructural heart disease is associated with a
JACC Vol. 47, No. 1, 2006 Roberts 11
January 3, 2006:9–21 Genomics and Cardiac Arrhythmias

family history of atrial fibrillation in 30% of patients (16). Screening for polymorphisms in polygenic disorders deter-
Much of susceptibility to acquired disease is thought to be mines whether there is a risk of increased susceptibility for
encoded primarily by SNPs that span the human genome. arrhythmias. Other critical features must be considered as
Even in single gene disorders, there are modifying genes we move into the genetic revolution and apply this infor-
(SNPs), which modulate the phenotype resulting from the mation to the care of patients. Despite having the genotype
interaction of the environment with the genotype. A third at birth, several factors modulate whether the phenotype
group of genes modulating arrhythmias in acquired and will ever develop and to what extent. First, is the feature of
inherited diseases, particularly coronary artery disease or penetrance which the risk of developing the phenotype in an
heart failure, are those whose expression is reprogrammed individual with the genetic defect. In the case of single gene
by the underlying disease. An excellent and well- disorders, those that are autosomal dominant have a high
characterized example is cardiac compensatory hypertrophy penetrance of probably 70% to 80% but seldom complete.
in response to pressure overload as occurs in hypertension, Thus, a percentage of individuals despite having the defec-
valvular disease, or heart failure. In pathological hypertro- tive gene do not develop the disease. The other modifier is
phy as opposed to physiological hypertrophy, expression of referred to as expressivity defined as the variability in
a host of genes is up-regulated as part of the so-called fetal phenotypic manifestations that occur across a population
program (18). The extent of hypertrophy may, in part, despite being due to the same genetic defect. The onset of
depend on the presence or absence of certain alleles. For the phenotype is often age-dependent. The severity of the
example, regardless of the cause, if the patient is homozy- phenotype and whether it develops are also influenced by
gous for the DD allele of the angiotensin-converting en- environmental factors such as exercise, temperature, gender,
zyme gene as apposed to the I-form, hypertrophy will be and other factors. Nevertheless, solutions to these problems
more extensive and the propensity for SCD greatly in- will be facilitated with routine genetic screening and in-
creased (19). Hypertrophy that deteriorates into dilated creased experience. Present knowledge clearly indicates that
cardiomyopathy induces the expression of other genes, a multidisciplinary approach is required in the future to
which could alter the predisposition to atrial or ventricular identify these genetic abnormalities and utilize them for
arrhythmias and SCD. All of the association studies sug- treatment and prevention. It does require careful clinical
gesting genetic predisposition remain suspect because of the characterization, genetic expertise, functional analysis, and
small sample size (6), but the technology is now available to genetic counseling along with drug discovery to significantly
pursue studies with adequate sample size. The availability of impact this worldwide scourge.
genome-wide scans with 500,000 SNPs properly selected is
likely to add a new dimension to our search for genetic
GENETICS OF SUPRAVENTICULAR ARRHYTHMIAS
predisposition and modifier genes.
Atrial fibrillation. Atrial fibrillation is the most common
sustained cardiac arrhythmia. It occurs in about 6% of the
GENETIC SCREENING FOR PREDICTION
population over the age of 60 years. It is estimated there are
AND PREVENTION OF ARRYTHMIAS
over 3 million people with persistent atrial fibrillation in the
Genes themselves do not engage in the work of the cell, but U.S. (20), which will increase to 5.6 million by 2050 (21).
rather their end product, the protein, performs the function At age 40 years and over, the chance of developing atrial
of the organism. In single gene disorders of adults, the most fibrillation in men and women is about 25% (22). It is
common mutation is a missense. This is a genetic defect in associated with significant morbidity and mortality, ac-
which a single base has been substituted for another counting for over one-third of all strokes over the age of 60
resulting in the encoding of a different amino acid, which, if years (23). In a recent Framingham Heart study of 2,243
occurring in an important domain of the protein, will be participants, 681 had at least one parent with atrial fibril-
associated with a gain- or loss-of-function. Other less lation (16). This indicates 30% of all patients with atrial
common defects would be a loss or gain of a base resulting fibrillation, with or without structural heart disease, have a
in a protein with many amino acids altered or the coding family history of the disease (16). The relative risk of atrial
may be changed to a start or stop codon resulting in a fibrillation was increased 85% in individuals with at least
truncated protein. Genetic defects occurring in non-protein one parent with a history of atrial fibrillation (16). We
coding regions will be manifested primarily as increased or identified the first locus for familial atrial fibrillation on
decreased expression of the protein. Thus, the major func- chromosome 10 (10q22), confirming the inheritance of this
tional change in the protein is loss- or gain-of-function due disease (24). Currently, seven chromosomal loci have been
to an alteration in the amino acid composition or the mapped and four of the genes identified. The genetic
expression of the protein. While genetic screening is pivotal features of each of these loci are summarized in Table 1, and
in the prevention of genetic disease, it unfortunately is not a brief discussion will follow of each of the genes.
the only variable that must be considered. Genetic screening KCNQ1 gene. KCNQ1, the first gene, was identified (25)
determines the presence or absence of the genetic defect and in a four-generation Chinese family showing autosomal
thus whether there is a risk of developing the disease. dominant inheritance. The loci was mapped to chromosome
12 Roberts JACC Vol. 47, No. 1, 2006
Genomics and Cardiac Arrhythmias January 3, 2006:9–21

Table 1. Genetic Loci Responsible for Atrial Fibrillation


Mode of Effect on
Locus Gene Inheritance Mutation Function Physiological Effect
11p15.5 KCNQ1 AD S140G Gain of function 2 AAP 2 ARP
21q22 KCNE2 AD R27C Gain of function 2 AAD 2 ARP
17q KCNJ2 AD V931 Gain of function 2 AAD 2 ARP
7q35–36 KCNH2 AD N588K Gain of function 2 AAP 2 ARP
5p13 Unknown Recessive Unknown Unknown Unknown
6q14–16 Unknown AD Unknown Unknown Unknown
10q22 Unknown AD Unknown Unknown Unknown
AAP ⫽ atrial action potential duration; AD ⫽ autosomal dominant; ARP ⫽ atrial effective refractory period.

11p15.5 and the gene, KCNQ1, encodes for the alpha KCNH2 gene exhibited a missense mutation at nucleotide
subunit of a cardiac potassium channel. The mutation 17645 with C going to G resulting in the substitution of
consisted of substitution of adenine to guanine at nucleotide asparagine for lysine at residue 588 (N588K). All affected
418 resulting in an amino acid substitution of serine for members upon electrophysiological testing showed a short-
glycine at residue 140 (S140g). Penetrance was close to ening of the atrial and ventricular refractory periods with
100%. Expression of the wild type and mutant KCNQ1 inducibility of atrial and ventricular fibrillation. The muta-
gene in COS cells showed increased potassium current tion confers a gain-of-function with a shortening of the
density. Thus, the mutation induced a gain-of-function, atrial AP duration and the effective atrial refractory period.
which shortens the atrial action potential (AP) duration and Genetic defects provide insight into the mechanism for
the effective atrial refractory period. Several investigators atrial fibrillation. There are several consistent features of
with multiple families had shown KCNQ1 is not responsi- the genetics of atrial fibrillation:
ble for atrial fibrillation in their families (26), suggesting this
1) Genes identified all encode for subunits of potassium
is an uncommon cause for atrial fibrillation.
channels
KCNE2 gene. A second locus was identified by the same
2) All mutations confer a gain-of-function
Chinese investigators (27) on 21q22 encoding for another
3) All of the mutations shorten the atrial AP duration and
potassium channel subunit (KCNE2). The mutation was
the atrial effective refractory period
shown to involve a C to T transition at nucleotide 79 of the
gene for KCNE2. This resulted in an arginine to cysteine at Atrial fibrillation is thus a channelopathy whereby the
residue 27 (r27C), which is present in all affected individuals genetic defects provide an ideal substrate for re-entry
but not in healthy individuals. Mode of transmission was arrhythmias. The consistency of the mechanism whereby
again, autosomal dominant. Functional studies, again, genetic defects induce atrial fibrillation should provide
showed that the mutant shortened the atrial AP duration specific targets for the development of novel drug therapy.
and the atrial effective refractory period. The penetrance
appeared to be incomplete although difficult to assess from
the limited number of studies performed. POLYMORPHISMS THAT
KCNJ2 gene. A third gene was identified in 30 unrelated PREDISPOSE TO ATRIAL FIBRILLATION IN
Chinese kindred of atrial fibrillation showing a missense
INDIVIDUALS WITH STRUCTURAL HEART DISEASE
mutation in the potassium channel KCNJ2 (28). KCNJ2
encodes for the Kir 2.1 channel, which mediates potassium Atrial fibrillation occurs mostly in association with acquired
current in the heart. It is an inward rectifier, which induces diseases such as coronary heart disease, value disease, or
early repolarization. The gene exhibited a missense muta- heart failure. In a recent study (1) involving 250 patients
tion with G to A at nucleotide 277 corresponding to a valine with structural heart disease and atrial fibrillation, 250
to isoleucine at residue 93 (V93I) of the protein. Functional controls expression of polymorphisms N235T, G-6A,
analysis again showed that this mutant gene shortens the G-217A of the angiotensin-converting enzyme gene were
atrial AP duration and the atrial refractory period. Overex- significantly increased in patients with atrial fibrillation over
pression of the wild type Kir 2.1 in the mouse induced atrial that of controls with a p value of 0.01, 0.005, and 0.002,
fibrillation (29). Acquired atrial fibrillation in humans is respectively. The odds ratio for atrial fibrillation was 2.5
associated with increased expression of the Kir 2.1 channel with M235/M235 ⫹ M235/T235 genotype, 3.3 for the
(30 –33). The investigators also analyzed KCNJ2 in 154 G6/G6 genotype, and 2.0 for the G217/G217 genotype.
other patients with lone atrial fibrillation, but no mutation Similar associations have been made with polymorphisms in
was identified (28). the connexin 40 gene. This gene encodes a cardiac gap
KCNH2 gene. Recently, Hong et al. (34) identified a junction protein, which mediates electrocoupling of cardio-
family with short QT syndrome (SQTS) in whom three myocytes (2). The polymorphisms in the promoter region
members presented with atrial fibrillation. The proband of upstream of the gene, which are ⫺44A and ⫺44AA
17 years of age presented with paroxysmal atrial fibrillation. genotypes, were more frequent in subjects with atrial fibril-
JACC Vol. 47, No. 1, 2006 Roberts 13
January 3, 2006:9–21 Genomics and Cardiac Arrhythmias

lation exhibiting a risk ratio of 5.3 and 6.2, respectively, for subunit has a binding site for AMP, which is essential for
atrial fibrillation. In another study (35), 108 patients with enzymatic activity. The mutation consists of a substitution
acquired atrial fibrillation compared to controls showed the of glutamine for arginine at residue 302. Six mutations have
K38G alleles of connexin 40 exhibited an odds risk ratio for since been described in several families all of which are
atrial fibrillation of 1.8 in patients with more than one missense mutations except for one, which is an insertion
K38G allele. These studies suggest that polymorphisms (38). The penetrance based on the few families so far
predispose to atrial fibrillation when occurring in association studied is in the range of 70% to 80%. The phenotype
with these diseases. It is important to recognize that these consists of pre-excitation on the ECG, varying degrees of
studies are case control association studies, and it is inade- conduction defects, frequently cardiac hypertrophy, and
quate to make definitive conclusions. The sample size is too excessive glycogen storage in the myocytes without sarco-
few and would require confirmation of the larger sample size mere or myocyte disarray (39). The most common arrhyth-
and also better matching of controls with affected individ- mia associated with WPW is that of paroxysmal supraven-
uals. However, the availability recently of genome-wide tricular tachycardia with a narrow QRS. In the remaining
scans with hundreds of thousands of SNPs will, in the 20%, the arrhythmia is generally atrial fibrillation. In this
future, make it possible to perform genomic studies to syndrome, however, the incidence of atrial fibrillation is
identify SNPs that predispose to diseases such as atrial much higher in the range of 40% to 50%; AMPK is a sensor
fibrillation. This is expected to be a major onslaught of of the body’s ATP level and is activated by an increase in the
genomic technology in the next few years. ratio of AMP to ATP (40,41). The availability of ATP is
Paroxysmal supraventricular tachycardia. Wolff-Parkinson- increased by AMPK through increasing glucose absorption,
White syndrome is the most common form of paroxysmal inhibiting glycogen synthesis, increasing fatty acid oxida-
supraventricular tachycardia in China and the second most tion, and decreasing fatty acid synthesis. Transgenic mice
common cause in the Western world (14,36). The incidence expressing either the R302q mutation (39) or the N4881
of WPW is about 1 in 3,000 of the population. This mutation (42) exhibit a typical phenotype as observed in
syndrome, named after the individuals described in 1930, humans for this disease. The mechanism whereby AMPK
has remained as a paradigm of a model for re-entry leads to accessory pathways or alters conduction remains to
arrhythmias. In this disease, there is an accessory bundle of be determined. In one study, it has been shown that R302Q
muscle tissue, which connects the atrium to the ventricle mutation inhibits the cardiac sodium inward current (41).
bypassing the atrioventricular node. Conduction through Utilizing these transgenic models, studies are now being
the accessory bundle is usually retrograde but under unusual performed to determine alternate forms of therapy. It also
circumstances may be antegrade at very rapid rates of 150 serves as a model to elucidate AMPK’s role in the develop-
beats/min. Rapid heart rates, whether retrograde or ante- ment of the heart and, in particular, that of the specialized
grade, if sustained lead to heart failure or even ventricular cardiac conduction system. The accepted form of treatment
tachycardia and SCD. The most common form of tachy- currently in patients with familial WPW is ablation of the
cardia associated with WPW is paroxysmal supraventricular accessory bundle. Therapy to prevent the excessive glycogen
tachycardia with rates of 120 to 160 beats/min. The role of storage may require replacement of AMPK enzyme.
WPW in SCD is difficult to assess. Autopsies performed to
assess cause of death do not examine the specialized con-
GENETICS OF VENTRICULAR ARRHYTHMIAS
duction system of the heart and thus would not detect
WPW. In studies in which the conduction system was Brugada syndrome. There remains a large group of indi-
evaluated, WPW was found to account for 33% of patients viduals who develop idiopathic ventricular fibrillation unas-
who died suddenly without structural heart disease (15). sociated with any structural heart disease. In 1995, a disease
The prevalence of familial WPW appears to be low with now referred to as Brugada syndrome (43) was described,
most due to a sporadic genetic defect; WPW is a genetic which is associated with SCD at a very young age and is
disease in that the formation of an accessory conducting responsible for a significant proportion of sudden infant
bundle or lack of remodeling of an existing bundle requires death syndrome. Brugada syndrome is characterized by a
genetic regulation. It is perhaps analogous to cancer, which history of sudden death and a typical ECG pattern of
is an obligatory genetic disease, but the familial form ST-segment elevation in leads V1 to V3 (Fig. 1) (13). It is
appears less common. claimed to be responsible for up to 12% of all sudden deaths
In 2001 we identified a family with WPW and showed and approximately 20% of deaths occurring in patients with
(37) the gene responsible for the disease in this family structurally normal hearts. In the Western world, the
encodes for the gamma 2 subunit of the enzyme adenosine prevalence of this disease appears to be in the order of 1 in
monophosphate-activated protein kinase (AMPK). The 5,000 individuals. In Southeast Asia, the disease is more
gene is referred to as PRKAG2 located on chromosome prevalent being a leading cause of death in males under the
7q36 and is inherited in an autosomal dominant pattern; age of 40, second only to car accidents (13). This disease
AMPK consists of three subunits, alpha, beta, and gamma, exhibits an autosomal dominant pattern of inheritance
with each subunit encoded by a distinct gene. The gamma indicating it affects men and women equally, and 50% of the
14 Roberts JACC Vol. 47, No. 1, 2006
Genomics and Cardiac Arrhythmias January 3, 2006:9–21

Figure 1. Variation in the precordial lead ST and T waves in a patient with Brugada syndrome. Reprinted with permission from Fuster V, Alexander RW,
O’Rourke RA. Hurst’s The Heart. 11th edition. McGraw-Hill Companies, 2004.

offspring will inherit the gene and be at risk of developing the splicing donor site of intron 7. Recently, the first
arrhythmias and SCD. This also means only one copy of the intronic mutation in SCN5A was described (46), which
gene is defective, and it acts as a dominant positive or involved a four base pair insertion in exon 27. This led to a
negative. All of the mutations identified have been in a truncated protein and a non-functional channel. Brugada
subunit of the sodium cardiac channel (SCN5A). syndrome and LQTS can both be caused by defects in the
The subunit referred to as SCN5A has a genomic SCN5A but are opposite in function, namely, the former is
structure (44) consisting of 28 exons spanning approxi- due to loss of function leading to an accelerated inactivation
mately 80 kb on chromosome 3p21 with a dinucleotide of the sodium channel resulting in more rapid depolariza-
repeat polymorphism in entron 16. The gene encodes for a tion (46). Patients with Brugada syndrome on becoming
protein with 2,016 amino acids. Sodium channels, like most febrile are more likely to develop arrhythmias and SCD
ion channels, are part of a complex. The pore forming alpha (13). It is evident there are many more mutations and other
subunit is encoded by SCN5A, and up to three beta
genes yet to be discovered considering that over 75% of
subunits have been described, as well as other attachments
cases are not due to known mutations. A second locus has
to the cytoskeleton such as caveolas (45). The mutations
been mapped to chromosome 3 (13) indicating at least
identified responsible for the Brugada syndrome have been
another gene; however, that gene has not been identified.
primarily missense mutations. More than 60 mutations have
Genetic screening, currently not available, will be essential
been described in the SCN5A of which all have been
localized to the protein coding sequences. Screening for for the management of this disease because most of these
these mutations show they account for only about 25% of people do not have symptoms. Even when someone is
patients with Brugada syndrome. This indicates there may identified to have a history of this disease, it will be
be other mutations in the SCN5A gene and most likely necessary to determine which 50% of the offspring have the
mutations in multiple other genes. Most of the genes mutation and are vulnerable to arrhythmias and SCD.
responsible for the ion channels of the cardiac AP have been Genetic screening is necessary for appropriate genetic coun-
cloned and are available as good candidates to screen for seling as well as to implement preventive therapy when it
these mutations. The mutations identified in SCN5A have becomes available. In the meantime, it is important to
been associated with a loss of current either by creating a recognize that most drugs are not effective in this syndrome.
truncated protein or by increasing sodium channel inactiva- An implantable cardioverter-defibrillator (ICD) is essen-
tion (13). One of the mutations, an insertion, occurred in tially the only recommended form of therapy.
JACC Vol. 47, No. 1, 2006 Roberts 15
January 3, 2006:9–21 Genomics and Cardiac Arrhythmias

Table 2. Genetic Loci Responsible for Long QT Syndrome


Arrhythmia Chromosome Gene Frequency
Autosomal Dominant Loci
LQT1 Ilp15 KCNQI ⫺50%
LQT2 7q35 KCNH2 30%–40%
LQT3 3p21 SCN5A 5%–10%
LQT4 4q25 ANK2 Rare
LQT5 21q22 KCNEI Rare
LQT6 21q22 KCNE2 Rare
LQT7 17 KCNJ2 Rare

Recessive (and Deafness)


LQT Ilp15 KCNQI Rare
LQT 21q22 KCNQ2 Rare

LQTS the cell with most of the activity occurring during the
upstroke of the AP. An increased amount of current active
Clinical features. The congenital LQTS is an inherited
during the plateau phase would prolong depolarization and
disease characterized by prolongation of ventricular repolar-
lengthen the AP. Potassium channels when active tend to
ization, which is manifested by prolongation of the QT
restore the internal negativity of the cell and shorten the
interval on the ECG (47). It is associated with a high risk of
AP. If potassium currents predominate during the plateau,
sudden death from ventricular arrhythmias. There are two
repolarization will occur shortening the AP as opposed to
forms of the disease, one autosomal dominant and the other
sodium currents if they predominate during plateau, it
recessive (48). The autosomal dominant form is far more
prolongs the AP. In LQTS due to defects in a potassium
common, and the rare recessive form is associated with
deafness. Clinically, the syndrome is characterized by syn- channel, there is a loss-of-function, whereas in LQTS due
copal episodes, malignant ventricular tachycardia, and fibril- to defects in sodium channel gene, there is a gain-of-
lation. The majority of patients with LQTS are asympto- function. Both are due to autosomal dominant patterns of
matic and are either discovered incidentally based on ECG, inheritance indicating only one copy of the mutated gene is
family history, or having survived an episode of syncope or necessary to produce the clinical syndrome.
severe ventricular arrhythmias. The prognosis of untreated The pathogenesis of LQTS due to defects in the ion
patients is considered to be quite poor. It is claimed that channels may be summarized as follows: mutations in the
about one-fifth of untreated patients presenting with syn- potassium channel lead to inadequate opening and decreased
cope die within 1 year and 50% within 10 years (13). Long outward current; mutations in the sodium channels lead to
QT syndrome can also be acquired due to a long list of inadequate closing of the channels and excessive sodium
drugs, primarily antiarrhythmics, antidepressants, and phe- inward current. This results in an imbalance with an inade-
nothiazides. In addition, electrolyte imbalance such as quate maintenance of the electrical gradient reflected in pro-
hypokalemia, hypomagnesemia, and hypocalcemia, can also longation of the AP reflected by long QT interval on the
cause LQTS. These drugs and electrolyte abnormalities play ECG.
a major triggering role in ventricular arrhythmias and SCD Genotype-phenotype correlations. The role of genetics
in patients with the inherited LQTS. with respect to prognosis and therapy for LQTS remains to
Pathogenesis of familial LQTS. Long QT syndrome is be defined. Studies relating a single mutation to survival or
now known to be due to nine different genes (Table 2). Of the to clinical events as well as therapy have not shown a
nine genes identified, seven encode potassium channel sub- clear-cut stratification (50). This may, in part, relate to lack
units, one, a sodium channel, and, most recently, a mutation of adequate sample size and the genetic background on
encoding for ankyrin B, a scaffolding protein. The latter is the which that particular mutation is interacting. Attempts,
only non-channel disease gene thus far described (49). however, to relate a particular gene to survival and how it
In LQTS, prolongation of the AP (Fig. 2) may be due to interacts with gender and the duration of the QT interval
an increase in the phase of depolarization or a decrease in have been performed (48). In the study by Priori et al. (48),
the current that induces repolarization. The molecular investigators assess the risk of the first cardiac event before
defect is determined by the specific genotype, but the final the age of 40 years and before initiating therapy in 580
common pathway is prolongation of the AP and decreased patients. The study involved 355 patients with a mutation in
repolarization reserve (47). This results in diminished ability the KCNQ1 gene, 176 patients with a mutation in the
of the ventricular myocyte to respond to stress such as KCNH2 gene, and 49 individuals with a mutation in the
hypoglycemia, hypomagnesemia, and drugs that prolonged SCN5A gene. These represent the most common causes of
the AP. The sodium and potassium channels share opposite the familial LQTS. Results, as illustrated in Figures 3A, 3B,
affects on the AP duration. The sodium channel depolarizes and 3C, indicate the cumulative event-free survival was
16 Roberts JACC Vol. 47, No. 1, 2006
Genomics and Cardiac Arrhythmias January 3, 2006:9–21

Figure 2. Schematic of inward and outward ionic currents, pumps, and exchangers that underlie atrial and ventricular action potentials in the mammalian
heart. Control and failing (bold line) action potential profiles are shown on top. Each phase of the action potential is labeled. Under the action potential,
a schematic of the time course of each current is shown, and the gene product (probable clone) that underlies the current is indicated. Reprinted with
permission from Fuster V, Alexander RW, O’Rourke RA. Hurst’s The Heart. 11th edition. McGraw-Hill Companies, 2004.

quite different among the three groups with the highest illustrate an interaction of the particular gene with gender and
event rate occurring in KCNH2 and SCN5A. Analysis for the duration of the QT interval.
gender within each subgroup showed that gender had no
influence on patients with a mutation in the KCNH2 gene,
whereas female patients with a mutation in the KCNH2 GENETIC IMPLICATIONS FOR THE
gene had a higher risk than male patients, and there was a THERAPEUTIC MANAGEMENT OF LQTS
trend towards a higher risk among patients with a mutation
Despite the large number of mutations (300) responsible for
in the SCN5A. Thus, a first cardiac arrest or sudden death
LQTS, there is still limited correlation with respect to specific
was highest among female patients with a mutation in the
therapy. It is evident that mutations in the KCNH2 or
KCNH2 gene (0.82 per year) and male patients with a
SCN5A genes are associated with more cardiac events. Given
mutation in the SCNA5 gene (0.96 per year). Thus, the role of the risk stratification provided by the three parameters previ-
gender varies according to the genetic locus. Correcting for the ously discussed, the investigators (48,50) recommend that
QT interval showed significant differences among the three prophylactic treatment is warranted in male and female pa-
subgroups. The mean QT was 466 ⫾ 44 ms among patients tients with a mutation in the KCNQ1 gene who have a QT
with a mutation at the KCNQ1 gene, 490 ⫾ 49 ms among above 500 ms or more, male patients with a mutation in the
those with a mutation in the KCNH2 gene, and 496 ⫾ 49 ms KCNH2 gene who have a QT of 500 ms or more, all female
among those with mutations in the SCN5A gene. In each patients with a mutation at the KCNH2 gene irrespective of
subgroup, the QT interval was longer in those who were the QT, and all patients with a mutation in the SCN5A gene.
symptomatic. The number of genetically affected patients with Schwartz et al. (51,52) reported cardiac events more
a normal QT interval was significantly higher in the KCNQ1 frequently occur during exercise (62%) and only rarely
group (36%) than in the KCNH2 group (19%) or the SCN5A during sleep and rest in LQT1 patients. Swimming is also a
group (10%). Analysis of the event-free survival, taking into common trigger in the LQT1 syndrome (52). In contrast, in
account the quartile of corrected QT interval, showed progres- LQT3 patients, cardiac events predominantly occur during
sive decrease in survival at longer QTc values. These data sleep and rest (39%) while exercise-induced events are rare
JACC Vol. 47, No. 1, 2006 Roberts 17
January 3, 2006:9–21 Genomics and Cardiac Arrhythmias

specific syndromes can be advised to avoid this form of


activity. Several studies have concluded that beta-blockers
are far more effective in LQT1 than LQT2 or LQT3.
Because it is more common to have a lethal event in LQT3
than in LQT1 or LQT2, some have suggested that an ICD
implantation is to be encouraged in patients with LQT3
syndrome (53). In 2002, Moss et al. (52) suggested LQT2
patients with mutations in the pore region of the KCNH2
gene have increased risk of arrhythmias over that of muta-
tions in the non-pore region. This has been disputed by
Zareba et al. (54), but a more recent Japanese study reports
findings similar to that of Moss et al. (55). Many patients
with LQTS do not respond to beta-blockers or other forms
of drug therapy and thus would be candidates for an ICD.
Nevertheless, in asymptomatic patients with borderline
prolonged QT, the decision remains difficult to insert an
ICD. The investigators strongly recommend that instituting
therapy in patients at low risk of becoming symptomatic
before the age of 40 years should be individualized. Further
studies are required in such patients before a robust recom-
mendation can be developed.

THE ROLE OF GENETICS IN DRUG-INDUCED LQTS


Long QT syndrome as a cause for ventricular tachycardia
and sudden death is more commonly associated with ac-
quired disorders than that of the familial LQTS. A major
cause of acquired long QT is drugs (56). The predominant
mechanism whereby the QT interval is prolonged, whether
it be genetic or acquired, is prolongation of sodium influx or
more commonly delayed outflux of potassium ions. There
are over 20 potassium channels expressed in the heart, most
of which participate in the AP (57). Antiarrhythmic drugs
can also induce long QT primarily those in class 1A (e.g.,
quinidine) or class 3 (e.g., sotolal). Of the 25 drugs for
which a mechanism has been elucidated, 24 of them are
blockers of the potassium channel, which delays repolariza-
tion. It is well recognized these drugs induce prolonged QT
only in individuals who are susceptible. It is the aim of
pharmacogenetics and pharmacogenomics to determine the
genetic basis for this susceptibility. One obvious avenue of
investigation is to determine whether these susceptible
Figure 3. Kaplan-Meier estimates of cumulative survival free of cardiac individuals have a mutation in one of the channel proteins
events among the 580 patients with the long QT syndrome in the risk that is aggravated by drugs. The extent to which this occurs
stratification analysis, according to gender and the QT interval corrected
for heart rate (QTc), in the group with a mutation at the LQT1 locus (A),
remains to be determined as screening for familial long QT
the group with a mutation at the LQT2 locus (B), and the group with a mutations remains experimental and time-consuming. A
mutation at the LQT3 locus (C). Printed with permission from N Engl recent study showed 10% to 15% of individuals with
J Med 2003;348:1866 –74.
drug-induced LQTS harbor ion channel gene mutations or
SNPs that predispose to the LQTS (58). Most of the
(51). In LQT2 patients, cardiac events occur equally during mutations or SNPs found so far that predispose to long QT
exercise or rest (51). Interestingly, a subtle startle in a form are in the potassium channels (59). Gene variants associated
of auditory stimulus is more likely to precipitate arrhythmias with acquired arrhythmias according to Anantharam et al.
in LQT2. Exercise or mental stress more often triggers (60) can be classified as indirect, direct, and compound.
ventricular arrhythmias in LQT4 patients (49). In LQT7, Indirect are mutations that impair the potassium channel at
hypokalemia is more likely to trigger ventricular arrhythmias baseline but do not affect drug sensitivity and are not
as well as periodic paralysis. Thus, patients with these associated with any prolongation of the QT interval until
18 Roberts JACC Vol. 47, No. 1, 2006
Genomics and Cardiac Arrhythmias January 3, 2006:9–21

drug administration is superimposed upon this inherited KCNH2 gene encoding an alpha subunit of the potassium
impairment. An example would be A116V-MIRPI with channel. The SQTS2 is caused by a gain-of-function mutation
quinidine (61). Another example is an Asian-specific SNP in the KCNQ1 gene encoding the alpha subunit of the
(G643S) in the KCNQ1 gene, which has been shown to potassium channel. The SQTS3 was reported recently (65) due
induce LQTS (62). The prevalence of G643S is 11% in the to a gain-of-function mutation in the KCNH2 gene encoding
Japanese population and was first identified after a patient for the inwardly rectifying potassium channel, which acceler-
being on a class 1A antiarrhythmia agent. Another example ates repolarization and shortens the AP duration. It is of some
of an SNP predisposing to prolonged long QT, which physiological significance that a loss of function mutation in
appears to be ethnic-related, is that of the S1102Y SNP these genes is associated with the opposite effect, namely,
found more commonly in the African population (63). This delayed repolarization, which induces familial LQTS. The
SNP in the SCN5A gene increases the risk of cardiac mechanism for arrhythmias in the SQTS is yet to be deter-
arrhythmias in the presence of drugs such as amiodarone. It mined. The prevalence of the SQTS has, until recently, been
is found primarily in West African, Caribbean, and African
considered rare. There is increasing evidence that many cases
American populations. It does not in itself cause LQTS but
of ventricular fibrillation and SCD previously considered idio-
only in the presence of other aggravating factors.
pathic may be due to SQTS. A rare disease referred to as
Direct mutations are perhaps the most difficult because
Anderson syndrome (66) is due to a loss-of-function mutation
they do not affect potassium current but increase sensitivity
in the gene encoding for a potassium channel subunit, which is
to drug blockade (64). An example of this is T8A-MIRPI
SNP present in 1.6% of the U.S. population was first associated with muscle paralysis, long QT, and ventricular
identified in a patient taking a combination of sulfamethox- arrhythmias. It is of interest that quinidine, a drug that
azole and trimethoprim. This SNP increases the sensitivity prolongs the QT interval, has been shown to favorably affect
of the sodium channel to blockage by the sulfa drugs. The this disease and is recommended as treatment to prevent
third category compound mutations are those that impair malignant arrhythmias.
channel function at baseline and also increase sensitivity to
the drug. An example is the Q9E MIRPI channel, which
was discovered in a female patient after taking clarithromy-
FAMILIAL CATECHOLAMINERGIC
cin, which precipitated torsades de pointes. Women are
POLYMORPHIC VENTRICULAR TACHYCARDIA
more vulnerable to develop prolonged QT than men if
caused by drugs that block the potassium channel (64). This is inherited in autosomal dominant and recessive forms
Ultimately, if we are to avoid sudden death associated with occurring without evidence of structural myocardial disease
prolonged QT, it will be necessary to screen for all known (67,68). It is characterized by bidirectional and polymorphic
mutations and SNPs that predispose to long QT. ventricular tachycardia in response to vigorous exercise. This
An equally fruitful area is the role of heredity with respect frequently deteriorates into ventricular fibrillation and
to the pharmacokinetics of these drugs. Most of these drugs death. Patients usually present with recurrent syncope,
are eliminated from the bloodstream by the cytochrome seizures, or sudden death after physical activity or emotional
CYP2D6 system in the liver which again, depending on stress (69). The mortality is significant approximating 30%
which particular polymorphism is inherited, can increase or by age 30 years (67,70). The first family was mapped to
decrease the blood concentration of that particular drug. 1q42– 43 (67), and missense mutations were identified in
Ultimately, it will be possible to determine whether some-
the gene encoding ryanodine receptor 2 (68). Subsequently,
one is a rapid or slow acetylator of these drugs, and the
mutations responsible for this disease were also shown to be
appropriate drug dose can be modified. It is well recognized
present in calsequestrin 2 (71). Thus, both genes are
in the case of procainamide that fast acetylators of this drug
involved with the release and handling of calcium.
are less likely to develop the lupus syndrome (56). Similar
The mechanism whereby these defects induce ventricular
studies are well recognized for the development of neurop-
athy in response to isoniazid therapy (56). tachycardia remains to be determined. It is well established
that the ectopic beats are initiated in discrete regions of the
epicardium. A recently proposed mechanism by Nam et al.
SQTS (72) suggests delayed afterdepolarization induces extrasys-
This is a rare disease associated with accelerated atrial and tolic activity, which triggers ventricular tachycardia under
ventricular depolarization and a high propensity for atrial and conditions of defective calcium handling. The origin of the
ventricular arrhythmias (13). Mutations have been identified in ectopic beats in the epicardium increases transmural disper-
three genes, all of which are a gain-of-function. The syndrome sion, which provides a substrate for re-entrant tachyarrhyth-
usually occurs in structurally normal hearts. The syndrome is mias. The reason for the ectopics originating in the epicar-
recognized by a short QT on the surface ECG. Three loci have dium is unknown but postulated to be because of the greater
been identified referred to as SQTS1 and SQTS2 and SQTS3 density of sarcoplasmic reticulum calcium content in the
(65); SQTS1 is caused by a gain-of-function mutation in the epicardium. The choice of treatment is a beta-blocker.
JACC Vol. 47, No. 1, 2006 Roberts 19
January 3, 2006:9–21 Genomics and Cardiac Arrhythmias

TISSUE GENE PROFILING USING MICROARRAYS TO and associate genes with certain responses to the environment
SEARCH FOR GENES THAT INDUCE OR PREDISPOSE such as remodeling with atrial fibrillation or hypertrophy in
TO VENTRICULAR AND ATRIAL ARRHYTHMIAS response to ischemic heart disease.

The human genome is estimated to contain about 25,000


distinct genes. Identification of these genes and their func- FUTURE OF GENOMICS IN
tion remains to be determined. Regulation of gene expres- CARDIOVASCULAR ARRHYTHMIAS
sion, recognized to play a major role in the response of the While cardiac arrhythmias may be associated with or
body to environmental stimuli including disease, is of without structural heart disease, the defect always involves
critical importance in the diagnosis and treatment of disease. some aspect of the electrical conduction system of the heart.
Genetic regulation of organ differentiation is perhaps even less The structural component is, of course, the sinus node,
understood. DNA or RNA microarray (73,74), a relatively new specialized atrial tracts, atrioventricular node, and bundle of
technique consisting of thousands of genes etched into a chip, HIS with the Purkinje system. After conducting within this
can be used to identify genes via hybridization to complimen- specialized electrical transmission, the current must pass
tary base pairing. Microarray chips are now available with into other cells such as myocytes. The important compo-
essentially all of the genes represented by expressed sequence nents are predominantly ionic currents, ion channels, struc-
tag or oligonucleotide. This technique is being utilized to tural proteins, and gap junctions. Most of the genetic
determine which genes are expressed in certain tissues and the defects currently identified involve some aspect of the
extent to which their expression is regulated in response to a subunits of the ion channels. We now recognize at least 429
variety of stimuli. It is well recognized in animal models and in genes that encode ion channel proteins in the human (79).
human disease that atrial fibrillation, once it occurs, tends to be Of these, 170 encode potassium channels, 38 are for calcium
self-perpetuating and is associated with significant tissue re- channels, 29 are for sodium channels, 58 are for chloride
modeling of the atria (75). Studies being performed in the goat channels, and 15 are glutemate receptors (80). It is of
(76) and in human atrial tissue removed at the time of surgery interest that ion channels represent only about 5% of the
(77) show a distinct gene expression profile associated with molecular targets of modern medicine (81,82). It is now
atrial remodeling in response to atrial fibrillation. A variety of recognized that in addition to the role of ion channels
genes have been observed to be up-regulated while others are regulating membrane potential they also regulate many
down-regulated during this remodeling process (78). It has also other functions including cell volume and hormone secre-
been shown in animal models that once atrial fibrillation is tion (83,84). The complexity of these channels, such as the
converted to sinus rhythm, this pattern will return to that of the voltage-gated cardiac potassium channels, is such that
normal atrium (76). This provides an opportunity to assess several domains exist within the integrated channel protein.
these genes for polymorphisms as well as perform genomic In addition to the pore domain, there are the domains that
function analysis to determine genes that play a pathogenic role provide ion selectivity, opening and closing of the gate
in sustaining atrial fibrillation. Such studies, while in their together with the sensing units whether it be voltage or
primitive state, are ongoing. Another area of interest is atrial chemical. The elucidation of mutations causing disease is
fibrillation that occurs after cardiac surgery in about 20% of rapidly enhancing our understanding of the topography and
individuals (77). Are they predisposed to genetic factors? The sequences involved with each domain. Approaches by the
ability to scan with a chip containing 25,000 genes should pharmaceutical industry to develop designer drugs for those
accelerate finding genes pivotal to the disease. domains, while in its infancy, are rapidly developing.
Similarly, it is well recognized that ventricular hypertrophy The characteristic feature of the electrical activity of the
increases one’s risk of sudden death two- to four-fold regardless heart is the AP of the atria and ventricles due to depolar-
of the stimulus (18). The mechanism whereby ventricular ization and repolarization with the characteristic long pla-
hypertrophy increases risk to arrhythmias have not been deter- teau phase involving multiple ion channels. Many of these
mined. It is thus reasonable to assume that certain genes with channels have now been cloned and will serve as prime
polymorphisms that predispose to sudden death may be up- candidates for the identification of genetic defects in the
regulated in hypertrophy, which accounts in part for the future. The major currents involved in the atrial and
vulnerability to arrhythmias. Microarray analysis, again, has ventricular AP are indicated in Figure 2. Cardiac electrical
already shown there is a differential display of genes in the atria activity is thus a complex process, which integrates the
and ventricle, and with the development of hypertrophy the electrical activity of multiple molecules. Any variant in
fetal program of genes is up-regulated along with a variety of either of these molecules whether it is due to SNPs or
other genes (18). The microarray also provides the opportunity greater defects can significantly alter cardiac activity. It is
to classify genes in clusters as well as identify sets of genes that well recognized that structural heart disease, associated with
play a role in cell signaling. Thus, if a particular gene with an either hypertrophy or cardiac dilatation, can structurally
SNP is shown to be important, one can assess for SNPs in affect electrical transmission or through altered gene expres-
other such genes that are part of the signaling network. sion induce arrhythmias. Hypertrophy is well documented
Nevertheless, it will significantly accelerate our ability to relate to increase the propensity for SCD by about four-fold.
20 Roberts JACC Vol. 47, No. 1, 2006
Genomics and Cardiac Arrhythmias January 3, 2006:9–21

Structural heart disease is associated with excessive fibrous tial stages of the ventricular growth response. J Cell Biol
1988;107:1911– 8.
tissue, which has a lower velocity of conduction and could
19. Marian AJ, Yu QT, Workman R, Greve G, Roberts R. Angiotensin
lead to re-entry arrhythmias. Common genetic variants have converting enzyme polymorphism in hypertrophic cardiomyopathy
been shown to increase the risk of cardiac arrhythmias, and and sudden cardiac death. Lancet 1993;342:1085– 6.
most recent studies show that even common genetic variant 20. Feinberg WM, Blackshear JL, Laupacis A, Kronmal R, Hart RG.
Prevalence, age distribution, and gender of patients with atrial fibril-
in genes referred to as SNPs can alter cardiac electrical lation. Analysis and implications. Arch Intern Med 1995;155:469 –73.
manifestations in certain populations. Atrial fibrillation is 21. Phillips K, Go AS, Hylek EM, et al. Prevalence of diagnosed atrial
associated with significant morbidity in the aging popula- fibrillation in adults: national implications for rhythm management
and stroke prevention: the AnTicoagulation and Risk Factors in Atrial
tion. Ventricular fibrillation is a lethal arrhythmia respon-
Fibrillation (ATRIA) study. JAMA 2001;285:2370 –5.
sible for over 600,000 deaths each year in the Western 22. The Boston Area Anticoagulation Trial for Atrial Fibrillation Inves-
world. Both atrial and ventricular fibrillation remain the tigators. The effect of low-dose warfarin on the risk of stroke in
most challenging of rhythm disorders. patients with nonrheumatic atrial fibrillation. N Engl J Med 1990;323:
1505–11.
23. Wolf PA, Abbott RD, Kannel WB. Atrial fibrillation: a major
Reprint requests and correspondence: Dr. Robert Roberts, contributor to stroke in the elderly: the Framingham study. Arch
University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, Intern Med 1987;147:1561– 4.
Ontario, K1Y 4W7 Canada. E-mail: rroberts@ottawaheart.ca. 24. Brugada R. Is atrial fibrillation a genetic disease? J Cardiovasc
Electrophysiol 2005;16:553– 6.
25. Wall JD, Pritchard JK. Haplotype blocks and linkage disequilibrium in
the human genome. Nat Rev Genet 2003;4:587–97.
REFERENCES 26. Ellinor PT, Moore RK, Patton KK, Ruskin JN, Pollack MR, MacRae
CA. Mutations in the long QT gene, KCNQ1, are an uncommon
1. Tsai CC, Lai L-P, Lin J, et al. Renin-angiotensin system gene cause of atrial fibrillation. Heart 2004;90:1487– 8.
polymorphisms and atrial fibrillation. Circulation 2004;109:1640 – 6. 27. Yang Y, Xia M, Jin Q, et al. Identification of a KCNE2 gain-of-
2. Lai L-P, Su M-J, Yeh H-M, et al. Association of the human minK
function mutation in patients with familial atrial fibrillation. Am Hum
gene 38G allele with atrial fibrillation: evidence of possible genetic
Genet 2004;75:899 –905.
control on the pathogenesis of atrial fibrillation. Am Heart J 2002;
28. Xia M, Jin Q, Bendahhou S, et al. A Kir2.1 gain-of-function mutation
144:485–90.
3. Chan L, Boerwinkle E. Gene-environment interactions and gene underlies familial atrial fibrillation. Biochem Biophys Res Commun
therapy in atherosclerosis. Cardiol Rev 1994;2:130 –7. 2005;332:1012–9.
4. American Heart Association. Heart and Stroke Statistical Update. 29. Li J, McLerie M, Lopatin AN. Transgenic upregulation of IK1 in the
2000 edition. Dallas, TX: American Heart Association, 2000. mouse heart leads to multiple abnormalities of cardiac excitability.
5. Patil N, Berno AJ, Hinds DA, et al. Blocks of limited haplotype Am J Physiol 2004;287:48289 –94.
diversity revealed by high-resolution scanning of human chromosome 30. Dobrev D, Graf E, Wettwer E, et al. Molecular basis of downregu-
21. Science 2001;294:1719 –23. lation of G-protein-coupled inward rectifying K(⫹) current
6. Hirschhorn JN, Daly MJ. Genome-wide association studies for com- (I(K,ACh) in chronic human atrial fibrillation: decrease in GIRK4
mon diseases and complex traits. Nat Rev Genet 2005;6:95–108. mRNA correlates with reduced I(K,ACh) and muscarinic receptor-
7. Gibbs RA. The international HapMap project. Nature 2003;426:789 – mediated shortening of action potentials. Circulation 2001;104:
96. 2551–7.
8. Kruglyak L. Prospects for whole-genome linkage disequilibrium map- 31. Dobrev D, Wettwer E, Kortner A, Knaut M, Schuler S, Ravens U.
ping of common disease genes. Nat Genet 1999;22:139 – 44. Human inward rectifier potassium channels in chronic and postoper-
9. Arking D, Chugh S, Chakravarti A, Spooner P. Genomics in sudden ative atrial fibrillation. Cardiovasc Res 2002;54:397– 404.
death. Circ Res 2004;94:712–23. 32. Van Wagoner DR, Pond AL, McCarthy PM, Trimmer JS, Nerbonne
10. Echt DS, Liebson PR, Mitchell LB, et al. Mortality and morbidity in JM. Outward K⫹ current densities and Kv1.5 expression are reduced
patients receiving encainide, flecainide, or placebo. The Cardiac in chronic human atrial fibrillation. Circ Res 1997;80:772– 81.
Arrhythmia Suppression Trial. N Engl J Med 1991;324:781– 8. 33. Van Wagoner DR. Electrophysiological remodeling in human atrial
11. Bristow MR, O’Connell JB. Myocardial diseases. In: Kelley WN, fibrillation. Pacing Clin Electrophysiol 2003;26:1572–5.
Watanabe AM, editors. Textbook of Internal Medicine. Philadelphia, 34. Hong K, Bjerregaard P, Gussak I, Brugada R. Short QT syndrome
PA: J. B. Lippincott Co., 1992:150 –7. and atrial fibrillation caused by mutation in KCNH2. J Cardiovasc
12. Maron BJ, Gardin JM, Flack JM, Gidding SS, Kurosaki TT, Bild DE. Electrophysiol 2005;4:394 – 6.
Prevalence of hypertrophic cardiomyopathy in a general population of 35. Firouzi M, Ramanna H, Kok B, et al. Association of human
young adults: echocardiographic analysis of 4,111 subjects in the connexin40 gene polymorphisms with atrial vulnerability as a risk
CARDIA study. Circulation 1995;92:785–9. factor for idiopathic atrial fibrillation. Circ Res 2004;95:e29 –33.
13. Marian AJ, Brugada R, Roberts R. Cardiovascular diseases due to
36. Guize L, Soria R, Chaouat JC. Prevalence et evolution du syndrome
genetic abnormalities. In: Fuster V, Alexander RW, O’Rourke RA,
de Wolff-Parkinson-White dans une population de 138,048 sujets.
editors. Hurst’s the Heart. 11th edition. New York, NY: McGraw-
Ann Med Interne 1985;136:474 – 8.
Hill, 2004.
37. Gollob MH, Green MS, Tang A, et al. Identification of a gene
14. Wan Q, Wu N, Fan W, Tang YY, Jin L, Fang Q. Clinical
manifestations and prevalence of different types of supraventricular responsible for familial Wolff-Parkinson-White syndrome. New Engl
tachycardia among Chinese. Chin Med J 1992;105:284 –5. J Med 2001;344:1823– 64.
15. Benson DW, Jr., Benditt DG, Anderson RW, et al. Cardiac arrest in 38. Arad M, Benson DW, Perez-Atayde A, et al. Constitutively active
young, ostensibly healthy patients: clinical, hemodynamic, and elec- AMP kinase mutations cause glycogen storage disease mimicking.
trophysiologic findings. Am J Cardiol 1983;52:65–9. Clin Invest 2002;109:357– 62.
16. Fox CS, Parise H, D’Agostino RB, et al. Parental atrial fibrillation as 39. Sidhu J, Yadavendra S, Rajawat YS, et al. Transgenic mouse model of
a risk factor for atrial fibrillation in offspring. J Am Cardiol 2004;292: ventricular preexcitation and atrioventricular reentrant tachycardia
1174 –5. induced by an AMP-activated protein kinase loss-of-function muta-
17. Splawski I, Timothy KW, Tateyama M, et al. Variant of SCN5A tion responsible for Wolff-Parkinson-White syndrome. Circulation
sodium channel implicated in risk of cardiac arrhythmia. Science 2005;111:21–9.
2002;297:1333– 6. 40. Cheung PC, Salt I, Davies DG, Hardie DG, Carling D. Character-
18. Ueno H, Perryman MB, Roberts R, Schneider MD. Differentiation of ization of AMP-activated protein kinase gamma-subunit isoforms and
cardiac myocytes following mitogen withdrawal exhibits three sequen- their role in AMP binding. Biochem J 2000;346:659 – 69.
JACC Vol. 47, No. 1, 2006 Roberts 21
January 3, 2006:9–21 Genomics and Cardiac Arrhythmias

41. Light PE, Wallace CHR, Dyck JRB. Constitutively-active AMP- susceptibility to life-threatening arrhythmias. J Cardiovasc
activated protein kinase regulates voltage-gated sodium channels in Electrophysiol 2001;12:1223–9.
ventricular myocytes. Circulation 2003;107:1962–5. 63. Splawski I, Timothy KW, Tateyama M, et al. Variant of SCN5A
42. Arad M, Moskowitz IP, Patel VV, et al. Transgenic mice overexpress- sodium channel implicated in risk of cardiac arrhythmia. Science
ing mutant PRKAG2 define the cause of Wolff-Parkinson-White 2002;297:1333– 6.
syndrome in glycogen storage cardiomyopathy. Circulation 2003;107: 64. Makkar RR, Fromm BS, Steinman RT, Meissner MD, Lehmann
2850 – 6. MH. Female gender as a risk factor for torsades de pointes associated
43. Brugada P, Brugada J. Right bundle branch block, persistent ST with cardiovascular drugs. J Am Med Assoc 1993;270:2590 –7.
segment elevation and sudden cardiac death: a distinct clinical and 65. Priori SG, Pandit SV, Rivolta I, et al. A novel form of short QT
electrocardiographic syndrome. A multicenter report (see comments). syndrome (SQT3) is caused by a mutation in the KCNJ2 gene. Circ
J Am Coll Cardiol 1992;20:1391– 6. Res 2005;96:800 –7.
44. Wang Q, Li Z, Shen J, Keating MT. Genomic organization of the 66. Schulze-Bahr E. Short QT syndrome or Andersen syndrome: yin and
human SCN5A gene encoding the cardiac sodium channel. Genomics yang of Kir2.1 channel dysfunction. Circ Res 2005;96:703– 4.
1996;34:9 –16. 67. Swan H, Pippo K, Viitasalo M, et al. Arrhythmic disorder mapped to
45. Yarborough TL, Lu T, Lee HCSEF. Localization of cardiac sodium chromosome 1q42-q43 causes malignant polymorphic ventricular
channels in caveolin-rich membrane domains; regulation of sodium tachycardia in structurally normal hearts. J Am Coll Cardiol 1999;34:
current amplitude. Circ Res 2002;90:443–9. 2035– 42.
46. Hong K, Guerchicoff A, Pollevick GD, et al. Cryptic 5 splice site 68. Priori SG, Napolitano C, Tiso N, et al. Mutations in the cardiac
activation in SCN5A associated with Brugada syndrome. J Mol Cell ryanodine receptor gene (hRyR2) underlie catecholaminergic polymor-
Cardiol 2005;38:555– 60. phic ventricular tachycardia. Circulation 2001;103:196 –200.
47. Keating MT, Sanguinetti MC. Molecular and cellular mechanisms of 69. Viskin S, Belhassen B. Polymorphic ventricular tachyarrhythmias in
cardiac arrhythmias. Cell 2001;104:569 – 80. the absence of organic heart disease: classification, differential diagno-
48. Priori SG, Schwartz PJ, Ronschetti E, et al. Risk stratification in the sis, and implications for therapy. Prog Cardiovasc Dis 1998;41:17–34.
long-QT syndrome. N Engl J Med 2005;348:1866 –74. 70. Leenhardt A, Lucet V, Denjoy I, Grau F, Ngoc DD, Coumel P.
49. Mohler PJ, Schott JJ, Gramolini AO, et al. Ankyrin-B mutation Catecholaminergic polymorphic ventricular tachycardia in children: a
causes type 4 long-QT cardiac arrhythmia and sudden cardiac death. 7-year follow-up of 21 patients. Circulation 1995;91:1512–9.
Nature 2003;421:634 –9. 71. Lahat H, Pras E, Olender T, et al. A missense mutation in a highly
50. Splawski I, Shen J, Timothy K, et al. Spectrum of mutations in conserved region of CASQ2 is associated with autosomal recessive
catecholamine-induced polymorphic ventricular tachycardia in Be-
long-QT syndrome genes. KVLQT1, HERG, SCN5A, KCNE1, and
douin families from Israel. Am J Hum Genet 2001;69:1378.
KCNE2. Circulation 2000;102:1178 – 85.
72. Nam G-B, Burashnikov B, Antzelevitch C. Cellular mechanisms
51. Schwartz PJ, Priori SG, Spazzolini C, et al. Genotype-phenotype
underlying the development of catecholaminergic ventricular tachycar-
correlation in the long-QT syndrome: gene-specific triggers for life-
dia. Circulation 2005;111:2727–33.
threatening arrhythmias. Circulation 2001;103:89 –95.
73. Kononen J, Bubendorf L, Kallioniemi A, et al. Tissue microarrays for
52. Moss AJ, Zareba W, Kaufman ES, et al. Increased risk of arrhythmic
high-throughput molecular profiling of tumor specimens. Nat Med
events in long-QT syndrome with mutations in the pore region of the
1998;4:844 –7.
human ether-a-go-go-related gene potassium channel. Circulation 74. Schena M, Shalon D, Davis RW, Brown PO. Quantitative monitor-
2002;105:794 –9. ing of gene expression patterns with a complementary DNA microar-
53. Zareba W, Moss AJ, Schwartz PJ, et al. Influence of the genotype on ray. Science 1995;270:467–70.
the clinical course of the long-QT syndrome. N Engl J Med 1998; 75. Lai L-P, Lin J-E, Lin C-S, et al. Functional genomic study on atrial
339:960 –5. fibrillation using cDNA microarray and two-dimensional protein
54. Zareba W, Moss AJ, Sheu G, et al. Location of mutation in the electrophoresis techniques and identification of the myosin regulatory
KCNQ1 and phenotypic presentation of long QT syndrome. J Car- light chain isoform reprogramming in atrial fibrillation. J Cardiovasc
diovasc Electrophysiol 2003;14:1149 –53. Electrophysiol 2004;15:214 –23.
55. Shimizu W, Horie M, Ohno S, et al. Mutation site-specific differences 76. Thijssen VL, van der Velden HMW, van Ankeren EP, et al. Analysis
in arrhythmic risk and sensitivity to sympathetic stimulation in LQT of altered gene expression during sustained atrial fibrillation in the
form of congenital long QT syndrome-multi-center study in Japan. goat. Cardiovasc Res 2002;54:427–37.
J Am Coll Cardiol 2004;44:117–25. 77. Ohki R, Yamamoto K, Ueno S, et al. Gene expression profiling of
56. Roden DM. Pharmacogenetics and drug-induced arrhythmias. Car- human atrial myocardium with atrial fibrillation by DNA microarray
diovasc Res 2001;50:224 –31. analysis. Int J Cardiol 2005;102:233– 8.
57. Escande D. Pharmacogenetics of cardiac K⫹ channels. Eur J Phar- 78. Kim Y-H, Lee J-H, Lim D-S, et al. Gene expression profiling of
macol 2000;410:281–7. oxidative stress on atrial fibrillation in humans. Exp Mol Med
58. Yang P, Kanki H, Drolet B, et al. Allelic variants in long-QT disease 2003;35:336 – 49.
genes in patients with drug-associated torsades de pointes. Circulation 79. Xu J, Chen Y, Li M. High-throughput technologies for studying
2002;105:1943– 8. potassium channels—progresses and challenges. Circ Res 2004;3:
59. Moule SK, Denton RM. The activation of p38 MAPK by the 32– 8.
beta-adrenergic agonist isoproterenol in rat epididymal fat cells. FEBS 80. Yellen G. The voltage-gated potassium channels and their relatives.
Lett 1998;439:287–90. Nature 2002;419:35– 42.
60. Anantharam A, Markowitz S, Abbott GW. Pharmacogenetic consid- 81. Hopkin AL, Groom CR. The druggable genome. Nat Rev Drug
erations in diseases of cardiac ion channels. J Pharmacol Exp Ther Discov 2002;1:727–30.
2003;307:831– 8. 82. Drews J. Drug discovery: a historical perspective. Science 2000;287:
61. Sesti F, Abbott GW, Wei JY, et al. A common polymorphism 1960 – 4.
associated with antibiotic-induced cardiac arrhythmia. Proc Natl Acad 83. Jan LY, Jan YN. Cloned potassium channels from eukaryotes and
Sci U S A 2000;97:10613– 8. prokaryotes. Annu Rev Neurosci 1997;20:91–123.
62. Kubota T, Horie M, Takao M, et al. Evidence for a single nucleotide 84. Jan LY, Jan YN. Structural elements involved in specific K⫹ channel
polymorphism in the KCNQ1 potassium channel that underlies functions. Annu Rev Physiol 1992;54:537–55.

You might also like