ChapterPDF Available

The Molecular Pathophysiology of Atrial Fibrillation

Authors:
1
The Molecular Pathophysiology of Atrial Fibrillation
Stanley Nattel,1, 2,4 Jordi Heijman,3 Niels Voigt,4 Xander H.T. Wehrens5
and Dobromir Dobrev4
1Research Center, Department of Medicine, Montreal Heart Institute and Université de Montréal,
Montreal, Quebec, Canada; 2Department of Pharmacology and Therapeutics, McGill University;
3Department of Cardiology, Maastricht University, Maastricht, The Netherlands; 4Institute of
Pharmacology, West-German Heart and Vascular Center, Faculty of Medicine, University
Duisburg-Essen, Essen, Germany; 5Cardiovascular Research Institute; Departments of Molecular
Physiology and Biophysics, Medicine (Cardiology), Pediatrics, Baylor College of Medicine,
Houston, USA
Correspondence to Stanley Nattel, 5000 Belanger St E, Montreal, Quebec, H1T 1C8, Canada;
Tel.: (514)-376-3330 ext. 3990. Fax: (514)-376-1355. E-mail: stanley.nattel@icm-mhi.org.
2
Atrial fibrillation (AF) is a highly prevalent and clinically relevant arrhythmia, for which all
current therapeutic approaches have important limitations. An improved understanding of the
mechanistic basis of AF has evolved over the past decades, particularly with respect to molecular
aspects. Mechanistic insights have contributed greatly to the contemporary management of AF
and are expected to further improve arrhythmia therapy in the future (1). The purpose of this
chapter is to review recent findings in the molecular pathophysiology of AF, and to discuss their
potential value for improving management.
We will begin with an overview of the etiological determinants of AF, then discuss briefly
the principal mechanisms contributing to AF, and finally review the molecular basis of specific
arrhythmia determinants.
Etiological Determinants
Heart Disease
The majority of AF-cases have associated cardiac disease (Figure 1). Cardiac senescence
is a major predisposing factor, largely mediated by structural remodeling which causes fibrotic
alterations and microconduction slowing, as well as atrial enlargement (2). Heart failure (HF),
hypertensive heart disease, valvular disease and ischemic heart disease are major contributors to
AF-occurrence. Less common conditions leading to AF include pericarditis, myocarditis, and
various cardiomyopathies. Cardiac surgery is followed by post-operative AF, with a typical
presentation and therapeutic response, in about 30% of cases.
3
Genetic Determinants
There has been a rapid increase in knowledge of genetic determinants of AF over the past
10 years (3-6). A wide range of disease-causing mutations has been established (Table 1) and
additional AF susceptibility gene-loci have been identified with genome-wide association studies
(GWASs; Table 2). Rare variants linked to monogenic forms of AF have high penetrance and
provide important insights into AF-mechanisms, whereas common genetic variants identified
using GWAS provide new insights into genetic-based population determinants of AF, while
raising challenging pathophysiological issues (6).
Extracardiac Contributors
A variety of extracardiac conditions can affect AF-occurrence. Heavy alcohol
consumption promotes AF (7), and hyperthyroidism is a well-recognized factor (8). Obesity is
increasingly recognized as an AF risk-factor (9), with obstructive sleep apnea, often associated
with obesity, also being an important contributor. Autonomic tone may set the conditions for AF
initiation and maintenance. The AF-promoting properties of vagal activation are well-known,
and there is increasing evidence for an important role of combined sympathovagal discharge (10).
General Mechanisms and AF Forms
Focal ectopic firing and reentrant activity are the primary AF arrhythmia mechanisms
(Figure 2A). Focal activity can be transient, producing isolated atrial extrasystoles or self-
limited tachycardias, and can contribute to AF-generation by acting as a trigger to initiate reentry
in a vulnerable substrate. In addition, sustained focal ectopic activity can produce rapid “driver”
activity that is conducted heterogeneously to generate fibrillatory conduction maintaining the
4
irregular activity typical of AF (11, 12). Clinical AF can be paroxysmal (self-terminating) or
persistent (terminating only with medical intervention). With increasing duration, persistent AF
becomes increasingly resistant to therapy and “long-standing persistent AF”, lasting >1 year, and
often becomes permanent when attempts to restore sinus rhythm fail or are abandoned.
Repetitively-firing focal ectopic drivers are believed to produce paroxysmal forms. Reentrant
activity generates more persistent AF, tending to become more fixed, therapy-resistant and
irreversible as the substrate evolves (13, 14). Besides progression of AF-associated
comorbidities, the evolution of the substrate is promoted by AF-induced remodeling related to the
rapid atrial rate, cardiac dysfunction, neurohumoral effects and consequences of atrial metabolic
disturbances (Figure 2B). The remodeling induced by long-standing AF involves both functional
and structural components that promote a transition towards complex reentrant mechanisms, as
quantified in recent noninvasive mapping studies (15, 16). These analyses have shown
pronounced temporal and inter-patient variability in AF activity, suggesting dynamic and
complex interactions between AF mechanisms.
Focal Ectopic Activity
Several mechanisms produce abnormal impulse formation and can cause focal ectopic
activity. Spontaneous automatic activity depends on the balance between inward and outward
currents during phase 4 of the action potential (AP). Increased phase 4 inward currents carried by
Na+ or Ca2+, particularly time-dependent activating currents like the “funny current” If, and/or
decreased phase 4 outward currents, produce spontaneous phase-4 depolarization that can reach
threshold and cause ectopic firing.
Focal ectopic activity may also result from afterdepolarizations, which are subdivided into
early afterdepolarizations (EADs; arising before the end of phase 3) or delayed
5
afterdepolarizations (DADs) occurring after full repolarization. Normal cardiomyocyte Ca2+
handling is crucial for cardiac contractility (Figure 3A). DADs (Figure 3B) are thought to be the
predominant cause of focal atrial ectopic firing. DADs are caused by a diastolic Ca2+ leak from
the sarcoplasmic reticulum (SR) via SR Ca2+-release channels or “ryanodine receptors” (RyRs,
RyR2 being the cardiac isoform). Systolic Ca2+-release through RyR2s mediates cardiac
excitation-contraction coupling. Relaxation is mediated by diastolic Ca2+ removal from the
cytosol into the SR by a Ca2+-uptake pump, the SR Ca2+-ATPase (SERCA2a). RyR2s are
sensitive to both cytosolic and intraluminal SR Ca2+ concentrations and diastolic releases result
from SR Ca2+-overload or when oversensitive RyR2s have an abnormally low Ca2+-threshold for
Ca2+-release. Excess cytosolic Ca2+ is handled by the sarcolemmal Na+, Ca2+-exchanger (NCX),
which moves three Na+ ions (charge +3) into the cell for each Ca2+ ion (charge +2) extruded into
the extracellular space, generating net inward current (called “transient inward current”, “Iti”) that
depolarizes the cell, producing a DAD. When DADs reach threshold they induce premature AP
firing (dashed line in Figure 3B), which promotes its own perpetuation by synchronizing release
events and allowing more Ca2+ to enter the cell (17). Repeated DAD-triggered APs can generate
focal atrial tachycardias. RyR2-function is regulated by channel-phosphorylation:
hyperphosphorylation enhances RyR2 Ca2+-sensitivity and promotes DAD-formation.
Calsequestrin (CSQ) is the principal Ca2+-storage buffer of the SR. Inadequate CSQ
function/expression increases free SR Ca2+-concentration and promotes diastolic RyR2 Ca2+-
release (18).
EADs generally occur when AP-duration (APD) is excessively prolonged. With very long
APs, L-type Ca2+-currents may have enough time to recover from inactivation carrying inward
Ca2+-current to generate an EAD and stimulate a spontaneous extrasystole.
6
Reentry
The two principal competing conceptual frameworks for understanding functional reentry (19)
are shown in Figure 4 (top). The leading-circle model (Figure 4A) posits reentry around a central
zone that is continuously activated by centripetal waves emanating from a reentering activation
wavefront. In this model, reentry establishes itself in a circuit with dimension equal to the
distance travelled during one refractory period (“wavelength”, refractory period times conduction
velocity; CV). When the wavelength is small because of slow conduction or brief refractoriness,
multiple circuits can be accommodated in the atria and spontaneous self-termination is unlikely.
In the spiral-wave model (Figure 4B), reentry is maintained by rotors established by tissue
excitability properties (depending on both conduction and refractoriness), which determine rotor
period, stability and size (greater excitability generates smaller, more stable, and faster rotors).
Anatomical obstacles or complexities favor reentry by anchoring reentry circuits. Figure 4C
illustrates the effect of structural remodeling. Progressive atrial dilation creates longer
conduction pathways for reentry. Tissue fibrosis slows conduction, makes it more heterogeneous
and creates conduction-barriers that favor the development of stable rotors and/or multiple
simultaneous irregular reentry circuits that can sustain AF. In addition, fibroblast proliferation
can promote arrhythmogenesis via cardiomyocyte-fibroblast interactions that alter AP-properties
and slow conduction.
Molecular control mechanisms
Molecular control of gene-expression in AF
There is extensive evidence for an important role of altered gene-control in AF
pathogenesis. Figure 5 presents a simplified schematic of dysregulated molecular gene-
7
expression control mechanisms that are implicated in AF. These are recognized to occur in at
least 4 different contexts: 1) AF-inducing mutations in transcription factors (TFs); 2) gene-
variants identified by GWAS that are in or related to TFs, which are the presumptive immediate
mediators of AF-substrate control; 3) AF-induced signaling changes that are involved in AF-
induced atrial remodeling; 4) changes in gene-expression that mediate the AF-promoting effects
of predisposing diseases and risk factors. Many of these are discussed in detail below, but we will
provide a general overview here. The tissue-specific cellular phenotype is controlled primarily by
selective expression of genes in the genome. Transcription-patterns from DNA to messenger-
RNA (mRNA) are tissue and disease specific, largely under the regulation of TFs that bind to
specific DNA-sequences and enhance or suppress transcription of associated sequences.
Mutations in the TFs GATA, NKX2-5 and TBX5 cause various forms of congenital heart disease
and predispose to AF (20). In addition, GWASs have identified a number of single-nucleotide
polymorphisms (SNPs) believed to control AF-risk via TFs, like the 4q25 variants for which
PITX2 is the closest gene, the 16q22 SNPs in an intron (non-coding segment) of ZFHX3 and the
1q24 variants in PRRX1 (Table 2). AF itself causes reprogramming of gene-expression, in
particular by increasing cellular Ca2+-loading and thereby activating a variety of signaling
systems (21). Prominent among these is the Nuclear Factor of Activated T-Lymphocytes
(NFAT)/calcineurin system, which regulates the expression of a range of important ion-channels
(21). AF-inducing conditions (in many cases along with AF itself) act through a number of cell-
membrane receptors controlling signaling pathways like the renin-angiotensin-aldosterone and
transforming growth-factor-β systems, which induce atrial remodeling through a range of
downstream TFs. Reactive oxygen species, particularly those derived from NADPH-oxidase, also
play a key role. Nuclear-delimited signaling likely also occurs, although we are just beginning to
understand these pathways and their role in disease states (22). Finally, a range of small non-
8
coding RNA-sequences called microRNAs, which control gene-expression principally by
blocking translation of target mRNAs, have been implicated in AF-inducing remodeling. Long
non-coding RNA (lncRNA) might also contribute (23), but this research is just in its infancy in
the atria.
Molecular control of cell Ca2+-handling and DAD generation
Normal cell Ca2+-handling is crucial for cellular contraction and relaxation (Figure 3A;
see also Chapter 16 of this book). Abnormal SR Ca2+-handling is seen in both paroxysmal and
chronic AF (pAF, cAF) patients (24-29), promoting spontaneous RyR2-mediated diastolic SR-
Ca2+ releases. Moreover, patients with rare inherited variants in RyR2 linked to
catecholaminergic polymorphic ventricular tachycardia (CPVT) also commonly exhibit AF as a
result of abnormal SR Ca2+ release (30).
Figure 6 summarizes the detailed molecular pathobiology of DAD-inducing diastolic
RyR2 Ca2+-release in non-genetic forms of AF. Protein-kinase A (PKA)-phosphorylation of
RyR2 at Ser2808 (26) and Ca2+-calmodulin dependent kinase-II (CaMKII) phosphorylation at
Ser2814 are increased in dogs and goats with pacing-induced AF and in AF-patients (24, 29-32).
CaMKII-activity is normally autoinhibited. Ca2+-calmodulin binding removes autoinhibition,
activating CaMKII and causing autophosphorylation that activates CaMKII and makes it Ca2+-
independent. Similar activation may result from CaMKII-oxidation. Changes in RyR2
phosphorylation-state at PKA and CaMKII sites may result not only from changed kinase-
activity, but also from alterations in dephosphorylating enzymes, phosphatases (31). Proteomic
studies revealed that the serine/threonine protein phosphatase type-1 (PP1), a major phosphatase
in the heart, is also dysregulated in pAF (33). In particular, extensive changes in PP1 regulatory
9
subunits were observed, which may underlie heterogeneous changes in the phosphorylation status
of Ca2+-handling proteins in AF patients.
These post-translational alterations increase RyR2 Ca2+-sensitivity, enhancing channel
open-probability (24, 26). Mice deficient in RyR2-inhibitory FK-505 binding protein 12.6, mice
with gain-of-function mutations in RyR2, and mice with constitutively phosphorylated RyR2
channels at S2814 (S2814D mice), all exhibit increased susceptibility to pacing-induced AF in
association with increased atrial-cell SR Ca2+-leak and triggered activity (24, 30, 34, 35).
Angiotensin-effects to promote AF may in part be due to oxidative stress acting via CaMKII-
oxidation on diastolic RyR2 Ca2+-release (36). RyR2-dysfunction can be induced by Ca2+-
overload resulting from phospholamban hyperphosphorylation, which removes phospholamban-
inhibition of SERCA and enhances SR Ca2+-uptake (31), as has been reported for pAF (29).
Phospholamban-hyperphosphorylation can be produced by enhanced PKA or CaMKII-activity,
or by decreased phosphatase-function. Reduced phosphatase-function can be a consequence of
increased activity of an inhibitory protein, I-1, typically caused by I-1 hyperphosphorylation (31).
Increases in NCX expression and/or function are also commonly noted in persistent AF
(24, 28, 31, 37), causing Iti resulting from any specific amount of diastolic SR Ca2+ leak to be
larger in AF, likely contributing to the increased risk of DADs and triggered ectopic activity (24,
38). Cardiac IP3-receptors (IP3R2) act as Ca2+-transporting pathways and can facilitate SR Ca2+-
leak to promote arrhythmogenesis. IP3R2 expression is increased by ATR and is greater in atria
than ventricles (39). IP3R2-coupled amplification of atrial SR Ca2+-release events and related
arrhythmogenesis may thus contribute to AF-related ectopic activity.
Congestive heart failure (CHF) is a very important cause of AF. Focal drivers and
triggered activity play a role in CHF-related AF (40). In experimental dilated cardiomyopathy,
CHF increases SR Ca2+-load and reduces calsequestrin expression, thereby promoting
10
spontaneous SR Ca2+-release (41). Increased SR Ca2+-load also contributes to arrhythmogenic SR
Ca2+-releases in pAF patients (29). Altered RyR2 activity due to an inherited mutation in the
structural protein junctophilin-2 was shown to cause AF in patients with hypertrophic
cardiomyopathy, and a relative lack of junctophilin-2 due to increased RyR2 protein expression
may contribute to RyR2 dysfunction in pAF (42). The increased RyR2 protein expression in pAF
is at least in part due to a reduction in the inhibitory miRNA-106b-25 complex (43). Coronary
artery disease (CAD) is also an important risk factor for AF. Atrial ischemia promotes AF-
maintenance (44). In a dog model of chronic occlusive coronary-artery disease affecting the
atrium, frequent spontaneous atrial ectopy is associated with an increased incidence of atrial-
cardiomyocyte triggered activity (44). Triggered activity is likely due to spontaneous SR Ca2+-
release events and increased NCX-function in cardiomyocytes from the ischemic border-zone
(44).
In addition to the rare genetic variants in RyR2, two AF-promoting genetic variants have
been linked to DAD-mechanisms: 1) A mutation of the gene encoding the adapter protein
ankyrin-B (Long-QT Syndrome-4, LQTS4), which causes multiple proteins to be poorly-
addressed to their membrane-targets, altering Ca2+ handling and leading to DADs/triggered
activity (45, 46); and 2) A predicted loss-of-function SNP of the SLN gene encoding the Ca2+-
binding protein sarcolipin, which could increase SR Ca2+-load and thereby affect DAD-
susceptibility (47).
Beta-adrenoceptor activation phosphorylates RyR2, promoting diastolic SR Ca2+-release
events (47). Conditions that directly cause DAD-promoting abnormalities in Ca2+-handling may
require adrenergic stimulation to induce Ca2+-sparks and triggered activity (44). Spontaneous AF
paroxysms occur in dog models of autonomic hyperinnervation (48), with sympathovagal-
discharge preceding AF-paroxysms (49). Vagal activation promotes arrhythmogenesis by
11
reducing APD, allowing afterdepolarizations induced by adrenergic stimulation to induce ectopic
firing in pulmonary veins (50-52). Finally, although Ca2+-handling abnormalities and DADs
occur in atrial cardiomyocytes from pAF and cAF patients, atrial aftercontractions are less
frequent in multicellular atrial trabeculae from AF patients compared to sinus rhythm controls
(53). Similarly, high atrial rates alone may not produce proarrhythmic Ca2+-handling
abnormalities, but may even cause Ca2+-handling silencing to counteract the potentially cytotoxic
effects of chronically elevated intracellular Ca2+ (54). Thus, much more work is needed to
precisely define the role of abnormal Ca2+-handling in AF pathophysiology.
Molecular control of L-type Ca2+-current changes
AF (55), and indeed all very rapid atrial tachyarrhythmias (56), remodel atrial electrical
properties to promote AF initiation and maintenance (atrial tachycardia remodeling, ATR). A
major AF-promoting component of ATR is refractory-period reduction due to APD-abbreviation
(Figure 4B). Reduced depolarizing L-type Ca2+-current (ICa,L), along with increased repolarizing
inward-rectifier K+-currents, underlie ATR-induced APD-shortening (57-64). The molecular
basis of ICa,L-reduction in persistent AF is illustrated in Figure 7A. Rapid atrial activation
induces Ca2+-loading, activating Ca2+-calmodulin/calcineurin/NFAT signalling that causes down-
regulation of Cav1.2 -subunit mRNA (65-67). Other contributors to ICa,L downregulation may
include decreased expression of accessory 1, 2a, 2b, 3 and 22 subunits (59, 68, 69), Cav1.2
dephosphorylation via PP1 and type-2A (PP2A) protein phosphatases (31, 59, 70), increased
Cav1.2 -subunit s-nitrosylation (71), and metabolic stress (72). MicroRNAs are centrally
involved in cardiac remodeling (73) and recent work implicates increased miRNA-328 and miR-
21 in AF-promotion due to ICa,L-downregulation mediated by inhibition of translation and mRNA
destabilization (74, 75). In 82 patients with Brugada syndrome/Short-QT ECG phenotypes,
12
loss-of-function mutations of the CACNA1C and CACNB2 genes, encoding ICa,L α- and
β-subunits, were observed along with AF in individual patients (76). Patients with full-blown
Short-QT syndromes of various phenotypes have reduced APDs and are also predisposed to AF
(6).
Molecular control of K+-current and basis of AF-promoting alterations
Inward-rectifier K+-current enhancement promotes AF maintenance, by reducing APD
(favouring reentry) and stabilizing/accelerating arrhythmia-maintaining rotors by removing
voltage-dependent INa-inactivation through membrane-hyperpolarization (11, 77). Upregulation
of other K+-currents like two-pore K+-currents, small-conductance Ca2+-dependent K+-currents
and Kv1.1-mediated K+-currents may further contribute to APD-shortening in persistent AF (78-
80). Figure 7B shows how IK1 and IK,AChc are upregulated in persistent AF. IK1 increases because
of upregulation of the underlying Kir2.1 subunit (60-64, 69, 81-83) caused by reduced Kir2.1-
inhibitory microRNAs, like miR-1, miR-26 and miR-101 (83, 84). Kir2.1-dephosphorylation
(activation) via increased PP1 and PP2A function (31, 59, 70, 85) may also contribute.
Congestive HF upregulates fibroblast Kir2.1 expression and related IK1 currents, thereby causing
membrane hyperpolarization, increasing Ca2+ entry, and enhancing atrial fibroblast proliferation
(86), contributing to structural remodeling (see below). The changes in fibroblast IK1 are likely
mediated by miRNA-26a downregulation, positioning miRNA-26a as a common determinant of
both electrical and structural remodeling.
Agonist-induced muscarinic-receptor mediated IK,ACh activation is reduced in AF (60, 61).
This involves a reduction in Kir3.1 and Kir3.4 channel subunits and is associated with a loss of
physiological Na+-mediated IK,ACh activation (87). However, increased agonist-independent
(constitutive) IK,AChc is enhanced, both in dog models (60, 63, 82, 88) and in AF-patients (62, 64,
13
81, 89). Increased IK,AChc is due to greater IKACh-channel opening-probability, with no change in
single-channel conductance, kinetics or density (62, 82). A key role is played by altered IK,AChc
protein-kinase C (PKC) phosphorylation, with increased phosphorylation by stimulatory Ca2+-
dependent isoforms and reduced inhibitory classical Ca2+-independent isoform function (88, 89).
IK,AChc inhibition suppresses atrial tachyarrhythmias in ATR-preparations (63), suggesting that
IK,AChc contributes to ATR-induced arrhythmogenic remodeling.
The most common AF-promoting monogenic paradigm is accelerated atrial
repolarization due to gain-of-function K+-channel mutations (Table 1). Vagal enhancement is
known to promote clinical AF, and is central in some cases (90). IK,ACh hyperpolarizes atrial
cardiomyocytes and reduces APD in a spatially-heterogeneous way. Vagal enhancement strongly
favors AF initiation and persistence by facilitating the initiation and subsequent stability of
reentrant rotors (90). Kir3.4 knock-out strongly suppresses IK,ACh and prevents cholinergic AF
(91).
Molecular determinants of atrial conduction disturbances
Conduction abnormalities due to ion-channel dysfunction
Conduction-abnormalities favor reentry. Gap junctions are essential for efficient cell
coupling and conduction. There are discrepant results about AF-related atrial gap-junctional
remodeling in the literature (69, 92, 93). Some of the variability may be due to differences in
AF-duration, underlying heart disease and species-related factors (94). Spatially-heterogeneous
connexin-40 remodeling occurs in the goat model of electrically-maintained AF, consistent with
clinical data indicating that gene-variants that affect connexin-40 promoter function may
predispose to AF (95-97). Connexin-43 dephosphorylation/lateralization occurs in CHF, but
CHF-induced conduction-slowing and AF-promotion are unchanged with CHF-recovery, despite
14
disappearance of connexin-abnormalities (98). Recent evidence indicates that connexin-43 gene
transfer can improve conduction and suppress AF in porcine models, supporting the importance
of gap-junction protein remodeling in AF (99, 100).
Atrial ischemic disease causes localized conduction slowing, which allows for AF-
sustaining local reentry stabilized around a line of conduction-block (44). With acute ischemia,
gap-junction uncoupling predominates (101).
A number of AF-associated gene-variants affect ion-channels that control cardiac
conduction. GJA5 encodes connexin-40, an atrial-selective gap junction ion-channel. Connexin-
40 knockout causes conduction abnormalities and atrial arrhythmia-susceptibility (102). An AF-
causing GJA5 missense somatic mutation was identified in idiopathic AF-patients (96). GJA5-
promoter variants believed to decrease gene-transcription increase AF-susceptibility (95, 97,
103).
INa provides the energy for conduction and governs CV. INa-density decreases in canine
ATR, with corresponding decreases in SCN5A-subunit mRNA and protein (57). In humans with
AF, SCN5A mRNA expression appears unchanged (69). Atrial cardiomyocytes from AF-patients
showed a slightly reduced INa (104).
Cardiac Na+-channel gene (SCN5A) loss-of-function mutations cause AF, presumably via
reentry-promoting conduction-slowing (Table 1). SCN5A mutations were initially associated
with AF in a family presenting a complex and variable phenotype including dilated
cardiomyopathy, AF, sinus node dysfunction, and conduction defects (105). SCN5A mutations
and SNPs were subsequently found in idiopathic-AF subjects (106, 107). Loss-of-function
SCN5A mutations are the most common cause of Brugada Syndrome (108), which typically
presents as VF/sudden death, but can also cause AF (96). Recently, mutations in SCN10A, as
15
well as in Na+-channel -subunits like SCN1B, 2B and 3B, have been implicated in AF (109-
112).
Structural remodeling
Atrial fibrosis plays an important role in AF of different etiologies (13, 44, 113, 114).
The underlying signalling pathways and clinical manifestations have recently been reviewed
(115). The development of atrial fibrosis is likely determined by multiple signals acting
simultaneously. Cardiomyocytes and fibroblasts interact extensively through autocrine and
paracrine factors, and possibly electrically as well (13, 116). Fibroblasts produce extracellular-
matrix (ECM) proteins and mediators that affect cardiomyocyte phenotype, whereas
cardiomyocytes generate products like reactive oxygen species (ROS), platelet-derived growth
factor (PDGF), transforming growth-factor-β (TGF-β) and connective tissue growth factor
(CTGF) that modulate fibroblast function.
Angiotensin-II (AT-II) plays an important role in AF (117), likely in large measure via
fibroblast-modulation. AT-II type-1 receptors (AT1Rs) promote fibrosis via enhanced actions of
TGF-β, Smad2/3, Smad4, Arkadia, and activated extracellular signal-regulated (ERK) mitogen-
activated protein-kinase (MAPK) (118). Arkadia promotes ubiquination and removal of Smad7,
thereby increasing TGF-β signaling by removing Smad7-antagonism (119). In addition, AT1Rs
act through Shc/Grb2/SOS to activate Ras, which enhances MAPK-phosphorylation (120), and
through phospholipase-C (PLC). PLC breaks down phosphatidylinositol 4,5-bisphosphate (PIP2),
yielding diacylglycerol (DAG) and inositol 1,4,5-trisphosphate (IP3). DAG activates PKC and
IP3 mobilizes intracellular Ca2+, both actions contributing to remodeling. The JAK/STAT
pathway, also AT1R-sensitive, activates transcription-factors such as AP-1 and NF-κB, which
cause further cardiomyocyte remodeling. AT2R-activation counters AT1R-mediated MAPK-
16
activation by enhancing dephosphorylation via PP2A and phosphotyrosine phosphatase (PTP)
(120).
TGF-β1 is a key player in cardiac fibrosis. It is secreted by both fibroblasts and
cardiomyocytes (121). Overactivity of cardiac TGF-β1 causes atrial-selective fibrosis,
conduction abnormalities, and AF promotion (122). TGF-β1 mediates AT-II effects in both
paracrine and autocrine fashions (123). TGF-β1 acts through SMADs to activate fibroblasts and
enhance collagen production (13, 116, 119). Rapidly-firing atrial cardiomyocytes produce
Ang-II and ROS, acting via enhanced TGF-β production to differentiate cardiac fibroblasts into
ECM-secreting myofibroblasts (124, 125). Progressive fibrosis likely contributes to conduction
disturbances that make long-lasting AF very difficult to treat (14).
PDGF stimulates fibroblast proliferation and differentiation (13, 116, 122). PDGF-
receptors contain two transmembrane-domains that dimerize upon stimulation and then activate
an internal tyrosine-kinase. Tyrosine-kinase autophosphorylation of PDGF-receptors induces
Ras/MEK1/2, MAPK, JAK/STAT, and PLC signaling. PDGF-overexpression induces cardiac
fibrosis and dysfunction (126). Atrial-selective PDGF expression and action may contribute to
the greater fibrotic responses typically seen for atria versus ventricles (121).
Connective-tissue growth factor (CTGF) lies downstream to TGF-β1 and AT-II in
profibrotic-signaling pathways. CTGF activates fibroblasts through Src-kinase and MAPKs
(127). CTGF has emerged as a potentially-central player in atrial structural remodeling (128-
130).
MicroRNAs appear to contribute importantly to atrial structural remodeling. MiR-29
inhibits collagen-gene expression (131) and its downregulation likely contributes to atrial fibrosis
in CHF (132). MiR-30 and miR-133 are also downregulated in CHF, suppress CTGF-translation
(133), and may participate in atrial fibrosis (134). In contrast to miR-29, miR-20 and miR-133,
17
atrial miR-21 expression increases in CHF (134). MiR-21 targets the Sprouty-1 (Spry-1) gene,
which promotes fibroblast MAPK-phosphorylation and enhances fibroblast survival (135)
Although the significance of this finding for ventricular remodeling has been disputed (136),
more recent work strongly suggests an important role of miR-21 upregulation in CHF-associated
atrial fibrosis and AF-promotion (137).
SNPs in genes determining atrial structural integrity, inflammation and neurohumoral
control, have been associated with AF by conventional approaches. Examples include genes
encoding angiotensin-converting-enzyme (ACE) (138, 139), matrix metalloproteinase-2
(MMP2), and interleukin-10 (140). However, these have not been replicated in hypothesis-free
large-scale population studies. GWASs have implicated SNPs on chromosome 4q25, with
PITX2c being the closest potential target gene, and an SNP in chromosome 16q22 near the zinc
finger homeobox 3 transcription factor gene (ZFHX3) (50, 51, 141). PITX2c is involved in
cardiac development, particularly sidedness and pulmonary-vein aspects (142, 143) implicating
possible structural abnormalities as the way in which it may be involved in AF. ZFHX3 is a
tumor-suppressor (144, 145), which induces expression of PDGF-receptors and protects against
oxidant stress (146), also suggesting structural-remodeling as a potential mechanism of AF-
promotion.
Atrial Ca2+-handling abnormalities may also contribute to reentry-promoting structural
remodeling. Increased fibroblast Ca2+-entry through TRP-channels promotes fibroblast
proliferation and structural remodeling (147). Genetic inhibition of RyR2-hyperphosphorylation
prevents excessive SR Ca2+-leak, suppresses atrial dilatation, and reduces atrial conduction
abnormalities in mice with cardiac-restricted overexpression of a repressor form of the cAMP-
response element modulator (148).
18
Future directions
The largest and most important challenge related to the molecular basis of AF is translating our
increased knowledge into practical clinical applications. It is widely hoped that targeting the
molecular mechanisms of AF will permit the development of novel, safer and more specific
treatment-approaches. Recent advances suggest that biological therapies capable of specifically
modifying the AF-substrate may be in sight(149). The challenges in moving from scientific
discovery to practical therapeutic innovation in this area are substantial, but the history of this
field suggests that conceptual advances do ultimately result in tangible improvements in patient
management options (1).
Acknowledgements
The authors thank Jennifer Bacchi for excellent secretarial help.
Funding
Supported by the Canadian Institutes of Health Research (MGP6957 and MOP44365), the
Quebec Heart and Stroke Foundation, the Foundation Leducq (European-North American Atrial
Fibrillation Research Alliance, ENAFRA, grant 07CVD03), the German Federal Ministry of
Education and Research through Atrial Fibrillation Competence Network (grant 01Gi0204) and
DZHK (German Centre for Cardiovascular Research), the Deutsche Forschungsgemeinschaft (Do
769/1-3), the European Union (European Network for Translational Research in Atrial
Fibrillation, EUTRAF, grant 261057). NIH-NHLBI grants (R01-HL089598, R01-HL091947,
R01-HL117641, R41-HL129570), and the American Heart Association (13EIA14560061).
19
20
References
1. Heijman, J., Algalarrondo, V., Voigt, N., Wehrens, X.H.T., and Nattel, S. The value of
basic research insights into atrial fibrillation mechanisms as a guide to therapeutic
innovation: A critical analysis. Cardiovasc Res. 2015:in revision.
2. Nattel, S. From guidlines to Bench: Implications of unresolved clinical issues for basic
investigations of atrial fibrillation mechanisms. Can J Cardiol. 2011;27(1):19-26.
3. Darbar, D. The Role of Pharmacogenetics in Atrial Fibrillation Therapeutics - Is
Personalized Therapy in Sight? J Cardiovasc Pharmacol. 2015.
4. Olesen, M.S., Nielsen, M.W., Haunso, S., and Svendsen, J.H. Atrial fibrillation: the role
of common and rare genetic variants. Eur J Hum Genet. 2014;22(3):297-306.
5. Sinner, M.F., Tucker, N.R., Lunetta, K.L., Ozaki, K., Smith, J.G., Trompet, S., Bis, J.C.,
Lin, H., Chung, M.K., Nielsen, J.B., Lubitz, S.A., Krijthe, B.P., Magnani, J.W., Ye, J.,
Gollob, M.H., Tsunoda, T., Muller-Nurasyid, M., Lichtner, P., Peters, A., Dolmatova, E.,
Kubo, M., Smith, J.D., Psaty, B.M., Smith, N.L., Jukema, J.W., Chasman, D.I., Albert,
C.M., Ebana, Y., Furukawa, T., Macfarlane, P.W., Harris, T.B., Darbar, D., Dorr, M.,
Holst, A.G., Svendsen, J.H., Hofman, A., Uitterlinden, A.G., Gudnason, V., Isobe, M.,
Malik, R., Dichgans, M., Rosand, J., Van Wagoner, D.R., Consortium, M., Consortium,
A.F., Benjamin, E.J., Milan, D.J., Melander, O., Heckbert, S.R., Ford, I., Liu, Y., Barnard,
J., Olesen, M.S., Stricker, B.H., Tanaka, T., Kaab, S., and Ellinor, P.T. Integrating
genetic, transcriptional, and functional analyses to identify 5 novel genes for atrial
fibrillation. Circulation. 2014;130(15):1225-1235.
6. Tucker, N.R., and Ellinor, P.T. Emerging directions in the genetics of atrial fibrillation.
Circ Res. 2014;114(9):1469-1482.
7. Mukamal, K.J., Tolstrup, J.S., Friberg, J., Jensen, G., and Gronbaek, M. Alcohol
consumption and risk of atrial fibrillation in men and women: the Copenhagen City Heart
Study. Circulation. 2005;112(12):1736-1742.
8. Auer, J., Scheibner, P., Mische, T., Langsteger, W., Eber, O., and Eber, B. Subclinical
hyperthyroidism as a risk factor for atrial fibrillation. Am Heart J. 2001;142(5):838-842.
9. Schoonderwoerd, B.A., Smit, M.D., Pen, L., and Van Gelder, I.C. New risk factors for
atrial fibrillation: causes of 'not-so-lone atrial fibrillation'. Europace. 2008;10(6):668-673.
10. Chou, C.C., and Chen, P.S. New concepts in atrial fibrillation: neural mechanisms and
calcium dynamics. Cardiol Clin. 2009;27(1):35-43, viii.
21
11. Nattel, S., Burstein, B., and Dobrev, D. Atrial remodeling and atrial fibrillation:
mechanisms and implications. Circ Arrhythm Electrophysiol. 2008;1(1):62-73.
12. Berenfeld, O., Zaitsev, A.V., Mironov, S.F., Pertsov, A.M., and Jalife, J. Frequency-
dependent breakdown of wave propagation into fibrillatory conduction across the
pectinate muscle network in the isolated sheep right atrium. Circ Res. 2002;90(11):1173-
1180.
13. Burstein, B., and Nattel, S. Atrial fibrosis: mechanisms and clinical relevance in atrial
fibrillation. J Am Coll Cardiol. 2008;51(8):802-809.
14. de Groot, N.M., Houben, R.P., Smeets, J.L., Boersma, E., Schotten, U., Schalij, M.J.,
Crijns, H., and Allessie, M.A. Electropathological substrate of longstanding persistent
atrial fibrillation in patients with structural heart disease: epicardial breakthrough.
Circulation. 2010;122(17):1674-1682.
15. Cuculich, P.S., Wang, Y., Lindsay, B.D., Faddis, M.N., Schuessler, R.B., Damiano, R.J.,
Jr., Li, L., and Rudy, Y. Noninvasive characterization of epicardial activation in humans
with diverse atrial fibrillation patterns. Circulation. 2010;122(14):1364-1372.
16. Haissaguerre, M., Hocini, M., Denis, A., Shah, A.J., Komatsu, Y., Yamashita, S., Daly,
M., Amraoui, S., Zellerhoff, S., Picat, M.Q., Quotb, A., Jesel, L., Lim, H., Ploux, S.,
Bordachar, P., Attuel, G., Meillet, V., Ritter, P., Derval, N., Sacher, F., Bernus, O.,
Cochet, H., Jais, P., and Dubois, R. Driver domains in persistent atrial fibrillation.
Circulation. 2014;130(7):530-538.
17. Lou, Q., Belevych, A.E., Radwanski, P.B., Liu, B., Kalyanasundaram, A., Knollmann,
B.C., Fedorov, V.V., and Gyorke, S. Alternating membrane potential/calcium interplay
underlies repetitive focal activity in a genetic model of calcium-dependent atrial
arrhythmias. J Physiol. 2015;593(6):1443-1458.
18. MacLennan, D.H., and Chen, S.R. Store overload-induced Ca2+ release as a triggering
mechanism for CPVT and MH episodes caused by mutations in RYR and CASQ genes. J
Physiol. 2009;587(Pt 13):3113-3115.
19. Comtois, P., Kneller, J., and Nattel, S. Of circles and spirals: bridging the gap between the
leading circle and spiral wave concepts of cardiac reentry. Europace. 2005;7 Suppl 2:10-
20.
20. Mahida, S. Transcription factors and atrial fibrillation. Cardiovasc Res. 2014;101(2):194-
202.
21. Nattel, S., and Dobrev, D. The multidimensional role of calcium in atrial fibrillation
pathophysiology: mechanistic insights and therapeutic opportunities. Eur Heart J.
2012;33(15):1870-1877.
22
22. Tadevosyan, A., Vaniotis, G., Allen, B.G., Hebert, T.E., and Nattel, S. G protein-coupled
receptor signalling in the cardiac nuclear membrane: evidence and possible roles in
physiological and pathophysiological function. J Physiol. 2012;590(Pt 6):1313-1330.
23. Ruan, Z., Sun, X., Sheng, H., and Zhu, L. Long non-coding RNA expression profile in
atrial fibrillation. Int J Clin Exp Pathol. 2015;8(7):8402-8410.
24. Voigt, N., Li, N., Wang, Q., Wang, W., Trafford, A.W., Abu-Taha, I., Sun, Q., Wieland,
T., Ravens, U., Nattel, S., Wehrens, X.H., and Dobrev, D. Enhanced sarcoplasmic
reticulum Ca2+ leak and increased Na+-Ca2+ exchanger function underlie delayed
afterdepolarizations in patients with chronic atrial fibrillation. Circulation.
2012;125(17):2059-2070.
25. Hove-Madsen, L., Llach, A., Bayes-Genis, A., Roura, S., Rodriguez Font, E., Aris, A.,
and Cinca, J. Atrial fibrillation is associated with increased spontaneous calcium release
from the sarcoplasmic reticulum in human atrial myocytes. Circulation.
2004;110(11):1358-1363.
26. Vest, J.A., Wehrens, X.H., Reiken, S.R., Lehnart, S.E., Dobrev, D., Chandra, P., Danilo,
P., Ravens, U., Rosen, M.R., and Marks, A.R. Defective cardiac ryanodine receptor
regulation during atrial fibrillation. Circulation. 2005;111(16):2025-2032.
27. Liang, X., Xie, H., Zhu, P.H., Hu, J., Zhao, Q., Wang, C.S., and Yang, C. Ryanodine
receptor-mediated Ca2+ events in atrial myocytes of patients with atrial fibrillation.
Cardiology. 2008;111(2):102-110.
28. Neef, S., Dybkova, N., Sossalla, S., Ort, K.R., Fluschnik, N., Neumann, K., Seipelt, R.,
Schondube, F.A., Hasenfuss, G., and Maier, L.S. CaMKII-dependent diastolic SR Ca2+
leak and elevated diastolic Ca2+ levels in right atrial myocardium of patients with atrial
fibrillation. Circ Res. 2010;106(6):1134-1144.
29. Voigt, N., Heijman, J., Wang, Q., Chiang, D.Y., Li, N., Karck, M., Wehrens, X.H., Nattel,
S., and Dobrev, D. Cellular and molecular mechanisms of atrial arrhythmogenesis in
patients with paroxysmal atrial fibrillation. Circulation. 2014;129(2):145-156.
30. Chelu, M.G., Sarma, S., Sood, S., Wang, S., van Oort, R.J., Skapura, D.G., Li, N.,
Santonastasi, M., Muller, F.U., Schmitz, W., Schotten, U., Anderson, M.E., Valderrabano,
M., Dobrev, D., and Wehrens, X.H. Calmodulin kinase II-mediated sarcoplasmic
reticulum Ca2+ leak promotes atrial fibrillation in mice. J Clin Invest. 2009;119(7):1940-
1951.
31. El-Armouche, A., Boknik, P., Eschenhagen, T., Carrier, L., Knaut, M., Ravens, U., and
Dobrev, D. Molecular determinants of altered Ca2+ handling in human chronic atrial
fibrillation. Circulation. 2006;114(7):670-680.
23
32. Greiser, M., Neuberger, H.R., Harks, E., El-Armouche, A., Boknik, P., de Haan, S.,
Verheyen, F., Verheule, S., Schmitz, W., Ravens, U., Nattel, S., Allessie, M.A., Dobrev,
D., and Schotten, U. Distinct contractile and molecular differences between two goat
models of atrial dysfunction: AV block-induced atrial dilatation and atrial fibrillation. J
Mol Cell Cardiol. 2009;46(3):385-394.
33. Chiang, D.Y., Lebesgue, N., Beavers, D.L., Alsina, K.M., Damen, J.M., Voigt, N.,
Dobrev, D., Wehrens, X.H., and Scholten, A. Alterations in the interactome of
serine/threonine protein phosphatase type-1 in atrial fibrillation patients. J Am Coll
Cardiol. 2015;65(2):163-173.
34. Sood, S., Chelu, M.G., van Oort, R.J., Skapura, D., Santonastasi, M., Dobrev, D., and
Wehrens, X.H. Intracellular calcium leak due to FKBP12.6 deficiency in mice facilitates
the inducibility of atrial fibrillation. Heart Rhythm. 2008;5(7):1047-1054.
35. Li, N., Wang, T., Wang, W., Cutler, M.J., Wang, Q., Voigt, N., Rosenbaum, D.S.,
Dobrev, D., and Wehrens, X.H. Inhibition of CaMKII phosphorylation of RyR2 prevents
induction of atrial fibrillation in FKBP12.6 knockout mice. Circ Res. 2012;110(3):465-
470.
36. Purohit, A., Swaminathan, P.D., Chen, B., Voigt, N., Koval, O.M., Luczak, E.D., Gao, Z.,
Li, N., Guan, X., He, B.J., El-Accaoui, R.N., Yang, J., Kutschke, W., Singh, M.V., Weiss,
R.M., Wehrens, X.H., Song, L.S., Dobrev, D., and Anerson, M.E. Angiotensin II
Promotes Atrial Fibrillation in Mice by CaMKII Oxidation. Circulation.
2011;124(Suppl):A14037.
37. Lenaerts, I., Bito, V., Heinzel, F.R., Driesen, R.B., Holemans, P., D'Hooge, J.,
Heidbuchel, H., Sipido, K.R., and Willems, R. Ultrastructural and functional remodeling
of the coupling between Ca2+ influx and sarcoplasmic reticulum Ca2+ release in right atrial
myocytes from experimental persistent atrial fibrillation. Circ Res. 2009;105(9):876-885.
38. Dobrev, D., Voigt, N., and Wehrens, X.H. The ryanodine receptor channel as a molecular
motif in atrial fibrillation: pathophysiological and therapeutic implications. Cardiovasc
Res. 2012;89(4):734-743.
39. Zhao, Z.H., Zhang, H.C., Xu, Y., Zhang, P., Li, X.B., Liu, Y.S., and Guo, J.H. Inositol-
1,4,5-trisphosphate and ryanodine-dependent Ca2+ signaling in a chronic dog model of
atrial fibrillation. Cardiology. 2007;107(4):269-276.
40. Ryu, K., Shroff, S.C., Sahadevan, J., Martovitz, N.L., Khrestian, C.M., and Stambler, B.S.
Mapping of atrial activation during sustained atrial fibrillation in dogs with rapid
ventricular pacing induced heart failure: evidence for a role of driver regions. J
Cardiovasc Electrophysiol. 2005;16(12):1348-1358.
24
41. Yeh, Y.H., Wakili, R., Qi, X.Y., Chartier, D., Boknik, P., Kaab, S., Ravens, U., Coutu, P.,
Dobrev, D., and Nattel, S. Calcium-handling abnormalities underlying atrial
arrhythmogenesis and contractile dysfunction in dogs with congestive heart failure. Circ
Arrhythm Electrophysiol. 2008;1(2):93-102.
42. Beavers, D.L., Wang, W., Ather, S., Voigt, N., Garbino, A., Dixit, S.S., Landstrom, A.P.,
Li, N., Wang, Q., Olivotto, I., Dobrev, D., Ackerman, M.J., and Wehrens, X.H. Mutation
E169K in junctophilin-2 causes atrial fibrillation due to impaired RyR2 stabilization. J
Am Coll Cardiol. 2013;62(21):2010-2019.
43. Chiang, D.Y., Kongchan, N., Beavers, D.L., Alsina, K.M., Voigt, N., Neilson, J.R., Jakob,
H., Martin, J.F., Dobrev, D., Wehrens, X.H., and Li, N. Loss of microRNA-106b-25
cluster promotes atrial fibrillation by enhancing ryanodine receptor type-2 expression and
calcium release. Circ Arrhythm Electrophysiol. 2014;7(6):1214-1222.
44. Nishida, K., Qi, X.Y., Wakili, R., Comptois, P., Chartier, D., Harada, M., Iwasaki, Y.,
Romeo, P., Maguy, A., Dobrev, D., Michael, G., Talajic, M., and Nattel, S. Mechanisms
of Atrial Tachyarrhythmias Associated with Coronary Occlusion in a Chronic Canine
Model. Circulation. 2011;123(2):137-146.
45. Cunha, S.R., Hund, T.J., Hashemi, S., Voigt, N., Li, N., Wright, P., Koval, O., Li, J.,
Gudmundsson, H., Gumina, R.J., Karck, M., Schott, J.J., Probst, V., Le Marec, H.,
Anderson, M.E., Dobrev, D., Wehrens, X.H., and Mohler, P.J. Defects in ankyrin-based
membrane protein targeting pathways underlie atrial fibrillation. Circulation.
2011;124(11):1212-1222.
46. Mohler, P.J., Schott, J.J., Gramolini, A.O., Dilly, K.W., Guatimosim, S., duBell, W.H.,
Song, L.S., Haurogne, K., Kyndt, F., Ali, M.E., Rogers, T.B., Lederer, W.J., Escande, D.,
Le Marec, H., and Bennett, V. Ankyrin-B mutation causes type 4 long-QT cardiac
arrhythmia and sudden cardiac death. Nature. 2003;421(6923):634-639.
47. Nyberg, M.T., Stoevring, B., Behr, E.R., Ravn, L.S., McKenna, W.J., and Christiansen,
M. The variation of the sarcolipin gene (SLN) in atrial fibrillation, long QT syndrome and
sudden arrhythmic death syndrome. Clin Chim Acta. 2007;375(1-2):87-91.
48. Tan, A.Y., Zhou, S., Ogawa, M., Song, J., Chu, M., Li, H., Fishbein, M.C., Lin, S.F.,
Chen, L.S., and Chen, P.S. Neural mechanisms of paroxysmal atrial fibrillation and
paroxysmal atrial tachycardia in ambulatory canines. Circulation. 2008;118(9):916-925.
49. Choi, E.K., Shen, M.J., Han, S., Kim, D., Hwang, S., Sayfo, S., Piccirillo, G., Frick, K.,
Fishbein, M.C., Hwang, C., Lin, S.F., and Chen, P.S. Intrinsic cardiac nerve activity and
paroxysmal atrial tachyarrhythmia in ambulatory dogs. Circulation. 2010;121(24):2615-
2623.
25
50. Burashnikov, A., and Antzelevitch, C. Reinduction of atrial fibrillation immediately after
termination of the arrhythmia is mediated by late phase 3 early afterdepolarization-
induced triggered activity. Circulation. 2003;107(18):2355-2360.
51. Patterson, E., Po, S.S., Scherlag, B.J., and Lazzara, R. Triggered firing in pulmonary
veins initiated by in vitro autonomic nerve stimulation. Heart Rhythm. 2005;2(6):624-631.
52. Chou, C.C., Nguyen, B.L., Tan, A.Y., Chang, P.C., Lee, H.L., Lin, F.C., Yeh, S.J.,
Fishbein, M.C., Lin, S.F., Wu, D., Wen, M.S., and Chen, P.S. Intracellular calcium
dynamics and acetylcholine-induced triggered activity in the pulmonary veins of dogs
with pacing-induced heart failure. Heart Rhythm. 2008;5(8):1170-1177.
53. Christ, T., Rozmaritsa, N., Engel, A., Berk, E., Knaut, M., Metzner, K., Canteras, M.,
Ravens, U., and Kaumann, A. Arrhythmias, elicited by catecholamines and serotonin,
vanish in human chronic atrial fibrillation. Proc Natl Acad Sci U S A.
2014;111(30):11193-11198.
54. Greiser, M., Kerfant, B.G., Williams, G.S., Voigt, N., Harks, E., Dibb, K.M., Giese, A.,
Meszaros, J., Verheule, S., Ravens, U., Allessie, M.A., Gammie, J.S., van der Velden, J.,
Lederer, W.J., Dobrev, D., and Schotten, U. Tachycardia-induced silencing of subcellular
Ca2+ signaling in atrial myocytes. J Clin Invest. 2014;124(11):4759-4772.
55. Wijffels, M.C., Kirchhof, C.J., Dorland, R., and Allessie, M.A. Atrial fibrillation begets
atrial fibrillation. A study in awake chronically instrumented goats. Circulation.
1995;92(7):1954-1968.
56. Shiroshita-Takeshita, A., Mitamura, H., Ogawa, S., and Nattel, S. Rate-dependence of
atrial tachycardia effects on atrial refractoriness and atrial fibrillation maintenance.
Cardiovasc Res. 2009;81(1):90-97.
57. Yue, L., Feng, J., Gaspo, R., Li, G.R., Wang, Z., and Nattel, S. Ionic remodeling
underlying action potential changes in a canine model of atrial fibrillation. Circ Res.
1997;81(4):512-525.
58. Van Wagoner, D.R., Pond, A.L., Lamorgese, M., Rossie, S.S., McCarthy, P.M., and
Nerbonne, J.M. Atrial L-type Ca2+ currents and human atrial fibrillation. Circ Res.
1999;85(5):428-436.
59. Christ, T., Boknik, P., Wohrl, S., Wettwer, E., Graf, E.M., Bosch, R.F., Knaut, M.,
Schmitz, W., Ravens, U., and Dobrev, D. L-type Ca2+ current downregulation in chronic
human atrial fibrillation is associated with increased activity of protein phosphatases.
Circulation. 2004;110(17):2651-2657.
60. Ehrlich, J.R., Cha, T.J., Zhang, L., Chartier, D., Villeneuve, L., Hebert, T.E., and Nattel,
S. Characterization of a hyperpolarization-activated time-dependent potassium current in
26
canine cardiomyocytes from pulmonary vein myocardial sleeves and left atrium. J
Physiol. 2004;557(Pt 2):583-597.
61. Dobrev, D., Graf, E., Wettwer, E., Himmel, H.M., Hala, O., Doerfel, C., Christ, T.,
Schuler, S., and Ravens, U. Molecular basis of downregulation of G-protein-coupled
inward rectifying K+ current (IK,ACh) in chronic human atrial fibrillation: decrease in
GIRK4 mRNA correlates with reduced IK,ACh and muscarinic receptor-mediated
shortening of action potentials. Circulation. 2001;104(21):2551-2557.
62. Dobrev, D., Friedrich, A., Voigt, N., Jost, N., Wettwer, E., Christ, T., Knaut, M., and
Ravens, U. The G protein-gated potassium current IK,ACh is constitutively active in
patients with chronic atrial fibrillation. Circulation. 2005;112(24):3697-3706.
63. Cha, T.J., Ehrlich, J.R., Chartier, D., Qi, X.Y., Xiao, L., and Nattel, S. Kir3-based inward
rectifier potassium current: potential role in atrial tachycardia remodeling effects on atrial
repolarization and arrhythmias. Circulation. 2006;113(14):1730-1737.
64. Voigt, N., Friedrich, A., Bock, M., Wettwer, E., Christ, T., Knaut, M., Strasser, R.H.,
Ravens, U., and Dobrev, D. Differential phosphorylation-dependent regulation of
constitutively active and muscarinic receptor-activated IK,ACh channels in patients with
chronic atrial fibrillation. Cardiovasc Res. 2007;74(3):426-437.
65. Yue, L., Melnyk, P., Gaspo, R., Wang, Z., and Nattel, S. Molecular mechanisms
underlying ionic remodeling in a dog model of atrial fibrillation. Circ Res.
1999;84(7):776-784.
66. Qi, X.Y., Yeh, Y.H., Xiao, L., Burstein, B., Maguy, A., Chartier, D., Villeneuve, L.R.,
Brundel, B.J., Dobrev, D., and Nattel, S. Cellular signaling underlying atrial tachycardia
remodeling of L-type calcium current. Circ Res. 2008;103(8):845-854.
67. Sun, H., Chartier, D., Leblanc, N., and Nattel, S. Intracellular calcium changes and
tachycardia-induced contractile dysfunction in canine atrial myocytes. Cardiovasc Res.
2001;49(4):751-761.
68. Bosch, R.F., Scherer, C.R., Rub, N., Wohrl, S., Steinmeyer, K., Haase, H., Busch, A.E.,
Seipel, L., and Kuhlkamp, V. Molecular mechanisms of early electrical remodeling:
transcriptional downregulation of ion channel subunits reduces ICa,L and Ito in rapid atrial
pacing in rabbits. J Am Coll Cardiol. 2003;41(5):858-869.
69. Gaborit, N., Steenman, M., Lamirault, G., Le Meur, N., Le Bouter, S., Lande, G., Leger,
J., Charpentier, F., Christ, T., Dobrev, D., Escande, D., Nattel, S., and Demolombe, S.
Human atrial ion channel and transporter subunit gene-expression remodeling associated
with valvular heart disease and atrial fibrillation. Circulation. 2005;112(4):471-481.
27
70. Greiser, M., Halaszovich, C.R., Frechen, D., Boknik, P., Ravens, U., Dobrev, D.,
Luckhoff, A., and Schotten, U. Pharmacological evidence for altered src kinase regulation
of ICa,L in patients with chronic atrial fibrillation. Naunyn Schmiedebergs Arch
Pharmacol. 2007;375(6):383-392.
71. Carnes, C.A., Janssen, P.M., Ruehr, M.L., Nakayama, H., Nakayama, T., Haase, H.,
Bauer, J.A., Chung, M.K., Fearon, I.M., Gillinov, A.M., Hamlin, R.L., and Van Wagoner,
D.R. Atrial glutathione content, calcium current, and contractility. J Biol Chem.
2007;282(38):28063-28073.
72. Harada, M., Tadevosyan, A., Qi, X., Xiao, J., Liu, T., Voigt, N., Karck, M., Kamler, M.,
Kodama, I., Murohara, T., Dobrev, D., and Nattel, S. Atrial Fibrillation Activates AMP-
Dependent Protein Kinase and its Regulation of Cellular Calcium Handling: Potential
Role in Metabolic Adaptation and Prevention of Progression. J Am Coll Cardiol.
2015;66(1):47-58.
73. Luo, X., Yang, B., and Nattel, S. MicroRNAs and atrial fibrillation: mechanisms and
translational potential. Nat Rev Cardiol. 2015;12(2):80-90.
74. Lu, Y., Zhang, Y., Wang, N., Pan, Z., Gao, X., Zhang, F., Shan, H., Luo, X., Bai, Y., Sun,
L., Song, W., Xu, C., Wang, Z., and Yang, B. MicroRNA-328 contributes to adverse
electrical remodeling in atrial fibrillation. Circulation. 2010;122(23):2378-2387.
75. Barana, A., Matamoros, M., Dolz-Gaiton, P., Perez-Hernandez, M., Amoros, I., Nunez,
M., Sacristan, S., Pedraz, A., Pinto, A., Fernandez-Aviles, F., Tamargo, J., Delpon, E.,
and Caballero, R. Chronic atrial fibrillation increases microRNA-21 in human atrial
myocytes decreasing L-type calcium current. Circ Arrhythm Electrophysiol.
2014;7(5):861-868.
76. Antzelevitch, C., Pollevick, G.D., Cordeiro, J.M., Casis, O., Sanguinetti, M.C., Aizawa,
Y., Guerchicoff, A., Pfeiffer, R., Oliva, A., Wollnik, B., Gelber, P., Bonaros, E.P., Jr.,
Burashnikov, E., Wu, Y., Sargent, J.D., Schickel, S., Oberheiden, R., Bhatia, A., Hsu,
L.F., Haissaguerre, M., Schimpf, R., Borggrefe, M., and Wolpert, C. Loss-of-function
mutations in the cardiac calcium channel underlie a new clinical entity characterized by
ST-segment elevation, short QT intervals, and sudden cardiac death. Circulation.
2007;115(4):442-449.
77. Pandit, S.V., Berenfeld, O., Anumonwo, J.M., Zaritski, R.M., Kneller, J., Nattel, S., and
Jalife, J. Ionic determinants of functional reentry in a 2-D model of human atrial cells
during simulated chronic atrial fibrillation. Biophys J. 2005;88(6):3806-3821.
78. Glasscock, E., Voigt, N., McCauley, M.D., Sun, Q., Li, N., Chiang, D.Y., Zhou, X.B.,
Molina, C.E., Thomas, D., Schmidt, C., Skapura, D.G., Noebels, J.L., Dobrev, D., and
Wehrens, X.H. Expression and function of Kv1.1 potassium channels in human atria from
patients with atrial fibrillation. Basic Res Cardiol. 2015;110(5):505.
28
79. Qi, X.Y., Diness, J.G., Brundel, B.J., Zhou, X.B., Naud, P., Wu, C.T., Huang, H., Harada,
M., Aflaki, M., Dobrev, D., Grunnet, M., and Nattel, S. Role of small-conductance
calcium-activated potassium channels in atrial electrophysiology and fibrillation in the
dog. Circulation. 2014;129(4):430-440.
80. Schmidt, C., Wiedmann, F., Voigt, N., Zhou, X.B., Heijman, J., Lang, S., Albert, V.,
Kallenberger, S., Ruhparwar, A., Szabo, G., Kallenbach, K., Karck, M., Borggrefe, M.,
Biliczki, P., Ehrlich, J.R., Baczko, I., Lugenbiel, P., Schweizer, P.A., Donner, B.C.,
Katus, H.A., Dobrev, D., and Thomas, D. Upregulation of K2P3.1 K+ Current Causes
Action Potential Shortening in Patients With Chronic Atrial Fibrillation. Circulation.
2015;132(2):82-92.
81. Voigt, N., Trausch, A., Knaut, M., Matschke, K., Varro, A., Van Wagoner, D.R., Nattel,
S., Ravens, U., and Dobrev, D. Left-to-right atrial inward rectifier potassium current
gradients in patients with paroxysmal versus chronic atrial fibrillation. Circ Arrhythm
Electrophysiol. 2010;3(5):472-480.
82. Voigt, N., Maguy, A., Yeh, Y.H., Qi, X., Ravens, U., Dobrev, D., and Nattel, S. Changes
in IK,ACh single-channel activity with atrial tachycardia remodelling in canine atrial
cardiomyocytes. Cardiovasc Res. 2008;77(1):35-43.
83. Girmatsion, Z., Biliczki, P., Bonauer, A., Wimmer-Greinecker, G., Scherer, M., Moritz,
A., Bukowska, A., Goette, A., Nattel, S., Hohnloser, S.H., and Ehrlich, J.R. Changes in
microRNA-1 expression and IK1 up-regulation in human atrial fibrillation. Heart Rhythm.
2009;6(12):1802-1809.
84. Luo, X., Pan, Z., Shan, H., Xiao, J., Sun, X., Wang, N., Lin, H., Xiao, L., Maguy, A., Qi,
X.Y., Li, Y., Gao, X., Dong, D., Zhang, Y., Bai, Y., Ai, J., Sun, L., Lu, H., Luo, X.Y.,
Wang, Z., Lu, Y., Yang, B., and Nattel, S. MicroRNA-26 governs profibrillatory inward-
rectifier potassium current changes in atrial fibrillation. J Clin Invest. 2013;123(5):1939-
1951.
85. Karle, C.A., Zitron, E., Zhang, W., Wendt-Nordahl, G., Kathofer, S., Thomas, D., Gut, B.,
Scholz, E., Vahl, C.F., Katus, H.A., and Kiehn, J. Human cardiac inwardly-rectifying K+
channel Kir2.1b is inhibited by direct protein kinase C-dependent regulation in human
isolated cardiomyocytes and in an expression system. Circulation. 2002;106(12):1493-
1499.
86. Qi, X.Y., Huang, H., Ordog, B., Luo, X., Naud, P., Sun, Y., Wu, C.T., Dawson, K.,
Tadevosyan, A., Chen, Y., Harada, M., Dobrev, D., and Nattel, S. Fibroblast inward-
rectifier potassium current upregulation in profibrillatory atrial remodeling. Circ Res.
2015;116(5):836-845.
87. Voigt, N., Heijman, J., Trausch, A., Mintert-Jancke, E., Pott, L., Ravens, U., and Dobrev,
D. Impaired Na+-dependent regulation of acetylcholine-activated inward-rectifier K+
29
current modulates action potential rate dependence in patients with chronic atrial
fibrillation. J Mol Cell Cardiol. 2013;61:142-152.
88. Makary, S., Voigt, N., Maguy, A., Wakili, R., Nishida, K., Harada, M., Dobrev, D., and
Nattel, S. Differential protein kinase C isoform regulation and increased constitutive
activity of acetylcholine-regulated potassium channels in atrial remodeling. Circ Res.
2011;109(9):1031-1043.
89. Voigt, N., Makary, S., Nattel, S., and Dobrev, D. Voltage-clamp-based methods for the
detection of constitutively active acetylcholine-gated IK,ACh channels in the diseased heart.
Methods Enzymol. 2010;484:653-675.
90. Kneller, J., Zou, R., Vigmond, E.J., Wang, Z., Leon, L.J., and Nattel, S. Cholinergic atrial
fibrillation in a computer model of a two-dimensional sheet of canine atrial cells with
realistic ionic properties. Circ Res. 2002;90(9):E73-87.
91. Kovoor, P., Wickman, K., Maguire, C.T., Pu, W., Gehrmann, J., Berul, C.I., and
Clapham, D.E. Evaluation of the role of IK,ACh in atrial fibrillation using a mouse
knockout model. J Am Coll Cardiol. 2001;37(8):2136-2143.
92. Nattel, S., Maguy, A., Le Bouter, S., and Yeh, Y.H. Arrhythmogenic ion-channel
remodeling in the heart: heart failure, myocardial infarction, and atrial fibrillation. Physiol
Rev. 2007;87(2):425-456.
93. Dhein, S., Hagen, A., Jozwiak, J., Dietze, A., Garbade, J., Barten, M., Kostelka, M., and
Mohr, F.W. Improving cardiac gap junction communication as a new antiarrhythmic
mechanism: the action of antiarrhythmic peptides. Naunyn Schmiedebergs Arch
Pharmacol. 2010;381(3):221-234.
94. Nishida, K., Michael, G., Dobrev, D., and Nattel, S. Animal models for atrial fibrillation:
clinical insights and scientific opportunities. Europace. 2010;12(2):160-172.
95. Firouzi, M., Ramanna, H., Kok, B., Jongsma, H.J., Koeleman, B.P., Doevendans, P.A.,
Groenewegen, W.A., and Hauer, R.N. Association of human connexin40 gene
polymorphisms with atrial vulnerability as a risk factor for idiopathic atrial fibrillation.
Circ Res. 2004;95(4):e29-33.
96. Gollob, M.H., Jones, D.L., Krahn, A.D., Danis, L., Gong, X.Q., Shao, Q., Liu, X., Veinot,
J.P., Tang, A.S., Stewart, A.F., Tesson, F., Klein, G.J., Yee, R., Skanes, A.C., Guiraudon,
G.M., Ebihara, L., and Bai, D. Somatic mutations in the connexin 40 gene (GJA5) in
atrial fibrillation. N Engl J Med. 2006;354(25):2677-2688.
97. Juang, J.M., Chern, Y.R., Tsai, C.T., Chiang, F.T., Lin, J.L., Hwang, J.J., Hsu, K.L.,
Tseng, C.D., Tseng, Y.Z., and Lai, L.P. The association of human connexin 40 genetic
polymorphisms with atrial fibrillation. Int J Cardiol. 2007;116(1):107-112.
30
98. Burstein, B., Comtois, P., Michael, G., Nishida, K., Villeneuve, L., Yeh, Y.H., and Nattel,
S. Changes in connexin expression and the atrial fibrillation substrate in congestive heart
failure. Circ Res. 2009;105(12):1213-1222.
99. Bikou, O., Thomas, D., Trappe, K., Lugenbiel, P., Kelemen, K., Koch, M., Soucek, R.,
Voss, F., Becker, R., Katus, H.A., and Bauer, A. Connexin 43 gene therapy prevents
persistent atrial fibrillation in a porcine model. Cardiovasc Res. 2011;92(2):218-225.
100. Igarashi, T., Finet, J.E., Takeuchi, A., Fujino, Y., Strom, M., Greener, I.D., Rosenbaum,
D.S., and Donahue, J.K. Connexin gene transfer preserves conduction velocity and
prevents atrial fibrillation. Circulation. 2012;125(2):216-225.
101. Dhein, S. Cardiac ischemia and uncoupling: gap junctions in ischemia and infarction. Adv
Cardiol. 2006;42:198-212.
102. Hagendorff, A., Schumacher, B., Kirchhoff, S., Luderitz, B., and Willecke, K. Conduction
disturbances and increased atrial vulnerability in Connexin40-deficient mice analyzed by
transesophageal stimulation. Circulation. 1999;99(11):1508-1515.
103. Wirka, R.C., Gore, S., Van Wagoner, D.R., Arking, D.E., Lubitz, S.A., Lunetta, K.L.,
Benjamin, E.J., Alonso, A., Ellinor, P.T., Barnard, J., Chung, M.K., and Smith, J.D. A
Common Connexin-40 Gene Promoter Variant Affects Connexin-40 Expression in
Human Atria and Is Associated with Atrial Fibrillation. Circ Arrhythm Electrophysiol.
4(1):87-93.
104. Sossalla, S., Kallmeyer, B., Wagner, S., Mazur, M., Maurer, U., Toischer, K., Schmitto,
J.D., Seipelt, R., Schondube, F.A., Hasenfuss, G., Belardinelli, L., and Maier, L.S. Altered
Na(+) currents in atrial fibrillation effects of ranolazine on arrhythmias and contractility
in human atrial myocardium. J Am Coll Cardiol. 2010;55(21):2330-2342.
105. Olson, T.M., Michels, V.V., Ballew, J.D., Reyna, S.P., Karst, M.L., Herron, K.J., Horton,
S.C., Rodeheffer, R.J., and Anderson, J.L. Sodium channel mutations and susceptibility to
heart failure and atrial fibrillation. JAMA. 2005;293(4):447-454.
106. Chen, L.Y., Ballew, J.D., Herron, K.J., Rodeheffer, R.J., and Olson, T.M. A common
polymorphism in SCN5A is associated with lone atrial fibrillation. Clin Pharmacol Ther.
2007;81(1):35-41.
107. Ellinor, P.T., Nam, E.G., Shea, M.A., Milan, D.J., Ruskin, J.N., and MacRae, C.A.
Cardiac sodium channel mutation in atrial fibrillation. Heart Rhythm. 2008;5(1):99-105.
108. Hedley, P.L., Jorgensen, P., Schlamowitz, S., Moolman-Smook, J., Kanters, J.K.,
Corfield, V.A., and Christiansen, M. The genetic basis of Brugada syndrome: a mutation
update. Hum Mutat. 2009;30(9):1256-1266.
31
109. Watanabe, H., Darbar, D., Kaiser, D.W., Jiramongkolchai, K., Chopra, S., Donahue, B.S.,
Kannankeril, P.J., and Roden, D.M. Mutations in sodium channel beta1- and beta2-
subunits associated with atrial fibrillation. Circ Arrhythm Electrophysiol. 2009;2(3):268-
275.
110. Olesen, M.S., Jespersen, T., Nielsen, J.B., Liang, B., Moller, D.V., Hedley, P.,
Christiansen, M., Varro, A., Olesen, S.P., Haunso, S., Schmitt, N., and Svendsen, J.H.
Mutations in sodium channel beta-subunit SCN3B are associated with early-onset lone
atrial fibrillation. Cardiovasc Res. 2011;89(4):786-793.
111. Wang, P., Yang, Q., Wu, X., Yang, Y., Shi, L., Wang, C., Wu, G., Xia, Y., Yang, B.,
Zhang, R., Xu, C., Cheng, X., Li, S., Zhao, Y., Fu, F., Liao, Y., Fang, F., Chen, Q., Tu,
X., and Wang, Q.K. Functional dominant-negative mutation of sodium channel subunit
gene SCN3B associated with atrial fibrillation in a Chinese GeneID population. Biochem
Biophys Res Commun. 2010;398(1):98-104.
112. Jabbari, J., Olesen, M.S., Yuan, L., Nielsen, J.B., Liang, B., Macri, V., Christophersen,
I.E., Nielsen, N., Sajadieh, A., Ellinor, P.T., Grunnet, M., Haunso, S., Holst, A.G.,
Svendsen, J.H., and Jespersen, T. Common and rare variants in SCN10A modulate the
risk of atrial fibrillation. Circ Cardiovasc Genet. 2015;8(1):64-73.
113. Lamirault, G., Gaborit, N., Le Meur, N., Chevalier, C., Lande, G., Demolombe, S.,
Escande, D., Nattel, S., Leger, J.J., and Steenman, M. Gene expression profile associated
with chronic atrial fibrillation and underlying valvular heart disease in man. J Mol Cell
Cardiol. 2006;40(1):173-184.
114. Li, D., Fareh, S., Leung, T.K., and Nattel, S. Promotion of atrial fibrillation by heart
failure in dogs: atrial remodeling of a different sort. Circulation. 1999;100(1):87-95.
115. Dzeshka, M.S., Lip, G.Y., Snezhitskiy, V., and Shantsila, E. Cardiac Fibrosis in Patients
With Atrial Fibrillation: Mechanisms and Clinical Implications. J Am Coll Cardiol.
2015;66(8):943-959.
116. Yue, L., Xie, J., and Nattel, S. Molecular determinants of cardiac fibroblast electrical
function and therapeutic implications for atrial fibrillation. Cardiovasc Res.
2011;89(4):744-753.
117. Ehrlich, J.R., Hohnloser, S.H., and Nattel, S. Role of angiotensin system and effects of its
inhibition in atrial fibrillation: clinical and experimental evidence. Eur Heart J.
2006;27(5):512-518.
118. He, X., Gao, X., Peng, L., Wang, S., Zhu, Y., Ma, H., Lin, J., and Duan, D.D. Atrial
Fibrillation Induces Myocardial Fibrosis Through Angiotensin II Type 1 Receptor-
Specific Arkadia-Mediated Downregulation of Smad7. Circ Res. 108(2):164-175.
32
119. Koinuma, D., Shinozaki, M., Komuro, A., Goto, K., Saitoh, M., Hanyu, A., Ebina, M.,
Nukiwa, T., Miyazawa, K., Imamura, T., and Miyazono, K. Arkadia amplifies TGF-beta
superfamily signalling through degradation of Smad7. EMBO J. 2003;22(24):6458-6470.
120. Hunyady, L., and Catt, K.J. Pleiotropic AT1 receptor signaling pathways mediating
physiological and pathogenic actions of angiotensin II. Mol Endocrinol. 2006;20(5):953-
970.
121. Burstein, B., Libby, E., Calderone, A., and Nattel, S. Differential behaviors of atrial
versus ventricular fibroblasts: a potential role for platelet-derived growth factor in atrial-
ventricular remodeling differences. Circulation. 2008;117(13):1630-1641.
122. Verheule, S., Sato, T., Everett, T.t., Engle, S.K., Otten, D., Rubart-von der Lohe, M.,
Nakajima, H.O., Nakajima, H., Field, L.J., and Olgin, J.E. Increased vulnerability to atrial
fibrillation in transgenic mice with selective atrial fibrosis caused by overexpression of
TGF-beta1. Circ Res. 2004;94(11):1458-1465.
123. Attisano, L., and Wrana, J.L. Signal transduction by the TGF-beta superfamily. Science.
2002;296(5573):1646-1647.
124. Burstein, B., Qi, X.Y., Yeh, Y.H., Calderone, A., and Nattel, S. Atrial cardiomyocyte
tachycardia alters cardiac fibroblast function: a novel consideration in atrial remodeling.
Cardiovasc Res. 2007;76(3):442-452.
125. Tsai, C.T., Tseng, C.D., Hwang, J.J., Wu, C.K., Yu, C.C., Wang, Y.C., Chen, W.P., Lai,
L.P., Chiang, F.T., and Lin, J.L. Tachycardia of atrial myocytes induces collagen
expression in atrial fibroblasts through transforming growth factor {beta}1. Cardiovasc
Res. 2011;89(4):805-815.
126. Ponten, A., Folestad, E.B., Pietras, K., and Eriksson, U. Platelet-derived growth factor D
induces cardiac fibrosis and proliferation of vascular smooth muscle cells in heart-specific
transgenic mice. Circ Res. 2005;97(10):1036-1045.
127. Gudbjartsson, D.F., Arnar, D.O., Helgadottir, A., Gretarsdottir, S., Holm, H., Sigurdsson,
A., Jonasdottir, A., Baker, A., Thorleifsson, G., Kristjansson, K., Palsson, A., Blondal, T.,
Sulem, P., Backman, V.M., Hardarson, G.A., Palsdottir, E., Helgason, A., Sigurjonsdottir,
R., Sverrisson, J.T., Kostulas, K., Ng, M.C., Baum, L., So, W.Y., Wong, K.S., Chan, J.C.,
Furie, K.L., Greenberg, S.M., Sale, M., Kelly, P., MacRae, C.A., Smith, E.E., Rosand, J.,
Hillert, J., Ma, R.C., Ellinor, P.T., Thorgeirsson, G., Gulcher, J.R., Kong, A.,
Thorsteinsdottir, U., and Stefansson, K. Variants conferring risk of atrial fibrillation on
chromosome 4q25. Nature. 2007;448(7151):353-357.
128. Cardin, S., Libby, E., Pelletier, P., Le Bouter, S., Shiroshita-Takeshita, A., Le Meur, N.,
Leger, J., Demolombe, S., Ponton, A., Glass, L., and Nattel, S. Contrasting gene
33
expression profiles in two canine models of atrial fibrillation. Circ Res. 2007;100(3):425-
433.
129. Adam, O., Lavall, D., Theobald, K., Hohl, M., Grube, M., Ameling, S., Sussman, M.A.,
Rosenkranz, S., Kroemer, H.K., Schafers, H.J., Bohm, M., and Laufs, U. Rac1-induced
connective tissue growth factor regulates connexin 43 and N-cadherin expression in atrial
fibrillation. J Am Coll Cardiol. 2010;55(5):469-480.
130. Ko, W.C., Hong, C.Y., Hou, S.M., Lin, C.H., Ong, E.T., Lee, C.F., Tsai, C.T., and Lai,
L.P. Elevated expression of connective tissue growth factor in human atrial fibrillation
and angiotensin II-treated cardiomyocytes. Circ J. 2011;75(7):1592-1600.
131. van Rooij, E., Sutherland, L.B., Thatcher, J.E., DiMaio, J.M., Naseem, R.H., Marshall,
W.S., Hill, J.A., and Olson, E.N. Dysregulation of microRNAs after myocardial infarction
reveals a role of miR-29 in cardiac fibrosis. Proc Natl Acad Sci U S A.
2008;105(35):13027-13032.
132. Dawson, K., Wakili, R., Ördög, B., Clauss, S., Chen, Y., Iwasaki, Y., Voigt, N., Qi, X.Y.,
Sinner, M.F., Dobrev, D., Kääb, S., and Nattel, S. MicroRNA29: A mechanistic
contributor and potential biomarker in atrial fibrillation. Circulation. 2013;127:1466-
1475.
133. Duisters, R.F., Tijsen, A.J., Schroen, B., Leenders, J.J., Lentink, V., van der Made, I.,
Herias, V., van Leeuwen, R.E., Schellings, M.W., Barenbrug, P., Maessen, J.G.,
Heymans, S., Pinto, Y.M., and Creemers, E.E. miR-133 and miR-30 regulate connective
tissue growth factor: implications for a role of microRNAs in myocardial matrix
remodeling. Circ Res. 2009;104(2):170-178, 176p following 178.
134. Chen, Y., Wakili, R., Luo, X., Dawson, K., Qi, X.Y., Clauss, S., Naud, P., Shi, Y.F.,
Tardif, J.C., Kääb, S., Dobrev, D., and Nattel, S. Distinct atrial and ventricular microRNA
expression changes in experimental dilated cardiomyopathy: cell-type specificity and time
course. J Mol Cell Cardiol. 2014;12:113-124.
135. Thum, T., Gross, C., Fiedler, J., Fischer, T., Kissler, S., Bussen, M., Galuppo, P., Just, S.,
Rottbauer, W., Frantz, S., Castoldi, M., Soutschek, J., Koteliansky, V., Rosenwald, A.,
Basson, M.A., Licht, J.D., Pena, J.T., Rouhanifard, S.H., Muckenthaler, M.U., Tuschl, T.,
Martin, G.R., Bauersachs, J., and Engelhardt, S. MicroRNA-21 contributes to myocardial
disease by stimulating MAP kinase signalling in fibroblasts. Nature. 2008;456(7224):980-
984.
136. Patrick, D.M., Montgomery, R.L., Qi, X., Obad, S., Kauppinen, S., Hill, J.A., van Rooij,
E., and Olson, E.N. Stress-dependent cardiac remodeling occurs in the absence of
microRNA-21 in mice. J Clin Invest. 2010;120(11):3912-3916.
34
137. Cardin, S., Guasch, E., Luo, X., Naud, P., Le Quang, K., Shi, Y.F., Tardif, J.C.,
Comptois, P., and Nattel, S. A role for microRNA in atrial profibrillatory fibrotic
remodeling associated with experimental postinfarction heart failure. Circ Arrhythm
Electrophyiol. 2012;5:1027-1035.
138. Fatini, C., Sticchi, E., Gensini, F., Gori, A.M., Marcucci, R., Lenti, M., Michelucci, A.,
Genuardi, M., Abbate, R., and Gensini, G.F. Lone and secondary nonvalvular atrial
fibrillation: role of a genetic susceptibility. Int J Cardiol. 2007;120(1):59-65.
139. Bedi, M., McNamara, D., London, B., and Schwartzman, D. Genetic susceptibility to
atrial fibrillation in patients with congestive heart failure. Heart Rhythm. 2006;3(7):808-
812.
140. Kato, K., Oguri, M., Hibino, T., Yajima, K., Matsuo, H., Segawa, T., Watanabe, S.,
Yoshida, H., Satoh, K., Nozawa, Y., Yokoi, K., and Yamada, Y. Genetic factors for lone
atrial fibrillation. Int J Mol Med. 2007;19(6):933-939.
141. Benjamin, E.J., Rice, K.M., Arking, D.E., Pfeufer, A., van Noord, C., Smith, A.V.,
Schnabel, R.B., Bis, J.C., Boerwinkle, E., Sinner, M.F., Dehghan, A., Lubitz, S.A.,
D'Agostino, R.B., Sr., Lumley, T., Ehret, G.B., Heeringa, J., Aspelund, T., Newton-Cheh,
C., Larson, M.G., Marciante, K.D., Soliman, E.Z., Rivadeneira, F., Wang, T.J.,
Eiriksdottir, G., Levy, D., Psaty, B.M., Li, M., Chamberlain, A.M., Hofman, A., Vasan,
R.S., Harris, T.B., Rotter, J.I., Kao, W.H., Agarwal, S.K., Stricker, B.H., Wang, K.,
Launer, L.J., Smith, N.L., Chakravarti, A., Uitterlinden, A.G., Wolf, P.A., Sotoodehnia,
N., Kottgen, A., van Duijn, C.M., Meitinger, T., Mueller, M., Perz, S., Steinbeck, G.,
Wichmann, H.E., Lunetta, K.L., Heckbert, S.R., Gudnason, V., Alonso, A., Kaab, S.,
Ellinor, P.T., and Witteman, J.C. Variants in ZFHX3 are associated with atrial fibrillation
in individuals of European ancestry. Nat Genet. 2009;41(8):879-881.
142. Mommersteeg, M.T., Brown, N.A., Prall, O.W., de Gier-de Vries, C., Harvey, R.P.,
Moorman, A.F., and Christoffels, V.M. Pitx2c and Nkx2-5 are required for the formation
and identity of the pulmonary myocardium. Circ Res. 2007;101(9):902-909.
143. Mommersteeg, M.T., Hoogaars, W.M., Prall, O.W., de Gier-de Vries, C., Wiese, C.,
Clout, D.E., Papaioannou, V.E., Brown, N.A., Harvey, R.P., Moorman, A.F., and
Christoffels, V.M. Molecular pathway for the localized formation of the sinoatrial node.
Circ Res. 2007;100(3):354-362.
144. Kim, C.J., Song, J.H., Cho, Y.G., Cao, Z., Lee, Y.S., Nam, S.W., Lee, J.Y., and Park,
W.S. Down-regulation of ATBF1 is a major inactivating mechanism in hepatocellular
carcinoma. Histopathology. 2008;52(5):552-559.
145. Sun, X., Frierson, H.F., Chen, C., Li, C., Ran, Q., Otto, K.B., Cantarel, B.L., Vessella,
R.L., Gao, A.C., Petros, J., Miura, Y., Simons, J.W., and Dong, J.T. Frequent somatic
35
mutations of the transcription factor ATBF1 in human prostate cancer. Nat Genet.
2005;37(4):407-412.
146. Kim, T.S., Kawaguchi, M., Suzuki, M., Jung, C.G., Asai, K., Shibamoto, Y., Lavin, M.F.,
Khanna, K.K., and Miura, Y. The ZFHX3 (ATBF1) transcription factor induces
PDGFRB, which activates ATM in the cytoplasm to protect cerebellar neurons from
oxidative stress. Dis Model Mech. 2010;3(11-12):752-762.
147. Harada, M., Luo, X., Qi, X.Y., Tadevosyan, A., Maguy, A., Ordog, B., Ledoux, J., Kato,
T., Naud, P., Voigt, N., Shi, Y., Kamiya, K., Murohara, T., Kodama, I., Tardif, J.C.,
Schotten, U., Van Wagoner, D.R., Dobrev, D., and Nattel, S. Transient receptor potential
canonical-3 channel-dependent fibroblast regulation in atrial fibrillation. Circulation.
2012;126(17):2051-2064.
148. Li, N., Chiang, D.Y., Wang, S., Wang, Q., Sun, L., Voigt, N., Respress, J.L., Ather, S.,
Skapura, D.G., Jordan, V.K., Horrigan, F.T., Schmitz, W., Muller, F.U., Valderrabano,
M., Nattel, S., Dobrev, D., and Wehrens, X.H. Ryanodine receptor-mediated calcium leak
drives progressive development of an atrial fibrillation substrate in a transgenic mouse
model. Circulation. 2014;129(12):1276-1285.
149. Donahue, J.K. Biological therapies for atrial fibrillation: ready for prime time? J
Cardiovasc Pharmacol. 2015.
36
Figure legends:
Figure 1: Etiological contributors to atrial fibrillation.
Figure 2: Mechanistic basis of atrial fibrillation and associated clinical forms. A, Focal
firing usually results from local ectopic activity. Organized discrete reentrant
activity and focal firing can maintain AF by producing regularly-firing drivers that
are conducted irregularly (fibrillatory conduction) in the heterogeneous atrial
substrate. B, AF can progress from paroxysmal (self-terminating) forms, believed to
result primarily from focal drivers, to persistent and longstanding persistent (>1
year) forms due to AF-related remodeling and/or disease progression, resulting in
development of an increasingly vulnerable substrate. Substrate vulnerability, AF-
related remodeling and disease progression are further modulated by genetic
predisposition. . There is overlap between mechanisms and pronounced patient-to-
patient heterogeneity.
Figure 3: Cellular Ca2+ handling and DADs. A, During the plateau phase of the action
potential (AP) Ca2+ enters the cell via L-type Ca2+ channels. This Ca2+ binds to
ryanodine-receptors (RyR2), triggering a much larger Ca2+ release from the
sarcoplasmic reticulum (SR), which initiates cellular contraction. SR Ca2+ stores are
maintained by pumping Ca2+ into the SR by the SR Ca2+-ATPase, SERCA. B,
Diastolic Ca2+-handling abnormalities underlie DADs. Spontaneous SR Ca2+-
releases through RyR2 elevate cytosolic Ca2+, which is exchanged for extracellular
Na+ by the Na+/Ca2+ exchanger (NCX), producing depolarizing transient inward-
current (Iti). Inappropriate diastolic RyR2 Ca2+-release is promoted by
RyR2-hyperphosphorylation, excess SR Ca2+, or decreased SR Ca2+-binding to
calsequestrin (CSQ; red arrows). Repolarizing conductances oppose Iti and suppress
37
diastolic depolarization, so reduced diastolic K+ current or increased NCX current
can favor DADs (orange arrows).
Figure 4: Determinants of reentry. Top, Basic concepts of reentry. The leading circle model
(A) posits reentry around a central zone that is continuously activated by centripetal
waves emanating from the reentering activation wavefront. The spiral wave model
(B) describes reentry as a “rotor” established by tissue excitability properties. C,
Structural remodeling (atrial enlargement and fibrosis, most typically affecting the
left atrium, LA) produces relatively-fixed reentry substrates that reverse poorly if at
all.
Figure 5: A schematic representation of gene-regulatory pathway dysregulation in AF.
Abbreviations: AT-II, angiotensin-II; ATR, angiotensin receptor; NFAT, nuclear
factor of activated T-cells; NOX, NADPH-oxidase; P, phosphate; ROS, reactive
oxygen species; TF, transcription factor; TGFβ, transforming growth factor-β;
TGFβR, TGFβ-receptor;.
Figure 6: Molecular basis of DAD-inducing diastolic Ca2+-releases. RyR-dysfunction is
caused by RyR-hyperphosphorylation or excess Ca2+-loads. Phospholamban (PLB)
inhibits SERCA. PLB-hyperphosphorylation removes this inhibitory effect,
enhances SERCA function and can lead to Ca2+-overload. High atrial rate during AF
enhances cellular Ca2+-entry. Increased cell-Ca2+ promotes Ca2+/calmodulin (CaM)
binding to Ca2+/calmodulin-dependent protein kinase-II (CaMKII), disinhibiting the
catalytic subunit. After CaMKII catalytic-subunit activation, oxidation at
Met281/282 or phosphorylation at Thr286 cause persistent CaMKII activity.
Inhibitor-1 (I-1) suppresses protein-phosphatase-1 (PP1) function in the SR and
contributes to PLN and RyR phosphorylation. These factors have been associated
38
with AF in samples from pAF patients (blue arrows), cAF patients (red arrows), or
animal models (yellow arrows).
Figure 7: Mechanisms of ionic-current remodeling in AF. A, L-type Ca2+-current (ICa,L)
downregulation. High atrial rates in AF enhance intracellular Ca2+-load, activating
calcineurin via the Ca2+/calmodulin (CaM) binding. Calcineurin dephosphorylates
Nuclear Factor of Activated T-Lymphocytes (NFAT), allowing it to translocate into
the nucleus and reduce mRNA-levels of the ICa,L alpha-subunit, Cav1.2. Breakdown
of Cav1.2 protein by calpains might also contribute to reduced Cav1.2 protein
expression. The protein Zinc transporter-1 (ZnT-1) impairs Cav1.2 membrane-
trafficking and is upregulated in AF. Increased protein-phosphatase-1/2a (PP1/PP2a)
activity dephosphorylates Cav1.2-phosphorylation and may also decrease ICa,L. B,
Inward-rectifier K+-current upregulation. Increased Kir2.1 subunit expression is
caused by decreases in inhibitory microRNAs like miR-101, miR-26, miR-1, at least
in part due to NFAT-mediated inhibition of miR-26/miR-1 expression.
Acetylcholine-regulated K+-current (IK,AChc) is increased because of increased
membrane-abundance of the stimulatory protein-kinase C (PKC) isoform PKC and
decreased cellular expression of the inhibitory isoform PKC due to enhanced
calpain-mediated breakdown.
Gene
(Presumed) Functional
target(s)
(Presumed) Arrhythmogenic mechanism
AAC variants affecting K+-currents
IKATP
LOF: Impaired APD adaptation during stress
IKur
GOF: ↓ ERP; LOF: ↑ APD, EADs, TA
Ito
GOF: ↓ ERP
IKs
GOF: ↓ ERP; LOF: ↑ APD, EADs
KCNQ1/KCNE2 K+-current
GOF: ↓ ERP
Ito / I Kr
GOF: ↓ ERP
IKs (?)
?
IKs
GOF: ↓ ERP
I
Kr
GOF: ↓ ERP; LOF: ?
IK2P
LOF: ↑ APD, SAN dysfunction
IK1
GOF: ↓ ERP, ↓ RMP
I
K1
?
IK,ACh
?
IKATP
GOF: ↓ ERP
I
K1
?
IKs
GOF: ↓ ERP; LOF: ?
IKs
GOF: ↓ ERP, ?
AAC variants affecting Na+ and related currents
If
LOF: SAN dysfunction
INa
GOF: ↑ excitability / TA; LOF: ↓ CV
INa
GOF: ↑ excitability / TA; LOF: ↓ CV
I
Na
LOF: ↓ CV
INa/I to
Mixed: ↓ INa , ↑ Ito , ↓ ERP
INa
LOF: ↓ CV
INa
LOF: ↓ CV
I
Na
?
AAC variants affecting Ca2+-handling and current
ICa,L
?
I
Ca,L
?
Ca2+-buffering
?
RyR2-channel
GOF: ↑ SCaEs, TA
RyR2-channel
GOF: ↑ SCaEs, TA, ↓ CV
AAC variants not directly affecting sarcolemmal ion currents
TF
LOF: ?
Connexin43
LOF: ↓ CV
Connexin40
LOF: ↓ CV
BMP antagonist
GOF: Abnormal cardiac development, ↓ CV
Lamin A/C
Structural remodeling, ↓ CV
Contractile proteins
?
TF
LOF: ?
Nuclear pore complex
LOF: Impaired nuclear permeability, remodeling,↓ APD
TF
LOF: ?
TF
?
Table 1. amino-acid coding (AAC) missense variants associated with atrial fibrillation. APD: action potential duration;
CV: slow/heterogeneous conduction; ERP: reduced effective refractory period; GOF: Gain of function variants; LOF: Loss
of function variants; RMP: resting membrane potential hyperpolarization; SCaEs: spontaneous diastolic Ca2+-release
events; TA: triggered activity; TF: transcription factor; ?: unknown mechanism. Based on [3,4,4a,4b].
(Presumed/nearest) Gene
Variant
(Presumed) Function
Relative risk
C9orf3
rs10821415
?
1.13 (1.08-1.18)
CAND2
rs4642101
Modulating atrial APD
1.10 (1.06-1.14)
CAV1
rs3807989
Cellular structure and signaling
0.88 (0.84-0.91)
GJA1 rs13216675
Electrical coupling controlling conduction
velocity
1.10 (1.06-1.14)
HCN4
rs7164883
Regulation of If and automaticity
1.16 (1.10-1.22)
KCNN3
rs6666258
Regulation of I
SK
, modulating APD
1.18 (1.13-1.23)
NEURL
rs12415501
Modulating atrial APD
1.18 (1.13-1.23)
PITX2 rs6817105
Left/right division and pulmonary vein sleeve
development
1.64 (1.54-2.21)
PRRX1
rs3903239
Development of great vessels
1.14 (1.10-1.18)
SYNE2
rs1152591
Sarcomere structural protein
1.13 (1.09-1.18)
SYNPO2L / MYOZ1
rs10824026
Regulating actin and cardiomyocyte structure
0.85 (0.81-0.9)
TBX5 rs10507248
Transcription factor controlling conduction
system development
1.12 (1.08-1.16)
ZFHX3
rs2106261
?
1.24 (1.17-1.30)
Table 2. Genetic variants associated with atrial fibrillation in genome-wide association and large-scale genotyping studies
in patients from European descent. Based on [3,4,4a,4b].
Causes of Atrial Fibrillation
Extrinsic factors
Heart disease
Genetic factors
ATRIAL FIBRILLATION
Abnormalities in cardiac
structure or function
Altered ion channel
function
Calcium-handling
abnormalities
Thyroid dysfunction
Sleep apnea
Autonomic toneAlcohol, drugs
Inflammation
Non-CV disease
Diabetes
Obesity
COPDHypertensive heart diseaseCoronary artery disease
Valvular heart disease Cardiomyopathies
Pericarditis /Myocarditis
Aging heart
Heart failure
Rare disease-
causing mutations
Common variants
with small effect size
Variants of unknown significance
Cigarette smoking
Reentry
Driver
Focal firing Trigger
Fibrillatory
conduction
Substrate+
Atrial Fibrillation Triggers and Substrates
Single
circuit
reentry
sustained
transient
remodeling
A
BAtrial Fibrillation Forms and Progression
Figure 2
Multiple
circuit
reentry
ATRIAL FIBRILLATION
remodeling
+
AF AF AFAF
0 4 8
days
AF
Vulnerability
triggers trigger triggertriggers
trigger
60 80
Age (years)
Paroxysmal
AF Persistent
AF Longstanding
persistent AF
AF-related remodeling Disease-related remodeling
Genetic predisposition
AF-related
remodeling
Disease-
related
remodeling
Genetics
Paroxysmal AF Persistent AF Longstanding perAF
progression progression
Figure 3
ICa,L
NCX
Cytosol
3Na+
RyR2
SR
Diastole
[Ca2+]
PP
Ca2+
B. Diastolic Ca2+-handling
abnormalities and DAD generation
Iti
DADs
200 ms
NCX
Ca2+
AF
CSQ
Extracellular
space Repolarizing
conductances
ICa,L
ICa,L
NCX
Cytosol
3Na+
RyR2
SR
Systole
[Ca2+]
PP
Ca2+
A. Systolic Ca2+-handling
producing cell contraction
200 ms
NCX
Ca2+
CSQ
Extracellular
space
SERCA SERCA
SLN
PLB
SLN
PLB
Structural Remodeling
Figure 4
LA enlargement
AF maintaining
substrate
C
Functional Determinants
Mechanisms of Reentry
Leading circle
Determined by wavelength = effective
refractory period ×conduction velocity
core
Spiral wave
Determined by excitability
and wave curvature
A B
refractory
RA LA
LA fibrosis
Cytosol
Nucleus
DNA
GATA-4
GATA-5
GATA-6 NKX2-5 TBX5
PITX2
ZFHX2
PRRX1
ATR AT-II
TGFβR
TGFβR
TGFβR
TGFβ
SMAD 2/3
STAT
JAK
Ca2+
ICa,L
CaM
NFATc3/c4
P
Calcineurin NFATc3/c4 P
AF
ROS ROS
SMAD 4
NF-κB
c-fos
NOX
AF-promoting conditions
miRNAs
mRNA
protein
Figure 5
AF-inducing mutations in transcription factors (TFs)
TF-related gene variants implicated in AF
Figure 6
RyR2
SR
PP
[Ca2+]
CSQ
Ser2814
Ser2808
PP
Ser16
Thr17
I-1
PP1
P
PP1
Atrial
rate
Thr35
Ca2+ Leak
P
Ox CaM
CaM Ca2+
Ca2+
(autoinhibited)
(autophosphorylated)(Ca2+/CaM-dependent)
(oxidized)
CaMKII activity
Factors promoting AF by inducing diastolic
Ca2+ leak through RyR2
Ang-II
Thr286
NADPHox
Cytosol
Extracellular space
SERCA
PLB
SLN PThr5
Met281/282
Associated with AF in:
pAF patients
cAF patients
Animal models
Spin-1
JPH2
DNAmRNA
Cav1.2
Ca2+
P
PP1/PP2A
ICa,L
GSH
NO
CaM
NFATc3/c4
NFATc3/c4 P
P
Calcineurin
Oxidative
Stress
Atrial
rate
APD
L-type Ca2+ current
remodeling
Trafficking
Transcription
Degradation
Calpains
Nucleus
Cytosol
ZnT-1
miR-328
mRNA
Kir2.1
IK1
Translation
miR-26
miR-101
miR-1
IK,AChc Basal IR current
APD
Hyperpolarization
PPKCε
Inward rectifier K+current
remodeling
AB
Figure 7
PKCα
AF
mRNA
Transcription miR-101
miR-26
Calpains
Ca2+
Cav1.2
Kir2.1
... AF can be predisposed by factors such as cardiac senescence (mediated by structural remodeling which causes fibrotic alterations), microconduction slowing and atrial enlargement, and most AF cases have associated a cardiac disease (heart failure, hypertensive heart disease, valvular disease and ischemic heart disease). There are genetic and hereditary factors linked to AF development, and a variety of extracardiac conditions (as alcohol consumption, hyperthyroidism and obesity) that can increase the risk of AF occurrence [2]. Reentrant activity and focal ectopic firing are the primary mechanisms of initiation and maintenance of AF. ...
... and N defined as in Eq. (2). Observe that, on the contrary of ApEn, FuzzyEn does not consider self-counting (j / = i). ...
Article
Abstract Background Classification of complex fractionated atrial electrograms is crucial for the study of atrial fibrillation and the development of treatment strategies, because these electrophysiological phenomena represent a common substrate for radiofrequency ablation in treatment of this arrythmia. Objective The objective of this work is the characterization of short term atrial electrograms using nonlinear dynamics measures, helping in the automatic classification of electrograms. Methods The dataset consists of 113 atrial electrograms recordings from left-atrial endocardial mapping. These signals were classified by three expert electrophysiologists into four classes, from C0 (non fractionated) to C3 (high degree of fractionation). The calculated features were Approximate entropy, Dispersion entropy, Fuzzy entropy, Permutation entropy, Tsallis entropy, Shannon entropy, Renyi entropy, and Lempel-Ziv complexity. Features were selected for classification using Neighborhood Component Analysis. Different classifiers were tested using selected features, and the one with maximum sensitivity and specificity in each task was reported. Results We obtained a classification performance that overcome previous works on this database and are comparable to the results of studies performed over bigger datasets. Separation between C3 signals from (C0, C1, C2) signals was performed with 99.98% sensitivity and 96.61% specificity. Non-fractionated signals (C0 + C1) were separated from fractionated signals (C2 + C3) with 96.72% sensitivity and 94.51% specificity. Moreover, the estimation times of the selected features are low enough to consider the online application of this scheme. Conclusions and significance Classification performance obtained using information and complexity measures shown better results than previous works over this dataset, encouraging the application of these features to characterize atrial electrograms.
... Although the two theories mentioned can satisfactorily explain the occurrence of PWD, since its emergence and up to the present time, researchers have given as a resounding fact that the local theory is correct [4,5]. Four main elements give weight to the local theory: 1) the demonstration that PWD is an independent predictor of atrial fibrillation in multiple studies, 2) the relationship of PWD with atrial conduction, 3) its decrease after interventiononal therapies that reduce atrial remodeling or remove atrial tissue from certain critical areas and 4) there is strong evidence that atrial depolarization asynchrony plays a crucial role in the genesis and maintenance of atrial fibrillation [6][7][8][9][10][11][12][13][14][15][16][17][18][19]. However, the datas that favor the local theory through the relationship of atrial conduction with PWD are weak, because they do not explore the essence of the theory that is the inhomogeneous conduction of the atrial electrical impulse. ...
Article
Background Local theory and the vectorial theory are used to explain the origin of P-wave dispersion (PWD). There are no previous studies that analyze both at the same time. Objectives We set out to determine the implication of local and vectorial theories in the origin of PWD. Methods Cross-sectional study in 153 randomly selected patients aged 18–70 years, undergoing electrophysiological study. Inhomogeneous atrial conduction was evaluated by atrial electrogram dispersion in terms of duration (EGMdurdis) and morphology (EGMmorph dis). P-distal coronary sinus interval (P-DCS) was also measured. P-wave was measured twice, firstly at a calibration of 20 mm/mV and a sweep speed of 50 mm/s, enhancement 10× (basic measurement [BM]), and second time at sweep speed of 150 mm/s, enhancement 80-160× (high precision measurement [HPM]). Results PWD with BM was 48 ms [36–54 ms] while with HPM it was 4 ms [0–10 ms], p < 0.001. With BM, maximum and minimum P- wave duration presented a moderate correlation (r = 0.342; p < 0.001), using HPM it becomes strong (r = 0.750; p < 0.001). In cases with P-DCS < 80 ms (r = 0.965; p < 0.001), but not with P-DCS ≥ 80 ms (r = 0.649; p < 0.001), the previous correlation became almost perfect with HPM. EGMdurdis and EGMmorphdis were weak but significantly correlated with PWD. This correlation became moderate in patients with P-DCS ≥ 80 ms and disappeared in those with P-DCS, using BM and HPM. Conclusion Vectorial theory explains almost entirely the PWD phenomenon. Inhomogeneous conduction could be an additional mechanism to explain PWD, but its contribution is small.
... The role of the parasympathetic nervous system or volume overload in the pathogenesis of AF has a long and rich history independently. 5,6 Whereas molecular control mechanisms of AF have been investigated widely, linked to gene expression, cell Ca 2þ handling, and conduction disturbance, 7 interrelation between volume overload and parasympathetic nervous system in triggering and maintaining AF remains elusive. Recently, we established a rat model to deliver long-term volume overload to the heart by aorto-venocaval shunt (AVS) operation, where initiating trigger of AF, namely, increased automaticity in the pulmonary-vein myocardium could be found. ...
Article
Full-text available
Whereas molecular mechanisms of atrial fibrillation (AF) have been widely investigated, there is limited information regarding interrelation between chronic volume overload and parasympathetic nervous system in the pathophysiology of AF. In this study, we investigated the influence of abdominal aorto-venocaval shunt (AVS) -induced atrial remodeling on electrophysiological responses to cholinergic receptor stimulation in the isolated rat atria. Interstitial fibrosis, cardiomyocyte hypertrophy and atrial enlargement, known as structural arrhythmogenic substrates for AF, took place after one month of AVS operation. Carbachol at 0.1 and 1 μM shortened the effective refractory period, acting as functional arrhythmogenic substrates, but increased the conduction velocity both in the atria of the sham-operated and AVS rats. The extents of the electrophysiological responses to carbachol in the atria of the AVS rat were greater than those in the sham-operated ones. Also, the higher inducibility and longer duration of carbachol-mediated AF were detected in the AVS atria than those in the sham-operated ones. These results showed that chronic volume overload-induced atrial remodeling promoted electrophysiological responses to cholinergic receptor stimulation in the isolated atria of rats, suggesting possible synergistic actions between structural arrhythmogenic substrate in the remodeled atria and functional arrhythmogenic substrates modulated by parasympathetic nerve activity.
... Although the two theories mentioned can satisfactorily explain the occurrence of PWD, since its emergence and up to the present time, researchers have given as a resounding fact that the local theory is correct [4,5]. Four main elements give weight to the local theory: 1) the demonstration that PWD is an independent predictor of atrial fibrillation in multiple studies, 2) the relationship of PWD with atrial conduction, 3) its decrease after interventiononal therapies that reduce atrial remodeling or remove atrial tissue from certain critical areas and 4) there is strong evidence that atrial depolarization asynchrony plays a crucial role in the genesis and maintenance of atrial fibrillation [6][7][8][9][10][11][12][13][14][15][16][17][18][19]. However, the datas that favor the local theory through the relationship of atrial conduction with PWD are weak, because they do not explore the essence of the theory that is the inhomogeneous conduction of the atrial electrical impulse. ...
Article
Full-text available
Atrial myopathy is a condition that consists of electrical, structural, contractile, and autonomic remodeling of the atria and is the substrate for development of atrial fibrillation, the most common arrhythmia. Pathophysiologic mechanisms driving atrial myopathy are inflammation, oxidative stress, atrial stretch, and neurohormonal signals, e.g., angiotensin-II and aldosterone. These mechanisms initiate the structural and functional remodeling of the atrial myocardium. Novel therapeutic strategies are being developed that target the pathophysiologic mechanisms of atrial myopathy. In this review, we will discuss the pathophysiology of atrial myopathy, as well as diagnostic and therapeutic strategies.
Chapter
Atrial fibrillation (AF) is the commonest arrhythmia seen in clinical practice and is responsible for thromboembolic strokes, substantial impairment in life quality, and mortality. This chapter discusses the epidemiology of AF, the different types of AF, and its mechanisms and treatment including stroke prevention and rate and rhythm control with special reference to current and future catheter ablation strategies.
Article
Full-text available
Atrial fibrillation (AF) is an extremely common clinical problem associated with increased morbidity and mortality. Current antiarrhythmic options include pharmacological, ablation and surgical therapies, and have significantly improved clinical outcomes. However, their efficacy remains suboptimal and their use is limited by a variety of potentially serious adverse effects. There is a clear need for improved therapeutic options. Several decades of research have substantially expanded our understanding of the basic mechanisms of AF. Ectopic firing and reentrant activity have been identified as the predominant mechanisms for arrhythmia initiation and maintenance. However, it has become clear that the clinical factors predisposing to AF and the cellular and molecular mechanisms involved are extremely complex. Moreover, all AF-promoting and maintaining mechanisms are dynamically regulated and subject to remodeling caused by both AF and cardiovascular disease. Accordingly, the initial presentation and clinical progression of AF patients is enormously heterogeneous. Improved understanding of arrhythmia mechanisms is likely needed to facilitate therapeutic innovation, but this assumption has not to our knowledge been carefully examined. Here, we review recent insights into the basic mechanisms of AF, critically analyze the role of basic-research insights in the development of presently-used anti-AF therapeutic options, and assess the potential value of contemporary experimental discoveries for future therapeutic innovation. Finally, we highlight some of the important challenges to the translation of basic science findings to clinical application.
Article
Full-text available
To investigate the expression profiles of long non-coding RNAs (lncRNAs) in atrial fibrillation (AF), atrial tissues from 3 AF patients and 3 non-AF patients that were collected for lncRNA expression microarray analyses to explore the role of lncRNA in the pathogenesis of AF. Gene Ontology (GO) categories and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed to identify the main functions of the differentially expressed genes and AF-related pathways. A total of 219 lncRNAs was found to be differentially expressed between AFs and controls. Among them, 156 were upregulated and 63 were downregulated. Eight out of 10 dysregulated lncRNAs such as uc001eqh.1 were validated by quantitative real-time PCR. GO categories, pathway analyses, and interaction network showed a consistent result that differentially expressed genes contribute to the pathogenesis of AF. In conclusion, the findings of our study provide a perspective on lncRNA in AF and the foundation for further study of the biological functions of lncRNAs in AF.
Article
Full-text available
Voltage-gated Kv1.1 channels encoded by the Kcna1 gene are traditionally regarded as being neural-specific with no known expression or intrinsic functional role in the heart. However, recent studies in mice reveal low-level Kv1.1 expression in heart and cardiac abnormalities associated with Kv1.1-deficiency suggesting that the channel may have a previously unrecognized cardiac role. Therefore, this study tests the hypothesis that Kv1.1 channels are associated with arrhythmogenesis and contribute to intrinsic cardiac function. In intra-atrial burst pacing experiments, Kcna1-null mice exhibited increased susceptibility to atrial fibrillation (AF). The atria of Kcna1-null mice showed minimal Kv1 family ion channel remodeling and fibrosis as measured by qRT-PCR and Masson's trichrome histology, respectively. Using RT-PCR, immunocytochemistry, and immunoblotting, KCNA1 mRNA and protein were detected in isolated mouse cardiomyocytes and human atria for the first time. Patients with chronic AF (cAF) showed no changes in KCNA1 mRNA levels relative to controls; however, they exhibited increases in atrial Kv1.1 protein levels, not seen in paroxysmal AF patients. Patch-clamp recordings of isolated human atrial myocytes revealed significant dendrotoxin-K (DTX-K)-sensitive outward current components that were significantly increased in cAF patients, reflecting a contribution by Kv1.1 channels. The concomitant increases in Kv1.1 protein and DTX-K-sensitive currents in atria of cAF patients suggest that the channel contributes to the pathological mechanisms of persistent AF. These findings provide evidence of an intrinsic cardiac role of Kv1.1 channels and indicate that they may contribute to atrial repolarization and AF susceptibility.
Article
Atrial fibrillation is a prominent cause of morbidity and mortality in developed countries. Treatment strategies center on controlling atrial rhythm or ventricular rate. The need for anticoagulation is an independent decision from the rate versus rhythm control debate. This review discusses novel biological strategies that have potential utility in the management of atrial fibrillation. Rate controlling strategies predominately rely on G-protein gene transfer to enhance cholinergic or suppress adrenergic signaling pathways in the atrioventricular node. Calcium channel blocking gene therapy and fibrosis enhancing cell therapy have also been reported. Rhythm controlling strategies focus on disrupting reentry by enhancing conduction or suppressing repolarization. Efforts to suppress inflammation and apoptosis are also under study. Resistance to blood clot formation has been shown with thrombomodulin. These strategies are in various stages of preclinical development.
Article
Atrial fibrillation (AF) is associated with structural, electrical, and contractile remodeling of the atria. Development and progression of atrial fibrosis is the hallmark of structural remodeling in AF and is considered the substrate for AF perpetuation. In contrast, experimental and clinical data on the effect of ventricular fibrotic processes in the pathogenesis of AF and its complications are controversial. Ventricular fibrosis seems to contribute to abnormalities in cardiac relaxation and contractility and to the development of heart failure, a common finding in AF. Given that AF and heart failure frequently coexist and that both conditions affect patient prognosis, a better understanding of the mutual effect of fibrosis in AF and heart failure is of particular interest. In this review paper, we provide an overview of the general mechanisms of cardiac fibrosis in AF, differences between fibrotic processes in atria and ventricles, and the clinical and prognostic significance of cardiac fibrosis in AF. Copyright © 2015 American College of Cardiology Foundation. Published by Elsevier Inc. All rights reserved.
Article
Atrial fibrillation is a prominent cause of morbidity and mortality in developed countries. Treatment strategies center on controlling atrial rhythm or ventricular rate. The need for anticoagulation is an independent decision from the rate vs. rhythm control debate. This review discusses novel biological strategies that have potential utility in the management of atrial fibrillation. Rate controlling strategies predominately rely on G-protein gene transfer to enhance cholinergic or suppress adrenergic signaling pathways in the atrioventricular node. Calcium channel blocking gene therapy and fibrosis enhancing cell therapy have also been reported. Rhythm controlling strategies focus on disrupting reentry by enhancing conduction or suppressing repolarization. Efforts to suppress inflammation and apoptosis are also under study. Resistance to blood clot formation has been shown with thrombomodulin. These strategies are in various stages of preclinical development.
Article
Background Atrial fibrillation (AF) is associated with metabolic stress, which activates adenosine monophosphate-regulated protein kinase (AMPK). Objectives This study sought to examine AMPK response to AF and associated metabolic stress, along with consequences for atrial cardiomyocyte Ca2+ handling. Methods Calcium ion (Ca2+) transients (CaTs) and cell shortening (CS) were measured in dog and human atrial cardiomyocytes. AMPK phosphorylation and AMPK association with Ca2+-handling proteins were evaluated by immunoblotting and immunoprecipitation. Results CaT amplitude and CS decreased at 4-min glycolysis inhibition (GI) but returned to baseline at 8 min, suggesting cellular adaptation to metabolic stress, potentially due to AMPK activation. GI increased AMPK-activating phosphorylation, and an AMPK inhibitor, compound C (CompC), abolished the adaptation of CaT and CS to GI. The AMPK activator 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) increased CaT amplitude and CS, restoring CompC-induced CaT and CS decreases. CompC decreased L-type calcium channel current (ICa,L), along with ICa,L-triggered CaT amplitude and sarcoplasmic reticulum (SR) Ca2+ content under voltage clamp conditions in dog cells and suppressed CaT and ICa,L in human cardiomyocytes. Small interfering ribonucleic acid-based AMPK knockdown decreased CaT amplitude in neonatal rat cardiomyocytes. L-type Ca2+ channel α subunits coimmunoprecipitated with AMPKα. Atrial AMPK-activating phosphorylation was enhanced by 1 week of electrically maintained AF in dogs; fractional AMPK phosphorylation was increased in paroxysmal AF and reduced in longstanding persistent AF patients. Conclusions AMPK is activated by metabolic stress and AF, and helps maintain the intactness of atrial ICa,L, Ca2+ handling, and cell contractility. AMPK contributes to the atrial compensatory response to AF-related metabolic stress; AF-related metabolic responses may be an interesting new therapeutic target.
Article
Classical concepts of atrial fibrillation (AF) have been rooted in Moe’s multiple-wavelet hypothesis and simple cellular-automaton computer model. Recent experimental work has raised questions about the multiple-wavelet mechanism, suggesting a discrete “driver region” underlying AF. We reexplored the theoretical basis for AF with a 2-dimensional computer model of a 5×10-cm sheet of atrial cells with realistic ionic and coupling properties. Vagal actions were formulated based on patch-clamp studies of acetylcholine (ACh) effects. In control, a single extrastimulus resulted in a highly meandering unstable spiral wave. Simulated electrograms showed fibrillatory activity, with a dominant frequency (DF, 6.5 Hz) that correlated with the mean rate. Uniform ACh reduced core meander of the spiral wave by ≈70% (as measured by the standard deviation of spiral-wave tip position) and accelerated the DF to 17.0 Hz. Simulated vagally induced refractoriness heterogeneity caused wavefront breakup as accelerated reentrant activity in regions of short refractoriness impinged on regions unable to respond in a 1:1 fashion because of longer refractoriness. In 7 simulations spanning the range of conditions giving sustained AF, 5 were maintained by single dominant spiral waves. On average, 3.0±1.3 wavelets were present (range, 1 to 7). Most wavelets were short-lived and did not contribute to AF maintenance. In contrast to predictions of the multiple-wavelet hypothesis, but in agreement with recent experimental evidence, our model indicates that AF can result from relatively stable primary spiral-wave generators and is significantly organized. Our results suggest that vagal AF may arise from ACh-induced stabilization of the primary spiral-wave generator and disorganization of the heterogeneous tissue response. The full text of this article is available at http://www.circresaha.org.
Article
Atrial fibrillation (AF) is the most common sustained cardiac arrhythmia worldwide requiring therapy. Despite recent advances in catheter-based and surgical therapy, antiarrhythmic drugs (AAD) remain the mainstay of treatment for symptomatic AF. However, response in individual patients is highly variable with over half the patients treated with rhythm control therapy experiencing recurrence of AF within a year. Contemporary AADs used to suppress AF are incompletely and unpredictably effective and associated with significant risks of proarrhythmia and non-cardiac toxicities. Furthermore, this 'one-size' fits all strategy for selecting antiarrhythmics is based largely on minimizing risk of adverse effects rather than on the likelihood of suppressing AF. The limited success of rhythm-control therapy is in part due to heterogeneity of the underlying substrate, interindividual differences in disease mechanisms, and our inability to predict response to AADs in individual patients. Genetic studies of AF over the last decade have revealed that susceptibility to and response to therapy for AF is modulated by the underlying genetic substrate. However, the bedside application of these new discoveries to the management of AF patients has thus far been disappointing. This may in part be related to our limited understanding about genetic predictors of drug response in general, the challenges associated with determining efficacy of response to AADs and lack of randomized genotype-directed clinical trials. Nonetheless, recent studies have shown that common AF susceptibility risk alleles at the chromosome 4q25 locus modulated response to AADs, electrical cardioversion and ablation therapy. This monograph discusses how genetic approaches to AF have not only provided important insights into underlying mechanisms but also identified AF sub-types that can be better targeted with more mechanism-based 'personalized' therapy.
Article
-Antiarrhythmic management of atrial fibrillation (AF) remains a major clinical challenge. Mechanism-based approaches to AF therapy are sought to increase effectiveness and to provide individualized patient care. K2P3.1 (TASK-1) two-pore-domain K(+) (K2P) channels have been implicated in action potential regulation in animal models. However, their role in the pathophysiology and treatment of paroxysmal and chronic AF patients is unknown. -Right and left atrial tissue was obtained from patients with paroxysmal (p) or chronic (c)AF and from sinus rhythm (SR) controls. Ion channel expression was analyzed by quantitative real-time PCR and Western blot. Membrane currents and action potentials were recorded using voltage- and current-clamp techniques. K2P3.1 subunits exhibited predominant atrial expression, and atrial K2P3.1 transcript levels were highest among functional K2P channels. K2P3.1 mRNA and protein levels were increased in cAF. Enhancement of corresponding currents in the right atrium resulted in shortened action potential duration at 90% of repolarization (APD90) compared to patients in SR. By contrast, K2P3.1 expression was not significantly affected in pAF subjects. Pharmacological K2P3.1 inhibition prolonged APD90 in atrial myocytes from cAF patients to values observed among SR subjects. -Enhancement of atrial-selective K2P3.1 currents contributes to APD shortening in cAF patients, and K2P3.1 channel inhibition reverses AF-related APD shortening. These results highlight the potential of K2P3.1 as a novel drug target for mechanism-based AF therapy.