Next Article in Journal
Markers of Metabolic Abnormalities in Vitiligo Patients
Previous Article in Journal
Unbalanced Expression of Structural Genes in Carotenoid Pathway Contributes to the Flower Color Formation of the Osmanthus Cultivar ‘Yanzhi Hong’
Previous Article in Special Issue
Enhanced Late INa Induces Intracellular Ion Disturbances and Automatic Activity in the Guinea Pig Pulmonary Vein Cardiomyocytes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Atrial Fibrillation and Underlying Structural and Electrophysiological Heterogeneity

Department of Cardiovascular Medicine, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(18), 10193; https://doi.org/10.3390/ijms251810193
Submission received: 19 August 2024 / Revised: 16 September 2024 / Accepted: 19 September 2024 / Published: 23 September 2024
(This article belongs to the Special Issue Cardiac Arrhythmia: Molecular Mechanisms and Therapeutic Strategies)

Abstract

:
As atrial fibrillation (AF) progresses from initial paroxysmal episodes to the persistent phase, maintaining sinus rhythm for an extended period through pharmacotherapy and catheter ablation becomes difficult. A major cause of the deteriorated treatment outcome is the atrial structural and electrophysiological heterogeneity, which AF itself can exacerbate. This heterogeneity exists or manifests in various dimensions, including anatomically segmental structural features, the distribution of histological fibrosis and the autonomic nervous system, sarcolemmal ion channels, and electrophysiological properties. All these types of heterogeneity are closely related to the development of AF. Recognizing the heterogeneity provides a valuable approach to comprehending the underlying mechanisms in the complex excitatory patterns of AF and the determining factors that govern the seemingly chaotic propagation. Furthermore, substrate modification based on heterogeneity is a potential therapeutic strategy. This review aims to consolidate the current knowledge on structural and electrophysiological atrial heterogeneity and its relation to the pathogenesis of AF, drawing insights from clinical studies, animal and cell experiments, molecular basis, and computer-based approaches, to advance our understanding of the pathophysiology and management of AF.

1. Introduction

Atrial fibrillation (AF), a common arrhythmia, has been associated with increased risks of heart failure and stroke, consequently exacerbating patient prognoses. Despite its prevalence, AF treatments have not yet been optimized. Pharmacotherapy for AF using antiarrhythmic drugs is characterized by limited efficacy, with no substantial evidence indicating improvements in long-term outcomes [1]. Upstream therapies, such as angiotensin-converting enzyme inhibitors (ACEis) and angiotensin-receptor blockers, have not been proven to prevent the occurrence of AF [2,3]. Despite the effectiveness of catheter ablation for paroxysmal AF, it has therapeutic limitations for persistent AF. As AF progresses from the paroxysmal to the persistent phase, it becomes necessary to focus not only on the triggers but also the vulnerable substrates that drive fibrillatory activity.
The detailed pathophysiological mechanisms underlying AF remain largely elusive. In particular, the processes by which the triggers induce electrical fibrillatory activity and the factors that sustain this abnormal activity remain poorly understood. Furthermore, the determining factors of the seemingly disorganized propagation pattern during AF remain unclear. These mechanisms have been investigated from various perspectives, such as anatomical structure, tissue, sarcolemmal ion currents, and electrophysiology. One of the contributing factors to the vulnerable substrates for AF, namely arrhythmogenic substrate, is heterogeneity. The electrical activity in an atrium is believed to repeat even and synchronous excitement unless heterogeneity exists. When a normal atrial excitation wave advances evenly and synchronously on a homogeneous substrate but encounters a heterogeneous point, it breaks up (wavebreak) and occasionally rotates. If a cascade of wavebreaks is induced, the wave begins to exhibit chaotic propagation. As the propagating wave rotates consistently around the point of the wavebreak, if sustained, it is termed a rotor, which typically has a short cycle length and produces rapid tachycardia. A rotor can be a driver of AF, which represents a focal source demonstrating repetitive and fast activity that propagates outward from the source. The process from the inception of a wavebreak to the sustenance of fibrillation can be attributed to heterogeneity [4]. Many studies have reported a correlation between atrial heterogeneity and susceptibility to AF; however, how these atrial heterogeneities correlate with AF remains unclear, resulting in the unawareness that the underlying heterogeneities are a causative factor of AF. Two main types of heterogeneity exist: structural and electrophysiological. Structural heterogeneity can be further viewed based on three subcategories: macroscopic (segmental), mesoscopic (tissue), and microscopic (cellular and molecular) structure. Each level of structural heterogeneity can affect electrophysiological heterogeneity, contributing to the substrate for AF [5,6,7] (Figure 1).
Highlighting atrial heterogeneity will help us understand its contribution to AF and investigate new therapeutic strategies for AF [8]. Herein, we will summarize the atrial structural and electrophysiological heterogeneities which are incorporated into the pathophysiology of AF, drawing insights from human clinical studies, animal experiments, cell experiments, and theoretical models.

2. Macroscopic Heterogeneity

The atria are complex structures composed of various components, including chambers divided by the atrial septum, venous elements, appendages, and vestibules, rather than a single uniform structure (Figure 2). At the boundaries of these segments or within each segment, inherent heterogeneities exist, even in the absence of pathophysiological conditions. In particular, venous components, such as the pulmonary vein (PV), vena cava, and coronary sinus (CS), have common peculiar features. They have in common myocardial tissue stretching from the atrium, which is referred to as a myocardial sleeve. The features of the myocardial sleeve differ from those of the atrial myocardium in terms of myocardial thickness and arrangement. Around the boundary between the veins and atrium, abrupt changes in musculature structure may cause electrical divergence or convergence, possibly resulting in an electrical source–sink mismatch. Meanwhile, the right atrial (RA) appendage is composed of pectinate muscles, which have an uneven and rough surface characterized by the branching and overlapping arrangement of trabeculae. Moreover, the crista terminalis, the connection between pectinate muscles and the relatively smooth muscle region derived from the sinus venosus, generates an electrical boundary. The complex structure of pectinate muscles may contribute to the arrhythmogenic substrates [9]. Although these distinct structural heterogeneities are not arrhythmogenic in most healthy individuals, they become arrhythmogenic and evoke AF when certain pathophysiological conditions arise. The causative factors of AF based on anatomical heterogeneity are listed in Table 1.

2.1. Pulmonary Vein

PVs play prominent roles in AF, and catheter ablation for the electrical isolation of PVs has become an established treatment because the majority of AF is initiated by ectopic discharges mostly within the PVs. Abnormal automaticity and triggered activity have been suggested as the triggers of AF. The architecture of the PV myocardial sleeve can be a trigger source for AF, and a few mechanisms have been advocated in canine experiments. Previous anatomic-electrophysiological studies in isolated PV specimens have demonstrated that PVs contain a mixture of pacemakers and myocardial cells [10]. These pacemaker cells with spontaneous activity have a significantly lower density of inward rectifier potassium current, leading to less negative diastolic potential, with which increased automaticity is generally associated [11]. Thus, pacemaker activity from these cells is thought to result in the formation of the ectopic beats that initiate AF. Several studies using the atria of autopsy patients support these findings in canine experiments. Light microscopy with periodic acid–Schiff staining and electron microscopy identified that the PV has cells that are histologically and morphologically similar to node, transitional, and Purkinje cells [12]. Not only enhanced automaticity, but also triggered activity is referred to as impulse initiation in cardiac fibers which is dependent on afterdepolarizations [13]. Animal experiments showed that isolated PV cardiomyocytes (CMs) from healthy dogs and rabbits manifested arrhythmogenic afterdepolarizations, suggesting that triggered activity may account for the trigger of AF within the PV [11,14]. In addition, PVs contain enriched autonomic innervation, and the autonomic nervous system is suggested to enhance ectopic triggers via sympathetic or vagal activation [15,16]. These findings indicate that the PV myocardial sleeve may act as a focal trigger. Furthermore, not only do PVs act as a trigger of AF, but they also contribute to the initiating and sustaining mechanism of AF [17]. The arrhythmogenicity of PVs can be evidenced in humans by persisting tachyarrhythmia confined to the PV during sinus rhythm after its isolation, suggesting that the PV myocardial sleeve is involved in the maintenance mechanism of AF. Furthermore, many of the focal left atrial (LA) tachycardias after segmental PV isolation have been reported to be caused by a focal reentrant circuit located at the PV ostium [18]. From a structural perspective, this is partly attributed to the arrangement and thickness of the myocyte bundles within the PV myocardial sleeves and at the PV-LA junction. The sleeves are composed of circularly or spirally oriented bundles of myocytes with additional bundles that are longitudinally or obliquely oriented [19]. These intricate arrangements of myocardial fibers in the PVs, which are often diagonal or perpendicular to LA CMs near the PV-LA junction, contribute to conduction slowing because of the conduction anisotropy, serving as a possible reason for their complexity within the PV and even at the PV-LA junction [19,20]. According to a histological study performed on human hearts, the thickness of the sleeves is not uniform. The myocardial thickness of the PV is almost uniformly thin (0.3–0.8 mm) in contrast to that of LA (1.1–2.6 mm). An abrupt change in myocardial thickness exists around the PV-LA junction (mean 1.1 mm), and the sleeve tapers distally [21]. The differential muscle narrowing and complex conduction patterns at the PV-LA junction can provide a robust anatomical basis for source–sink mismatch and conduction anisotropy. The arrangement of muscle bundles between different ipsilateral PVs, the carina, is also convoluted and interwoven, which may produce non-uniform anisotropic properties [22,23]. The catheter ablation strategy of PV antrum isolation to encircle ipsilateral PVs by ablating on the atrial side of the PV-LA junction has been widely accepted. PV antrum isolation has achieved a higher clinical success rate and a lower incidence of postprocedural atrial arrhythmias [24], since it can be attributed not only to the isolation of PVs but also to the isolation of the PV-LA junction and the ipsilateral carina. These structural characteristics of the PV myocardium potentially represent a primary substrate for AF.

2.2. Vena Cava

Myocardial sleeves within the superior vena cava (SVC) can also serve as the origins of AF [25], and the SVC accounts for the major portion of non-PV foci [26,27,28]. Unlike PVs, limited data regarding the morphology of SVC myocardial sleeves have been published. However, a previous canine study revealed that half of the isolated CMs from SVC myocardial sleeves have pacemaker activity, showing spontaneous depolarization and less negative resting membrane potentials [29]. The presence of CMs with pacemaker activity suggests that automaticity plays a role in the arrhythmogenicity of the SVC. This study also demonstrated that the infusion of autonomic agents, such as isoproterenol, atropine, and phenylephrine, into SVC CMs accelerated the spontaneous activity and induced afterdepolarizations, indicating that the enhanced automaticity and afterdepolarization are involved in the arrhythmogenic activity of the SVC [29]. Additionally, a recent study showed that the muscle fibers of SVC myocardial sleeves exhibit a morphology similar to that of Purkinje fibers [30], supporting the indication of possible arrhythmogenicity in the SVC. In addition to the cell types in SVC myocardial sleeves, the geometric size of SVC myocardial sleeves can be an important element of SVC arrhythmogenicity. In a clinical study, an SVC myocardial sleeve longer than 30 mm has been reported as an independent risk factor for arrhythmogenic trigger sources from the SVC [31]. Long SVC myocardial sleeves were also reported to be correlated with the SVC potential amplitude, reflecting greater myocardial volume. Thus, these findings suggest that long myocardial extension and large amounts of CMs in the SVC can be a source of arrhythmogenicity. SVC myocardial sleeves may serve not only as a trigger of AF, but also as a perpetuator that initiates and sustains AF. Similar to PV myocardial sleeves, the arrangement of musculature in myocardial sleeves within the SVC is intricate. The cardiac fibers are bundled and disposed in longitudinal, oblique, or circumferential directions in the venous wall, which may cause anisotropic conduction within the SVC. Based on high-resolution electroanatomic mapping, slow and heterogeneous conduction has been observed during sinus rhythm and is exaggerated by premature stimulation, indicating that SVC myocardial sleeves have the potential for a reentrant mechanism [32]. Several observations that the SVC acts not only as an AF trigger but also as a driver have been reported [33,34]. Miyazaki et al. reported that SVC fibrillation was confined to the SVC after its electrical isolation, indicating the existence of the driver within the SVC [34]. Therefore, the intricate structure and heterogeneity of the SVC may be involved in the AF substrate, which could potentially facilitate the heterogeneity of the underlying electrophysiological property. For non-responders to PV isolation and patients undergoing repeated ablation, SVC isolation should be considered, as it can separate a highly heterogeneous region with a possible trigger or an AF perpetuator from the RA [35].
The musculature extension into the inferior vena cava (IVC) is shorter than that into the SVC, rendering a lower possibility of an AF source [36,37]. However, a few reports have shown that the IVC serves as a driver for the maintenance of AF in addition to being a trigger [38,39]. The optimal IVC ablation strategy for AF remains unknown because of the limited studies, although focal ablation and IVC isolation have been reported [39].

2.3. Coronary Sinus

The CS occupies the atrioventricular groove, and its wall is covered with a myocardial sleeve. The CS is a remnant of the sinus venosus, and its muscle sleeve can be an extension of the RA myocardium over the CS. Histological studies have shown that the CS myocardial sleeve has muscular connections with both RA and LA fibers [40,41]. Muscular fibers of varying thickness arise from the myocardial sleeve along the inferior mitral annulus, providing electrical continuity with the LA. Striated myocardial connections exist between the CS and LA ranging from one to two fascicles surrounded by insulating compartments of adipose tissue [42]. The CS muscle sleeve and its branching connections to the atrium have been implicated in the genesis of atrial tachyarrhythmias. Non-PV ectopic beats arising from the CS have also been reported in clinical studies, suggesting that AF may originate within the CS [27,43]. Little evidence regarding the abnormal automaticity within the CS has been reported; however, some previous reports on CMs isolated from canine CS have confirmed the triggered activity [44,45]. Conversely, several studies have demonstrated that the CS myocardial sleeve may serve not only as a source generating a focal trigger but also as a part of a reentrant circuit [46,47]. The complex orientation of muscular fibers around the CS and their discrete insertion sites within the LA construct a heterogeneous structure and may facilitate the source–sink mismatch, leading to conduction disturbance within the connections [48]. These electrical connections between the CS and both atria may be involved in arrhythmogenesis by forming a part of the reentrant circuit [49]. Optical mapping in a canine model showed a functional conduction block at the RA-CS junction, and this site was involved in the rapid pacing-induced reentrant circuit [50]. Activation maps derived from another canine model demonstrated that the CS musculature developed unstable reentry and AF, which were prevented by the isolation of CS musculature from LA tissue [51]. Similarly, in a clinical study, electrical dissociation of the CS from the LA led to less inducibility of sustained AF after PV isolation, which suggests that the CS is involved in perpetuating AF [46].

2.4. Crista Terminalis and Pectinate Muscles

The RA appendage is readily distinguished from the RA because of the crista terminalis and pectinate muscles [52]. The crista terminalis has been demonstrated as a conduction barrier during typical atrial flutter. The investigations of the conduction properties within the crista terminalis demonstrated that it has marked anisotropic conduction with enhanced conduction velocity in the longitudinal direction compared to that in the transverse direction [53]. Meanwhile, in terms of the genesis of AF, animal experiments reported that isolated cells from the crista terminalis in rabbits have spontaneous pacemaker activity, whereas those in dogs showed delayed afterdepolarization induced by norepinephrine [53,54,55]. In a clinical study, the non-PV ectopic triggers of paroxysmal AF originating from the crista terminalis were observed [27]. These findings endorse the view that spontaneous activation may be initiated at the crista terminalis. Moreover, the complex branching structure of the pectinate muscle network has been proposed to provide a substrate for complex patterns of propagation during AF. Pectinate muscles spread throughout the wall of the atrial appendage with a branching and overlapping arrangement. The pectinate muscles are composed of ridges; however, the wall in the groove between the ridges is very thin [36]. This complex structure results in a source–sink mismatch at the branching site of bundles. An electrophysiological study of canine RA tissue showed that a premature stimulus induced conduction delay along the ridge of large pectinate muscles, leading to a wavebreak and resulting in the initiation of reentry [56]. The direction of the propagation in the RA appendage was also demonstrated to be rate-dependent with variability from beat to beat at the site of pectinate bundles, leading to a transformation of fibrillatory conduction [9]. Thus, pectinate muscles may contribute to the vulnerable substrate, allowing the sustenance of AF. Owing to the recent progress in electrode mapping techniques, AF rotational drivers were identified and described within the RA appendage [57,58].
Pectinate muscles are much less extensive in the LA appendage, and the LA appendage lacks a muscular bundle equivalent to the crista terminalis [36]. Despite these anatomical characteristics of the LA appendage being distinct from the RA appendage, the LA appendage has been identified as a possible trigger origin and substrate for AF maintenance after PV isolation [59,60]. Previous studies have demonstrated that LA appendage isolation in addition to PV isolation by catheter ablation reduced AF recurrence compared with PV isolation alone [61,62]. However, procedures with LA appendage isolation had higher rates of thromboembolism compared to those without it [63].

2.5. Left Atrial Posterior Wall

The LA posterior wall is a major non-PV trigger that initiates paroxysmal AF [27]. Previous studies have shown that the shortest AF cycle length and highest dominant frequency during AF are found in the LA posterior wall in dogs, sheep, and patients with chronic AF, implying that the LA posterior wall can be a potential source of AF [64,65,66,67]. However, to date, the reason why the LA posterior wall is so arrhythmogenic remains unclear. Of note, the LA posterior wall has a common embryologic origin with PVs, which may contribute to the factors that make the LA posterior wall distinct from other areas of the atrial wall [68,69,70,71,72]. However, the way in which the embryologic origin of the LA posterior wall is associated with the sources of AF remains elusive. On the other hand, an anatomical factor potentially characterizes the arrhythmogenicity in the LA posterior wall, despite the lack of evidence regarding the involvement of abnormal automaticity and triggered activity. The fiber orientation and myocardial thickness of the LA posterior wall abruptly change with partial involvement of the septopulmonary bundle. This bundle arises from the anterior interatrial raphe, ascends obliquely, and combines with longitudinal fibers from the anterior vestibule to run between the left and right PVs on the LA posterior wall, forming heterogeneous fiber orientation [73]. Furthermore, the LA posterior wall exhibits varying degrees of thickness, with the largest increase in myocardial thickness occurring at the border of the septopulmonary bundle [73]. These abrupt changes in fiber orientation and thickness may characterize the electrophysiological properties within the LA posterior wall. Markides et al. demonstrated that a line of functional conduction block in the LA posterior wall exists, running craniocaudally between the PVs during sinus rhythm and PV ectopy in humans [74]. They also demonstrated that patients with AF developed this line of conduction delay, corresponding to a change in fiber orientation in this region. Wavefronts entering the atrium from the PV interact with this functional line of conduction block, resulting in the reentry formation of daughter wavefronts. In an ovine study, electric source–sink mismatch and the subsequent wavebreaks mostly appeared at the septopulmonary bundle near the right superior PV, where the myocardial thickness dramatically expanded [73]. These findings indicate that the structural peculiarity may promote arrhythmogenicity in the LA posterior wall.
To summarize this section, macroscopic views show several common features throughout the atrial region. The pacemaker activity underlying myocardial sleeves within PV and SVC, and the arrhythmogenic afterdepolarization in the myocytes of PV, SVC, and CS, are potential triggers of AF. The intricate and anisotropic arrangement, and varying thickness of muscle fibers can provide an anatomical basis for source–sink mismatch and anisotropic conduction, which are causative mechanisms of the reentrant circuit, located in the PV (including PV-LA junction), SVC, CS-LA junction, crista terminalis, pectinate muscles, and septopulmonary bundle of the LA posterior wall.
Table 1. Macroscopic heterogeneities predisposing to AF. RA: right atrium, RAA: right atrial appendage, LA: left atrium, and LAA: left atrial appendage.
Table 1. Macroscopic heterogeneities predisposing to AF. RA: right atrium, RAA: right atrial appendage, LA: left atrium, and LAA: left atrial appendage.
Macroscopic Structure
RegionStructureCausative FactorSpecies and ReferenceMechanism for ArrhythmiaSection
Pulmonary vein (PV)PV myocardial sleevepacemaker activitydog [10,11], human [12]arrhythmogenic trigger2.1
afterdepolarizationdog, rabbit [11,14]arrhythmogenic trigger
intricate muscular bundle arrangementhuman [19]reentry
autonomic innervationhuman [15,16]arrhythmogenic trigger
PV-LA junctionvarying thickness of muscular fiberhuman [19], dog [20]reentry
intricate muscular bundle arrangementhuman [21]reentry
carinaintricate muscular bundle arrangementhuman [22,23]reentry
Superior vena cava (SVC)SVC myocardial sleevepacemaker activitydog [29], human [30]arrhythmogenic trigger2.2
afterdepolarizationdog [29]arrhythmogenic trigger
intricate muscular bundle arrangementhuman [33,34]reentry
Coronary sinus (CS)CS myocardial sleeveafterdepolarizationdog [44,45]arrhythmogenic trigger2.3
RA-CS connectionfunctional conduction blockdog [50]reentry
CS-LA connectionintricate muscular bundle arrangementhuman [46,47,48]reentry
varying thickness of muscular fiberreentry
Crista terminalis (CT)CTpacemaker activityrabbit [54,55]arrhythmogenic trigger2.4
triggered activitydog [53]arrhythmogenic trigger
anisotropic conductiondog [53]reentry
Pectinate muscleRAA (LAA)branching structure (ridge and groove)human [36], dog [56]reentry
Left atrial posterior wallseptopulmonary bundleintricate muscular bundle arrangementsheep [73], human [74]reentry2.5
varying thickness of muscular fiberreentry

3. Mesoscopic Heterogeneity

Anatomical structural heterogeneity at the segmental level, as described above, is inherently present and may become a potential factor of triggers and substrates for AF. However, most AFs are believed to occur when combined with other atrial pathophysiological conditions. Particularly in heart failure, increased atrial volume overload leads to the elevation of atrial pressure and stretching of atrial muscle, resulting in the reconstruction of tissue and the alteration of electrophysiological properties, known as atrial remodeling [6]. Atrial remodeling histopathologically involves the activation and proliferation of myofibroblasts (MFs), which are responsible for the uncontrolled deposition of the extracellular matrix. The enhanced fibrosis leads to tissue anisotropy, and obstructs the electrical wave propagation [75,76]. The pathological sources of AF have been proposed from the view of tissue-level heterogeneity.

3.1. Myofibroblasts and Fibrosis

Fibrosis refers to an increased deposition of collagen and other extracellular matrix proteins in the interstitial space. The excessive deposition of collagen enlarges interstitial spaces and decouples electrical cell-to-cell communication at gap junctions, resulting in the reduction in inter-myocyte electrical coupling. As described above, PVs are a major segment that proposes high arrhythmogenicity. An important tissue feature of PVs is the patchy areas of fibrosis, which can be detected even within healthy PVs [19]. In addition, a higher degree of fibrosis in the atrial myocardium extends to the PVs in patients with AF [21]. The inherent and acquired fibrosis within PVs is a possible source of arrhythmogenicity. Furthermore, fibroblast proliferation not only promotes collagen production which serves as an electrical insulator, but also mediates the electrical coupling with CMs. MFs are basically unexcitable cells, but they can form hetero-cellular gap junctions with CMs [77]. Some in vitro experiments using CMs isolated from animal hearts or in silico experiments demonstrated that the electrical coupling with MFs depolarizes the resting membrane potential of CMs [77,78,79]. According to a numerical study, these actions of MFs alter the electrophysiological characteristics of CMs, such as the shortening of myocyte action potential duration (APD) and a decrease in myocyte maximum depolarization velocity, leading to slow conduction velocity [80]. As a result, both the extracellular matrix and MFs bring about further complex electrophysiological changes, potentially causing arrhythmogenicity.

3.2. Heterogeneous Distribution of Fibrosis

As atrial remodeling progresses, the heterogeneous distribution of fibrosis is exacerbated, which is one of the factors promoting the pathophysiology of AF. To assess the tissue heterogeneity caused by fibrosis in an entire atrium, one non-invasive method for visualizing atrial fibrosis is cardiac magnetic resonance (CMR) imaging. Late gadolinium-enhanced CMR (LGE-CMR) has been recognized as an indicator to quantify the scar areas in an atrium [81,82], making LGE-CMR a promising tool for visualizing atrial fibrosis. LGE-CMR in patients with AF showed that the fibrotic area is heterogeneously distributed, and preferentially located at the LA posterior wall and around the antrum of the left inferior PV [83,84,85]. Previous studies have demonstrated that more advanced fibrosis detected by LGE-CMR is related to the recurrence of AF after PV isolation [86,87]. Therefore, to investigate the efficacy of catheter ablation targeting atrial fibrosis detected by LGE-CMR, the DECAAF II trial executed the LGE-CMR-guided strategy of identifying fibrotic regions compared with conventional methods for persistent AF [88]. However, no significant difference in the recurrence rate of AF was observed between the two groups. Much debate remains regarding the therapeutic applications based on the LGE-CMR. Another method currently used to estimate fibrotic tissue is electroanatomic voltage mapping. Low-voltage zone (LVZ), defined as an area with less than a certain cutoff using bipolar voltage mapping, has been widely used as a surrogate for local fibrotic tissue [89]. Several previous studies have shown that the most frequent localization of the LVZ tends to be within the LA anterior wall, followed by the septum and LA posterior wall [85,90]. Another observational study suggested that this segmental LVZ distribution pattern advances as AF perpetuates; initially in the LA anterior wall, followed by the LA posterior wall [91]. The severity of fibrosis estimated by the LVZ during sinus rhythm is associated with poor outcomes after PV isolation [92,93]. Several meta-analyses have reported that LVZ-guided ablation for persistent AF provides a significant reduction in recurrence rates [94,95,96]. In a study comparing the fibrotic areas shown by LGE-CMR and LVZ in the LA, LGE-CMR and LVZ covered 55% and 24% of the LA, respectively, while 61% of the LVZ was co-located with the LGE area, and only 28% of the LGE area displayed LVZ [85]. A high mismatch was observed in the distribution and volume of fibrosis detected by the LGE-CMR compared with that detected by the LVZ. The cause of this significant spatial mismatch is still under debate. Considering the association of the fibrotic area detected by these modalities and the electrophysiological properties, both of them correlate with a decrease in atrial conduction velocity [97,98,99]. On the other hand, the relationship between the LGE area and local reentry or rotor remains debated at present. While some reports suggest a higher prevalence of reentry in areas where enhancement is observed on LGE-CMR, other reports do not support this correlation [100,101,102,103]. Similarly, a wide discrepancy exists in the distribution of the LVZ and sites of identified rotors [104]. In fact, several studies have reported that reentrant AF sources were identified at only 8–37% of the LVZ [85,104].

3.3. Fibrosis and Arrhythmogenicity

Although the heterogeneous distribution of fibrosis does not necessarily allow for identifying the site of AF sources, the factors causing local conduction disturbances are believed to be partly ascribed to increased fibrosis [105]. Fibrosis has been indicated as the primary factor causing AF. However, among the fibrotic volume, distribution, or both, the factor that contributes the most to arrhythmogenesis remains unclear. Previous studies have implied that the heterogeneity of fibrotic distribution is a more contributive factor to the development of AF [106,107]. In a recent study with a porcine ischemic model, heterogeneous fibrosis in the LA rather than the overall level of fibrosis was associated with an increased AF susceptibility [108]. Several studies have demonstrated that the areas adjacent to dense fibrosis, in relatively healthy zones, show high levels of arrhythmogenic activity [109,110]. Another study also reported that the patchy and heterogeneous distribution of fibrous tissue can lead to conduction delays and the fragmentation of signals [111]. Jadidi et al. reported fractionated continuous activity in the vicinity of the border zone within the patchy fibrotic area, rather than within the dense fibrotic area [109]. However, determining the exact role of fibrotic tissue in arrhythmogenesis experimentally is difficult; therefore, experimental studies have been substituted with some numerical studies. A simulation study demonstrated that an important factor determining the formation and dynamics of arrhythmia in heterogeneous fibrosis stems from the maximum local fibrosis density occurring within the heterogeneous tissue [112]. A highly heterogeneous distribution can induce wavebreaks resulting in the formation of meandering rotors [113]. Other theoretical studies demonstrated that the degree and distribution of fibrosis had a large effect on rotor locations [114], and AF can be perpetuated by rotors meandering in the border zones of patchy fibrosis [115,116]. These numerical findings support the experimental results that the heterogeneous fibrotic distribution provides a certain mechanism of arrhythmogenesis and plays a key role in local fibrillatory activity.

3.4. Autonomic Ganglion Plexus

Electrophysiological properties in atria are modified by the autonomic nervous system, in which AF is often mediated by adrenergic or vagal activation [117]. Autonomic ganglia in the atrium are heterogeneously distributed, and ganglion plexuses (GPs) are concentrated at the SVC, CS, PV, and LA posterior wall in humans [118]. Preliminary studies have shown that GP stimulation induces the shortening of APD observed within PV sleeves in dogs [119,120]. Additionally, they demonstrated that injecting acetylcholine into the GP in adipose tissue next to a PV leads to rapid firing from the ipsilateral PV, transitioning to AF, followed by the cessation of AF by ablating the GP at the same site [119,121]. Excessive activity of the autonomic ganglia is now believed to be partially involved in the generation of AF, at least in cases showing focal firing. In clinical practice, attempts have been made to achieve catheter ablation for GP denervation [122]. Recent meta-analyses have shown a reduction in arrhythmia recurrence by adjunctive GP ablation plus PV isolation compared with PV isolation alone [123,124]. When it comes to autonomic nervous activity and AF, the oblique ligament of Marshall (LOM) also plays a key role. It is a remnant of the left SVC and is located between the LA appendage and the left superior/inferior PVs. It runs inferiorly along the inferior atrial wall, while only its intracardiac portion remains patent as the vein of Marshall which drains into the CS. The LOM contains fat and fibrotic tissues, vessels, muscle bundles, nerve fibers, and ganglia. Not only can the LOM’s complex structure serve as an arrhythmogenic substrate, but also its rich innervation by sympathetic nerves may serve as a source of catecholamine-sensitive focal automaticity. Ectopic activity and focal automaticity in the LOM induced by isoproterenol have been demonstrated in dogs, indicating that it contributes to the breakout of AF [125]. The effectiveness of LOM ablation has been reported in patients with persistent AF after PV isolation, implying that enhanced atrial denervation, elimination of AF triggers, or conduction block correlate with a reduction in the vulnerability to AF [126,127].

4. Microscopic Heterogeneity

Since cardiac action potentials are governed mainly by sarcolemmal ion currents, the alteration of these ion currents can help understand the arrhythmogenic mechanism. Once arrhythmogenic substrates result in AF, AF further adversely progresses the arrhythmogenic substrates, which is termed “AF begets AF”. In some reports, electrical changes in sarcolemmal ion currents resulting from AF were investigated using a whole-cell patch-clamp system with CMs isolated from the appendages of patients with persistent AF [128,129,130,131,132]. Despite the overall changes in ion currents in AF, to the best of our knowledge, the localized inter- or intra-cellular heterogeneous distribution of ion channels has not been discussed so far. However, a better understanding of the structural and electrophysiological heterogeneity underlying AF at a microscopic level will lead to advancements in treatment strategies. Regardless of the underlying diverse causes, abnormal discharge and electrical reentry are the two main determinants of ectopic firing, initiation, and maintenance of AF. Herein, we will review the microscopic heterogeneities at a cellular level and the molecular basis of the connexins, the profibrotic process, and genetics in association with their predisposition to AF.

4.1. Connexin Remodeling and Regulation

As regards the conduction heterogenicity exacerbated by AF, the redistribution of connexin has been mainly argued. Electrical intercellular conduction is highly dependent on connexins, which facilitate cell-to-cell connections (gap junctions) and are known to be involved in the electrochemical coupling to adjacent cells at the intercalated disk [133]. An action potential in a CM propagates longitudinally into the adjacent CM via gap junctions localized normally in the intercalated disk. The heterogeneous distribution in a tissue or lateralization of connexin distribution in a CM has often been discussed. Of note, heterogeneous distribution is termed a patchy distribution in a tissue, and lateralization is termed a lateralized intracellular distribution. Connexin remodeling in a CM, such as reduced expression and lateralization, can decrease longitudinal cell-to-cell electrical coupling resulting in a change in anisotropic conduction. van der Velden et al. investigated the role of gap junctions in a goat AF model [134,135]. They found no changes in the overall gene expression and protein levels of Cx40 and Cx43; however, according to immunohistochemical studies and confocal laser scanning microscopy, a heterogeneous distribution of Cx40 in tissue was observed in AF. Subsequently, Polontchouk et al. demonstrated that in atrial CMs obtained from patients with AF, while the protein level of Cx40 increased, its lateralization in the CMs also increased [136,137]. In patients with AF undergoing a biopsy procedure, the reduced expression and lateralized distribution of Cx40 were observed [138]. Overall, these results showed inconsistent expression levels of Cx40, but the intracellular lateralization of Cx40 was commonly observed. Previous findings on the expression levels and intercellular lateralization of Cx43 in AF were similar to those of Cx40 [133,139]. van der Velden et al. also demonstrated that the total levels and localization of Cx43 remained unchanged in a goat AF model; however, several studies with canine AF models reported increased Cx43 expression together with increased lateralization [140,141]. In humans, a study showed that the LA in patients with lone AF exhibited an increase in the protein expression of Cx43, while another study demonstrated the lateralization of Cx43 in patients with persistent AF [136]. In summary, the heterogeneous expression level and the heterogeneous distribution, including lateralization, of Cx40 and Cx43 may result in heterogeneous intercellular coupling, leading to conduction defects and anisotropy, which can facilitate wavebreaks and pathophysiological substrates of AF [142].
Abnormalities in connexin assembly, permeability, or localization impair electrical propagation and can lead to conduction disturbance which contributes to the development of AF [143]. Those abnormalities manifest in the form of phosphorylation, Cx40/Cx43 protein ratios, and lateralization to the surface membrane [143]. Connexin phosphorylation has been thought to switch on several types of functions. For instance, it regulates gap junctional protein trafficking and assembly. The increase in lateralized connexins can lead to a reduction in gap junctional conductance. Cx43 is phosphorylated specifically by various kinases including protein kinase A (PKA), protein kinase C (PKC), Ca2+/calmodulin-dependent kinase II (CaMKII), and mitogen-activated protein kinases (MAPKs) [144]. The sympathetic and β-adrenergic induction of activated cyclic adenosine monophosphate/PKA promote the synthesis and the phosphorylation of Cx43 (Ser364) [145]. PKA enhances the assembly of connexins into the gap junction and suppresses proteolytic degradation. As a result, PKA increases channel conductance. On the other hand, angiotensin II (Ang II) induces PKC activation, which augments the phosphorylation of Cx43 (Ser368) [146], inhibits the assembly of the connexins into the gap junction, and accelerates proteolytic degradation, followed by the impairment of the gap junctional communication. A remarkable reduction in connexins in the intercalated disk, together with an increase in the lateralized delocalization, attenuates cell-to-cell coupling. Ca2+ is also an important factor that decreases the gap junctional conductance. Ca2+ overload elevated the resistance of gap junctions by hindering the PKA-mediated phosphorylation of Cx43. In addition, Ca2+/calmodulin binding activates CaMKII. Not only does CaMKII phosphorylate the ryanodine receptor 2 and increase its Ca2+ sensitivity to exhibit Ca2+ leak and triggered activity via an increase in channel open probability, but it also exerts an indirect effect by regulating Cx43 expression and subcellular localization in the intercalated disk [147]. Furthermore, inflammatory cytokines activate MAPK to upregulate phosphorylated Cx43 and impair cell-to-cell communication because Cx43 becomes extensively dispersed at the intercalated disks [148,149]. Taken together, the phosphorylation of connexins causes the abnormal distribution and expression of the gap junction, which promotes conduction heterogeneity. Remodeling of the gap junction can be an arrhythmogenic substrate.

4.2. Molecular Mechanism of Atrial Fibrosis

Atrial fibrosis is a major factor contributing to atrial structural and electrophysiological heterogeneity. The proliferation of the extracellular matrix (ECM) is promoted by the neurohormonal dysregulation of the renin–angiotensin–aldosterone system (RAAS) and the activation of fibrotic pathways, mainly initiated by the transforming growth factor beta (TGFβ), connective tissue growth factor, and platelet-derived growth factor [150]. These profibrotic signaling molecules activate fibroblasts, resulting in proliferation and differentiation into secretory myofibroblasts, often accompanied by the upregulation of matrix metalloproteinases (MMPs) and the downregulation of the tissue inhibitors of metalloproteinases (TIMPs). AF can activate inflammatory cells, such as macrophages, leading to oxidative stress and the production of reactive oxygen species (ROS). Consequently, the RAAS is activated, and Ang II contributes to a profibrotic process by binding to Ang II type 1 receptor [151], followed by the further stimulation of phospholipase C, which acts through inositol 1,4,5-trisphosphate (IP3) and diacylglycerol (DAG). IP3 mediates an increase in Ca2+ levels in the cytoplasm. The intracellular Ca2+ overload promotes fibroblast proliferation and differentiation. DAG activates PKC, which activates the downstream signaling of MAPK. Ang II serves as a potent nicotinamide adenine dinucleotide phosphate oxidase activator, leading to ROS overproduction, which activates MAPK as well. The activation of the MAPK signaling pathway promotes the secretion of various transcription factors such as TGFβ and MMP. TGFβ binds to serine/threonine kinase receptors, accompanied by the suppressor of mother against decapentaplegic protein-mediated signal transduction, thereby further promoting the profibrotic process. These processes in cooperation accelerate the formation of vulnerable substrates for AF. In terms of the association between the profibrotic molecular process and heterogeneity, AF vulnerability has been demonstrated to be associated with a segmental heterogeneous pattern of atrial ECM remodeling (molecular MMP/TIMP profile) in a porcine AF model with myocardial infarction [152]. This result implied that a substantial regional heterogeneity exists at the molecular level. Furthermore, atrial inflammation and adipose tissue depots play an important role in AF substrate formation [153,154]. In particular, many studies have suggested an association between epicardial adipose tissue (EAT) and the fibrosis of the underlying atrial myocardium [155,156,157]. EAT contains various cell types, especially adipocytes and immune cells such as macrophages and lymphocytes. Both the immune cells and adipocytes within EAT can release various cytokines, contributing to pathogenic inflammation. In patients with coronary artery disease, EAT has increased levels of inflammatory cytokines [158]. Another study with resected LA appendage and associated EAT from cardiac surgery patients with AF demonstrated that the fibrosis of EAT was associated with LA fibrosis [157]. The fibrosis and inflammation of EAT can predispose to the fibrosis of the adjacent atrial myocardium through the proinflammatory and profibrotic bioactive secretome from EAT [154]. Adipocytes can also have a remote effect on the myocardium by the secretion of adipokines. Profibrotic secretome analysis identified an adipokine, activin A, which is a member of the TGFβ superfamily. Ventedlef et al. demonstrated that atrial fibrosis was promoted with a high concentration of activin A secreted from adipocytes [155]. Taken together, a great amount of EAT correlates with the development of AF through the profibrotic secretome. In terms of the distribution of EAT in the LA, a study evaluating the location of EAT using computed tomography in patients with AF showed that large clusters of EAT are observed adjacent to the anterior roof, LA appendage, and lateral mitral isthmus [159]. This result implies that the heterogeneous distribution of EAT can be a causative factor of the regionally dependent severity of fibrosis in atria.

4.3. Genetics of Atrial Fibrillation

Many studies have demonstrated that AF has substantial genetic factors predisposing to the development of AF [160]. The linkage analyses of AF have shown various causative genes for familial AF. Mutations in the potassium channels were discovered early as causes of familial AF: KCNQ1, which encodes a subunit of KvLQT1; KCNH2 which encodes HERG; and KCNJ2 which encodes Kir2.1 [161,162,163]. An increase in potassium currents due to a gain of function, which shortens the action potential and refractory period of atrial CMs, forms a reentry substrate that promotes the occurrence and maintenance of AF. However, AF is rarely a monogenic disorder; AF is often caused by environmental factors and multiple common genetic predispositions that affect AF susceptibility. Genome-wide association studies have clarified the genetic polymorphisms strongly associated with AF. Multiple variants were found in genes responsible for cardiac development (NKX2-5, TBX5, and PITX2) and electrophysiology, including the coding genes of potassium ion channels (KCNH2 and KCNJ2) and connexins (GJA1 and GJA5) [164]. Although the susceptibility to AF is different across ancestries, some of the variants found across different ancestries are concentrated in the 4q25 region of the long arm of chromosome 4, in which the transcription factor PITX2 is closest [165]. PITX2 is expressed exclusively in the LA and has a critical function of regulating the left–right differentiation of the embryonic heart [166,167]. The asymmetrical organ morphogenesis by PITX2 confines the sinoatrial node, pacemaker cells, to the RA. PITX2C is the major isoform expressed in the heart, particularly in the LA, and PITX2C expression was significantly downregulated in human patients with AF [168]. Thus, it is probable that a deficiency in PITX2 causes the incomplete suppression of pacemaker activity in the LA, resulting in enhanced pacemaker activity in the LA. Furthermore, a transgenic murine model with a Pitx2c deficiency, which exhibited a decrease in Pitx2c expression in only the LA compared to the wild-type, showed the shortening of the LA action potential, and more susceptibility to AF compared to the wild-type [169]. Similarly, the chamber-specific Pitx2 conditional mutant mice revealed that the LA displayed a more depolarized resting membrane potential and a smaller action potential amplitude [168]. PITX2 has also been reported to regulate the extension of myocardial sleeves into PVs from the LA during development [170]. The differentiation of dorsal mesenchymal cells into the pulmonary myocardium requires PITX2, and transgenic Pitx2-deficient mice were found to have normally developed PVs, but absent PV myocardial sleeves [170]. In summary, these findings indicate that genetic predisposition increases susceptibility to AF, either electrophysiologically or anatomically. The regionally heterogeneous expression patterns of these genes can facilitate the basis of electrophysiological heterogeneity.

5. Electrophysiological Heterogeneity

The progression of AF is believed to require an ectopic or reentrant basis. When a focal discharge exceeds the threshold to excite the surrounding muscle, the propagation from this ectopic focus possibly acts as a trigger. If the electrophysiological heterogeneity meets the sufficient need for the AF substrate, the wavefront of the propagation wave can trigger a cascade of wavebreaks, leading to continuous reentry and the initiation of AF. The maintenance of continuous activity depends on the existing electrophysiological properties with refractory and excitability determinants; shortened refractory period and reduced conduction velocity (CV). As described above, segmentally differentiated tissue properties, fiber arrangements, and redistributed intra- and inter-cellular connexins facilitate the structural heterogeneities, which in turn connect with electrophysiological heterogeneities in the refractoriness and conduction of action potential.

5.1. Repolarization Heterogeneity

The heterogeneity of repolarization in the atria has been reported as a source of AF [171], similar to that in the ventricles where repolarization heterogeneity has been widely reported to cause ventricular tachycardia and fibrillation [172,173]. The analyses of atrial action potentials derived from the patch-clamp technique, monophasic action potential (MAP) method [174], endo- or epicardial electrophysiological studies, or optical mapping [175] are helpful to assess atrial repolarization. According to studies that used these methods, the heterogeneity in the APD or effective refractory period (ERP) serves as a substrate for AF [176,177,178]. In a study comparing action potential morphology using isolated CMs from various atrial segments in adult or aged dogs, and with sinus rhythm or chronic AF, patch-clamp recordings showed a tendency for APD shortening and increased APD heterogeneity in aged dogs with chronic AF compared to that in adult dogs with sinus rhythm [179]. Another canine study investigating the specific cellular electrophysiological properties using a patch-clamp system showed that resting membrane potential was more depolarized, and the APD was shorter in PVs than in the LA [180]. MAP studies in patients with paroxysmal AF showed a significant increase in the segmental dispersion of APD and ERP compared to those in patients with sinus rhythm [171,181,182]. In a canine model, an electrophysiological study demonstrated that the regional heterogeneity of refractory periods increases in AF [176]. Optical mapping with isolated hearts of diabetic rats demonstrated that an increase in the spatial dispersion of APD corresponds to the vulnerability of atrial arrhythmia [183]. Another optical mapping study in canine models showed the progressive shortening of APD and the depolarization of the resting membrane from the LA to the distal PVs [120]. This study also substantiated that the heterogeneity of the APD in PVs provided a favorable substrate for reentry formation. Simulation studies have supported these findings that the heterogeneity in refractoriness can account for the initiation of AF [176,184]. They revealed the presence of segmental dispersion or heterogeneity of APD in chronic AF and even paroxysmal AF. Further studies numerically showed that wavebreaks occur at sites with the steepest gradients of APD dispersion [185,186,187]. Another computer-based study with a 3D human model, which incorporated the regional distribution of APD from previous experimental data, showed that marked regional differences in the APD co-exist with shortened APD, facilitating the initiation and maintenance of reentrant waves. In particular, reentrant excitation is stabilized in the presence of pronounced regional differences in APD at the PV-LA junction and the crista terminalis/pectinate muscle junction in the RA appendage [188].
Many reports have suggested that an increase in inward rectifier potassium current is crucial in AF by shortening repolarization and causing hyperpolarization [128,129,130,131,132]. Consistent with the dispersion of APD, an in vitro assay with isolated CMs from different segments in an atrium showed the heterogeneous expression of the inward rectifier potassium channel [189,190]. Since the functional effects of structural remodeling induced by AF on the regional heterogeneity of APD have not yet been elucidated, computational models have been proposed to characterize these relationships. Several theoretical studies have compared the remodeled ionic currents with AF-induced reduction in APD in a human atrium, demonstrating that the up-regulation of inward rectifier potassium current has a greater influence on APD than any changes in other ion currents [191]. Moreover, Berenfeld et al. introduced the ionically regional gradient model to investigate the relation between the distribution of various types of ion channels and rotor stability. Among them, the gradient of the inward rectifier potassium current had the most significant impact on rotor stability [192,193]. The variation in the conductance of the inward rectifier potassium current resulted in gradients of APD, leading to the trapping of rotors at sites with the steepest gradients of APD [114].
Of note, what complicates the understanding of the reentrant mechanism is that the determinants of electrophysiological properties may change, not only spatially but also temporally [194,195,196]. It is not easy to thoroughly visualize spatio-temporally heterogeneous electrophysiological factors. APD is not just a static electrophysiological parameter but a dynamically changing one that can be influenced by the preceding diastolic interval (DI). If the preceding DI is short, the following APD tends to shorten, whereas if it is long, the following APD tends to be prolonged. The relationship between APD and DI is defined as APD restitution [197]. A steep curve in the APD restitution slope can cause oscillations in APD alternans, a phenomenon in which APD (or repolarization time) alternates between longer and shorter durations over time. When APD alternans occurs in a spatially inconsistent pattern, it is termed discordant APD alternans. Discordant alternans is a phenomenon in which two spatially distinct regions exhibit APD alternance of opposite phases. At the boundaries between the regions in which APD oscillates discordantly, there turns out to be an increased APD gap. The APD gap can induce the functional conduction block at the border, and thus yield heterogeneous conduction patterns, resulting in a possible source of fibrillatory conduction. Previous studies have demonstrated that discordant alternance is associated with reentrant activation and susceptibility to AF [198,199,200]. This substantiates that APD possesses the property of spatio-temporal variability, which can be a cause of electrophysiological heterogeneity and serve as the substrate for arrhythmogenicity. Taken together, repolarization heterogeneity can be a strong determinant of arrhythmogenic substrate.

5.2. Conduction Heterogeneity

Atrial conduction disorder can be decomposed into two elements: altered conduction propagation (direction) and reduced magnitude (velocity). When longitudinal propagation along the muscular fiber is disturbed because of the reduced electrical coupling between myocytes or collagenous obstacles, conduction in the transverse direction through lateral cell connections gets involved in the propagation. Additionally, tissue discontinuities due to fibrosis or anisotropic fiber orientation can cause electrical source–sink mismatch along these pathways, resulting in the local slowing of conduction or conduction block, and the tortuous conduction pathway. Furthermore, the hetero-cellular electrical coupling of myofibroblasts and myocytes can increase heterogeneity in excitability, refractoriness, and electrical load, potentially inducing conduction slowing [201]. This can lead to a pivoting or zigzag trajectory of activation. Electroanatomic maps during sinus rhythm of patients with paroxysmal or persistent AF showed that the atrial conduction properties in persistent AF were characterized by a higher wavefront curvature and a larger number of pivoting points than those in paroxysmal AF [202]. Several animal studies with optical mapping recognized zigzag conduction in pathological states such as stretch-induced and age-related atria [203,204]. Slow conduction with pivoting or zigzag pathways has become a factor of localized reentrant circuits [204]. Slow conduction or pivoting points can be viewed as fragmented potential or complex fractionated atrial electrograms (CFAEs) in the local potentials obtained from the electrode catheter during catheter ablation procedures [205,206]. Ablating the localized regions that represent CFAE in addition to PV isolation has been advocated to increase the procedural success rate. A meta-analysis demonstrated that the adjunctive CFAE ablation could provide additional benefits, such as reducing the recurrence of AF for patients with persistent AF [207]. Of note, a CFAE during AF is not always an indicator of an abnormal substrate; however, CFAE during sinus rhythm is a preferential marker of slow conduction, wavefront collision, or pivoting sites. Furthermore, to understand atrial arrhythmogenicity, transmural conduction should also be considered. de Groot et al. demonstrated the asynchronous activation of the endo-epicardial wall during AF in humans using simultaneous endo-epicardial mapping, which was not apparently observed during sinus rhythm [208,209]. These results implied that the dissociation of endo-epicardial activation may be important in the maintenance of AF. Optical mapping studies are helpful to assess the localized CV and visualize the heterogeneity. In animal experiments, atrial heterogeneous CV has been reported using isolated hearts in models such as pressure-overload mice [210], diabetic rats [183], and rabbits with atrial stretch stress [211]. These studies substantiated that incremental atrial heterogeneity in CV is closely related to the increased occurrence of AF. In addition, just as APD depends on the preceding DI, CV depends on it as well [212]. This dynamic is referred to as CV restitution, similar to APD restitution. When static conduction abnormalities are combined with dynamic conduction abnormalities due to CV restitution, conduction becomes more heterogeneous. In other words, the source of AF is present in regions where the decrease in atrial CV and the increase in the heterogeneity of CV with rate dependence are prominent [210,213,214] (Figure 3). To date, while a positive relation between the severity of the conduction heterogeneity and the susceptibility of AF has been suggested, how the conduction heterogeneity is linked to the initiation and sustenance of AF and the propagation pattern during AF remains unclear.

5.3. Ectopic Excitement

Ectopic premature contractions are widely acknowledged to serve as critical triggers for AF. The underlying mechanism by which these focal discharges act as triggers is their ability to induce conduction disturbances, thereby manifesting the AF substrate. Mapping studies have demonstrated that an atrial extrasystole can cause a functional conduction block [215,216]. Not only can a premature firing with a short-coupling interval trigger the initiation of AF via heterogenous APD or CV restitution as described in the previous sections, but conduction-directional effects can also predispose to functional conduction block and conduction slowing. In terms of the directional dependence of conduction, Kumagai et al. showed that the conduction delay from the distal PV to the PV-LA junction was significantly longer than that from the PV-LA junction to the distal PV, and a short-coupled extra-stimulus from the PV formed a PV-LA reciprocating reentrant circuit involving exit and entrance breakthrough points at the PV-LA junction [217]. Spach et al. demonstrated with human and canine models that premature stimuli resulted in slower conduction in the transverse direction, leading to anisotropic conduction [218,219,220]. They showed that ectopic premature excitation provokes more conduction disorders and reentries. An intra-operative epicardial mapping during 503 premature atrial contractions revealed that the CV decreases within the PV area, and the local directional conduction heterogeneity increases [216]. Furthermore, they demonstrated that patients with AF have a slower CV and more conduction heterogeneity during sinus rhythm, which becomes more pronounced during premature atrial contractions than in patients without AF.

6. Clinical Applications for Reducing Heterogeneity

Three therapeutic approaches can be considered to alleviate the heterogeneity. The first approach is to electrically separate the area with severe heterogeneity from the healthy area (Figure 4A). Isolating not only the trigger source but also regions with significant heterogeneity that deeply contribute to the initiation and maintenance of AF can be considered beneficial. In this context, PV isolation, PV antrum isolation, SVC isolation, and LA posterior wall isolation have been performed, with demonstrated efficacy. To further improve this approach, developing novel procedural techniques and devices for isolating anatomically challenging regions and advancing imaging and mapping technologies for more accurate identification of areas that need isolation is essential. While isolating a larger area can potentially be more effective by surrounding a greater amount of AF substrate with a conduction block, the isolated atrial regions lose their physiological contractile function, which can lead to thrombus formation. Therefore, the isolation should be limited to a necessary and sufficient extent. The second way is to ablate the individual distinctive features contributing to triggers, drivers, or other factors that are considered strongly correlated to AF sources using catheter interventions (Figure 4B). To this end, catheter ablation targeting sites with distinctive local features, such as reentrant circuit with conduction delay [221,222], fragmented potential [206,207], ganglionated plexi [122], local driver [206], and rotor [223], have been developed. However, the indicators to be targeted to achieve optimal electrical modification remain unclear. Particularly in the cases of persistent AF, where target regions are widely distributed throughout the atria, the complete ablation of all the target areas is difficult. Furthermore, the ablation process itself can contribute to myocardial damage and the formation of new conduction disturbances, presenting numerous challenges that need to be addressed. In this approach, developing new methods to identify the required and sufficient targeted areas and innovative interventions to modify myocardial conduction properties without proposing another source of AF is highly desired. The third approach is to decrease the heterogeneity or restore it to the original state with pharmacotherapy (Figure 4C). The development of new pharmacotherapy to reduce heterogeneity, including the profibrotic process or connexin remodeling, which is acquired under pathological conditions, is anticipated. To date, several drugs for atrial arrhythmia and heart failure have been proven to reduce the atrial heterogeneity in animal experiments: sodium channel blockers [217], calcium channel blockers [224], ACEis [225], and neprilysin and angiotensin receptor blockers [210]. However, these drugs have not been proven to reduce the occurrence of AF in clinical settings. While a new treatment is being strongly anticipated, sodium–glucose cotransporter-2 (SGLT-2) inhibitors are currently used for heart failure, and their effectiveness on AF has been gradually uncovered [226]. In short, therapeutic interventions for advanced AF are not established yet since the treatment outcomes decline as heterogeneity is exacerbated. Thus, finding strategies through catheter-based procedures or pharmacotherapy to reduce the heterogeneity could potentially lead to improved treatment outcomes. In addition to elucidating the underlying cause, a new approach to heterogeneity is anticipated.

7. Conclusions

The experimental and clinical evidence that the structural and electrophysiological heterogeneity contributes to the substrate for AF is accumulating. Advances in the recognition of the underlying heterogeneity with mapping technologies have elucidated the substrates incorporated into the pathophysiology of AF. Further studies on the underlying heterogeneity would provide more profound insights into the mechanisms of the sustenance and dynamics of AF.

Author Contributions

Conceptualization, S.I., K.I. and G.N.; methodology, S.I.; software, S.I.; validation, S.I. and K.I.; formal analysis, S.I.; investigation, S.I.; resources, S.I.; data curation, S.I.; writing—original draft preparation, S.I., K.I. and G.N.; writing—review and editing, K.I. and T.S.; visualization, S.I.; supervision, T.S.; project administration, T.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Wyse, D.G.; Waldo, A.L.; DiMarco, J.P.; Domanski, M.J.; Rosenberg, Y.; Schron, E.B.; Kellen, J.C.; Greene, H.L.; Mickel, M.C.; Dalquist, J.E.; et al. A comparison of rate control and rhythm control in patients with atrial fibrillation. N. Engl. J. Med. 2002, 347, 1825–1833. [Google Scholar] [CrossRef] [PubMed]
  2. Disertori, M.; Latini, R.; Barlera, S.; Franzosi, M.G.; Staszewsky, L.; Maggioni, A.P.; Lucci, D.; Di Pasquale, G.; Tognoni, G. Valsartan for prevention of recurrent atrial fibrillation. N. Engl. J. Med. 2009, 360, 1606–1617. [Google Scholar] [CrossRef] [PubMed]
  3. Mohammad, Z.; Ahmad, J.; Sultan, A.; Penagaluri, A.; Morin, D.; Dominic, P. Effect of sacubitril-valsartan on the incidence of atrial fibrillation: A meta-analysis. J. Cardiovasc. Electrophysiol. 2023, 34, 1037–1042. [Google Scholar] [CrossRef] [PubMed]
  4. Rabinovitch, A.; Rabinovitch, R.; Biton, Y.; Braunstein, D.; Thieberger, R. A possible new cardiac heterogeneity as an arrhythmogenic driver. Sci. Rep. 2023, 13, 7571. [Google Scholar] [CrossRef]
  5. Allessie, M.A.; Boyden, P.A.; Camm, A.J.; Kléber, A.G.; Lab, M.J.; Legato, M.J.; Rosen, M.R.; Schwartz, P.J.; Spooner, P.M.; Van Wagoner, D.R.; et al. Pathophysiology and prevention of atrial fibrillation. Circulation 2001, 103, 769–777. [Google Scholar] [CrossRef]
  6. Nattel, S.; Burstein, B.; Dobrev, D. Atrial remodeling and atrial fibrillation: Mechanisms and implications. Circ. Arrhythm. Electrophysiol. 2008, 1, 62–73. [Google Scholar] [CrossRef]
  7. Dobrev, D.; Carlsson, L.; Nattel, S. Novel molecular targets for atrial fibrillation therapy. Nat. Rev. Drug Discov. 2012, 11, 275–291. [Google Scholar] [CrossRef]
  8. Avula, U.M.R.; Abrams, J.; Katchman, A.; Zakharov, S.; Mironov, S.; Bayne, J.; Roybal, D.; Gorti, A.; Yang, L.; Iyer, V.; et al. Heterogeneity of the action potential duration is required for sustained atrial fibrillation. JCI Insight 2019, 4, e128765. [Google Scholar] [CrossRef]
  9. Berenfeld, O.; Zaitsev, A.V.; Mironov, S.F.; Pertsov, A.M.; 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, 1173–1180. [Google Scholar] [CrossRef]
  10. Chen, Y.J.; Chen, S.A.; Chang, M.S.; Lin, C.I. Arrhythmogenic activity of cardiac muscle in pulmonary veins of the dog: Implication for the genesis of atrial fibrillation. Cardiovasc. Res. 2000, 48, 265–273. [Google Scholar] [CrossRef]
  11. Chen, Y.J.; Chen, S.A.; Chen, Y.C.; Yeh, H.I.; Chan, P.; Chang, M.S.; Lin, C.I. Effects of rapid atrial pacing on the arrhythmogenic activity of single cardiomyocytes from pulmonary veins: Implication in initiation of atrial fibrillation. Circulation 2001, 104, 2849–2854. [Google Scholar] [CrossRef] [PubMed]
  12. Perez-Lugones, A.; McMahon, J.T.; Ratliff, N.B.; Saliba, W.I.; Schweikert, R.A.; Marrouche, N.F.; Saad, E.B.; Navia, J.L.; McCarthy, P.M.; Tchou, P.; et al. Evidence of specialized conduction cells in human pulmonary veins of patients with atrial fibrillation. J. Cardiovasc. Electrophysiol. 2003, 14, 803–809. [Google Scholar] [CrossRef] [PubMed]
  13. Wit, A.L.; Boyden, P.A. Triggered activity and atrial fibrillation. Heart Rhythm 2007, 4, S17–S23. [Google Scholar] [CrossRef] [PubMed]
  14. Chen, Y.J.; Chen, S.A.; Chen, Y.C.; Yeh, H.I.; Chang, M.S.; Lin, C.I. Electrophysiology of single cardiomyocytes isolated from rabbit pulmonary veins: Implication in initiation of focal atrial fibrillation. Basic Res. Cardiol. 2002, 97, 26–34. [Google Scholar] [CrossRef] [PubMed]
  15. Vaitkevicius, R.; Saburkina, I.; Rysevaite, K.; Vaitkeviciene, I.; Pauziene, N.; Zaliunas, R.; Schauerte, P.; Jalife, J.; Pauza, D.H. Nerve supply of the human pulmonary veins: An anatomical study. Heart Rhythm 2009, 6, 221–228. [Google Scholar] [CrossRef]
  16. Nguyen, B.L.; Fishbein, M.C.; Chen, L.S.; Chen, P.S.; Masroor, S. Histopathological substrate for chronic atrial fibrillation in humans. Heart Rhythm 2009, 6, 454–460. [Google Scholar] [CrossRef]
  17. Iwasaki, Y.K.; Nishida, K.; Kato, T.; Nattel, S. Atrial fibrillation pathophysiology: Implications for management. Circulation 2011, 124, 2264–2274. [Google Scholar] [CrossRef]
  18. Gerstenfeld, E.P.; Callans, D.J.; Sauer, W.; Jacobson, J.; Marchlinski, F.E. Reentrant and nonreentrant focal left atrial tachycardias occur after pulmonary vein isolation. Heart Rhythm 2005, 2, 1195–1202. [Google Scholar] [CrossRef]
  19. Ho, S.Y.; Cabrera, J.A.; Tran, V.H.; Farré, J.; Anderson, R.H.; Sánchez-Quintana, D. Architecture of the pulmonary veins: Relevance to radiofrequency ablation. Heart 2001, 86, 265–270. [Google Scholar] [CrossRef]
  20. Hamabe, A.; Okuyama, Y.; Miyauchi, Y.; Zhou, S.; Pak, H.N.; Karagueuzian, H.S.; Fishbein, M.C.; Chen, P.S. Correlation between anatomy and electrical activation in canine pulmonary veins. Circulation 2003, 107, 1550–1555. [Google Scholar] [CrossRef]
  21. Hassink, R.J.; Aretz, H.T.; Ruskin, J.; Keane, D. Morphology of atrial myocardium in human pulmonary veins: A postmortem analysis in patients with and without atrial fibrillation. J. Am. Coll. Cardiol. 2003, 42, 1108–1114. [Google Scholar] [CrossRef] [PubMed]
  22. Valles, E.; Fan, R.; Roux, J.F.; Liu, C.F.; Harding, J.D.; Dhruvakumar, S.; Hutchinson, M.D.; Riley, M.; Bala, R.; Garcia, F.C.; et al. Localization of atrial fibrillation triggers in patients undergoing pulmonary vein isolation: Importance of the carina region. J. Am. Coll. Cardiol. 2008, 52, 1413–1420. [Google Scholar] [CrossRef] [PubMed]
  23. Cabrera, J.A.; Ho, S.Y.; Climent, V.; Fuertes, B.; Murillo, M.; Sánchez-Quintana, D. Morphological evidence of muscular connections between contiguous pulmonary venous orifices: Relevance of the interpulmonary isthmus for catheter ablation in atrial fibrillation. Heart Rhythm 2009, 6, 1192–1198. [Google Scholar] [CrossRef] [PubMed]
  24. Arentz, T.; Weber, R.; Bürkle, G.; Herrera, C.; Blum, T.; Stockinger, J.; Minners, J.; Neumann, F.J.; Kalusche, D. Small or large isolation areas around the pulmonary veins for the treatment of atrial fibrillation? Results from a prospective randomized study. Circulation 2007, 115, 3057–3063. [Google Scholar] [CrossRef]
  25. Yeh, H.I.; Lai, Y.J.; Lee, S.H.; Lee, Y.N.; Ko, Y.S.; Chen, S.A.; Severs, N.J.; Tsai, C.H. Heterogeneity of myocardial sleeve morphology and gap junctions in canine superior vena cava. Circulation 2001, 104, 3152–3157. [Google Scholar] [CrossRef]
  26. Watanabe, K.; Nitta, J.; Inaba, O.; Sato, A.; Inamura, Y.; Kato, N.; Suzuki, M.; Goya, M.; Hirao, K.; Sasano, T. Predictors of non-pulmonary vein foci in paroxysmal atrial fibrillation. J. Interv. Card. Electrophysiol. 2021, 61, 71–78. [Google Scholar] [CrossRef]
  27. Lin, W.S.; Tai, C.T.; Hsieh, M.H.; Tsai, C.F.; Lin, Y.K.; Tsao, H.M.; Huang, J.L.; Yu, W.C.; Yang, S.P.; Ding, Y.A.; et al. Catheter ablation of paroxysmal atrial fibrillation initiated by non-pulmonary vein ectopy. Circulation 2003, 107, 3176–3183. [Google Scholar] [CrossRef]
  28. Yamaguchi, T.; Tsuchiya, T.; Miyamoto, K.; Nagamoto, Y.; Takahashi, N. Characterization of non-pulmonary vein foci with an EnSite array in patients with paroxysmal atrial fibrillation. Europace 2010, 12, 1698–1706. [Google Scholar] [CrossRef]
  29. Chen, Y.J.; Chen, Y.C.; Yeh, H.I.; Lin, C.I.; Chen, S.A. Electrophysiology and arrhythmogenic activity of single cardiomyocytes from canine superior vena cava. Circulation 2002, 105, 2679–2685. [Google Scholar] [CrossRef]
  30. Kugler, S.; Nagy, N.; Rácz, G.; Tőkés, A.M.; Dorogi, B.; Nemeskéri, Á. Presence of cardiomyocytes exhibiting Purkinje-type morphology and prominent connexin45 immunoreactivity in the myocardial sleeves of cardiac veins. Heart Rhythm 2018, 15, 258–264. [Google Scholar] [CrossRef]
  31. Higuchi, K.; Yamauchi, Y.; Hirao, K.; Sasaki, T.; Hachiya, H.; Sekiguchi, Y.; Nitta, J.; Isobe, M. Superior vena cava as initiator of atrial fibrillation: Factors related to its arrhythmogenicity. Heart Rhythm 2010, 7, 1186–1191. [Google Scholar] [CrossRef] [PubMed]
  32. Shah, D.C.; Haïssaguerre, M.; Jaïs, P.; Clémenty, J. High-resolution mapping of tachycardia originating from the superior vena cava: Evidence of electrical heterogeneity, slow conduction, and possible circus movement reentry. J. Cardiovasc. Electrophysiol. 2002, 13, 388–392. [Google Scholar] [CrossRef] [PubMed]
  33. Miyazaki, S.; Kuwahara, T.; Takahashi, A. Confined driver of atrial fibrillation in the superior vena cava. J. Cardiovasc. Electrophysiol. 2012, 23, 440. [Google Scholar] [CrossRef] [PubMed]
  34. Miyazaki, S.; Takigawa, M.; Kusa, S.; Kuwahara, T.; Taniguchi, H.; Okubo, K.; Nakamura, H.; Hachiya, H.; Hirao, K.; Takahashi, A.; et al. Role of arrhythmogenic superior vena cava on atrial fibrillation. J. Cardiovasc. Electrophysiol. 2014, 25, 380–386. [Google Scholar] [CrossRef] [PubMed]
  35. Simu, G.; Deneke, T.; Ene, E.; Nentwich, K.; Berkovitz, A.; Sonne, K.; Halbfass, P.; Arvaniti, E.; Waechter, C.; Müller, J. Empirical superior vena cava isolation in patients undergoing repeat catheter ablation procedure after recurrence of atrial fibrillation. J. Interv. Card. Electrophysiol. 2022, 65, 551–558. [Google Scholar] [CrossRef]
  36. Ho, S.Y.; Sánchez-Quintana, D. The importance of atrial structure and fibers. Clin. Anat. 2009, 22, 52–63. [Google Scholar] [CrossRef]
  37. Hashizume, H.; Ushiki, T.; Abe, K. A histological study of the cardiac muscle of the human superior and inferior venae cavae. Arch. Histol. Cytol. 1995, 58, 457–464. [Google Scholar] [CrossRef]
  38. Alonso-Martín, C.; García Mancebo, S.; Campos García, B.; Guerra Ramos, J.; Moreno Weidman, Z.; Méndez Zurita, F.; Montiel Quintero, R.; Betancur Gutiérrez, A.; Viñolas, X.; Rodríguez Font, E. Atrial Fibrillation Originating in the Inferior Vena Cava: A Typical Presentation of an Atypical Location. JACC Case Rep. 2021, 3, 1918–1923. [Google Scholar] [CrossRef]
  39. Tao, Y.; Yang, D.; Chen, L. Paroxysmal Atrial Fibrillation Originating from the Inferior Vena Cava: A Case Report and Literature Review. Front. Cardiovasc. Med. 2022, 9, 935524. [Google Scholar] [CrossRef]
  40. Habib, A.; Lachman, N.; Christensen, K.N.; Asirvatham, S.J. The anatomy of the coronary sinus venous system for the cardiac electrophysiologist. Europace 2009, 11 (Suppl. S5), v15–v21. [Google Scholar] [CrossRef]
  41. Antz, M.; Otomo, K.; Arruda, M.; Scherlag, B.J.; Pitha, J.; Tondo, C.; Lazzara, R.; Jackman, W.M. Electrical conduction between the right atrium and the left atrium via the musculature of the coronary sinus. Circulation 1998, 98, 1790–1795. [Google Scholar] [CrossRef] [PubMed]
  42. Chauvin, M.; Shah, D.C.; Haïssaguerre, M.; Marcellin, L.; Brechenmacher, C. The anatomic basis of connections between the coronary sinus musculature and the left atrium in humans. Circulation 2000, 101, 647–652. [Google Scholar] [CrossRef] [PubMed]
  43. Volkmer, M.; Antz, M.; Hebe, J.; Kuck, K.H. Focal atrial tachycardia originating from the musculature of the coronary sinus. J. Cardiovasc. Electrophysiol. 2002, 13, 68–71. [Google Scholar] [CrossRef] [PubMed]
  44. Johnson, N.; Danilo, P., Jr.; Wit, A.L.; Rosen, M.R. Characteristics of initiation and termination of catecholamine-induced triggered activity in atrial fibers of the coronary sinus. Circulation 1986, 74, 1168–1179. [Google Scholar] [CrossRef]
  45. Tseng, G.N.; Wit, A.L. Characteristics of a transient inward current that causes delayed afterdepolarizations in atrial cells of the canine coronary sinus. J. Mol. Cell Cardiol. 1987, 19, 1105–1119. [Google Scholar] [CrossRef]
  46. Oral, H.; Ozaydin, M.; Chugh, A.; Scharf, C.; Tada, H.; Hall, B.; Cheung, P.; Pelosi, F.; Knight, B.P.; Morady, F. Role of the coronary sinus in maintenance of atrial fibrillation. J. Cardiovasc. Electrophysiol. 2003, 14, 1329–1336. [Google Scholar] [CrossRef]
  47. Katritsis, D.; Ioannidis, J.P.; Giazitzoglou, E.; Korovesis, S.; Anagnostopoulos, C.E.; Camm, A.J. Conduction delay within the coronary sinus in humans: Implications for atrial arrhythmias. J. Cardiovasc. Electrophysiol. 2002, 13, 859–862. [Google Scholar] [CrossRef]
  48. Ho, S.Y.; Sanchez-Quintana, D.; Cabrera, J.A.; Anderson, R.H. Anatomy of the left atrium: Implications for radiofrequency ablation of atrial fibrillation. J. Cardiovasc. Electrophysiol. 1999, 10, 1525–1533. [Google Scholar] [CrossRef]
  49. Ahmed, N.; Perveen, S.; Mehmood, A.; Rani, G.F.; Molon, G. Coronary Sinus Ablation Is a Key Player Substrate in Recurrence of Persistent Atrial Fibrillation. Cardiology 2019, 143, 107–113. [Google Scholar] [CrossRef]
  50. Morita, H.; Zipes, D.P.; Morita, S.T.; Wu, J. The role of coronary sinus musculature in the induction of atrial fibrillation. Heart Rhythm 2012, 9, 581–589. [Google Scholar] [CrossRef]
  51. Morita, H.; Zipes, D.P.; Morita, S.T.; Wu, J. Isolation of canine coronary sinus musculature from the atria by radiofrequency catheter ablation prevents induction of atrial fibrillation. Circ. Arrhythm. Electrophysiol. 2014, 7, 1181–1188. [Google Scholar] [CrossRef] [PubMed]
  52. Zhao, J.; Trew, M.L.; Legrice, I.J.; Smaill, B.H.; Pullan, A.J. A tissue-specific model of reentry in the right atrial appendage. J. Cardiovasc. Electrophysiol. 2009, 20, 675–684. [Google Scholar] [CrossRef] [PubMed]
  53. Zhao, Q.Y.; Huang, H.; Tang, Y.H.; Wang, X.; Okello, E.; Liang, J.J.; Jiang, H.; Huang, C.X. Relationship between autonomic innervation in crista terminalis and atrial arrhythmia. J. Cardiovasc. Electrophysiol. 2009, 20, 551–557. [Google Scholar] [CrossRef]
  54. Yamashita, T.; Nakajima, T.; Hazama, H.; Hamada, E.; Murakawa, Y.; Sawada, K.; Omata, M. Regional differences in transient outward current density and inhomogeneities of repolarization in rabbit right atrium. Circulation 1995, 92, 3061–3069. [Google Scholar] [CrossRef] [PubMed]
  55. Giles, W.R.; van Ginneken, A.C. A transient outward current in isolated cells from the crista terminalis of rabbit heart. J. Physiol. 1985, 368, 243–264. [Google Scholar] [CrossRef]
  56. Wu, T.J.; Yashima, M.; Xie, F.; Athill, C.A.; Kim, Y.H.; Fishbein, M.C.; Qu, Z.; Garfinkel, A.; Weiss, J.N.; Karagueuzian, H.S.; et al. Role of pectinate muscle bundles in the generation and maintenance of intra-atrial reentry: Potential implications for the mechanism of conversion between atrial fibrillation and atrial flutter. Circ. Res. 1998, 83, 448–462. [Google Scholar] [CrossRef]
  57. Baptiste, F.; Kalifa, J.; Durand, C.; Gitenay, E.; Bremondy, M.; Ayari, A.; Maillot, N.; Taormina, A.; Fofana, A.; Penaranda, G.; et al. Right atrial appendage firing in atrial fibrillation. Front. Cardiovasc. Med. 2022, 9, 997998. [Google Scholar] [CrossRef]
  58. Marcus, M.B.; Shein, J.A.; Vaishnav, A.S.; Mountantonakis, S.E. Paroxysmal Atrial Fibrillation with Both Triggers and Rotational Drivers within the Right Atrial Appendage. JACC Case Rep. 2019, 1, 607–611. [Google Scholar] [CrossRef]
  59. Takahashi, Y.; Sanders, P.; Rotter, M.; Haïssaguerre, M. Disconnection of the left atrial appendage for elimination of foci maintaining atrial fibrillation. J. Cardiovasc. Electrophysiol. 2005, 16, 917–919. [Google Scholar] [CrossRef]
  60. Di Biase, L.; Burkhardt, J.D.; Mohanty, P.; Sanchez, J.; Mohanty, S.; Horton, R.; Gallinghouse, G.J.; Bailey, S.M.; Zagrodzky, J.D.; Santangeli, P.; et al. Left atrial appendage: An underrecognized trigger site of atrial fibrillation. Circulation 2010, 122, 109–118. [Google Scholar] [CrossRef]
  61. AlTurki, A.; Huynh, T.; Dawas, A.; AlTurki, H.; Joza, J.; Healey, J.S.; Essebag, V. Left atrial appendage isolation in atrial fibrillation catheter ablation: A meta-analysis. J. Arrhythm. 2018, 34, 478–484. [Google Scholar] [CrossRef] [PubMed]
  62. Di Biase, L.; Burkhardt, J.D.; Mohanty, P.; Mohanty, S.; Sanchez, J.E.; Trivedi, C.; Güneş, M.; Gökoğlan, Y.; Gianni, C.; Horton, R.P.; et al. Left Atrial Appendage Isolation in Patients with Longstanding Persistent AF Undergoing Catheter Ablation: BELIEF Trial. J. Am. Coll. Cardiol. 2016, 68, 1929–1940. [Google Scholar] [CrossRef] [PubMed]
  63. Ghannam, M.; Jongnarangsin, K.; Emami, H.; Yokokawa, M.; Liang, J.J.; Saeed, M.; Oral, H.; Morady, F.; Chugh, A. Incidental left atrial appendage isolation after catheter ablation of persistent atrial fibrillation: Mechanisms and long-term risk of thromboembolism. J. Cardiovasc. Electrophysiol. 2023, 34, 1152–1161. [Google Scholar] [CrossRef] [PubMed]
  64. Morillo, C.A.; Klein, G.J.; Jones, D.L.; Guiraudon, C.M. Chronic rapid atrial pacing. Structural, functional, and electrophysiological characteristics of a new model of sustained atrial fibrillation. Circulation 1995, 91, 1588–1595. [Google Scholar] [CrossRef]
  65. Huang, J.L.; Tai, C.T.; Lin, Y.J.; Ting, C.T.; Chen, Y.T.; Chang, M.S.; Lin, F.Y.; Lai, W.T.; Chen, S.A. The mechanisms of an increased dominant frequency in the left atrial posterior wall during atrial fibrillation in acute atrial dilatation. J. Cardiovasc. Electrophysiol. 2006, 17, 178–188. [Google Scholar] [CrossRef]
  66. Kalifa, J.; Tanaka, K.; Zaitsev, A.V.; Warren, M.; Vaidyanathan, R.; Auerbach, D.; Pandit, S.; Vikstrom, K.L.; Ploutz-Snyder, R.; Talkachou, A.; et al. Mechanisms of wave fractionation at boundaries of high-frequency excitation in the posterior left atrium of the isolated sheep heart during atrial fibrillation. Circulation 2006, 113, 626–633. [Google Scholar] [CrossRef]
  67. Wu, T.J.; Doshi, R.N.; Huang, H.L.; Blanche, C.; Kass, R.M.; Trento, A.; Cheng, W.; Karagueuzian, H.S.; Peter, C.T.; Chen, P.S. Simultaneous biatrial computerized mapping during permanent atrial fibrillation in patients with organic heart disease. J. Cardiovasc. Electrophysiol. 2002, 13, 571–577. [Google Scholar] [CrossRef]
  68. Bliss, D.F., 2nd; Hutchins, G.M. The dorsal mesocardium and development of the pulmonary veins in human embryos. Am. J. Cardiovasc. Pathol. 1995, 5, 55–67. [Google Scholar]
  69. Douglas, Y.L.; Jongbloed, M.R.; Gittenberger-de Groot, A.C.; Evers, D.; Dion, R.A.; Voigt, P.; Bartelings, M.M.; Schalij, M.J.; Ebels, T.; DeRuiter, M.C. Histology of vascular myocardial wall of left atrial body after pulmonary venous incorporation. Am. J. Cardiol. 2006, 97, 662–670. [Google Scholar] [CrossRef]
  70. Sherif, H.M. The developing pulmonary veins and left atrium: Implications for ablation strategy for atrial fibrillation. Eur. J. Cardiothorac. Surg. 2013, 44, 792–799. [Google Scholar] [CrossRef]
  71. Tessari, A.; Pietrobon, M.; Notte, A.; Cifelli, G.; Gage, P.J.; Schneider, M.D.; Lembo, G.; Campione, M. Myocardial Pitx2 differentially regulates the left atrial identity and ventricular asymmetric remodeling programs. Circ. Res. 2008, 102, 813–822. [Google Scholar] [CrossRef] [PubMed]
  72. Galli, D.; Domínguez, J.N.; Zaffran, S.; Munk, A.; Brown, N.A.; Buckingham, M.E. Atrial myocardium derives from the posterior region of the second heart field, which acquires left-right identity as Pitx2c is expressed. Development 2008, 135, 1157–1167. [Google Scholar] [CrossRef] [PubMed]
  73. Klos, M.; Calvo, D.; Yamazaki, M.; Zlochiver, S.; Mironov, S.; Cabrera, J.A.; Sanchez-Quintana, D.; Jalife, J.; Berenfeld, O.; Kalifa, J. Atrial septopulmonary bundle of the posterior left atrium provides a substrate for atrial fibrillation initiation in a model of vagally mediated pulmonary vein tachycardia of the structurally normal heart. Circ. Arrhythm. Electrophysiol. 2008, 1, 175–183. [Google Scholar] [CrossRef] [PubMed]
  74. Markides, V.; Schilling, R.J.; Ho, S.Y.; Chow, A.W.; Davies, D.W.; Peters, N.S. Characterization of left atrial activation in the intact human heart. Circulation 2003, 107, 733–739. [Google Scholar] [CrossRef] [PubMed]
  75. Burstein, B.; Nattel, S. Atrial fibrosis: Mechanisms and clinical relevance in atrial fibrillation. J. Am. Coll. Cardiol. 2008, 51, 802–809. [Google Scholar] [CrossRef]
  76. Xu, J.; Cui, G.; Esmailian, F.; Plunkett, M.; Marelli, D.; Ardehali, A.; Odim, J.; Laks, H.; Sen, L. Atrial extracellular matrix remodeling and the maintenance of atrial fibrillation. Circulation 2004, 109, 363–368. [Google Scholar] [CrossRef]
  77. Miragoli, M.; Salvarani, N.; Rohr, S. Myofibroblasts induce ectopic activity in cardiac tissue. Circ. Res. 2007, 101, 755–758. [Google Scholar] [CrossRef]
  78. Rohr, S. Myofibroblasts in diseased hearts: New players in cardiac arrhythmias? Heart Rhythm 2009, 6, 848–856. [Google Scholar] [CrossRef]
  79. Yue, L.; Xie, J.; Nattel, S. Molecular determinants of cardiac fibroblast electrical function and therapeutic implications for atrial fibrillation. Cardiovasc. Res. 2011, 89, 744–753. [Google Scholar] [CrossRef]
  80. Sánchez, J.; Gomez, J.F.; Martinez-Mateu, L.; Romero, L.; Saiz, J.; Trenor, B. Heterogeneous Effects of Fibroblast-Myocyte Coupling in Different Regions of the Human Atria under Conditions of Atrial Fibrillation. Front. Physiol. 2019, 10, 847. [Google Scholar] [CrossRef]
  81. Oakes, R.S.; Badger, T.J.; Kholmovski, E.G.; Akoum, N.; Burgon, N.S.; Fish, E.N.; Blauer, J.J.; Rao, S.N.; DiBella, E.V.; Segerson, N.M.; et al. Detection and quantification of left atrial structural remodeling with delayed-enhancement magnetic resonance imaging in patients with atrial fibrillation. Circulation 2009, 119, 1758–1767. [Google Scholar] [CrossRef] [PubMed]
  82. Marrouche, N.F.; Wilber, D.; Hindricks, G.; Jais, P.; Akoum, N.; Marchlinski, F.; Kholmovski, E.; Burgon, N.; Hu, N.; Mont, L.; et al. Association of atrial tissue fibrosis identified by delayed enhancement MRI and atrial fibrillation catheter ablation: The DECAAF study. JAMA 2014, 311, 498–506. [Google Scholar] [CrossRef] [PubMed]
  83. Benito, E.M.; Cabanelas, N.; Nuñez-Garcia, M.; Alarcón, F.; Figueras, I.V.R.M.; Soto-Iglesias, D.; Guasch, E.; Prat-Gonzalez, S.; Perea, R.J.; Borràs, R.; et al. Preferential regional distribution of atrial fibrosis in posterior wall around left inferior pulmonary vein as identified by late gadolinium enhancement cardiac magnetic resonance in patients with atrial fibrillation. Europace 2018, 20, 1959–1965. [Google Scholar] [CrossRef] [PubMed]
  84. Higuchi, K.; Cates, J.; Gardner, G.; Morris, A.; Burgon, N.S.; Akoum, N.; Marrouche, N.F. The Spatial Distribution of Late Gadolinium Enhancement of Left Atrial Magnetic Resonance Imaging in Patients with Atrial Fibrillation. JACC Clin. Electrophysiol. 2018, 4, 49–58. [Google Scholar] [CrossRef]
  85. Chen, J.; Arentz, T.; Cochet, H.; Müller-Edenborn, B.; Kim, S.; Moreno-Weidmann, Z.; Minners, J.; Kohl, P.; Lehrmann, H.; Allgeier, J.; et al. Extent and spatial distribution of left atrial arrhythmogenic sites, late gadolinium enhancement at magnetic resonance imaging, and low-voltage areas in patients with persistent atrial fibrillation: Comparison of imaging vs. electrical parameters of fibrosis and arrhythmogenesis. Europace 2019, 21, 1484–1493. [Google Scholar] [CrossRef]
  86. Peters, D.C.; Wylie, J.V.; Hauser, T.H.; Nezafat, R.; Han, Y.; Woo, J.J.; Taclas, J.; Kissinger, K.V.; Goddu, B.; Josephson, M.E.; et al. Recurrence of atrial fibrillation correlates with the extent of post-procedural late gadolinium enhancement: A pilot study. JACC Cardiovasc. Imaging 2009, 2, 308–316. [Google Scholar] [CrossRef]
  87. McGann, C.; Akoum, N.; Patel, A.; Kholmovski, E.; Revelo, P.; Damal, K.; Wilson, B.; Cates, J.; Harrison, A.; Ranjan, R.; et al. Atrial fibrillation ablation outcome is predicted by left atrial remodeling on MRI. Circ. Arrhythm. Electrophysiol. 2014, 7, 23–30. [Google Scholar] [CrossRef]
  88. Marrouche, N.F.; Wazni, O.; McGann, C.; Greene, T.; Dean, J.M.; Dagher, L.; Kholmovski, E.; Mansour, M.; Marchlinski, F.; Wilber, D.; et al. Effect of MRI-Guided Fibrosis Ablation vs Conventional Catheter Ablation on Atrial Arrhythmia Recurrence in Patients with Persistent Atrial Fibrillation: The DECAAF II Randomized Clinical Trial. JAMA 2022, 327, 2296–2305. [Google Scholar] [CrossRef]
  89. Sanders, P.; Morton, J.B.; Davidson, N.C.; Spence, S.J.; Vohra, J.K.; Sparks, P.B.; Kalman, J.M. Electrical remodeling of the atria in congestive heart failure: Electrophysiological and electroanatomic mapping in humans. Circulation 2003, 108, 1461–1468. [Google Scholar] [CrossRef]
  90. Huo, Y.; Gaspar, T.; Pohl, M.; Sitzy, J.; Richter, U.; Neudeck, S.; Mayer, J.; Kronborg, M.B.; Piorkowski, C. Prevalence and predictors of low voltage zones in the left atrium in patients with atrial fibrillation. Europace 2018, 20, 956–962. [Google Scholar] [CrossRef]
  91. Liu, Z.; Xia, Y.; Guo, C.; Li, X.; Fang, P.; Yin, X.; Yang, X. Low-Voltage Zones as the Atrial Fibrillation Substrates: Relationship with Initiation, Perpetuation, and Termination. Front. Cardiovasc. Med. 2021, 8, 705510. [Google Scholar] [CrossRef] [PubMed]
  92. Verma, A.; Wazni, O.M.; Marrouche, N.F.; Martin, D.O.; Kilicaslan, F.; Minor, S.; Schweikert, R.A.; Saliba, W.; Cummings, J.; Burkhardt, J.D.; et al. Pre-existent left atrial scarring in patients undergoing pulmonary vein antrum isolation: An independent predictor of procedural failure. J. Am. Coll. Cardiol. 2005, 45, 285–292. [Google Scholar] [CrossRef]
  93. Vlachos, K.; Efremidis, M.; Letsas, K.P.; Bazoukis, G.; Martin, R.; Kalafateli, M.; Lioni, L.; Georgopoulos, S.; Saplaouras, A.; Efremidis, T.; et al. Low-voltage areas detected by high-density electroanatomical mapping predict recurrence after ablation for paroxysmal atrial fibrillation. J. Cardiovasc. Electrophysiol. 2017, 28, 1393–1402. [Google Scholar] [CrossRef] [PubMed]
  94. Moustafa, A.; Karim, S.; Kahaly, O.; Elzanaty, A.; Meenakshisundaram, C.; Abi-Saleh, B.; Eltahawy, E.; Chacko, P. Low voltage area guided substrate modification in nonparoxysmal atrial fibrillation: A systematic review and meta-analysis. J. Cardiovasc. Electrophysiol. 2023, 34, 455–464. [Google Scholar] [CrossRef] [PubMed]
  95. Zhou, Y.; Zhang, H.; Yan, P.; Zhou, P.; Wang, P.; Li, X. Efficacy of left atrial low-voltage area-guided catheter ablation of atrial fibrillation: An updated systematic review and meta-analysis. Front. Cardiovasc. Med. 2022, 9, 993790. [Google Scholar] [CrossRef]
  96. Junarta, J.; Siddiqui, M.U.; Riley, J.M.; Dikdan, S.J.; Patel, A.; Frisch, D.R. Low-voltage area substrate modification for atrial fibrillation ablation: A systematic review and meta-analysis of clinical trials. Europace 2022, 24, 1585–1598. [Google Scholar] [CrossRef]
  97. Miyamoto, K.; Tsuchiya, T.; Narita, S.; Yamaguchi, T.; Nagamoto, Y.; Ando, S.; Hayashida, K.; Tanioka, Y.; Takahashi, N. Bipolar electrogram amplitudes in the left atrium are related to local conduction velocity in patients with atrial fibrillation. Europace 2009, 11, 1597–1605. [Google Scholar] [CrossRef]
  98. Ali, R.L.; Qureshi, N.A.; Liverani, S.; Roney, C.H.; Kim, S.; Lim, P.B.; Tweedy, J.H.; Cantwell, C.D.; Peters, N.S. Left Atrial Enhancement Correlates with Myocardial Conduction Velocity in Patients with Persistent Atrial Fibrillation. Front. Physiol. 2020, 11, 570203. [Google Scholar] [CrossRef]
  99. Fukumoto, K.; Habibi, M.; Ipek, E.G.; Zahid, S.; Khurram, I.M.; Zimmerman, S.L.; Zipunnikov, V.; Spragg, D.; Ashikaga, H.; Trayanova, N.; et al. Association of Left Atrial Local Conduction Velocity with Late Gadolinium Enhancement on Cardiac Magnetic Resonance in Patients with Atrial Fibrillation. Circ. Arrhythm. Electrophysiol. 2016, 9, e002897. [Google Scholar] [CrossRef]
  100. Cochet, H.; Dubois, R.; Yamashita, S.; Al Jefairi, N.; Berte, B.; Sellal, J.M.; Hooks, D.; Frontera, A.; Amraoui, S.; Zemoura, A.; et al. Relationship between Fibrosis Detected on Late Gadolinium-Enhanced Cardiac Magnetic Resonance and Re-Entrant Activity Assessed with Electrocardiographic Imaging in Human Persistent Atrial Fibrillation. JACC Clin Electrophysiol. 2018, 4, 17–29. [Google Scholar] [CrossRef]
  101. Chrispin, J.; Gucuk Ipek, E.; Zahid, S.; Prakosa, A.; Habibi, M.; Spragg, D.; Marine, J.E.; Ashikaga, H.; Rickard, J.; Trayanova, N.A.; et al. Lack of regional association between atrial late gadolinium enhancement on cardiac magnetic resonance and atrial fibrillation rotors. Heart Rhythm 2016, 13, 654–660. [Google Scholar] [CrossRef] [PubMed]
  102. Sohns, C.; Lemes, C.; Metzner, A.; Fink, T.; Chmelevsky, M.; Maurer, T.; Budanova, M.; Solntsev, V.; Schulze, W.H.W.; Staab, W.; et al. First-in-Man Analysis of the Relationship between Electrical Rotors from Noninvasive Panoramic Mapping and Atrial Fibrosis from Magnetic Resonance Imaging in Patients with Persistent Atrial Fibrillation. Circ. Arrhythm. Electrophysiol. 2017, 10, e004419. [Google Scholar] [CrossRef] [PubMed]
  103. Nakamura, T.; Kiuchi, K.; Fukuzawa, K.; Takami, M.; Watanabe, Y.; Izawa, Y.; Suehiro, H.; Akita, T.; Takemoto, M.; Sakai, J.; et al. Late-gadolinium enhancement properties associated with atrial fibrillation rotors in patients with persistent atrial fibrillation. J. Cardiovasc. Electrophysiol. 2021, 32, 1005–1013. [Google Scholar] [CrossRef] [PubMed]
  104. Schade, A.; Nentwich, K.; Costello-Boerrigter, L.C.; Halbfass, P.; Mueller, P.; Roos, M.; Barth, S.; Krug, J.; Szoelloesi, G.A.; Lapp, H.; et al. Spatial Relationship of Focal Impulses, Rotors and Low Voltage Zones in Patients with Persistent Atrial Fibrillation. J. Cardiovasc. Electrophysiol. 2016, 27, 507–514. [Google Scholar] [CrossRef]
  105. King, J.H.; Huang, C.L.; Fraser, J.A. Determinants of myocardial conduction velocity: Implications for arrhythmogenesis. Front. Physiol. 2013, 4, 154. [Google Scholar] [CrossRef]
  106. Guichard, J.B.; Naud, P.; Xiong, F.; Qi, X.; L’Heureux, N.; Hiram, R.; Tardif, J.C.; Cartier, R.; Da Costa, A.; Nattel, S. Comparison of Atrial Remodeling Caused by Sustained Atrial Flutter versus Atrial Fibrillation. J. Am. Coll. Cardiol. 2020, 76, 374–388. [Google Scholar] [CrossRef]
  107. Nattel, S. Molecular and Cellular Mechanisms of Atrial Fibrosis in Atrial Fibrillation. JACC Clin. Electrophysiol. 2017, 3, 425–435. [Google Scholar] [CrossRef]
  108. Zhang, Z.; Vlcek, J.; Pauly, V.; Hesse, N.; Bauer, J.; Chataut, K.R.; Maderspacher, F.; Volz, L.S.; Buchberger, K.; Xia, R.; et al. Atrial fibrosis heterogeneity is a risk for atrial fibrillation in pigs with ischaemic heart failure. Eur. J. Clin. Investig. 2024, 54, e14137. [Google Scholar] [CrossRef]
  109. Jadidi, A.S.; Cochet, H.; Shah, A.J.; Kim, S.J.; Duncan, E.; Miyazaki, S.; Sermesant, M.; Lehrmann, H.; Lederlin, M.; Linton, N.; et al. Inverse relationship between fractionated electrograms and atrial fibrosis in persistent atrial fibrillation: Combined magnetic resonance imaging and high-density mapping. J. Am. Coll. Cardiol. 2013, 62, 802–812. [Google Scholar] [CrossRef]
  110. Haissaguerre, M.; Shah, A.J.; Cochet, H.; Hocini, M.; Dubois, R.; Efimov, I.; Vigmond, E.; Bernus, O.; Trayanova, N. Intermittent drivers anchoring to structural heterogeneities as a major pathophysiological mechanism of human persistent atrial fibrillation. J. Physiol. 2016, 594, 2387–2398. [Google Scholar] [CrossRef]
  111. Tanaka, K.; Zlochiver, S.; Vikstrom, K.L.; Yamazaki, M.; Moreno, J.; Klos, M.; Zaitsev, A.V.; Vaidyanathan, R.; Auerbach, D.S.; Landas, S.; et al. Spatial distribution of fibrosis governs fibrillation wave dynamics in the posterior left atrium during heart failure. Circ. Res. 2007, 101, 839–847. [Google Scholar] [CrossRef] [PubMed]
  112. Kazbanov, I.V.; ten Tusscher, K.H.; Panfilov, A.V. Effects of Heterogeneous Diffuse Fibrosis on Arrhythmia Dynamics and Mechanism. Sci. Rep. 2016, 6, 20835. [Google Scholar] [CrossRef] [PubMed]
  113. Xie, F.; Qu, Z.; Garfinkel, A.; Weiss, J.N. Electrophysiological heterogeneity and stability of reentry in simulated cardiac tissue. Am. J. Physiol. Heart Circ. Physiol. 2001, 280, H535–H545. [Google Scholar] [CrossRef] [PubMed]
  114. Saha, M.; Roney, C.H.; Bayer, J.D.; Meo, M.; Cochet, H.; Dubois, R.; Vigmond, E.J. Wavelength and Fibrosis Affect Phase Singularity Locations during Atrial Fibrillation. Front. Physiol. 2018, 9, 1207. [Google Scholar] [CrossRef] [PubMed]
  115. Zahid, S.; Cochet, H.; Boyle, P.M.; Schwarz, E.L.; Whyte, K.N.; Vigmond, E.J.; Dubois, R.; Hocini, M.; Haïssaguerre, M.; Jaïs, P.; et al. Patient-derived models link re-entrant driver localization in atrial fibrillation to fibrosis spatial pattern. Cardiovasc. Res. 2016, 110, 443–454. [Google Scholar] [CrossRef] [PubMed]
  116. Morgan, R.; Colman, M.A.; Chubb, H.; Seemann, G.; Aslanidi, O.V. Slow Conduction in the Border Zones of Patchy Fibrosis Stabilizes the Drivers for Atrial Fibrillation: Insights from Multi-Scale Human Atrial Modeling. Front. Physiol. 2016, 7, 474. [Google Scholar] [CrossRef]
  117. Lu, Z.; Scherlag, B.J.; Lin, J.; Niu, G.; Fung, K.M.; Zhao, L.; Ghias, M.; Jackman, W.M.; Lazzara, R.; Jiang, H.; et al. Atrial fibrillation begets atrial fibrillation: Autonomic mechanism for atrial electrical remodeling induced by short-term rapid atrial pacing. Circ. Arrhythm. Electrophysiol. 2008, 1, 184–192. [Google Scholar] [CrossRef]
  118. Armour, J.A.; Murphy, D.A.; Yuan, B.X.; Macdonald, S.; Hopkins, D.A. Gross and microscopic anatomy of the human intrinsic cardiac nervous system. Anat. Rec. 1997, 247, 289–298. [Google Scholar] [CrossRef]
  119. Patterson, E.; Po, S.S.; Scherlag, B.J.; Lazzara, R. Triggered firing in pulmonary veins initiated by in vitro autonomic nerve stimulation. Heart Rhythm 2005, 2, 624–631. [Google Scholar] [CrossRef]
  120. Po, S.S.; Li, Y.; Tang, D.; Liu, H.; Geng, N.; Jackman, W.M.; Scherlag, B.; Lazzara, R.; Patterson, E. Rapid and stable re-entry within the pulmonary vein as a mechanism initiating paroxysmal atrial fibrillation. J. Am. Coll. Cardiol. 2005, 45, 1871–1877. [Google Scholar] [CrossRef]
  121. Po, S.S.; Scherlag, B.J.; Yamanashi, W.S.; Edwards, J.; Zhou, J.; Wu, R.; Geng, N.; Lazzara, R.; Jackman, W.M. Experimental model for paroxysmal atrial fibrillation arising at the pulmonary vein-atrial junctions. Heart Rhythm 2006, 3, 201–208. [Google Scholar] [CrossRef] [PubMed]
  122. Choi, E.K.; Zhao, Y.; Everett, T.H.t.; Chen, P.S. Ganglionated plexi as neuromodulation targets for atrial fibrillation. J. Cardiovasc. Electrophysiol. 2017, 28, 1485–1491. [Google Scholar] [CrossRef] [PubMed]
  123. Rackley, J.; Nudy, M.; Gonzalez, M.D.; Naccarelli, G.; Maheshwari, A. Pulmonary vein isolation with adjunctive left atrial ganglionic plexus ablation for treatment of atrial fibrillation: A meta-analysis of randomized controlled trials. J. Interv. Card. Electrophysiol. 2023, 66, 333–342. [Google Scholar] [CrossRef] [PubMed]
  124. Kampaktsis, P.N.; Oikonomou, E.K.; Choi, D.Y.; Cheung, J.W. Efficacy of ganglionated plexi ablation in addition to pulmonary vein isolation for paroxysmal versus persistent atrial fibrillation: A meta-analysis of randomized controlled clinical trials. J. Interv. Card. Electrophysiol. 2017, 50, 253–260. [Google Scholar] [CrossRef]
  125. Doshi, R.N.; Wu, T.J.; Yashima, M.; Kim, Y.H.; Ong, J.J.; Cao, J.M.; Hwang, C.; Yashar, P.; Fishbein, M.C.; Karagueuzian, H.S.; et al. Relation between ligament of Marshall and adrenergic atrial tachyarrhythmia. Circulation 1999, 100, 876–883. [Google Scholar] [CrossRef]
  126. Lam, A.; Küffer, T.; Hunziker, L.; Nozica, N.; Asatryan, B.; Franzeck, F.; Madaffari, A.; Haeberlin, A.; Mühl, A.; Servatius, H.; et al. Efficacy and safety of ethanol infusion into the vein of Marshall for mitral isthmus ablation. J. Cardiovasc. Electrophysiol. 2021, 32, 1610–1619. [Google Scholar] [CrossRef]
  127. Valderrábano, M.; Peterson, L.E.; Swarup, V.; Schurmann, P.A.; Makkar, A.; Doshi, R.N.; DeLurgio, D.; Athill, C.A.; Ellenbogen, K.A.; Natale, A.; et al. Effect of Catheter Ablation with Vein of Marshall Ethanol Infusion vs Catheter Ablation Alone on Persistent Atrial Fibrillation: The VENUS Randomized Clinical Trial. JAMA 2020, 324, 1620–1628. [Google Scholar] [CrossRef]
  128. Bosch, R.F.; Zeng, X.; Grammer, J.B.; Popovic, K.; Mewis, C.; Kühlkamp, V. Ionic mechanisms of electrical remodeling in human atrial fibrillation. Cardiovasc. Res. 1999, 44, 121–131. [Google Scholar] [CrossRef]
  129. Workman, A.J.; Kane, K.A.; Rankin, A.C. The contribution of ionic currents to changes in refractoriness of human atrial myocytes associated with chronic atrial fibrillation. Cardiovasc. Res. 2001, 52, 226–235. [Google Scholar] [CrossRef]
  130. Van Wagoner, D.R.; Pond, A.L.; Lamorgese, M.; Rossie, S.S.; McCarthy, P.M.; Nerbonne, J.M. Atrial L-type Ca2+ currents and human atrial fibrillation. Circ. Res. 1999, 85, 428–436. [Google Scholar] [CrossRef]
  131. Skasa, M.; Jüngling, E.; Picht, E.; Schöndube, F.; Lückhoff, A. L-type calcium currents in atrial myocytes from patients with persistent and non-persistent atrial fibrillation. Basic Res. Cardiol. 2001, 96, 151–159. [Google Scholar] [CrossRef] [PubMed]
  132. Yue, L.; Melnyk, P.; Gaspo, R.; Wang, Z.; Nattel, S. Molecular mechanisms underlying ionic remodeling in a dog model of atrial fibrillation. Circ. Res. 1999, 84, 776–784. [Google Scholar] [CrossRef] [PubMed]
  133. Severs, N.J.; Bruce, A.F.; Dupont, E.; Rothery, S. Remodelling of gap junctions and connexin expression in diseased myocardium. Cardiovasc. Res. 2008, 80, 9–19. [Google Scholar] [CrossRef] [PubMed]
  134. van der Velden, H.M.; Ausma, J.; Rook, M.B.; Hellemons, A.J.; van Veen, T.A.; Allessie, M.A.; Jongsma, H.J. Gap junctional remodeling in relation to stabilization of atrial fibrillation in the goat. Cardiovasc. Res. 2000, 46, 476–486. [Google Scholar] [CrossRef]
  135. van der Velden, H.M.; van Kempen, M.J.; Wijffels, M.C.; van Zijverden, M.; Groenewegen, W.A.; Allessie, M.A.; Jongsma, H.J. Altered pattern of connexin40 distribution in persistent atrial fibrillation in the goat. J. Cardiovasc. Electrophysiol. 1998, 9, 596–607. [Google Scholar] [CrossRef]
  136. Polontchouk, L.; Haefliger, J.A.; Ebelt, B.; Schaefer, T.; Stuhlmann, D.; Mehlhorn, U.; Kuhn-Regnier, F.; De Vivie, E.R.; Dhein, S. Effects of chronic atrial fibrillation on gap junction distribution in human and rat atria. J. Am. Coll. Cardiol. 2001, 38, 883–891. [Google Scholar] [CrossRef]
  137. Yeh, H.I.; Lai, Y.J.; Lee, S.H.; Chen, S.T.; Ko, Y.S.; Chen, S.A.; Severs, N.J.; Tsai, C.H. Remodeling of myocardial sleeve and gap junctions in canine superior vena cava after rapid pacing. Basic Res. Cardiol. 2006, 101, 269–280. [Google Scholar] [CrossRef]
  138. Kostin, S.; Klein, G.; Szalay, Z.; Hein, S.; Bauer, E.P.; Schaper, J. Structural correlate of atrial fibrillation in human patients. Cardiovasc. Res. 2002, 54, 361–379. [Google Scholar] [CrossRef]
  139. Jennings, M.M.; Donahue, J.K. Connexin Remodeling Contributes to Atrial Fibrillation. J. Atr. Fibrillation 2013, 6, 839. [Google Scholar] [CrossRef]
  140. Sakabe, M.; Fujiki, A.; Nishida, K.; Sugao, M.; Nagasawa, H.; Tsuneda, T.; Mizumaki, K.; Inoue, H. Enalapril prevents perpetuation of atrial fibrillation by suppressing atrial fibrosis and over-expression of connexin43 in a canine model of atrial pacing-induced left ventricular dysfunction. J. Cardiovasc. Pharmacol. 2004, 43, 851–859. [Google Scholar] [CrossRef]
  141. Elvan, A.; Huang, X.D.; Pressler, M.L.; Zipes, D.P. Radiofrequency catheter ablation of the atria eliminates pacing-induced sustained atrial fibrillation and reduces connexin 43 in dogs. Circulation 1997, 96, 1675–1685. [Google Scholar] [CrossRef] [PubMed]
  142. Chaldoupi, S.M.; Loh, P.; Hauer, R.N.; de Bakker, J.M.; van Rijen, H.V. The role of connexin40 in atrial fibrillation. Cardiovasc. Res. 2009, 84, 15–23. [Google Scholar] [CrossRef] [PubMed]
  143. Nattel, S.; Heijman, J.; Zhou, L.; Dobrev, D. Molecular Basis of Atrial Fibrillation Pathophysiology and Therapy: A Translational Perspective. Circ. Res. 2020, 127, 51–72. [Google Scholar] [CrossRef] [PubMed]
  144. Imanaga, I.; Hai, L.; Ogawa, K.; Matsumura, K.; Mayama, T. Phosphorylation of connexin in functional regulation of the cardiac gap junction. Exp. Clin. Cardiol. 2004, 9, 161–164. [Google Scholar] [PubMed]
  145. TenBroek, E.M.; Lampe, P.D.; Solan, J.L.; Reynhout, J.K.; Johnson, R.G. Ser364 of connexin43 and the upregulation of gap junction assembly by cAMP. J. Cell Biol. 2001, 155, 1307–1318. [Google Scholar] [CrossRef]
  146. Saidi Brikci-Nigassa, A.; Clement, M.J.; Ha-Duong, T.; Adjadj, E.; Ziani, L.; Pastre, D.; Curmi, P.A.; Savarin, P. Phosphorylation controls the interaction of the connexin43 C-terminal domain with tubulin and microtubules. Biochemistry 2012, 51, 4331–4342. [Google Scholar] [CrossRef]
  147. Takanari, H.; Bourgonje, V.J.; Fontes, M.S.; Raaijmakers, A.J.; Driessen, H.; Jansen, J.A.; van der Nagel, R.; Kok, B.; van Stuijvenberg, L.; Boulaksil, M.; et al. Calmodulin/CaMKII inhibition improves intercellular communication and impulse propagation in the heart and is antiarrhythmic under conditions when fibrosis is absent. Cardiovasc. Res. 2016, 111, 410–421. [Google Scholar] [CrossRef]
  148. Zhong, C.; Chang, H.; Wu, Y.; Zhou, L.; Wang, Y.; Wang, M.; Wu, P.; Qi, Z.; Zou, J. Up-regulated Cx43 phosphorylation at Ser368 prolongs QRS duration in myocarditis. J. Cell Mol. Med. 2018, 22, 3537–3547. [Google Scholar] [CrossRef]
  149. Forrester, S.J.; Booz, G.W.; Sigmund, C.D.; Coffman, T.M.; Kawai, T.; Rizzo, V.; Scalia, R.; Eguchi, S. Angiotensin II Signal Transduction: An Update on Mechanisms of Physiology and Pathophysiology. Physiol. Rev. 2018, 98, 1627–1738. [Google Scholar] [CrossRef]
  150. Nattel, S.; Dobrev, D. Electrophysiological and molecular mechanisms of paroxysmal atrial fibrillation. Nat. Rev. Cardiol. 2016, 13, 575–590. [Google Scholar] [CrossRef]
  151. Wakili, R.; Voigt, N.; Kääb, S.; Dobrev, D.; Nattel, S. Recent advances in the molecular pathophysiology of atrial fibrillation. J. Clin. Investig. 2011, 121, 2955–2968. [Google Scholar] [CrossRef] [PubMed]
  152. Stacy, M.R.; Lin, B.A.; Thorn, S.L.; Lobb, D.C.; Maxfield, M.W.; Novack, C.; Zellars, K.N.; Freeburg, L.; Akar, J.G.; Sinusas, A.J.; et al. Regional heterogeneity in determinants of atrial matrix remodeling and association with atrial fibrillation vulnerability postmyocardial infarction. Heart Rhythm 2022, 19, 847–855. [Google Scholar] [CrossRef] [PubMed]
  153. Ihara, K.; Sasano, T. Role of Inflammation in the Pathogenesis of Atrial Fibrillation. Front. Physiol. 2022, 13, 862164. [Google Scholar] [CrossRef] [PubMed]
  154. Willar, B.; Tran, K.V.; Fitzgibbons, T.P. Epicardial adipocytes in the pathogenesis of atrial fibrillation: An update on basic and translational studies. Front. Endocrinol. 2023, 14, 1154824. [Google Scholar] [CrossRef]
  155. Venteclef, N.; Guglielmi, V.; Balse, E.; Gaborit, B.; Cotillard, A.; Atassi, F.; Amour, J.; Leprince, P.; Dutour, A.; Clément, K.; et al. Human epicardial adipose tissue induces fibrosis of the atrial myocardium through the secretion of adipo-fibrokines. Eur. Heart. J. 2015, 36, 795–805a. [Google Scholar] [CrossRef]
  156. Nalliah, C.J.; Bell, J.R.; Raaijmakers, A.J.A.; Waddell, H.M.; Wells, S.P.; Bernasochi, G.B.; Montgomery, M.K.; Binny, S.; Watts, T.; Joshi, S.B.; et al. Epicardial Adipose Tissue Accumulation Confers Atrial Conduction Abnormality. J. Am. Coll. Cardiol. 2020, 76, 1197–1211. [Google Scholar] [CrossRef]
  157. Abe, I.; Teshima, Y.; Kondo, H.; Kaku, H.; Kira, S.; Ikebe, Y.; Saito, S.; Fukui, A.; Shinohara, T.; Yufu, K.; et al. Association of fibrotic remodeling and cytokines/chemokines content in epicardial adipose tissue with atrial myocardial fibrosis in patients with atrial fibrillation. Heart Rhythm 2018, 15, 1717–1727. [Google Scholar] [CrossRef]
  158. Mazurek, T.; Zhang, L.; Zalewski, A.; Mannion, J.D.; Diehl, J.T.; Arafat, H.; Sarov-Blat, L.; O’Brien, S.; Keiper, E.A.; Johnson, A.G.; et al. Human epicardial adipose tissue is a source of inflammatory mediators. Circulation 2003, 108, 2460–2466. [Google Scholar] [CrossRef]
  159. Tsao, H.M.; Hu, W.C.; Wu, M.H.; Tai, C.T.; Lin, Y.J.; Chang, S.L.; Lo, L.W.; Hu, Y.F.; Tuan, T.C.; Wu, T.J.; et al. Quantitative analysis of quantity and distribution of epicardial adipose tissue surrounding the left atrium in patients with atrial fibrillation and effect of recurrence after ablation. Am. J. Cardiol. 2011, 107, 1498–1503. [Google Scholar] [CrossRef]
  160. Tsai, C.T.; Lai, L.P.; Hwang, J.J.; Lin, J.L.; Chiang, F.T. Molecular genetics of atrial fibrillation. J. Am. Coll. Cardiol. 2008, 52, 241–250. [Google Scholar] [CrossRef]
  161. Chen, Y.H.; Xu, S.J.; Bendahhou, S.; Wang, X.L.; Wang, Y.; Xu, W.Y.; Jin, H.W.; Sun, H.; Su, X.Y.; Zhuang, Q.N.; et al. KCNQ1 gain-of-function mutation in familial atrial fibrillation. Science 2003, 299, 251–254. [Google Scholar] [CrossRef] [PubMed]
  162. Hong, K.; Bjerregaard, P.; Gussak, I.; Brugada, R. Short QT syndrome and atrial fibrillation caused by mutation in KCNH2. J. Cardiovasc. Electrophysiol. 2005, 16, 394–396. [Google Scholar] [CrossRef] [PubMed]
  163. Xia, M.; Jin, Q.; Bendahhou, S.; He, Y.; Larroque, M.M.; Chen, Y.; Zhou, Q.; Yang, Y.; Liu, Y.; Liu, B.; et al. A Kir2.1 gain-of-function mutation underlies familial atrial fibrillation. Biochem. Biophys. Res. Commun. 2005, 332, 1012–1019. [Google Scholar] [CrossRef] [PubMed]
  164. Roselli, C.; Rienstra, M.; Ellinor, P.T. Genetics of Atrial Fibrillation in 2020: GWAS, Genome Sequencing, Polygenic Risk, and Beyond. Circ. Res. 2020, 127, 21–33. [Google Scholar] [CrossRef]
  165. Gudbjartsson, D.F.; Arnar, D.O.; Helgadottir, A.; Gretarsdottir, S.; Holm, H.; Sigurdsson, A.; Jonasdottir, A.; Baker, A.; Thorleifsson, G.; Kristjansson, K.; et al. Variants conferring risk of atrial fibrillation on chromosome 4q25. Nature 2007, 448, 353–357. [Google Scholar] [CrossRef]
  166. Ryan, A.K.; Blumberg, B.; Rodriguez-Esteban, C.; Yonei-Tamura, S.; Tamura, K.; Tsukui, T.; de la Peña, J.; Sabbagh, W.; Greenwald, J.; Choe, S.; et al. Pitx2 determines left-right asymmetry of internal organs in vertebrates. Nature 1998, 394, 545–551. [Google Scholar] [CrossRef]
  167. Campione, M.; Ros, M.A.; Icardo, J.M.; Piedra, E.; Christoffels, V.M.; Schweickert, A.; Blum, M.; Franco, D.; Moorman, A.F. Pitx2 expression defines a left cardiac lineage of cells: Evidence for atrial and ventricular molecular isomerism in the iv/iv mice. Dev. Biol. 2001, 231, 252–264. [Google Scholar] [CrossRef]
  168. Chinchilla, A.; Daimi, H.; Lozano-Velasco, E.; Dominguez, J.N.; Caballero, R.; Delpón, E.; Tamargo, J.; Cinca, J.; Hove-Madsen, L.; Aranega, A.E.; et al. PITX2 insufficiency leads to atrial electrical and structural remodeling linked to arrhythmogenesis. Circ. Cardiovasc. Genet. 2011, 4, 269–279. [Google Scholar] [CrossRef]
  169. Kirchhof, P.; Kahr, P.C.; Kaese, S.; Piccini, I.; Vokshi, I.; Scheld, H.H.; Rotering, H.; Fortmueller, L.; Laakmann, S.; Verheule, S.; et al. PITX2c is expressed in the adult left atrium, and reducing Pitx2c expression promotes atrial fibrillation inducibility and complex changes in gene expression. Circ. Cardiovasc. Genet. 2011, 4, 123–133. [Google Scholar] [CrossRef]
  170. Mommersteeg, M.T.; Brown, N.A.; Prall, O.W.; de Gier-de Vries, C.; Harvey, R.P.; Moorman, A.F.; Christoffels, V.M. Pitx2c and Nkx2-5 are required for the formation and identity of the pulmonary myocardium. Circ. Res. 2007, 101, 902–909. [Google Scholar] [CrossRef]
  171. Li, Z.; Hertervig, E.; Yuan, S.; Yang, Y.; Lin, Z.; Olsson, S.B. Dispersion of atrial repolarization in patients with paroxysmal atrial fibrillation. Europace 2001, 3, 285–291. [Google Scholar] [CrossRef] [PubMed]
  172. Yuan, S.; Wohlfart, B.; Olsson, S.B.; Blomström-Lundqvist, C. The dispersion of repolarization in patients with ventricular tachycardia. A study using simultaneous monophasic action potential recordings from two sites in the right ventricle. Eur. Heart J. 1995, 16, 68–76. [Google Scholar] [CrossRef] [PubMed]
  173. Garfinkel, A.; Kim, Y.H.; Voroshilovsky, O.; Qu, Z.; Kil, J.R.; Lee, M.H.; Karagueuzian, H.S.; Weiss, J.N.; Chen, P.S. Preventing ventricular fibrillation by flattening cardiac restitution. Proc. Natl. Acad. Sci. USA 2000, 97, 6061–6066. [Google Scholar] [CrossRef] [PubMed]
  174. Pop, T.; Fleischmann, D. Alternans in human atrial monophasic action potenial. Br. Heart. J. 1977, 39, 1273–1275. [Google Scholar] [CrossRef] [PubMed]
  175. Ihara, K.; Sugiyama, K.; Takahashi, K.; Yamazoe, M.; Sasano, T.; Furukawa, T. Electrophysiological Assessment of Murine Atria with High-Resolution Optical Mapping. J. Vis. Exp. 2018, 32, e56478. [Google Scholar] [CrossRef]
  176. Fareh, S.; Villemaire, C.; Nattel, S. Importance of refractoriness heterogeneity in the enhanced vulnerability to atrial fibrillation induction caused by tachycardia-induced atrial electrical remodeling. Circulation 1998, 98, 2202–2209. [Google Scholar] [CrossRef]
  177. Wood, M.A.; Mangano, R.A.; Schieken, R.M.; Baumgarten, C.M.; Simpson, P.M.; Ellenbogen, K.A. Modulation of atrial repolarization by site of pacing in the isolated rabbit heart. Circulation 1996, 94, 1465–1470. [Google Scholar] [CrossRef]
  178. Arora, R.; Verheule, S.; Scott, L.; Navarrete, A.; Katari, V.; Wilson, E.; Vaz, D.; Olgin, J.E. Arrhythmogenic substrate of the pulmonary veins assessed by high-resolution optical mapping. Circulation 2003, 107, 1816–1821. [Google Scholar] [CrossRef]
  179. Anyukhovsky, E.P.; Sosunov, E.A.; Chandra, P.; Rosen, T.S.; Boyden, P.A.; Danilo, P., Jr.; Rosen, M.R. Age-associated changes in electrophysiologic remodeling: A potential contributor to initiation of atrial fibrillation. Cardiovasc. Res. 2005, 66, 353–363. [Google Scholar] [CrossRef]
  180. Ehrlich, J.R.; Cha, T.J.; Zhang, L.; Chartier, D.; Melnyk, P.; Hohnloser, S.H.; Nattel, S. Cellular electrophysiology of canine pulmonary vein cardiomyocytes: Action potential and ionic current properties. J. Physiol. 2003, 551, 801–813. [Google Scholar] [CrossRef]
  181. Diker, E.; Ozdemir, M.; Aydoğdu, S.; Tezcan, U.K.; Korkmaz, S.; Kütük, E.; Göksel, S. Dispersion of repolarization in paroxysmal atrial fibrillation. Int. J. Cardiol. 1998, 63, 281–286. [Google Scholar] [CrossRef] [PubMed]
  182. Namba, T.; Ashihara, T.; Nakazawa, K.; Ohe, T. Spatial heterogeneity in refractoriness as a proarrhythmic substrate: Theoretical evaluation by numerical simulation. Jpn Circ. J. 2000, 64, 121–129. [Google Scholar] [CrossRef] [PubMed]
  183. Watanabe, M.; Yokoshiki, H.; Mitsuyama, H.; Mizukami, K.; Ono, T.; Tsutsui, H. Conduction and refractory disorders in the diabetic atrium. Am. J. Physiol. Heart Circ. Physiol. 2012, 303, H86–H95. [Google Scholar] [CrossRef] [PubMed]
  184. Qu, Z.; Xie, F.; Garfinkel, A.; Weiss, J.N. Origins of spiral wave meander and breakup in a two-dimensional cardiac tissue model. Ann. Biomed. Eng. 2000, 28, 755–771. [Google Scholar] [CrossRef] [PubMed]
  185. Ciaccio, E.J.; Peters, N.S.; Garan, H. Effects of refractory gradients and ablation on fibrillatory activity. Comput. Biol. Med. 2018, 95, 175–187. [Google Scholar] [CrossRef]
  186. Seitz, J.; Bars, C.; Théodore, G.; Beurtheret, S.; Lellouche, N.; Bremondy, M.; Ferracci, A.; Faure, J.; Penaranda, G.; Yamazaki, M.; et al. AF Ablation Guided by Spatiotemporal Electrogram Dispersion without Pulmonary Vein Isolation: A Wholly Patient-Tailored Approach. J. Am. Coll. Cardiol. 2017, 69, 303–321. [Google Scholar] [CrossRef]
  187. Qu, Z.; Garfinkel, A.; Chen, P.S.; Weiss, J.N. Mechanisms of discordant alternans and induction of reentry in simulated cardiac tissue. Circulation 2000, 102, 1664–1670. [Google Scholar] [CrossRef]
  188. Colman, M.A.; Aslanidi, O.V.; Kharche, S.; Boyett, M.R.; Garratt, C.; Hancox, J.C.; Zhang, H. Pro-arrhythmogenic effects of atrial fibrillation-induced electrical remodelling: Insights from the three-dimensional virtual human atria. J. Physiol. 2013, 591, 4249–4272. [Google Scholar] [CrossRef]
  189. Gaborit, N.; Le Bouter, S.; Szuts, V.; Varro, A.; Escande, D.; Nattel, S.; Demolombe, S. Regional and tissue specific transcript signatures of ion channel genes in the non-diseased human heart. J. Physiol. 2007, 582, 675–693. [Google Scholar] [CrossRef]
  190. Voigt, N.; Trausch, A.; Knaut, M.; Matschke, K.; Varró, A.; Van Wagoner, D.R.; Nattel, S.; Ravens, U.; Dobrev, D. Left-to-right atrial inward rectifier potassium current gradients in patients with paroxysmal versus chronic atrial fibrillation. Circ. Arrhythm. Electrophysiol. 2010, 3, 472–480. [Google Scholar] [CrossRef]
  191. Zhang, H.; Garratt, C.J.; Zhu, J.; Holden, A.V. Role of up-regulation of IK1 in action potential shortening associated with atrial fibrillation in humans. Cardiovasc. Res. 2005, 66, 493–502. [Google Scholar] [CrossRef] [PubMed]
  192. Berenfeld, O. The Major Role of I(K1) in Mechanisms of Rotor Drift in the Atria: A Computational Study. Clin. Med. Insights Cardiol. 2016, 10, 71–79. [Google Scholar] [CrossRef] [PubMed]
  193. Calvo, C.J.; Deo, M.; Zlochiver, S.; Millet, J.; Berenfeld, O. Attraction of rotors to the pulmonary veins in paroxysmal atrial fibrillation: A modeling study. Biophys. J. 2014, 106, 1811–1821. [Google Scholar] [CrossRef] [PubMed]
  194. Honarbakhsh, S.; Schilling, R.J.; Orini, M.; Providencia, R.; Keating, E.; Finlay, M.; Sporton, S.; Chow, A.; Earley, M.J.; Lambiase, P.D.; et al. Structural remodeling and conduction velocity dynamics in the human left atrium: Relationship with reentrant mechanisms sustaining atrial fibrillation. Heart Rhythm 2019, 16, 18–25. [Google Scholar] [CrossRef]
  195. Qu, Z.; Weiss, J.N. Cardiac Alternans: From Bedside to Bench and Back. Circ. Res. 2023, 132, 127–149. [Google Scholar] [CrossRef]
  196. Al Abed, A.; Lovell, N.H.; Dokos, S. Local Heterogeneous Electrical Restitution Properties of Rabbit Atria. J. Cardiovasc. Electrophysiol. 2016, 27, 743–753. [Google Scholar] [CrossRef]
  197. Kim, B.S.; Kim, Y.H.; Hwang, G.S.; Pak, H.N.; Lee, S.C.; Shim, W.J.; Oh, D.J.; Ro, Y.M. Action potential duration restitution kinetics in human atrial fibrillation. J. Am. Coll. Cardiol. 2002, 39, 1329–1336. [Google Scholar] [CrossRef]
  198. Narayan, S.M.; Franz, M.R.; Clopton, P.; Pruvot, E.J.; Krummen, D.E. Repolarization alternans reveals vulnerability to human atrial fibrillation. Circulation 2011, 123, 2922–2930. [Google Scholar] [CrossRef]
  199. Hiromoto, K.; Shimizu, H.; Furukawa, Y.; Kanemori, T.; Mine, T.; Masuyama, T.; Ohyanagi, M. Discordant repolarization alternans-induced atrial fibrillation is suppressed by verapamil. Circ. J. 2005, 69, 1368–1373. [Google Scholar] [CrossRef]
  200. Liu, T.; Xiong, F.; Qi, X.Y.; Xiao, J.; Villeneuve, L.; Abu-Taha, I.; Dobrev, D.; Huang, C.; Nattel, S. Altered calcium handling produces reentry-promoting action potential alternans in atrial fibrillation-remodeled hearts. JCI Insight 2020, 5, e133754. [Google Scholar] [CrossRef]
  201. Maesen, B.; Zeemering, S.; Afonso, C.; Eckstein, J.; Burton, R.A.; van Hunnik, A.; Stuckey, D.J.; Tyler, D.; Maessen, J.; Grau, V.; et al. Rearrangement of atrial bundle architecture and consequent changes in anisotropy of conduction constitute the 3-dimensional substrate for atrial fibrillation. Circ. Arrhythm. Electrophysiol. 2013, 6, 967–975. [Google Scholar] [CrossRef] [PubMed]
  202. Frontera, A.; Pagani, S.; Limite, L.R.; Peirone, A.; Fioravanti, F.; Enache, B.; Cuellar Silva, J.; Vlachos, K.; Meyer, C.; Montesano, G.; et al. Slow Conduction Corridors and Pivot Sites Characterize the Electrical Remodeling in Atrial Fibrillation. JACC Clin. Electrophysiol. 2022, 8, 561–577. [Google Scholar] [CrossRef] [PubMed]
  203. Ueda, N.; Yamamoto, M.; Honjo, H.; Kodama, I.; Kamiya, K. The role of gap junctions in stretch-induced atrial fibrillation. Cardiovasc. Res. 2014, 104, 364–370. [Google Scholar] [CrossRef] [PubMed]
  204. Koura, T.; Hara, M.; Takeuchi, S.; Ota, K.; Okada, Y.; Miyoshi, S.; Watanabe, A.; Shiraiwa, K.; Mitamura, H.; Kodama, I.; et al. Anisotropic conduction properties in canine atria analyzed by high-resolution optical mapping: Preferential direction of conduction block changes from longitudinal to transverse with increasing age. Circulation 2002, 105, 2092–2098. [Google Scholar] [CrossRef] [PubMed]
  205. Konings, K.T.; Smeets, J.L.; Penn, O.C.; Wellens, H.J.; Allessie, M.A. Configuration of unipolar atrial electrograms during electrically induced atrial fibrillation in humans. Circulation 1997, 95, 1231–1241. [Google Scholar] [CrossRef]
  206. Nademanee, K.; McKenzie, J.; Kosar, E.; Schwab, M.; Sunsaneewitayakul, B.; Vasavakul, T.; Khunnawat, C.; Ngarmukos, T. A new approach for catheter ablation of atrial fibrillation: Mapping of the electrophysiologic substrate. J. Am. Coll. Cardiol. 2004, 43, 2044–2053. [Google Scholar] [CrossRef]
  207. Wu, S.H.; Jiang, W.F.; Gu, J.; Zhao, L.; Wang, Y.L.; Liu, Y.G.; Zhou, L.; Gu, J.N.; Xu, K.; Liu, X. Benefits and risks of additional ablation of complex fractionated atrial electrograms for patients with atrial fibrillation: A systematic review and meta-analysis. Int. J. Cardiol. 2013, 169, 35–43. [Google Scholar] [CrossRef]
  208. de Groot, N.; van der Does, L.; Yaksh, A.; Lanters, E.; Teuwen, C.; Knops, P.; van de Woestijne, P.; Bekkers, J.; Kik, C.; Bogers, A.; et al. Direct Proof of Endo-Epicardial Asynchrony of the Atrial Wall During Atrial Fibrillation in Humans. Circ. Arrhythm. Electrophysiol. 2016, 9, e003648. [Google Scholar] [CrossRef]
  209. Kharbanda, R.K.; Kik, C.; Knops, P.; Bogers, A.; de Groot, N.M.S. First Evidence of Endo-Epicardial Asynchrony of the Left Atrial Wall in Humans. JACC Case Rep. 2020, 2, 745–749. [Google Scholar] [CrossRef]
  210. Iwamiya, S.; Ihara, K.; Furukawa, T.; Sasano, T. Sacubitril/valsartan attenuates atrial conduction disturbance and electrophysiological heterogeneity with ameliorating fibrosis in mice. Front. Cardiovasc. Med. 2024, 11, 1341601. [Google Scholar] [CrossRef]
  211. Eijsbouts, S.C.; Majidi, M.; van Zandvoort, M.; Allessie, M.A. Effects of acute atrial dilation on heterogeneity in conduction in the isolated rabbit heart. J. Cardiovasc. Electrophysiol. 2003, 14, 269–278. [Google Scholar] [CrossRef] [PubMed]
  212. Bian, W.; Tung, L. Structure-related initiation of reentry by rapid pacing in monolayers of cardiac cells. Circ. Res. 2006, 98, e29–e38. [Google Scholar] [CrossRef] [PubMed]
  213. Lalani, G.G.; Schricker, A.; Gibson, M.; Rostamian, A.; Krummen, D.E.; Narayan, S.M. Atrial conduction slows immediately before the onset of human atrial fibrillation: A bi-atrial contact mapping study of transitions to atrial fibrillation. J. Am. Coll. Cardiol. 2012, 59, 595–606. [Google Scholar] [CrossRef] [PubMed]
  214. Schricker, A.A.; Lalani, G.G.; Krummen, D.E.; Rappel, W.J.; Narayan, S.M. Human atrial fibrillation initiates via organized rather than disorganized mechanisms. Circ. Arrhythm. Electrophysiol. 2014, 7, 816–824. [Google Scholar] [CrossRef] [PubMed]
  215. Duytschaever, M.; Danse, P.; Eysbouts, S.; Allessie, M. Is there an optimal pacing site to prevent atrial fibrillation?: An experimental study in the chronically instrumented goat. J. Cardiovasc. Electrophysiol. 2002, 13, 1264–1271. [Google Scholar] [CrossRef] [PubMed]
  216. van Schie, M.S.; Ramdat Misier, N.L.; Razavi Ebrahimi, P.; Heida, A.; Kharbanda, R.K.; Taverne, Y.; de Groot, N.M.S. Premature atrial contractions promote local directional heterogeneities in conduction velocity vectors. Europace 2023, 25, 1162–1171. [Google Scholar] [CrossRef]
  217. Kumagai, K.; Tojo, H.; Noguchi, H.; Yasuda, T.; Ogawa, M.; Nakashima, H.; Zhang, B.; Saku, K. Effects of the NA+ channel blocker pilsicainide on the electrophysiologic properties of pulmonary veins in patients with atrial fibrillation. J. Cardiovasc. Electrophysiol. 2004, 15, 1396–1401. [Google Scholar] [CrossRef]
  218. Spach, M.S.; Dolber, P.C.; Heidlage, J.F. Influence of the passive anisotropic properties on directional differences in propagation following modification of the sodium conductance in human atrial muscle. A model of reentry based on anisotropic discontinuous propagation. Circ. Res. 1988, 62, 811–832. [Google Scholar] [CrossRef]
  219. Spach, M.S.; Dolber, P.C.; Heidlage, J.F. Interaction of inhomogeneities of repolarization with anisotropic propagation in dog atria. A mechanism for both preventing and initiating reentry. Circ. Res. 1989, 65, 1612–1631. [Google Scholar] [CrossRef]
  220. Spach, M.S.; Boineau, J.P. Microfibrosis produces electrical load variations due to loss of side-to-side cell connections: A major mechanism of structural heart disease arrhythmias. Pacing Clin. Electrophysiol. 1997, 20, 397–413. [Google Scholar] [CrossRef]
  221. Jaïs, P.; Hsu, L.F.; Rotter, M.; Sanders, P.; Takahashi, Y.; Rostock, T.; Sacher, F.; Hocini, M.; Clémenty, J.; Haïssaguerre, M. Mitral isthmus ablation for atrial fibrillation. J. Cardiovasc. Electrophysiol 2005, 16, 1157–1159. [Google Scholar] [CrossRef] [PubMed]
  222. Hocini, M.; Jaïs, P.; Sanders, P.; Takahashi, Y.; Rotter, M.; Rostock, T.; Hsu, L.F.; Sacher, F.; Reuter, S.; Clémenty, J.; et al. Techniques, evaluation, and consequences of linear block at the left atrial roof in paroxysmal atrial fibrillation: A prospective randomized study. Circulation 2005, 112, 3688–3696. [Google Scholar] [CrossRef] [PubMed]
  223. Narayan, S.M.; Baykaner, T.; Clopton, P.; Schricker, A.; Lalani, G.G.; Krummen, D.E.; Shivkumar, K.; Miller, J.M. Ablation of rotor and focal sources reduces late recurrence of atrial fibrillation compared with trigger ablation alone: Extended follow-up of the CONFIRM trial (Conventional Ablation for Atrial Fibrillation with or without Focal Impulse and Rotor Modulation). J. Am. Coll. Cardiol. 2014, 63, 1761–1768. [Google Scholar] [CrossRef] [PubMed]
  224. Fareh, S.; Bénardeau, A.; Thibault, B.; Nattel, S. The T-type Ca2+ channel blocker mibefradil prevents the development of a substrate for atrial fibrillation by tachycardia-induced atrial remodeling in dogs. Circulation 1999, 100, 2191–2197. [Google Scholar] [CrossRef]
  225. Li, D.; Shinagawa, K.; Pang, L.; Leung, T.K.; Cardin, S.; Wang, Z.; Nattel, S. Effects of angiotensin-converting enzyme inhibition on the development of the atrial fibrillation substrate in dogs with ventricular tachypacing-induced congestive heart failure. Circulation 2001, 104, 2608–2614. [Google Scholar] [CrossRef]
  226. Liao, J.; Ebrahimi, R.; Ling, Z.; Meyer, C.; Martinek, M.; Sommer, P.; Futyma, P.; Di Vece, D.; Schratter, A.; Acou, W.J.; et al. Effect of SGLT-2 inhibitors on arrhythmia events: Insight from an updated secondary analysis of  >80,000 patients (the SGLT2i-Arrhythmias and Sudden Cardiac Death). Cardiovasc. Diabetol. 2024, 23, 78. [Google Scholar] [CrossRef]
Figure 1. Macro-, meso-, microscopic structural and electrophysiological heterogeneities. RA: right atrium, RAA: right atrial appendage, LA: left atrium, PV: pulmonary vein, SVC: superior vena cava, IVC: inferior vena cava, Macro: macroscopic, Meso: mesoscopic, and Micro: microscopic. Numbers in parentheses indicate the corresponding section numbers in the text.
Figure 1. Macro-, meso-, microscopic structural and electrophysiological heterogeneities. RA: right atrium, RAA: right atrial appendage, LA: left atrium, PV: pulmonary vein, SVC: superior vena cava, IVC: inferior vena cava, Macro: macroscopic, Meso: mesoscopic, and Micro: microscopic. Numbers in parentheses indicate the corresponding section numbers in the text.
Ijms 25 10193 g001
Figure 2. Anatomy of human atria in 3D computed tomography. The endocardial aspects of the atria and surrounding structures above the mitral and tricuspid annulus are cropped using 3D computed tomography. With the 3D images of contrast-enhanced computed tomography reflecting the endocardial aspects of the atria, the endocardial structures, crista terminalis, and pectinate muscles, in particular, are recognizable. Of note, the endocardial aspects of an RAA demonstrate a rough surface due to pectinate muscles, while the body of an RA has a smooth surface. The marks of the crista terminalis and pectinate muscles from the endocardial aspect are shown by the arrows. The images are separated by colors: RA, SVC, and IVC (white); LA, LAA, and PVs (brown); and CS (yellow). AP view: anterior–posterior view, RL view: right lateral view, PA view: posterior–anterior view, RA: right atrium, RAA: right atrial appendage, LA: left atrium, LAA: left atrial appendage, LSPV: left superior pulmonary vein, LIPV: left inferior pulmonary vein, RSPV: right superior pulmonary vein, RIPV: right inferior pulmonary vein, SVC: superior vena cava, IVC: inferior vena cava, CS: coronary sinus, MA: mitral annulus, and TA: tricuspid annulus.
Figure 2. Anatomy of human atria in 3D computed tomography. The endocardial aspects of the atria and surrounding structures above the mitral and tricuspid annulus are cropped using 3D computed tomography. With the 3D images of contrast-enhanced computed tomography reflecting the endocardial aspects of the atria, the endocardial structures, crista terminalis, and pectinate muscles, in particular, are recognizable. Of note, the endocardial aspects of an RAA demonstrate a rough surface due to pectinate muscles, while the body of an RA has a smooth surface. The marks of the crista terminalis and pectinate muscles from the endocardial aspect are shown by the arrows. The images are separated by colors: RA, SVC, and IVC (white); LA, LAA, and PVs (brown); and CS (yellow). AP view: anterior–posterior view, RL view: right lateral view, PA view: posterior–anterior view, RA: right atrium, RAA: right atrial appendage, LA: left atrium, LAA: left atrial appendage, LSPV: left superior pulmonary vein, LIPV: left inferior pulmonary vein, RSPV: right superior pulmonary vein, RIPV: right inferior pulmonary vein, SVC: superior vena cava, IVC: inferior vena cava, CS: coronary sinus, MA: mitral annulus, and TA: tricuspid annulus.
Ijms 25 10193 g002
Figure 3. Increased heterogeneity with rate dependence in a diseased atrium. (A) Conduction velocity maps (during sinus rhythm) in a control and a diseased murine atrium with heart failure. (B) Increase in heterogeneity of conduction velocity dependent on pacing cycle length in a diseased atrium. The dotted arrow indicates an increase in the heterogeneity of conduction velocity. Taken from the reference [210] with partial modification.
Figure 3. Increased heterogeneity with rate dependence in a diseased atrium. (A) Conduction velocity maps (during sinus rhythm) in a control and a diseased murine atrium with heart failure. (B) Increase in heterogeneity of conduction velocity dependent on pacing cycle length in a diseased atrium. The dotted arrow indicates an increase in the heterogeneity of conduction velocity. Taken from the reference [210] with partial modification.
Ijms 25 10193 g003
Figure 4. Therapeutic approach of ablation or pharmacotherapy for the heterogeneity. (A) Isolation strategy by ablation. (B) Ablation strategy for the individual structural and electrophysiological features. (C) Pharmacotherapy to reduce the overall heterogeneity by inhibiting the progression of pathophysiology such as profibrotic process or connexin remodeling. RA: right atrium, LA: left atrium, PV: pulmonary vein, SVC: superior vena cava, and IVC: inferior vena cava.
Figure 4. Therapeutic approach of ablation or pharmacotherapy for the heterogeneity. (A) Isolation strategy by ablation. (B) Ablation strategy for the individual structural and electrophysiological features. (C) Pharmacotherapy to reduce the overall heterogeneity by inhibiting the progression of pathophysiology such as profibrotic process or connexin remodeling. RA: right atrium, LA: left atrium, PV: pulmonary vein, SVC: superior vena cava, and IVC: inferior vena cava.
Ijms 25 10193 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Iwamiya, S.; Ihara, K.; Nitta, G.; Sasano, T. Atrial Fibrillation and Underlying Structural and Electrophysiological Heterogeneity. Int. J. Mol. Sci. 2024, 25, 10193. https://doi.org/10.3390/ijms251810193

AMA Style

Iwamiya S, Ihara K, Nitta G, Sasano T. Atrial Fibrillation and Underlying Structural and Electrophysiological Heterogeneity. International Journal of Molecular Sciences. 2024; 25(18):10193. https://doi.org/10.3390/ijms251810193

Chicago/Turabian Style

Iwamiya, Satoshi, Kensuke Ihara, Giichi Nitta, and Tetsuo Sasano. 2024. "Atrial Fibrillation and Underlying Structural and Electrophysiological Heterogeneity" International Journal of Molecular Sciences 25, no. 18: 10193. https://doi.org/10.3390/ijms251810193

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop