Abstract

Aims

The concept of “atrial cardiomyopathy” (AtCM) had been percolating through the literature since its first mention in 1972. Since then, publications using the term were sporadic until the decision was made to convene an expert working group with representation from four multinational arrhythmia organizations to prepare a consensus document on atrial cardiomyopathy in 2016 (EHRA/HRS/APHRS/SOLAECE expert consensus on atrial cardiomyopathies: definition, characterization, and clinical implication). Subsequently, publications on AtCM have increased progressively.

Methods and results

The present consensus document elaborates the 2016 AtCM document further to implement a simple AtCM staging system (AtCM stages 1–3) by integrating biomarkers, atrial geometry, and electrophysiological changes. However, the proposed AtCM staging needs clinical validation. Importantly, it is clearly stated that the presence of AtCM might serve as a substrate for the development of atrial fibrillation (AF) and AF may accelerates AtCM substantially, but AtCM per se needs to be viewed as a separate entity.

Conclusion

Thus, the present document serves as a clinical consensus statement of the European Heart Rhythm Association (EHRA) of the ESC, the Heart Rhythm Society (HRS), the Asian Pacific Heart Rhythm Society (APHRS), and the Latin American Heart Rhythm Society (LAHRS) to contribute to the evolution of the AtCM concept.

Table of contents

  • 1. Background, definition and classification

  • 2. Important recent basic mechanistic insights

  •  2.1. Early-onset atrial fibrillation due to underlying genetic cardiomyopathy and arrhythmia syndromes

  •  2.2. Atrial amyloidosis

  •  2.3. Inflammatory signalling

  •  2.4. Gut microbiome

  •  2.5. Reactive lipid mediators of oxidative stress

  •  2.6. Atrial adipose tissue and fibro-fatty infiltration

  • 3. New findings in the pathophysiology of atrial cardiomyopathy

  •  3.1. Ageing and atrial cardiomyopathy

  •  3.2. Gender and atrial cardiomyopathy

  •  3.3. Atrial failure: definitions, causes, and consequences

  •  3.4. Atrial functional valvular regurgitation

  •  3.5. Metabolic disorders and atrial cardiomyopathy

  •  3.6. Atrial cardiomyopathy and atrial ablation

  •  3.7. The development of atrial fibrosis

  • 4. Imaging advances

  •  4.1. Atrial imaging tools in the evaluation of atrial cardiomyopathy

  •  4.2. Atrial size and function

  •  4.3. LA late gadolinium-enhanced cardiac magnetic resonance imaging

  •  4.4. Electroanatomic mapping

  •  4.5. Prediction of incident atrial fibrillation by cardiac imaging

  •  4.6. Recurrent atrial fibrillation

  •  4.7. Reverse atrial remodelling and atrial remodelling

  •  4.8. Risk stratification

  • 5. Biomarkers, omics, ECG parameters

  •  5.1. Biomarkers

  •  5.2. Blood-based biomarkers

  •  5.3. Limitations of biomarkers

  •  5.4. Multi-omics and atrial cardiomyopathy

  •  5.5. ECG parameters

  •  5.6. Artificial intelligence

  •  5.7. Future directions

  • 6. Stroke and cognitive decline

  •  6.1. Empiric evidence that atrial cardiomyopathy leads to stroke

  •  6.2. Mechanisms underlying the relationship between atrial cardiomyopathy and stroke

  •  6.3. Does atrial cardiomyopathy lead to cognitive decline?

  • 7. Therapeutic implications

  •  7.1. Primary prevention of atrial cardiomyopathy

  •  7.2. Lifestyle interventions to reverse atrial cardiomyopathy

  •  7.3. Pharmacological therapies to reduce progression of atrial cardiomyopathy

  •  7.4. Impact of rhythm control strategies on atrial cardiomyopathy

  •  7.5. Rhythm control and atrial cardiomyopathy

  •  7.6. Reversal of atrial cardiomyopathy by rhythm control?

  •  7.7. Device therapy to reverse atrial cardiomyopathy

  • 8. Conclusions and future directions

  • Supplementary material

  • Acknowledgements

  • Data availability

1. Background, definition and classification

The concept of ‘atrial cardiomyopathy’ (AtCM) had been percolating through the literature since its first mention in 1972.1 Since then, publications using the term were sporadic until the decision was made to convene an expert working group with representation from four multinational arrhythmia organizations to prepare a consensus document on atrial cardiomyopathy (EHRA/HRS/APHRS/SOLAECE expert consensus on atrial cardiomyopathies: definition, characterization, and clinical implication).2 Subsequent to the publication of the working group report,2–4 the number of publications on AtCM has increased progressively (Figure 1). In 2022, progress in the area motivated the decision to reconstitute the working group in order to take stock of a range of important developments. The present manuscript presents the results of that reflection.

The colour shading is pretty but could be confusing, if readers think the red colour designates something different from the blue. Consider keeping simple monochrome (which avoids this potential confusion) or else adding a note to the figure legend indicating that each vertical bar represents the number of publications each year, with the colours simply being decorative.2–4
Figure 1

The colour shading is pretty but could be confusing, if readers think the red colour designates something different from the blue. Consider keeping simple monochrome (which avoids this potential confusion) or else adding a note to the figure legend indicating that each vertical bar represents the number of publications each year, with the colours simply being decorative.2–4

In the first consensus document on AtCM, the working group recommended the following working definition of atrial cardiomyopathy: ‘Any complex of structural, architectural, contractile, or electrophysiological changes affecting the atria with the potential to produce clinically relevant manifestations’ (Table 1). This definition is of necessity broad, as AtCM is the basis for clinical manifestations of atrial arrhythmia (e.g. atrial fibrillation (AF) and atrial flutter), atrial thrombogenesis and clinically important functional–structural manifestations like atrial failure and atrio-ventricular valve dysfunction secondary to atrial dilation.

Table 1

Definition of atrial cardiomyopathy (AtCM)

‘Any complex of structural, architectural, contractile or electrophysiological changes affecting the atria with the potential to produce clinically relevant manifestations.’
‘Any complex of structural, architectural, contractile or electrophysiological changes affecting the atria with the potential to produce clinically relevant manifestations.’
Table 1

Definition of atrial cardiomyopathy (AtCM)

‘Any complex of structural, architectural, contractile or electrophysiological changes affecting the atria with the potential to produce clinically relevant manifestations.’
‘Any complex of structural, architectural, contractile or electrophysiological changes affecting the atria with the potential to produce clinically relevant manifestations.’

Accordingly, AtCM encompasses all functionally relevant complexes of atrial tissue changes, including those in contractility, electrophysiology and chamber architecture. Several systemic diseases or disease states can influence overall atrial function, causing pathophysiological changes and overt pathology. We also proposed a histological classification scheme for atrial cardiomyopathies in the consensus document, with the acronym EHRAS (for EHRA/HRS/APHRS/SOLAECE), defining four classes: I (principally cardiomyocyte changes); II (principally fibrotic changes); III (combined cardiomyocyte-pathology/fibrosis); IV primarily non-collagen infiltration (with or without cardiomyocyte changes). This simple classification might help to convey the primary underlying atrial pathology in various clinical conditions; however, it has not, to date, been widely accepted.5–7 A reason for this current situation might be that systematic analyses of atrial biopsies is not feasible in vivo, and therefore, histological validation of clinical findings/atrial imaging is rarely done.

Members of the working group considered that Table 2 might present a simple working system for staging AtCM by integrating biomarkers, atrial geometry, and electrophysiological changes to define different stages of AtCM. Importantly, AtCM has to be differentiated from AF, which might be a consequence of AtCM: AtCM commonly causes AF, however, AF may also accelerate AtCM. To concretize the suggested AtCM staging system, prospective trials are needed to validate the stages and cut-off values.

Table 2

Classification of different stages of atrial cardiomyopathies

1.Mild (subclinical) atrial cardiomyopathyDetected by electrophysiological measures or imaging technologies without overt arrythmia or significant mechanical atrial dysfunction
2. Moderate (clinically manifest) atrial cardiomyopathySignificant structural abnormalities or atrial mechanical dysfunction and elevated biomarkers (BNP, ANP, etc.) and /or manifestation of atrial fibrillation
3. Severe atrial cardiomyopathy:Atrial systolic failure (severely reduced left atrial ejection fraction ≤35%) associated with substantially impaired atrial contractility (flow velocities: ≤20 cm/sec within LAA/tissue strain), and/or major degrees (≥35% of LA wall volume) of interstitial alterations (atrial fibrosis, fatty infiltrates, amyloid infiltration, inflammation), and/or severe atrial enlargement (LA diameter ≥5.0 cm, LA volume index ≥50 mL/m2), and/or long-standing persistent/permanent atrial fibrillation
1.Mild (subclinical) atrial cardiomyopathyDetected by electrophysiological measures or imaging technologies without overt arrythmia or significant mechanical atrial dysfunction
2. Moderate (clinically manifest) atrial cardiomyopathySignificant structural abnormalities or atrial mechanical dysfunction and elevated biomarkers (BNP, ANP, etc.) and /or manifestation of atrial fibrillation
3. Severe atrial cardiomyopathy:Atrial systolic failure (severely reduced left atrial ejection fraction ≤35%) associated with substantially impaired atrial contractility (flow velocities: ≤20 cm/sec within LAA/tissue strain), and/or major degrees (≥35% of LA wall volume) of interstitial alterations (atrial fibrosis, fatty infiltrates, amyloid infiltration, inflammation), and/or severe atrial enlargement (LA diameter ≥5.0 cm, LA volume index ≥50 mL/m2), and/or long-standing persistent/permanent atrial fibrillation

Functional measurements (LAEF etc.) in sinus rhythm; validation of suggested AtCM stages and cut-off values need to be done in prospective trials.

LA, left atrium; LAA, left atrial appendage.

Table 2

Classification of different stages of atrial cardiomyopathies

1.Mild (subclinical) atrial cardiomyopathyDetected by electrophysiological measures or imaging technologies without overt arrythmia or significant mechanical atrial dysfunction
2. Moderate (clinically manifest) atrial cardiomyopathySignificant structural abnormalities or atrial mechanical dysfunction and elevated biomarkers (BNP, ANP, etc.) and /or manifestation of atrial fibrillation
3. Severe atrial cardiomyopathy:Atrial systolic failure (severely reduced left atrial ejection fraction ≤35%) associated with substantially impaired atrial contractility (flow velocities: ≤20 cm/sec within LAA/tissue strain), and/or major degrees (≥35% of LA wall volume) of interstitial alterations (atrial fibrosis, fatty infiltrates, amyloid infiltration, inflammation), and/or severe atrial enlargement (LA diameter ≥5.0 cm, LA volume index ≥50 mL/m2), and/or long-standing persistent/permanent atrial fibrillation
1.Mild (subclinical) atrial cardiomyopathyDetected by electrophysiological measures or imaging technologies without overt arrythmia or significant mechanical atrial dysfunction
2. Moderate (clinically manifest) atrial cardiomyopathySignificant structural abnormalities or atrial mechanical dysfunction and elevated biomarkers (BNP, ANP, etc.) and /or manifestation of atrial fibrillation
3. Severe atrial cardiomyopathy:Atrial systolic failure (severely reduced left atrial ejection fraction ≤35%) associated with substantially impaired atrial contractility (flow velocities: ≤20 cm/sec within LAA/tissue strain), and/or major degrees (≥35% of LA wall volume) of interstitial alterations (atrial fibrosis, fatty infiltrates, amyloid infiltration, inflammation), and/or severe atrial enlargement (LA diameter ≥5.0 cm, LA volume index ≥50 mL/m2), and/or long-standing persistent/permanent atrial fibrillation

Functional measurements (LAEF etc.) in sinus rhythm; validation of suggested AtCM stages and cut-off values need to be done in prospective trials.

LA, left atrium; LAA, left atrial appendage.

2. Important recent basic mechanistic insights

2.1. Early-onset atrial fibrillation due to underlying genetic cardiomyopathy and arrhythmia syndromes

Recent evidence indicates that AF developing in younger patients is often the initial manifestation of an atrial cardiomyopathy caused by rare variants in cardiomyopathy (CM) or arrhythmia syndrome genes.8 In one study, patients with AF-onset at ages ≤65 years underwent whole-exome sequencing for 145 genes commonly included on commercial cardiomyopathy and arrhythmia syndrome panels including dilated cardiomyopathy, hypertrophic cardiomyopathy, Brugada syndromes and other inherited arrhythmia syndromes.9 In a cohort of 1293 patients with a median age of 50 years, 10.1% had a disease-causing variant, with 62.8% carrying a variant of unknown significance (VUS), and 7.1% heterozygous for an autosomal recessive disease. Patients with AF-onset at <30 years of age are most likely to have a disease-associated variant (16.8%), with the most common genes being TTN, followed by MYH7, MYH6, LMNA, and KCNQ1. During a median follow-up of 9.9 years in the same cohort, the presence of a disease-causing variant was associated with a 50% increase in mortality that included 73 deaths related to cardiomyopathy, 40 sudden deaths, and 10 stroke-related deaths.10 Thus, patients with early-onset AF have a significant risk of a disease-associated variant, primarily in cardiomyopathy genes, with increased mortality, supporting a potential role for genetic testing in these patients. However, while there are data pointing towards the potential value of genetic testing for AtCM, no major society guidelines presently recommend such testing, and it is not covered by most health insurers.11 Further work in this promising area is certainly needed. A systematic multicenter registry of atrial-specific cardiomyopathies is a very-much needed first step.12

2.2. Atrial amyloidosis

Increasing evidence indicates that the prevalence of atrial amyloidosis is greater than previously suspected, particularly in the elderly.13–16 The prevalence of heart failure with a preserved ejection fraction (HFpEF) increases with ageing, and senile amyloidosis due to wild-type transthyretin (ATTRwt) accounts for up to 25% of HFpEF in patients >80 years of age, commonly with atrial involvement and AF.13,16 In a recent study of HFpEF patients aged >60 years with LV hypertrophy,99m Tc-DPD scintigraphy was highly suggestive of ATTRwt in 13% of patients.14 Isolated atrial amyloidosis (IAA) is a different entity mediated by atrial natriuretic peptide (ANP), and deposits containing both TTR and ANP have been detected in the same atria, indicating that both forms of senile atrial amyloidosis can co-exist.15 Although some forms of amyloid were previously considered organ specific [e.g. amyloid β (Aβ) in Alzheimer's disease], cardiac involvement with Aβ deposition and diastolic dysfunction has been identified in patients with Alzheimer's disease.17 Mechanistically, there is increasing evidence for the cardiotoxicity of soluble pre-amyloid oligomers. Light-chain variable domain proteins from cardiac light-chain amyloidosis (AL) patients are highly toxic to atrial HL-1 cardiomyocytes,18 while natriuretic peptide oligomers cause mitochondrial dysfunction, superoxide generation, and electrophysiological alterations in atrial cells.19

2.3. Inflammatory signalling

Recent findings showed that atrial NLRP3-inflammasome signalling creates AF-promoting AtCM in mouse models, and is increased in patients with common forms of AF including paroxysmal, persistent, and post-operative AF20,21 as well as in conditions that promote AF (obesity,22diabetes,22 gut dysbiosis,23 sepsis,24 and HFpEF25–27). Expression (‘priming’) and assembly (‘triggering’) of the NLRP3-inflammasome complex consisting of NLRP3, apoptosis-associated speck-like protein containing a C-terminal caspase activation and recruitment domain (ASC) and pro-caspase-1 accelerates auto-cleavage of pro-caspase-1 in atrial and immune cells. The resulting increase in active caspase-1 cleaves the inactive precursors of IL-1β and IL-18 to their active forms. In addition, caspase-1 degrades gasdermin-D, releasing the N-terminal fragment that forms plasma membrane pores, which allow IL-1β and IL-18 to leave the cells to exert autocrine and paracrine effects.25,26 Mice with cardiomyocyte-restricted constitutive activation of the NLRP3 inflammasome show increased atrial ectopy associated with abnormal Ca2+-handling due to increased RyR2 expression, as well as reentry-promoting APD shortening (electrical remodelling), likely due to upregulation of atrial-selective ultra-rapid delayed-rectifier (IKur) and acetylcholine-activated inward-rectifier (IK,ACh) K+-current, as well as structural remodelling involving hypertrophy and fibrosis.20 The activation of the NLRP3 inflammasome is likely mediated by increased activity of CaMKII-related pathways21 and impaired AMPK signalling.27 Gut microbiota-derived lipopolysaccharide may constitute a biologically relevant upstream activator of the atrial NLRP3 inflammasome in AtCM, particularly during ageing.23 Recent data have linked another inflammasome, the Absent-in-Melanoma 2 (AIM2) inflammasome, to AtCM, and AF susceptibility.28 This study showed that high-protein diets create AF-promoting AtCM by activating the AIM2-inflammasome, which is associated with mitochondrial oxidative stress along with proarrhythmic sarcoplasmic reticulum Ca2+-release events.28 These findings position the NLRP3 and AIM2 inflammasomes as potential novel drug targets for AF treatment.

Recent data point to an important role of recruited macrophages in the development of inflammatory signalling leading to AtCM and AF,29 with macrophage targets being of potential interest as novel AF therapeutic compounds.30 There is also evidence that genetic and epigenetic regulation of innate immunity may play a major role in conditioning AF susceptibility by controlling inflammatory responses.31 Active resolution of inflammation is emerging as a theme in combatting inflammation and preventing its chronicization.26 Pro-resolution mediators like resolvin D1 have shown effectiveness in prevention of AF substrate development in animal models of both right32 and left heart disease.32,33 Nevertheless, specific differences between various types of inflammation, like sterile inflammatory processes or viral/bacterial infections, cannot be made at present.

2.4. Gut microbiome

The gut microbiota is a dynamic system containing microorganisms that produce bioactive metabolites, the type and quality of which depend on lifestyle-related factors (e.g. diet, exercise), among others. This microbiota contributes to the pathophysiology of many cardiovascular conditions like heart failure, hypertension, obesity, and coronary artery disease, and there is accumulating evidence for a role of gut microbiota in AtCM and related arrhythmogenesis.34 Although many gut microbiota-derived metabolites could play a role in atrial arrhythmogenesis,34 recent evidence suggests that gut dysbiosis may promote AtCM and AF partly through increased levels of circulating lipopolysaccharide and glucose, along with enhanced activity of the atrial NLRP3 inflammasome, which causes atrial fibrosis.23 As a proof of causality, faecal microbiota transplantation (FMT) from young (2–3 months) to old (22–24 months) rats reduced atrial fibrosis and the age-related increase in AF susceptibility by attenuating atrial NLRP3-inflammasome activity. Selective inhibition of both lipopolysaccharide and the NLRP3 inflammasome also reduced AF susceptibility and atrial fibrosis, suggesting lipopolysaccharide as a key upstream driver of the atrial NLRP3 inflammasome and the associated arrhythmogenesis.25,26 Although gut microbiota could create inflammatory AtCM, the precise cellular and molecular mechanisms, as well as the clinical contexts in which this system is active in AtCM, require further investigation.

2.5. Reactive lipid mediators of oxidative stress

During inflammation and oxidative stress, lipid peroxidation generates dicarbonyl compounds that adduct to proteins, DNA, and other lipids to promote molecular dysfunction and amyloid formation.35 The most reactive dicarbonyls are isolevuglandins (IsoLGs),36 and small molecule scavengers that selectively inactivate these and other lipid dicarbonyls are beneficial in multiple preclinical studies, indicating that IsoLGs are a major component of oxidative stress-related injury.37 In a murine model of hypertension, IsoLG protein adducts and preamyloid oligomers (PAOs) developed in the atria prior to structural or histologic abnormalities coincident with AF susceptibility. These abnormalities and arrhythmia vulnerability were prevented by administration of a dicarbonyl scavenger 2-hydroxybenzylamine (2-HOBA), but not by an inactive analog 4-hydroxybenzylamine (4-HOBA). These findings support the concept of pre-emptively scavenging reactive downstream mediators, rather than targeting generation of reactive oxidative species per se, as a potential therapeutic approach to prevent AtCM. Preclinical and Phase I studies of 2-HOBA have demonstrated its safety and tolerability, and clinical trials in humans are in progress.38,39

2.6. Atrial adipose tissue and fibro-fatty infiltration

Altered metabolism and increased abundance of epicardial adipose tissue (EAT) in the atria is associated with a higher risk of AF.40–42 EAT43–47 could contribute to the formation of the AF substrate by several mechanisms. First, EAT continuously secrete fatty acids that fuel the myocardium, together with a variety of cytokines and peptides, as well as extracellular vesicles.48,49 Some of these secreted molecules, like TGFα, KIL-1B, and FAPB4, can regulate myocardial electrophysiology, others like adiponectin regulate the oxidative stress of the atrial myocardium.50–52 Furthermore, during AF, heart failure or in diabetic patients, EAT secretes profibrotic cytokines such as activin-A and metalloproteases that contribute to the spreading of fibrosis from the epicardial layer to the neighbouring subepicardial myocardium.53–55 Clusters of mononuclear inflammatory cells including cytotoxic CD8+ T-lymphocytes are often observed at the junction between subepicardial fat and fibrotic tissue, resembling lymphocytic myocarditis of patients with AtCM.56,57 Furthermore, the secretome of abnormal EAT contains several inflammatory chemokines. Therefore, EAT is an important potential contributor to atrial inflammation.58,59

The replacement of the EAT beneath the epicardial layer by fibrosis causes fibro-fatty infiltration of the subepicardial atrial layers in AF patients and in aged patients with heart failure or mitral regurgitation.55,58,60 It is established that subepicardial fibro-fatty infiltration is a major arrhythmogenic substrate in both the ventricles and the atria, favouring epicardial–endocardial asynchrony with local conduction block, breakthroughs, and rotor formation.61–63 Moreover, 3D computational modelling of human atria integrating MR images and histo-anatomical data suggests that there is a relationship between the slowing of electrical conduction and the degree of fibro-fatty infiltration.64

The precise mechanisms regulating the accumulation and shaping of the biological properties of EAT are poorly understood. EAT originates mainly from epicardial progenitor cells (EPC). EPC are normally quiescent in the adult heart, but can be reactivated to differentiate into various lineages, including adipocytes.65,66 In human atrial myocardium, subsets of cells derived from EPC (EPDC) are confined to the sub-epicardium where they can differentiate into adipocytes or fibroblasts in response to local stimuli. For instance, the ANP secreted by stretched atrial cardiomyocytes induces, at low concentrations, the adipogenic differentiation of EPDC by activating cGMP-dependent signalling pathways.67 Angiotensin-II produced by the atria can also induce the differentiation of EPDC into fibroblasts, primarily via activation of canonical SMAD2/3, and p38-MAPK signalling pathways.68 Adipogenic differentiation of EPDC is related to lipid metabolism and the capacity of EAT to store fat, for example when submitted to an excess dietary fatty acid load.69 The multiple mechanisms causing the accumulation of EAT likely contribute to the formation of an arrhythmogenic AF substrate over time.

In conclusion, the formation of EAT is a complex interplay between metabolism, inflammation, and oxidative stress, promoting AtCM and AF by local and systemic mechanisms, and may offer new opportunities for management of AtCM/AF.

3. New findings in the pathophysiology of atrial cardiomyopathy

Many animal model and human studies have revealed a close interplay among AtCM, AF, and stroke, via various mechanisms such as ageing, inflammation, oxidative stress and stretch, which, in turn, lead to fibrosis, electrical, and autonomic remodelling and a pro-thrombotic state.70,71 The complex interplay between the mechanisms creates a feedback loop of ever-worsening AtCM and higher risk of more sustained AF and strokes.70 However, there is often a lack of a clear temporal relationship between the onset of AF and stroke. In fact, the current paradigm of selecting individuals at elevated risk of stroke and therefore warranting oral anticoagulation (OAC) therapy, endorsed by major international societies,71–73 by CHA2DS2-VASc (including congestive heart failure, hypertension, age, diabetes mellitus, prior stroke/transient ischaemic attack/thromboembolism, vascular disease) or related scores, are all variables related to the risk of AtCM but not the properties of AF per se (frequency, duration, ventricular rates, etc.). This observation suggests that additional factors inherent in AtCM may be important contributors to stroke occurrence and that the presence of AF per se may not be necessary.74,75 Of note, the recently published Apixaban to Prevent Recurrence After Cryptogenic Stroke in Patients With Atrial Cardiopathy (ARCADIA) trial failed to observe benefit from the oral anticoagulant apixaban in patients with cryptogenic stroke and biomarker evidence of AtCM.76

3.1. Ageing and atrial cardiomyopathy

One major factor leading to AtCM may be ageing, which leads to progressive decline in the structure and function of the heart and is a leading risk factor for cardiovascular diseases. There is strong epidemiological evidence for a close link between ageing, AF, and stroke. The prevalence of many AF and stroke risk factors, including diabetes, hypertension, and coronary/peripheral atherosclerotic vascular disease, steadily increases with age. All these risk factors contribute to the development of atrial cardiomyopathy, and there is mounting evidence supporting the concept that AtCM not only leads to stasis, but endothelial/endocardial dysfunction and a hypercoagulable state, three key factors in thrombogenesis, as originally described by Virchow.77

Tissue remodelling, characterized by myocyte loss, reactive cellular hypertrophy, fibrosis and autonomic nervous system dysregulation, is part of the pathophysiology of AtCM, and a possible common denominator of ageing, stroke, and other AF risk factors (Figures 2 and 3). On the other hand, recent evidence indicates that AF is a major accelerator of ageing of the atrial myocardium (Table 3). At the molecular level, the association between AtCM and ageing (Figure 4) appears to be mediated by oxidative stress,79 calcium handling dysregulation,80 electrophysiologic changes, and structural remodelling with apoptosis and fibrosis,70 all of which contribute to the initiation and/or maintenance of AF, but the mechanisms have been inadequately explored.81 In fact, it is unknown why some ageing individuals with AtCM develop AF while others do not. Chronic inflammation is associated with several age-related diseases such as atherosclerosis, Alzheimer's disease, sarcopenia, and arthritis.82 The genesis of chronic inflammation with ageing (so-called ‘inflammaging’) is unclear but inflammation could be an underlying mechanism connecting ageing to AtCM and AF.83 Furthermore, several markers of cellular ageing, such as telomere length, SA-β-gal, and p16a are associated with AF.81 DNA damage, a mechanism underlying cell senescence, has been shown in tachypaced atrial myocytes and patients in persistent AF.84,85 Clearly, a comprehensive understanding of the molecular mechanisms of intrinsic cardiac ageing, including atrial ageing, will be required to improve understanding of the relation between ageing, inflammation, AtCM, and AF. Such new understanding should guide the development and future translation of novel therapies to clinical application.

Age-related changes in atrial tissue structure. Commonly found age-related changes in atrial tissue structure (Masson's trichrome staining) which represent the structural bases of atrial ageing. yo, years old.
Figure 2

Age-related changes in atrial tissue structure. Commonly found age-related changes in atrial tissue structure (Masson's trichrome staining) which represent the structural bases of atrial ageing. yo, years old.

(A) Light microscopy image (Masson's trichrome stain) of atrial tissue with normal intercellular space (41 yo) and (B) cardiomyocyte loss with expansion of interstitial fibrous tissue (82 yo). (C) and (D): myofibrillar loss and fibrotic replacement in the aged left atrium. yo, years old.
Figure 3

(A) Light microscopy image (Masson's trichrome stain) of atrial tissue with normal intercellular space (41 yo) and (B) cardiomyocyte loss with expansion of interstitial fibrous tissue (82 yo). (C) and (D): myofibrillar loss and fibrotic replacement in the aged left atrium. yo, years old.

Histological factor associated with atrial structural remodelling characterized by electroanatomic mapping. The upper panel shows light microscopy images of atrial biopsy samples obtained from patients with non-valvular atrial fibrillation. The lower panel shows examples of high-density bipolar voltage map of the left atrium created during high right atrial pacing at 100 beats per minute. Not only fibrosis, but also increased intercellular space preceding fibrosis, myofibrillar loss, decrease in myocardial nuclear density, and amyloid deposition are associated with the progression of atrial structural remodelling.78 The rightmost image in the upper panel shows Congo Red staining, the second from the right shows haematoxylin and eosin staining, and the others show Masson's trichrome staining. The colour gradation in the lower panel voltage maps indicates differences in voltage amplitude from purple at 1.5 mV to grey at 0.1 mV, as shown in the colour bar. AtCM, atrial cardiomyopathy (see Table 2 for explanation of AtCM stages).
Figure 4

Histological factor associated with atrial structural remodelling characterized by electroanatomic mapping. The upper panel shows light microscopy images of atrial biopsy samples obtained from patients with non-valvular atrial fibrillation. The lower panel shows examples of high-density bipolar voltage map of the left atrium created during high right atrial pacing at 100 beats per minute. Not only fibrosis, but also increased intercellular space preceding fibrosis, myofibrillar loss, decrease in myocardial nuclear density, and amyloid deposition are associated with the progression of atrial structural remodelling.78 The rightmost image in the upper panel shows Congo Red staining, the second from the right shows haematoxylin and eosin staining, and the others show Masson's trichrome staining. The colour gradation in the lower panel voltage maps indicates differences in voltage amplitude from purple at 1.5 mV to grey at 0.1 mV, as shown in the colour bar. AtCM, atrial cardiomyopathy (see Table 2 for explanation of AtCM stages).

Table 3

Age-related changes in atrial tissue structure

1. Age-dependent cardiomyocyte loss and myocyte hypertrophy
2. Interstitial changes and increase in intercellular space
3. Enhancement of the interstitial adipose and fibrosis tissue
3. Endocardial fibrosis
4. Inflammatory infiltrates
6. Vacuolar degeneration and Aschoff bodies
8. Iron depositions
9. Amyloidosis
1. Age-dependent cardiomyocyte loss and myocyte hypertrophy
2. Interstitial changes and increase in intercellular space
3. Enhancement of the interstitial adipose and fibrosis tissue
3. Endocardial fibrosis
4. Inflammatory infiltrates
6. Vacuolar degeneration and Aschoff bodies
8. Iron depositions
9. Amyloidosis
Table 3

Age-related changes in atrial tissue structure

1. Age-dependent cardiomyocyte loss and myocyte hypertrophy
2. Interstitial changes and increase in intercellular space
3. Enhancement of the interstitial adipose and fibrosis tissue
3. Endocardial fibrosis
4. Inflammatory infiltrates
6. Vacuolar degeneration and Aschoff bodies
8. Iron depositions
9. Amyloidosis
1. Age-dependent cardiomyocyte loss and myocyte hypertrophy
2. Interstitial changes and increase in intercellular space
3. Enhancement of the interstitial adipose and fibrosis tissue
3. Endocardial fibrosis
4. Inflammatory infiltrates
6. Vacuolar degeneration and Aschoff bodies
8. Iron depositions
9. Amyloidosis

3.2. Gender and atrial cardiomyopathy

Based on epidemiologic investigations, women seem less vulnerable to AF development than men,86 with the prevalence-discrepancy between genders becoming less clear with advancing age, in part perhaps due to the higher life expectancy in women or increased amount of epicardial fat.86–88 A substantial reason for the lower prevalence of AF in women vs. men appears to be their smaller size, since body size (and particularly lean body mass) is a major determinant of AF risk.89 Furthermore, compared to men, women suffering from AF appear to be exposed to significantly greater risks of all-cause and cardiovascular death,90 as well as reporting graver symptoms and a worse quality of life, all of which suggest more serious AF-related structural/functional impairment.91

Recent studies on AF ablation have also shown significant gender-based differences between males and females, with the latter showing a higher risk of arrhythmia recurrence92,93 and increased periprocedural complications and hospitalization rates.94 In addition, a recent large systematic review and meta-analysis of post-ablation outcomes found that, independently of age, comorbidities, and hospital factors, women display higher rates of complications and readmissions following AF ablation.95 In 2022, Wong et al. explored whether gender-based electrophysiological substrate differences might explain the aforementioned worse AF ablation outcomes in women. Intriguingly, they found that females are characterized by significantly more conduction velocity slowing, a greater proportion of complex fractionated signals, and at 22-month follow-up, notably poorer single-procedure and multi-procedure arrhythmia-free survival. These results strongly suggest more advanced atrial remodelling in women.96 Clinical studies have suggested that oestrogen deficiency or oestrogen–progestin imbalance (as that in the luteal phase of the menstrual cycle, during oral contraceptive treatment, during pregnancy, and menopause) may be associated with an increased occurrence of cardiac arrhythmias. The RETAC-group, in a recent study involving 220 AF patients that underwent high-density voltage mapping in SR prior to their first PVI procedure, found that women had greater low-voltage LA voltage-mapping scores than men, with this propensity driven by older women (>60 years) with persistent AF while absent in younger women.97 Moreover, it has already been demonstrated that administration of oestrogen, whose receptors are significantly expressed in cardiomyocytes, is linked with prolonged action potential duration, atrial and atrio-ventricular nodal conduction time, as well as atrial refractoriness, with these functional abnormalities likely due to increased calcium influx leading to weaker repolarizing currents or potassium channel downregulation.98,99 Moreover, prior investigations had shown that, compared to men, and in individuals with or without AF, women possess greater amounts of atrial fibrosis both in histopathological and cardiac magnetic resonance (CMR) studies.100,101

3.3. Atrial failure: definitions, causes, and consequences

Over the past few years, various definitions of atrial failure have been proposed, which, in turn, have been based on functional, morphological, or morpho/functional approaches.102–104 Generally, all of these are based on the concept of ‘atrial cardiomyopathy’ proposed by the original EHRA/HRS/APHRS/SOLAECE expert consensus document.2 Based on the AtCM staging proposed in Table 2, atrial failure would primarily feature in stage 3 AtCM (Figure 3).

The causes of atrial failure are multiple: (i) Atrial rhythm disorders. Rapid atrial arrhythmias—such as AF—induce ineffective active muscle contraction, with both systolic and diastolic LV dysfunction. A similar LV impairment may also be a consequence of suboptimal electrical AV coupling [e.g. due to long PR interval, left bundle branch block, advanced interatrial block (IAB)]102; (ii) Atrial myocardial structural remodelling. Morphological changes at the histological and ultrastructural levels (cardiomyocyte and/or interstitial modifications), often as a consequence of associated cardiac or non-cardiac diseases, along with AF in a vicious cycle (‘AF begets AF’); (iii) Atrial ischaemia, myocarditis, and induced post-ablation stiff LA syndrome. These can lead to increased atrial stiffness, reduced contractility, and electromechanical dissociation.2,105–108 As for the consequences of atrial failure, heart failure, increased thrombogenicity, and pulmonary hypertension may be caused by altered flow dynamics and suboptimal LV filling; AF may also result from atrial failure and contribute to the consequences.102,109 Recent investigations have shown that both LA dysfunction and remodelling are independently associated with, or may even herald, the onset of HF in an asymptomatic healthy population, in which reduced atrial reserve during exercise can indicate the first evidence of a failing LA.110,111 In addition to these pathogenetic mechanisms, atrial electrical dysfunction—and impaired LA–LV coupling—could play a key part in patients suffering from HFpEF.110,112 By the same token, atrial failure may reveal previously latent LV dysfunction and precipitate a HF syndrome. According to the Frank–Starling law, in the initial stages of LV dysfunction, adaptive atrial chamber dilation develops to counteract higher preload, while an increase in active pump function contributes to preserving adequate LV filling. Further impairment in LA function does nothing to mitigate the above-mentioned disproportionate atrial volume/pressure, leading to higher LA and pulmonary-wedge pressures and manifest atrial failure syndrome, as well as perpetuating a harmful vicious cycle.102

Recent evidence suggests a relationship between atrial disease and the risk of stroke independent of AF.74,75 The Asymptomatic Atrial Fibrillation and Stroke Evaluation in Pacemaker Patients and the Atrial Fibrillation Reduction Atrial Pacing Trial (ASSERT) followed 2580 patients aged ≥65 years with a pacemaker and defibrillator and a history of hypertension but without a history of AF. The authors found that only 15% of patients with subclinical atrial fibrillation (SCAF)-associated embolic events had evidence of SCAF >6 min in duration during the month before their stroke or systemic embolism. In addition, most SCAF that had occurred before embolic events was far shorter than 48 h (the minimum duration which, in the past, was believed to be required for thrombus formation in the LAA before cardioversion).113,114 Based on the results of this trial, it appears that some thromboembolic events may be secondary to stasis from an actual AF episode, some to persistent endothelial changes (e.g. due to previous AF episodes), and, lastly, some to mechanisms not directly related to AF, in which the supraventricular arrhythmia may simply represent a risk marker or, if causal, related via complex indirect pathogenetic mechanisms within the context of AF-related AtCM.115 Two additional clinical studies (TRENDS and IMPACT), have found similar results to those of the ASSERT study on the lack of temporal correlation between AF episodes and cerebrovascular events.74,75 Interestingly, analysis of data from the Multi-Ethnic Study of Atherosclerosis (MESA) displayed an inverse association between LA reservoir function (as total LA emptying fractions), and the risk of cerebrovascular events, with this association being independent of acknowledged cerebrovascular risk factors and AF itself.102 These findings suggest value of LA functional parameters in the stratification of individuals at risk for ischaemic cerebrovascular events, particularly in those with no clear evidence of AF.111 In addition to the risk of thrombosis in AF, the amount of atrial fibrosis in patients with stroke of unknown aetiology seems to be greater than in patients with an identified cause and comparable to those suffering from AF.102,116–118 Endothelial changes and regional/global wall motion abnormalities associated with increased fibrosis have been proposed as potential risk factors for atrial thrombosis and cerebrovascular events.102,116,119 Müller-Edenborn et al., in a large cohort of AF patients undergoing TEE prior to electrical cardioversion for AF, found that in individuals with LAA thrombi, 97% of them had a p-wave-based diagnosis of AtCM, which was associated with reduced LAA-flow velocity at TEE. This study adds further evidence that AtCM is associated with both prolonged atrial conduction time and reduced contractile function with reduction in LAA-flow velocity, especially in those who develop LAA thrombi.120 The observation of the ARCADIA trial of no benefit from apixaban in patients with cryptogenic stroke and biomarker evidence of AtCM argues against the idea that AtCM without clinical AF increases the risk of stroke appreciably, but further consideration of the results and design of the trial is needed before any definite conclusions can be made.76

The rhythm and contraction of the atria contribute to efficient coronary blood flow. Controlling for other known risk factors, patients with AF have a three-fold increased risk of myocardial infarction,121 possibly because irregular, shortened diastole with altered flow reserve is a common cause of ventricular ischaemia and a contributor to type 2 myocardial infarction.122,123 Additional mechanisms involved in the pathogenesis of atrial ischaemia/infarction in individuals with atrial myopathy/AF include increased inflammation/oxidative stress, altered platelet activation, and particularly atrial thromboembolism, which can occur even in the absence of AF.111,124

Atrial dysfunction secondary to supraventricular arrhythmias involves activation of neurohormonal pathways, (e.g. RAA system and vasoactive peptides) and can induce ventricular chamber dilation with altered systolic and diastolic cardiac function. In this scenario, the LV myocardium shows inflammation, cardiomyocyte histopathological changes, and loss of normal myocardial extracellular matrix (ECM) structure, composition, and function.125

3.4. Atrial functional valvular regurgitation

Atrial functional mitral and tricuspid regurgitation are clinicopathological conditions, which are closely related to atrial failure and atrial dilatation.126–129 Secondary or functional mitral and tricuspid regurgitation (FMR; FTR) is defined as valvular regurgitation caused by abnormal left/right heart function as opposed to primary structural valve changes.130 While FMR/FTR is most often the result of abnormalities in left/right ventricular remodelling, more recently recognized is the entity of atrial FMR (AFMR/AFTR). In this scenario, ventricular geometry and function are relatively preserved, the culprit being mitral/tricuspid annular enlargement associated with left/right atrial dilatation, which results in poor coaptation of the valve leaflets causing AFMR and AFTR. Compared to ventricular FMR, the entity of AFMR/AFTR, including its prevalence, pathophysiology, prognostic implication, and potential therapy, is less clear.126,127,129 Variability in the literature as to the definition of AFMR is a contributing factor. Recently, an expert opinion paper has been communicated that presents a unifying definition of AFMR.131 The major criteria for AFMR include normal LV cavity size and systolic function (global and regional), and the presence of both mitral annular dilation and LA enlargement. The LV may dilate secondarily in later stages of significant AFMR. AFMR is primarily rooted in excessive LA and mitral annular dilation which pulls both leaflets apart and thereby causes leaflet tethering and malcoaptation.132,133 Generally, two common clinical scenarios may lead to excessive LA and subsequent mitral annular dilation in the context of preserved LV function: AF and/or HFpEF.132 Both syndromes are linked by LA remodelling and dysfunction. AF is known to be both cause and consequence of atrial remodelling.102 In addition, chronically elevated filling pressures in HFpEF may induce adverse anatomical and functional LA remodelling. Furthermore, AF and HFpEF are strongly inter-related as they share common cardiovascular risk factors and potentiate each other. In patients with AF, restoration of sinus rhythm is associated with reduction in LA volumes, mitral annular size, and the degree of MR.134,135 Although this observation suggests that rhythm rather than rate control in AF might mitigate AFMR, this idea has not been formally tested.

Recent data point to a high prevalence of occult HFpEF among patients with AF and dyspnoea, especially in those who remain symptomatic after sinus rhythm restoration.136,137 This observation has led several groups to discern an AF-predominant HFpEF subtype with more advanced LA myopathy, disproportionate to the degree of LV dysfunction.137–139 The occurrence of AFMR in HFpEF is another marker of this subtype of advanced AtCM, leading to worse haemodynamics and poorer exercise reserve.134 A recent community study of moderate-to-severe isolated MR found that only 50% of patients with AFMR had a history of AF.135 Even when mild-to-moderate, the presence of AFMR tended to be associated with worse outcomes, though this association faded after correcting for LA cardiomyopathy. While this latter finding may suggest that the degree of AtCM is of greater importance in prognostic significance compared to AFMR per se, it does not refute the concept that treating AFMR in HFpEF could be beneficial by improving AtCM and thereby outcomes. In addition, recent cross-sectional data cannot disentangle the sequence of causality between AFMR, LA cardiomyopathy, and HFpEF. Presumably, a bidirectional relationship exists between AFMR and AtCM.134 Future prospective studies are needed to answer these questions.

3.5. Metabolic disorders and atrial cardiomyopathy

Metabolic disorders are the most common modifiable risk factors for AtCM and AF.43–47 This relationship is well established for obesity, diabetes, and metabolic syndrome, whereas it is still debated for liver steatosis-associated diseases.140,141 In the Framingham heart study, diabetes is associated with 40% increased risk of AF, and the risk increases with the duration of AF.142,143 High BMI is also associated with a higher incidence of AF.144

Metabolic diseases can directly impact the progression of AF substrate and AtCM through several mechanisms and signalling pathways.43,45 For instance, diabetes and glucose intolerance are characterized by inefficient energy production and metabolic inflexibility of the atrial myocardium with predominance of fatty acid beta-oxidation, mitochondrial dysfunction, and ROS production.145 This metabolic remodelling has a marked impact on atrial excitability and contractile properties, for example, explaining the decreased peak longitudinal strain function and left atrial deformation reported in diabetes mellitus patients.111 The excitation–contraction coupling process of cardiomyocytes is tightly controlled by cellular metabolism. Abnormal activity of several calcium and potassium channels of atrial cardiomyocytes have been reported in experimental models of diabetes.146 K-ATP channels that are normally closed and preferentially coupled to glycolysis over beta-oxidation lipolysis, are open in atrial myocytes of dysmetabolic mice, contributing to action potential shortening.69

An excess of dietary fatty acids can induce an adaptive metabolic signature of the atrial myocardium characterized by impaired glycolysis and predominance of fatty acid oxidation.67 Furthermore, when the uptake of lipid by atrial cardiomyocytes overwhelms their beta-oxidation capacity, a process of lipid storage ensues, activated through elongation of fatty acids, recruitment of adipogenic transcription factors, and adipogenic differentiation of progenitor cells.69 Altogether, these phenomena result in accumulation of intramyocardial lipid and expansion of EAT,147 whose role in the progression of AtCM and risk of AF is detailed elsewhere in this article. However, it must be noted here that metabolic disorders have specific impacts on EAT properties. For instance, in obese or diabetic patients, EAT secretes inflammatory cytokines which favour myocardial fibrosis and contribute to altered excitability.148 By analogy with what is known for ventricle remodelling or atherosclerotic plaque, another potential interface between metabolic disorders and myocardial inflammation are macrophages.149,150 Clinical studies are warranted to investigate the impact of improving metabolic status of patients on the risk of AF and AtCM.

3.6. Atrial cardiomyopathy and atrial ablation

Structural remodelling, including histological and anatomical-structural changes, contributes to the trigger and maintenance mechanisms of AF151 (Figure 4). Conversely, long-lasting AF itself also contributes to the progression of AF, leading to AtCM.152 The most effective way to interrupt this vicious cycle between AF maintenance and AtCM is to restore sinus rhythm. AF ablation is the most effective rhythm control strategy; it significantly reduces AF burden and prevents the progression of AtCM.153,154 Using bipolar voltage mapping and MRI, Sugumar et al.155 reported that rhythm control by AF ablation induces recovery of atrial electrical and structural changes, suggesting that AF ablation may result in some reversal of AF-induced AtCM (see Section 7).

3.7. The development of atrial fibrosis

Fibrosis is a central, causally related pathophysiological component of AtCM.156,157 It is now well established that the extent of LA interstitial fibrosis roughly parallels the biological/clinical history of AF; i.e. from early structural remodelling in most sinus rhythm patients, to paroxysmal, persistent, and, ultimately, long-standing lasting persistent AF.157,158 Additionally, interstitial fibrosis is often a progressive multi-step event in anatomical/architectural terms, with, in most cases, the pulmonary vein outlet area being the predominant initial location, with subsequent progression extending the pathology to the rest of the LA.105,159,160

The above-mentioned pathogenic peculiarities have raised various interesting questions, such as (i) Why is the pulmonary vein area—and, in a broader sense, the left atrial posterior wall—the area most prone to fibrosis (and structural remodelling in general)? (ii) Assuming that interstitial collagen deposition occurs through autocrine/paracrine mechanisms, what are the key cellular sources and cell-networks involved? (iii) Is collagen deposition in the atria a ‘local affair’, i.e. are atrial mesenchymal cell-components the only players in this process? Do any extra-cardiac mesenchymal components cooperate in the development of interstitial structural remodelling?

There are several possible explanations for why fibrosis begins in the LA posterior wall: (i) According to Laplace's law, the left atrial posterior wall is prone to greater stress than the left atrial appendage or other LA free walls. This is because the LA posterior wall is the thinnest part of the LA, blood pressure is similar throughout the LA and LA appendage, and the LA chamber volume is greater than that of the LA appendage; (ii) Anatomically, the LA posterior wall is close to the pulmonary vein orifices, a region subject to intense and continuous mechanical stretching; (iii) Embryologically, the LA appendage develops from the embryonic LA, whereas the LA proper originates from an outgrowth of the pulmonary vein.105,159 It has also recently been suggested that, in AF patients, atrial fibrosis (assessed by magnetic resonance imaging (MRI) with late gadolinium enhancement) might be more extensive in the area adjacent to the descending thoracic aorta, with its extent inversely correlated with the distance between LA and the aorta.161,162 A recent study explored fibrotic remodelling in LA biopsies from a series of patients with and without AF.158 In the vast majority of cases, collagen fibre deposition begins unevenly throughout the atrial myocardium, with separate fronts of fibrosis progressing from both the adventitial/periadventitial tissues and the thickened sub endocardium into the inter-cardiomyocyte interstitium. This tendency might be explained by the fact that both the adventitia and sub endocardium are already richer in collagen-producing cells under normal conditions. Subsequently, fibrosis becomes increasingly widespread and more homogeneous by gradually filling in the preserved myocardial areas. On these grounds, fibrosis stages might be defined as follows: early, presence of sparse single myocardiosclerotic foci originating from the sub endocardium or the periadventitial tissues; intermediate, formation of multiple single myocardiosclerotic foci originating from the subendocardium or the periadventitial tissues; advanced, generation of confluent myocardiosclerotic foci; and very advanced, development of areas of diffuse fibrosis (Figure 5).163 In agreement with this concept, elevated circulating fibrocytes have been proposed as a marker of left atrial fibrosis (in low-voltage regions) and AF recurrence.164,165

Microphotographs captured from left atrial biopsies belonging to patients suffering from long-lasting atrial fibrillation associated with severe mitral-valve regurgitation. These histopathological sections have been stained with a combined Weigert/Van Gieson dye where cardiomyocytes are highlighted in yellow-brown, collagen fibres in red, and elastic fibres in black. (A) Medium-power view of a developing Early-Stage histopathological remodelling (ATCM stage 1) where multiple foci of interstitial fibrosis (arrowheads) stem from the adventitia of a penetrating coronary branch (asterisk). (B) Low-power view of an incipient Early-Stage histopathological remodelling with multiple interstitial fibrosis foci (arrowheads) originating from a longitudinally oriented blood vessel (asterisks). (C) An initial Early-Stage AtCM histopathological remodelling with a fibroelastotic endocardial thickening (asterisk) from which newly deposed collagen fibres enlarge the adventitia of subendocardial blood vessels (arrowheads). (D) Incipient Early-Stage histopathological remodelling interstitial fibrosis (arrowheads) stemming from an endocardium thickened by fibroelastosis which had also enlarged local blood vessel adventitia (asterisk). (E) Early-Stage histopathological remodelling characterized by multiple foci of interstitial fibrosis (arrowheads) that had originated from sparse penetrating blood vessel adventitias (asterisks). (F) Another type of Intermediate-Stage histopathological remodelling (AtCM stage 2) compound interstitial fibrosis (arrowheads) which takes origin from two blood vessels and a thickened endocardium (asterisks). (G) Advanced-Stage histopathological remodelling characterized by confluent myocardiosclerotic foci (asterisks). H. Compact and diffuse areas of interstitial fibrosis (Very Advanced-Stage histopathological remodelling; AtCM stage 3). Staining. (A-G) Weigert/Van Gieson combined dye. Original magnifications. (A): ×20 (scale bar is 150μm); (B-G): ×10 (scale bar in (B) is 300μm).
Figure 5

Microphotographs captured from left atrial biopsies belonging to patients suffering from long-lasting atrial fibrillation associated with severe mitral-valve regurgitation. These histopathological sections have been stained with a combined Weigert/Van Gieson dye where cardiomyocytes are highlighted in yellow-brown, collagen fibres in red, and elastic fibres in black. (A) Medium-power view of a developing Early-Stage histopathological remodelling (ATCM stage 1) where multiple foci of interstitial fibrosis (arrowheads) stem from the adventitia of a penetrating coronary branch (asterisk). (B) Low-power view of an incipient Early-Stage histopathological remodelling with multiple interstitial fibrosis foci (arrowheads) originating from a longitudinally oriented blood vessel (asterisks). (C) An initial Early-Stage AtCM histopathological remodelling with a fibroelastotic endocardial thickening (asterisk) from which newly deposed collagen fibres enlarge the adventitia of subendocardial blood vessels (arrowheads). (D) Incipient Early-Stage histopathological remodelling interstitial fibrosis (arrowheads) stemming from an endocardium thickened by fibroelastosis which had also enlarged local blood vessel adventitia (asterisk). (E) Early-Stage histopathological remodelling characterized by multiple foci of interstitial fibrosis (arrowheads) that had originated from sparse penetrating blood vessel adventitias (asterisks). (F) Another type of Intermediate-Stage histopathological remodelling (AtCM stage 2) compound interstitial fibrosis (arrowheads) which takes origin from two blood vessels and a thickened endocardium (asterisks). (G) Advanced-Stage histopathological remodelling characterized by confluent myocardiosclerotic foci (asterisks). H. Compact and diffuse areas of interstitial fibrosis (Very Advanced-Stage histopathological remodelling; AtCM stage 3). Staining. (A-G) Weigert/Van Gieson combined dye. Original magnifications. (A): ×20 (scale bar is 150μm); (B-G): ×10 (scale bar in (B) is 300μm).

As mentioned above, the deposition of collagen in the extracellular space is an important hallmark of atrial fibrosis, and AtCM develops as a consequence of many pathophysiological processes, including inflammation, oxidative stress, ischaemia, and AF itself (Figure 5).2,43,45 However, the process of fibrosis is broader than just the deposition of collagens and encompasses the proliferation of fibroblasts and other mesenchymal cells and their differentiation into α-smooth muscle actin (α-SMA) expressing myofibroblasts, and the deposition of significant amounts of a range of ECM proteins, including various molecular species of collagen as well as other matricellular proteins (Figure 5).166–168 In a proteomic analysis of ECM proteins in left and right atria of patients with and without AF, the glycoprotein decorin was found to be most altered.169 Decorin is implicated in collagen fibrillogenesis and the production of a variety of other ECM molecules involved in cellular signalling; cleaved decorin suppress myostatin activity and increases myocyte size. These effects are similar to observations in a goat model of pacing-induced AF.170 Another analysis of the full proteome of atrial tissue of patients with and without AF identified neutrophil degranulation as the most upregulated process in AF, and the reactive oxygen donor myeloperoxidase as the most abundantly upregulated protein. Myeloperoxidae, in turn activates atrial fibroblasts and promotes ECM expansion.171,172 A whole-genome RNA sequencing analysis of LA tissue of patients with no AF, paroxysmal or persistent AF suggested epithelial-to-mesenchymal transition as an important driver of fibrosis formation and AF progression. Interestingly, increased (or decreased) proteins in paroxysmal vs. non-AF tended to increase (or decrease) further in persistent vs. paroxysmal AF.173 It has been shown in experimental models that early molecular and electrophysiological abnormalities precede LA structural remodelling and the formation of fibrosis. Several kinases, including liver kinase B1 (LKB1) and atrial AMP-activated protein kinase (AMPK) regulate transcription factors involved in the expression of ion channels and fibrosis and contribute to AtCM.43,45,174 These studies provide a link between inflammation and metabolic stress and the development of AtCM. The PREDICT AF trial demonstrated upregulation of various ECM proteins, including collagen1A1, collagen 3A1, collagen8A2, tenascin-C, thrombospondin-2, and biglycan in patients without a history of AF or AtCM who develop AF during the subsequent months or years.175

4. Imaging advances

The multimodality evaluation of atrial size and function using cardiac imaging is key to the diagnosis of AtCM and the characterization of the degree of atrial damage.176

4.1. Atrial imaging tools in the evaluation of atrial cardiomyopathy

AtCM can be evaluated with echocardiography, CMR imaging, and cardiac computed tomography (CCT). Echocardiography and CMR allow continuous recording of atrial volumes through the cardiac cycle. CCT evaluates accurately atrial volume at a single point in the cardiac cycle. Three-dimensional transthoracic echocardiography (TTE) avoids the problem of foreshortening of the atria that can occur with two-dimensional TTE.177 Left and right atrial appendages are more specifically characterized by transoesophageal echocardiography (TEE) and CCT. CCT is also validated in the evaluation of EAT and has been recently implemented with deep learning algorithms.178 The progression of LA roof shape, as determined by CT, is associated with the development of non-PV arrhythmic substrates in patients undergoing AF ablation.179,180 Pre-AF ablation CCT-based regional wall deformations were better predictors of low-voltage area than other remodelling surrogates, suggesting potential clinical utility.181

CMR has been applied to evaluate atrial fibrosis with late gadolinium enhancement analysis and atrial velocity based on 4D flow imaging (see detailed discussion below).182,183 The specific characteristics of each technique in atrial evaluation are summarized in Table 4.

Table 4

Atrial imaging tools: utility of multimodality imaging

Atrial parametersTTETEECardiac MRICardiac CTElectroanatomic mappinga
Atrial Size++ (more accurate in 3D TTE)++++++
Atrial Function+++ (more accurate in 3D TTE)++++
Atrial appendage size+++++++
Thrombus+++++++++ (delayed acquisition)b
Epicardial adipose tissue+++++
Atrial fibrosis+ (LGE)b+
Atrial velocity++ (exclusively in atrial appendage)+++ (4D flow imaging)
Atrial activation/conduction time+ p-wave duration
Atrial parametersTTETEECardiac MRICardiac CTElectroanatomic mappinga
Atrial Size++ (more accurate in 3D TTE)++++++
Atrial Function+++ (more accurate in 3D TTE)++++
Atrial appendage size+++++++
Thrombus+++++++++ (delayed acquisition)b
Epicardial adipose tissue+++++
Atrial fibrosis+ (LGE)b+
Atrial velocity++ (exclusively in atrial appendage)+++ (4D flow imaging)
Atrial activation/conduction time+ p-wave duration

LGE, late gadolinium enhancement; 3D, Three-dimensional; TTE, transthoracic echocardiography; TEE, transoesophageal echocardiography; MRI, magnetic resonance imaging; CT, computed tomography.

aElectroanatomic mapping includes voltage mapping, activation sequence mapping, and evaluation of conduction velocity and fractionated electrograms.

bContrast agents are necessary.

Table 4

Atrial imaging tools: utility of multimodality imaging

Atrial parametersTTETEECardiac MRICardiac CTElectroanatomic mappinga
Atrial Size++ (more accurate in 3D TTE)++++++
Atrial Function+++ (more accurate in 3D TTE)++++
Atrial appendage size+++++++
Thrombus+++++++++ (delayed acquisition)b
Epicardial adipose tissue+++++
Atrial fibrosis+ (LGE)b+
Atrial velocity++ (exclusively in atrial appendage)+++ (4D flow imaging)
Atrial activation/conduction time+ p-wave duration
Atrial parametersTTETEECardiac MRICardiac CTElectroanatomic mappinga
Atrial Size++ (more accurate in 3D TTE)++++++
Atrial Function+++ (more accurate in 3D TTE)++++
Atrial appendage size+++++++
Thrombus+++++++++ (delayed acquisition)b
Epicardial adipose tissue+++++
Atrial fibrosis+ (LGE)b+
Atrial velocity++ (exclusively in atrial appendage)+++ (4D flow imaging)
Atrial activation/conduction time+ p-wave duration

LGE, late gadolinium enhancement; 3D, Three-dimensional; TTE, transthoracic echocardiography; TEE, transoesophageal echocardiography; MRI, magnetic resonance imaging; CT, computed tomography.

aElectroanatomic mapping includes voltage mapping, activation sequence mapping, and evaluation of conduction velocity and fractionated electrograms.

bContrast agents are necessary.

4.2. Atrial size and function

The atria serve a variety of important functions, e.g. (i) as a reservoir for blood prior to ventricular filling, (ii) as a transport chamber for the conduit of blood during diastole, and (iii) as an active booster pump during atrial systole. Three main atrial volumes reflect these three main functional phases of the atria during the cardiac cycle: (i) maximum atrial volume, evaluated in end-systole during the reservoir phase, (ii) pre-atrial contraction volume evaluated in mid-diastole during the conduit phase and (iii) minimum atrial volume evaluated in late diastole during the booster-pump phase (Figure 6). Atrial function indices are calculated from the different atrial volumes. Atrial total emptying fraction and expansion index consider minimal and maximal atrial volumes, atrial passive emptying fraction considers maximal and pre-atrial contraction volumes, atrial active emptying fraction considers pre-atrial contraction and minimal volumes. Atrial strain is evaluated using TTE and cardiac CMR during the three phases of the atria: reservoir strain, conduit strain and contractile strain (Figure 6; supplementary material online, Figure  S1 and S1.1). Notably, CCT emerges as a valuable tool for comprehensively evaluating atrial function in AtCM. CCT provides unparalleled spatial resolution when compared to other imaging methods, allowing detailed assessment of atrial function and morphology.184 Different studies define normal values of size and function for the left and right atria in the different phases of the cardiac cycle in TTE, CMR, and CCT (see Supplementary material online, Table S1).185–192 Recently, data in the literature suggest that LA reservoir strain <18% could be useful in heart failure to define high left ventricular filling pressures. However, to the best of our knowledge, there is no cut-off value defining irreversible atrial dysfunction in AtCM (Table 2; Table 5).

Atrial size and function during the cardiac cycle: volume (A) and strain (B) curve analysis. Precise calculations: Total EF = [(maxAV − minAV)/ maxAV]; Expansion Volume = [(maxAV − minAV)/minAV]; Passive EF = [(maxAV − preAV)/maxAV]; Active EF = [(preAV − minAV)/preAV]. Abbreviations: maxAV, Maximal Atrial Volume; minAV, Minimal Atrial Volume; preAV, Pre–atrial contraction Volume; EF, Emptying Fraction.
Figure 6

Atrial size and function during the cardiac cycle: volume (A) and strain (B) curve analysis. Precise calculations: Total EF = [(maxAV − minAV)/ maxAV]; Expansion Volume = [(maxAV − minAV)/minAV]; Passive EF = [(maxAV − preAV)/maxAV]; Active EF = [(preAV − minAV)/preAV]. Abbreviations: maxAV, Maximal Atrial Volume; minAV, Minimal Atrial Volume; preAV, Pre–atrial contraction Volume; EF, Emptying Fraction.

Table 5

Multimodal assessment of cardiac chamber anatomy and function in AF: assessment of stroke risk prediction using LA strain study

Author, year, ReferencePopulationTechnique, methods of evaluating parametersResults
Stroke risk prediction with LA deformation studies
Shih, 2011178
  • 66 patients

  • Permanent AF

  • two groups according to the presence of a history of stroke or not

  • 2D TTE

  • LA Strain

  • Compared to AF patients without stroke, AF + stroke patients have a lower atrial strain (all components)

  • In multivariate analysis adjusted for age, indexed LA volume, and LVEF, active and reservoir phases of strain were independently associated with stroke risk, (association not present for conduit strain, E', and E/E')

Azemi, 2012182
  • Retrospective study

  • Patients with AF, stroke or TIA, and CHADS2 scores ≤1 before their event were retrospectively identified and compared with age- and sex-matched controls

  • 2D TTE

  • LA Strain

Compared to controls, patients with AF, stroke or TIA and CHADS2 scores ≤1 before their event had:
  • Lower LVEF

  • Larger LA size and LAV (32.3 ± 13.3 vs. 24.4 ± 11.9 mL/m2; P = 0.012)

  • A lower peak of LA negative strain (−3.2 ± 1.2% vs. −6.9 ± 4.2%; P < 0.001) and a lower peak of LA positive strain (14 ± 11% vs. 25 ± 12%; P < 0.001)

  • Peak negative of LA strain was significantly associated with stroke (OR 2.15; P < 0.001)

Obokata, 2014183
  • Prospective study

  • Paroxysmal AF or persistent AF with an acute embolic event (82 patients) or without (204 controls)

  • 2D TTE

  • LA Strain

  • 26 patients with acute embolism died during follow-up with a median follow-up time of 425 days

  • The global LA strain was lower in patients with acute embolism compared to the control group (P < 0.001)

  • In multivariate analysis, global LA strain was independently associated with acute embolism (OR 0.74; 95% CI 0.67, 0.82; P < 0.001) and had an incremental value in addition to the CHA2DS2-VASc score (P < 0.0001)

  • Global LA strain independently predicts mortality after embolism

Leung, 2018184
  • 1361 patients

  • First diagnosis of AF

  • 7.9 years follow-up on stroke occurrence

  • 2D TTE

  • LAV, LA strain, LV GLS

  • PA-TDI, reflects the total atrial conduction time

100 patients (7%) developed an ischemic stroke
Despite the similarity of LV dimensions, LVEF, longitudinal global strain, LA volumes patients with AF and stroke have in comparison with patients without stroke have:
  • Lower LA reservoir strain (14.5% vs. 18.9%; P = 0.005)

  • Lower conducted strain (10.5% vs. 13.5%; P = 0.013)

  • Longer PA-TDI (166 vs. 141 ms; P < 0.001)

Author, year, ReferencePopulationTechnique, methods of evaluating parametersResults
Stroke risk prediction with LA deformation studies
Shih, 2011178
  • 66 patients

  • Permanent AF

  • two groups according to the presence of a history of stroke or not

  • 2D TTE

  • LA Strain

  • Compared to AF patients without stroke, AF + stroke patients have a lower atrial strain (all components)

  • In multivariate analysis adjusted for age, indexed LA volume, and LVEF, active and reservoir phases of strain were independently associated with stroke risk, (association not present for conduit strain, E', and E/E')

Azemi, 2012182
  • Retrospective study

  • Patients with AF, stroke or TIA, and CHADS2 scores ≤1 before their event were retrospectively identified and compared with age- and sex-matched controls

  • 2D TTE

  • LA Strain

Compared to controls, patients with AF, stroke or TIA and CHADS2 scores ≤1 before their event had:
  • Lower LVEF

  • Larger LA size and LAV (32.3 ± 13.3 vs. 24.4 ± 11.9 mL/m2; P = 0.012)

  • A lower peak of LA negative strain (−3.2 ± 1.2% vs. −6.9 ± 4.2%; P < 0.001) and a lower peak of LA positive strain (14 ± 11% vs. 25 ± 12%; P < 0.001)

  • Peak negative of LA strain was significantly associated with stroke (OR 2.15; P < 0.001)

Obokata, 2014183
  • Prospective study

  • Paroxysmal AF or persistent AF with an acute embolic event (82 patients) or without (204 controls)

  • 2D TTE

  • LA Strain

  • 26 patients with acute embolism died during follow-up with a median follow-up time of 425 days

  • The global LA strain was lower in patients with acute embolism compared to the control group (P < 0.001)

  • In multivariate analysis, global LA strain was independently associated with acute embolism (OR 0.74; 95% CI 0.67, 0.82; P < 0.001) and had an incremental value in addition to the CHA2DS2-VASc score (P < 0.0001)

  • Global LA strain independently predicts mortality after embolism

Leung, 2018184
  • 1361 patients

  • First diagnosis of AF

  • 7.9 years follow-up on stroke occurrence

  • 2D TTE

  • LAV, LA strain, LV GLS

  • PA-TDI, reflects the total atrial conduction time

100 patients (7%) developed an ischemic stroke
Despite the similarity of LV dimensions, LVEF, longitudinal global strain, LA volumes patients with AF and stroke have in comparison with patients without stroke have:
  • Lower LA reservoir strain (14.5% vs. 18.9%; P = 0.005)

  • Lower conducted strain (10.5% vs. 13.5%; P = 0.013)

  • Longer PA-TDI (166 vs. 141 ms; P < 0.001)

2D, two-dimensional; 3D, three-dimensional; AF, atrial fibrillation, TTE, transthoracic echocardiography; LAV, LA volume; LA, left atrium; LVEF, left ventricular ejection fraction; CI, confidence interval; GLS, global longitudinal strain; HR, hazard ratio; LV, left ventricle; OR, odds ratio; PA-TDI, P-wave to A' duration on tissue Doppler imaging.

Table 5

Multimodal assessment of cardiac chamber anatomy and function in AF: assessment of stroke risk prediction using LA strain study

Author, year, ReferencePopulationTechnique, methods of evaluating parametersResults
Stroke risk prediction with LA deformation studies
Shih, 2011178
  • 66 patients

  • Permanent AF

  • two groups according to the presence of a history of stroke or not

  • 2D TTE

  • LA Strain

  • Compared to AF patients without stroke, AF + stroke patients have a lower atrial strain (all components)

  • In multivariate analysis adjusted for age, indexed LA volume, and LVEF, active and reservoir phases of strain were independently associated with stroke risk, (association not present for conduit strain, E', and E/E')

Azemi, 2012182
  • Retrospective study

  • Patients with AF, stroke or TIA, and CHADS2 scores ≤1 before their event were retrospectively identified and compared with age- and sex-matched controls

  • 2D TTE

  • LA Strain

Compared to controls, patients with AF, stroke or TIA and CHADS2 scores ≤1 before their event had:
  • Lower LVEF

  • Larger LA size and LAV (32.3 ± 13.3 vs. 24.4 ± 11.9 mL/m2; P = 0.012)

  • A lower peak of LA negative strain (−3.2 ± 1.2% vs. −6.9 ± 4.2%; P < 0.001) and a lower peak of LA positive strain (14 ± 11% vs. 25 ± 12%; P < 0.001)

  • Peak negative of LA strain was significantly associated with stroke (OR 2.15; P < 0.001)

Obokata, 2014183
  • Prospective study

  • Paroxysmal AF or persistent AF with an acute embolic event (82 patients) or without (204 controls)

  • 2D TTE

  • LA Strain

  • 26 patients with acute embolism died during follow-up with a median follow-up time of 425 days

  • The global LA strain was lower in patients with acute embolism compared to the control group (P < 0.001)

  • In multivariate analysis, global LA strain was independently associated with acute embolism (OR 0.74; 95% CI 0.67, 0.82; P < 0.001) and had an incremental value in addition to the CHA2DS2-VASc score (P < 0.0001)

  • Global LA strain independently predicts mortality after embolism

Leung, 2018184
  • 1361 patients

  • First diagnosis of AF

  • 7.9 years follow-up on stroke occurrence

  • 2D TTE

  • LAV, LA strain, LV GLS

  • PA-TDI, reflects the total atrial conduction time

100 patients (7%) developed an ischemic stroke
Despite the similarity of LV dimensions, LVEF, longitudinal global strain, LA volumes patients with AF and stroke have in comparison with patients without stroke have:
  • Lower LA reservoir strain (14.5% vs. 18.9%; P = 0.005)

  • Lower conducted strain (10.5% vs. 13.5%; P = 0.013)

  • Longer PA-TDI (166 vs. 141 ms; P < 0.001)

Author, year, ReferencePopulationTechnique, methods of evaluating parametersResults
Stroke risk prediction with LA deformation studies
Shih, 2011178
  • 66 patients

  • Permanent AF

  • two groups according to the presence of a history of stroke or not

  • 2D TTE

  • LA Strain

  • Compared to AF patients without stroke, AF + stroke patients have a lower atrial strain (all components)

  • In multivariate analysis adjusted for age, indexed LA volume, and LVEF, active and reservoir phases of strain were independently associated with stroke risk, (association not present for conduit strain, E', and E/E')

Azemi, 2012182
  • Retrospective study

  • Patients with AF, stroke or TIA, and CHADS2 scores ≤1 before their event were retrospectively identified and compared with age- and sex-matched controls

  • 2D TTE

  • LA Strain

Compared to controls, patients with AF, stroke or TIA and CHADS2 scores ≤1 before their event had:
  • Lower LVEF

  • Larger LA size and LAV (32.3 ± 13.3 vs. 24.4 ± 11.9 mL/m2; P = 0.012)

  • A lower peak of LA negative strain (−3.2 ± 1.2% vs. −6.9 ± 4.2%; P < 0.001) and a lower peak of LA positive strain (14 ± 11% vs. 25 ± 12%; P < 0.001)

  • Peak negative of LA strain was significantly associated with stroke (OR 2.15; P < 0.001)

Obokata, 2014183
  • Prospective study

  • Paroxysmal AF or persistent AF with an acute embolic event (82 patients) or without (204 controls)

  • 2D TTE

  • LA Strain

  • 26 patients with acute embolism died during follow-up with a median follow-up time of 425 days

  • The global LA strain was lower in patients with acute embolism compared to the control group (P < 0.001)

  • In multivariate analysis, global LA strain was independently associated with acute embolism (OR 0.74; 95% CI 0.67, 0.82; P < 0.001) and had an incremental value in addition to the CHA2DS2-VASc score (P < 0.0001)

  • Global LA strain independently predicts mortality after embolism

Leung, 2018184
  • 1361 patients

  • First diagnosis of AF

  • 7.9 years follow-up on stroke occurrence

  • 2D TTE

  • LAV, LA strain, LV GLS

  • PA-TDI, reflects the total atrial conduction time

100 patients (7%) developed an ischemic stroke
Despite the similarity of LV dimensions, LVEF, longitudinal global strain, LA volumes patients with AF and stroke have in comparison with patients without stroke have:
  • Lower LA reservoir strain (14.5% vs. 18.9%; P = 0.005)

  • Lower conducted strain (10.5% vs. 13.5%; P = 0.013)

  • Longer PA-TDI (166 vs. 141 ms; P < 0.001)

2D, two-dimensional; 3D, three-dimensional; AF, atrial fibrillation, TTE, transthoracic echocardiography; LAV, LA volume; LA, left atrium; LVEF, left ventricular ejection fraction; CI, confidence interval; GLS, global longitudinal strain; HR, hazard ratio; LV, left ventricle; OR, odds ratio; PA-TDI, P-wave to A' duration on tissue Doppler imaging.

4.3. LA late gadolinium-enhanced cardiac magnetic resonance imaging

Late gadolinium-enhanced CMR (LGE-CMR) methods play a pivotal role for diagnostic and prognostic assessment of ventricular cardiomyopathies. LGE may be caused by conditions that significantly increase the interstitial space or that slow down gadolinium exit.193 LGE is a marker of myocardial fibrosis in chronic myocardial infarction and various non-ischaemic cardiomyopathies. In addition, LGE may represent an increase in the interstitial space caused by interstitial oedema or by tissue infiltration of inflammatory cells, like acute myocarditis or cardiac sarcoidosis.193,194 Interstitial overload of amyloid proteins is also a potential cause of LGE.193 In recent years, LGE-CMR methods have been developed in an attempt to systematically image and quantify LA fibrosis.181 However, LGE-CMR presents challenges for the detection of LA fibrosis, including lack of standardized acquisition protocols and processing techniques, and low image resolution related to the thin LA wall,2,195 and is still available only in a limited number of research centers. Recent studies comparing the most common LGE-CMR methods revealed considerable discrepancies in global and regional LGE extent.196,197 LGE-CMR cannot be used to visualize diffuse fibrosis because ‘normal’ myocardium with diffuse fibrosis is nulled to highlight focal scar, resulting in loss of information on any background interstitial expansion.198 This limitation is potentially quite problematic in light of an autopsy study showed that atrial fibrosis is often a diffuse process.199 LGE regions detected in the atria may represent relatively advanced fibrotic or non-fibrotic lesions. Studies using LGE-CMR to quantify LA fibrosis have provided interesting observations such as anchoring of LA rotors at the border between fibrotic and non-fibrotic zones,200 a correlation between greater fibrosis and more AF recurrence post-AF ablation201 and a relationship between the extent of LA fibrosis and stroke risk.202 Two recent randomized controlled studies reported that targeting LA fibrosis detected on LGE-MRI in addition to PV isolation, compared to PV isolation alone, resulted in no significant improvement in atrial arrhythmia recurrence rates.203,204

4.4. Electroanatomic mapping

Electroanatomic mapping has been widely used during catheter ablation (CA) procedures to define AtCM. Electroanatomical mapping provides voltage mapping, activation sequence mapping, and evaluation of conduction velocity and fractionated electrograms.78,196,205,206 A reduction in endocardial bipolar voltage amplitudes, represented as low-voltage areas on electroanatomic maps (LVA, commonly defined as an area with bipolar voltage <0.5 mV), has been widely used as a marker for AtCM. Diffuse voltage reductions are associated with increase in left atrial volume and other cardiovascular risk factors (Figure 4).205 LVAs have been considered a surrogate of local fibrosis. An electroanatomical mapping study showed that atrial bipolar voltage reduction is a diffuse process; with LVAs being local reflections of diffuse voltage reduction associated with histological fibrosis.205 However, a recent atrial biopsy study showed that not only fibrosis, but also other histological factors including increased extracellular space, myofibrillar loss, decreased myocardial nuclear density, and amyloid deposition are associated with atrial voltage reduction, reflecting associated complex pathological changes (Figure 4).78

4.5. Prediction of incident atrial fibrillation by cardiac imaging

At present, no specific AF treatment decision is based solely on cardiac imaging. In large cohort studies using TTE and CMR, increases in LA volumes and decreases in LA function were associated with incident AF.207–211 In a large multi-ethnic population, larger RA volume indices were also independently associated with incident AF after adjustment for cardiovascular risk factors and LA parameters.212 CCT-determined EAT mass appears correlated with the risk of incident AF.213,214 In addition, EAT volume is highly associated with paroxysmal and persistent AF independent of traditional cardiovascular risk factors.41 Impaired LA function and LA enlargement were associated with the number of premature atrial contractions per hour on extended ambulatory electrocardiographic monitoring.215,216

4.6. Recurrent atrial fibrillation

The progression of LA roof shape, as determined by CT, is associated with the development of non-PV arrhythmic substrates in patients undergoing AF ablation.179,180 Pre-AF ablation CCT-based regional wall deformations were better predictors of LVA presence than other remodelling surrogates, suggesting potential clinical utility.181 LA fibrosis extent estimated by LGE-CMR is significantly and independently associated with recurrent arrhythmia post-AF ablation.201 However, two recent randomized controlled studies reported that targeting LA fibrosis detected on LGE-MRI in addition to PV isolation, compared to PV isolation alone, resulted in no significant improvement in atrial arrhythmia recurrence rates.203,204 The authors suggested that the lack of benefit from fibrosis-guided ablation is explained by limited overall fibrosis burden in the cohort204 and technical challenges, including lack of standardized and established end-points for fibrosis ablation.203 They also hypothesized that different types of fibrosis can occur in atrial tissue, including interstitial and reparative fibrosis, unequally contributing to AF development.203

4.7. Reverse atrial remodelling and atrial remodelling

Cardiac imaging could be useful to predict reverse cardiac remodelling following treatment of AF217 and to evaluate anatomical and functional components of reverse remodelling, as well as improvement in functional atrio-ventricular valve regurgitation after restoration of sinus rhythm.218 Using CCT, Huang et al.219 observed that atrial remodelling begins with increased atrial (LA and RA) volumes after 6 min of atrial high-rate episodes (AHRE), and atrial contractile dysfunction (both LA and RA) manifests when the duration of AHRE exceeded 6 h.

4.8. Risk stratification

Using CCT, Tsao et al. 220, in their investigation of atrial remodelling in relation to AF-related stroke, found that increased age and reduced booster-pump function of the atria are independently associated with stroke occurrence, suggesting that substrate-based assessments might enhance stroke risk stratification in AF patients. LA strain analysis appeared to improve the risk stratification of thromboembolism in patients with AF183 and to discriminate the presence of LA or LA appendage (LAA) thrombus on TEE.221,222 LAA morphologies and volume are correlated with the history of ischaemic stroke and transient ischaemic attack.223,224 AF resulted in overall impaired flow dynamics in both LA and LAA.225 Regardless of history of AF, patients at moderate-to-high risk of stroke exhibit modified flow characteristics in the LA on CMR.225,226 A greater atrial FDG uptake on PET-scanning was associated with a greater prevalence of stroke in AF patients.227 The presence of LVA has been reported to be associated with history of stroke and subclinical silent cerebral ischaemia (SCI)228 and concurrent heart failure with systolic dysfunction in patients with non-valvular AF.229 In addition, a recent observational study enrolling 1488 patients undergoing AF ablation showed that LVA presence and its extent were both associated with poor long-term composite end-points of death, heart failure, and stroke, irrespective of AF recurrence or other confounders.230 Cardiac imaging allows identification of early atrial dysfunction and its systematic integration in the characterization of AtCM, and as a diagnostic and prognostic imaging tool merits active investigation.227

5. Biomarkers, omics, ECG parameters

5.1. Biomarkers

A whole range of biomarkers, whether urine, blood, imaging, or ECG-based, have been proposed as surrogates of an ‘AtCM state’ that may predict the occurrence and progression of AF. Blood-based biomarkers can be used in combination with ECG/electrophysiological or echocardiographic data, to define such a state. These biomarkers can be electrophysiological, structural, haemodynamic, serological, and genetic (Table 6). Importantly, these biomarkers of atrial cardiomyopathy have all been associated with the risk of stroke, even in the absence of overt AF. Hence, they have been used as inclusion criteria in some studies investigating AtCM in relation to stroke or cerebrovascular risk, as defining criteria for the condition. For example, in the ARCADIA trial,232 which investigated patients with cryptogenic ischaemic stroke and atrial cardiomyopathy, the inclusion criteria included patients ≥45 years of age with embolic stroke of undetermined source (ESUS) and evidence of AtCM defined as ≥1 of the following markers: P-wave terminal force >5000 µV × ms in ECG lead V1, serum NT-proBNP > 250 pg/mL, and left atrial diameter index ≥3 cm/m2 on the echocardiogram. Of note, the ARCADIA trial did not confirm effectiveness of these biomarkers in selecting patients with cryptogenic stroke for OAC therapy.76 The ARIC (Atherosclerosis Risk in Communities) study evaluated the association of echocardiographic left atrial function (reservoir, conduit, and contractile strain) and left atrial size (left atrial volume index) with ischaemic stroke.233 A further analysis from ARIC found that the presence of AtCM (based on P-wave terminal, NT-proBNP, and LA volume index criteria) was significantly associated with an increased risk of dementia, with limited impact of the effect by AF or stroke.234

Table 6

Potential biomarkers associated with atrial cardiomyopathy and stroke risk

Biomarkers classified into categoriesSpecific examples
Electrophysiological
Atrial fibrillationSubclinical atrial fibrillation
P-wave featuresP-wave terminal force in lead V1
Duration of amplified p-wave
P-wave morphology
Paroxysmal supraventricular tachycardia
Atrial ectopyExcessive supraventricular ectopic activity
Structural
Left atrial sizeLeft atrial enlargement, left atrial volume index
LAA morphologyNon-chicken wing type
Myocardial fibrosisRegions of delayed gadolinium enhancement on cardiac MRI
Haemodynamic
LAA-flow velocityLow flow velocity
Serological
NT-proBNP, BNP
Hs-cTnT
Genetic
Polymorphismsrs2200733, rs10033464 in chromosome 4q25
Biomarkers classified into categoriesSpecific examples
Electrophysiological
Atrial fibrillationSubclinical atrial fibrillation
P-wave featuresP-wave terminal force in lead V1
Duration of amplified p-wave
P-wave morphology
Paroxysmal supraventricular tachycardia
Atrial ectopyExcessive supraventricular ectopic activity
Structural
Left atrial sizeLeft atrial enlargement, left atrial volume index
LAA morphologyNon-chicken wing type
Myocardial fibrosisRegions of delayed gadolinium enhancement on cardiac MRI
Haemodynamic
LAA-flow velocityLow flow velocity
Serological
NT-proBNP, BNP
Hs-cTnT
Genetic
Polymorphismsrs2200733, rs10033464 in chromosome 4q25

LAA, left atrial appendage; MRI, magnetic resonance imaging; NT-proBNP, N-terminal pro-brain natriuretic peptide; BNP, brain natriuretic peptide; Hs-cTnT, high-sensitivity cardiac troponin T.231

Table 6

Potential biomarkers associated with atrial cardiomyopathy and stroke risk

Biomarkers classified into categoriesSpecific examples
Electrophysiological
Atrial fibrillationSubclinical atrial fibrillation
P-wave featuresP-wave terminal force in lead V1
Duration of amplified p-wave
P-wave morphology
Paroxysmal supraventricular tachycardia
Atrial ectopyExcessive supraventricular ectopic activity
Structural
Left atrial sizeLeft atrial enlargement, left atrial volume index
LAA morphologyNon-chicken wing type
Myocardial fibrosisRegions of delayed gadolinium enhancement on cardiac MRI
Haemodynamic
LAA-flow velocityLow flow velocity
Serological
NT-proBNP, BNP
Hs-cTnT
Genetic
Polymorphismsrs2200733, rs10033464 in chromosome 4q25
Biomarkers classified into categoriesSpecific examples
Electrophysiological
Atrial fibrillationSubclinical atrial fibrillation
P-wave featuresP-wave terminal force in lead V1
Duration of amplified p-wave
P-wave morphology
Paroxysmal supraventricular tachycardia
Atrial ectopyExcessive supraventricular ectopic activity
Structural
Left atrial sizeLeft atrial enlargement, left atrial volume index
LAA morphologyNon-chicken wing type
Myocardial fibrosisRegions of delayed gadolinium enhancement on cardiac MRI
Haemodynamic
LAA-flow velocityLow flow velocity
Serological
NT-proBNP, BNP
Hs-cTnT
Genetic
Polymorphismsrs2200733, rs10033464 in chromosome 4q25

LAA, left atrial appendage; MRI, magnetic resonance imaging; NT-proBNP, N-terminal pro-brain natriuretic peptide; BNP, brain natriuretic peptide; Hs-cTnT, high-sensitivity cardiac troponin T.231

5.2. Blood-based biomarkers

While AF-related biomarkers have been studied extensively,235,236 relatively few biomarkers reflecting AtCM per se (without AF) have been assessed.237,238 Nonetheless, many biomarkers have been assessed in both scenarios, reflecting the non-specificity of many biomarkers (see later). Progression of AF from paroxysmal to persistent is sometimes taken to indicate underlying atrial substrate abnormalities, and some biomarkers have been tested in this scenario.

The natriuretic peptides closely associated with HF239 are also associated with risk of incident AF and its complications.240,241 Cardiac troponin (cTnT), a biomarker of myocardial damage used to diagnose myocardial ischaemic events, was associated cardioembolic stroke In the ARIC study.242 Soluble suppressor of tumorigenicity-2 protein (sST2) and tissue inhibitor of matrix metalloproteinase (TIMP)-1 are associated with AF progression, independent of clinical characteristics and biomarkers.243 Fibrosis biomarkers may predict AF recurrence after cardioversion.238 Recently, elevated serum levels of bone morphogenetic protein 10 (BMP10), primarily expressed by atrial myocytes, was shown to be associated with recurrences of AF following CA.244 Cohort studies in patients with AF demonstrated an association with ischaemic stroke,245 all-cause mortality and major adverse cerebrovascular and cardiovascular events.246

C-reactive protein (CRP) is associated with AF and AF burden in nonsurgical patients, suggesting that inflammatory processes contribute to atrial remodelling.247 The association of van Willebrand factor and soluble P-selectin with stroke supports the role of hypercoagulability and endothelial damage to the risk for stroke in AF patients.248

5.3. Limitations of biomarkers

Some biomarkers are non-specific, perhaps reflecting a sick patient or a sick heart.249,250 For example, Growth Differentiation Factor 15 (GDF-15) is a stress-responsive biomarker associated with several types of cardiovascular diseases, and has been proposed as a specific biomarker of bleeding risk in AF.251 Nonetheless, GDF-15 is also associated with mortality and MACE in non-AF subjects.252 Assay variability, diurnal variation of biomarker levels and costs may limit practical usability of both urine and blood biomarkers. A balance is needed between potentially useful information they can provide and simplicity/practical application in busy clinical settings.253 Thus far, there are no biomarkers that are validated for clinical management guidance in AtCM.

5.4. Multi-omics and atrial cardiomyopathy

Primary atrial cardiomyopathy arises from atrial-specific gene variants, as well as genes that associate with extra-atrial features that are functional in the atrial myocardium.12,254 One atrial-specific gene variant that gives rise to atrial electromechanical defects is the MYL4 gene, which encodes the highly atrial-selective essential myosin light chain.255,256 Another example is the NPPA gene, which encodes ANP A: NPPA variants have been associated with extreme bi-atrial dilation, thromboembolic risk and atrial standstill.254,257 Examples of gene variants that associate with ventricular and/or systemic disorders, but can also produce atrial dysfunction include HCN4, encoding the cardiac pacemaker channel;258 SCN5A, encoding the main cardiac voltage-gated sodium channel;259 SCN1B, which encodes the voltage-gated sodium channel β1/β1B subunits;260 and LMNA, which encodes Lamin A/C.261

Most of our knowledge related to AtCM genomics, transcriptomics and epigenomics derives from studies on AF, which also leads to atrial dysfunction. Large-scale, genome-wide association studies (GWAS) have identified chromosomal loci harboring common variants that influence atrial ECG parameters, including heart rate, P-wave duration, and PR interval,262 as well as variants associated with cardiac size, structure and contractile function.263 Altogether, GWAS studies have revealed over 100 genetic loci associated with AF susceptibility (Figure 7).264,265 It is unknown whether the expression of any of these variants also results in primary AtCM, but their association with AF may lead to secondary AtCM.

Manhattan plot showing known loci and novel loci associated with atrial fibrillation. A total of 34 740 186 genetic variants (each represented by a dot) were tested, comparing 60 620 atrial fibrillation cases and 970 216 controls free of atrial fibrillation. The x axis represents the genome in physical order, and the y axis represents P values (−log10(P value)) of association. The black horizontal dotted line represents a Bonferroni-corrected threshold of statistical significance corresponding to 1 000 000 independent tests (P < 5 × 10−8). Reprinted by permission from Nielsen et al.264
Figure 7

Manhattan plot showing known loci and novel loci associated with atrial fibrillation. A total of 34 740 186 genetic variants (each represented by a dot) were tested, comparing 60 620 atrial fibrillation cases and 970 216 controls free of atrial fibrillation. The x axis represents the genome in physical order, and the y axis represents P values (−log10(P value)) of association. The black horizontal dotted line represents a Bonferroni-corrected threshold of statistical significance corresponding to 1 000 000 independent tests (P < 5 × 10−8). Reprinted by permission from Nielsen et al.264

Transcriptional studies in human atrial samples using both microarrays and RNA-seq have identified genes responsible for atrial remodelling,266 among them NPPA and NPPB,267 GPR22 and RGS6, encoding G-protein signalling components; NTM, that encodes neurotrimin, a promising novel marker of HF,268 or COLQ, which is found in both left and right atria, and encodes a variant collagen that anchors acetylcholinesterase to the basal lamina.269 A recent mRNA sequencing study in atrial specimens from patients undergoing cardiac surgery identified 35 genes robustly associated with AF independently of the clinical diagnose of heart failure.267 Genes were involved in regulation of cardiomyocyte structure, conduction properties, fibrosis, inflammation, and endothelial dysfunction.270 A large-animal model study on AF progression showed that significant differential transcriptomic and proteomic expression changes are established early during the transition from paroxysmal to persistent AF.271

It is now clear that AF is a condition in which both environmental and genetic factors contribute to disease pathogenesis.265,272 Most AF-associated variants identified by GWAS lie within non-coding regions of the genome,273 where they affect gene-expression by altering transcription factor activity and the epigenetic state of chromatin. Epigenetics refers to heritable alterations that are not due to changes in DNA. Methylation of cytosine-phosphate-guanine (CpG) islands by DNA methyltransferases (DNMTs) occurs in paroxysmal and permanent AF.274,275 A recent article used 4 DNA methylation-based ageing biomarkers to provide an epigenetic basis for the known association between chronological age and incident AF (Figure 8).276,277 Other studies demonstrated histone modifications associated with AF.278,279 Specifically, histone deacetylases (HDAC) and histone transferases (HAT) appear to play an important role in AF.278 Patients suffering from AF have increased expression of EZH2, encoding the histone methyltransferase responsible for the H3K27me3 signature, in both atrial cardiomyocytes and fibroblasts.279 It is clear that progress in the understanding of the mechanisms of AtCM will require a new generation of multiomic studies in patients with AtCM independent of AF.

Although chronological age progresses at an immutable rate, biological ageing, measured by epigenetic age in this article, is modifiable by various exposures and life experiences. Thus, establishing a relationship between epigenetic age and atrial fibrillation (AF) risk gives a unique insight into modifiable ageing associated atrial fibrillation risks. Reprinted from Ward-Caviness et al.277
Figure 8

Although chronological age progresses at an immutable rate, biological ageing, measured by epigenetic age in this article, is modifiable by various exposures and life experiences. Thus, establishing a relationship between epigenetic age and atrial fibrillation (AF) risk gives a unique insight into modifiable ageing associated atrial fibrillation risks. Reprinted from Ward-Caviness et al.277

5.5. ECG parameters

In AtCM, structural, architectural, contractile, and electrophysiologic changes in the atria manifest as ECG abnormalities. Table 7 provides examples of studies supporting these associations, which have been reviewed in detail elsewhere.301–303

Table 7

ECG parameters

AssociationsReferences
Amplified digital p-wave duration (APWD)Atrial fibrosis/low voltage
Atrial contractile dysfunction, LAA thrombogenesis
Reduced LAA-velocity
AF
AF recurrence after catheter ablation
280,281
120,221
120
282
280,281
PTFV1Low LA appendage ejection velocity283
Functional atrial remodelling284
AF285
AF recurrence after catheter ablation286
P-wave axisAF287
P-wave voltageAF288
AF recurrence after catheter ablation289
P-wave areaLA enlargement290
AF in mitral stenosis291
P-wave dispersionIdiopathic paroxysmal AF292,293
AF recurrence after cardioversion294
Fibrillatory wave
(F-wave)
LA enlargement295–297
Low LA appendage flow296
Long-standing persistent AF297,298
Heart failure297
AF recurrence after catheter ablation299,300
AssociationsReferences
Amplified digital p-wave duration (APWD)Atrial fibrosis/low voltage
Atrial contractile dysfunction, LAA thrombogenesis
Reduced LAA-velocity
AF
AF recurrence after catheter ablation
280,281
120,221
120
282
280,281
PTFV1Low LA appendage ejection velocity283
Functional atrial remodelling284
AF285
AF recurrence after catheter ablation286
P-wave axisAF287
P-wave voltageAF288
AF recurrence after catheter ablation289
P-wave areaLA enlargement290
AF in mitral stenosis291
P-wave dispersionIdiopathic paroxysmal AF292,293
AF recurrence after cardioversion294
Fibrillatory wave
(F-wave)
LA enlargement295–297
Low LA appendage flow296
Long-standing persistent AF297,298
Heart failure297
AF recurrence after catheter ablation299,300

LA, left atrial; AF, atrial fibrillation.

Table 7

ECG parameters

AssociationsReferences
Amplified digital p-wave duration (APWD)Atrial fibrosis/low voltage
Atrial contractile dysfunction, LAA thrombogenesis
Reduced LAA-velocity
AF
AF recurrence after catheter ablation
280,281
120,221
120
282
280,281
PTFV1Low LA appendage ejection velocity283
Functional atrial remodelling284
AF285
AF recurrence after catheter ablation286
P-wave axisAF287
P-wave voltageAF288
AF recurrence after catheter ablation289
P-wave areaLA enlargement290
AF in mitral stenosis291
P-wave dispersionIdiopathic paroxysmal AF292,293
AF recurrence after cardioversion294
Fibrillatory wave
(F-wave)
LA enlargement295–297
Low LA appendage flow296
Long-standing persistent AF297,298
Heart failure297
AF recurrence after catheter ablation299,300
AssociationsReferences
Amplified digital p-wave duration (APWD)Atrial fibrosis/low voltage
Atrial contractile dysfunction, LAA thrombogenesis
Reduced LAA-velocity
AF
AF recurrence after catheter ablation
280,281
120,221
120
282
280,281
PTFV1Low LA appendage ejection velocity283
Functional atrial remodelling284
AF285
AF recurrence after catheter ablation286
P-wave axisAF287
P-wave voltageAF288
AF recurrence after catheter ablation289
P-wave areaLA enlargement290
AF in mitral stenosis291
P-wave dispersionIdiopathic paroxysmal AF292,293
AF recurrence after cardioversion294
Fibrillatory wave
(F-wave)
LA enlargement295–297
Low LA appendage flow296
Long-standing persistent AF297,298
Heart failure297
AF recurrence after catheter ablation299,300

LA, left atrial; AF, atrial fibrillation.

P-wave duration (IAB): a p-wave duration ≥120 ms indicates partial IAB.304 Biphasic P-wave morphology in inferior leads (II, III, aVF) indicates complete block of Bachmann's bundle (advanced IAB).302,303 IAB is strongly associated with LA enlargement.305 Advanced IAB is associated with fibrotic atrial cardiomyopathy, an environment for re-entry, and premature atrial beats, which promote the development and progression of AF.306,307 Patients with prolonged P-wave duration301 or advanced IAB have a higher risk of recurrent AF after CA.308 Paradoxically, abnormally short P-wave duration is also a risk factor for incident309 or recurrent AF.310 Amplified (150–200 mm/sec, 80–100 mm/mV) digital P-wave duration can predict arrhythmia recurrences after PVI, detection of HF patients at risk for AF, and determine different AtCM-severity stages (discrete stage of AtCM: APWD 140–150 ms; moderate AtCM-stage: APWD 150–180 ms and advanced AtCM-stage: APWD > 180 ms or presence of pos-negative p-waves in two of three inferior leads) implicating LAA slow flow velocity and LAA thrombogenesis and major adverse event rates.120,280–282

Besides P-wave duration, P-wave terminal force in V1 (PTFV1) is a commonly used marker for AtCM. PTFV1 is calculated as the product of its duration component (PTDV1) and its absolute amplitude component (PTAV1). An absolute PTFV1 value >4 mV·ms is considered pathological.287 Abnormal PTFV1 is associated with low LA appendage ejection velocity in TEE.283 Originally, a pathological PTFV1 was suggested as a marker of LA enlargement and posterior displacement, but more recent studies suggest that it is caused by conduction abnormalities resulting in predominantly posterior terminal LA activation.284,311 LA strain measured by speckle tracking echocardiography, an indicator of functional atrial remodelling, is reduced in the presence of PTFV1 abnormalities.284 A recent meta-analysis showed that PTFV1 is associated with risk of AF and is a predictor of AF in patients with and without cardiovascular disease285 and of AF recurrence after CA.286 Recently, it was shown that amplified P-wave duration of digital 12-lead ECG is associated with left atrial low-voltage substrate and can identify patients with persistent AF who are at high risk for arrhythmia recurrence after CA312 along with new-onset AF in patients with HFpEF.3,313

P-wave axis: a normal frontal plane p-wave axis is between 0 and +75.302 A recent meta-analysis indicates that an abnormal P-wave axis is associated with future detection of AF.287

P-wave voltage: P-wave voltage ≤0.1 mV in lead I is considered abnormal.302 A score incorporating abnormal p-wave voltage predicts new-onset AF.288 Lead I P-wave amplitude ≤0.1 mV is associated with recurrence of AF after radiofrequency ablation.289

P-wave area: P-wave area is calculated in lead II using the following formula: P-wave area = ½P-wave duration × P-wave voltage.302 Abnormal P-wave area is defined as ≥4 ms × mV, and has been found to be associated with LA enlargement.290 In patients with mitral stenosis, P-wave area in V3 predicts new-onset AF.291 P-wave dispersion: P-wave dispersion is defined as the difference between P-wave maximum and P-wave minimum duration on the 12-lead ECG.302 Greater P-wave dispersion was associated with incident AF and AF recurrence after cardioversion.292,294 In patients with cryptogenic stroke and an implantable loop recorder, P-wave dispersion >40 ms is a predictor of subsequent AF detection.293

Fibrillatory wave (f-wave): lower-amplitude f-waves are associated with larger LA size and lower LA appendage flow.295,296 A low f-wave amplitude in lead II and high dominant rate in lead V1 independently discriminates long-standing persistent AF from other AF-types (paroxysmal/persistent).298 In a community-based prospective survey, fine f-waves were more prevalent than coarse in patients with a larger LA diameter or with persistent AF, and was independently associated with a higher risk of HF.297 Lower preprocedural f-wave amplitudes are associated with AF recurrence in patients undergoing AF ablation.299,300

Signal averaged P-wave analysis: technological advancement of ECG analysis is currently evolving rapidly and has the potential to further improve the prognostic value of ECG recordings in patients with atrial cardiomyopathy. Signal averaged P-wave analysis of ECG recording for several minutes gives more precise and reproducible information about atrial spread of activation. Prolonged signal averaged P-wave durations were found in patients with paroxysmal AF vs. controls without history of AF314 and in patients with recurrences of AF after CA.315 P-wave parameters from prolonged recordings better distinguish between patients with paroxysmal vs. persistent AF compared to standard 10-s ECGs.316

Premature atrial contractions: several studies have identified premature atrial contractions as marker for AtCM, AF and increased stroke risk.317,318

5.6. Artificial intelligence

Artificial intelligence (AI)-based P-wave wavelet analysis promises to provide significant advancements in the field.302 Deep learning approaches have shown that patients at risk for AF can be identified by their ECG in sinus rhythm with very high accuracy,319 suggesting that the commonly used arsenal of P-wave metrics for prediction of incident AF is most likely incomplete. In a pragmatic study to test the efficacy of an AI-ECG guided screening for AF,320 the AF detection rate was five-fold higher in the high-risk AI-ECG group compared to the low-risk group, indicating that AI might increase the effectiveness of ECG-based AF screening. Other studies have successfully applied AI to quantify the substrate for AF and detect targets for AF ablation.321,322

5.7. Future directions

ECG analysis has the potential to identify patients at risk for AtCM, AF, AF recurrences and strokes, particularly if used in conjunction with imaging or circulating biomarkers. The potential clinical impact is significant. Patients at risk for AF or stroke might undergo intensified rhythm monitoring. Patients with high risk for AF recurrences might undergo more aggressive ablation strategies or the treating electrophysiologist might refrain from recommending AF ablation. However, successful implementation of these strategies will depend on finding solutions for structured collection of digital ECGs, standardization of analysis, identification of robust thresholds for risk assessment and ultimately the integration of ECG-derived parameters in prospective trials for clinical decision making.302 Prospective randomized clinical trials will be needed to evaluate the prognostic value of pre-specified ECG parameters as inclusion criteria.

6. Stroke and cognitive decline

6.1. Empiric evidence that atrial cardiomyopathy leads to stroke

There is substantial and growing evidence that AtCM, even in the absence of AF, predicts (and presumably causes) stroke.117,118 Multiple P-wave indices, as discussed above, have been associated with an increased stroke risk.323 Frequent atrial ectopy predicts stroke even without interim evidence of AF.324 While inter-observer variability and changes over time may affect reliability,325 P-wave terminal force in V1 appears to be especially predictive of embolic stroke, even in the absence of evident AF.326,327 Echocardiographic measurements demonstrating a larger LA similarly predict increased risk of stroke, again even when no AF is present.328,329 When P-wave terminal force in V1, left atrial diameter, and incident AF were included in the same multivariable model in a population-based longitudinal cohort, LA diameter did not emerge as an independent risk factor for stroke.330 More recent investigations have focused on dynamic assessments of LA function, with evidence that, among community-dwelling individuals without AF, echocardiographically-ascertained reduced LA strain is associated with incident stroke and adds predictive value to the CHA2DS2-VASc score.233

6.2. Mechanisms underlying the relationship between atrial cardiomyopathy and stroke

Even in AtCM without AF, atrial-wall substrate changes may be evident in association with risk factors such as hypertension, coronary disease, diabetes mellitus, and HF.331–334 Flow abnormalities associated with AtCM117,118 may relate to the presence of diastolic dysfunction and (relative) atrial stasis, most evident as TEE-detected LAA spontaneous echo contrast, even in sinus rhythm,335 or when a paroxysm of AF occurs. Global LA longitudinal strain assessed by speckle tracking TTE discriminates the presence of LA or LA appendage thrombus on TEE in patients with acute ischaemic stroke. Abnormal blood constituents include abnormalities of coagulation and platelets, which characterize the risk factors mentioned above.336,337 Indeed, the concept of ‘thrombogenic endocardial remodelling’ suggests that a diseased atrial endocardium may facilitate the production of thrombi independent of the presence of AF.2 In this circumstance, AF may be an epiphenomenon, with the true causal pathway between AtCM and stroke attributed more directly to pro-thrombotic properties of the atria itself.338–342

6.3. Does atrial cardiomyopathy lead to cognitive decline?

AF might induce cognitive decline through multiple mechanisms: hypoperfusion due to reduced cardiac output, silent brain infarction, inflammation, or platelet dysfunction.343–346 Cerebral microbleeds associated with anticoagulation can also cause cognitive decline.347 Finally, beat-to-beat variability seen in patients with AtCM and AF reduces cardiac output,343,348 and brain hypoperfusion is associated with cognitive dysfunction, brain ageing, and low brain volume.343,348,349 There are selected reports that have evaluated the potential association between abnormal atrial function and cognitive dysfunction, independent of AF or stroke occurrence. In an early consecutive series of 180 older individuals without AF, stroke, or a diagnosis of dementia, LA enlargement on echocardiography was significantly associated with cognitive impairment.350 Greater LA volumes and reduced LA reservoir function are associated with subclinical cerebrovascular disease on brain MRI. The ARIC Study found that the association of LA enlargement with cognitive function was borderline,351 and identified in cross-sectional analyses but not longitudinally.352 Adverse LA electrical remodelling as measured by abnormal P-wave indices was associated with an increased risk of dementia after adjusting for incident AF and stroke.351 In the most recent ARIC report focused on this question, LA enlargement was not associated with dementia risk,353 but abnormal LA strain and emptying fraction were significantly associated with an increased risk of dementia independent of AF or stroke. It is also possible that AtCM and dementia commonly co-exist without a direct causal connection and without relationship to thromboembolism per se. For example, a propensity to amyloid deposits in both the atria and the brain may lead to a concomitant AtCM and Alzheimer's disease.15,354 Left atrial dysfunction as a predictor of cognitive decline may similarly suggest a general predilection to profibrotic remodelling that adversely effects both the heart and the brain.353 In summary, based on existing data there is some evidence linking AtCM to risk of incident dementia, but no clear evidence of a strong independent association between AtCM and cognitive decline.

7. Therapeutic implications

7.1. Primary prevention of atrial cardiomyopathy

Pathological atrial remodelling may be prevented, or possibly even reversed, by addressing the initiating stimulus/disease (e.g. elevated pressure, volume, AF) or targeting the key underlying mechanisms.

Primary prevention refers to preventive therapies before the disease phenotype has emerged, for example preventing AF by preventing or reversing an AtCM.355 Lifestyle modification including achieving/maintaining a healthy bodyweight, has been shown to reduce AF burden and prevent AtCM in patients with AF,356,357 but there is limited evidence on primary prevention of AtCM (i.e. before AF presentation). Similarly, there are few data on the treatment of obstructive sleep apnoea for primary prevention of AtCM. Interventions in the renin-angiotensin-aldosterone system (RAAS) have yielded mixed results,358 with particularly convincing data supporting mineralocorticoid receptor antagonists (MRA)359,360 and RAAS inhibitors in general in primary prevention.352 A meta-analysis of 14 studies (5 RCTs) showed an odds ratio of 0.52 for new-onset AF in patients treated with MRA.359 An updated meta-analysis pooled data from 20 trials (21 791 participants, mean age: 65.2 years, 31.3% women) showed that MRA reduce AF events (risk ratio = 0.76; 95% CI: 0.67–0.87) in both patients with and without prior AF.361 Our current inability to detect the early development of AtCM limits the options to provide effective preventive medicine.

7.2. Lifestyle interventions to reverse atrial cardiomyopathy

Lifestyle-related factors (obesity, smoking, alcohol excess, hypertension and obstructive sleep apnoea) are important contributors to adverse LA remodelling and AF risk.140,141,362–364 The 2020 ESC guidelines list several risk factors and comorbidities, including cardiovascular and non-cardiovascular comorbidities and lifestyle factors, that increase the risk of AF and of AtCM.140 Randomized controlled trials examining weight loss, cardiometabolic risk factor management,141 and alcohol reduction365,366 have demonstrated reduction in AF burden and recurrence rate. In the Framingham cohort, each 10-g/day increase in alcohol intake was associated with a 0.16-mm (95%CI: 0.10–0.21-mm) larger LA dimension367 and among those with established AF, habitual drinkers had larger atria than lifelong non-drinkers (LAVi: 50 ± 13 mL/m2 vs. 43 ± 12 mL/m2). A randomized trial on alcohol abstinence vs. continuation in alcohol users with AF demonstrated later and less frequent recurrence of AF in abstainers.366 A meta-analysis of 10 prospective studies demonstrated a reduction in LA size upon obstructive sleep apnoea treatment with continuous positive airway pressure (CPAP).368 A direct effect of CPAP on electrophysiological remodelling (LA voltage, conduction velocity and LVAs) was also demonstrated in a small randomized study.369

7.3. Pharmacological therapies to reduce progression of atrial cardiomyopathy

Inhibition of the renin-angiotensin aldosterone system has shown some efficacy in preventing new-onset AF, but mostly in those with a history of HF. In a meta-analysis of 14 randomized controlled trials (92 817 patients), angiotensin receptor blockers (RR: 0.78; 95% CI: 0.66–0.92), and angiotensin-converting enzyme inhibitors (RR: 0.79; 95% CI: 0.65–1.00) both reduced the risk of incident AF.370 Another meta-analysis showed that recurrent AF was less frequent (OR 0.37; 95%CI: 0.24–0.57 P < 0.001) in MRA treated patients.359 MRA most prominently suppressed recurrent AF or AF in patients with reduced left ventricular function.371 In a meta-analysis of 5 randomized trials involving HF patients, MRA were associated with modest reductions in LAVI (−1.12 mL/m2; 95%CI: −1.91–0.33 mL/m2).372 Animal studies suggest that angiotensin converting enzyme inhibitors, angiotensin receptor blockers, and aldosterone antagonists can prevent LA enlargement, atrial fibrosis and markers (TGFβ), and atrial dysfunction.372–376 There was no difference in AF burden, nor on AF burden in 425 patients with paroxysmal AF treated with the angiotensin receptor antagonist olmesartan, but this is perhaps not surprising given the lack of angiotensin-renin system activation and generally limited structural remodelling in paroxysmal AF patients.358 SGLT-2 inhibitors are now often used as part of comprehensive therapy in systolic HF, with beneficial effects including improved cardiac energy metabolism, prevention of inflammation, oxidative stress, and adverse cardiac remodelling.377 Treatment of diabetic rats for 8 weeks with empagliflozin prevented LA enlargement, fibrosis, atrial mitochondrial dysfunction, inflammation, and AF inducibility.378 In type 2 diabetes, treatment with dapagliflozin was associated with a 19% reduction in AF events.379

7.4. Impact of rhythm control strategies on atrial cardiomyopathy

Although there is a large body of evidence linking atrial remodelling to the risk of AF and the lower success of medical rhythm control therapy or LA ablation, the opposite, i.e. studies linking reversal of atrial remodelling to improvement in AF-indices, is less well documented (Figure 9).

Interaction of risk factors, atrial cardiomyopathy, occurrence of atrial fibrillation and outcome. AtCM, atrial cardiomyopathy; AF, atrial fibrillation; CV, cardiovascular; TIA, transient ischaemic attack.
Figure 9

Interaction of risk factors, atrial cardiomyopathy, occurrence of atrial fibrillation and outcome. AtCM, atrial cardiomyopathy; AF, atrial fibrillation; CV, cardiovascular; TIA, transient ischaemic attack.

7.5. Rhythm control and atrial cardiomyopathy

The ATHENA trial demonstrated that dronedarone was associated with a reduction in the combined endpoint of first hospitalization or death.380 The use of dronedarone was also associated with fewer cases of progression to permanent AF HR: 0.65; 95% CI: 0.56–0.75 (P < 0.001).381 The EAST-AFNET 4 trial demonstrated a reduction in adverse cardiovascular outcomes in patients with recently diagnosed AF treated with early (mostly medical) rhythm control, compared to standard of care (mostly rate control).382 The effects were most pronounced in patients with cardiovascular comorbidities (CHA2DS2-VASc score ≥4).383 Restoration of sinus rhythm in a selected population appeared to reduce cardiovascular complications, potentially through reduction or reversal of atrial remodelling/AtCM.

7.6. Reversal of atrial cardiomyopathy by rhythm control?

Catheter ablation by means of pulmonary vein isolation (PVI) is the most effective rhythm control strategy for AF and can partially reverse atrial structural (LA volume increases and mitral regurgitation).133,384 and electrical remodelling. Patients with sinus rhythm after ablation showed a decrease in circulating C-reactive protein, whereas there was no change in patients with AF recurrence in the first 3 months after ablation.385 Recent work suggests that effective rhythm control can prevent progression of AF to more persistent forms.153,386 Potential mechanisms for post-ablation reduction in LA volume include the shrinkage of the left atrium through contraction of the ablation scar,387 but also reduction of mitral regurgitation and reversal of AF-induced atrial remodelling/cardiomyopathy. In the absence of AF recurrence, there was a reduction in LAVI from 31 ± 4 to 28 ± 4 mL/m2 and in atrial electromechanical conduction delay (19 ± 9–13 ± 5 msec), while no reversal of remodelling occurred in patients with AF recurrence after cryoballoon ablation.388 In 106 patients with AF (43 paroxysmal and 63 persistent AF) undergoing PVI, a significant reduction in LAVi from 43 ± 13 to 33 ± 12 mL/m2 was observed.389 Although thoracoscopic AF ablation can be effective in patients with severe LA enlargement, it is uncertain whether reduced LA volume is due to restoration of sinus rhythm per se.133,387,390 In a meta-analysis of 24 studies (2040 patients), maximum LA volume decreased by 6.5 mL (95%CI: 4.4–8.6 mL) after ablation, regardless of whether AF was paroxysmal or persistent.391 Even in the presence of severe LA dilatation (LA volume >200 mL), a 10% reduction in LA size was observed in 52% and was a strong predictor of freedom from AF post-ablation.392 In a substudy of 15 patients with systolic dysfunction who underwent AF ablation (the CAMERA-MRI [Catheter Ablation Versus Medical Rate Control in Atrial Fibrillation and Systolic Dysfunction-Magnetic Resonance Imaging] trial), RA and LA area decreased by 27% and 19%, respectively, after 23 ± 12 months of follow-up, associated with improvements in LV systolic function and atrial electrical reverse remodelling.155 In 86 patients undergoing ablation for persistent AF, those who maintained sinus rhythm at 12 months had a greater reduction in RA area, which correlated with reduced FTR.393 CMR studies have shown that both LA and RA emptying fraction increased significantly by 30–40% after 6 months of follow-up after ablation for persistent AF in 20 consecutive patients.394 LA functional reserve during exercise was lower in 61 patients with AF recurrence after ablation.395 LA strain improved after CA (21.3% ± 1.7% to 28.6% ± 1.7%), whereas it declined with medical management (26.3% ± 1.2% to 21.7% ± 1.2%), driven in part by AF burden.154 Restoration of sinus rhythm was associated with greater reductions in LA volume, presence and extent of MR and annular size.133 Electrical cardioversion of AF did result in return of atrial systolic function, but did not alter the elevated levels of plasma Von Willebrandfactor, fibrinogen and soluble P-selectin concentrations.396 Overall, the evidence strongly suggests that SR restoration and maintenance is effective in AtCM reversal.

7.7. Device therapy to reverse atrial cardiomyopathy

Cardiac resynchronization therapy (CRT) in patients with HFrEF can partially reverse atrial remodelling. In 107 patients (3 months post-CRT), LA volume was reduced, with improvements in emptying fraction; benefits were largely due to pressure and volume unloading effects from LV reverse remodelling.397 Conduction system pacing is emerging as a more physiological alternative to RV apex pacing, but did not reduce left atrial size in AF patients with HFrEF or HFpEF.398,399 Mitral-valve disease-induced LA enlargement and electrical remodelling can be partially reversed with valve surgery. In patients with severe mitral regurgitation, valve repair resulted in reductions in maximum LAVi (43 ± 14–25 ± 8 mL/m2).400 In patients undergoing mitral commissurotomy, immediate reductions in LA pressure (23 ± 7 to 10 ± 4 mm Hg) were accompanied by reductions in LA volume (75 ± 20 mL to 52 ± 18 mL) with associated improvements in conduction.401

8. Conclusions and future directions

In this document, we have summarized the extensive progress in the area of AtCM since the first consensus document.2 This substantial progress has provided new insights in many important areas, but important knowledge gaps remain, as summarized in Table 8. Additional experimental and clinical studies, registries and big data analyses are needed to fill these knowledge gaps in order to harness the full potential of the AtCM concept for improving the prevention and management of AF.402 The proposed AtCM classification system (AtCM stages; Table 2) and the synopsis about the development/progression of AtCM (Figure 9) warrant further study (for stylistic symmetry), reflection and testing.

Table 8

Knowledge gaps in AtCM

Knowledge Gaps in Atrial Cardiomyopathy (AtCM)
1.Optimal monitoring frequency and duration to measure development of AtCM
2Threshold of AtCM that results in an increased risk of stroke, heart failure, dementia, and other AF-related outcomes
3.Prevalence of subclinical AtCM in broad community-based cohorts
4.Risk factors and determinants of AtCM in broad community-based cohorts
5.Temporal relationship between different types of AtCM
6.Precise correlation of various biomarkers (ECG, blood parameters etc.) with AtCM
7.Threshold of AtCM that indicates need for anticoagulation in patients without the presence of AF (e.g. CHA2DS2-VASc score ≥4?)
8.Reversibility of AtCM in response to different therapies, and time course
9.Diversity of structural alterations in atrial tissue in patients with AtCM
10.Association of AtCM (and its subtypes) with CV comorbidities.
11.Molecular mechanisms underlying the development and progression of AtCM
12.Histological findings associated with AtCM in patients without AF
13.Relationships between atrial function and histological findings in AtCM
14.Relationship between LGE-MRI and histology in the diagnosis of AtCM
15.Multiomic analysis to elucidate the mechanisms of development and progression of AtCM
16.Impact of incretins (GLP-1 analogues, GIP etc.) on AtCM development and regression
Knowledge Gaps in Atrial Cardiomyopathy (AtCM)
1.Optimal monitoring frequency and duration to measure development of AtCM
2Threshold of AtCM that results in an increased risk of stroke, heart failure, dementia, and other AF-related outcomes
3.Prevalence of subclinical AtCM in broad community-based cohorts
4.Risk factors and determinants of AtCM in broad community-based cohorts
5.Temporal relationship between different types of AtCM
6.Precise correlation of various biomarkers (ECG, blood parameters etc.) with AtCM
7.Threshold of AtCM that indicates need for anticoagulation in patients without the presence of AF (e.g. CHA2DS2-VASc score ≥4?)
8.Reversibility of AtCM in response to different therapies, and time course
9.Diversity of structural alterations in atrial tissue in patients with AtCM
10.Association of AtCM (and its subtypes) with CV comorbidities.
11.Molecular mechanisms underlying the development and progression of AtCM
12.Histological findings associated with AtCM in patients without AF
13.Relationships between atrial function and histological findings in AtCM
14.Relationship between LGE-MRI and histology in the diagnosis of AtCM
15.Multiomic analysis to elucidate the mechanisms of development and progression of AtCM
16.Impact of incretins (GLP-1 analogues, GIP etc.) on AtCM development and regression

AtCM, atrial cardiomyopathy; AF, atrial fibrillation; CHA2DS2-VASc score, congestive heart failure, hypertension, age ≥75 (doubled), diabetes, stroke (doubled), vascular disease, age 65–74 and sex category (female). CV, cardiovascular; ECG, electrocardiogram.

Table 8

Knowledge gaps in AtCM

Knowledge Gaps in Atrial Cardiomyopathy (AtCM)
1.Optimal monitoring frequency and duration to measure development of AtCM
2Threshold of AtCM that results in an increased risk of stroke, heart failure, dementia, and other AF-related outcomes
3.Prevalence of subclinical AtCM in broad community-based cohorts
4.Risk factors and determinants of AtCM in broad community-based cohorts
5.Temporal relationship between different types of AtCM
6.Precise correlation of various biomarkers (ECG, blood parameters etc.) with AtCM
7.Threshold of AtCM that indicates need for anticoagulation in patients without the presence of AF (e.g. CHA2DS2-VASc score ≥4?)
8.Reversibility of AtCM in response to different therapies, and time course
9.Diversity of structural alterations in atrial tissue in patients with AtCM
10.Association of AtCM (and its subtypes) with CV comorbidities.
11.Molecular mechanisms underlying the development and progression of AtCM
12.Histological findings associated with AtCM in patients without AF
13.Relationships between atrial function and histological findings in AtCM
14.Relationship between LGE-MRI and histology in the diagnosis of AtCM
15.Multiomic analysis to elucidate the mechanisms of development and progression of AtCM
16.Impact of incretins (GLP-1 analogues, GIP etc.) on AtCM development and regression
Knowledge Gaps in Atrial Cardiomyopathy (AtCM)
1.Optimal monitoring frequency and duration to measure development of AtCM
2Threshold of AtCM that results in an increased risk of stroke, heart failure, dementia, and other AF-related outcomes
3.Prevalence of subclinical AtCM in broad community-based cohorts
4.Risk factors and determinants of AtCM in broad community-based cohorts
5.Temporal relationship between different types of AtCM
6.Precise correlation of various biomarkers (ECG, blood parameters etc.) with AtCM
7.Threshold of AtCM that indicates need for anticoagulation in patients without the presence of AF (e.g. CHA2DS2-VASc score ≥4?)
8.Reversibility of AtCM in response to different therapies, and time course
9.Diversity of structural alterations in atrial tissue in patients with AtCM
10.Association of AtCM (and its subtypes) with CV comorbidities.
11.Molecular mechanisms underlying the development and progression of AtCM
12.Histological findings associated with AtCM in patients without AF
13.Relationships between atrial function and histological findings in AtCM
14.Relationship between LGE-MRI and histology in the diagnosis of AtCM
15.Multiomic analysis to elucidate the mechanisms of development and progression of AtCM
16.Impact of incretins (GLP-1 analogues, GIP etc.) on AtCM development and regression

AtCM, atrial cardiomyopathy; AF, atrial fibrillation; CHA2DS2-VASc score, congestive heart failure, hypertension, age ≥75 (doubled), diabetes, stroke (doubled), vascular disease, age 65–74 and sex category (female). CV, cardiovascular; ECG, electrocardiogram.

Supplementary material

Supplementary material is available at Europace online.

Acknowledgements

We thank Dr. Anjaly Vijayan and IPPMed for their excellent technical assistance and support.

The authors thank the EHRA Scientific Documents Committee:

Prof. Katja Zeppenfeld, Prof. Jens Cosedis Nielsen, Prof. Isabel Deisenhofer, Dr. Luigi di Biase, Prof. Kristina Hermann Haugaa, Dr. Daniel Keene, Prof. Christian Meyer, Prof. Petr Peichl, Prof. Silvia Priori, Dr. Alireza Sepehri Shamloo, Prof. Markus Stühlinger, Prof. Jacob Tfelt Hansen, Prof. Arthur Wilde

(2023) Payment from healthcare industry: speaker fees, honoraria, consultancy, advisory board fees, investigator, committee member, etc., including travel funding related to these activities. Biosense Webster: Atrial Fibrillation (AF); Boston Scientific, Medtronic, Abbott Medical: Atrial Fibrillation (AF), Device Therapy. Research funding from healthcare industry received by your department/institution or an entity controlled by your department/institution, under your direct/personal responsibility. Johnson & Johnson: Research projects, PI; Boston Scientific, Medtronic, Biotronik, Abbott Medical: Research projects, PI.

J.-A.C.: (2021) Direct personal payment from healthcare industry: speaker fees, honoraria, consultancy, advisory board fees, investigator, committee member, etc. Biotronik, Abbott, Medtronic, Johnson & Johnson: Speaker fees.

(2023) Payment from healthcare industry: speaker fees, honoraria, consultancy, advisory board fees, investigator, committee member, etc., including travel funding related to these activities. Abbott: Arrhythmias, General; Johnson & Johnson: Arrhythmias, General.

J.R.d.G.: (2021) Direct personal payment from healthcare industry: speaker fees, honoraria, consultancy, advisory board fees, investigator, committee member, etc. Johnson & Johnson, AtrianMedical: Ablation of atrial fibrillation; Bayer, Daiichi Sankyo: Stroke prevention in atrial fibrillation. Research funding from healthcare industry under your direct/personal responsibility (to department or institution). Bayer, Daiichi Sankyo: Atrial fibrillation; Boston Scientific, Johnson & Johnson: Electrophysiology/atrial fibrillation. Any other interest (financial or otherwise) that should be declared in view of holding an ESC position. Owner of RhythmCARE, personal consultancy firm specialising in consulting, advice, research, and education on the broad field of atrial fibrillation and cardiac electrophysiology. The parties for which RhythmCARE provided services are listed above.

(2023) Ongoing research funding from public and/or not-for-profit organisations (which started in 2023 or continues in 2023). This information is not mandatory and does not impact one's ability to join an ESC activity. ERA4Health. Payment from healthcare industry: speaker fees, honoraria, consultancy, advisory board fees, investigator, committee member, etc., including travel funding related to these activities. Daiichi Sankyo: Atrial Fibrillation (AF), Stroke. AtrianMedical: Other. Payment from healthcare industry to your department or institution or any other body for your personal services: speaker fees, honoraria, consultancy, advisory board fees, investigator, committee member, etc., including travel funding related to these activities. Bayer, Daiichi Sankyo, Atricure: Atrial Fibrillation (AF). Research funding from healthcare industry received by your department/institution or an entity controlled by your department/institution, under your direct/personal responsibility. Johnson & Johnson: AF ablation, PI, Boston Scientific, Medtronic: ICD, PI; Bayer, Daiichi Sankyo: Stroke prevention in AF, PI.

D.D.: (2021) Any other interest (financial or otherwise) that should be declared in view of holding an ESC position. I am Editor-in-Chief of Int J Cardiol Heart Vasc and Deputy-Editor of Int J Cardiol. For my services on both journals, I get compensation from Elsevier.

(2023) Payment from healthcare industry: speaker fees, honoraria, consultancy, advisory board fees, investigator, committee member, etc., including travel funding related to these activities. TMA Foundation: Atrial Fibrillation (AF); Daiichi Sankyo: Pharmacology and Pharmacotherapy, Other.

G.F.: (2021) Direct personal payment from healthcare industry: speaker fees, honoraria, consultancy, advisory board fees, investigator, committee member, etc. Libbs: Pharma—antiarrhythmic drugs; Daiichi Sankyo: Pharma—DOACs.

(2023) Payment from healthcare industry: speaker fees, honoraria, consultancy, advisory board fees, investigator, committee member, etc., including travel funding related to these activities. Libbs Pharma: Arrhythmias, General.

J.J.: (2021) Payment from healthcare industry to your department or institution or any other body for your personal services: speaker fees, honoraria, consultancy, advisory board fees, investigator, committee member, etc. Stembiosys: Scientific Advisory Board. Direct ownership of shares or direct financial interest in healthcare, media, education companies or in companies related to (suppliers), or in competition with the ESC and its mission: to reduce the burden of cardiovascular disease. Healthcare—ARRHYTHMIAS—Direct ownership of shares in Corify and Stembiosys.

G.Y.L.: (2021) Payment from healthcare industry to your department or institution or any other body for your personal services: speaker fees, honoraria, consultancy, advisory board fees, investigator, committee member, etc. Daiichi-Sankyo: Anticoagulation; Boehringer Ingelheim: Anticoagulation; Registries; Steering Committees; Pfizer: Anticoagulation; Registries; BMS: Antithrombotic therapy. Research funding from healthcare industry under your direct/personal responsibility (to department or institution). Boehringer-Ingelheim: AF registries [unrestricted educational grant]. Any other interest (financial or otherwise) that should be declared in view of holding an ESC position. Shares in private limited company (a legal separate entity in UK), but no salary/dividends/income/personal renumeration received.

(2023) Research funding from healthcare industry received by your department/institution or an entity controlled by your department/institution, under your direct/personal responsibility. Boehringer-Ingelheim: AF registries [unrestricted educational grant]. Academic, non-promotional research work on registries by fellow, Principal Investigator. Anthos: AF registries [unrestricted educational grant]. Academic, non-promotional research work on registries by fellow, Principal Investigator. Any other interest (financial or otherwise) that should be declared in view of holding an ESC position (e.g. editorial role in non-ESC publications—editor in chief or other editorial position—and/or officer position in other organisations). Shares in private limited company (a legal separate entity in UK), but no salary/income/personal renumeration received.

G.M.: (2021) Direct personal payment from healthcare industry: speaker fees, honoraria, consultancy, advisory board fees, investigator, committee member, etc. Johnson and Johnson: Anticoagulation for atrial fibrillation; InCarda: Novel antiarrhythmic drug delivery system. Research funding from healthcare industry under your direct/personal responsibility (to department or institution). Baylis Medical: Transseptal puncture procedures.

(2023) Payment from healthcare industry: speaker fees, honoraria, consultancy, advisory board fees, investigator, committee member, etc., including travel funding related to these activities. InCarda: Atrial Fibrillation (AF).

K.M.: (2021) Receipt of royalties for intellectual property. Metabolic Technologies Inc: Pharmaceutical company. Research funding from healthcare industry under your direct/personal responsibility (to department or institution). NIH: American Heart Association: Atrial fibrillation.

(2023) Receipt of royalties for intellectual property. Metabolic Technologies Inc: Atrial Fibrillation (AF), Basic Science.

S.N.: (2021) Direct personal payment from healthcare industry: speaker fees, honoraria, consultancy, advisory board fees, investigator, committee member, etc. Johnson & Johnson: Inflammation in Heart Cellws; GlaxoSmithKline: Novel AF Therapies; LQT Therapeutics: Novel Long QT therapies.

(2023) Any other interest (financial or otherwise) that should be declared in view of holding an ESC position (e.g. editorial role in non-ESC publications—editor in chief or other editorial position—and/or officer position in other organisations). Editor-in-Chief, Canadian Journal of Cardiology Section Editor, Rhythm Disorders, JACC.

U.S.: (2023) Ongoing research funding from public and/or not-for-profit organisations (which started in 2023 or continues in 2023). This information is not mandatory and does not impact one's ability to join an ESC activity. Dutch Heart Foundation, EP Solutions, European Union, Roche.

Payment from healthcare industry: speaker fees, honoraria, consultancy, advisory board fees, investigator, committee member, etc., including travel funding related to these activities. YourRhythmics: Atrial Fibrillation (AF). Direct financial interest in healthcare, media, education companies, or in companies related to or in competition with the ESC and its mission: to reduce the burden of cardiovascular disease. Healthcare—Atrial Fibrillation (AF)—Shareholder of YourRhythmics BV.

L.S.-D.: (2021) Declaration not submitted.

(2023) Payment from healthcare industry: speaker fees, honoraria, consultancy, advisory board fees, investigator, committee member, etc., including travel funding related to these activities. Pfizer: Atrial Fibrillation (AF), Acute Heart Failure; Bristol Myers Squibb: Atrial Fibrillation (AF), Acute Heart Failure.

T.Y.: (2021) Direct personal payment from healthcare industry: speaker fees, honoraria, consultancy, advisory board fees, investigator, committee member, etc. Bristol Myers Squibb, Bayer, Boehringer-Ingelheim, Daiichi Sankyo, Takeda Pharmaceuticals, Otuska Pharmaceutical: atrial fibrillation; Abbott, Medtronic, Japan lifeline: catheter ablation. Payment from healthcare industry to your department or institution or any other body for your personal services: speaker fees, honoraria, consultancy, advisory board fees, investigator, committee member, etc. Abbott: catheter ablation. Research funding from healthcare industry under your direct/personal responsibility (to department or institution). Boston Scientific Japan, Japan lifeline, Abbott Medical Japan, Nihon Kohden Corpatation, Japan Medtronic, Fides-ONE Corporation: arrhythmia management.

(2023) Payment from healthcare industry: speaker fees, honoraria, consultancy, advisory board fees, investigator, committee member, etc., including travel funding related to these activities. Abbott, Novartis, NIHON KOHDEN: Arrhythmias, General; Bayer, Daiichi Sankyo, Medtronic, Bristol Myers Squibb: Atrial Fibrillation (AF). Research funding from healthcare industry received by your department/institution or an entity controlled by your department/institution, under your direct/personal responsibility. Abbott, Boston Scientific, Medtronic, NIHON KOHDEN, Japan lifeline, Fides-One corporation: research funding for atrial cardiomyopathy, atrial fibrillation, and heart failure, principal investigator.

A.G.: (2021) Direct personal payment from healthcare industry: speaker fees, honoraria, consultancy, advisory board fees, investigator, committee member, etc. Abbott, Boston Scientific, Medtronic: Ablation; Boehringer-Ingelheim, Daiichi Sankyo, Pfizer, Bayer Healthcare, Bristol Myers Squibb: anticoagulation; Berlin Chemie AG: antiischemic drugs; OMEICOS: Atrial fibrillation; Viofor: Heart Failure; Astra Zeneca: PCI, antiplatelets.

(2023) Payment from healthcare industry: speaker fees, honoraria, consultancy, advisory board fees, investigator, committee member, etc., including travel funding related to these activities. Daiichi Sankyo, Bristol Myers Squibb: Atrial Fibrillation (AF); Medtronic: Device Therapy; Boston Scientific, Bayer Healthcare: History of Cardiology, Chronic Heart Failure.

K.Z.: (2021) Research funding from healthcare industry under your direct/personal responsibility (to department or institution). Biosense Webster: Research electrophysiology.

(2022) Payment from healthcare industry to your department or institution or any other body for your personal services: speaker fees, honoraria, consultancy, advisory board fees, investigator, committee member, etc. Biosense Webster: Ventricular Arrhythmias and Sudden Cardiac Death (SCD). Research funding from healthcare industry under your direct/personal responsibility (to department or institution). Biosense Webster: investigator initiated research, PI.

(2023) Payment from healthcare industry to your department or institution or any other body for your personal services: speaker fees, honoraria, consultancy, advisory board fees, investigator, committee member, etc., including travel funding related to these activities. Biosense Webster: Ventricular Arrhythmias and Sudden Cardiac Death (SCD). Research funding from healthcare industry received by your department/institution or an entity controlled by your department/institution, under your direct/personal responsibility. Biosense Webster: Investigator initiated research, ventricular arrhythmias, PI.

N.A.: (2022) Nothing to be declared.

(2023) Ongoing research funding from public and/or not-for-profit organisations (which started in 2023 or continues in 2023). This information is not mandatory and does not impact one's ability to join an ESC activity. National Institutes of Health.

R.T.A.: (2022) Travel and meeting support from healthcare industry, independent of the above activities. Spectranetics: Device Therapy.

(2023) Payment from healthcare industry: speaker fees, honoraria, consultancy, advisory board fees, investigator, committee member, etc., including travel funding related to these activities. Daiichi Sankyo: Clinical Skills; Boston Scientific, Biotronik, Spectranetics: Device Therapy.

T.F.A.: (2022) Research funding from healthcare industry under your direct/personal responsibility (to department or institution). Biosense Webster: Investigator-initiated trial on High power-short duration RF ablatoin (QDOT), Principal Investigator.

(2023) Ongoing research funding from public and/or not-for-profit organisations (which started in 2023 or continues in 2023). This information is not mandatory and does not impact one's ability to join an ESC activity. German Research Foundation. Payment from healthcare industry: speaker fees, honoraria, consultancy, advisory board fees, investigator, committee member, etc., including travel funding related to these activities. CorifyCare: Atrial Fibrillation (AF). Funding from healthcare industry for travel to, and attendance of events and meetings, unrelated to the activities listed in 2.A and 3.A. Biosense Webster: Arrhythmias, General.

J.D.: (2022) Direct personal payment from healthcare industry: speaker fees, honoraria, consultancy, advisory board fees, investigator, committee member, etc. Astra Zeneca, Medtronic: Device Therapy, Chronic Heart Failure; Boston Scientific: Other. Travel and meeting support from healthcare industry, independent of the above activities. Boston Scientific: Other.

(2023) Payment from healthcare industry: speaker fees, honoraria, consultancy, advisory board fees, investigator, committee member, etc., including travel funding related to these activities. Astra Zeneca: Chronic Heart Failure; Medtronic: Device Therapy, Chronic Heart Failure; Boston Scientific: Other. Funding from healthcare industry for travel to, and attendance of events and meetings, unrelated to the activities listed in 2.A and 3.A. Boston Scientific: Other.

J.-B.G.: (2022) Direct personal payment from healthcare industry: speaker fees, honoraria, consultancy, advisory board fees, investigator, committee member, etc. Abbott Laboratories: Arrhythmias, General, Atrial Fibrillation (AF), Device Therapy; Microport: Arrhythmias, General, Atrial Fibrillation (AF), Supraventricular Tachycardia (Non-Atrial Fibrillation), Device Therapy. Travel and meeting support from healthcare industry, independent of the above activities. Abbott Laboratories: six items checked.

(2023) Payment from healthcare industry: speaker fees, honoraria, consultancy, advisory board fees, investigator, committee member, etc., including travel funding related to these activities. Abbott: Arrhythmias, General, Atrial Fibrillation (AF), Syncope and Bradycardia, Ventricular Arrhythmias and Sudden Cardiac Death (SCD).

J.K.: (2022) Research funding from healthcare industry under your direct/personal responsibility (to department or institution). Biosense Webster, Medtronic: Fellowship and AF research support, Principle investigator; Zoll Medical: Randomised trial of CPAP treatment for sleep apnoea patients with AF, Principal investigator.

(2023) Payment from healthcare industry to your department or institution or any other body for your personal services: speaker fees, honoraria, consultancy, advisory board fees, investigator, committee member, etc., including travel funding related to these activities. Biosense Webster: Atrial Fibrillation (AF). Research funding from healthcare industry received by your department/institution or an entity controlled by your department/institution, under your direct/personal responsibility. Zoll Medical: Atrial fibrillation and sleep apnoea, Chief investigator; Biosense Webster: Atrial fibrillation mapping and ablation, Principal investigator; Medtronic: Atrial fibrillation mapping, Chief investigator; Abbott: Atrial fibrillation PFA ablation, Principal investigator. Any other interest (financial or otherwise) that should be declared in view of holding an ESC position (e.g. editorial role in non-ESC publications—editor in chief or other editorial position—and/or officer position in other organisations). Associate editor Journal of the American College of Cardiology-Cardiac Electrophysiology Chair of the Heart Rhythm Council, Cardiac Society of Australia and New Zealand.

H.L.: (2022) Direct personal payment from healthcare industry: speaker fees, honoraria, consultancy, advisory board fees, investigator, committee member, etc. Abbott: Arrhythmias, General. Research funding from healthcare industry under your direct/personal responsibility (to department or institution). Abbott, Medtronic: Research Funding for Electrophysiology Fellow, Supervisor.

(2023) Payment from healthcare industry: speaker fees, honoraria, consultancy, advisory board fees, investigator, committee member, etc., including travel funding related to these activities. Abbott: Arrhythmias, General. Research funding from healthcare industry received by your department/institution or an entity controlled by your department/institution, under your direct/personal responsibility. Abbott, Medtronic: Research Funding for Electrophysiology Fellow, Supervisor.

E.L.A.A., S.C., S.H., Y.-J.L., H.-N.P., N.T., S.A.J.: (2021, 2023) Nothing to be declared.

J.J., D.C.: (2023) Nothing to be declared.

Data availability

No new data were generated or analysed in support of this research.

References

1

Williams
 
DO
,
Jones
 
EL
,
Nagle
 
RE
,
Smith
 
BS
.
Familial atrial cardiomyopathy with heart block
.
Q J Med
 
1972
;
41
:
491
508
.

2

Goette
 
A
,
Kalman
 
JM
,
Aguinaga
 
L
,
Akar
 
J
,
Cabrera
 
JA
,
Chen
 
SA
 et al.  
EHRA/HRS/APHRS/SOLAECE expert consensus on atrial cardiomyopathies: definition, characterization, and clinical implication
.
Europace
 
2016
;
18
:
1455
90
.

3

Goette
 
A
,
Kalman
 
JM
,
Aguinaga
 
L
,
Akar
 
J
,
Cabrera
 
JA
,
Chen
 
SA
 et al.  
EHRA/HRS/APHRS/SOLAECE expert consensus on atrial cardiomyopathies: definition, characterization, and clinical implication
.
Heart Rhythm
 
2017
;
14
:
e3
40
.

4

Goette
 
A
,
Kalman
 
JM
,
Aguinaga
 
L
,
Akar
 
J
,
Cabrera
 
JA
,
Chen
 
SA
 et al.  
EHRA/HRS/APHRS/SOLAECE expert consensus on atrial cardiomyopathies: definition, characterisation, and clinical implication
.
J Arrhythm
 
2016
;
32
:
247
78
.

5

Guichard
 
JB
,
Nattel
 
S
.
Atrial cardiomyopathy: a useful notion in cardiac disease management or a passing fad?
 
J Am Coll Cardiol
 
2017
;
70
:
756
65
.

6

Winters
 
J
,
Isaacs
 
A
,
Zeemering
 
S
,
Kawczynski
 
M
,
Maesen
 
B
,
Maessen
 
J
 et al.  
Heart failure, female sex, and atrial fibrillation are the main drivers of human atrial cardiomyopathy: results from the CATCH ME consortium
.
J Am Heart Assoc
 
2023
;
12
:
e031220
.

7

Chahine
 
Y
,
Askari-Atapour
 
B
,
Kwan
 
KT
,
Anderson
 
CA
,
Macheret
 
F
,
Afroze
 
T
 et al.  
Epicardial adipose tissue is associated with left atrial volume and fibrosis in patients with atrial fibrillation
.
Front Cardiovasc Med
 
2022
;
9
:
1045730
.

8

Abou Ziki
 
MD
,
Bhat
 
N
,
Neogi
 
A
,
Driscoll
 
TP
,
Ugwu
 
N
,
Liu
 
Y
 et al.  
Epistatic interaction of PDE4DIP and DES mutations in familial atrial fibrillation with slow conduction
.
Hum Mutat
 
2021
;
42
:
1279
93
.

9

Yoneda
 
ZT
,
Anderson
 
KC
,
Quintana
 
JA
,
O'Neill
 
MJ
,
Sims
 
RA
,
Glazer
 
AM
 et al.  
Early-onset atrial fibrillation and the prevalence of rare variants in cardiomyopathy and arrhythmia genes
.
JAMA Cardiol
 
2021
;
6
:
1371
9
.

10

Yoneda
 
ZT
,
Anderson
 
KC
,
Ye
 
F
,
Quintana
 
JA
,
O'Neill
 
MJ
,
Sims
 
RA
 et al.  
Mortality among patients with early-onset atrial fibrillation and rare variants in cardiomyopathy and arrhythmia genes
.
JAMA Cardiol
 
2022
;
7
:
733
41
.

11

Roberts
 
JD
,
Chalazan
 
B
,
Andrade
 
JG
,
Macle
 
L
,
Nattel
 
S
,
Tadros
 
R
.
Clinical genetic testing for atrial fibrillation: are we there yet?
 
Can J Cardiol
 
2024
;
40
:
540
53
.

12

Marcoux
 
E
,
Sosnowski
 
D
,
Ninni
 
S
,
Mackasey
 
M
,
Cadrin-Tourigny
 
J
,
Roberts
 
JD
 et al.  
Genetic atrial cardiomyopathies: common features, specific differences, and broader relevance to understanding atrial cardiomyopathy
.
Circ Arrhythm Electrophysiol
.
2023
;
16
:
675
98
.

13

Cornwell
 
GG
, 3rd,
Murdoch
 
WL
,
Kyle
 
RA
,
Westermark
 
P
,
Pitkänen
 
P
.
Frequency and distribution of senile cardiovascular amyloid. A clinicopathologic correlation
.
Am J Med
 
1983
;
75
:
618
23
.

14

González-López
 
E
,
Gallego-Delgado
 
M
,
Guzzo-Merello
 
G
,
de Haro-Del Moral
 
FJ
,
Cobo-Marcos
 
M
,
Robles
 
C
 et al.  
Wild-type transthyretin amyloidosis as a cause of heart failure with preserved ejection fraction
.
Eur Heart J
 
2015
;
36
:
2585
94
.

15

Röcken
 
C
,
Peters
 
B
,
Juenemann
 
G
,
Saeger
 
W
,
Klein
 
HU
,
Huth
 
C
 et al.  
Atrial amyloidosis: an arrhythmogenic substrate for persistent atrial fibrillation
.
Circulation
 
2002
;
106
:
2091
7
.

16

Tanskanen
 
M
,
Peuralinna
 
T
,
Polvikoski
 
T
,
Notkola
 
IL
,
Sulkava
 
R
,
Hardy
 
J
 et al.  
Senile systemic amyloidosis affects 25% of the very aged and associates with genetic variation in alpha2-macroglobulin and tau: a population-based autopsy study
.
Ann Med
 
2008
;
40
:
232
9
.

17

Troncone
 
L
,
Luciani
 
M
,
Coggins
 
M
,
Wilker
 
EH
,
Ho
 
CY
,
Codispoti
 
KE
 et al.  
Aβ amyloid pathology affects the hearts of patients with Alzheimer's disease: mind the heart
.
J Am Coll Cardiol
 
2016
;
68
:
2395
407
.

18

Sikkink
 
LA
,
Ramirez-Alvarado
 
M
.
Cytotoxicity of amyloidogenic immunoglobulin light chains in cell culture
.
Cell Death Dis
 
2010
;
1
:
e98
.

19

Yang
 
Z
,
Subati
 
T
,
Kim
 
K
,
Murphy
 
MB
,
Dougherty
 
OP
,
Christopher
 
IL
 et al.  
Natriuretic peptide oligomers cause proarrhythmic metabolic and electrophysiological effects in atrial myocytes
.
Circ Arrhythm Electrophysiol
 
2022
;
15
:
e010636
.

20

Yao
 
C
,
Veleva
 
T
,
Scott
 
L
, Jr.
,
Cao
 
S
,
Li
 
L
,
Chen
 
G
 et al.  
Enhanced cardiomyocyte NLRP3 inflammasome signaling promotes atrial fibrillation
.
Circulation
 
2018
;
138
:
2227
42
.

21

Heijman
 
J
,
Muna
 
AP
,
Veleva
 
T
,
Molina
 
CE
,
Sutanto
 
H
,
Tekook
 
M
 et al.  
Atrial myocyte NLRP3/CaMKII nexus forms a substrate for postoperative atrial fibrillation
.
Circ Res
 
2020
;
127
:
1036
55
.

22

Scott
 
L
, Jr.,
Fender
 
AC
,
Saljic
 
A
,
Li
 
L
,
Chen
 
X
,
Wang
 
X
 et al.  
NLRP3 inflammasome is a key driver of obesity-induced atrial arrhythmias
.
Cardiovasc Res
 
2021
;
117
:
1746
59
.

23

Zhang
 
Y
,
Zhang
 
S
,
Li
 
B
,
Luo
 
Y
,
Gong
 
Y
,
Jin
 
X
 et al.  
Gut microbiota dysbiosis promotes age-related atrial fibrillation by lipopolysaccharide and glucose-induced activation of NLRP3-inflammasome
.
Cardiovasc Res
 
2022
;
118
:
785
97
.

24

Bapat
 
A
,
Schloss
 
MJ
,
Yamazoe
 
M
,
Grune
 
J
,
Hulsmans
 
M
,
Milan
 
DJ
 et al.  
A mouse model of atrial fibrillation in sepsis
.
Circulation
 
2023
;
147
:
1047
9
.

25

Scott
 
L
, Jr.,
Li
 
N
,
Dobrev
 
D
.
Role of inflammatory signaling in atrial fibrillation
.
Int J Cardiol
 
2019
;
287
:
195
200
.

26

Dobrev
 
D
,
Heijman
 
J
,
Hiram
 
R
,
Li
 
N
,
Nattel
 
S
.
Inflammatory signalling in atrial cardiomyocytes: a novel unifying principle in atrial fibrillation pathophysiology
.
Nat Rev Cardiol
 
2023
;
20
:
145
67
.

27

Tong
 
D
,
Schiattarella
 
GG
,
Jiang
 
N
,
Daou
 
D
,
Luo
 
Y
,
Link
 
MS
 et al.  
Impaired AMP-activated protein kinase signaling in heart failure with preserved ejection fraction-associated atrial fibrillation
.
Circulation
 
2022
;
146
:
73
6
.

28

Song
 
J
,
Wu
 
J
,
Robichaux
 
DJ
,
Li
 
T
,
Wang
 
S
,
Arredondo Sancristobal
 
MJ
 et al.  
A high-protein diet promotes atrial arrhythmogenesis via absent-in-melanoma 2 inflammasome
.
Cells
 
2024
;
13
:
108
.

29

Hulsmans
 
M
,
Schloss
 
MJ
,
Lee
 
IH
,
Bapat
 
A
,
Iwamoto
 
Y
,
Vinegoni
 
C
 et al.  
Recruited macrophages elicit atrial fibrillation
.
Science
 
2023
;
381
:
231
9
.

30

Huynh
 
K
.
Novel macrophage targets for the treatment of atrial fibrillation
.
Nat Rev Cardiol
 
2023
;
20
:
648
.

31

Ninni
 
S
,
Dombrowicz
 
D
,
de Winther
 
M
,
Staels
 
B
,
Montaigne
 
D
,
Nattel
 
S
.
Inherited and acquired genetic factors altering innate immune responses: an emerging actor in atrial fibrillation
.
J Am Coll Cardiol
 
2024
;
83
:
1163
76
.

32

Hiram
 
R
,
Xiong
 
F
,
Naud
 
P
,
Xiao
 
J
,
Sirois
 
M
,
Tanguay
 
JF
 et al.  
The inflammation-resolution promoting molecule resolvin-D1 prevents atrial proarrhythmic remodelling in experimental right heart disease
.
Cardiovasc Res
 
2021
;
117
:
1776
89
.

33

Hiram
 
R
,
Xiong
 
F
,
Naud
 
P
,
Xiao
 
J
,
Sosnowski
 
DK
,
Saljic
 
A
 et al.  
An inflammation resolution-promoting intervention prevents atrial fibrillation due to left-ventricular dysfunction
.
Cardiovasc Res
.
2024
;
120
:
345
59
.

34

Gawałko
 
M
,
Agbaedeng
 
TA
,
Saljic
 
A
,
Müller
 
DN
,
Wilck
 
N
,
Schnabel
 
R
 et al.  
Gut microbiota, dysbiosis and atrial fibrillation. Arrhythmogenic mechanisms and potential clinical implications
.
Cardiovasc Res
 
2021
;
118
:
2415
27
.

35

Boutaud
 
O
,
Montine
 
TJ
,
Chang
 
L
,
Klein
 
WL
,
Oates
 
JA
.
PGH2-derived levuglandin adducts increase the neurotoxicity of amyloid beta1–42
.
J Neurochem
 
2006
;
96
:
917
23
.

36

Davies
 
SS
,
May-Zhang
 
LS
,
Boutaud
 
O
,
Amarnath
 
V
,
Kirabo
 
A
,
Harrison
 
DG
.
Isolevuglandins as mediators of disease and the development of dicarbonyl scavengers as pharmaceutical interventions
.
Pharmacol Ther
 
2020
;
205
:
107418
.

37

May-Zhang
 
LS
,
Kirabo
 
A
,
Huang
 
J
,
Linton
 
MF
,
Davies
 
SS
,
Murray
 
KT
.
Scavenging reactive lipids to prevent oxidative injury
.
Annu Rev Pharmacol Toxicol
 
2021
;
61
:
291
308
.

38

Pitchford
 
LM
,
Driver
 
PM
,
Fuller
 
JC
, Jr.
,
Akers
 
WS
,
Abumrad
 
NN
,
Amarnath
 
V
 et al.  
Safety, tolerability, and pharmacokinetics of repeated oral doses of 2-hydroxybenzylamine acetate in healthy volunteers: a double-blind, randomized, placebo-controlled clinical trial
.
BMC Pharmacol Toxicol
 
2020
;
21
:
3
.

39

Pitchford
 
LM
,
Rathmacher
 
JA
,
Fuller
 
JC
, Jr.
,
Daniels
 
JS
,
Morrison
 
RD
,
Akers
 
WS
 et al.  
First-in-human study assessing safety, tolerability, and pharmacokinetics of 2-hydroxybenzylamine acetate, a selective dicarbonyl electrophile scavenger, in healthy volunteers
.
BMC Pharmacol Toxicol
 
2019
;
20
:
1
.

40

Wong
 
CX
,
Abed
 
HS
,
Molaee
 
P
,
Nelson
 
AJ
,
Brooks
 
AG
,
Sharma
 
G
 et al.  
Pericardial fat is associated with atrial fibrillation severity and ablation outcome
.
J Am Coll Cardiol
 
2011
;
57
:
1745
51
.

41

Al Chekakie
 
MO
,
Welles
 
CC
,
Metoyer
 
R
,
Ibrahim
 
A
,
Shapira
 
AR
,
Cytron
 
J
 et al.  
Pericardial fat is independently associated with human atrial fibrillation
.
J Am Coll Cardiol
 
2010
;
56
:
784
8
.

42

Tsao
 
HM
,
Hu
 
WC
,
Wu
 
MH
,
Tai
 
CT
,
Lin
 
YJ
,
Chang
 
SL
 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
503
.

43

Su
 
KN
,
Ma
 
Y
,
Cacheux
 
M
,
Ilkan
 
Z
,
Raad
 
N
,
Muller
 
GK
 et al.  
Atrial AMP-activated protein kinase is critical for prevention of dysregulation of electrical excitability and atrial fibrillation
.
JCI insight
 
2022
;
7
:
e141213
.

44

Hulsurkar
 
MM
,
Lahiri
 
SK
,
Moore
 
O
,
Moreira
 
LM
,
Abu-Taha
 
I
,
Kamler
 
M
 et al.  
Atrial-specific LKB1 knockdown represents a novel mouse model of atrial cardiomyopathy with spontaneous atrial fibrillation
.
Circulation
 
2021
;
144
:
909
12
.

45

Kim
 
GE
,
Ross
 
JL
,
Xie
 
C
,
Su
 
KN
,
Zaha
 
VG
,
Wu
 
X
 et al.  
LKB1 deletion causes early changes in atrial channel expression and electrophysiology prior to atrial fibrillation
.
Cardiovasc Res
 
2015
;
108
:
197
208
.

46

Perike
 
S
,
Gonzalez-Gonzalez
 
FJ
,
Abu-Taha
 
I
,
Damen
 
FW
,
Hanft
 
LM
,
Lizama
 
KS
 et al.  
PPP1R12C promotes atrial hypocontractility in atrial fibrillation
.
Circ Res
 
2023
;
133
:
758
71
.

47

Rommel
 
C
,
Rösner
 
S
,
Lother
 
A
,
Barg
 
M
,
Schwaderer
 
M
,
Gilsbach
 
R
 et al.  
The transcription factor ETV1 induces atrial remodeling and arrhythmia
.
Circ Res
 
2018
;
123
:
550
63
.

48

Iacobellis
 
G
.
Epicardial adipose tissue in contemporary cardiology
.
Nat Rev Cardiol
 
2022
;
19
:
593
606
.

49

Gan
 
L
,
Xie
 
D
,
Liu
 
J
,
Bond Lau
 
W
,
Christopher
 
TA
,
Lopez
 
B
 et al.  
Small extracellular microvesicles mediated pathological communications between dysfunctional adipocytes and cardiomyocytes as a novel mechanism exacerbating ischemia/reperfusion injury in diabetic mice
.
Circulation
 
2020
;
141
:
968
83
.

50

Gómez-Hurtado
 
N
,
Domínguez-Rodríguez
 
A
,
Mateo
 
P
,
Fernández-Velasco
 
M
,
Val-Blasco
 
A
,
Aizpún
 
R
 et al.  
Beneficial effects of leptin treatment in a setting of cardiac dysfunction induced by transverse aortic constriction in mouse
.
J Physiol
 
2017
;
595
:
4227
43
.

51

Zhao
 
Y
,
Sun
 
Q
,
Zeng
 
Z
,
Li
 
Q
,
Zhou
 
S
,
Zhou
 
M
 et al.  
Regulation of SCN3B/scn3b by interleukin 2 (IL-2): IL-2 modulates SCN3B/scn3b transcript expression and increases sodium current in myocardial cells
.
BMC Cardiovasc Disord
 
2016
;
16
:
1
.

52

Ernault
 
AC
,
Meijborg
 
VMF
,
Coronel
 
R
.
Modulation of cardiac arrhythmogenesis by epicardial adipose tissue: JACC state-of-the-art review
.
J Am Coll Cardiol
 
2021
;
78
:
1730
45
.

53

Mazurek
 
T
,
Zhang
 
L
,
Zalewski
 
A
,
Mannion
 
JD
,
Diehl
 
JT
,
Arafat
 
H
 et al.  
Human epicardial adipose tissue is a source of inflammatory mediators
.
Circulation
 
2003
;
108
:
2460
6
.

54

Venteclef
 
N
,
Guglielmi
 
V
,
Balse
 
E
,
Gaborit
 
B
,
Cotillard
 
A
,
Atassi
 
F
 et al.  
Human epicardial adipose tissue induces fibrosis of the atrial myocardium through the secretion of adipo-fibrokines
.
Eur Heart J
 
2015
;
36
:
795
805
.

55

Wang
 
Q
,
Yu
 
Y
,
Zhang
 
P
,
Chen
 
Y
,
Li
 
C
,
Chen
 
J
 et al.  
The crucial role of activin A/ALK4 pathway in the pathogenesis of Ang-II-induced atrial fibrosis and vulnerability to atrial fibrillation
.
Basic Res Cardiol
 
2017
;
112
:
47
.

56

Haemers
 
P
,
Hamdi
 
H
,
Guedj
 
K
,
Suffee
 
N
,
Farahmand
 
P
,
Popovic
 
N
 et al.  
Atrial fibrillation is associated with the fibrotic remodelling of adipose tissue in the subepicardium of human and sheep atria
.
Eur Heart J
 
2017
;
38
:
53
61
.

57

Chaumont
 
C
,
Suffee
 
N
,
Gandjbakhch
 
E
,
Balse
 
E
,
Anselme
 
F
,
Hatem
 
SN
.
Epicardial origin of cardiac arrhythmias: clinical evidences and pathophysiology
.
Cardiovasc Res
 
2022
;
118
:
1693
702
.

58

Tarsitano
 
MG
,
Pandozzi
 
C
,
Muscogiuri
 
G
,
Sironi
 
S
,
Pujia
 
A
,
Lenzi
 
A
 et al.  
Epicardial adipose tissue: a novel potential imaging marker of comorbidities caused by chronic inflammation
.
Nutrients
 
2022
;
14
:
2926
.

59

Abe
 
I
,
Teshima
 
Y
,
Kondo
 
H
,
Kaku
 
H
,
Kira
 
S
,
Ikebe
 
Y
 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
27
.

60

Ishii
 
Y
,
Abe
 
I
,
Kira
 
S
,
Harada
 
T
,
Takano
 
M
,
Oniki
 
T
 et al.  
Detection of fibrotic remodeling of epicardial adipose tissue in patients with atrial fibrillation: imaging approach based on histological observation
.
Heart Rhythm O2
 
2021
;
2
:
311
23
.

61

Ravelli
 
F
,
Masè
 
M
,
Cristoforetti
 
A
,
Avogaro
 
L
,
D'Amato
 
E
,
Tessarolo
 
F
 et al.  
Quantitative assessment of transmural fibrosis profile in the human atrium: evidence for a three-dimensional arrhythmic substrate by slice-to-slice histology
.
Europace
 
2023
;
25
:
739
47
.

62

Gharaviri
 
A
,
Bidar
 
E
,
Potse
 
M
,
Zeemering
 
S
,
Verheule
 
S
,
Pezzuto
 
S
 et al.  
Epicardial fibrosis explains increased endo-epicardial dissociation and epicardial breakthroughs in human atrial fibrillation
.
Front Physiol
 
2020
;
11
:
68
.

63

Nalliah
 
CJ
,
Bell
 
JR
,
Raaijmakers
 
AJA
,
Waddell
 
HM
,
Wells
 
SP
,
Bernasochi
 
GB
 et al.  
Epicardial adipose tissue accumulation confers atrial conduction abnormality
.
J Am Coll Cardiol
 
2020
;
76
:
1197
211
.

64

De Coster
 
T
,
Claus
 
P
,
Kazbanov
 
IV
,
Haemers
 
P
,
Willems
 
R
,
Sipido
 
KR
 et al.  
Arrhythmogenicity of fibro-fatty infiltrations
.
Sci Rep
 
2018
;
8
:
2050
.

65

Zhou
 
B
,
Ma
 
Q
,
Rajagopal
 
S
,
Wu
 
SM
,
Domian
 
I
,
Rivera-Feliciano
 
J
 et al.  
Epicardial progenitors contribute to the cardiomyocyte lineage in the developing heart
.
Nature
 
2008
;
454
:
109
13
.

66

Yamaguchi
 
Y
,
Cavallero
 
S
,
Patterson
 
M
,
Shen
 
H
,
Xu
 
J
,
Kumar
 
SR
 et al.  
Adipogenesis and epicardial adipose tissue: a novel fate of the epicardium induced by mesenchymal transformation and PPARγ activation
.
Proc Natl Acad Sci U S A
 
2015
;
112
:
2070
5
.

67

Suffee
 
N
,
Moore-Morris
 
T
,
Farahmand
 
P
,
Rücker-Martin
 
C
,
Dilanian
 
G
,
Fradet
 
M
 et al.  
Atrial natriuretic peptide regulates adipose tissue accumulation in adult atria
.
Proc Natl Acad Sci U S A
 
2017
;
114
:
E771
e80
.

68

Suffee
 
N
,
Moore-Morris
 
T
,
Jagla
 
B
,
Mougenot
 
N
,
Dilanian
 
G
,
Berthet
 
M
 et al.  
Reactivation of the epicardium at the origin of myocardial fibro-fatty infiltration during the atrial cardiomyopathy
.
Circ Res
 
2020
;
126
:
1330
42
.

69

Suffee
 
N
,
Baptista
 
E
,
Piquereau
 
J
,
Ponnaiah
 
M
,
Doisne
 
N
,
Ichou
 
F
 et al.  
Impacts of a high-fat diet on the metabolic profile and the phenotype of atrial myocardium in mice
.
Cardiovasc Res
 
2022
;
118
:
3126
39
.

70

Shen
 
MJ
,
Arora
 
R
,
Jalife
 
J
.
Atrial myopathy
.
JACC Basic Transl Sci
 
2019
;
4
:
640
54
.

71

January
 
CT
,
Wann
 
LS
,
Alpert
 
JS
,
Calkins
 
H
,
Cigarroa
 
JE
,
Cleveland
 
JC
, Jr.
 et al.  
2014 AHA/ACC/HRS guideline for the management of patients with atrial fibrillation: a report of the American College of Cardiology/American Heart Association Task Force on practice guidelines and the Heart Rhythm Society
.
Circulation
 
2014
;
130
:
e199
267
.

72

Kirchhof
 
P
,
Benussi
 
S
,
Kotecha
 
D
,
Ahlsson
 
A
,
Atar
 
D
,
Casadei
 
B
 et al.  
2016 ESC guidelines for the management of atrial fibrillation developed in collaboration with EACTS
.
Eur Heart J
 
2016
;
37
:
2893
962
.

73

Andrade
 
JG
,
Aguilar
 
M
,
Atzema
 
C
,
Bell
 
A
,
Cairns
 
JA
,
Cheung
 
CC
 et al.  
The 2020 Canadian Cardiovascular Society/Canadian Heart Rhythm Society Comprehensive Guidelines for the Management of Atrial Fibrillation
.
Can J Cardiol
 
2020
;
36
:
1847
948
.

74

Daoud
 
EG
,
Glotzer
 
TV
,
Wyse
 
DG
,
Ezekowitz
 
MD
,
Hilker
 
C
,
Koehler
 
J
 et al.  
Temporal relationship of atrial tachyarrhythmias, cerebrovascular events, and systemic emboli based on stored device data: a subgroup analysis of TRENDS
.
Heart Rhythm
 
2011
;
8
:
1416
23
.

75

Martin
 
DT
,
Bersohn
 
MM
,
Waldo
 
AL
,
Wathen
 
MS
,
Choucair
 
WK
,
Lip
 
GY
 et al.  
Randomized trial of atrial arrhythmia monitoring to guide anticoagulation in patients with implanted defibrillator and cardiac resynchronization devices
.
Eur Heart J
 
2015
;
36
:
1660
8
.

76

Kamel
 
H
,
Longstreth
 
WT
, Jr.
,
Tirschwell
 
DL
,
Kronmal
 
RA
,
Marshall
 
RS
,
Broderick
 
JP
 et al.  
Apixaban to prevent recurrence after cryptogenic stroke in patients with atrial cardiopathy: the ARCADIA randomized clinical trial
.
JAMA
 
2024
;
331
:
573
81
.

77

Watson
 
T
,
Shantsila
 
E
,
Lip
 
GY
.
Mechanisms of thrombogenesis in atrial fibrillation: Virchow's triad revisited
.
Lancet
 
2009
;
373
:
155
66
.

78

Takahashi
 
Y
,
Yamaguchi
 
T
,
Otsubo
 
T
,
Nakashima
 
K
,
Shinzato
 
K
,
Osako
 
R
 et al.  
Histological validation of atrial structural remodelling in patients with atrial fibrillation
.
Eur Heart J
 
2023
;
44
:
3339
53
.

79

Kudryavtseva
 
AV
,
Krasnov
 
GS
,
Dmitriev
 
AA
,
Alekseev
 
BY
,
Kardymon
 
OL
,
Sadritdinova
 
AF
 et al.  
Mitochondrial dysfunction and oxidative stress in aging and cancer
.
Oncotarget
 
2016
;
7
:
44879
905
.

80

Unni
 
PA
,
Pillai
 
GG
,
Sajithalulu
 
S
.
Biological processes and key druggable targets involved in age-associated memory loss: a systematic review
.
Life Sci
 
2021
;
270
:
119079
.

81

Mehdizadeh
 
M
,
Aguilar
 
M
,
Thorin
 
E
,
Ferbeyre
 
G
,
Nattel
 
S
.
The role of cellular senescence in cardiac disease: basic biology and clinical relevance
.
Nat Rev Cardiol
 
2022
;
19
:
250
64
.

82

Chung
 
HY
,
Cesari
 
M
,
Anton
 
S
,
Marzetti
 
E
,
Giovannini
 
S
,
Seo
 
AY
 et al.  
Molecular inflammation: underpinnings of aging and age-related diseases
.
Ageing Res Rev
 
2009
;
8
:
18
30
.

83

Kalstad
 
AA
,
Myhre
 
PL
,
Laake
 
K
,
Opstad
 
TB
,
Tveit
 
A
,
Solheim
 
S
 et al.  
Biomarkers of ageing and cardiac remodeling are associated with atrial fibrillation
.
Scand Cardiovasc J
 
2021
;
55
:
213
9
.

84

Ramos
 
KS
,
Brundel
 
B
.
DNA damage, an innocent bystander in atrial fibrillation and other cardiovascular diseases?
 
Front Cardiovasc Med
 
2020
;
7
:
67
.

85

Adili
 
A
,
Zhu
 
X
,
Cao
 
H
,
Tang
 
X
,
Wang
 
Y
,
Wang
 
J
 et al.  
Atrial fibrillation underlies cardiomyocyte senescence and contributes to deleterious atrial remodeling during disease progression
.
Aging Dis
 
2022
;
13
:
298
312
.

86

Rahman
 
F
,
Kwan
 
GF
,
Benjamin
 
EJ
.
Global epidemiology of atrial fibrillation
.
Nat Rev Cardiol
 
2014
;
11
:
639
54
.

87

Chen
 
M
,
Li
 
C
,
Liao
 
P
,
Cui
 
X
,
Tian
 
W
,
Wang
 
Q
 et al.  
Epidemiology, management, and outcomes of atrial fibrillation among 30 million citizens in Shanghai, China from 2015 to 2020: a medical insurance database study
.
Lancet Reg Health West Pac
 
2022
;
23
:
100470
.

88

Westaby
 
JD
,
Zullo
 
E
,
Bicalho
 
LM
,
Anderson
 
RH
,
Sheppard
 
MN
.
Effect of sex, age and body measurements on heart weight, atrial, ventricular, valvular and sub-epicardial fat measurements of the normal heart
.
Cardiovasc Pathol
 
2023
;
63
:
107508
.

89

Fenger-Grøn
 
M
,
Overvad
 
K
,
Tjønneland
 
A
,
Frost
 
L
.
Lean body mass is the predominant anthropometric risk factor for atrial fibrillation
.
J Am Coll Cardiol
 
2017
;
69
:
2488
97
.

90

Emdin
 
CA
,
Wong
 
CX
,
Hsiao
 
AJ
,
Altman
 
DG
,
Peters
 
SA
,
Woodward
 
M
 et al.  
Atrial fibrillation as risk factor for cardiovascular disease and death in women compared with men: systematic review and meta-analysis of cohort studies
.
Bmj
 
2016
;
532
:
h7013
.

91

Volgman
 
AS
,
Benjamin
 
EJ
,
Curtis
 
AB
,
Fang
 
MC
,
Lindley
 
KJ
,
Naccarelli
 
GV
 et al.  
Women and atrial fibrillation
.
J Cardiovasc Electrophysiol
 
2021
;
32
:
2793
807
.

92

Cheng
 
X
,
Hu
 
Q
,
Gao
 
L
,
Liu
 
J
,
Qin
 
S
,
Zhang
 
D
.
Sex-related differences in catheter ablation of atrial fibrillation: a systematic review and meta-analysis
.
Europace
 
2019
;
21
:
1509
18
.

93

Kuck
 
KH
,
Brugada
 
J
,
Fürnkranz
 
A
,
Chun
 
KRJ
,
Metzner
 
A
,
Ouyang
 
F
 et al.  
Impact of female sex on clinical outcomes in the FIRE AND ICE trial of catheter ablation for atrial fibrillation
.
Circ Arrhythm Electrophysiol
 
2018
;
11
:
e006204
.

94

Cheung
 
JW
,
Cheng
 
EP
,
Wu
 
X
,
Yeo
 
I
,
Christos
 
PJ
,
Kamel
 
H
 et al.  
Sex-based differences in outcomes, 30–day readmissions, and costs following catheter ablation of atrial fibrillation: the United States Nationwide Readmissions Database 2010–14
.
Eur Heart J.
 
2019
;
40
:
3035
43
.

95

Wang
 
L
,
Selzman
 
KA
,
Shah
 
RU
.
Peri-procedural complications in women: an alarming and consistent trend
.
Eur Heart J
 
2019
;
40
:
3044
5
.

96

Wong
 
GR
,
Nalliah
 
CJ
,
Lee
 
G
,
Voskoboinik
 
A
,
Chieng
 
D
,
Prabhu
 
S
 et al.  
Sex-related differences in atrial remodeling in patients with atrial fibrillation: relationship to ablation outcomes
.
Circ Arrhythm Electrophysiol
 
2022
;
15
:
e009925
.

97

Müller-Edenborn
 
B
,
Moreno-Weidmann
 
Z
,
Venier
 
S
,
Defaye
 
P
,
Park
 
CI
,
Guerra
 
J
 et al.  
Determinants of fibrotic atrial cardiomyopathy in atrial fibrillation. A multicenter observational study of the RETAC (reseau européen de traîtement d'arrhythmies cardiaques)-group
.
Clin Res Cardiol
 
2022
;
111
:
1018
27
.

98

Rosano
 
GM
,
Leonardo
 
F
,
Dicandia
 
C
,
Sheiban
 
I
,
Pagnotta
 
P
,
Pappone
 
C
 et al.  
Acute electrophysiologic effect of estradiol 17beta in menopausal women
.
Am J Cardiol
 
2000
;
86
:
1385
7
.
A5-6
.

99

Tadros
 
R
,
Ton
 
AT
,
Fiset
 
C
,
Nattel
 
S
.
Sex differences in cardiac electrophysiology and clinical arrhythmias: epidemiology, therapeutics, and mechanisms
.
Can J Cardiol
 
2014
;
30
:
783
92
.

100

Li
 
Z
,
Wang
 
Z
,
Yin
 
Z
,
Zhang
 
Y
,
Xue
 
X
,
Han
 
J
 et al.  
Gender differences in fibrosis remodeling in patients with long-standing persistent atrial fibrillation
.
Oncotarget
 
2017
;
8
:
53714
29
.

101

Cochet
 
H
,
Mouries
 
A
,
Nivet
 
H
,
Sacher
 
F
,
Derval
 
N
,
Denis
 
A
 et al.  
Age, atrial fibrillation, and structural heart disease are the main determinants of left atrial fibrosis detected by delayed-enhanced magnetic resonance imaging in a general cardiology population
.
J Cardiovasc Electrophysiol
 
2015
;
26
:
484
92
.

102

Bisbal
 
F
,
Baranchuk
 
A
,
Braunwald
 
E
,
Bayés de Luna
 
A
,
Bayés-Genís
 
A
.
Atrial failure as a clinical entity: JACC review topic of the week
.
J Am Coll Cardiol
 
2020
;
75
:
222
32
.

103

Thomas
 
L
,
Abhayaratna
 
WP
.
Left atrial reverse remodeling: mechanisms, evaluation, and clinical significance
.
JACC Cardiovasc imaging
 
2017
;
10
:
65
77
.

104

Maron
 
BJ
,
Towbin
 
JA
,
Thiene
 
G
,
Antzelevitch
 
C
,
Corrado
 
D
,
Arnett
 
D
 et al.  
Contemporary definitions and classification of the cardiomyopathies: an American Heart Association Scientific Statement from the Council on Clinical Cardiology, Heart Failure and Transplantation Committee; Quality of Care and Outcomes Research and Functional Genomics and Translational Biology Interdisciplinary Working Groups; and Council on Epidemiology and Prevention
.
Circulation
 
2006
;
113
:
1807
16
.

105

Corradi
 
D
,
Callegari
 
S
,
Maestri
 
R
,
Benussi
 
S
,
Alfieri
 
O
.
Structural remodeling in atrial fibrillation
.
Nat Clin Pract Cardiovas Med
 
2008
;
5
:
782
96
.

106

Kühl
 
U
,
Pauschinger
 
M
,
Noutsias
 
M
,
Seeberg
 
B
,
Bock
 
T
,
Lassner
 
D
 et al.  
High prevalence of viral genomes and multiple viral infections in the myocardium of adults with “idiopathic” left ventricular dysfunction
.
Circulation
 
2005
;
111
:
887
93
.

107

Gibson
 
DN
,
Di Biase
 
L
,
Mohanty
 
P
,
Patel
 
JD
,
Bai
 
R
,
Sanchez
 
J
 et al.  
Stiff left atrial syndrome after catheter ablation for atrial fibrillation: clinical characterization, prevalence, and predictors
.
Heart Rhythm
 
2011
;
8
:
1364
71
.

108

Enríquez-Vázquez
 
D
,
Quintanilla
 
JG
,
García-Escolano
 
A
,
Couselo-Seijas
 
M
,
Simón-Chica
 
A
,
Lee
 
P
 et al.  
Non-invasive electromechanical assessment during atrial fibrillation identifies underlying atrial myopathy alterations with early prognostic value
.
Nat Commun
 
2023
;
14
:
4613
.

109

Pessoa-Amorim
 
G
,
Mancio
 
J
,
Vouga
 
L
,
Ribeiro
 
J
,
Gama
 
V
,
Bettencourt
 
N
 et al.  
Impaired left atrial strain as a predictor of new-onset atrial fibrillation after aortic valve replacement independently of left atrial size
.
Rev Esp Cardiol (Engl Ed)
 
2018
;
71
:
466
76
.

110

Zakeri
 
R
,
Moulay
 
G
,
Chai
 
Q
,
Ogut
 
O
,
Hussain
 
S
,
Takahama
 
H
 et al.  
Left atrial remodeling and atrioventricular coupling in a canine model of early heart failure with preserved ejection fraction
.
Circ Heart Fail
 
2016
;
9
:
e003238
.

111

Habibi
 
M
,
Zareian
 
M
,
Ambale Venkatesh
 
B
,
Samiei
 
S
,
Imai
 
M
,
Wu
 
C
 et al.  
Left atrial mechanical function and incident ischemic cerebrovascular events independent of AF: insights from the MESA study
.
JACC Cardiovasc imaging
 
2019
;
12
:
2417
27
.

112

Eicher
 
JC
,
Laurent
 
G
,
Mathé
 
A
,
Barthez
 
O
,
Bertaux
 
G
,
Philip
 
JL
 et al.  
Atrial dyssynchrony syndrome: an overlooked phenomenon and a potential cause of ‘diastolic’ heart failure
.
Eur J Heart Fail
 
2012
;
14
:
248
58
.

113

You
 
JJ
,
Singer
 
DE
,
Howard
 
PA
,
Lane
 
DA
,
Eckman
 
MH
,
Fang
 
MC
 et al.  
Antithrombotic therapy for atrial fibrillation: antithrombotic therapy and prevention of thrombosis, 9th ed: American college of chest physicians evidence-based clinical practice guidelines
.
Chest.
 
2012
;
141
:
e531S
75S
.

114

Weigner
 
MJ
,
Caulfield
 
TA
,
Danias
 
PG
,
Silverman
 
DI
,
Manning
 
WJ
.
Risk for clinical thromboembolism associated with conversion to sinus rhythm in patients with atrial fibrillation lasting less than 48 hours
.
Ann Intern Med
 
1997
;
126
:
615
20
.

115

Brambatti
 
M
,
Connolly
 
SJ
,
Gold
 
MR
,
Morillo
 
CA
,
Capucci
 
A
,
Muto
 
C
 et al.  
Temporal relationship between subclinical atrial fibrillation and embolic events
.
Circulation
 
2014
;
129
:
2094
9
.

116

Fonseca
 
AC
,
Alves
 
P
,
Inácio
 
N
,
Marto
 
JP
,
Viana-Baptista
 
M
,
Pinho
 
EMT
 et al.  
Patients with undetermined stroke have increased atrial fibrosis: a cardiac magnetic resonance imaging study
.
Stroke
 
2018
;
49
:
734
7
.

117

Tandon
 
K
,
Tirschwell
 
D
,
Longstreth
 
WT
, Jr.
,
Smith
 
B
,
Akoum
 
N
.
Embolic stroke of undetermined source correlates to atrial fibrosis without atrial fibrillation
.
Neurology
 
2019
;
93
:
e381
7
.

118

Kühnlein
 
P
,
Mahnkopf
 
C
,
Majersik
 
JJ
,
Wilson
 
BD
,
Mitlacher
 
M
,
Tirschwell
 
D
 et al.  
Atrial fibrosis in embolic stroke of undetermined source: a multicenter study
.
Eur J Neurol
 
2021
;
28
:
3634
9
.

119

Nattel
 
S
.
Atrial fibrosis, endocardial damage, and thrombosis in atrial fibrillation: association with underlying conditions or causal?
 
JACC Clin Electrophysiol
 
2023
;
9
:
1169
71
.

120

Müller-Edenborn
 
B
,
Minners
 
J
,
Keyl
 
C
,
Eichenlaub
 
M
,
Jander
 
N
,
Abdelrazek
 
S
 et al.  
Electrocardiographic diagnosis of atrial cardiomyopathy to predict atrial contractile dysfunction, thrombogenesis and adverse cardiovascular outcomes
.
Sci Rep
 
2022
;
12
:
576
.

121

Lee
 
HY
,
Yang
 
PS
,
Kim
 
TH
,
Uhm
 
JS
,
Pak
 
HN
,
Lee
 
MH
 et al.  
Atrial fibrillation and the risk of myocardial infarction: a nation-wide propensity-matched study
.
Sci Rep
 
2017
;
7
:
12716
.

122

Goette
 
A
,
Borof
 
K
,
Breithardt
 
G
,
Camm
 
AJ
,
Crijns
 
H
,
Kuck
 
KH
 et al.  
Presenting pattern of atrial fibrillation and outcomes of early rhythm control therapy
.
J Am Coll Cardiol
 
2022
;
80
:
283
95
.

123

Goette
 
A
,
Bukowska
 
A
,
Dobrev
 
D
,
Pfeiffenberger
 
J
,
Morawietz
 
H
,
Strugala
 
D
 et al.  
Acute atrial tachyarrhythmia induces angiotensin II type 1 receptor-mediated oxidative stress and microvascular flow abnormalities in the ventricles
.
Eur Heart J
 
2009
;
30
:
1411
20
.

124

Shibata
 
T
,
Kawakami
 
S
,
Noguchi
 
T
,
Tanaka
 
T
,
Asaumi
 
Y
,
Kanaya
 
T
 et al.  
Prevalence, clinical features, and prognosis of acute myocardial infarction attributable to coronary artery embolism
.
Circulation
 
2015
;
132
:
241
50
.

125

Mueller
 
KAL
,
Heinzmann
 
D
,
Klingel
 
K
,
Fallier-Becker
 
P
,
Kandolf
 
R
,
Kilias
 
A
 et al.  
Histopathological and immunological characteristics of tachycardia-induced cardiomyopathy
.
J Am Coll Cardiol
 
2017
;
69
:
2160
72
.

126

Ortiz-Leon
 
XA
,
Posada-Martinez
 
EL
,
Trejo-Paredes
 
MC
,
Ivey-Miranda
 
JB
,
Pereira
 
J
,
Crandall
 
I
 et al.  
Understanding tricuspid valve remodelling in atrial fibrillation using three-dimensional echocardiography
.
Eur Heart J Cardiovasc Imaging
 
2020
;
21
:
747
55
.

127

Kwak
 
S
,
Lim
 
J
,
Yang
 
S
,
Rhee
 
TM
,
Choi
 
YJ
,
Lee
 
HJ
 et al.  
Atrial functional tricuspid regurgitation: importance of atrial fibrillation and right atrial remodeling and prognostic significance
.
JACC Cardiovasc imaging
 
2023
;
16
:
575
87
.

128

Arndt
 
M
,
Lendeckel
 
U
,
Röcken
 
C
,
Nepple
 
K
,
Wolke
 
C
,
Spiess
 
A
 et al.  
Altered expression of ADAMs (a disintegrin and metalloproteinase) in fibrillating human atria
.
Circulation
 
2002
;
105
:
720
5
.

129

Guta
 
AC
,
Badano
 
LP
,
Tomaselli
 
M
,
Mihalcea
 
D
,
Bartos
 
D
,
Parati
 
G
 et al.  
The pathophysiological link between right atrial remodeling and functional tricuspid regurgitation in patients with atrial fibrillation: a three-dimensional echocardiography study
.
J Am Soc Echocardiogr
 
2021
;
34
:
585
94.e1
.

130

Levine
 
RA
,
Schwammenthal
 
E
.
Ischemic mitral regurgitation on the threshold of a solution: from paradoxes to unifying concepts
.
Circulation
 
2005
;
112
:
745
58
.

131

Zoghbi
 
WA
,
Levine
 
RA
,
Flachskampf
 
F
,
Grayburn
 
P
,
Gillam
 
L
,
Leipsic
 
J
 et al.  
Atrial functional mitral regurgitation: a JACC: cardiovascular imaging expert panel viewpoint
.
JACC Cardiovasc imaging
 
2022
;
15
:
1870
82
.

132

Deferm
 
S
,
Bertrand
 
PB
,
Verbrugge
 
FH
,
Verhaert
 
D
,
Rega
 
F
,
Thomas
 
JD
 et al.  
Atrial functional mitral regurgitation: JACC review topic of the week
.
J Am Coll Cardiol
 
2019
;
73
:
2465
76
.

133

Gertz
 
ZM
,
Raina
 
A
,
Saghy
 
L
,
Zado
 
ES
,
Callans
 
DJ
,
Marchlinski
 
FE
 et al.  
Evidence of atrial functional mitral regurgitation due to atrial fibrillation: reversal with arrhythmia control
.
J Am Coll Cardiol
 
2011
;
58
:
1474
81
.

134

Tamargo
 
M
,
Obokata
 
M
,
Reddy
 
YNV
,
Pislaru
 
SV
,
Lin
 
G
,
Egbe
 
AC
 et al.  
Functional mitral regurgitation and left atrial myopathy in heart failure with preserved ejection fraction
.
Eur J Heart Fail
 
2020
;
22
:
489
98
.

135

Dziadzko
 
V
,
Dziadzko
 
M
,
Medina-Inojosa
 
JR
,
Benfari
 
G
,
Michelena
 
HI
,
Crestanello
 
JA
 et al.  
Causes and mechanisms of isolated mitral regurgitation in the community: clinical context and outcome
.
Eur Heart J
 
2019
;
40
:
2194
202
.

136

Reddy
 
YNV
,
Obokata
 
M
,
Gersh
 
BJ
,
Borlaug
 
BA
.
High prevalence of occult heart failure with preserved ejection fraction among patients with atrial fibrillation and dyspnea
.
Circulation
 
2018
;
137
:
534
5
.

137

Patel
 
RB
,
Shah
 
SJ
.
Therapeutic targeting of left atrial myopathy in atrial fibrillation and heart failure with preserved ejection fraction
.
JAMA Cardiol
 
2020
;
5
:
497
9
.

138

Reddy
 
YNV
,
Obokata
 
M
,
Verbrugge
 
FH
,
Lin
 
G
,
Borlaug
 
BA
.
Atrial dysfunction in patients with heart failure with preserved ejection fraction and atrial fibrillation
.
J Am Coll Cardiol
 
2020
;
76
:
1051
64
.

139

Packer
 
M
,
Lam
 
CSP
,
Lund
 
LH
,
Redfield
 
MM
.
Interdependence of atrial fibrillation and heart failure with a preserved ejection fraction reflects a common underlying atrial and ventricular myopathy
.
Circulation
 
2020
;
141
:
4
6
.

140

Hindricks
 
G
,
Potpara
 
T
,
Dagres
 
N
,
Arbelo
 
E
,
Bax
 
JJ
,
Blomström-Lundqvist
 
C
 et al.  
2020 ESC guidelines for the diagnosis and management of atrial fibrillation developed in collaboration with the European Association for Cardio-Thoracic Surgery (EACTS): The Task Force for the diagnosis and management of atrial fibrillation of the European Society of Cardiology (ESC) developed with the special contribution of the European Heart Rhythm Association (EHRA) of the ESC
.
Eur Heart J
 
2021
;
42
:
373
498
.

141

Abed
 
HS
,
Wittert
 
GA
,
Leong
 
DP
,
Shirazi
 
MG
,
Bahrami
 
B
,
Middeldorp
 
ME
 et al.  
Effect of weight reduction and cardiometabolic risk factor management on symptom burden and severity in patients with atrial fibrillation: a randomized clinical trial
.
JAMA
 
2013
;
310
:
2050
60
.

142

Benjamin
 
EJ
,
Levy
 
D
,
Vaziri
 
SM
,
D'Agostino
 
RB
,
Belanger
 
AJ
,
Wolf
 
PA
.
Independent risk factors for atrial fibrillation in a population-based cohort: the Framingham heart study
.
JAMA
 
1994
;
271
:
840
4
.

143

Dublin
 
S
,
Glazer
 
NL
,
Smith
 
NL
,
Psaty
 
BM
,
Lumley
 
T
,
Wiggins
 
KL
 et al.  
Diabetes mellitus, glycemic control, and risk of atrial fibrillation
.
J Gen Intern Med
 
2010
;
25
:
853
8
.

144

Fenger-Grøn
 
M
,
Vinter
 
N
,
Frost
 
L
.
Body mass and atrial fibrillation risk: status of the epidemiology concerning the influence of fat versus lean body mass
.
Trends Cardiovasc Med
 
2020
;
30
:
205
11
.

145

Gong
 
M
,
Yuan
 
M
,
Meng
 
L
,
Zhang
 
Z
,
Tse
 
G
,
Zhao
 
Y
 et al.  
Wenxin Keli regulates mitochondrial oxidative stress and homeostasis and improves atrial remodeling in diabetic rats
.
Oxid Med Cell Longev
 
2020
;
2020
:
2468031
.

146

Aromolaran
 
AS
,
Colecraft
 
HM
,
Boutjdir
 
M
.
High-fat diet-dependent modulation of the delayed rectifier K(+) current in adult Guinea pig atrial myocytes
.
Biochem Biophys Res Commun
 
2016
;
474
:
554
9
.

147

Goldberg
 
IJ
,
Trent
 
CM
,
Schulze
 
PC
.
Lipid metabolism and toxicity in the heart
.
Cell Metab
 
2012
;
15
:
805
12
.

148

Vyas
 
V
,
Blythe
 
H
,
Wood
 
EG
,
Sandhar
 
B
,
Sarker
 
SJ
,
Balmforth
 
D
 et al.  
Obesity and diabetes are major risk factors for epicardial adipose tissue inflammation
.
JCI insight
 
2021
;
6
:
e145495
.

149

Guo
 
Y
,
Lip
 
GY
,
Apostolakis
 
S
.
Inflammation in atrial fibrillation
.
J Am Coll Cardiol
 
2012
;
60
:
2263
70
.

150

Smorodinova
 
N
,
Bláha
 
M
,
Melenovský
 
V
,
Rozsívalová
 
K
,
Přidal
 
J
,
Ďurišová
 
M
 et al.  
Analysis of immune cell populations in atrial myocardium of patients with atrial fibrillation or sinus rhythm
.
PLoS One
 
2017
;
12
:
e0172691
.

151

Nattel
 
S
,
Harada
 
M
.
Atrial remodeling and atrial fibrillation: recent advances and translational perspectives
.
J Am Coll Cardiol
 
2014
;
63
:
2335
45
.

152

Heijman
 
J
,
Voigt
 
N
,
Nattel
 
S
,
Dobrev
 
D
.
Cellular and molecular electrophysiology of atrial fibrillation initiation, maintenance, and progression
.
Circ Res
 
2014
;
114
:
1483
99
.

153

Andrade
 
JG
,
Deyell
 
MW
,
Macle
 
L
,
Wells
 
GA
,
Bennett
 
M
,
Essebag
 
V
 et al.  
Progression of atrial fibrillation after cryoablation or drug therapy
.
N Engl J Med
 
2023
;
388
:
105
16
.

154

Walters
 
TE
,
Nisbet
 
A
,
Morris
 
GM
,
Tan
 
G
,
Mearns
 
M
,
Teo
 
E
 et al.  
Progression of atrial remodeling in patients with high-burden atrial fibrillation: implications for early ablative intervention
.
Heart Rhythm
 
2016
;
13
:
331
9
.

155

Sugumar
 
H
,
Prabhu
 
S
,
Voskoboinik
 
A
,
Young
 
S
,
Gutman
 
SJ
,
Wong
 
GR
 et al.  
Atrial remodeling following catheter ablation for atrial fibrillation-mediated cardiomyopathy: long-term follow-up of CAMERA-MRI study
.
JACC Clin Electrophysiol
 
2019
;
5
:
681
8
.

156

Li
 
D
,
Fareh
 
S
,
Leung
 
TK
,
Nattel
 
S
.
Promotion of atrial fibrillation by heart failure in dogs: atrial remodeling of a different sort
.
Circulation
 
1999
;
100
:
87
95
.

157

Nattel
 
S
.
Molecular and cellular mechanisms of atrial fibrosis in atrial fibrillation
.
JACC Clin Electrophysiol
 
2017
;
3
:
425
35
.

158

Corradi
 
D
.
Atrial fibrillation from the pathologist's perspective
.
Cardiovasc Pathol
 
2014
;
23
:
71
84
.

159

Corradi
 
D
,
Callegari
 
S
,
Benussi
 
S
,
Nascimbene
 
S
,
Pastori
 
P
,
Calvi
 
S
 et al.  
Regional left atrial interstitial remodeling in patients with chronic atrial fibrillation undergoing mitral-valve surgery
.
Virchows Arch
 
2004
;
445
:
498
505
.

160

Teh
 
AW
,
Kistler
 
PM
,
Lee
 
G
,
Medi
 
C
,
Heck
 
PM
,
Spence
 
SJ
 et al.  
Electroanatomic remodeling of the left atrium in paroxysmal and persistent atrial fibrillation patients without structural heart disease
.
J Cardiovasc Electrophysiol
 
2012
;
23
:
232
8
.

161

Caixal
 
G
,
Althoff
 
T
,
Garre
 
P
,
Alarcón
 
F
,
NuñezGarcia
 
M
,
Benito
 
EM
 et al.  
Proximity to the descending aorta predicts regional fibrosis in the adjacent left atrial wall: aetiopathogenic and prognostic implications
.
Europace
 
2021
;
23
:
1559
67
.

162

Hopman
 
L
,
Bhagirath
 
P
,
Mulder
 
MJ
,
Eggink
 
IN
,
van Rossum
 
AC
,
Allaart
 
CP
 et al.  
Quantification of left atrial fibrosis by 3D late gadolinium-enhanced cardiac magnetic resonance imaging in patients with atrial fibrillation: impact of different analysis methods
.
Eur Heart J Cardiovasc Imaging
 
2022
;
23
:
1182
90
.

163

Callegari
 
S
,
Macchi
 
E
,
Monaco
 
R
,
Magnani
 
L
,
Tafuni
 
A
,
Croci
 
S
 et al.  
Clinicopathological bird's-eye view of left atrial myocardial fibrosis in 121 patients with persistent atrial fibrillation: developing architecture and main cellular players
.
Circ Arrhythm Electrophysiol
 
2020
;
13
:
e007588
.

164

Liu
 
Y
,
Niu
 
XH
,
Yin
 
X
,
Liu
 
YJ
,
Han
 
C
,
Yang
 
J
 et al.  
Elevated circulating fibrocytes is a marker of left atrial fibrosis and recurrence of persistent atrial fibrillation
.
J Am Heart Assoc
 
2018
;
7
:
e008083
.

165

Travers
 
JG
,
Kamal
 
FA
,
Robbins
 
J
,
Yutzey
 
KE
,
Blaxall
 
BC
.
Cardiac fibrosis: the fibroblast awakens
.
Circ Res
 
2016
;
118
:
1021
40
.

166

Frangogiannis
 
NG
.
Cardiac fibrosis
.
Cardiovasc Res
 
2021
;
117
:
1450
88
.

167

Yue
 
L
,
Xie
 
J
,
Nattel
 
S
.
Molecular determinants of cardiac fibroblast electrical function and therapeutic implications for atrial fibrillation
.
Cardiovasc Res
 
2011
;
89
:
744
53
.

168

Christensen
 
G
,
Herum
 
KM
,
Lunde
 
IG
.
Sweet, yet underappreciated: proteoglycans and extracellular matrix remodeling in heart disease
.
Matrix Biol
 
2019
;
75–76
:
286
99
.

169

Barallobre-Barreiro
 
J
,
Gupta
 
SK
,
Zoccarato
 
A
,
Kitazume-Taneike
 
R
,
Fava
 
M
,
Yin
 
X
 et al.  
Glycoproteomics reveals decorin peptides with anti-myostatin activity in human atrial fibrillation
.
Circulation
 
2016
;
134
:
817
32
.

170

Ausma
 
J
,
Wijffels
 
M
,
Thoné
 
F
,
Wouters
 
L
,
Allessie
 
M
,
Borgers
 
M
.
Structural changes of atrial myocardium due to sustained atrial fibrillation in the goat
.
Circulation
 
1997
;
96
:
3157
63
.

171

Kawasaki
 
M
,
Meulendijks
 
ER
,
van den Berg
 
NWE
,
Nariswari
 
FA
,
Neefs
 
J
,
Wesselink
 
R
, et al.  
Neutrophil degranulation interconnects over-represented biological processes in atrial fibrillation
.
Sci Rep
.
2021
;
11
:
2972
.

172

Meulendijks
 
ER
,
Al-Shama
 
RFM
,
Kawasaki
 
M
,
Fabrizi
 
B
,
Neefs
 
J
,
Wesselink
 
R
 et al.  
Atrial epicardial adipose tissue abundantly secretes myeloperoxidase and activates atrial fibroblasts in patients with atrial fibrillation
.
J Transl Med
 
2023
;
21
:
366
.

173

van den Berg
 
NWE
,
Kawasaki
 
M
,
Fabrizi
 
B
,
Nariswari
 
FA
,
Verduijn
 
AC
,
Neefs
 
J
 et al.  
Epicardial and endothelial cell activation concurs with extracellular matrix remodeling in atrial fibrillation
.
Clin Transl Med
 
2021
;
11
:
e558
.

174

Harada
 
M
,
Tadevosyan
 
A
,
Qi
 
X
,
Xiao
 
J
,
Liu
 
T
,
Voigt
 
N
 et al.  
Atrial fibrillation activates AMP-dependent protein kinase and its regulation of cellular calcium handling: potential role in metabolic adaptation and prevention of progression
.
J Am Coll Cardiol
 
2015
;
66
:
47
58
.

175

van den Berg
 
NWE
,
Neefs
 
J
,
Kawasaki
 
M
,
Nariswari
 
FA
,
Wesselink
 
R
,
Fabrizi
 
B
 et al.  
Extracellular matrix remodeling precedes atrial fibrillation: results of the PREDICT-AF trial
.
Heart rhythm
 
2021
;
18
:
2115
25
.

176

Inoue
 
K
,
Khan
 
FH
,
Remme
 
EW
,
Ohte
 
N
,
García-Izquierdo
 
E
,
Chetrit
 
M
 et al.  
Determinants of left atrial reservoir and pump strain and use of atrial strain for evaluation of left ventricular filling pressure
.
Eur Heart J Cardiovasc Imaging
 
2021
;
23
:
61
70
.

177

Smiseth
 
OA
,
Morris
 
DA
,
Cardim
 
N
,
Cikes
 
M
,
Delgado
 
V
,
Donal
 
E
 et al.  
Multimodality imaging in patients with heart failure and preserved ejection fraction: an expert consensus document of the European Association of Cardiovascular Imaging
.
Eur Heart J Cardiovasc Imaging
 
2022
;
23
:
e34
61
.

178

Shih
 
JY
,
Tsai
 
WC
,
Huang
 
YY
,
Liu
 
YW
,
Lin
 
CC
,
Huang
 
YS
 et al.  
Association of decreased left atrial strain and strain rate with stroke in chronic atrial fibrillation
.
J Am Soc Echocardiogr
 
2011
;
24
:
513
9
.

179

Kurotobi
 
T
,
Iwakura
 
K
,
Inoue
 
K
,
Kimura
 
R
,
Toyoshima
 
Y
,
Ito
 
N
 et al.  
The significance of the shape of the left atrial roof as a novel index for determining the electrophysiological and structural characteristics in patients with atrial fibrillation
.
Europace
 
2011
;
13
:
803
8
.

180

Mangiafico
 
V
,
Saberwal
 
B
,
Lavalle
 
C
,
Raharja
 
A
,
Ahmed
 
Z
,
Papageorgiou
 
N
 et al.  
The role of CT in detecting AF substrate
.
Trends Cardiovasc Med
 
2021
;
31
:
457
66
.

181

Nedios
 
S
,
Sanatkhani
 
S
,
Oladosu
 
M
,
Seewöster
 
T
,
Richter
 
S
,
Arya
 
A
 et al.  
Association of low-voltage areas with the regional wall deformation and the left atrial shape in patients with atrial fibrillation: a proof of concept study
.
Int J Cardiol Heart Vasc
 
2021
;
33
:
100730
.

182

Azemi
 
T
,
Rabdiya
 
VM
,
Ayirala
 
SR
,
McCullough
 
LD
,
Silverman
 
DI
.
Left atrial strain is reduced in patients with atrial fibrillation, stroke or TIA, and low risk CHADS(2) scores
.
J Am Soc Echocardiogr
 
2012
;
25
:
1327
32
.

183

Obokata
 
M
,
Negishi
 
K
,
Kurosawa
 
K
,
Tateno
 
R
,
Tange
 
S
,
Arai
 
M
 et al.  
Left atrial strain provides incremental value for embolism risk stratification over CHA₂DS₂-VASc score and indicates prognostic impact in patients with atrial fibrillation
.
J Am Soc Echocardiogr
 
2014
;
27
:
709
16.e4
.

184

Leung
 
M
,
van Rosendael
 
PJ
,
Abou
 
R
,
Ajmone Marsan
 
N
,
Leung
 
DY
,
Delgado
 
V
 et al.  
Left atrial function to identify patients with atrial fibrillation at high risk of stroke: new insights from a large registry
.
Eur Heart J
 
2018
;
39
:
1416
25
.

185

Lang
 
RM
,
Badano
 
LP
,
Mor-Avi
 
V
,
Afilalo
 
J
,
Armstrong
 
A
,
Ernande
 
L
 et al.  
Recommendations for cardiac chamber quantification by echocardiography in adults: an update from the American Society of Echocardiography and the European Association of Cardiovascular Imaging
.
J Am Soc Echocardiogr
 
2015
;
28
:
1
39.e14
.

186

Singh
 
A
,
Carvalho Singulane
 
C
,
Miyoshi
 
T
,
Prado
 
AD
,
Addetia
 
K
,
Bellino
 
M
 et al.  
Normal values of left atrial size and function and the impact of age: results of the World Alliance Societies of Echocardiography Study
.
J Am Soc Echocardiogr
 
2022
;
35
:
154
64.e3
.

187

Kou
 
S
,
Caballero
 
L
,
Dulgheru
 
R
,
Voilliot
 
D
,
De Sousa
 
C
,
Kacharava
 
G
 et al.  
Echocardiographic reference ranges for normal cardiac chamber size: results from the NORRE study
.
Eur Heart J Cardiovasc Imaging
 
2014
;
15
:
680
90
.

188

Soulat-Dufour
 
L
,
Addetia
 
K
,
Miyoshi
 
T
,
Citro
 
R
,
Daimon
 
M
,
Fajardo
 
PG
 et al.  
Normal values of right atrial size and function according to age, sex, and ethnicity: results of the world alliance societies of echocardiography study
.
J Am Soc Echocardiogr
 
2021
;
34
:
286
300
.

189

Krittanawong
 
C
,
Maitra
 
NS
,
Hassan Virk
 
HU
,
Farrell
 
A
,
Hamzeh
 
I
,
Arya
 
B
 et al.  
Normal ranges of right atrial strain: a systematic review and meta-analysis
.
JACC Cardiovasc imaging
 
2023
;
16
:
282
94
.

190

Kawel-Boehm
 
N
,
Hetzel
 
SJ
,
Ambale-Venkatesh
 
B
,
Captur
 
G
,
Francois
 
CJ
,
Jerosch-Herold
 
M
 et al.  
Reference ranges (“normal values”) for cardiovascular magnetic resonance (CMR) in adults and children: 2020 update
.
J Cardiovasc Magn Reson
 
2020
;
22
:
87
.

191

Doria de Vasconcellos
 
H
,
Win
 
TT
,
Chamera
 
E
,
Hong
 
SY
,
Venkatesh
 
BA
,
Young
 
P
 et al.  
References values for left atrial volumes, emptying fractions, strains, and strain rates and their determinants by age, gender, and ethnicity: the multiethnic study of atherosclerosis (MESA)
.
Acad Radiol
 
2021
;
28
:
356
63
.

192

Stojanovska
 
J
,
Cronin
 
P
,
Patel
 
S
,
Gross
 
BH
,
Oral
 
H
,
Chughtai
 
K
 et al.  
Reference normal absolute and indexed values from ECG-gated MDCT: left atrial volume, function, and diameter
.
AJR Am J Roentgenol
 
2011
;
197
:
631
7
.

193

Aquaro
 
GD
,
De Gori
 
C
,
Faggioni
 
L
,
Parisella
 
ML
,
Cioni
 
D
,
Lencioni
 
R
 et al.  
Diagnostic and prognostic role of late gadolinium enhancement in cardiomyopathies
.
Eur Heart J Suppl
 
2023
;
25
:
C130
c6
.

194

Okasha
 
O
,
Kazmirczak
 
F
,
Chen
 
KA
,
Farzaneh-Far
 
A
,
Shenoy
 
C
.
Myocardial involvement in patients with histologically diagnosed cardiac sarcoidosis: a systematic review and meta-analysis of gross pathological images from autopsy or cardiac transplantation cases
.
J Am Heart Assoc
 
2019
;
8
:
e011253
.

195

Donal
 
E
,
Lip
 
GY
,
Galderisi
 
M
,
Goette
 
A
,
Shah
 
D
,
Marwan
 
M
 et al.  
EACVI/EHRA expert consensus document on the role of multi-modality imaging for the evaluation of patients with atrial fibrillation
.
Eur Heart J Cardiovasc Imaging
 
2016
;
17
:
355
83
.

196

Eichenlaub
 
M
,
Mueller-Edenborn
 
B
,
Minners
 
J
,
Figueras
 
IVRM
,
Forcada
 
BR
,
Colomer
 
AV
 et al.  
Comparison of various late gadolinium enhancement magnetic resonance imaging methods with high-definition voltage and activation mapping for detection of atrial cardiomyopathy
.
Europace
 
2022
;
24
:
1102
11
.

197

Boyle
 
PM
,
Sarairah
 
S
,
Kwan
 
KT
,
Scott
 
GD
,
Mohamedali
 
F
,
Anderson
 
CA
 et al.  
Elevated fibrosis burden as assessed by MRI predicts cryoballoon ablation failure
.
J Cardiovasc Electrophysiol
 
2023
;
34
:
302
12
.

198

Flett
 
AS
,
Hayward
 
MP
,
Ashworth
 
MT
,
Hansen
 
MS
,
Taylor
 
AM
,
Elliott
 
PM
 et al.  
Equilibrium contrast cardiovascular magnetic resonance for the measurement of diffuse myocardial fibrosis: preliminary validation in humans
.
Circulation
 
2010
;
122
:
138
44
.

199

Platonov
 
PG
,
Mitrofanova
 
LB
,
Orshanskaya
 
V
,
Ho
 
SY
.
Structural abnormalities in atrial walls are associated with presence and persistency of atrial fibrillation but not with age
.
J Am Coll Cardiol
 
2011
;
58
:
2225
32
.

200

Zahid
 
S
,
Cochet
 
H
,
Boyle
 
PM
,
Schwarz
 
EL
,
Whyte
 
KN
,
Vigmond
 
EJ
 et al.  
Patient-derived models link re-entrant driver localization in atrial fibrillation to fibrosis spatial pattern
.
Cardiovasc Res
 
2016
;
110
:
443
54
.

201

Marrouche
 
NF
,
Wilber
 
D
,
Hindricks
 
G
,
Jais
 
P
,
Akoum
 
N
,
Marchlinski
 
F
 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
.

202

King
 
JB
,
Azadani
 
PN
,
Suksaranjit
 
P
,
Bress
 
AP
,
Witt
 
DM
,
Han
 
FT
 et al.  
Left atrial fibrosis and risk of cerebrovascular and cardiovascular events in patients with atrial fibrillation
.
J Am Coll Cardiol
 
2017
;
70
:
1311
21
.

203

Marrouche
 
NF
,
Wazni
 
O
,
McGann
 
C
,
Greene
 
T
,
Dean
 
JM
,
Dagher
 
L
 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
305
.

204

Bisbal
 
F
,
Benito
 
E
,
Teis
 
A
,
Alarcón
 
F
,
Sarrias
 
A
,
Caixal
 
G
 et al.  
Magnetic resonance imaging-guided fibrosis ablation for the treatment of atrial fibrillation: the ALICIA trial
.
Circ Arrhythm Electrophysiol
 
2020
;
13
:
e008707
.

205

Yamaguchi
 
T
,
Otsubo
 
T
,
Takahashi
 
Y
,
Nakashima
 
K
,
Fukui
 
A
,
Hirota
 
K
 et al.  
Atrial structural remodeling in patients with atrial fibrillation is a diffuse fibrotic process: evidence from high-density voltage mapping and atrial biopsy
.
J Am Heart Assoc
 
2022
;
11
:
e024521
.

206

Chen
 
J
,
Arentz
 
T
,
Cochet
 
H
,
Müller-Edenborn
 
B
,
Kim
 
S
,
Moreno-Weidmann
 
Z
 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
93
.

207

Hirose
 
T
,
Kawasaki
 
M
,
Tanaka
 
R
,
Ono
 
K
,
Watanabe
 
T
,
Iwama
 
M
 et al.  
Left atrial function assessed by speckle tracking echocardiography as a predictor of new-onset non-valvular atrial fibrillation: results from a prospective study in 580 adults
.
Eur Heart J Cardiovasc Imaging
 
2012
;
13
:
243
50
.

208

Hauser
 
R
,
Nielsen
 
AB
,
Skaarup
 
KG
,
Lassen
 
MCH
,
Duus
 
LS
,
Johansen
 
ND
 et al.  
Left atrial strain predicts incident atrial fibrillation in the general population: the Copenhagen city heart study
.
Eur Heart J Cardiovasc Imaging
 
2021
;
23
:
52
60
.

209

Nielsen
 
AB
,
Skaarup
 
KG
,
Hauser
 
R
,
Johansen
 
ND
,
Lassen
 
MCH
,
Jensen
 
GB
 et al.  
Normal values and reference ranges for left atrial strain by speckle-tracking echocardiography: the Copenhagen City Heart Study
.
Eur Heart J Cardiovasc Imaging
 
2021
;
23
:
42
51
.

210

Habibi
 
M
,
Samiei
 
S
,
Ambale Venkatesh
 
B
,
Opdahl
 
A
,
Helle-Valle
 
TM
,
Zareian
 
M
 et al.  
Cardiac magnetic resonance-measured left atrial volume and function and incident atrial fibrillation: results from MESA (multi-ethnic study of atherosclerosis)
.
Circ Cardiovasc Imaging
 
2016
;
9
:
51
56
.

211

Lim
 
DJ
,
Ambale-Ventakesh
 
B
,
Ostovaneh
 
MR
,
Zghaib
 
T
,
Ashikaga
 
H
,
Wu
 
C
 et al.  
Change in left atrial function predicts incident atrial fibrillation: the multi-ethnic study of atherosclerosis
.
Eur Heart J Cardiovasc Imaging
 
2019
;
20
:
979
87
.

212

Xie
 
E
,
Yu
 
R
,
Ambale-Venkatesh
 
B
,
Bakhshi
 
H
,
Heckbert
 
SR
,
Soliman
 
EZ
 et al.  
Association of right atrial structure with incident atrial fibrillation: a longitudinal cohort cardiovascular magnetic resonance study from the Multi-Ethnic Study of Atherosclerosis (MESA)
.
J Cardiovasc Magn Reson
 
2020
;
22
:
36
.

213

Chong
 
B
,
Jayabaskaran
 
J
,
Ruban
 
J
,
Goh
 
R
,
Chin
 
YH
,
Kong
 
G
 et al.  
Epicardial adipose tissue assessed by computed tomography and echocardiography are associated with adverse cardiovascular outcomes: a systematic review and meta-analysis
.
Circ Cardiovasc Imaging
 
2023
;
16
:
e015159
.

214

West
 
HW
,
Siddique
 
M
,
Williams
 
MC
,
Volpe
 
L
,
Desai
 
R
,
Lyasheva
 
M
 et al.  
Deep-learning for epicardial adipose tissue assessment with computed tomography: implications for cardiovascular risk prediction
.
JACC Cardiovasc Imaging
 
2023
;
16
:
800
16
.

215

Heckbert
 
SR
,
Jensen
 
PN
,
Austin
 
TR
,
Chen
 
LY
,
Post
 
WS
,
Ambale Venkatesh
 
B
 et al.  
Associations of left atrial function and structure with supraventricular ectopy: the multi-ethnic study of atherosclerosis
.
J Am Heart Assoc
 
2021
;
10
:
e018093
.

216

Huber
 
MP
,
Pandit
 
JA
,
Jensen
 
PN
,
Wiggins
 
KL
,
Patel
 
RB
,
Freed
 
BH
 et al.  
Left atrial strain and the risk of atrial arrhythmias from extended ambulatory cardiac monitoring: MESA
.
J Am Heart Assoc
 
2022
;
11
:
e026875
.

217

Tops
 
LF
,
Delgado
 
V
,
Bertini
 
M
,
Marsan
 
NA
,
Den Uijl
 
DW
,
Trines
 
SA
 et al.  
Left atrial strain predicts reverse remodeling after catheter ablation for atrial fibrillation
.
J Am Coll Cardiol
 
2011
;
57
:
324
31
.

218

Soulat-Dufour
 
L
,
Lang
 
S
,
Addetia
 
K
,
Ederhy
 
S
,
Adavane-Scheuble
 
S
,
Chauvet-Droit
 
M
 et al.  
Restoring sinus rhythm reverses cardiac remodeling and reduces valvular regurgitation in patients with atrial fibrillation
.
J Am Coll Cardiol
 
2022
;
79
:
951
61
.

219

Huang
 
SH
,
Tsao
 
HM
,
Liao
 
CF
,
Chen
 
ZY
,
Chao
 
TF
,
Chen
 
SA
.
Multimodality imaging assessment of the Biatrial remodeling of the burden of atrial high-rate episodes in patients with cardiac implanted electronic devices
.
Int J Cardiol
 
2023
;
371
:
175
83
.

220

Tsao
 
HM
,
Hu
 
WC
,
Tsai
 
PH
,
Lee
 
CL
,
Wang
 
HH
,
Chang
 
SL
 et al.  
Functional remodeling of both atria is associated with occurrence of stroke in patients with paroxysmal and persistent atrial fibrillation
.
Acta Cardiol Sin
 
2017
;
33
:
50
7
.

221

Huang
 
T
,
Patrick
 
S
,
Mayer
 
LK
,
Müller-Edenborn
 
B
,
Eichenlaub
 
M
,
Allgeier
 
M
 et al.  
Echocardiographic and electrocardiographic determinants of atrial cardiomyopathy identify patients with atrial fibrillation at risk for left atrial thrombogenesis
.
J Clin Med
 
2022
;
11
:
1332
.

222

Kim
 
D
,
Shim
 
CY
,
Hong
 
GR
,
Kim
 
MH
,
Seo
 
J
,
Cho
 
IJ
 et al.  
Clinical implications and determinants of left atrial mechanical dysfunction in patients with stroke
.
Stroke
 
2016
;
47
:
1444
51
.

223

Di Biase
 
L
,
Santangeli
 
P
,
Anselmino
 
M
,
Mohanty
 
P
,
Salvetti
 
I
,
Gili
 
S
 et al.  
Does the left atrial appendage morphology correlate with the risk of stroke in patients with atrial fibrillation? Results from a multicenter study
.
J Am Coll Cardiol
 
2012
;
60
:
531
8
.

224

Burrell
 
LD
,
Horne
 
BD
,
Anderson
 
JL
,
Muhlestein
 
JB
,
Whisenant
 
BK
.
Usefulness of left atrial appendage volume as a predictor of embolic stroke in patients with atrial fibrillation
.
Am J Cardiol
 
2013
;
112
:
1148
52
.

225

Markl
 
M
,
Lee
 
DC
,
Furiasse
 
N
,
Carr
 
M
,
Foucar
 
C
,
Ng
 
J
 et al.  
Left atrial and left atrial appendage 4D blood flow dynamics in atrial fibrillation
.
Circ Cardiovasc Imaging
 
2016
;
9
:
e004984
.

226

Spartera
 
M
,
Stracquadanio
 
A
,
Pessoa-Amorim
 
G
,
Von Ende
 
A
,
Fletcher
 
A
,
Manley
 
P
 et al.  
The impact of atrial fibrillation and stroke risk factors on left atrial blood flow characteristics
.
Eur Heart J Cardiovasc Imaging
 
2021
;
23
:
115
23
.

227

Ayubcha
 
C
,
Amanullah
 
A
,
Patel
 
KH
,
Teichner
 
E
,
Gokhale
 
S
,
Marquez-Valenzuela
 
U
 et al.  
Stroke and molecular imaging: a focus on FDG-PET
.
Am J Nucl Med Mol Imaging
 
2023
;
13
:
51
63
.

228

Müller
 
P
,
Makimoto
 
H
,
Dietrich
 
JW
,
Fochler
 
F
,
Nentwich
 
K
,
Krug
 
J
 et al.  
Association of left atrial low-voltage area and thromboembolic risk in patients with atrial fibrillation
.
Europace
 
2018
;
20
:
f359
f65
.

229

Prabhu
 
S
,
Voskoboinik
 
A
,
McLellan
 
AJA
,
Peck
 
KY
,
Pathik
 
B
,
Nalliah
 
CJ
 et al.  
Biatrial electrical and structural atrial changes in heart failure: electroanatomic mapping in persistent atrial fibrillation in humans
.
JACC Clin Electrophysiol
 
2018
;
4
:
87
96
.

230

Masuda
 
M
,
Matsuda
 
Y
,
Uematsu
 
H
,
Sugino
 
A
,
Ooka
 
H
,
Kudo
 
S
 et al.  
Prognostic impact of atrial cardiomyopathy: long-term follow-up of patients with and without low-voltage areas following atrial fibrillation ablation
.
Heart Rhythm
 
2023
;
21
:
378
86
.

231

Kato
 
Y
,
Takahashi
 
S
.
Atrial cardiopathy and cryptogenic stroke
.
Front Neurol
 
2022
;
13
:
839398
.

232

Kamel
 
H
,
Longstreth
 
WT
, Jr.
,
Tirschwell
 
DL
,
Kronmal
 
RA
,
Broderick
 
JP
,
Palesch
 
YY
 et al.  
The AtRial cardiopathy and antithrombotic drugs in prevention after cryptogenic stroke randomized trial: rationale and methods
.
Int J Stroke
 
2019
;
14
:
207
14
.

233

Maheshwari
 
A
,
Norby
 
FL
,
Inciardi
 
RM
,
Wang
 
W
,
Zhang
 
MJ
,
Soliman
 
EZ
 et al.  
Left atrial mechanical dysfunction and the risk for ischemic stroke in people without prevalent atrial fibrillation or stroke : a prospective cohort study
.
Ann Intern Med
 
2023
;
176
:
39
48
.

234

Johansen
 
MC
,
Wang
 
W
,
Zhang
 
M
,
Knopman
 
DS
,
Ndumele
 
C
,
Mosley
 
TH
 et al.  
Risk of dementia associated with atrial cardiopathy: the ARIC study
.
J Am Heart Assoc
 
2022
;
11
:
e025646
.

235

Ding
 
WY
,
Gupta
 
D
,
Lip
 
GYH
.
Atrial fibrillation and the prothrombotic state: revisiting Virchow's triad in 2020
.
Heart
 
2020
;
106
:
1463
8
.

236

Khan
 
AA
,
Lip
 
GYH
.
The prothrombotic state in atrial fibrillation: pathophysiological and management implications
.
Cardiovasc Res
 
2019
;
115
:
31
45
.

237

Goette
 
A
,
Jentsch-Ullrich
 
K
,
Lendeckel
 
U
,
Röcken
 
C
,
Agbaria
 
M
,
Auricchio
 
A
 et al.  
Effect of atrial fibrillation on hematopoietic progenitor cells: a novel pathophysiological role of the atrial natriuretic peptide?
 
Circulation
 
2003
;
108
:
2446
9
.

238

Mukherjee
 
R
,
Akar
 
JG
,
Wharton
 
JM
,
Adams
 
DK
,
McClure
 
CD
,
Stroud
 
RE
 et al.  
Plasma profiles of matrix metalloproteinases and tissue inhibitors of the metalloproteinases predict recurrence of atrial fibrillation following cardioversion
.
J Cardiovasc Transl Res
 
2013
;
6
:
528
35
.

239

Lee
 
KK
,
Doudesis
 
D
,
Anwar
 
M
,
Astengo
 
F
,
Chenevier-Gobeaux
 
C
,
Claessens
 
YE
 et al.  
Development and validation of a decision support tool for the diagnosis of acute heart failure: systematic review, meta-analysis, and modelling study
.
BMJ
 
2022
;
377
:
e068424
.

240

Nasab Mehrabi
 
E
,
Toupchi-Khosroshahi
 
V
,
Athari
 
SS
.
Relationship of atrial fibrillation and N terminal pro brain natriuretic peptide in heart failure patients
.
ESC Heart Fail
 
2023
;
10
:
3250
7
.

241

Mu
 
X
,
Qiu
 
M
,
Li
 
Y
,
Li
 
Z
,
Qi
 
B
,
Jing
 
Z
 et al.  
N-terminal pro-B-type natriuretic peptide improves the predictive value of CHA(2) DS(2) -VASc risk score for long-term cardiovascular events in acute coronary syndrome patients with atrial fibrillation
.
Clin Cardiol
 
2023
;
46
:
810
7
.

242

Folsom
 
AR
,
Nambi
 
V
,
Bell
 
EJ
,
Oluleye
 
OW
,
Gottesman
 
RF
,
Lutsey
 
PL
 et al.  
Troponin T, N-terminal pro-B-type natriuretic peptide, and incidence of stroke: the atherosclerosis risk in communities study
.
Stroke
 
2013
;
44
:
961
7
.

243

Sun
 
WP
,
Du
 
X
,
Chen
 
JJ
.
Biomarkers for predicting the occurrence and progression of atrial fibrillation: soluble suppression of tumorigenicity 2 protein and tissue inhibitor of matrix metalloproteinase-1
.
Int J Clin Pract
 
2022
;
2022
:
6926510
.

244

Reyat
 
JS
,
Chua
 
W
,
Cardoso
 
VR
,
Witten
 
A
,
Kastner
 
PM
,
Kabir
 
SN
 et al.  
Reduced left atrial cardiomyocyte PITX2 and elevated circulating BMP10 predict atrial fibrillation after ablation
.
JCI insight
 
2020
;
5
:
e139179
.

245

Hijazi
 
Z
,
Benz
 
AP
,
Lindbäck
 
J
,
Alexander
 
JH
,
Connolly
 
SJ
,
Eikelboom
 
JW
 et al.  
Bone morphogenetic protein 10: a novel risk marker of ischaemic stroke in patients with atrial fibrillation
.
Eur Heart J
 
2023
;
44
:
208
18
.

246

Hennings
 
E
,
Blum
 
S
,
Aeschbacher
 
S
,
Coslovsky
 
M
,
Knecht
 
S
,
Eken
 
C
 et al.  
Bone morphogenetic protein 10-A novel biomarker to predict adverse outcomes in patients with atrial fibrillation
.
J Am Heart Assoc
 
2023
;
12
:
e028255
.

247

Chung
 
MK
,
Martin
 
DO
,
Sprecher
 
D
,
Wazni
 
O
,
Kanderian
 
A
,
Carnes
 
CA
 et al.  
C-reactive protein elevation in patients with atrial arrhythmias: inflammatory mechanisms and persistence of atrial fibrillation
.
Circulation
 
2001
;
104
:
2886
91
.

248

Conway
 
DS
,
Pearce
 
LA
,
Chin
 
BS
,
Hart
 
RG
,
Lip
 
GY
.
Prognostic value of plasma von Willebrand factor and soluble P-selectin as indices of endothelial damage and platelet activation in 994 patients with nonvalvular atrial fibrillation
.
Circulation
 
2003
;
107
:
3141
5
.

249

Camelo-Castillo
 
A
,
Rivera-Caravaca
 
JM
,
Marín
 
F
,
Vicente
 
V
,
Lip
 
GYH
,
Roldán
 
V
.
Predicting adverse events beyond stroke and bleeding with the ABC-stroke and ABC-bleeding scores in patients with atrial fibrillation: the Murcia AF project
.
Thromb Haemost
 
2020
;
120
:
1200
7
.

250

Esteve-Pastor
 
MA
,
Roldán
 
V
,
Rivera-Caravaca
 
JM
,
Ramírez-Macías
 
I
,
Lip
 
GYH
,
Marín
 
F
.
The use of biomarkers in clinical management guidelines: a critical appraisal
.
Thromb Haemost
 
2019
;
119
:
1901
19
.

251

Gorog
 
DA
,
Gue
 
YX
,
Chao
 
TF
,
Fauchier
 
L
,
Ferreiro
 
JL
,
Huber
 
K
 et al.  
Assessment and mitigation of bleeding risk in atrial fibrillation and venous thromboembolism: executive summary of a European and Asia-Pacific expert consensus paper
.
Thromb Haemost
 
2022
;
122
:
1625
52
.

252

Xie
 
S
,
Li
 
Q
,
Luk
 
AOY
,
Lan
 
HY
,
Chan
 
PKS
,
Bayés-Genís
 
A
 et al.  
Major adverse cardiovascular events and mortality prediction by circulating GDF-15 in patients with type 2 diabetes: a systematic review and meta-analysis
.
Biomolecules
 
2022
;
12
:
934
.

253

Ding
 
WY
,
McDowell
 
G
,
Lip
 
GYH
.
Utilizing biomarkers in atrial fibrillation: the pros and cons
.
Cardiovasc Res
 
2022
;
118
:
2035
6
.

254

Fatkin
 
D
,
Huttner
 
IG
,
Johnson
 
R
.
Genetics of atrial cardiomyopathy
.
Curr Opin Cardiol
 
2019
;
34
:
275
81
.

255

Gudbjartsson
 
DF
,
Arnar
 
DO
,
Helgadottir
 
A
,
Gretarsdottir
 
S
,
Holm
 
H
,
Sigurdsson
 
A
 et al.  
Variants conferring risk of atrial fibrillation on chromosome 4q25
.
Nature
 
2007
;
448
:
353
7
.

256

Orr
 
N
,
Arnaout
 
R
,
Gula
 
LJ
,
Spears
 
DA
,
Leong-Sit
 
P
,
Li
 
Q
 et al.  
A mutation in the atrial-specific myosin light chain gene (MYL4) causes familial atrial fibrillation
.
Nat Commun
 
2016
;
7
:
11303
.

257

Disertori
 
M
,
Quintarelli
 
S
,
Grasso
 
M
,
Pilotto
 
A
,
Narula
 
N
,
Favalli
 
V
 et al.  
Autosomal recessive atrial dilated cardiomyopathy with standstill evolution associated with mutation of Natriuretic Peptide Precursor A
.
Circ Cardiovasc Genet
 
2013
;
6
:
27
36
.

258

Schulze-Bahr
 
E
,
Neu
 
A
,
Friederich
 
P
,
Kaupp
 
UB
,
Breithardt
 
G
,
Pongs
 
O
 et al.  
Pacemaker channel dysfunction in a patient with sinus node disease
.
J Clin Invest
 
2003
;
111
:
1537
45
.

259

Tan
 
RB
,
Gando
 
I
,
Bu
 
L
,
Cecchin
 
F
,
Coetzee
 
W
.
A homozygous SCN5A mutation associated with atrial standstill and sudden death
.
Pacing Clin Electrophysiol
 
2018
;
41
:
1036
42
.

260

Ramos-Mondragon
 
R
,
Edokobi
 
N
,
Hodges
 
SL
,
Wang
 
S
,
Bouza
 
AA
,
Canugovi
 
C
 et al.  
Neonatal Scn1b-null mice have sinoatrial node dysfunction, altered atrial structure, and atrial fibrillation
.
JCI Insight
 
2022
;
7
:
e152050
.

261

Duparc
 
A
,
Cintas
 
P
,
Somody
 
E
,
Bieth
 
E
,
Richard
 
P
,
Maury
 
P
 et al.  
A cardio-neurological form of laminopathy: dilated cardiomyopathy with permanent partial atrial standstill and axonal neuropathy
.
Pacing Clin Electrophysiol
 
2009
;
32
:
410
5
.

262

van Setten
 
J
,
Brody
 
JA
,
Jamshidi
 
Y
,
Swenson
 
BR
,
Butler
 
AM
,
Campbell
 
H
 et al.  
PR interval genome-wide association meta-analysis identifies 50 loci associated with atrial and atrioventricular electrical activity
.
Nat Commun
 
2018
;
9
:
2904
.

263

Wild
 
PS
,
Felix
 
JF
,
Schillert
 
A
,
Teumer
 
A
,
Chen
 
MH
,
Leening
 
MJG
 et al.  
Large-scale genome-wide analysis identifies genetic variants associated with cardiac structure and function
.
J Clin Invest
 
2017
;
127
:
1798
812
.

264

Nielsen
 
JB
,
Thorolfsdottir
 
RB
,
Fritsche
 
LG
,
Zhou
 
W
,
Skov
 
MW
,
Graham
 
SE
 et al.  
Biobank-driven genomic discovery yields new insight into atrial fibrillation biology
.
Nat Genet
 
2018
;
50
:
1234
9
.

265

Roselli
 
C
,
Chaffin
 
MD
,
Weng
 
LC
,
Aeschbacher
 
S
,
Ahlberg
 
G
,
Albert
 
CM
 et al.  
Multi-ethnic genome-wide association study for atrial fibrillation
.
Nat Genet
 
2018
;
50
:
1225
33
.

266

Steenman
 
M
.
Insight into atrial fibrillation through analysis of the coding transcriptome in humans
.
Biophys Rev
 
2020
;
12
:
817
26
.

267

Man
 
JCK
,
van Duijvenboden
 
K
,
Krijger
 
PHL
,
Hooijkaas
 
IB
,
van der Made
 
I
,
de Gier-de Vries
 
C
 et al.  
Genetic dissection of a super enhancer controlling the Nppa-Nppb cluster in the heart
.
Circ Res
 
2021
;
128
:
115
29
.

268

Cao
 
TH
,
Quinn
 
PA
,
Sandhu
 
JK
,
Voors
 
AA
,
Lang
 
CC
,
Parry
 
HM
 et al.  
Identification of novel biomarkers in plasma for prediction of treatment response in patients with heart failure
.
Lancet
 
2015
;
385
:
S26
.

269

Çubukçuoğlu Deniz
 
G
,
Durdu
 
S
,
Doğan
 
Y
,
Erdemli
 
E
,
Özdağ
 
H
,
Akar
 
AR
.
Molecular signatures of human chronic atrial fibrillation in primary mitral regurgitation
.
Cardiovasc Ther
 
2021
;
2021
:
5516185
.

270

Zeemering
 
S
,
Isaacs
 
A
,
Winters
 
J
,
Maesen
 
B
,
Bidar
 
E
,
Dimopoulou
 
C
 et al.  
Atrial fibrillation in the presence and absence of heart failure enhances expression of genes involved in cardiomyocyte structure, conduction properties, fibrosis, inflammation, and endothelial dysfunction
.
Heart Rhythm
 
2022
;
19
:
2115
24
.

271

Alvarez-Franco
 
A
,
Rouco
 
R
,
Ramirez
 
RJ
,
Guerrero-Serna
 
G
,
Tiana
 
M
,
Cogliati
 
S
 et al.  
Transcriptome and proteome mapping in the sheep atria reveal molecular featurets of atrial fibrillation progression
.
Cardiovasc Res
 
2021
;
117
:
1760
75
.

272

Roselli
 
C
,
Rienstra
 
M
,
Ellinor
 
PT
.
Genetics of atrial fibrillation in 2020: GWAS, genome sequencing, polygenic risk, and beyond
.
Circ Res
 
2020
;
127
:
21
33
.

273

van Ouwerkerk
 
AF
,
Bosada
 
FM
,
van Duijvenboden
 
K
,
Hill
 
MC
,
Montefiori
 
LE
,
Scholman
 
KT
 et al.  
Identification of atrial fibrillation associated genes and functional non-coding variants
.
Nat Commun
 
2019
;
10
:
4755
.

274

Doñate Puertas
 
R
,
Meugnier
 
E
,
Romestaing
 
C
,
Rey
 
C
,
Morel
 
E
,
Lachuer
 
J
 et al.  
Atrial fibrillation is associated with hypermethylation in human left atrium, and treatment with decitabine reduces atrial tachyarrhythmias in spontaneously hypertensive rats
.
Transl Res
 
2017
;
184
:
57
67.e5
.

275

Shen
 
K
,
Tu
 
T
,
Yuan
 
Z
,
Yi
 
J
,
Zhou
 
Y
,
Liao
 
X
 et al.  
DNA methylation dysregulations in valvular atrial fibrillation
.
Clin Cardiol
 
2017
;
40
:
686
91
.

276

Roberts
 
JD
,
Vittinghoff
 
E
,
Lu
 
AT
,
Alonso
 
A
,
Wang
 
B
,
Sitlani
 
CM
 et al.  
Epigenetic age and the risk of incident atrial fibrillation
.
Circulation
 
2021
;
144
:
1899
911
.

277

Ward-Caviness
 
CK
.
Accelerated epigenetic aging and incident atrial fibrillation: new outlook on an immutable risk factor?
 
Circulation
 
2021
;
144
:
1912
4
.

278

Brundel
 
B
,
Li
 
J
,
Zhang
 
D
.
Role of HDACs in cardiac electropathology: therapeutic implications for atrial fibrillation
.
Biochim Biophys Acta Mol Cell Res
 
2020
;
1867
:
118459
.

279

Song
 
S
,
Zhang
 
R
,
Mo
 
B
,
Chen
 
L
,
Liu
 
L
,
Yu
 
Y
 et al.  
EZH2 as a novel therapeutic target for atrial fibrosis and atrial fibrillation
.
J Mol Cell Cardiol
 
2019
;
135
:
119
33
.

280

Müller-Edenborn
 
B
,
Chen
 
J
,
Allgeier
 
J
,
Didenko
 
M
,
Moreno-Weidmann
 
Z
,
Neumann
 
FJ
 et al.  
Amplified sinus-P-wave reveals localization and extent of left atrial low-voltage substrate: implications for arrhythmia freedom following pulmonary vein isolation
.
Europace
 
2020
;
22
:
240
9
.

281

Jadidi
 
A
,
Müller-Edenborn
 
B
,
Chen
 
J
,
Keyl
 
C
,
Weber
 
R
,
Allgeier
 
J
 et al.  
The duration of the amplified sinus-P-wave identifies presence of left atrial low voltage substrate and predicts outcome after pulmonary vein isolation in patients with persistent atrial fibrillation
.
JACC Clin Electrophysiol
 
2018
;
4
:
531
43
.

282

Huang
 
T
,
Schurr
 
P
,
Muller-Edenborn
 
B
,
Pilia
 
N
,
Mayer
 
L
,
Eichenlaub
 
M
 et al.  
Analysis of the amplified p-wave enables identification of patients with atrial fibrillation during sinus rhythm
.
Front Cardiovasc Med
 
2023
;
10
:
1095931
.

283

McConkey
 
N
,
Malamas
 
P
,
Norby
 
FL
,
Plamenac
 
J
,
Park
 
R
,
Weigel
 
F
 et al.  
Abnormal P-wave terminal force in lead V1 is associated with low left atrial appendage ejection velocity
.
J Electrocardiol
 
2021
;
67
:
142
7
.

284

Lebek
 
S
,
Wester
 
M
,
Pec
 
J
,
Poschenrieder
 
F
,
Tafelmeier
 
M
,
Fisser
 
C
 et al.  
Abnormal P-wave terminal force in lead V(1) is a marker for atrial electrical dysfunction but not structural remodelling
.
ESC Heart Fail
 
2021
;
8
:
4055
66
.

285

Huang
 
Z
,
Zheng
 
Z
,
Wu
 
B
,
Tang
 
L
,
Xie
 
X
,
Dong
 
R
 et al.  
Predictive value of P wave terminal force in lead V1 for atrial fibrillation: a meta-analysis
.
Ann Noninvasive Electrocardiol
 
2020
;
25
:
e12739
.

286

Li
 
R
,
Yang
 
X
,
Jia
 
M
,
Wang
 
D
,
Cui
 
X
,
Bai
 
L
 et al.  
Effectiveness of P-wave ECG index and left atrial appendage volume in predicting atrial fibrillation recurrence after first radiofrequency catheter ablation
.
BMC Cardiovasc Disord
 
2021
;
21
:
164
.

287

German
 
DM
,
Kabir
 
MM
,
Dewland
 
TA
,
Henrikson
 
CA
,
Tereshchenko
 
LG
.
Atrial fibrillation predictors: importance of the electrocardiogram
.
Ann Noninvasive Electrocardiol
 
2016
;
21
:
20
9
.

288

Alexander
 
B
,
Milden
 
J
,
Hazim
 
B
,
Haseeb
 
S
,
Bayes-Genis
 
A
,
Elosua
 
R
 et al.  
New electrocardiographic score for the prediction of atrial fibrillation: the MVP ECG risk score (morphology-voltage-P-wave duration)
.
Ann Noninvasive Electrocardiol
 
2019
;
24
:
e12669
.

289

Park
 
JK
,
Park
 
J
,
Uhm
 
JS
,
Joung
 
B
,
Lee
 
MH
,
Pak
 
HN
.
Low P-wave amplitude (<0.1
Mv) in lead I is associated with displaced inter-atrial conduction and clinical recurrence of paroxysmal atrial fibrillation after radiofrequency catheter ablation
.
Europace
.
2016
;
18
:
384
91
.

290

Zeng
 
C
,
Wei
 
T
,
Zhao
 
R
,
Wang
 
C
,
Chen
 
L
,
Wang
 
L
.
Electrocardiographic diagnosis of left atrial enlargement in patients with mitral stenosis: the value of the P-wave area
.
Acta Cardiol
 
2003
;
58
:
139
41
.

291

Tse
 
G
,
Lakhani
 
I
,
Zhou
 
J
,
Li
 
KHC
,
Lee
 
S
,
Liu
 
Y
 et al.  
P-wave area predicts new onset atrial fibrillation in mitral stenosis: a machine learning approach
.
Front Bioeng Biotechnol
 
2020
;
8
:
479
.

292

Dilaveris
 
PE
,
Gialafos
 
EJ
,
Sideris
 
SK
,
Theopistou
 
AM
,
Andrikopoulos
 
GK
,
Kyriakidis
 
M
 et al.  
Simple electrocardiographic markers for the prediction of paroxysmal idiopathic atrial fibrillation
.
Am Heart J
 
1998
;
135
:
733
8
.

293

Marks
 
D
,
Ho
 
R
,
Then
 
R
,
Weinstock
 
JL
,
Teklemariam
 
E
,
Kakadia
 
B
 et al.  
Real-world experience with implantable loop recorder monitoring to detect subclinical atrial fibrillation in patients with cryptogenic stroke: the value of p wave dispersion in predicting arrhythmia occurrence
.
Int J Cardiol
 
2021
;
327
:
86
92
.

294

Dilaveris
 
PE
,
Gialafos
 
EJ
,
Andrikopoulos
 
GK
,
Richter
 
DJ
,
Papanikolaou
 
V
,
Poralis
 
K
 et al.  
Clinical and electrocardiographic predictors of recurrent atrial fibrillation
.
Pacing Clin Electrophysiol
 
2000
;
23
:
352
8
.

295

Aysha
 
MH
,
Hassan
 
AS
.
Diagnostic importance of fibrillatory wave amplitude: a clue to echocardiographic left atrial size and etiology of atrial fibrillation
.
J Electrocardiol
 
1988
;
21
:
247
51
.

296

İçen
 
YK
,
Koca
 
H
,
Sümbül
 
HE
,
Yıldırım
 
A
,
Koca
 
F
,
Yıldırım
 
A
 et al.  
Relationship between coarse F waves and thromboembolic events in patients with permanent atrial fibrillation
.
J Arrhythm
 
2020
;
36
:
1025
31
.

297

Kawaji
 
T
,
Ogawa
 
H
,
Hamatani
 
Y
,
Kato
 
M
,
Yokomatsu
 
T
,
Miki
 
S
 et al.  
Fine fibrillatory wave as a risk factor for heart failure events in patients with atrial fibrillation: the fushimi atrial fibrillation (AF) registry
.
J Am Heart Assoc
 
2022
;
11
:
e024341
.

298

Park
 
J
,
Lee
 
C
,
Leshem
 
E
,
Blau
 
I
,
Kim
 
S
,
Lee
 
JM
 et al.  
Early differentiation of long-standing persistent atrial fibrillation using the characteristics of fibrillatory waves in surface ECG multi-leads
.
Sci Rep
 
2019
;
9
:
2746
.

299

Lankveld
 
T
,
Zeemering
 
S
,
Scherr
 
D
,
Kuklik
 
P
,
Hoffmann
 
BA
,
Willems
 
S
 et al.  
Atrial fibrillation complexity parameters derived from surface ECGs predict procedural outcome and long-term follow-up of stepwise catheter ablation for atrial fibrillation
.
Circ Arrhythm Electrophysiol
 
2016
;
9
:
e003354
.

300

Zarzoso
 
V
,
Latcu
 
DG
,
Hidalgo-Muñoz
 
AR
,
Meo
 
M
,
Meste
 
O
,
Popescu
 
I
 et al.  
Non-invasive prediction of catheter ablation outcome in persistent atrial fibrillation by fibrillatory wave amplitude computation in multiple electrocardiogram leads
.
Arch Cardiovasc Dis
 
2016
;
109
:
679
88
.

301

Intzes
 
S
,
Zagoridis
 
K
,
Symeonidou
 
M
,
Spanoudakis
 
E
,
Arya
 
A
,
Dinov
 
B
 et al.  
P-wave duration and atrial fibrillation recurrence after catheter ablation: a systematic review and meta-analysis
.
Europace
 
2023
;
25
:
450
9
.

302

Chen
 
LY
,
Ribeiro
 
ALP
,
Platonov
 
PG
,
Cygankiewicz
 
I
,
Soliman
 
EZ
,
Gorenek
 
B
 et al.  
P wave parameters and indices: a critical appraisal of clinical utility, challenges, and future research-A consensus document endorsed by the International Society of Electrocardiology and the International Society for Holter and Noninvasive Electrocardiology
.
Circ Arrhythm Electrophysiol
 
2022
;
15
:
e010435
.

303

Kreimer
 
F
,
Gotzmann
 
M
.
Left atrial cardiomyopathy—a challenging diagnosis
.
Front Cardiovasc Med
 
2022
;
9
:
942385
.

304

Bayés de Luna
 
A
,
Platonov
 
P
,
Cosio
 
FG
,
Cygankiewicz
 
I
,
Pastore
 
C
,
Baranowski
 
R
 et al.  
Interatrial blocks. A separate entity from left atrial enlargement: a consensus report
.
J Electrocardiol
 
2012
;
45
:
445
51
.

305

Tsao
 
CW
,
Josephson
 
ME
,
Hauser
 
TH
,
O'Halloran
 
TD
,
Agarwal
 
A
,
Manning
 
WJ
 et al.  
Accuracy of electrocardiographic criteria for atrial enlargement: validation with cardiovascular magnetic resonance
.
J Cardiovasc Magn Reson
 
2008
;
10
:
7
.

306

Bayés de Luna
 
A
,
Cladellas
 
M
,
Oter
 
R
,
Torner
 
P
,
Guindo
 
J
,
Martí
 
V
 et al.  
Interatrial conduction block and retrograde activation of the left atrium and paroxysmal supraventricular tachyarrhythmia
.
Eur Heart J
 
1988
;
9
:
1112
8
.

307

Power
 
DA
,
Lampert
 
J
,
Camaj
 
A
,
Bienstock
 
SW
,
Kocovic
 
N
,
Bayes-Genis
 
A
 et al.  
Cardiovascular complications of interatrial conduction block: JACC state-of-the-art review
.
J Am Coll Cardiol
 
2022
;
79
:
1199
211
.

308

Wu
 
JT
,
Long
 
DY
,
Dong
 
JZ
,
Wang
 
SL
,
Fan
 
XW
,
Yang
 
HT
 et al.  
Advanced interatrial block predicts clinical recurrence of atrial fibrillation after catheter ablation
.
J Cardiol
 
2016
;
68
:
352
6
.

309

Nielsen
 
JB
,
Kühl
 
JT
,
Pietersen
 
A
,
Graff
 
C
,
Lind
 
B
,
Struijk
 
JJ
 et al.  
P-wave duration and the risk of atrial fibrillation: results from the Copenhagen ECG study
.
Heart rhythm
 
2015
;
12
:
1887
95
.

310

Auricchio
 
A
,
Özkartal
 
T
,
Salghetti
 
F
,
Neumann
 
L
,
Pezzuto
 
S
,
Gharaviri
 
A
 et al.  
Short P-wave duration is a marker of higher rate of atrial fibrillation recurrences after pulmonary vein isolation: new insights into the pathophysiological mechanisms through computer simulations
.
J Am Heart Assoc
 
2021
;
10
:
e018572
.

311

Truong
 
QA
,
Charipar
 
EM
,
Ptaszek
 
LM
,
Taylor
 
C
,
Fontes
 
JD
,
Kriegel
 
M
 et al.  
Usefulness of electrocardiographic parameters as compared with computed tomography measures of left atrial volume enlargement: from the ROMICAT trial
.
J Electrocardiol
 
2011
;
44
:
257
64
.

312

Leshem
 
E
,
Zilberman
 
I
,
Tschabrunn
 
CM
,
Barkagan
 
M
,
Contreras-Valdes
 
FM
,
Govari
 
A
 et al.  
High-power and short-duration ablation for pulmonary vein isolation: biophysical characterization
.
JACC Clin Electrophysiol
 
2018
;
4
:
467
79
.

313

Müller-Edenborn
 
B
,
Minners
 
J
,
Kocher
 
S
,
Chen
 
J
,
Zeh
 
W
,
Lehrmann
 
H
 et al.  
Amplified P-wave duration predicts new-onset atrial fibrillation in patients with heart failure with preserved ejection fraction
.
Clin Res Cardiol
 
2020
;
109
:
978
87
.

314

Dhala
 
A
,
Underwood
 
D
,
Leman
 
R
,
Madu
 
E
,
Baugh
 
D
,
Ozawa
 
Y
 et al.  
Signal-averaged P-wave analysis of normal controls and patients with paroxysmal atrial fibrillation: a study in gender differences, age dependence, and reproducibility
.
Clin Cardiol
 
2002
;
25
:
525
31
.

315

Okumura
 
Y
,
Watanabe
 
I
,
Ohkubo
 
K
,
Ashino
 
S
,
Kofune
 
M
,
Hashimoto
 
K
 et al.  
Prediction of the efficacy of pulmonary vein isolation for the treatment of atrial fibrillation by the signal-averaged P-wave duration
.
Pacing Clin Electrophysiol
 
2007
;
30
:
304
13
.

316

Zink
 
MD
,
Laureanti
 
R
,
Hermans
 
BJM
,
Pison
 
L
,
Verheule
 
S
,
Philippens
 
S
 et al.  
Extended ECG improves classification of paroxysmal and persistent atrial fibrillation based on P- and f-waves
.
Front Physiol
 
2022
;
13
:
779826
.

317

Farinha
 
JM
,
Gupta
 
D
,
Lip
 
GYH
.
Frequent premature atrial contractions as a signalling marker of atrial cardiomyopathy, incident atrial fibrillation, and stroke
.
Cardiovasc Res
 
2023
;
119
:
429
39
.

318

Py
 
A
,
Schaaf
 
M
,
Duhamel
 
S
,
Si-Mohamed
 
S
,
Daher
 
J
,
Altman
 
M
 et al.  
Atrial premature activity detected after an ischaemic stroke unveils atrial myopathy
.
Arch Cardiovasc Dis
 
2020
;
113
:
227
36
.

319

Attia
 
ZI
,
Noseworthy
 
PA
,
Lopez-Jimenez
 
F
,
Asirvatham
 
SJ
,
Deshmukh
 
AJ
,
Gersh
 
BJ
 et al.  
An artificial intelligence-enabled ECG algorithm for the identification of patients with atrial fibrillation during sinus rhythm: a retrospective analysis of outcome prediction
.
Lancet
 
2019
;
394
:
861
7
.

320

Noseworthy
 
PA
,
Attia
 
ZI
,
Behnken
 
EM
,
Giblon
 
RE
,
Bews
 
KA
,
Liu
 
S
 et al.  
Artificial intelligence-guided screening for atrial fibrillation using electrocardiogram during sinus rhythm: a prospective non-randomised interventional trial
.
Lancet
 
2022
;
400
:
1206
12
.

321

Liao
 
S
,
Ragot
 
D
,
Nayyar
 
S
,
Suszko
 
A
,
Zhang
 
Z
,
Wang
 
B
 et al.  
Deep learning classification of unipolar electrograms in human atrial fibrillation: application in focal source mapping
.
Front Physiol
 
2021
;
12
:
704122
.

322

Lebert
 
J
,
Ravi
 
N
,
Fenton
 
FH
,
Christoph
 
J
.
Rotor localization and phase mapping of cardiac excitation waves using deep neural networks
.
Front Physiol
 
2021
;
12
:
782176
.

323

He
 
J
,
Tse
 
G
,
Korantzopoulos
 
P
,
Letsas
 
KP
,
Ali-Hasan-Al-Saegh
 
S
,
Kamel
 
H
 et al.  
P-wave indices and risk of ischemic stroke: a systematic review and meta-analysis
.
Stroke
 
2017
;
48
:
2066
72
.

324

Larsen
 
BS
,
Kumarathurai
 
P
,
Falkenberg
 
J
,
Nielsen
 
OW
,
Sajadieh
 
A
.
Excessive atrial ectopy and short atrial runs increase the risk of stroke beyond incident atrial fibrillation
.
J Am Coll Cardiol
 
2015
;
66
:
232
41
.

325

Sajeev
 
JK
,
Koshy
 
AN
,
Dewey
 
H
,
Kalman
 
JM
,
Bhatia
 
M
,
Roberts
 
L
 et al.  
Poor reliability of P-wave terminal force V(1) in ischemic stroke
.
J Electrocardiol
 
2019
;
52
:
47
52
.

326

Lattanzi
 
S
,
Cagnetti
 
C
,
Pulcini
 
A
,
Morelli
 
M
,
Maffei
 
S
,
Provinciali
 
L
 et al.  
The P-wave terminal force in embolic strokes of undetermined source
.
J Neurol Sci
 
2017
;
375
:
175
8
.

327

Stalikas
 
N
,
Doundoulakis
 
I
,
Karagiannidis
 
E
,
Kartas
 
A
,
Gavriilaki
 
M
,
Sofidis
 
G
 et al.  
Prevalence of markers of atrial cardiomyopathy in embolic stroke of undetermined source: a systematic review
.
Eur J Intern Med
 
2022
;
99
:
38
44
.

328

Li
 
Z
,
Wang
 
X
,
Liu
 
Q
,
Li
 
C
,
Gao
 
J
,
Yang
 
Y
 et al.  
Atrial cardiomyopathy markers predict ischemic cerebrovascular events independent of atrial fibrillation in patients with acute myocardial infarction
.
Front Cardiovasc Med
 
2022
;
9
:
1025842
.

329

Froehlich
 
L
,
Meyre
 
P
,
Aeschbacher
 
S
,
Blum
 
S
,
Djokic
 
D
,
Kuehne
 
M
 et al.  
Left atrial dimension and cardiovascular outcomes in patients with and without atrial fibrillation: a systematic review and meta-analysis
.
Heart
 
2019
;
105
:
1884
91
.

330

Kamel
 
H
,
Bartz
 
TM
,
Elkind
 
MSV
,
Okin
 
PM
,
Thacker
 
EL
,
Patton
 
KK
 et al.  
Atrial cardiopathy and the risk of ischemic stroke in the CHS (Cardiovascular Health Study)
.
Stroke
 
2018
;
49
:
980
6
.

331

Tan
 
S
,
Ho
 
C
,
Teo
 
YN
,
Teo
 
YH
,
Chan
 
MY
,
Lee
 
CH
 et al.  
Prevalence and incidence of stroke, white matter hyperintensities, and silent brain infarcts in patients with chronic heart failure: a systematic review, meta-analysis, and meta-regression
.
Front Cardiovasc Med
 
2022
;
9
:
967197
.

332

Mosenzon
 
O
,
Cheng
 
AY
,
Rabinstein
 
AA
,
Sacco
 
S
.
Diabetes and stroke: what are the connections?
 
J Stroke
 
2023
;
25
:
26
38
.

333

Jung
 
H
,
Yang
 
PS
,
Kim
 
D
,
Jang
 
E
,
Yu
 
HT
,
Kim
 
TH
 et al.  
Associations of hypertension burden on subsequent dementia: a population-based cohort study
.
Sci Rep
 
2021
;
11
:
12291
.

334

Lee
 
SR
,
Park
 
CS
,
Choi
 
EK
,
Ahn
 
HJ
,
Han
 
KD
,
Oh
 
S
 et al.  
Hypertension burden and the risk of new-onset atrial fibrillation: a nationwide population-based study
.
Hypertension
 
2021
;
77
:
919
28
.

335

Okamoto
 
M
,
Fujii
 
Y
,
Makita
 
Y
,
Kajihara
 
K
,
Yamasaki
 
S
,
Iwamoto
 
A
 et al.  
Left atrial appendage function in patients with systemic embolism in spite of in sinus rhythm
.
J Am Soc Echocardiogr
 
2006
;
19
:
211
4
.

336

Lip
 
GY
.
Hypertension and the prothrombotic state
.
J Hum Hypertens
 
2000
;
14
:
687
90
.

337

Lip
 
GY
,
Gibbs
 
CR
.
Does heart failure confer a hypercoagulable state? Virchow's triad revisited
.
J Am Coll Cardiol
 
1999
;
33
:
1424
6
.

338

Ruf
 
L
,
Bukowska
 
A
,
Gardemann
 
A
,
Goette
 
A
.
Coagulation factor Xa has no effects on the expression of PAR1, PAR2, and PAR4 and no proinflammatory effects on HL-1 cells
.
Cells.
 
2023
;
12
:
2849
.

339

Goette
 
A
,
Lendeckel
 
U
.
Atrial cardiomyopathy: pathophysiology and clinical consequences
.
Cells
 
2021
;
10
:
2605
.

340

Orlandi
 
F
,
Jakil
 
C
,
Damiani
 
G
,
Lauricella
 
S
,
Maggio
 
A
,
D'Alcamo
 
E
 et al.  
First trimester fetal blood sampling
.
Acta Eur Fertil
 
1988
;
19
:
23
4
.

341

Goette
 
A
.
Antithrombotic therapy after coronary artery stenting in atrial fibrillation: dual therapy encompassing NOAC plus P2Y12 inhibitor is ready for prime time!
.
Ann Transl Med
 
2019
;
7
:
S270
.

342

Bukowska
 
A
,
Hammwöhner
 
M
,
Corradi
 
D
,
Mahardhika
 
W
,
Goette
 
A
.
Atrial thrombogenesis in atrial fibrillation : results from atrial fibrillation models and AF-patients
.
Herzschrittmacherther Elektrophysiol
 
2018
;
29
:
76
83
.

343

Farina
 
E
,
Magni
 
E
,
Ambrosini
 
F
,
Manfredini
 
R
,
Binda
 
A
,
Sina
 
C
 et al.  
Neuropsychological deficits in asymptomatic atrial fibrillation
.
Acta Neurol Scand
 
1997
;
96
:
310
6
.

344

Gaita
 
F
,
Corsinovi
 
L
,
Anselmino
 
M
,
Raimondo
 
C
,
Pianelli
 
M
,
Toso
 
E
 et al.  
Prevalence of silent cerebral ischemia in paroxysmal and persistent atrial fibrillation and correlation with cognitive function
.
J Am Coll Cardiol
 
2013
;
62
:
1990
7
.

345

Lappegård
 
KT
,
Pop-Purceleanu
 
M
,
van Heerde
 
W
,
Sexton
 
J
,
Tendolkar
 
I
,
Pop
 
G
.
Improved neurocognitive functions correlate with reduced inflammatory burden in atrial fibrillation patients treated with intensive cholesterol lowering therapy
.
J Neuroinflammation
 
2013
;
10
:
78
.

346

Barber
 
M
,
Tait
 
RC
,
Scott
 
J
,
Rumley
 
A
,
Lowe
 
GD
,
Stott
 
DJ
.
Dementia in subjects with atrial fibrillation: hemostatic function and the role of anticoagulation
.
J Thromb Haemost
 
2004
;
2
:
1873
8
.

347

Poels
 
MM
,
Ikram
 
MA
,
van der Lugt
 
A
,
Hofman
 
A
,
Niessen
 
WJ
,
Krestin
 
GP
 et al.  
Cerebral microbleeds are associated with worse cognitive function: the Rotterdam Scan Study
.
Neurology
 
2012
;
78
:
326
33
.

348

Lavy
 
S
,
Stern
 
S
,
Melamed
 
E
,
Cooper
 
G
,
Keren
 
A
,
Levy
 
P
.
Effect of chronic atrial fibrillation on regional cerebral blood flow
.
Stroke
 
1980
;
11
:
35
8
.

349

Jefferson
 
AL
,
Himali
 
JJ
,
Beiser
 
AS
,
Au
 
R
,
Massaro
 
JM
,
Seshadri
 
S
 et al.  
Cardiac index is associated with brain aging: the Framingham Heart Study
.
Circulation
 
2010
;
122
:
690
7
.

350

Karadag
 
B
,
Ozyigit
 
T
,
Ozben
 
B
,
Kayaoglu
 
S
,
Altuntas
 
Y
.
Relationship between left atrial volume index and cognitive decline in elderly patients with sinus rhythm
.
J Clin Neurosci
 
2013
;
20
:
1074
8
.

351

Gutierrez
 
A
,
Norby
 
FL
,
Maheshwari
 
A
,
Rooney
 
MR
,
Gottesman
 
RF
,
Mosley
 
TH
 et al.  
Association of abnormal P-wave indices with dementia and cognitive decline over 25 years: ARIC-NCS (the atherosclerosis risk in communities neurocognitive study)
.
J Am Heart Assoc
 
2019
;
8
:
e014553
.

352

Zhang
 
MJ
,
Norby
 
FL
,
Lutsey
 
PL
,
Mosley
 
TH
,
Cogswell
 
RJ
,
Konety
 
SH
 et al.  
Association of left atrial enlargement and atrial fibrillation with cognitive function and decline: the ARIC-NCS
.
J Am Heart Assoc
 
2019
;
8
:
e013197
.

353

Wang
 
W
,
Zhang
 
MJ
,
Inciardi
 
RM
,
Norby
 
FL
,
Johansen
 
MC
,
Parikh
 
R
 et al.  
Association of echocardiographic measures of left atrial function and size with incident dementia
.
JAMA
 
2022
;
327
:
1138
48
.

354

Goette
 
A
,
Röcken
 
C
.
Atrial amyloidosis and atrial fibrillation: a gender-dependent “arrhythmogenic substrate”?
 
Eur Heart J
 
2004
;
25
:
1185
6
.

355

Savelieva
 
I
,
Kakouros
 
N
,
Kourliouros
 
A
,
Camm
 
AJ
.
Upstream therapies for management of atrial fibrillation: review of clinical evidence and implications for European Society of Cardiology guidelines. Part I: primary prevention
.
Europace
 
2011
;
13
:
308
28
.

356

Pathak
 
RK
,
Elliott
 
A
,
Middeldorp
 
ME
,
Meredith
 
M
,
Mehta
 
AB
,
Mahajan
 
R
 et al.  
Impact of CARDIOrespiratory FITness on arrhythmia recurrence in obese individuals with atrial fibrillation: the CARDIO-FIT study
.
J Am Coll Cardiol
 
2015
;
66
:
985
96
.

357

Pathak
 
RK
,
Middeldorp
 
ME
,
Meredith
 
M
,
Mehta
 
AB
,
Mahajan
 
R
,
Wong
 
CX
 et al.  
Long-term effect of goal-directed weight management in an atrial fibrillation cohort: a long-term follow-up study (LEGACY)
.
J Am Coll Cardiol
 
2015
;
65
:
2159
69
.

358

Goette
 
A
,
Schön
 
N
,
Kirchhof
 
P
,
Breithardt
 
G
,
Fetsch
 
T
,
Häusler
 
KG
 et al.  
Angiotensin II-antagonist in paroxysmal atrial fibrillation (ANTIPAF) trial
.
Circ Arrhythm Electrophysiol
 
2012
;
5
:
43
51
.

359

Neefs
 
J
,
van den Berg
 
NW
,
Limpens
 
J
,
Berger
 
WR
,
Boekholdt
 
SM
,
Sanders
 
P
 et al.  
Aldosterone pathway blockade to prevent atrial fibrillation: a systematic review and meta-analysis
.
Int J Cardiol
 
2017
;
231
:
155
61
.

360

Rienstra
 
M
,
Hobbelt
 
AH
,
Alings
 
M
,
Tijssen
 
JGP
,
Smit
 
MD
,
Brügemann
 
J
 et al.  
Targeted therapy of underlying conditions improves sinus rhythm maintenance in patients with persistent atrial fibrillation: results of the RACE 3 trial
.
Eur Heart J
 
2018
;
39
:
2987
96
.

361

Oraii
 
A
,
Healey
 
JS
,
Kowalik
 
K
,
Pandey
 
AK
,
Benz
 
AP
,
Wong
 
JA
 et al.  
Mineralocorticoid receptor antagonists and atrial fibrillation: a meta-analysis of clinical trials
.
Eur Heart J
 
2024
;
45
:
756
74
.

362

Goette
 
A
,
Lendeckel
 
U
,
Kuchenbecker
 
A
,
Bukowska
 
A
,
Peters
 
B
,
Klein
 
HU
 et al.  
Cigarette smoking induces atrial fibrosis in humans via nicotine
.
Heart
 
2007
;
93
:
1056
63
.

363

Bukowska
 
A
,
Nikonova
 
Y
,
Wolke
 
C
,
Lendeckel
 
U
,
Kockskämper
 
J
,
Goette
 
A
.
Anti-inflammatory effects of endothelin receptor blockade in left atrial tissue of spontaneously hypertensive rats
.
Int J Cardiol Heart Vasc
 
2022
;
42
:
101088
.

364

Goette
 
A
,
Hammwöhner
 
M
,
Bukowska
 
A
,
Scalera
 
F
,
Martens-Lobenhoffer
 
J
,
Dobrev
 
D
 et al.  
The impact of rapid atrial pacing on ADMA and endothelial NOS
.
Int J Cardiol
 
2012
;
154
:
141
6
.

365

McManus
 
DD
,
Yin
 
X
,
Gladstone
 
R
,
Vittinghoff
 
E
,
Vasan
 
RS
,
Larson
 
MG
 et al.  
Alcohol consumption, left atrial diameter, and atrial fibrillation
.
J Am Heart Assoc
 
2016
;
5
:
e004060
.

366

Voskoboinik
 
A
,
Kalman
 
JM
,
De Silva
 
A
,
Nicholls
 
T
,
Costello
 
B
,
Nanayakkara
 
S
 et al.  
Alcohol abstinence in drinkers with atrial fibrillation
.
N Engl J Med
 
2020
;
382
:
20
8
.

367

Deng
 
F
,
Raza
 
A
,
Guo
 
J
.
Treating obstructive sleep apnea with continuous positive airway pressure reduces risk of recurrent atrial fibrillation after catheter ablation: a meta-analysis
.
Sleep Med
 
2018
;
46
:
5
11
.

368

Khatib
 
R
,
Joseph
 
P
,
Briel
 
M
,
Yusuf
 
S
,
Healey
 
J
.
Blockade of the renin-angiotensin-aldosterone system (RAAS) for primary prevention of non-valvular atrial fibrillation: a systematic review and meta analysis of randomized controlled trials
.
Int J Cardiol
 
2013
;
165
:
17
24
.

369

Nalliah
 
CJ
,
Wong
 
GR
,
Lee
 
G
,
Voskoboinik
 
A
,
Kee
 
K
,
Goldin
 
J
 et al.  
Impact of CPAP on the atrial fibrillation substrate in obstructive sleep apnea: the SLEEP-AF study
.
JACC Clin Electrophysiol
 
2022
;
8
:
869
77
.

370

Kapelios
 
CJ
,
Murrow
 
JR
,
Nührenberg
 
TG
,
Montoro Lopez
 
MN
.
Effect of mineralocorticoid receptor antagonists on cardiac function in patients with heart failure and preserved ejection fraction: a systematic review and meta-analysis of randomized controlled trials
.
Heart Fail Rev
 
2019
;
24
:
367
77
.

371

Sampaio Rodrigues
 
T
,
Garcia Quarto
 
LJ
,
Nogueira
 
SC
,
Koshy
 
AN
,
Mahajan
 
R
,
Sanders
 
P
 et al.  
Incidence and progression of atrial fibrillation in patients with and without heart failure using mineralocorticoid receptor antagonists: a meta-analysis
.
Clin Res Cardiol
 
2024
;
113
:
884
97
.

372

Kumagai
 
K
,
Nakashima
 
H
,
Urata
 
H
,
Gondo
 
N
,
Arakawa
 
K
,
Saku
 
K
.
Effects of angiotensin II type 1 receptor antagonist on electrical and structural remodeling in atrial fibrillation
.
J Am Coll Cardiol
 
2003
;
41
:
2197
204
.

373

Li
 
Y
,
Li
 
WM
,
Gong
 
YT
,
Li
 
BX
,
Liu
 
W
,
Han
 
W
 et al.  
The effects of cilazapril and valsartan on the mRNA and protein expressions of atrial calpains and atrial structural remodeling in atrial fibrillation dogs
.
Basic Res Cardiol
 
2007
;
102
:
245
56
.

374

Takemoto
 
Y
,
Ramirez
 
RJ
,
Kaur
 
K
,
Salvador-Montañés
 
O
,
Ponce-Balbuena
 
D
,
Ramos-Mondragón
 
R
 et al.  
Eplerenone reduces atrial fibrillation burden without preventing atrial electrical remodeling
.
J Am Coll Cardiol
 
2017
;
70
:
2893
905
.

375

Du
 
L
,
Qin
 
M
,
Yi
 
Y
,
Chen
 
X
,
Jiang
 
W
,
Zhou
 
L
 et al.  
Eplerenone prevents atrial fibrosis via the TGF-β signaling pathway
.
Cardiology
 
2017
;
138
:
55
62
.

376

Li
 
D
,
Shinagawa
 
K
,
Pang
 
L
,
Leung
 
TK
,
Cardin
 
S
,
Wang
 
Z
 et al.  
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
14
.

377

Mancini
 
GBJ
,
O'Meara
 
E
,
Zieroth
 
S
,
Bernier
 
M
,
Cheng
 
AYY
,
Cherney
 
DZI
 et al.  
2022 Canadian Cardiovascular Society Guideline for use of GLP-1 receptor agonists and SGLT2 inhibitors for cardiorenal risk reduction in adults
.
Can J Cardiol
 
2022
;
38
:
1153
67
.

378

Shao
 
Q
,
Meng
 
L
,
Lee
 
S
,
Tse
 
G
,
Gong
 
M
,
Zhang
 
Z
 et al.  
Empagliflozin, a sodium glucose co-transporter-2 inhibitor, alleviates atrial remodeling and improves mitochondrial function in high-fat diet/streptozotocin-induced diabetic rats
.
Cardiovasc Diabetol
 
2019
;
18
:
165
.

379

Zelniker
 
TA
,
Bonaca
 
MP
,
Furtado
 
RHM
,
Mosenzon
 
O
,
Kuder
 
JF
,
Murphy
 
SA
 et al.  
Effect of dapagliflozin on atrial fibrillation in patients with type 2 diabetes mellitus: insights from the DECLARE-TIMI 58 trial
.
Circulation
 
2020
;
141
:
1227
34
.

380

Hohnloser
 
SH
,
Crijns
 
HJ
,
van Eickels
 
M
,
Gaudin
 
C
,
Page
 
RL
,
Torp-Pedersen
 
C
 et al.  
Effect of dronedarone on cardiovascular events in atrial fibrillation
.
N Engl J Med
 
2009
;
360
:
668
78
.

381

Blomström-Lundqvist
 
C
,
Naccarelli
 
GV
,
McKindley
 
DS
,
Bigot
 
G
,
Wieloch
 
M
,
Hohnloser
 
SH
.
Effect of dronedarone vs. placebo on atrial fibrillation progression: a post hoc analysis from ATHENA trial
.
Europace
 
2023
;
25
:
845
54
.

382

Kirchhof
 
P
,
Camm
 
AJ
,
Goette
 
A
,
Brandes
 
A
,
Eckardt
 
L
,
Elvan
 
A
 et al.  
Early rhythm-control therapy in patients with atrial fibrillation
.
N Engl J Med
 
2020
;
383
:
1305
16
.

383

Rillig
 
A
,
Borof
 
K
,
Breithardt
 
G
,
Camm
 
AJ
,
Crijns
 
H
,
Goette
 
A
 et al.  
Early rhythm control in patients with atrial fibrillation and high comorbidity burden
.
Circulation
 
2022
;
146
:
836
47
.

384

Masuda
 
M
,
Sekiya
 
K
,
Asai
 
M
,
Iida
 
O
,
Okamoto
 
S
,
Ishihara
 
T
 et al.  
Influence of catheter ablation for atrial fibrillation on atrial and ventricular functional mitral regurgitation
.
ESC Heart Fail
 
2022
;
9
:
1901
13
.

385

Rotter
 
M
,
Jaïs
 
P
,
Vergnes
 
MC
,
Nurden
 
P
,
Takahashi
 
Y
,
Sanders
 
P
 et al.  
Decline in C-reactive protein after successful ablation of long-lasting persistent atrial fibrillation
.
J Am Coll Cardiol
 
2006
;
47
:
1231
3
.

386

Kuck
 
KH
,
Lebedev
 
DS
,
Mikhaylov
 
EN
,
Romanov
 
A
,
Gellér
 
L
,
Kalējs
 
O
 et al.  
Catheter ablation or medical therapy to delay progression of atrial fibrillation: the randomized controlled atrial fibrillation progression trial (ATTEST)
.
Europace
 
2021
;
23
:
362
9
.

387

Hof
 
IE
,
Velthuis
 
BK
,
Chaldoupi
 
SM
,
Wittkampf
 
FH
,
van Driel
 
VJ
,
van der Heijden
 
JF
 et al.  
Pulmonary vein antrum isolation leads to a significant decrease of left atrial size
.
Europace
 
2011
;
13
:
371
5
.

388

Canpolat
 
U
,
Aytemir
 
K
,
Özer
 
N
,
Oto
 
A
.
The impact of cryoballoon-based catheter ablation on left atrial structural and potential electrical remodeling in patients with paroxysmal atrial fibrillation
.
J Interv Card Electrophysiol
 
2015
;
44
:
131
9
.

389

Yoshida
 
K
,
Yui
 
Y
,
Kimata
 
A
,
Koda
 
N
,
Kato
 
J
,
Baba
 
M
 et al.  
Troponin elevation after radiofrequency catheter ablation of atrial fibrillation: relevance to AF substrate, procedural outcomes, and reverse structural remodeling
.
Heart rhythm
 
2014
;
11
:
1336
42
.

390

Neefs
 
J
,
Wesselink
 
R
,
van den Berg
 
NWE
,
de Jong
 
J
,
Piersma
 
FR
,
van Boven
 
WP
 et al.  
Thoracoscopic surgical atrial fibrillation ablation in patients with an extremely enlarged left atrium
.
J Interv Card Electrophysiol
 
2022
;
64
:
469
78
.

391

Xiong
 
B
,
Li
 
D
,
Wang
 
J
,
Gyawali
 
L
,
Jing
 
J
,
Su
 
L
.
The effect of catheter ablation on left atrial size and function for patients with atrial fibrillation: an updated meta-analysis
.
PLoS One
 
2015
;
10
:
e0129274
.

392

Pump
 
A
,
Di Biase
 
L
,
Price
 
J
,
Mohanty
 
P
,
Bai
 
R
,
Santangeli
 
P
 et al.  
Efficacy of catheter ablation in nonparoxysmal atrial fibrillation patients with severe enlarged left atrium and its impact on left atrial structural remodeling
.
J Cardiovasc Electrophysiol
 
2013
;
24
:
1224
31
.

393

Bansilal
 
S
,
Bloomgarden
 
Z
,
Halperin
 
JL
,
Hellkamp
 
AS
,
Lokhnygina
 
Y
,
Patel
 
MR
 et al.  
Efficacy and safety of rivaroxaban in patients with diabetes and nonvalvular atrial fibrillation: the rivaroxaban once-daily, oral, direct factor Xa inhibition compared with vitamin K antagonism for prevention of stroke and embolism trial in atrial fibrillation (ROCKET AF trial)
.
Am Heart J
 
2015
;
170
:
675
82.e8
.

394

Muellerleile
 
K
,
Groth
 
M
,
Steven
 
D
,
Hoffmann
 
BA
,
Saring
 
D
,
Radunski
 
UK
 et al.  
Cardiovascular magnetic resonance demonstrates reversible atrial dysfunction after catheter ablation of persistent atrial fibrillation
.
J Cardiovasc Electrophysiol
 
2013
;
24
:
762
7
.

395

Cho
 
DH
,
Kim
 
YG
,
Choi
 
J
,
Kim
 
HD
,
Kim
 
MN
,
Shim
 
J
 et al.  
Atrial cardiomyopathy with impaired functional reserve in patients with paroxysmal atrial fibrillation
.
J Am Soc Echocardiogr
 
2023
;
36
:
180
8
.

396

Li-Saw-Hee
 
FL
,
Blann
 
AD
,
Gurney
 
D
,
Lip
 
GY
.
Plasma von Willebrand factor, fibrinogen and soluble P-selectin levels in paroxysmal, persistent and permanent atrial fibrillation. Effects of cardioversion and return of left atrial function
.
Eur Heart J
 
2001
;
22
:
1741
7
.

397

Yu
 
CM
,
Fang
 
F
,
Zhang
 
Q
,
Yip
 
GW
,
Li
 
CM
,
Chan
 
JY
 et al.  
Improvement of atrial function and atrial reverse remodeling after cardiac resynchronization therapy for heart failure
.
J Am Coll Cardiol
 
2007
;
50
:
778
85
.

398

Burri
 
H
,
Jastrzebski
 
M
,
Cano
 
Ó
,
Čurila
 
K
,
de Pooter
 
J
,
Huang
 
W
 et al.  
EHRA clinical consensus statement on conduction system pacing implantation: executive summary. Endorsed by the Asia-Pacific Heart Rhythm Society (APHRS), Canadian Heart Rhythm Society (CHRS) and Latin-American Heart Rhythm Society (LAHRS)
.
Europace
 
2023
;
25
:
1237
48
.

399

Su
 
L
,
Cai
 
M
,
Wu
 
S
,
Wang
 
S
,
Xu
 
T
,
Vijayaraman
 
P
 et al.  
Long-term performance and risk factors analysis after permanent his-bundle pacing and atrioventricular node ablation in patients with atrial fibrillation and heart failure
.
Europace
 
2020
;
22
:
ii19
26
.

400

Marsan
 
NA
,
Maffessanti
 
F
,
Tamborini
 
G
,
Gripari
 
P
,
Caiani
 
E
,
Fusini
 
L
 et al.  
Left atrial reverse remodeling and functional improvement after mitral valve repair in degenerative mitral regurgitation: a real-time 3-dimensional echocardiography study
.
Am Heart J
 
2011
;
161
:
314
21
.

401

John
 
B
,
Stiles
 
MK
,
Kuklik
 
P
,
Brooks
 
AG
,
Chandy
 
ST
,
Kalman
 
JM
 et al.  
Reverse remodeling of the atria after treatment of chronic stretch in humans: implications for the atrial fibrillation substrate
.
J Am Coll Cardiol
 
2010
;
55
:
1217
26
.

402

Goette
 
A
,
Auricchio
 
A
,
Boriani
 
G
,
Braunschweig
 
F
,
Terradellas
 
JB
,
Burri
 
H
 et al.  
EHRA white paper: knowledge gaps in arrhythmia management-status 2019
.
Europace
 
2019
;
21
:
993
4
.

Author notes

Developed in partnership with the European Heart Rhythm Association (EHRA), (a registered branch of the European Society of Cardiology (ESC)) the Heart Rhythm Society (HRS), the Asia Pacific Heart Rhythm Society (APHRS), the Latin America Heart Rhythm Society (LAHRS).

Conflict of interest: L.M.: (2022) Direct personal payment from healthcare industry: speaker fees, honoraria, consultancy, advisory board fees, investigator, committee member, etc. Biosense Webster: Atrial Fibrillation (AF); Boston Scientific, Medtronic, Abbott Medical: Atrial Fibrillation (AF), Device Therapy. Research funding from healthcare industry under your direct/personal responsibility (to department or institution). Johnson & Johnson: Research projects, PI; Boston Scientific, Medtronic, Biotronik, Abbott Medical: Research projects, PI.

This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial License (https://creativecommons.org/licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact reprints@oup.com for reprints and translation rights for reprints. All other permissions can be obtained through our RightsLink service via the Permissions link on the article page on our site—for further information please contact journals.permissions@oup.com.

Supplementary data