IMR Press / RCM / Volume 24 / Issue 4 / DOI: 10.31083/j.rcm2404119
Open Access Review
Gene Polymorphism and Recurrent Atrial Fibrillation after Catheter Ablation: A Comprehensive Review
Show Less
1 Department of Cardiology, The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, 213000 Changzhou, Jiangsu, China
2 State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 100037 Beijing, China
*Correspondence: zzymm1983@163.com (Zhen-Yan Zhu); lijianjun938@126.com (Jian-Jun Li)
These authors contributed equally.
Rev. Cardiovasc. Med. 2023, 24(4), 119; https://doi.org/10.31083/j.rcm2404119
Submitted: 3 November 2022 | Revised: 11 December 2022 | Accepted: 19 December 2022 | Published: 18 April 2023
Copyright: © 2023 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Atrial fibrillation (AF) is one of the most common cardiac arrhythmias, but its pathogenesis is still poorly understood. Catheter ablation is one of the most effective treatments for AF, but recurrence after ablation remains a challenge. There has been much research into the association of AF recurrence with several factors, including genetics. Over the past decade or so, significant advances have been made in the genetic architecture of atrial fibrillation. Genome-wide association studies (GWAS) have identified over 100 loci for genetic variants associated with atrial fibrillation. However, there is relatively little information on the systematic assessment of the genes related to AF recurrence after ablation. In this review article, we highlight the value of genetic polymorphisms in atrial fibrillation recurrence after catheter ablation and their potential mechanisms in the recurrence process to enhance our understanding of atrial fibrillation recurrence and contribute to individualized treatment strategies for patients with AF.

Keywords
gene polymorphism
atrial fibrillation
recurrence
catheter ablation
1. Introduction

Atrial fibrillation (AF) is one of the most common cardiac arrhythmias. The prevalence of AF varies slightly according to studies in different populations. Evidence suggests that the majority of AF in the general population is in the range of 1–5% [1, 2, 3]. According to the Framingham Heart Study (FHS), the prevalence of AF has increased threefold in the last 50 years [4]. The Global Burden of Disease project estimated the global prevalence of AF to be approximately 46.3 million in 2016 [5]. Patients with AF are at risk for cognitive decline, ischemic stroke, heart failure, myocardial infarction, and even death due to asynchronous atrial contractions, altered hemodynamics, and thromboembolism [6]. This has a tremendous negative impact on the patient, the whole family, and society.

Catheter ablation is currently one of the main tools for treating atrial fibrillation. It is mainly used to improve symptoms and control the heart rhythm in patients who have failed antiarrhythmic drug therapy or are intolerant to drugs [7]. In the 2020 ESC AF guidelines, catheter ablation has been identified as the first-line treatment for rhythm control in AF, especially in patients with paroxysmal AF and those need to improve their symptoms [8]. However, arrhythmia recurrence, defined as AF/atrial tachycardia/atrial flutter for 30 seconds or longer three months after ablation, remains a significant limitation of catheter ablation, according to the 2017 consensus. Late recurrence occurs in 50% or more of patients within five years [9]. The common risk factors known to influence ablation success include sex, age, duration of AF, body mass index, left atrial diameter, degree of left atrial scarring, and coexisting conditions, including hypertension, metabolic syndrome, heart failure, and sleep apnea [10], as shown in Fig. 1.

Fig. 1.

Risk factors associated with recurrence of atrial fibrillation. AF, atrial fibrillation; PV, pulmonary vein.

Genomics is a recently emerging field, and some evidence of family aggregation suggests that AF may be a genetically related disease [11]. This was further supported by a cohort study based on more than 5000 Icelandic patients with AF [12]. Over the past decades, more than 100 genes that are associated with AF have been identified. Some of these have been identified through classical linkage and family studies, while most were identified through functional or genome-wide association studies (GWAS). GWAS has identified many genetic variants (single nucleotide polymorphisms [SNPs]) associated with AF, and the role of genetic factors in the mechanism of AF is being increasingly recognized. Even a growing number of studies have found recurrence after AF ablation can be predicted using SNPs [13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26]. Predicting the outcome of catheter ablation by individual genetic characteristics has excellent potential to guide AF treatment strategies. In the following, we review the genetic variants associated with recurrence after AF ablation (See Table 1 (Ref. [13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28]) for relevant experimental studies and Fig. 2 for related genes and potential mechanisms).

Table 1.Genetic polymorphisms associated with recurrence of atrial fibrillation.
Author, year, and country AF type Study design SNP Adjacent gene OR/HR p-value Key fingdings
95% CI
Husser, 2010, Germany [13] drug-refractory paroxysmal or persistent AF Cohort study rs2200733 PITX2 ERAF: 2.108 (1.135–3.917) 0.018 The rs2200733 variant was independently associated with atrial fibrillation recurrence.
LRAF: 2.458 (1.061–5.693) 0.036
Chen, 2016, China [14] lone AF; paroxysmal AF; persistent AF Cohort study rs2200733 PITX2 OR: 1.714 0.01 The rs2200733 variant was independently associated with atrial fibrillation recurrence.
1.137–2.585
Zhao, 2017, China [15] paroxysmal or persistent AF Cohort study rs2200733 PITX2 HR: 1.766 (1.062–2.936) 0.028 The rs2200733 variant was independently associated with atrial fibrillation recurrence.
Choi, 2015, Korea [27] paroxysmal or persistent AF Observational study rs2200733 PITX2 HR: 1.01 (0.80–1.26) 0.963 The rs2200733 variant was not associated with atrial fibrillation recurrence.
Hu, 2016, China [28] paroxysmal or persistent AF Observational study rs2200733 PITX2 HR: 1.23 (0.87–1.76) 0.25 The rs2200733 variant was not associated with atrial fibrillation recurrence.
Park, 2017, Korea [16] long-standing persistent AF (L-PeAF) Observational study rs2106261 ZFHX3 OR: 2.70 (1.41–5.14) 0.003 The rs2106216 variant was independently associated with good responders after radiofrequency ablation for L-PeAF.
Tomomori, 2018, Japan [17] paroxysmal AF Retrospective single-center study rs2106261 ZFHX3 HR: 0.53 (0.29–0.98) 0.04 Patients with the minor (T) allele have a lower recurrence rate after ablation for paroxysmal atrial fibrillation.
Choi, 2015, Korea [27] paroxysmal or persistent AF Observational study rs2106261 ZFHX3 HR: 0.86 (0.71–1.04) 0.128 The rs2106261 variant was not associated with atrial fibrillation recurrence.
Ueberham, 2013, Germany [18] paroxysmal or persistent AF Cohort study I/D ACE OR: 2.25 (1.056–4.798) 0.036 ACE DD gene polymorphism was independently associated with atrial fibrillation recurrence.
Zhang, 2012, China [19] lone AF Prospective study I/D ACE RR: 2.35 (1.10–5.04) 0.028 The ACE gene DD genotype had a 2.35-fold increased risk for AF recurrence compared with the ACE gene II + ID genotype.
Hong, 2020, Korea [20] paroxysmal or persistent AF Prospective study rs3807989 CAV1 HR: 1.15 (1.02–1.31) 0.024 The rs3807989 variant was independently associated with atrial fibrillation recurrence.
Park, 2020, Korea [21] early-onset AF, <40 y Retrospective study rs11047543 SOX5 HR: 2.723 (1.358–5.461) 0.005 The rs11047543 variant was independently associated with atrial fibrillation recurrence.
Wutzler, 2013, Germany [22] drug-refractory AF Cohort study rs751141 EPHX2 12M: OR: 3.2 (1.237–8.276) 12M: 0.016 The rs751141 variant was independently associated with atrial fibrillation recurrence.
24M: OR: 6.076 (2.244–16.451) 24M: <0.0001
Shim, 2015, Korea [23] paroxysmal or persistent AF Cohort study rs1799983 (Glu298Asp) eNOS3 OR: 1.75 (1.07–2.86) 0.026 The rs1799983 variant was associated with early recurrence of AF (<3 months).
Wu, 2014, China [24] drug-refractory AF or permanent AF Retrospective study rs4845625 IL-6R ERAF: OR: 1.71 (1.20–2.38) 2.96 × 10-3 The rs4845625 variant was independently associated with atrial fibrillation recurrence.
LRAF: OR: 1.80 (1.28–2.55) 0.007
Hu, 2013, China [25] drug-refractory AF Unclear HO-1 promoter GT repeats HO-1 OR: 0.94 (0.90–0.99) 0.01 GT repeats polymorphism was independently associated with atrial fibrillation recurrence.
Amioka, 2019, Japan [26] paroxysmal AF Retrospective study rs3745297 (T > G, Ser96Ala) HRC HR: 2.66 (1.32–5.0) 0.007 Ser96Ala was independently associated with atrial fibrillation recurrence.
AF, atrial fibrillation; ACE, angiotensin-converting enzyme; CAV1, caveolin-1; eNOS3, endothelial nitric oxide synthase 3; IL-6 R, interleukin-6 receptor; HO-1, heme oxygenase-1; SNP, single nucleotide polymorphism; OR, odds ratio; HR, hazard ratio; RR, relative risk; ERAF, early recurrence of atrial fibrillation; LRAF, late recurrence of atrial fibrillation. L-PeAF, long-standing persistent AF.
Fig. 2.

Genes and potential mechanisms associated with recurrence of atrial fibrillation. AF, atrial fibrillation; PV, pulmonary vein; ACE, angiotensin-converting enzyme; Ang II, angiotensin II; TGF-β1, transforming growth factor β1; CAV1, caveolin-1; STAT3, signal transducer and activator of transcription 3; sEH, soluble epoxide hydrolase; EETs, epoxyeicosatrienoic acids; eNOS3, endothelial nitric oxide synthase3; IL-6 R, interleukin-6 receptor; HO-1, heme oxygenase-1; SR, sarcoplasmic reticulum.

2. Gene Polymorphisms
2.1 PITX2 SNP rs2200733

rs2200733 is located in the intron of chromosome 4q25 region. In recent years, several studies have suggested that SNP rs2200733 (genotypes CC, CT, and TT) may be associated with the risk of recurrence after radiofrequency ablation of atrial fibrillation [13, 14, 15, 29, 30, 31, 32, 33]. One hundred ninety-five patients with AF were studied in a German population for the first time by Husser et al. [13]. SNP rs2200733 (4q25) and SNP rs10033464 (4q25) were found to be associated with early recurrence (within seven days after ablation) and late recurrence (three-six months after ablation) after radiofrequency ablation of AF. Patients with the associated allelic variant had a 2-fold and 4-fold risk of early and late recurrence, respectively, compared with patients without the variant. This finding indicates the potential role of genotyping in the prediction of atrial fibrillation ablation therapy and in the management of the peri-interventional period. Shoemaker et al. [29] studied 378 patients in the Vanderbilt AF registry for atrial fibrillation ablation. By multivariate analysis, patients containing the risk allele (T) at the rs2200733 locus had a 24% reduction in recurrence-free time survival compared with patients with normal genes (survival time ratio 0.76; 95% CI 0.6–0.95; p = 0.016). The recurrence rate after radiofrequency (RF) ablation in this group of AF patients was 87.5%, which was significantly higher than that in patients with the normal genotype (40%). This further suggests that SNP rs220073 has some predictive value in the development of atrial fibrillation or atrial tachycardia after radiofrequency ablation of AF. Chen et al. [14] conducted a detailed study of 235 Chinese Han patients with atrial fibrillation using COX regression analysis. They found that the rs2200733 T allele was associated with the risk of recurrence after radiofrequency ablation in patients with atrial fibrillation. The association remained significant after correcting for various factors such as age, sex, and hypertension. They also found that rs2200733 was positively correlated with the right atrial diameter (RAD) and right superior pulmonary vein (RSPV) diameter. Patients with the TT genotype had larger right atrial volumes and thicker right upper pulmonary vein internal diameters than patients with the CC genotype. Zhao et al. [15] arrived at a similar conclusion. They also found that the TT gene group in rs2200733 was more likely to have AF recurrence than the TC + CC group and that this gene group increased the risk of AF recurrence 1.766-fold; in multivariate regression analysis, rs2200733 was an independent risk factor for AF recurrence. Shoemaker et al. [30] showed that SNP rs2200733 was associated with recurrence after RF ablation in patients with AF in a dominant model multivariate analysis (HR 1.3, 95% CI 1.1–1.6, p = 0.011), while SNP rs10033464 (4q25) was not significantly associated with recurrence after AF ablation in a meta-analysis of patients with AF undergoing RF ablation at three medical centers in Germany. Similarly, Rattanawong et al. [31] included 3322 patients with AF, performed a meta-analysis of their data, and found a strong association between SNP rs2200733 and recurrence after AF ablation ( relative risk (RR) 1.45, 95% CI 1.15–1.83, p = 0.002). The meta-analysis by Hu and He also supported these results [32, 33].

Additionally, the rs2200733 polymorphism has been shown to have no significant effect on recurrence after RF ablation of AF. Choi et al. [27] found that SNP rs2200733 did not correlate with recurrence after atrial fibrillation ablation in 1068 patients with atrial fibrillation undergoing radiofrequency ablation in a Korean population; SNP rs6843082 (4q25), SNP rs2106261 (16q22), and SNP rs13376333 (1q21) were also confirmed to be uncorrelated with recurrence in patients with AF. However, Hu et al. [28] used 189 Chinese patients with AF as their study population. They found that the rs2200733 variant was not associated with AF recurrence, but combining the rs7193343 risk allele enhanced the predictive value of AF recurrence.

In summary, there is no agreement about the rs2200733 polymorphism affecting the outcome after AF ablation. How these variants exert functional effects and how they predict rhythm outcomes after catheter ablation are not clear. rs2200733 is located in the intron region of chromosome 4q25 [34], and the gene closest to it upstream, PITX2, plays a crucial role in cardiac development. In humans, PITX2 is expressed in several isoforms, namely, PITX2a, PITX2b, PITX2c, and PITX2d. Only PITX2c is expressed asymmetrically in the heart and embryonic development. Deletion of this isoform may lead to malformations of the heart, abnormalities of the conduction system, and defects in cardiopulmonary muscle [35]. In earlier studies, it was found that PITX2c confers the morphology of the left atrium, and the deletion of PITX2c can give the left atrium the morphology of the right atrium [36]. Mouse embryos defective in the PITX2 gene have ectopic sinus nodes. Hill et al. [37] further revealed changes in the composition of PITX2-deficient atrial cardiomyocytes and the basis for asymmetric left-right defects in atrial cardiomyocytes. The pulmonary vein sleeve is a myofibrillar connection between the pulmonary veins and the left atrium and plays an essential role in atrial fibrillation. PITX2c is involved in the formation, differentiation, and proliferation of the pulmonary vein sleeve, and PITX2c-deficient mice lack the pulmonary vein sleeve [38]. Based on this finding, we hypothesized that carriers of the chromosome 4q25 variant might have a different pulmonary vein phenotype, which may affect the outcome of pulmonary vein catheter ablation. In addition, PITX2 regulates potassium and calcium channels; altered action potentials and changes in resting membrane potential are observed in PITX2 mutant models [39]. Recently, PITX2c-deficient mutants were found to lead to early monoidal and metabolic defects and electrical instability in a zebrafish study model, which led to the development of arrhythmias [40]. Taken together, we found that PITX2 is involved in the embryonic development of the atria, the formation of the pulmonary vein sleeve, and the regulation of atrial electricity. All of these structural and electrical remodelings may be risk factors for the recurrence of atrial fibrillation. Therefore, we speculate that rs2200733 may be involved in atrial fibrillation recurrence by regulating the expression of PITX2c.

2.2 ZFHX3 SNP rs2106261

The ZFHX3 gene (16q22) is the second gene highly associated with atrial fibrillation (AF), and it is associated with inflammation, stromal deposition, fibrosis, and atrial structure. Its SNPs are thought to increase susceptibility to AF [41, 42]. Interestingly, some recent reports suggest that SNP rs2106261 has predictive value for a good response after atrial fibrillation ablation. Park et al. [16] used multivariate logistic regression to study 141 patients with persistent AF and found that SNP rs2106216 had an excellent predictive value for patients with long-standing persistent AF without recurrence after radiofrequency ablation. In contrast, SNP rs7193343 (16q22) did not have this value. Tomomori et al. [17] genotyped rs2106261 (CC, CT, CT) and retrospectively studied 362 patients with paroxysmal AF. They found a lower AF recurrence rate in patients with the minor (T) allele using multivariate regression analysis (HR 0.53, p = 0.04). In addition, patients with paroxysmal AF with the minor allele of SNP rs2106261 (TT + TC) had lower levels of C reactive protein (CRP), neutrophil/lymphocyte (N/L) ratios, and interleukin 6 (IL-6) expression than patients with the CC type.

Similarly, studies on the rs2106261 polymorphism have had different findings. Choi et al. [27] found no correlation between SNP rs2106261 and postoperative recurrence in patients with atrial fibrillation in a study of 1068 patients with atrial fibrillation undergoing radiofrequency ablation in a Korean population. A meta-analysis by Jiang et al. [43] reached the same conclusion.

Thus, the impact of the SNP rs2106261 on postablation AF is also controversial. rs2106261 is located within the intron of the ZFHX3 gene, which encodes ZFHX3 ((or AT motif binding factor 1, (ATBF1)), a transcription factor containing multiple homologous structures and zinc finger motifs [44]. Although the ZFHX3gene is the second most crucial gene associated with AF, there is also uncertainty about its predictive value after ablation, and its underlying mechanisms have not been fully elucidated. (ZFHX3 was involved in inflammation, Ca2+ regulation, and electrical remodeling in many in vitro and ex vivo studies. In vitro, by rapidly pacing HL-1 cells, the expression of ZFHX3 can be reduced, thereby reducing its binding to PIAS3 (STAT3 inhibitor) and increasing the transcriptional activity of signal transducer and activator of transcription 3 (STAT3) [45]. While STATs are the main downstream substances of inflammatory signaling, they may mediate the inflammatory response and play an essential role in the process of atrial fibrillation [46]. Downregulation of (ZFHX3 has been reported to increase sarcoplasmic reticulum (SR) Ca2+ content, Ca2+ leakage, and Ca2+ transients; shorten action potential duration (APD); increase Kir3.4 and IKAch expression; and increase STAT3 and phosphorylated STAT3 in HL-1 atrial myocytes [47]. All of these factors contribute to forming and maintaining AF [48, 49, 50]. Huang et al. [51] also found that ZFHX3 and PITX2c can regulate each other, with ZFHX3 positively regulating PITX2c and PITX2c positively regulating ZFHX3. In addition, both ZFHX3 and PITX2c regulate the expression of the NPPA, TBX5, and NKX2.5 genes. The NAAP gene encodes cardiac natriuretic peptide (ANP), which governs cardiac ion channels and the autonomic nervous system and plays a vital role in cardiac electrophysiological activity [52, 53]. Both TBX5 (encoding Tbox5) and NKX2.5 (encoding NK2 transcription-related factors) also play a key role in cardiac electrophysiology and the development of AF [51, 54]. Recent studies have also found that the ZFHX3 transcription factor regulates INa channels [55]. This suggests that ZFHX3 may be involved in the recurrence mechanism of AF through its involvement in the inflammatory response as well as its regulation of ion channels. In addition, Husser et al. [56] and others found that ZFHX3 was associated with left atrial internal diameter, leading to the inference that ZFHX3 may be involved in AF recurrence through structural remodeling; however, there are few studies in this area. Hwang et al. [57] even performed a retrospective study of 14 single nucleotides of ZFHX3. They found the ZFHX3 gene polymorphisms (rs13336412, rs2106259, rs61208973, rs1858801, and rs12927436) were associated with extrapulmonary venous triggers but not with recurrence of AF. It is evident that although ZFHX3 may be involved in the mechanism of atrial fibrillation recurrence, how these mononucleotides affect it remains to be further investigated.

2.3 Angiotensin-Converting Enzyme (ACE) SNP ACE I/D

The renin-angiotensin-aldosterone system (RAAS) is an important system that regulates cardiovascular function. Angiotensin-converting enzyme (ACE) is the key enzyme of the RAAS. Its primary function is to convert inactive angiotensin I (Ang I) into active angiotensin II (Ang II), which constricts blood vessels, raises blood pressure, and promotes the secretion of aldosterone from the adrenal cortex, of which aldosterone plays a role in sodium retention, water retention, and potassium excretion. The ACE gene is located on the long arm of chromosome 17 (17q23) and encodes ACE, which exerts biochemical effects. Ueberham et al. [18] found that ACE DD genotype was a risk factor for the recurrence of atrial fibrillation using multivariate logistic regression analysis in a study of 238 patients with atrial fibrillation. The risk of postoperative recurrence was 2.251 times higher in patients with the DD genotype than in those with the ID + II genotype. It was suggested that atrial fibrosis due to variants in the ACE gene might be associated with recurrence after AF ablation. Zhang et al. [19] studied 193 patients with isolated AF in a Chinese Han population. They found a 2.35-fold increased risk of recurrence in patients with ACE DD genotype AF compared with patients with ID + II genotype using a multivariate logistic regression analysis. This study also found a larger left atrial internal diameter in patients with the DD genotype, suggesting that the effect of the ACE gene I/D polymorphism on isolated AF recurrence may be through left atrium diameter.

In past studies, a large body of evidence has shown that the RAAS is associated with cardiovascular disease. It has pro-inflammatory, antioxidant, and fibrotic effects. The mechanism of the ACE gene for recurrence after atrial fibrillation ablation is unclear. The ACE gene has a 287-bp repetitive sequence fragment in intron 16, and this fragment is often present as an insertion (I) or deletion (D) of this polymorphism [58]. Studies show elevated serum ACE levels and cardiac ACE activity in people with the ACE DD genotype [59]. Therefore, angiotensin II levels will also increase accordingly. Evidence suggests that angiotensin II induces fibroblast proliferation and collagen synthesis by increasing transforming growth factor β1 (TGF-β1) expression, leading to cardiac fibrosis [60]. Liu et al. [61] also showed that platelet-derived TGF-β1 promotes AngII-induced atrial fibrosis and the development of atrial fibrillation. They also found that structural remodeling and electrical conduction induced by AngII could be attenuated by platelet inhibitors [61]. Of course, increased TGF-β1 expression has been observed in animal models of atrial fibrillation [62]. Furthermore, a study by Watanabe et al. [63] found that the ACE D allele was associated with cardiac conduction abnormalities, as evidenced by prolonged PR intervals and P-wave duration. These two factors are also recognized as risk factors for increased recurrence of AF [64, 65]. Therefore, several lines of evidence suggest that patients with the ACE DD genotype may have more myocardial fibrosis and cardiac conduction abnormalities than patients with the II/ID genotype and are more likely to have AF recurrence.

2.4 Caveolin-1 (CAV1) SNP rs3807989

rs3807989 is located in an intron on the Caveolin-1 (CAV1) gene, which is associated with PR interval and atrial fibrillation in European populations [66, 67]. Chen et al. [68] first found similar results in a Chinese population, and they found that SNP rs3807989 in CAV1 was related to lone AF. A prospective study of 1722 patients who underwent catheter ablation found that the rs3807989 polymorphism was associated with AF recurrence. In addition, they corrected for confounding factors such as age, sex, LA diameter, LA volume, and QTc interval in 1290 AF patients entered into a Mendelian randomization analysis model. The PR interval was found to be highly associated with recurrent AF in terms of CAV1 (rs3807989, HR 1.04, 95% CI 1.01–1.07, p = 0.006) [20]. Ulus et al. [69] also found that the SNP rs3807989 predicted AF recurrence using multiple COX regression model analysis in Turkish patients with atrial fibrillation undergoing frozen balloon pulmonary vein isolation (rs3807989 G: OR 4.5, 95% CI 1.04–19.31, p = 0.043).

The caveolae are characterized by a 50–100 nm invagination of the cell surface plasma membrane. The fossa proteins are the major components of the fossa and are its main structural proteins. There are three isoforms of caveolin: caveolin-1, caveolin-2, and caveolin-3. Caveolin-1 is usually expressed in fibroblasts, endothelial cells, and smooth muscle cells [70]. The CAV1 gene encodes caveolin-1. The mechanism of the CAV1 gene in atrial fibrillation recurrence is also unclear. First, caveolin-1 affects ion channels and may have an important role in the development of AF. Caveolin-1 interacts with both the potassium channel Kir2.1 [71] and the pacemaker (HCN) channels HCN4 [72]. Second, caveolin-1 has also been found to have an important role in antifibrosis. Zhang et al. [73] demonstrated that caveolin-1 inhibits the TGF-β1 signaling pathway by regulating mitsugumin53 (MG53) and exerts an anti-fibrotic effect. Similarly, caveolin-1 depletes activation of the STAT3 signaling pathway and TGF-β1 signaling pathway, which leads to extracellular matrix deposition [74]. These results suggest caveolin-1 is involved in atrial fibrillation recurrence through its effects on ion channels and myocardial fibrosis. Therefore, we speculate that rs3807989 may be interested in atrial fibrillation recurrence through down-regulation of caveolin-1 expression, but the exact mechanism remains to be further investigated.

2.5 SOX5 SNP rs11047543

rs11047543 is located near the SOX5 gene, which encodes a transcription factor that plays an important role in cellular regulation through its transcriptional activity [75]. Park et al. [21] retrospectively analyzed 89 patients (age <40 years) with early-onset AF with at least 12 months of follow-up and found by multivariate analysis that the SNP rs11047543 for SOX5 (12p12) was an independent predictor of recurrence after AF ablation. Moreover, the recurrence rate was significantly higher in patients with the rs11047543 GA genotype than in patients with the GG genotype (72.2% vs 37.7%) in this study [21].

There are few reports on the association of rs11047543 with AF. Olesen et al. [76] found that SNP rs11047543 was associated with early-onset AF. Seifert et al. [77] found that SNP rs11047543 was associated with P-wave morphology. A meta-analysis based on 1010 patients showed that prolonged P-wave duration was significantly associated with postoperative recurrence of AF and that 149.5 ms was its potential threshold value [65]. More recently, a meta-analysis based on significant European and Asian populations similarly proposed a correlation between P-wave duration and AF and found associated genes suggesting a role of genetic factors in P-wave duration [78]. Although P-wave morphology and P-wave duration are not identical concepts, both are responsive to impulse conduction in the atria, and abnormalities in atrial conduction may be a factor in the recurrence of AF.

In addition, six single-nucleotide polymorphisms, including SOX5 SNP rs11047543, were associated with the PR interval in a genome-wide association meta-analysis [79]. Park et al. [64] found that a prolonged PR interval predicted atrial fibrillation recurrence. Additionally, they found that as the PR interval increased, the P-wave duration lengthened, the left atrial volume increased, and the intra-left atrial voltage decreased, suggesting that the PR interval may be associated with late atrial remodeling [64]. Another study based on GWAS data found that SOX5 was associated with PR interval, and SOX5 was significantly associated with the left atrial internal diameter and left atrial low voltage region [80]. PR interval represents intra-atrial conduction and atrial to interventricular conduction time, and its prolongation can increase the risk of arrhythmia and is influenced by genetic factors [81]. Thus, we speculate that rs11047543 may promote AF recurrence by regulating SOX5, affecting P-wave morphology, P-wave duration, and the PR interval.

2.6 EPHX2 SNP rs751141

Epoxyeicosatrienoic acid (EET) is a product of cytochrome P450 cyclooxygenase and exerts cardioprotective effects in various experimental models [82]. EET has been shown to modulate inflammatory pathways (reducing inflammatory response) [83], cardiac ion channels [84, 85] (inhibiting Na and L-Ca ion channels), and oxidative stress [86], which all have effects on AF susceptibility. EET has also been shown to protect mitochondria, resist apoptosis, fight cardiac fibrosis, and combat cardiac hypertrophy [87, 88]. EET is metabolized by soluble epoxide hydrolase (sEH) to a less biologically active diol (dihydroxyeicosatrienoic acid, DHET) [89]. Most experimental models demonstrate that sEH inhibition also benefits the heart [90, 91, 92]. Several exonic variants on EPHX2 (the gene encoding sEH) have been shown to increase or decrease sEH activity in vivo, including rs 751141 [93]. Wutzler et al. [22] recruited 218 patients with drug-refractory atrial fibrillation undergoing catheter ablation treatment. The EPHX2 gene SNP rs751141 was found to be an independent predictor of AF recurrence after catheter ablation by multivariate analysis. The presence of its variants increased the risk of recurrence by 3-fold at 12 months and 6-fold at 24 months after catheter ablation, respectively [22].

From the above description, we know that sEH inhibition and increased EET activity benefit the heart. In cellular experiments, sEH activity was lower in cells containing the rs751141 mutant because its mutation may have affected the stability of she [93]. It is assumed that patients containing this mutant have higher levels of EETs in their blood and tissues. Therefore, the protective effect of rs751141 is what we expected, but the results are surprising. On the other hand, some experimental and clinical findings have conflicting results with reduced sEH activity, which has a protective effect on the heart. In the CARDIA study, the rs751141 allele was associated with an increased risk of coronary calcification in African Americans [94]. Another study found no effect of the rs751141 allele on either ischemic stroke or ischemic heart disease in a Danish population [95]. We think that the positive results of sEH inhibition and increased EET levels in experimental models (inflammation, ischemia, and heart failure) may not fully apply to arrhythmias. There is certainly no doubt about the protective role of EETs in cardiac injury. EET accelerates wound neovascularization and healing [96], and this property may prevent scar formation between the pulmonary veins and the atria such that the recurrence of AF is higher after ablation. On the other hand, the modulation of cardiac ion channels by regional heterodimers of EETs also impacts AF recurrence, but more studies are needed to confirm this [84, 85, 97, 98, 99]. Therefore, we conjecture that the rs751141 variant may affect AF recurrence by regulating sEH activity in vivo, affecting EET levels, although more studies are needed to prove the exact process.

2.7 eNOS3 SNP rs1799983

The eNOS gene is located on chromosome 7 [100], contains 26 exons, and encodes a nitric oxide synthase (NOS) protein [101, 102]. There are three isoforms of nitric oxide synthase: NOS1, NOS2, and NOS3. NOS3 is also known as endothelial NOS (eNOS). In the heart, NOS1 and NOS3 are usually expressed [103]. On exon 7 of the eNOS gene, there is a clinically relevant variant called the G894T variant (eNOS3 SNP rs1799983) that corresponds to the glutamate-aspartate substitution (Glu298Asp) [104]. The rs1799983 variant (Glu298Asp) is associated with reduced basal nitric oxide (NO) production [105]. Shim et al. [23] recruited 500 patients with atrial fibrillation for radiofrequency ablation therapy. At a mean follow-up of 17 months, SNP rs1799983 of the eNOS3 gene was associated with early AF recurrence (within three months). After multiple logistic regression analyses, SNP rs1799983 was an independent predictor of early atrial fibrillation recurrence and was not associated with late recurrence [23].

Nitric oxide synthase (eNOS) and its product nitric oxide (NO) play an essential role in vasodilation and maintenance of cardiovascular homeostasis. NOS produces NO in the heart, affecting almost all mechanotransduction pathways in cardiac myocytes. It mediates mechanosensing, mechanical-electrical feedback (by modulating ion channel activity, including K+, Na+, and Ca2+ channels, etc.), and Ca2+ handling [106, 107]. eNOS and NO contribute to atrial myocardial superoxide production [108]. In addition, eNOS can interact with CAV1, inhibiting eNOS activity and reducing NO release, as evidenced by enhanced endothelium-dependent diastolic function in CAV1-deficient mice. In contrast, eNOS can also act directly on CAV1; this is only a result of in vitro experiments and is not based in vivo [109].

The exact mechanism by which the eNOS3 SNP rs1799983 is associated with early recurrence of AF is unclear, but the possibilities have been considered. First, acute inflammatory responses due to tissue damage formation should be widely present after catheter ablation of atrial fibrillation. In turn, these inflammatory responses cause the activation of signaling pathways that contribute to structural, electrical, and mechanical heterogeneity of atrial muscle, thereby promoting arrhythmogenesis [110]. The detrimental effects of inflammation and oxidative stress on atrial electrical and structural remodeling were mentioned in the summary of Karam et al. [111], which suggested that oxidative stress and inflammation could be targeted through some pathways to help control atrial fibrillation. The rs1799983 variant (Glu298Asp) is associated with reduced basal nitric oxide (NO) production [105]. In contrast, Shim et al. [23] found a higher rate of early recurrence in patients containing the rs1799983 variant; this may be due to reduce NO activity leading to slower inflammatory regression after atrial fibrillation ablation (RFCA), thus promoting recurrence of AF. Second, dysfunction of NOS and NO in atrial oxidative damage and electrical remodeling may also contribute to the reproduction of AF.

2.8 IL-6R SNP rs4845625

There is much evidence that inflammation plays a vital role in the pathophysiology of AF, including C-reactive protein, interleukins, and tumor necrosis factor-α [112]. In a meta-analysis, a variant of intron rs4845625 of the interleukin-6 receptor (IL-6 R) gene was found to be associated with AF in Caucasians [113]. At the same time, Smit et al. [114] found baseline levels of IL-6 to be an independent predictor of early recurrence of AF by multifactorial analysis (HR 1.3, 95% CI 1.0–1.7, p = 0.02). Wu et al. [24] studied 278 patients with AF in a retrospective study in a Chinese Han population with approximately one year of follow-up. They found that the SNP rs4845625 for IL-6R was associated with the recurrence of AF both early (4 weeks post-ablation) and late (3–12 months post-ablation). This relationship remained significant after correction for hypertension, age, sex, and diabetes mellitus. And the probability of recurrence was higher in patients with the T allele than in patients with the C allele in the dominant, recessive, and additive models.

The mechanism of how the rs4845625 polymorphism is involved in recurrence after atrial fibrillation ablation is still not well understood. Inflammation is one of the mechanisms by which AF occurs. Frustaci et al. [115] performed biopsies on the hearts of 12 patients with lone AF and found inflammatory lymphomonuclear infiltration and peripheral cell necrosis in their atrial myocytes, which was not observed in patients with sinus rhythm. This was supported in an animal model with increased susceptibility to AF in a sterile pericardial model in dogs, where the incidence of AF was reduced by cortisol steroid hormone treatment and where the inflammatory markers (IL-6, CRP, and TNF-α) were significantly lower in this study group than in the control group [116]. Specific localization of C-reactive protein was also found in the cytoplasm of atrial myocytes in patients with AF [117]. Several studies based on European and Asian populations have demonstrated that elevated indicators of inflammation can increase the risk of atrial fibrillation [118, 119, 120]. All of the above evidence supports the possible involvement of inflammation in the development of AF.

There is certainly some evidence that inflammation is an essential mechanism for recurrence after atrial fibrillation ablation. A meta-analysis based on large-scale data found that high-sensitivity C-reactive protein and interleukin-6 were strongly associated with atrial fibrillation recurrence [121]. Elevated serum MMP-2 levels were an independent predictor of atrial fibrillation recurrence [122]. A study of marathon runners found increased P-wave duration after exercise accompanied by increased levels of IL-6, CRP, and neutrophils, suggesting that acute changes in inflammatory cytokines may be associated with interatrial cell conduction [123]. Another study suggested that inflammation is associated with atrial structural remodeling. They found that IL-6 and CRP were associated with increased left atrial internal diameter [124]. From this, IL-6 might increase the recurrence of AF through electrical and structural remodeling. Nevertheless, unfortunately, Wu et al. [24] did not find a relationship between variant rs4845625 and serum IL-6 levels. The reason was that they did not use serum specimens from AF patients, only serum samples from the healthy population, and the sample size was minimal. Therefore, using sera from the AF population may yield different results if replicated in a larger cohort. Thus the mechanism of SNP rs4845625 involvement in the risk of recurrence after AF ablation also needs further exploration.

2.9 Heme Oxygenase-1 GT Repeat Polymorphism

Heme oxygenase-1 (HO-1) is a vital rate-limiting enzyme in the degradation of heme that degrades heme to iron ions, carbon monoxide (CO), and biliverdin; the latter is then converted to bilirubin by biliverdin reductase. HO-1 and its products can be activated by various oxidizing substances and act as an antioxidant system to protect the body [125]. Hu et al. [25] recruited 205 patients with drug-refractory AF undergoing radiofrequency ablation. They used multifactorial regression analysis and found that the HO-1 promoter polymorphism (GT repeat sequence) was associated with recurrence after paroxysmal AF ablation, with shorter GT repeat sequences in the recurrence group than in the nonrecurrence group [25].

HO-1 is a protective factor with anti-inflammatory, antioxidant, and anti-apoptotic effects [126, 127]. In transgenic mice, HO-1 prevents oxidative stress in myocardial tissue and improves smooth intimal proliferation and inflammatory responses in coronary arteries [128]. In an infarct model, human HO-1 (h HO-1) was injected intracardially into the rat myocardium by adenovirus, causing an increase in HO-1 expression. They found a significant decrease in infarct size and myocardial lipid peroxidation levels, pro-apoptotic Bax and pro-inflammatory interleukin-1β proteins, and an increase in anti-apoptotic Bcl-2 protein levels [129]. Another recent study found that HO-1 has a protective effect on atrial structural remodeling. Hsu et al. [130] found that patients with shorter HO-1 promoter GT repeat sequences had HO-1 overexpression, while patients with a reduced degree of atrial oxidative stress decreased collagen fiber production and reduced myogenic fiber degradation. Yeh et al. [131] found similar results in animal experiments.

Radiofrequency ablation causes a conduction block between the pulmonary veins and the atria through thermal injury. The GT repeat sequence was shorter, and the HO-1 expression level was higher in the recurrence group. HO-1 plays a vital role in antioxidative stress, anti-inflammation, anti-apoptosis, and improvement of atrial remodeling; thus, HO-1 may help restore electrical conduction between the pulmonary vein and atrial myocytes and reduce scar formation between them by reducing the damage to cardiomyocytes by RF ablation, thus leading to AF recurrence. In addition, the experiment by Hu et al. [25] did not find an effect of HO-1 promoter polymorphism on the outcome of persistent atrial fibrillation recurrence. Because they considered more factors affecting persistent AF recurrence, including more extensive atrial fibrosis, more atrial myocardial remodeling, and extrapulmonary vein trigger foci and had a limited number of cases, their ability to statistically analyze persistent AF cases was limited [25].

2.10 HRC SNP rs3745297 (T > G, Ser96Ala)

Sarcoplasmic reticulum (SR.) Ca2+ overload due to protein kinase A (PKA) hyperphosphorylation of the cardiac ryanodine receptor (RyR2) plays an essential role in the development and maintenance of atrial fibrillation [132]. HRC is a vital regulator of SR Ca2+ homeostasis in cardiac myocytes [133]. A study showed that the human HRC variant Ser96Ala was overexpressed in cardiomyocytes by gene transfer to adenovirus, resulting in increased SR Ca2+ overload and Ca2+ spark frequency [134]. Japanese scholar Amioka et al. [26] recruited 334 patients with paroxysmal AF who underwent radiofrequency ablation and found that HRC SNP rs3745297 (Ser96Ala) was an independent risk factor for AF recurrence by multifactorial regression analysis. The frequency of minor allele G was significantly higher in the recurrence group than in the nonrecurrence group (allele frequency model OR 1.8, p = 0.006; recessive model OR 3.55, p = 0.0009). Only 16 of the 57 recurrent patients in this study underwent secondary ablation. Their trigger sites were nine pulmonary veins (PV), one superior vena cava (SCV), one septum, and five unknown sites. Six of these patients had the HRC variant (Ser96Ala), but, unfortunately, no correlation was found between the HRC variant and the AF trigger site [26].

The HRC SNP rs3745297 (Ser96Ala) has been well studied in the field of heart failure and ventricular arrhythmias [134, 135]. Amioka was the first to identify the HRC SNP rs3745297 (Ser96Ala) associated with AF recurrence in this observational study. However, the mechanism of recurrence in patients with paroxysmal AF with the HRC Ser96Ala variant has not been clarified. The conjectured mechanisms are considered as follows. First, nonpulmonary vein trigger foci are a risk factor for AF recurrence. However, the frequency of HRC variants was similar in patients with AF of the pulmonary vein and nonpulmonary vein origin in the study by Amioka et al. [26], and the origin of AF recurrence was not found to be associated with HRC Ser96Ala. Therefore, the effect of HRC variants on nonpulmonary vein trigger foci, and thus the mechanism of AF recurrence may not be explained. Second, changes in Ca2+ and Ca2+ processing play an important role in the mechanism of triggering and maintaining AF [136]. Ion channels, including L-type Ca2+, and Ito, are involved in the alteration of the atrial nonresponse period that maintains AF and thus have an important role in the electrical remodeling of AF [137]. In addition, tachycardia-induced Ca2+ handling plays an essential role in the electrical remodeling of the atria [138]. Because the HRC variant Ser96Ala causes dysregulation of SR Ca2+ cycling [139], we speculate that patients with paroxysmal AF who possess the HRC Ser96Ala variant may be continuously exposed to Ca2+ overload, leading to inactivation of L-type Ca2+ channels and shortened action potentials, causing electrical remodeling of the atria and ultimately leading to AF recurrence.

3. Gene Polymorphisms and New-Onset Atrial Fibrillation

New-onset atrial fibrillation (new AF) is defined as atrial fibrillation that occurs during critical illness without a known previous history of atrial fibrillation. Myocardial ischemia, myocardial strain, electrolyte disturbances, sympathetic activation, and oxidative stress may be the causes of new AF [140, 141]. Genetic genes have been studied less frequently and with inconsistent results in this population of new AF. Kerchberger et al. [140] identified several single nucleotide polymorphisms associated with new AF in 1936 critically ill ICU patients, showing after controlling for clinical factors that rs3853445 (near PITX2, OR 0.47, 95% CI 0.30–0.73, p = 0.001) and rs12415501 (in NEURL, OR 1.72, 95% CI 1.27–2.59, p = 0.01) were associated with new-onset atrial fibrillation. Adding genetic factors to clinical factors in multivariate regression models helps identify new-onset atrial fibrillation [140]. Siebert et al. [142] conducted a prospective study of 203 patients with coronary artery disease who underwent coronary artery bypass grafting and did not find that the Scal polymorphism in the ANP gene predicted new-onset atrial fibrillation after coronary artery bypass. Kertai et al. [143] similarly selected coronary artery bypass graft patients and found that only SNP rs10504554, located in the LY96 intron region, was associated with a reduced risk of new-onset postoperative atrial fibrillation based on a genome-wide association study (OR 0.48, 95% CI 0.34–0.68, p = 2.9 × 10-5). Plante et al. [144] identified two single nucleotide polymorphisms (R87Q and P307S) in the voltage-gated channel hKv1.5 in a French-Canadian coronary artery bypass grafting population that altered the expression of gating processes and hKv1.5 channels; and patients with new-onset atrial fibrillation were more likely to have this polymorphism compared with controls (6.25% vs 3.37%; p = 0.42). A retrospective study by Kiliszek et al. [145] analyzing patients with an acute heart attack from two major centers in Poland found that SNP rs10757278 was associated with the development of new AF in such patients, with a protective effect of the minor allele (G) (OR 0.41, 95% CI 0.17–0.97, p = 0.025), which persisted after controlling for the Grace scale and age. This shows that the results of studies correlating new AF with genetic polymorphisms are inconsistent, probably because of the different populations studied. There are certainly not many large prospective and retrospective studies on new AF, and we think this may be an area we need to focus on in the future, and we look forward to more in-depth studies on this population.

4. Limitations and Outlooks

In the past decade, considerable progress has been made in identifying rare and common genetic variants associated with atrial fibrillation. However, there is still no conclusive evidence that the available data from genetic studies can be used for clinical decision-making in patients with atrial fibrillation. The reasons for this may be as follows: the first is the lack of a more comprehensive understanding of the potential genetic substrates of AF; the second is the fact that many genetic studies are based on retrospective studies and that extensive prospective randomized controlled studies are still lacking; and the third is the fact that there are differences by race and ethnicity and that many of the current study data are based on European, American, and Asian populations and populations in more economically developed regions. Although many retrospective studies have shown that outcomes after atrial fibrillation ablation are influenced by genetic variation, we consider the following points for improvement in how to apply the genetic findings to clinical practice better.

First, we found through extensively reading the literature that although different genetic variants or different variants of the same gene have some predictive value for outcome after AF ablation, it may be more reasonable to use the genetic risk score (GRS) as a risk assessment for patients with AF, more beneficial to identify patients at higher risk of developing AF, and thus more helpful to identify those prone to recurrence after AF ablation [146].

Second, many AF centers have data based on the more economically developed Caucasian and Asian populations, which is not representative of the general global population. In addition, we serve people from all levels of society and certainly from socioeconomically deprived areas. Hence, databases with broader applicable populations must be built, and more prospective randomized controlled studies must be conducted.

Third, the translation of genetic data to the clinic is also limited by technology. GWAS and GRS are only available in advanced vascular disease research centers. The ability to test quickly and obtain results is still debatable. Therefore, whether genetic testing can be made cheaper and more convenient through competitive mechanisms or technological improvements is a question that deserves to be addressed to enable its more comprehensive application.

5. Conclusions

Overall, although current research and cognitive gaps still need to be addressed, the application of genetics to the prediction of outcomes after AF ablation holds great promise. Both individual patients, their families, and even society as a whole will benefit from this.

Search Strategy and Selection Criteria

This review used the Pubmed database to identify references for this review. The search terms covered all parts of this review, but were not limited to “atrial fibrillation recurrence” and “genetic polymorphisms”. Three authors (MFW, CX, and SYS) searched for the full text of these studies and performed an evaluation. Disagreements were resolved by discussion.

Author Contributions

MFW, CX, SYS, LY, ZYZ and JJL jointly participated in drafting, conceptualizing, and designing the manuscript; MFW, CX, and SYS participated in data collection for this work and performed preliminary analysis; LY further analyzed and interpreted the data; when disagreements arose, all authors jointly analyzed the data and discussed to resolve them. ZYZ and JJL reviewed and provided feedback on important knowledge points; preliminary revisions were made to important knowledge points. The remaining four authors (MFW, CX, SYS, and LY) further critically improved the revisions. All authors have read and agreed to the final version and are responsible for the published content. All authors have fully participated in the work and agreed to take responsibility for all aspects of the work.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

Not applicable.

Funding

This research received no external funding.

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Ohsawa M, Okayama A, Sakata K, Kato K, Itai K, Onoda T, et al. Rapid increase in estimated number of persons with atrial fibrillation in Japan: an analysis from national surveys on cardiovascular diseases in 1980, 1990 and 2000. Journal of Epidemiology. 2005; 15: 194–196.
[2]
Heeringa J, van der Kuip DAM, Hofman A, Kors JA, van Herpen G, Stricker BHC, et al. Prevalence, incidence and lifetime risk of atrial fibrillation: the Rotterdam study. European Heart Journal. 2006; 27: 949–953.
[3]
Wilke T, Groth A, Mueller S, Pfannkuche M, Verheyen F, Linder R, et al. Incidence and prevalence of atrial fibrillation: an analysis based on 8.3 million patients. Europace. 2012; 15: 486–493.
[4]
Schnabel RB, Yin X, Gona P, Larson MG, Beiser AS, McManus DD, et al. 50 year trends in atrial fibrillation prevalence, incidence, risk factors, and mortality in the Framingham Heart Study: a cohort study. The Lancet. 2015; 386: 154–162.
[5]
Benjamin EJ, Muntner P, Alonso A, Bittencourt MS, Callaway CW, Carson AP, et al. Heart Disease and Stroke Statistics-2019 Update: A Report From the American Heart Association. Circulation. 2019; 139: e56–e528.
[6]
Benjamin EJ, Go AS, Desvigne-Nickens P, Anderson CD, Casadei B, Chen LY, et al. Research Priorities in Atrial Fibrillation Screening: A Report From a National Heart, Lung, and Blood Institute Virtual Workshop. Circulation. 2021; 143: 372–388.
[7]
January CT, Wann LS, Alpert JS, Calkins H, Cigarroa JE, Cleveland JC, et al. 2014 AHA/ACC/HRS guideline for the management of patients with atrial fibrillation: executive summary: a report of the American College of Cardiology/American Heart Association Task Force on practice guidelines and the Heart Rhythm Society. Circulation. 2014; 130: 2071–2104.
[8]
Sepehri Shamloo A, Dagres N, Hindricks G. 2020 ESC guidelines on atrial fibrillation: Summary of the most relevant recommendations and innovations. Herz. 2021; 46: 28–37. (In German)
[9]
Calkins H, Hindricks G, Cappato R, Kim Y, Saad EB, Aguinaga L, et al. 2017 HRS/EHRA/ECAS/APHRS/SOLAECE expert consensus statement on catheter and surgical ablation of atrial fibrillation. Europace. 2018; 20: e1–e160.
[10]
Habibi M, Calkins H. Atrial fibrillation catheter ablation: an updated review of current guidelines and expert consensus documents. Herzschrittmachertherapie & Elektrophysiologie. 2019; 30: 371–376.
[11]
Darbar D, Herron KJ, Ballew JD, Jahangir A, Gersh BJ, Shen W, et al. Familial atrial fibrillation is a genetically heterogeneous disorder. Journal of the American College of Cardiology. 2003; 41: 2185–2192.
[12]
Arnar DO, Thorvaldsson S, Manolio TA, Thorgeirsson G, Kristjansson K, Hakonarson H, et al. Familial aggregation of atrial fibrillation in Iceland. European Heart Journal. 2006; 27: 708–712.
[13]
Husser D, Adams V, Piorkowski C, Hindricks G, Bollmann A. Chromosome 4q25 variants and atrial fibrillation recurrence after catheter ablation. Journal of the American College of Cardiology. 2010; 55: 747–753.
[14]
Chen F, Yang Y, Zhang R, Zhang S, Dong Y, Yin X, et al. Polymorphism rs2200733 at chromosome 4q25 is associated with atrial fibrillation recurrence after radiofrequency catheter ablation in the Chinese Han population. American Journal of Translational Research. 2016; 8: 688–697.
[15]
Zhao L, Zhang G, Wen Z, Huang C, Wu H, Xu J, et al. Common variants predict recurrence after nonfamilial atrial fibrillation ablation in Chinese Han population. International Journal of Cardiology. 2017; 227: 360–366.
[16]
Park J, Lee J, Yang P, Kim T, Shin E, Park J, et al. Good responders to catheter ablation for long-standing persistent atrial fibrillation: Clinical and genetic characteristics. Journal of Cardiology. 2017; 69: 584–590.
[17]
Tomomori S, Nakano Y, Ochi H, Onohara Y, Sairaku A, Tokuyama T, et al. Maintenance of low inflammation level by the ZFHX3 SNP rs2106261 minor allele contributes to reduced atrial fibrillation recurrence after pulmonary vein isolation. PLoS ONE. 2018; 13: e0203281.
[18]
Ueberham L, Bollmann A, Shoemaker MB, Arya A, Adams V, Hindricks G, et al. Genetic ACE I/D polymorphism and recurrence of atrial fibrillation after catheter ablation. Circulation: Arrhythmia and Electrophysiology. 2013; 6: 732–737.
[19]
Zhang X, Wu L, Liu X, Yang Y, Tan H, Wang X, et al. Association of angiotensin-converting enzyme gene I/D and CYP11B2 gene -344T/C polymorphisms with lone atrial fibrillation and its recurrence after catheter ablation. Experimental and Therapeutic Medicine. 2012; 4: 741–747.
[20]
Hong M, Hwang I, Yu H, Kim T, Uhm J, Joung B, et al. Potential causal association of a prolonged PR interval and clinical recurrence of atrial fibrillation after catheter ablation: a Mendelian randomization analysis. Journal of Human Genetics. 2020; 65: 813–821.
[21]
Park YM, Roh SY, Lee DI, Shim J, Choi JI, Park SW, et al. The Effects of Single Nucleotide Polymorphisms in Korean Patients with Early-onset Atrial Fibrillation after Catheter Ablation. Journal of Korean Medical Science. 2020; 35: e411.
[22]
Wutzler A, Kestler C, Perrot A, Loehr L, Huemer M, Parwani AS, et al. Variations in the human soluble epoxide hydrolase gene and recurrence of atrial fibrillation after catheter ablation. International Journal of Cardiology. 2013; 168: 3647–3651.
[23]
Shim J, Park JH, Lee J, Uhm JS, Joung B, Lee M, et al. eNOS3 Genetic Polymorphism Is Related to Post-Ablation Early Recurrence of Atrial Fibrillation. Yonsei Medical Journal. 2015; 56: 1244–1250.
[24]
Wu G, Cheng M, Huang H, Yang B, Jiang H, Huang C. A variant of IL6R is associated with the recurrence of atrial fibrillation after catheter ablation in a Chinese Han population. PLoS ONE. 2014; 9: e99623.
[25]
Hu Y, Lee K, Wang H, Ueng K, Yeh H, Chao T, et al. The association between heme oxygenase-1 gene promoter polymorphism and the outcomes of catheter ablation of atrial fibrillation. PLoS ONE. 2013; 8: e56440.
[26]
Amioka M, Nakano Y, Ochi H, Onohara Y, Sairaku A, Tokuyama T, et al. Ser96Ala genetic variant of the human histidine-rich calcium-binding protein is a genetic predictor of recurrence after catheter ablation in patients with paroxysmal atrial fibrillation. PLoS ONE. 2019; 14: e0213208.
[27]
Choi E, Park JH, Lee J, Nam CM, Hwang MK, Uhm J, et al. Korean Atrial Fibrillation (AF) Network: Genetic Variants for AF Do Not Predict Ablation Success. Journal of the American Heart Association. 2015; 4: e002046.
[28]
Hu Y, Wang H, Yeh H, Lee K, Lin Y, Chang S, et al. Association of Single Nucleotide Polymorphisms with Atrial Fibrillation and the Outcome after Catheter Ablation. Acta Cardiologica Sinica. 2016; 32: 523–531.
[29]
Benjamin Shoemaker M, Muhammad R, Parvez B, White BW, Streur M, Song Y, et al. Common atrial fibrillation risk alleles at 4q25 predict recurrence after catheter-based atrial fibrillation ablation. Heart Rhythm. 2013; 10: 394–400.
[30]
Shoemaker MB, Bollmann A, Lubitz SA, Ueberham L, Saini H, Montgomery J, et al. Common genetic variants and response to atrial fibrillation ablation. Circulation: Arrhythmia and Electrophysiology. 2015; 8: 296–302.
[31]
Rattanawong P, Chenbhanich J, Vutthikraivit W, Chongsathidkiet P. A Chromosome 4q25 Variant is Associated with Atrial Fibrillation Recurrence After Catheter Ablation: A Systematic Review and Meta-Analysis. Journal of Atrial Fibrillation. 2018; 10: 1666.
[32]
Hu Z, Zou D. Genotype-phenotype associations in atrial fibrillation: meta-analysis. Journal of Interventional Cardiac Electrophysiology. 2018; 54: 283–288.
[33]
He J, Zhu W, Yu Y, Hu J, Hong K. Variant rs2200733 and rs10033464 on chromosome 4q25 are associated with increased risk of atrial fibrillation after catheter ablation: Evidence from a meta-analysis. Cardiology Journal. 2018; 25: 628–638.
[34]
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: 353–357.
[35]
Kirchhof P, Kahr PC, Kaese S, Piccini I, Vokshi I, Scheld H, et al. PITX2c is expressed in the adult left atrium, and reducing Pitx2c expression promotes atrial fibrillation inducibility and complex changes in gene expression. Circulation-Cardiovascular Genetics. 2011; 4: 123–133.
[36]
Liu C, Liu W, Lu MF, Brown NA, Martin JF. Regulation of left-right asymmetry by thresholds of Pitx2c activity. Development. 2001; 128: 2039–2048.
[37]
Hill MC, Kadow ZA, Li L, Tran TT, Wythe JD, Martin JF. A cellular atlas of Pitx2-dependent cardiac development. Development. 2019; 146: dev180398.
[38]
Mommersteeg MTM, Brown NA, Prall OWJ, de Gier-de Vries C, Harvey RP, Moorman AFM, et al. Pitx2c and Nkx2-5 are required for the formation and identity of the pulmonary myocardium. Circulation Research. 2007; 101: 902–909.
[39]
Syeda F, Kirchhof P, Fabritz L. PITX2-dependent gene regulation in atrial fibrillation and rhythm control. The Journal of Physiology. 2017; 595: 4019–4026.
[40]
Collins MM, Ahlberg G, Hansen CV, Guenther S, Marín-Juez R, Sokol AM, et al. Early sarcomere and metabolic defects in a zebrafish pitx2c cardiac arrhythmia model. Proceedings of the National Academy of Sciences of the United States of America. 2019; 116: 24115–24121.
[41]
Benjamin EJ, Rice KM, Arking DE, Pfeufer A, van Noord C, Smith AV, et al. Variants in ZFHX3 are associated with atrial fibrillation in individuals of European ancestry. Nature Genetics. 2009; 41: 879–881.
[42]
Li C, Wang F, Yang Y, Fu F, Xu C, Shi L, et al. Significant association of SNP rs2106261 in the ZFHX3 gene with atrial fibrillation in a Chinese Han GeneID population. Human Genetics. 2010; 129: 239–246.
[43]
Jiang T, Wang Y, Qu Q, Qi T, Chen Y, Qu J. Association between gene variants and the recurrence of atrial fibrillation: An updated meta-analysis. Medicine. 2019; 98: e15953.
[44]
Yasuda H, Mizuno A, Tamaoki T, Morinaga T. ATBF1, a multiple-homeodomain zinc finger protein, selectively down-regulates AT-rich elements of the human alpha-fetoprotein gene. Molecular and Cellular Biology. 1994; 14: 1395–1401.
[45]
Jiang Q, Ni B, Shi J, Han Z, Qi R, Xu W, et al. Down-regulation of ATBF1 activates STAT3 signaling via PIAS3 in pacing-induced HL-1 atrial myocytes. Biochemical and Biophysical Research Communications. 2014; 449: 278–283.
[46]
Yeh M, Gharavi NM, Choi J, Hsieh X, Reed E, Mouillesseaux KP, et al. Oxidized phospholipids increase interleukin 8 (IL-8) synthesis by activation of the c-src/signal transducers and activators of transcription (STAT)3 pathway. The Journal of Biological Chemistry. 2004; 279: 30175–30181.
[47]
Kao Y, Hsu J, Chen Y, Lin Y, Lkhagva B, Chen S, et al. ZFHX3 knockdown increases arrhythmogenesis and dysregulates calcium homeostasis in HL-1 atrial myocytes. International Journal of Cardiology. 2016; 210: 85–92.
[48]
Voigt N, Heijman J, Wang Q, Chiang DY, Li N, Karck M, et al. Cellular and molecular mechanisms of atrial arrhythmogenesis in patients with paroxysmal atrial fibrillation. Circulation. 2014; 129: 145–156.
[49]
Burashnikov A, Antzelevitch C. Reinduction of atrial fibrillation immediately after termination of the arrhythmia is mediated by late phase 3 early afterdepolarization-induced triggered activity. Circulation. 2003; 107: 2355–2360.
[50]
Kovoor P, Wickman K, Maguire CT, Pu W, Gehrmann J, Berul CI, et al. Evaluation of the role of I(KACh) in atrial fibrillation using a mouse knockout model. Journal of the American College of Cardiology. 2001; 37: 2136–2143.
[51]
Huang Y, Wang C, Yao Y, Zuo X, Chen S, Xu C, et al. Molecular Basis of Gene-Gene Interaction: Cyclic Cross-Regulation of Gene Expression and Post-GWAS Gene-Gene Interaction Involved in Atrial Fibrillation. PLoS Genetics. 2015; 11: e1005393.
[52]
Perrin MJ, Gollob MH. The role of atrial natriuretic peptide in modulating cardiac electrophysiology. Heart Rhythm. 2012; 9: 610–615.
[53]
Goetze JP, Bruneau BG, Ramos HR, Ogawa T, de Bold MK, de Bold AJ. Cardiac natriuretic peptides. Nature Reviews Cardiology. 2020; 17: 698–717.
[54]
Nadadur RD, Broman MT, Boukens B, Mazurek SR, Yang X, van den Boogaard M, et al. Pitx2 modulates a Tbx5-dependent gene regulatory network to maintain atrial rhythm. Science Translational Medicine. 2016; 8: 354ra115.
[55]
Rubio-Alarcón M, Cámara-Checa A, Dago M, Crespo-García T, Nieto-Marín P, Marín M, et al. Zfhx3 Transcription Factor Represses the Expression of SCN5A Gene and Decreases Sodium Current Density (INa). International Journal of Molecular Sciences. 2021; 22: 13031.
[56]
Husser D, Büttner P, Ueberham L, Dinov B, Sommer P, Arya A, et al. Association of atrial fibrillation susceptibility genes, atrial fibrillation phenotypes and response to catheter ablation: a gene-based analysis of GWAS data. Journal of Translational Medicine. 2017; 15: 71.
[57]
Hwang I, Kwon O, Hong M, Yang S, Park J, Yu HT, et al. Association of ZFHX3 Genetic Polymorphisms and Extra-Pulmonary Vein Triggers in Patients With Atrial Fibrillation Who Underwent Catheter Ablation. Frontiers in Physiology. 2022; 12: 807545.
[58]
van den Berg MP, Van Gelder IC. New insight into the association among atrial fibrillation, electrophysiological remodeling, and the ACE insertion/deletion polymorphism—toward a more patient-tailored therapy? Heart Rhythm. 2009; 6: 1333–1334.
[59]
Danser AH, Schalekamp MA, Bax WA, van den Brink AM, Saxena PR, Riegger GA, et al. Angiotensin-converting enzyme in the human heart. Effect of the deletion/insertion polymorphism. Circulation. 1995; 92: 1387–1388.
[60]
Kupfahl C, Pink D, Friedrich K, Zurbrügg HR, Neuss M, Warnecke C, et al. Angiotensin II directly increases transforming growth factor beta1 and osteopontin and indirectly affects collagen mRNA expression in the human heart. Cardiovascular Research. 2000; 46: 463–475.
[61]
Liu Y, Lv H, Tan R, An X, Niu X, Liu Y, et al. Platelets Promote Ang II (Angiotensin II)-Induced Atrial Fibrillation by Releasing TGF-β1 (Transforming Growth Factor-β1) and Interacting With Fibroblasts. Hypertension. 2020; 76: 1856–1867.
[62]
Chan Y, Chang G, Lai Y, Chen W, Chang S, Hung L, et al. Atrial fibrillation and its arrhythmogenesis associated with insulin resistance. Cardiovascular Diabetology. 2019; 18: 125.
[63]
Watanabe H, Kaiser DW, Makino S, MacRae CA, Ellinor PT, Wasserman BS, et al. ACE I/D polymorphism associated with abnormal atrial and atrioventricular conduction in lone atrial fibrillation and structural heart disease: implications for electrical remodeling. Heart Rhythm. 2009; 6: 1327–1332.
[64]
Park J, Kim T, Lee JS, Park JK, Uhm JS, Joung B, et al. Prolonged PR interval predicts clinical recurrence of atrial fibrillation after catheter ablation. Journal of the American Heart Association. 2014; 3: e001277.
[65]
Wang Y, Chen G, Li X, Zhou X, Li Y. Prolonged P-wave duration is associated with atrial fibrillation recurrence after radiofrequency catheter ablation: A systematic review and meta-analysis. International Journal of Cardiology. 2017; 227: 355–359.
[66]
Pfeufer A, van Noord C, Marciante KD, Arking DE, Larson MG, Smith AV, et al. Genome-wide association study of PR interval. Nature Genetics. 2010; 42: 153–159.
[67]
Holm H, Gudbjartsson DF, Arnar DO, Thorleifsson G, Thorgeirsson G, Stefansdottir H, et al. Several common variants modulate heart rate, PR interval and QRS duration. Nature Genetics. 2010; 42: 117–122.
[68]
Chen S, Wang C, Wang X, Xu C, Wu M, Wang P, et al. Significant Association Between CAV1 Variant rs3807989 on 7p31 and Atrial Fibrillation in a Chinese Han Population. Journal of the American Heart Association. 2015; 4: e001980.
[69]
Ulus T, Dural M, Meşe P, Yetmiş F, Mert KU, Görenek B, et al. Genetic variants associated with atrial fibrillationand long-term recurrence after catheter ablation for atrialfibrillation in Turkish patients. Anatolian Journal of Cardiology. 2021; 25: 129–138.
[70]
Tian J, Popal MS, Huang R, Zhang M, Zhao X, Zhang M, et al. Caveolin as a Novel Potential Therapeutic Target in Cardiac and Vascular Diseases: A Mini Review. Aging and Disease. 2020; 11: 378–389.
[71]
Ambrosini E, Sicca F, Brignone MS, D’Adamo MC, Napolitano C, Servettini I, et al. Genetically induced dysfunctions of Kir2.1 channels: implications for short QT3 syndrome and autism-epilepsy phenotype. Human Molecular Genetics. 2014; 23: 4875–4886.
[72]
Barbuti A, Scavone A, Mazzocchi N, Terragni B, Baruscotti M, Difrancesco D. A caveolin-binding domain in the HCN4 channels mediates functional interaction with caveolin proteins. Journal of Molecular and Cellular Cardiology. 2012; 53: 187–195.
[73]
Zhang M, Wang H, Wang X, Bie M, Lu K, Xiao H. MG53/CAV1 regulates transforming growth factor-β1 signaling-induced atrial fibrosis in atrial fibrillation. Cell Cycle. 2020; 19: 2734–2744.
[74]
Zhang M, Wang H, Bie M, Wang X, Lu K, Xiao H. Caveolin-1 Deficiency Induces Atrial Fibrosis and Increases Susceptibility to Atrial Fibrillation by the STAT3 Signaling Pathway. Journal of Cardiovascular Pharmacology. 2021; 78: 175–183.
[75]
Lefebvre V. The SoxD transcription factors–Sox5, Sox6, and Sox13–are key cell fate modulators. The International Journal of Biochemistry & Cell Biology. 2010; 42: 429–432.
[76]
Olesen MS, Holst AG, Jabbari J, Nielsen JB, Christophersen IE, Sajadieh A, et al. Genetic loci on chromosomes 4q25, 7p31, and 12p12 are associated with onset of lone atrial fibrillation before the age of 40 years. The Canadian Journal of Cardiology. 2012; 28: 191–195.
[77]
Seifert MB, Olesen MS, Christophersen IE, Nielsen JB, Carlson J, Holmqvist F, et al. Genetic variants on chromosomes 7p31 and 12p12 are associated with abnormal atrial electrical activation in patients with early-onset lone atrial fibrillation. Annals of Noninvasive Electrocardiology. 2019; 24: e12661.
[78]
Weng L, Hall AW, Choi SH, Jurgens SJ, Haessler J, Bihlmeyer NA, et al. Genetic Determinants of Electrocardiographic P-Wave Duration and Relation to Atrial Fibrillation. Circulation-Genomic and Precision Medicine. 2020; 13: 387–395.
[79]
Sinner MF, Ellinor PT, Meitinger T, Benjamin EJ, Kääb S. Genome-wide association studies of atrial fibrillation: past, present, and future. Cardiovascular Research. 2011; 89: 701–709.
[80]
Husser D, Büttner P, Stübner D, Ueberham L, Platonov PG, Dinov B, et al. PR Interval Associated Genes, Atrial Remodeling and Rhythm Outcome of Catheter Ablation of Atrial Fibrillation-A Gene-Based Analysis of GWAS Data. Frontiers in Genetics. 2017; 8: 224.
[81]
Cheng S, Keyes MJ, Larson MG, McCabe EL, Newton-Cheh C, Levy D, et al. Long-term outcomes in individuals with prolonged PR interval or first-degree atrioventricular block. The Journal of the American Medical Association. 2009; 301: 2571–2577.
[82]
Imig JD, Hammock BD. Soluble epoxide hydrolase as a therapeutic target for cardiovascular diseases. Nature Reviews Drug Discovery. 2009; 8: 794–805.
[83]
Node K, Huo Y, Ruan X, Yang B, Spiecker M, Ley K, et al. Anti-inflammatory properties of cytochrome P450 epoxygenase-derived eicosanoids. Science. 1999; 285: 1276–1279.
[84]
Pidkovka N, Rao R, Mei S, Gong Y, Harris RC, Wang W, et al. Epoxyeicosatrienoic acids (EETs) regulate epithelial sodium channel activity by extracellular signal-regulated kinase 1/2 (ERK1/2)-mediated phosphorylation. The Journal of Biological Chemistry. 2013; 288: 5223–5231.
[85]
Chen J, Capdevila JH, Zeldin DC, Rosenberg RL. Inhibition of cardiac L-type calcium channels by epoxyeicosatrienoic acids. Molecular Pharmacology. 1999; 55: 288–295.
[86]
Imig JD. Prospective for cytochrome P450 epoxygenase cardiovascular and renal therapeutics. Pharmacology & Therapeutics. 2018; 192: 1–19.
[87]
Jamieson KL, Endo T, Darwesh AM, Samokhvalov V, Seubert JM. Cytochrome P450-derived eicosanoids and heart function. Pharmacology & Therapeutics. 2017; 179: 47–83.
[88]
Lai J, Chen C. The Role of Epoxyeicosatrienoic Acids in Cardiac Remodeling. Frontiers in Physiology. 2021; 12: 642470.
[89]
Zeldin DC. Epoxygenase pathways of arachidonic acid metabolism. The Journal of Biological Chemistry. 2001; 276: 36059–36062.
[90]
Merkel MJ, Liu L, Cao Z, Packwood W, Young J, Alkayed NJ, et al. Inhibition of soluble epoxide hydrolase preserves cardiomyocytes: role of STAT3 signaling. American Journal of Physiology-Heart and Circulatory Physiology. 2010; 298: H679–H687.
[91]
Fang Q, Liu X, Ding J, Zhang Z, Chen G, Du T, et al. Soluble Epoxide Hydrolase Inhibition Protected against Diabetic Cardiomyopathy through Inducing Autophagy and Reducing Apoptosis Relying on Nrf2 Upregulation and Transcription Activation. Oxidative Medicine and Cellular Longevity. 2022; 2022: 3773415.
[92]
Sosnowski DK, Jamieson KL, Gruzdev A, Li Y, Valencia R, Yousef A, et al. Cardiomyocyte-specific disruption of soluble epoxide hydrolase limits inflammation to preserve cardiac function. American Journal of Physiology. Heart and Circulatory Physiology. 2022; 323: H670–H687.
[93]
Przybyla-Zawislak BD, Srivastava PK, Vazquez-Matias J, Mohrenweiser HW, Maxwell JE, Hammock BD, et al. Polymorphisms in human soluble epoxide hydrolase. Molecular Pharmacology. 2003; 64: 482–490.
[94]
Fornage M, Boerwinkle E, Doris PA, Jacobs D, Liu K, Wong ND. Polymorphism of the soluble epoxide hydrolase is associated with coronary artery calcification in African-American subjects: The Coronary Artery Risk Development in Young Adults (CARDIA) study. Circulation. 2004; 109: 335–339.
[95]
Lee J, Dahl M, Grande P, Tybjaerg-Hansen A, Nordestgaard BG. Genetically reduced soluble epoxide hydrolase activity and risk of stroke and other cardiovascular disease. Stroke. 2010; 41: 27–33.
[96]
Sander AL, Jakob H, Sommer K, Sadler C, Fleming I, Marzi I, et al. Cytochrome P450-derived epoxyeicosatrienoic acids accelerate wound epithelialization and neovascularization in the hairless mouse ear wound model. Langenbeck’s Archives of Surgery. 2011; 396: 1245–1253.
[97]
Xiao YF, Huang L, Morgan JP. Cytochrome P450: a novel system modulating Ca2+ channels and contraction in mammalian heart cells. The Journal of Physiology. 1998; 508: 777–792.
[98]
Xiao Y, Ke Q, Seubert JM, Bradbury JA, Graves J, Degraff LM, et al. Enhancement of cardiac L-type Ca2+ currents in transgenic mice with cardiac-specific overexpression of CYP2J2. Molecular Pharmacology. 2004; 66: 1607–1616.
[99]
Xiao Y. Cyclic AMP-dependent modulation of cardiac L-type Ca2+ and transient outward K+ channel activities by epoxyeicosatrienoic acids. Prostaglandins and Other Lipid Mediators. 2007; 82: 11–18.
[100]
Marangoni K, Araújo TG, Neves AF, Goulart LR. The -786T>C promoter polymorphism of the NOS3 gene is associated with prostate cancer progression. BMC Cancer. 2008; 8: 273.
[101]
Brenol CV, Chies JAB, Brenol JCT, Monticielo OA, Franciscatto P, Birriel F, et al. Endothelial nitric oxide synthase T-786C polymorphism in rheumatoid arthritis: association with extraarticular manifestations. Clinical Rheumatology. 2009; 28: 201–205.
[102]
Nagassaki S, Herculano RD, Graeff CFO, Tanus-Santos JE. eNOS T-786C polymorphism affects atorvastatin-induced changes in erythrocyte membrane fluidity. European Journal of Clinical Pharmacology. 2008; 65: 385–392.
[103]
Bonilla IM, Sridhar A, Györke S, Cardounel AJ, Carnes CA. Nitric oxide synthases and atrial fibrillation. Frontiers in Physiology. 2012; 3: 105.
[104]
Tesauro M, Thompson WC, Rogliani P, Qi L, Chaudhary PP, Moss J. Intracellular processing of endothelial nitric oxide synthase isoforms associated with differences in severity of cardiopulmonary diseases: cleavage of proteins with aspartate vs. glutamate at position 298. Proceedings of the National Academy of Sciences of the United States of America. 2000; 97: 2832–2835.
[105]
Veldman BA, Spiering W, Doevendans PA, Vervoort G, Kroon AA, de Leeuw PW, et al. The Glu298Asp polymorphism of the NOS 3 gene as a determinant of the baseline production of nitric oxide. Journal of Hypertension. 2002; 20: 2023–2027.
[106]
Boycott HE, Nguyen M, Vrellaku B, Gehmlich K, Robinson P. Nitric Oxide and Mechano-Electrical Transduction in Cardiomyocytes. Frontiers in Physiology. 2020; 11: 606740.
[107]
Hong L, Zhang M, Ly OT, Chen H, Sridhar A, Lambers E, et al. Human induced pluripotent stem cell-derived atrial cardiomyocytes carrying an SCN5A mutation identify nitric oxide signaling as a mediator of atrial fibrillation. Stem Cell Reports. 2021; 16: 1542–1554.
[108]
Lenaerts I, Driesen RB, Hermida N, Holemans P, Heidbüchel H, Janssens S, et al. Role of nitric oxide and oxidative stress in a sheep model of persistent atrial fibrillation. Europace. 2013; 15: 754–760.
[109]
Garcia V, Sessa WC. Endothelial NOS: perspective and recent developments. British Journal of Pharmacology. 2018; 176: 189–196.
[110]
Grune J, Yamazoe M, Nahrendorf M. Electroimmunology and cardiac arrhythmia. Nature Reviews Cardiology. 2021; 18: 547–564.
[111]
Karam BS, Chavez-Moreno A, Koh W, Akar JG, Akar FG. Oxidative stress and inflammation as central mediators of atrial fibrillation in obesity and diabetes. Cardiovascular Diabetology. 2017; 16: 120.
[112]
Oikonomou E, Zografos T, Papamikroulis G, Siasos G, Vogiatzi G, Theofilis P, et al. Biomarkers in Atrial Fibrillation and Heart Failure. Current Medicinal Chemistry. 2019; 26: 873–887.
[113]
Schnabel RB, Kerr KF, Lubitz SA, Alkylbekova EL, Marcus GM, Sinner MF, et al. Large-scale candidate gene analysis in whites and African Americans identifies IL6R polymorphism in relation to atrial fibrillation: the National Heart, Lung, and Blood Institute’s Candidate Gene Association Resource (CARe) project. Circulation-Cardiovascular Genetics. 2011; 4: 557–564.
[114]
Smit MD, Maass AH, De Jong AM, Muller Kobold AC, Van Veldhuisen DJ, Van Gelder IC. Role of inflammation in early atrial fibrillation recurrence. Europace. 2012; 14: 810–817.
[115]
Frustaci A, Chimenti C, Bellocci F, Morgante E, Russo MA, Maseri A. Histological substrate of atrial biopsies in patients with lone atrial fibrillation. Circulation. 1997; 96: 1180–1184.
[116]
Zhang Z, Zhang C, Wang H, Zhao J, Liu L, Lee J, et al. n-3 polyunsaturated fatty acids prevents atrial fibrillation by inhibiting inflammation in a canine sterile pericarditis model. International Journal of Cardiology. 2011; 153: 14–20.
[117]
Narducci ML, Pelargonio G, Dello Russo A, Casella M, Biasucci LM, La Torre G, et al. Role of tissue C-reactive protein in atrial cardiomyocytes of patients undergoing catheter ablation of atrial fibrillation: pathogenetic implications. Europace. 2011; 13: 1133–1140.
[118]
Tanaka M, Imano H, Kubota Y, Yamagishi K, Umesawa M, Muraki I, et al. Serum High-Sensitivity C-Reactive Protein Levels and the Risk of Atrial Fibrillation in Japanese Population: the Circulatory Risk in Communities Study. Journal of Atherosclerosis and Thrombosis. 2021; 28: 194–202.
[119]
Conen D, Ridker PM, Everett BM, Tedrow UB, Rose L, Cook NR, et al. A multimarker approach to assess the influence of inflammation on the incidence of atrial fibrillation in women. European Heart Journal. 2010; 31: 1730–1736.
[120]
Kwon CH, Kang JG, Lee HJ, Kim NH, Sung J, Cheong E, et al. C-reactive protein and risk of atrial fibrillation in East Asians. Europace. 2017; 19: 1643–1649.
[121]
Boyalla V, Harling L, Snell A, Kralj-Hans I, Barradas-Pires A, Haldar S, et al. Biomarkers as predictors of recurrence of atrial fibrillation post ablation: an updated and expanded systematic review and meta-analysis. Clinical Research in Cardiology. 2022; 111: 680–691.
[122]
Okumura Y, Watanabe I, Nakai T, Ohkubo K, Kofune T, Kofune M, et al. Impact of biomarkers of inflammation and extracellular matrix turnover on the outcome of atrial fibrillation ablation: importance of matrix metalloproteinase-2 as a predictor of atrial fibrillation recurrence. Journal of Cardiovascular Electrophysiology. 2011; 22: 987–993.
[123]
Wilhelm M, Zueger T, De Marchi S, Rimoldi SF, Brugger N, Steiner R, et al. Inflammation and atrial remodeling after a mountain marathon. Scandinavian Journal of Medicine & Science in Sports. 2012; 24: 519–525.
[124]
Psychari SN, Apostolou TS, Sinos L, Hamodraka E, Liakos G, Kremastinos DT. Relation of elevated C-reactive protein and interleukin-6 levels to left atrial size and duration of episodes in patients with atrial fibrillation. The American Journal of Cardiology. 2005; 95: 764–767.
[125]
Ryter SW, Alam J, Choi AMK. Heme oxygenase-1/carbon monoxide: from basic science to therapeutic applications. Physiological Reviews. 2006; 86: 583–650.
[126]
Exner M, Minar E, Wagner O, Schillinger M. The role of heme oxygenase-1 promoter polymorphisms in human disease. Free Radical Biology & Medicine. 2004; 37: 1097–1104.
[127]
Mason JC, Ali F. Heme Oxygenase-1 and Atherosclerosis. In George SJ, Johnson J (eds.) Atherosclerosis: Molecular and Cellular Mechanisms (pp. 301–323). Wiley: Weinheim, Germany. 2010.
[128]
Morita T, Imai T, Sugiyama T, Katayama S, Yoshino G. Heme oxygenase-1 in vascular smooth muscle cells counteracts cardiovascular damage induced by angiotensin II. Current Neurovascular Research. 2005; 2: 113–120.
[129]
Melo LG, Agrawal R, Zhang L, Rezvani M, Mangi AA, Ehsan A, et al. Gene therapy strategy for long-term myocardial protection using adeno-associated virus-mediated delivery of heme oxygenase gene. Circulation. 2002; 105: 602–607.
[130]
Hsu L, Yeh Y, Kuo C, Chen Y, Chang G, Tsai F, et al. Microsatellite polymorphism in the heme oxygenase-1 gene promoter and the risk of atrial fibrillation in Taiwanese. PLoS ONE. 2014; 9: e108773.
[131]
Yeh Y, Hsu L, Chen Y, Kuo C, Chang G, Chen W. Protective role of heme oxygenase-1 in atrial remodeling. Basic Research in Cardiology. 2016; 111: 58.
[132]
Vest JA, Wehrens XHT, Reiken SR, Lehnart SE, Dobrev D, Chandra P, et al. Defective cardiac ryanodine receptor regulation during atrial fibrillation. Circulation. 2005; 111: 2025–2032.
[133]
Arvanitis DA, Vafiadaki E, Johnson DM, Kranias EG, Sanoudou D. The Histidine-Rich Calcium Binding Protein in Regulation of Cardiac Rhythmicity. Frontiers in Physiology. 2018; 9: 1379.
[134]
Han P, Cai W, Wang Y, Lam CK, Arvanitis DA, Singh VP, et al. Catecholaminergic-induced arrhythmias in failing cardiomyocytes associated with human HRCS96A variant overexpression. American Journal of Physiology-Heart and Circulatory Physiology. 2011; 301: H1588–H1595.
[135]
Tzimas C, Johnson DM, Santiago DJ, Vafiadaki E, Arvanitis DA, Davos CH, et al. Impaired calcium homeostasis is associated with sudden cardiac death and arrhythmias in a genetic equivalent mouse model of the human HRC-Ser96Ala variant. Cardiovascular Research. 2017; 113: 1403–1417.
[136]
Nattel S, Dobrev D. The multidimensional role of calcium in atrial fibrillation pathophysiology: mechanistic insights and therapeutic opportunities. European Heart Journal. 2012; 33: 1870–1877.
[137]
Yue L, Feng J, Gaspo R, Li GR, Wang Z, Nattel S. Ionic remodeling underlying action potential changes in a canine model of atrial fibrillation. Circulation Research. 1997; 81: 512–525.
[138]
Kneller J, Sun H, Leblanc N, Nattel S. Remodeling of Ca(2+)-handling by atrial tachycardia: evidence for a role in loss of rate-adaptation. Cardiovascular Research. 2002; 54: 416–426.
[139]
Pollak AJ, Haghighi K, Kunduri S, Arvanitis DA, Bidwell PA, Liu G, et al. Phosphorylation of serine96 of histidine-rich calcium-binding protein by the Fam20C kinase functions to prevent cardiac arrhythmia. Proceedings of the National Academy of Sciences of the United States of America. 2017; 114: 9098–9103.
[140]
Kerchberger VE, Huang Y, Koyama T, Shoemaker MB, Darbar D, Bastarache JA, et al. Clinical and Genetic Contributors to New-Onset Atrial Fibrillation in Critically Ill Adults. Critical Care Medicine. 2020; 48: 22–30.
[141]
Rizvi F, Mirza M, Olet S, Albrecht M, Edwards S, Emelyanova L, et al. Noninvasive biomarker-based risk stratification for development of new onset atrial fibrillation after coronary artery bypass surgery. International Journal of Cardiology. 2020; 307: 55–62.
[142]
Siebert J, Lewicki Ł, Myśliwska J, Młotkowska M, Rogowski J. ScaI atrial natriuretic peptide gene polymorphisms and their possible association with postoperative atrial fibrillation - a preliminary report. Archives of Medical Science. 2017; 13: 568–574.
[143]
Kertai MD, Li YJ, Ji Y, Qi W, Lombard FW, Shah SH, et al. Genome-wide association study of new-onset atrial fibrillation after coronary artery bypass grafting surgery. American Heart Journal. 2015; 170: 580–590.e28.
[144]
Plante I, Fournier D, Mathieu P, Daleau P. A pilot study to estimate the feasibility of assessing the relationships between polymorphisms in hKv1.5 and atrial fibrillation in patients following coronary artery bypass graft surgery. The Canadian Journal of Cardiology. 2008; 24: 41–44.
[145]
Kiliszek M, Szpakowicz A, Franaszczyk M, Pepinski W, Waszkiewicz E, Skawronska M, et al. The 9p21 polymorphism is linked with atrial fibrillation during acute phase of ST-segment elevation myocardial infarction. Heart and Vessels. 2015; 31: 1590–1594.
[146]
Muse ED, Wineinger NE, Spencer EG, Peters M, Henderson R, Zhang Y, et al. Validation of a genetic risk score for atrial fibrillation: A prospective multicenter cohort study. PLoS Medicine. 2018; 15: e1002525.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top