Next Article in Journal
Plaque Rupture in a Hodgkin Lymphoma Survivor without Cardiovascular Risk Factors 20 Years after Thoracic Radiotherapy: A Case Report
Previous Article in Journal
SGLT2 Inhibitors vs. GLP-1 Agonists to Treat the Heart, the Kidneys and the Brain
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Obesity-Related Atrial Fibrillation: Cardiac Manifestation of a Systemic Disease

by
Worawan B. Limpitikul
1 and
Saumya Das
1,2,*
1
Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
2
Demoulas Family Foundation Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA 02114, USA
*
Author to whom correspondence should be addressed.
J. Cardiovasc. Dev. Dis. 2023, 10(8), 323; https://doi.org/10.3390/jcdd10080323
Submission received: 30 June 2023 / Revised: 27 July 2023 / Accepted: 27 July 2023 / Published: 30 July 2023
(This article belongs to the Section Cardiovascular Clinical Research)

Abstract

:
Atrial fibrillation (AF) is the most common arrhythmia worldwide and is associated with increased morbidity and mortality. The mechanisms underlying AF are complex and multifactorial. Although it is well known that obesity is a strong risk factor for AF, the mechanisms underlying obesity-related AF are not completely understood. Current evidence proposes that in addition to overall hemodynamic changes due to increased body weight, excess adiposity raises systemic inflammation and oxidative stress, which lead to adverse atrial remodeling. This remodeling includes atrial fibrosis, atrial dilation, decreased electrical conduction between atrial myocytes, and altered ionic currents, making atrial tissue more vulnerable to both the initiation and maintenance of AF. However, much remains to be learned about the mechanistic links between obesity and AF. This knowledge will power the development of novel diagnostic tools and treatment options that will help combat the rise of the global AF burden among the obesity epidemic.

1. Introduction

Atrial fibrillation (AF) is the most common arrhythmia in adults. It is associated with increased morbidity and mortality due to an increased risk of stroke, heart failure, death [1], increased hospitalization [2], and decreased quality of life [3]. AF constitutes a significant public health and economic burden. AF-related healthcare costs account for about 1% of the National Health Service budget in the United Kingdom [4] and between 6 and 26 billion dollars of annual healthcare spending in the United States [5,6]. The etiology of AF is multifactorial, encompassing genetic, environmental, and lifestyle-related risk factors. Most notably, obesity is known to be strongly associated with increased risk of AF, while strategies to induce weight loss may be associated with decreased risk of future AF. However, the mechanistic underpinning of obesity-related AF remains incompletely understood and is evolving. This review aims to provide a comprehensive synthesis of the current understanding of the mechanisms underlying obesity-related AF and discuss potential future directions of AF research.

2. Epidemiology of Obesity and AF

The prevalence of AF worldwide is around 33.5 million patients, and it affects 2.5–3.5% of all individuals across the globe [7]. The AF burden is high across races and sexes. For example, the lifetime risk of AF in adults is 36% in Caucasian men, 30% in Caucasian women, 21% in African American men, and 22% in African American women [8]. Although there is slightly more limited data on the Asian population, a nationwide cohort in Taiwan showed a 16% and 14% lifetime risk of AF in men and women, respectively [9]. The incidence of AF is also predicted to double, from 1.2 million cases in 2010 to 2.6 million cases in 2030, while the prevalence is projected to increase from 5.2 million in 2010 to 12.1 million cases in 2030 [10]. Importantly, as one-third of the total AF population is asymptomatic [11], these numbers are likely to be underestimated.
The key question is what drives the increase in the AF burden. Although improved detection rates in the era of consumer-driven usage of smartphones and wearables may partly explain this increase [12], changes in intrinsic characteristics of the population likely remain the major driver of increased AF prevalence and incidence. For example, as age is one of the strongest risk factors for AF [13,14], the aging of the overall population significantly contributes to the increased prevalence of AF. Among modifiable risk factors (hypertension, diabetes mellitus, obstructive sleep apnea, myocardial infarction, and heart failure), obesity is strongly associated with AF.
Obesity is defined by the World Health Organization as increased adiposity or excessive accumulation of fat that may impair health. Clinically, multiple metrics are used to quantify and define obesity. A commonly used metric is body mass index (BMI). For adults, a BMI of 25.0–29.9 kg/m2 indicates overweight, while a BMI over 30 kg/m2 indicates obesity. Three classes of obesity, by BMI, include classes I, II, and III for BMI of 30.0–34.9, 35.0–39.9, and 40 kg/m2 or higher, respectively. Additionally, obesity can be measured by waist circumference (≥102 cm in men and ≥88 cm in women in the Caucasian populations, and ≥90 cm in men and ≥80 cm in women in South Asian, Chinese, and Japanese populations [15]), waist-to-hip ratio, skin fold thickness, and percentage of body fat, etc. [16,17,18]. There is much debate about which metrics are better for defining obesity [19]. However, regardless of the metrics used to define obesity, there is a consistent trend of increased mortality with an increased degree of adiposity [20]. For example, a meta-analysis of 57 prospective studies with 900,000 adults showed that there was a 30% increase in overall mortality per 5 BMI units [21]. Unfortunately, due to changes in diet and lifestyle in modern society, the worldwide prevalence of overweight and obesity has increased significantly over the past decades. It is estimated that by 2030, one in three adults will be overweight, and one in five adults will be obese [22]. Additionally, a rapid rise in the prevalence of childhood obesity, which has increased eightfold over the past four decades in children aged 5–19 years old [23], will surely worsen the prevalence of obesity in future adult populations.
There are multiple lines of evidence linking obesity and AF. Numerous epidemiological studies have shown that obesity is associated with new-onset AF even after adjustment for other comorbidities associated with obesity [24,25,26]. A meta-analysis of 51 studies with over 600,000 individuals by Wong et al. showed a 29% greater risk of overall incident AF and a 10% greater risk of postoperative AF for each 5-unit increase in BMI [19,27]. Additionally, obesity is also associated with the progression of AF (change in AF type to more advanced types) [28,29]. For instance, a prospective cohort study of 3248 patients with a median follow-up period of 5.1 years demonstrated that class I obesity (BMI 30–34.9 kg/m2) is associated with a 54% increased risk of progression from paroxysmal to permanent AF [29]. Moreover, obesity increases the rate of recurrence after AF ablation [27,30] and cardioversion [31]. Specifically, there was a 13% increase in AF recurrence after an AF ablation and a 10% increase in recurrence after cardioversion per 5 units increase of BMI, as shown by a meta-analysis by Wong et al. [27] and an observational study of 2518 patients (a sub-study of the Atrial Fibrillation Follow-Up Investigation of Rhythm Management (AFFIRM) trial) [31], respectively.
Additionally, several Mendelian randomization studies have shown an association between obesity and AF. For instance, a cohort study of over 51,646 adults of European ancestry showed an association between BMI and incident AF [32]. Another Mendelian randomization study by Larsson et al. also demonstrated an association between BMI and AF in over 300,000 individuals of White British descent in the UK Biobank [33]. Moreover, there is an association between childhood obesity and AF in adults. A Mendelian randomization study of over 63,000 children showed that a higher BMI in childhood is associated with an increased risk of AF in adults [34]. Interestingly, an analysis by Zhou et al., which pooled data from 6 genome-wide association studies, demonstrated that the risk of AF in adulthood increases with earlier onset of obesity. They found that a 1 standard deviation increase in adult obesity, childhood BMI, and birth weight was associated with a 13%, 18%, and 26% increased risk of AF in adulthood, respectively [35]. These interesting findings may thus point to a causal relationship between obesity and AF.
Therefore, with the increase in the obesity burden and its association with AF, it is important to understand the mechanisms underlying obesity-associated AF. These understandings are a critical step in devising strategies for the treatment and prevention of AF.

3. Conceptual Models of AF

To understand the pathophysiology underlying obesity-related AF, we must first understand how AF is initiated and maintained.
There are multiple theories regarding the mechanisms of AF, dating back to the early 1900s. Classical models of AF include rapidly firing atrial ectopic foci, single re-entry circuits, and multiple functional re-entrant circuits [36]. Over the past several decades, the multiple re-entrant circuit theory has been the dominant conceptual model of AF. In the re-entry circuit model, there must be (1) triggers to initiate and (2) vulnerable substrates to sustain the circuits [37]. AF triggers are mainly located in the pulmonary veins and originate from sleeves of the atrial muscle that extend from the left atrium (LA) several centimeters into the pulmonary veins [38]. Ectopic activities that initiate re-entry circuits can be caused either by triggered activity or automaticity. There are two types of triggered activities: early afterdepolarizations (EADs) and delayed afterdepolarizations (DADs). EADs are spontaneous depolarization of the cell membrane during phases 2 and 3 of action potentials and are caused by abnormal openings of calcium channels or sodium channels. DADs are spontaneous depolarization during phase 4 of action potentials and are caused by spontaneous releases of calcium from an internal calcium store into cytosol, which leads to activation of the sodium-calcium exchanger. On the other hand, automaticity is caused by diastolic depolarization from a net inward current during phase 4 of the action potentials, most commonly from the funny current (HCN channels). Unlike cells of the sinoatrial node, which spontaneously initiate action potentials by diastolic depolarization, mature atrial and ventricular myocytes do not express HCN channels and thus lack this capability. In normal atrial tissue, the most common mechanism of ectopic activities that initiates AF is DADs. In animal studies, EADs became important only in the background of existing long-QT syndrome from abnormal sodium channel function [39] while automaticity is more likely to contribute to initiation of AF in an end-stage heart failure where current from HCN channels is upregulated as the atrial tissue reverts to a fetal phenotype [39].
To understand how vulnerable atrial substrate allows the perpetuation of AF, we will discuss two proposed models of the re-entrant circuits. One is the “leading circle” hypothesis, where a functional re-entry establishes itself in a circuit the size of the wavelength (WL). Here, WL is a product of conduction velocity (CV), how fast the action potential propagates from cells to cells, and refractory period (RP), the period immediately following stimulation during which cardiomyocytes cannot respond to further stimulation. In this hypothesis, the number of re-entrant circuits is a function of the atrial size and the WL of the re-entry circuits. For example, atrial dilation, a common finding in atria facing chronic volume and/or pressure overload, can support more re-entrant circuits compared to normal-sized atria. Additionally, a shorter WL, either from decreased CV or decreased RP, allows more re-entrant circuits to be sustained in the atria of the same size. Ultimately, having more circuits makes simultaneous termination of all circuits more difficult and thus increases the chance of AF to continue [36]. The second model is the “spiral wave” hypothesis, where maintenance of re-entry spirals depends on tissue excitability, propagation strength, RP, and the angle of curvature of the wavefront (the greater wavefront curvature, the slower CV). For instance, lower excitability and slower propagation (e.g., the use of a sodium channel blocker) limit the wavefront curvature, which mandates larger spirals [40,41].
In AF, there are both electrical and structural remodeling of atria that increase the propensity for the initiation and maintenance of AF. We will next discuss the dysregulation of adipocytes in the obese state and how this maladaptation leads to atrial electro-anatomical remodeling.

4. Adipose Tissue Biology

4.1. The Basics of Adipose Tissue

There are three major types of adipose tissue in mammals, each with unique physical and biochemical properties. These are brown, white, and beige adipose tissue. Brown adipose tissue (BAT) accounts for approximately 4% of total body fat. It resides mainly in the supraclavicular, para-aortic, para-vertebral, and supra-renal regions. The main function of BAT is thermogenesis. Therefore, brown adipocytes are small, multilocular, contain a large number of mitochondria, and express uncoupling protein-1, which allows heat generation by uncoupling the mitochondrial electron transport chain, thereby blocking ATP production and dissipating energy in the form of heat [42].
White adipose tissue (WAT), on the other hand, is the most abundant type of adipose tissue in the human body. The main function of WAT is storing energy in the form of triglycerides and secreting hormones that regulate hunger/satiety, metabolism, and inflammatory response. White adipocytes are bigger than brown adipocytes. They contain large fat droplets and few mitochondria. WAT can be subdivided into subcutaneous (SAT) and visceral (VAT) adipose tissue. While SAT occupies the space just beneath the layer of the skin, VAT resides deeper and wraps around internal organs, including the heart and blood vessels [42]. Compared to SAT, VAT is more vascularized and innervated [43]. It is also more metabolically active and plays a major role in energy balance and plasma glucose homeostasis. Adipocytes in VAT express more receptors to glucocorticoids, a key regulatory hormone for serum glucose levels via modifying metabolic adaptations during stress, such as fasting and starvation. VAT is also more sensitive to catecholamine-induced lipolysis due to the increased function of β3-adrenergic receptors [44]. Additionally, VAT contains more immune cells, including macrophages, which are key regulators of a chronic low-grade systemic inflammatory state in obesity. A special type of VAT includes epicardial fat, which lies directly over the surface of the myocardium with no intervening fascial plane and shares the same microcirculation as the epicardium. Additionally, epicardial adipose tissue also contains ganglionated plexi, an intrinsic cardiac nervous system that directly innervates the myocardium. Its unique position allows epicardial fat to exert direct influence on underlying atrial tissue.
The third type of adipose tissue is beige or brown-in-white adipose tissue. Beige adipocytes are more similar to brown adipocytes in that they contain multilocular lipid droplets and numerous mitochondria. However, they reside inside WAT depots. In humans, it is postulated that beige adipocytes are derived from the “browning” of white adipocytes in response to stimulation of β3-adrenergic receptors, for example, during chronic cold exposure or exercise [45].

4.2. Remodeling of Adipose Tissue during Obesity

In a state of positive energy balance, such as obesity, adipose tissue undergoes several remodeling processes to increase its lipid storage capacity. Adipocytes in WAT undergo hypertrophy (increase in cell size) of existing mature adipocytes and hyperplasia (increase in cell number) of resident adipocyte precursors. Adipocyte hypertrophy that exceeds angiogenesis capacity creates areas of local hypoxia, leading to cell necrosis and subsequently recruitment of macrophages and other immune cells [46]. These immune cells adopt proinflammatory phenotypes in an obese state, creating a chronic low-grade systemic inflammatory state leading to a spectrum of metabolic abnormalities, including insulin resistance, type 2 diabetes mellitus, and fatty liver disease [47,48]. In addition to cellular changes, the extracellular matrix surrounding these cells also has excess deposition of collagen, elastin, and fibronectin. These changes lead to increased fibrosis and thus rigidity of the extracellular matrix, which physically limits the expansion of adipocyte mass. As obesity progresses, the maximal lipid storage capacity is exceeded, and lipid storage shifts to ectopic sites, such as the viscera, heart, and vasculature [49].

5. Mechanisms Underlying Obesity-Related AF

The pathophysiology of obesity-related AF is multifactorial and remains incompletely understood. One approach to conceptualizing this complex process is to use the framework of AF mechanisms (Section 3) and consider changes that increase the propensity of AF initiation (triggers) and/or create a more vulnerable substrate for AF maintenance.

5.1. Hemodynamic Changes

To keep up with the metabolic demand from excess body weight, obese individuals have increased circulating blood volume and thus increased cardiac output via increased stroke volume [15,50]. Furthermore, the left ventricle (LV) in obese individuals with commonly associated hypertension also faces an additional elevation of an afterload. Chronic increases in wall tension from both increased pressure and chamber dilation lead to LV hypertrophy and LV diastolic dysfunction. Chronically stiff LV and elevated LV filling pressure ultimately cause increased LA filling pressure and LA dilation. This chronic LA stretch leads to a release of atrial natriuretic peptide, angiotensin II, endothelin 1, and transforming growth factor-β, inflammatory cytokines that ultimately lead to atrial myocyte hypertrophy and atrial fibrosis [51]. Overall atrial remodeling has a synergistic effect in promoting AF. That is, atrial dilation can support more re-entry circuits while atrial fibrosis causes a reduction of CV and thus WL, which also permits more re-entrant circuits to be sustained in the atrium.
In addition to directly altering hemodynamics, obesity also increases the risk of developing AF via obstructive sleep apnea (OSA), a highly prevalent sleep disorder among obese individuals [52]. OSA is strongly associated with AF [53]. Repetitive forced inspiration against a closed glottis results in a sudden drop in the intrathoracic pressure, which is transmitted to the LA. Repetitive LA stretch may result in remodeling of LA itself and pulmonary vein ostia [54], which are the most common sites of AF triggers [38]. Additionally, intermittent hypoxemia and hypercapnia can result in sympathetic activation, which increases the frequency of atrial arrhythmia triggering the initiation of AF [55].

5.2. Roles of Adipokines

Adipokines are paracrines secreted by adipocytes. In addition to controlling homeostasis of serum glucose levels and lipid metabolism, these adipokines also modulate the degree of systemic inflammation and could affect atrial remodeling and thus the risk of AF. There are numerous studies investigating the role of various adipokines in obesity-related AF, although some with conflicting results. Here, we will briefly discuss several key adipokines with the most evidence in relation to AF. Further details of clinical studies describing the association between various adipokines and AF can be found in a meta-analysis by Agbaedeng et al. [56].
Adiponectin is the most abundant adipokine in the body. It has anti-inflammatory and anti-fibrotic effects and is considered “cardioprotective” to vascular endothelial and cardiomyocytes [57]. A series of 100 patients undergoing cardiac surgeries demonstrated that increased epicardial fat volume, measured by cardiac computed tomography (CT), was associated with decreased expression of adiponectin in epicardial adipose tissue [58], hinting at the role of adiponectin in atrial remodeling and perhaps AF. However, there are mixed findings in terms of the relationship between serum adiponectin and AF. For example, a case-control study of 84 individuals with AF and 84 individuals in sinus rhythm showed that individuals with AF had significantly higher levels of serum adiponectin than those in sinus rhythm [59]. A sub-analysis of a prospective randomized AVOCADO (Aspirin Vs./Or Clopidogrel in Aspirin-resistant Diabetics inflammation Outcomes) trial with 273 type 2 diabetic participants demonstrated that participants with AF at baseline (12%) had higher levels of serum adiponectin compared to those without AF at baseline [60]. On the other hand, a case-control study of 80 patients hospitalized with AF and 165 controls showed no correlation between the absolute level of serum adiponectin and AF, but showed a negative correlation between the serum level adiponectin normalized with fat mass or visceral adipose tissue index and AF [61]. Lastly, an analysis of a community-based prospective cohort, the Cardiovascular Health Study (CHS), of 3,190 participants with a median follow-up of 11.4 years and 28% incident AF showed that incident AF was associated with an elevated level of serum adiponectin measured at the baseline visit [62]. Conflicting observations of the association between serum adiponectin levels and AF among various studies, even after adjusting for confounding factors, could indicate nuances of incompletely understood roles of this adipokine in AF. For example, the discrepancy of adiponectin level in serum versus epicardial adipose tissue may hint at a potentially larger influence of epicardial fat depots, which share the same microcirculation as underlying cardiomyocytes, compared to other VAT in AF.
Similarly, current evidence for the role of leptin in AF remains conflicting. In a diet-induced obese mouse model, leptin mediates atrial fibrosis and AF [63] and a higher level of serum leptin is associated with obesity [64]. However, there are conflicting reports from observational studies on the association between serum leptin levels and AF. For instance, a case-control study of 80 patients with AF showed an increased level of serum leptin compared to the control [61] while a sub-analysis of a prospective cohort of 273 patients with type 2 diabetes mellitus showed no association between serum leptin level and AF [60].
Apelin, a known cardioprotective adipokine, has been shown in an animal study to reduce angiotensin II-induced atrial fibrosis and atrial fibrillation [65]. In humans, a lower serum level of apelin is associated with an increased risk of developing postoperative AF, as observed in a retrospective case-control study of 508 patients who underwent off-pump coronary artery bypass graft (CABG) surgeries [66]. Moreover, a lower serum level of apelin is shown to be associated with AF recurrence after an intervention. For example, in a series of 93 patients with persistent AF who underwent cardioversion, individuals with lower levels of apelin at baseline had a higher rate of AF recurrence at the 6-month follow-up [67]. Another series of 61 patients with AF who underwent pulmonary vein isolation showed that patients with lower serum levels of apelin at baseline had a higher rate of AF recurrence at 6 months post-procedure [68]. Interestingly, comparing 200 obese women to 200 lean women, Zaki et al. found that obese women had a higher level of serum apelin compared to their lean counterparts [69]. Additionally, a cross-sectional study of 740 patients who underwent weight loss from bariatric surgeries showed a lower level of messenger RNA expression of apelin in omental and subcutaneous adipose tissue compared to their baseline level [70]. This finding may raise the possibility of a compensatory increase of leptin in patients with AF to counteract cardiac remodeling from obesity and requires further studies. Given the potential therapeutic targets of adipokines in obesity-induced AF, further studies are warranted.

5.3. Oxidative Stress

In a chronic state of excess energy balance, where the lipid storage capacity of fat depots is exceeded, there is an elevated level of circulating free fatty acids and triglycerides. In this state, cardiomyocytes have an increased uptake of free fatty acids, leading to increased fatty acid β-oxidation [71]. Additionally, elevated plasma level of free fatty acids also increases expression of mitochondrial uncoupling protein-3 [72], which leads to further increase in mitochondrial fatty acid oxidation and thus increased production of reactive oxygen species (ROS) [73]. This overall increase in oxidative stress leads to both structural and electrical remodeling of atrial tissue. For example, diet-induced obesity is associated with activation of endoplasmic reticulum oxidative stress, which leads to atrial fibrosis and increased AF inducibility in a mouse model [74]. High oxidative stress also leads to alteration of intracellular calcium homeostasis and alteration of CV, which will be further discussed in the following section.

5.4. Roles of Epicardial Fat in Obesity-Related AF

As discussed previously, the unique anatomical location of the epicardial fat depot with a shared circulation with the underlying epicardium allows epicardial adipose tissue to exert direct mechanical and chemical influences on underlying atrial tissue. Obese individuals have an increased volume of epicardial fat, as evidenced by an autopsy series dating back to 1933, which showed increased excess of epicardial fat in 136 obese individuals [75]. Moreover, an observational study of obese (n = 99) and lean (n = 96) patients with heart failure with preserved ejection fraction showed that epicardial fat thickness measured by transthoracic echocardiogram was 20% thicker in obese patients compared to lean individuals [76].
Multiple studies have shown that increased epicardial fat thickness and/or volume measured by various imaging modalities is associated with an increased incidence of AF. For example, two observational studies using a subset of the Framingham Heart Study cohort showed that increased epicardial fat volume measured by CT was associated with 40% higher odds of prevalent AF (n = 3217) [77] and abnormal atrial conduction (n = 1946) [78]. Additionally, a case-control study of 53 adults with AF and 52 adults without AF showed that the odds of AF increase by 42% for each milliliter increase of epicardial fat volume measured by cardiac magnetic resonance imaging (MRI), even after adjusting for LA volume [79]. A meta-analysis of 63 observational studies, including MRI, CT, and echocardiography, showed that a one standard deviation increase in epicardial fat volume was associated with 2.6-fold higher odds of AF [80]. Additionally, a series of 115 patients undergoing AF ablation showed that atrial remodeling, including lower voltage, slower CV, and greater fractionation of electrograms, was more pronounced in the regions of the atria adjacent to epicardial adipose tissue [81].
The mechanistic links between epicardial fat and AF are likely multifactorial. In addition to the potential effects of adipokines similar to those secreted from other VAT depots, epicardial adipose tissue poses several other mechanisms due to its unique location relative to atrial myocytes. For example, the proximity of epicardial fat allows direct mechanical compression of the atria [82]. Moreover, excess adiposity can lead to the direct infiltration of epicardial adipose tissue into atrial tissue. This is observed in both obesity-induced animal models [83] and in humans. In a series of 92 patients undergoing CABG surgeries, analysis of atrial tissue samples demonstrated significant fibro-fatty infiltration in tissue from patients with AF compared to those without AF [83]. Additionally, an autopsy series of 30 individuals showed increased fatty infiltration of atrial tissue in patients with a history of AF compared to controls [84]. Interestingly, in a series of 90 adults in sinus rhythm, the amount of intra-atrial fat measured by cardiac MRI monotonically increased with the growing risk of developing AF as measured by the ARIC risk score [85]. Ultimately, this fatty infiltration leads to atrial fibrosis and gap-junction remodeling with subsequent reduction of atrial CV and increase in conduction heterogeneity, which promote re-entry circuits [86].

5.5. Atrial Tissue Electro-Anatomical Remodeling in Obesity

Physiologic changes brought on by obesity alter both the electrical and structural properties of atrial tissue at a molecular level, which ultimately create a substrate primed for the initiation and maintenance of AF (Figure 1 and Table 1).
In terms of AF initiation or triggers, obesity is linked to increased oxidative stress in cardiomyocytes. In this state, ROS activates calcium/calmodulin-dependent protein kinase II (CaMKII), which leads to hyperphosphorylation of ryanodine receptors (RyR) on the sarcoplasmic reticulum (SR), a major intracellular calcium store [87,88]. In addition to increased phosphorylation, RyR is oxidized in a high oxidative stress environment [89]. Both changes lead to increased calcium leak from the SR, creating more DADs, a major source of triggers for AF initiation.
In terms of vulnerable substrates, studies conducted in various animal models demonstrated that diet-induced obese animals have increased LA fibrosis [90,91,92,93], increased atrial CV heterogeneity [94,95,96], decreased atrial RP [91,92], and thus, overall increased AF inducibility [90,91,92,93]. At the molecular level, atrial fibrosis is linked to multiple signaling pathways. For example, it is linked to increased expression of cadherin-11 [90], an adhesion molecule secreted by adipose tissue fibroblasts with major roles in the regulation of atrial tissue elasticity via altering collagen and elastin synthesis [97] and regulation of adipose tissue inflammation in obesity [98]. In a series of 43 patients undergoing CABG surgeries, expression of cadherin-11 in LA tissue excised intraoperatively was the highest in obese patients with AF, followed by lean patients with AF, and patients in normal sinus rhythm [90]. Moreover, atrial fibrosis is also associated with decreased expression of myeloid differentiation protein 1 (MD1), a key regulator in atrial modeling in response to inflammation [99]. Additionally, atrial fibrosis and inflammation are also associated with serum glucocorticoid kinase 1 (SGK1), a kinase molecule downstream of both insulin and mineralocorticoid signaling pathways [100]. Diet-induced obese mice with dominant-negative SGK1 demonstrated a reduction of AF fibrosis and proinflammatory signaling, while obese mice with constitutively active SGK1 had increased expression of proinflammatory and fibrotic molecules, compared to obese wild-type mice. Overall, atrial fibrosis leads to a reduction in CV and increased CV heterogeneity, both of which increase AF inducibility and maintenance.
A similar disturbance in electrical conduction has also been observed in humans. For example, in 212 patients without AF at baseline who underwent cardiac surgeries, intraoperative epicardial electrical mapping of the right and left atria showed more conduction delay and a higher incidence of postoperative AF in obese compared to lean patients [101]. Interestingly, conduction delay was the most severe in pulmonary veins and the Bachmann bundle, an inter-atrial connection that ensures bi-atrial synchronous contraction. In addition to atrial fibrosis, CV reduction observed in diet-induced obese animal models is associated with decreased voltage-gated sodium current (INa or NaV1.5), which is responsible for phase 0 (upstroke phase) of action potentials [102] and decreased expression of connexin 40 and 43, key proteins in gap junctions [90].
Lastly, electrical remodeling of atrial tissue in obese individuals also includes alteration of ion channel expression and/or membrane current density, which ultimately alters WL and thus AF inducibility. The atrial effective refractory period (ERP) is a term used in clinical electrophysiological studies and is defined as the longest interval between two pacing stimuli that fails to capture the atrium. In a series of 63 patients with AF undergoing catheter ablation, obese patients were found to have shorter ERP compared to lean patients [103]. At the cellular level, ERP spans phases 1, 2, and 3 of cardiac action potentials. The duration of phases 1, 2, and 3 is determined by the balance between the efflux of various types of potassium currents and the influx of calcium currents. A reduction in ERP was observed in multiple diet-induced animal models and was associated with an increase in ultra-rapid delayed rectifier potassium current (IKur or KV1.5) [91,102,104,105], increase in rapid and slow delayed rectifier potassium current (IKr or KV11.1 or hERG and IKs or KV7.1 or KCNQ1) [104], increase in transient outward potassium current (Ito or KV4.2/4.3) [105], and decrease in L-type calcium current (ICa,L or CaV1.2) [102,104]. Overall, a reduction in CV and RP leads to a reduction in WL and thus an increased probability of AF maintenance.

6. Reversibility of Obesity-Induced AF and Remodeling

Multiple studies have demonstrated an association between weight loss and AF burden and progression. An observational study of 1,415 patients with AF who achieved weight loss after participation in a physician-led lifestyle modification program showed that long-term sustained weight loss was associated with maintenance of sinus rhythm [106], reduction of AF-associated symptoms [107], and delay in progression of AF from paroxysmal to persistent [15,108]. Another observational study showed that 239 morbidly obese patients who underwent bariatric surgery with resultant weight loss prior to AF catheter ablation had a lower rate of AF recurrence than those who did not undergo bariatric surgery [109]. A meta-analysis of patients with weight loss, either by lifestyle modification or bariatric surgery, showed a dose-response reduction in the frequency of AF recurrence after AF ablation [110]. Mechanistically, this may be explained by atrial reverse remodeling. In an obese sheep model, weight reduction was associated with decreased atrial fibrosis, increased atrial ERP, improved CV, decreased CV heterogeneity, and decreased AF inducibility [111]. As obesity plays a critical role in incident and recurrent AF, the reversibility of this process has significant public health implications as the rate of obesity continues to grow worldwide.
Table 1. Summary of studies with plausible mechanisms of obesity-related atrial remodeling.
Table 1. Summary of studies with plausible mechanisms of obesity-related atrial remodeling.
StudiesModelsResultsPotential Signaling Pathways Involved
Mahajan et al., 2015 [95]Sheep ± calorie-dense diet- Obese (vs. lean): ↑ LA volume, ↑ LA pressure, ↓ CV, ↑ CV heterogeneity, ↔ ERP, ↔ ERP heterogeneity, ↑ AF burden, ↑ epicardial fat infiltrate in posterior LA and ↓ voltage in this region, ↑ atrial fibrosis and TGF-β TGF-β (profibrotic)
Mahajan et al., 2021 [111]Sheep ± calorie-dense diet ± weight loss (by decreasing diet intake)- Sustained obesity (vs. lean): ↑ LA pressure, ↑ inflammation, ↑ atrial TGF-β, ↑ endothelin-B receptor expression, ↑ atrial fibrosis, ↑ epicardial fat infiltration, ↓ ERP, ↓ Cx43 and ↓ CV, ↑ CV heterogeneity, ↑ duration of induced AF
- 30% weight loss (vs. obese) had ↓ LA pressure, ↓ inflammation, ↓ endothelin-B receptor expression, ↓ atrial fibrosis, ↑ ERP, ↑ Cx43 and ↑ CV, ↓ CV heterogeneity, ↓ duration of induced AF
TGF-β (profibrotic)
Endothelin-1 (profibrotic)
Fang et al., 2021 [90]Human LA tissues from patients in sinus rhythm vs. obesity + AF vs. lean + AF- ↑ Cad-11 in LA tissue: obesity + AF > lean + AF > sinus rhythmCad-11 (profibrotic, proinflammatory) via MAPK-κB and NF-κB
Mice ± high-fat diet;
WT vs. Cad-11−/−
- Obese WT (vs. lean WT): ↑ LA diameter, ↑ LA fibrosis, ↑ Cad-11 expression in LA, ↑ p-wave duration on ECG, ↓ Cx40 and Cx43 expression, ↑ lateralization of Cx, ↑ AF inducibility, and duration
- Obese Cad-11−/− (vs. obese WT): ↓ LA diameter, ↓ LA fibrosis, ↓ expression of proinflammatory cytokines, ↑ Cx40 and Cx43 expression, ↓ AF inducibility and duration
Scott et al., 2021 [91]Human RAA tissue from obese vs. lean patients- Obese (vs. lean): ↑ NLRP3 inflammasome activation in RAA tissueNLRP3 inflammasome (proinflammatory) increases IL-1β and activates CaMKII which phosphorylates SR
Sheep ± calorie-dense diet- Obese (vs. lean): ↑ NLRP3 inflammasome activation, ↑ AF inducibility and duration, ↑ ERP
Mice ± high-fat diet;
WT vs. NLRP3−/−
- Obese WT (vs. lean WT): ↑ AF inducibility and AF duration, ↓ EP, ↓ APD, ↑ IKur, ↑ calcium leak frequency from SR, ↑ atrial fibrosis
- Obese NLRP3−/− (vs. obese WT): ↓ AF inducibility, ↓ IKur, ↓ calcium leak frequency from SR
Otsuka et al., 2021 [92]Dogs ± high-fat diet ± rapid RA pacing- Obese (vs. lean): ↑ AF inducibility, ↑ epicardial fat size, ↑ myocardial fat infiltrate, ↑ atrial tissue interstitial fibrosisNot discussed
Okumura et al., 2015 [93]Pigs ± high-fat diet- Obese (vs. lean): ↑ LA pressure, ↓ ERP of PVs, ↑ duration of induced AFNot discussed
Abed et al., 2013 [94]Sheep ± calorie-dense diet- Obese (vs. lean): ↑ LA volume, ↑ LA fibrosis, ↑ LA inflammatory infiltrates, ↑ myocardial fat infiltrate, ↓ CV, ↔ ERP, ↑ endothelin-A and -B receptors, ↑ endothelin-1Endothelin-1 (profibrotic)
Shuai et al., 2019 [99]WT vs. MD1−/− mice + high-fat diet- Obese MD1−/− (vs. obese WT): ↑ LA fibrosis, ↑ IL-1β, IL-6, and TNF-α (proinflammatory), ↓ IL-10 (anti-inflammatory), ↑ AF inducibilityMD1 via TLR4/NF-κB pathway (proinflammatory)
Bapat et al., 2022 [100]Mice ± high-fat diet; WT vs. SGK1−/− vs. SGK1CA- Obese WT (vs. lean WT): ↑ SGK1 expression, ↑ PACs on continuous telemetry, ↑ AF inducibility, ↔ CV, ↔ APD,
- Obese SGK1−/− (vs. obese WT): ↓ AF inducibility, ↓ APD, ↔ INa current density, rightwards/depolarizing shift of INa inactivation kinetics, ↓ atrial fibrosis, ↓ atrial proinflammatory signaling (↓ NLRP3, ↓ IL-1β), ↓ collagen-I expression
- Obese SGK1CA (vs. obese WT): ↑ PACs on continuous telemetry, ↑ AF inducibility, ↑ APD, ↑ CV, ↑ NLRP3 expression, ↑ collagen-I expression
SGK1 (part of insulin signaling and mineralocorticoid pathways)
McCauley et al., 2020 [102]Mice ± high-fat diet ± mitochondrial antioxidant- Obese (vs. lean): ↑ AF inducibility, ↓ APD, ↓ INa expression and current density, ↓ ICa,L expression and current density, ↑ IKur expression and current density, ↑ atrial fibrosis, ↑ F2-IsoPs (↑ oxidative stress), ↑ NOX2 expression, ↑ PKC-α/δ expression
- Mitochondrial antioxidant: ↓ AF burden, ↑ INa, ↓ IKur, ↑ ICa,L, ↑ APD, reverse atrial fibrosis
Mitochondrial oxidative stress
Martinez-Mateu et al., 2019 [104]Guinea pigs ± high-fat diet- Obese (vs. lean): ↑ IKr, + IKs, ↓ ICa,L current density, ↔ IK1Not discussed
Zhang et al., 2016 [105]Mice ± high-fat diet- Obese (vs. lean): shorter PR-interval on ECG, ↑ SAN recovery time, ↓ ERP, ↑ IKur expression, ↑ Ito expressionNot discussed
Abbreviations: ↑, increased; ↓, decreased; ↔, unchanged; −/−, homozygous knockout; CA, constitutive activation; LA, left atrium; CV, conduction velocity; (E)RP, (effective) refractory period; AF, atrial fibrillation; TGF-β, transforming growth factor-β; Cx, connexin; WT, wild-type; Cad-11, cadherin-11; ECG, electrocardiogram; MAPK-κB, mitogen activated protein kinase-κB; NF-κB, nuclear factor-κB; RAA, right atrial appendage; NLRP3, NACHT, LRR, and PYD domains-containing Protein 3; APD, action potential duration; IKur, ultra-rapid delayed rectifier potassium current; IL, interleukin; SR, sarcoplasmic reticulum; CaMKII, calcium/calmodulin-dependent protein kinase II; PVs, pulmonary veins; MD1, Myeloid differentiation protein 1; TNF-α, tumor necrosis factor-α; TLR4, Toll-like receptor-4; SGK1, serum glucocorticoid kinase 1; PACs, premature atrial complexes; INa, voltage-gated sodium channel; ICa,L, L-type voltage-gated calcium channel; F2-IsoPs, F2-isoprostanes; NOX2, NADPH oxidase 2; PKC, protein kinase C; IKr, rapid delayed rectifier potassium current; IKs, slow delayed rectifier potassium current; IK1, inwardly rectifying potassium current; SAN, sinoatrial node; Ito, transient outward potassium current.

7. Perspective on Future Areas of Research

While the AF burden continues to rise, there are currently limited tools available for the diagnostics and treatment of AF. Although the gold standard of AF diagnosis is an electrocardiogram (ECG), paroxysmal AF may be missed in a routine 10-s 12-lead ECG or even a continuous heart monitor with a maximal duration of approximately two weeks. In patients with high suspicion for AF, for example, those with cryptogenic stroke, one may choose to obtain a longer-term recording (up to 5 years) using an implantable loop recorder, which involves a semi-invasive procedure and thus is without risk. Alternatively, biomarkers obtained non-invasively from serum or other secretions, such as saliva or urine, can prove to be an invaluable tool to predict incident AF and/or prognosticate AF progression. There has been a tremendous effort to identify various biomarkers specific to AF, including serum microRNAs, other non-coding RNAs [112,113], proteins/hormones [114,115], and metabolites [116,117]. However, the accuracy of these biomarkers is not yet sufficient for day-to-day clinical usage.
In terms of therapeutics, two current options are antiarrhythmics and AF ablation. Though non-invasive, utilization of antiarrhythmics is not without risk, as most of these medications are also pro-arrhythmic. Additionally, a widely utilized potent antiarrhythmic amiodarone has both short- and long-term pulmonary, thyroid, and liver toxicity. AF ablation involves intentionally creating scars to electrically isolate pulmonary veins, the most common site of AF triggers, from the left atrial tissue. Although more effective than antiarrhythmic medications in maintaining normal sinus rhythm, AF ablation is invasive, with complication rates up to 2.5% even in medical centers with experience in AF ablation [118]. Most importantly, although pulmonary vein isolation is very effective in AF treatment, it does not treat the root cause of AF, which is atrial myopathy, and AF can recur when atria become sicker over time.
Therefore, more effort should be focused on discovering novel diagnostic and therapeutic strategies for AF. A potentially appealing target is extracellular vesicles (EVs). EVs are lipid bilayer-bound particles released from cells and contain numerous cargos, including DNA, RNA, proteins, and metabolites. They can be found in various types of biological fluids, including serum, saliva, urine, and nasal secretion [119]. EVs have several advantages as a potential non-invasive diagnostic and therapeutic tool. In addition to the natural enrichment of target molecules inside EVs, these molecules are protected by a lipid bilayer and are more stable than free molecules in biological fluids. These features allow ease of sample handling and result reproducibility, and thus potentially facilitate translation from bench to bedside use. Multiple studies have identified an association between the content of circulating EVs and AF [120,121]. A study by Shaihov-Teper et al. analyzed EVs derived from the epicardial fat of individuals with (n = 32) and without AF (n = 30) [122]. EVs from patients with AF contained more proinflammatory and profibrotic cytokines and profibrotic microRNAs compared to those derived from the controls. More importantly, induced pluripotent stem-derived cardiomyocytes treated with epicardial fat-derived EVs from patients with AF had a short action potential duration and had more inducible sustained re-entry, both of which are markers of AF susceptibility. Additionally, as EVs have been shown to facilitate intra- and inter-organ communication [123,124], they could participate in communication between adipose tissue and atrial tissue and thus facilitate obesity-related AF. Moreover, there is an elevated serum level of adipocyte-derived EVs in obese individuals [125]. The cargo of these EVs has been shown to facilitate diseases associated with obesity. For example, hepatocytes treated with EVs isolated from VAT of obese individuals developed dysregulation of the transforming growth factor pathway, which is known to be associated with fatty liver disease [126]. Additionally, EVs derived from adipose tissue macrophages in obese mice contain microRNAs associated with signaling pathways involving insulin resistance [127].
In addition to their diagnostic potential, EVs may prove to be an invaluable therapeutic tool. EV-mimic nanovesicles are synthetic lipid bilayers containing small molecules, drugs, or nucleic acids. The properties of these nanovesicles, such as sizes, surface charges, and surface peptides, can be adjusted to target specific organs or tissues [128]. Therapeutic agents, such as microRNAs, can then be loaded into these synthetic nanovesicles. Therefore, these unique properties make EV-mimic nanovesicles an appealing tool for targeted AF treatment.
Lastly, it is important to consider AF as a heterogenous disease, a common phenotype resulting from multiple pathologies beyond valvular versus non-valvular AF. This may also explain why current standard-of-care treatments for all types of AF fail to sustain sinus rhythm. An analogy can be drawn between AF and heart failure with preserved ejection fraction (HFpEF), which accounts for 50% of overall heart failure cases. Despite great success in clinical trials for the treatment of heart failure with reduced ejection fraction (HFrEF), the results of clinical trials for the treatment of HFpEF fail to reach a similar degree of positive outcomes. As HFpEF is composed of diverse phenotypes, future effort are directed toward phenotype-specific treatment design [129]. This framework should be adapted to future AF research such that diagnostic tools and treatments are designed to target the unique pathophysiology underlying the sub-phenotypes of AF in each patient.

8. Conclusions

The global burden of AF is rising with the aging of the population and its associated comorbidities. Obesity-related AF significantly contributes to this burden, as the worldwide prevalence and incidence of obesity in adults and children are also increasing at an alarming rate. Therefore, it is critical to understand the mechanisms underlying obesity-related AF. Not only will this knowledge allow discoveries of novel diagnostic/prognostic tools and more effective treatments, but it will also lead to the development of new strategies for primary and secondary AF prevention. Lastly, it is critical to emphasize the importance of primordial prevention, which includes multidisciplinary effort [130] to decrease the worldwide obesity burden.

Author Contributions

W.B.L. performed the literature review and wrote the manuscript. S.D. provided feedback and guidance on the content of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

S.D. is a founding member and has equity in Thryv Therapeutics and Switch Therapeutics. He also consults for Thryv Therapeutics. He has received research funding from Abbott and Bristol Myers Squib. None of these are relevant to this manuscript.

References

  1. Bordignon, S.; Chiara Corti, M.; Bilato, C. Atrial Fibrillation Associated with Heart Failure, Stroke and Mortality. J. Atr. Fibrillation 2012, 5, 467. [Google Scholar] [CrossRef]
  2. Benjamin, E.J.; Muntner, P.; Alonso, A.; Bittencourt, M.S.; Callaway, C.W.; Carson, A.P.; Chamberlain, A.M.; Chang, A.R.; Cheng, S.; Das, S.R.; et al. Heart Disease and Stroke Statistics-2019 Update: A Report from the American Heart Association. Circulation 2019, 139, e56–e528. [Google Scholar] [CrossRef]
  3. Aliot, E.; Botto, G.L.; Crijns, H.J.; Kirchhof, P. Quality of life in patients with atrial fibrillation: How to assess it and how to improve it. Europace 2014, 16, 787–796. [Google Scholar] [CrossRef]
  4. Stewart, S.; Murphy, N.F.; Walker, A.; McGuire, A.; McMurray, J.J. Cost of an emerging epidemic: An economic analysis of atrial fibrillation in the UK. Heart 2004, 90, 286–292. [Google Scholar] [CrossRef] [Green Version]
  5. Kim, M.H.; Johnston, S.S.; Chu, B.C.; Dalal, M.R.; Schulman, K.L. Estimation of total incremental health care costs in patients with atrial fibrillation in the United States. Circ. Cardiovasc. Qual. Outcomes 2011, 4, 313–320. [Google Scholar] [CrossRef] [Green Version]
  6. Lee, W.C.; Lamas, G.A.; Balu, S.; Spalding, J.; Wang, Q.; Pashos, C.L. Direct treatment cost of atrial fibrillation in the elderly American population: A Medicare perspective. J. Med. Econ. 2008, 11, 281–298. [Google Scholar] [CrossRef]
  7. Ball, J.; Carrington, M.J.; McMurray, J.J.; Stewart, S. Atrial fibrillation: Profile and burden of an evolving epidemic in the 21st century. Int. J. Cardiol. 2013, 167, 1807–1824. [Google Scholar] [CrossRef]
  8. Mou, L.; Norby, F.L.; Chen, L.Y.; O’Neal, W.T.; Lewis, T.T.; Loehr, L.R.; Soliman, E.Z.; Alonso, A. Lifetime Risk of Atrial Fibrillation by Race and Socioeconomic Status: ARIC Study (Atherosclerosis Risk in Communities). Circ. Arrhythmia Electrophysiol. 2018, 11, e006350. [Google Scholar] [CrossRef]
  9. Chao, T.F.; Liu, C.J.; Tuan, T.C.; Chen, T.J.; Hsieh, M.H.; Lip, G.Y.H.; Chen, S.A. Lifetime Risks, Projected Numbers, and Adverse Outcomes in Asian Patients with Atrial Fibrillation: A Report from the Taiwan Nationwide AF Cohort Study. Chest 2018, 153, 453–466. [Google Scholar] [CrossRef] [Green Version]
  10. Colilla, S.; Crow, A.; Petkun, W.; Singer, D.E.; Simon, T.; Liu, X. Estimates of current and future incidence and prevalence of atrial fibrillation in the U.S. adult population. Am. J. Cardiol. 2013, 112, 1142–1147. [Google Scholar] [CrossRef]
  11. Dilaveris, P.E.; Kennedy, H.L. Silent atrial fibrillation: Epidemiology, diagnosis, and clinical impact. Clin. Cardiol. 2017, 40, 413–418. [Google Scholar] [CrossRef]
  12. Wang, Y.C.; Xu, X.; Hajra, A.; Apple, S.; Kharawala, A.; Duarte, G.; Liaqat, W.; Fu, Y.; Li, W.; Chen, Y.; et al. Current Advancement in Diagnosing Atrial Fibrillation by Utilizing Wearable Devices and Artificial Intelligence: A Review Study. Diagnostics 2022, 12, 689. [Google Scholar] [CrossRef]
  13. Amar, D.; Zhang, H.; Leung, D.H.; Roistacher, N.; Kadish, A.H. Older age is the strongest predictor of postoperative atrial fibrillation. Anesthesiology 2002, 96, 352–356. [Google Scholar] [CrossRef]
  14. Riordan, M.; Opaskar, A.; Yoruk, A.; Younis, A.; Ali, A.; McNitt, S.; Sahin, B.; Rosero, S.; Goldenberg, I.; Aktas, M.K. Predictors of Atrial Fibrillation during Long-Term Implantable Cardiac Monitoring Following Cryptogenic Stroke. J. Am. Heart Assoc. 2020, 9, e016040. [Google Scholar] [CrossRef]
  15. Lopez-Jimenez, F.; Almahmeed, W.; Bays, H.; Cuevas, A.; Di Angelantonio, E.; le Roux, C.W.; Sattar, N.; Sun, M.C.; Wittert, G.; Pinto, F.J.; et al. Obesity and cardiovascular disease: Mechanistic insights and management strategies. A joint position paper by the World Heart Federation and World Obesity Federation. Eur. J. Prev. Cardiol. 2022, 29, 2218–2237. [Google Scholar] [CrossRef]
  16. Misra, A.; Wasir, J.S.; Vikram, N.K. Waist circumference criteria for the diagnosis of abdominal obesity are not applicable uniformly to all populations and ethnic groups. Nutrition 2005, 21, 969–976. [Google Scholar] [CrossRef]
  17. Ross, R.; Neeland, I.J.; Yamashita, S.; Shai, I.; Seidell, J.; Magni, P.; Santos, R.D.; Arsenault, B.; Cuevas, A.; Hu, F.B.; et al. Waist circumference as a vital sign in clinical practice: A Consensus Statement from the IAS and ICCR Working Group on Visceral Obesity. Nat. Rev. Endocrinol. 2020, 16, 177–189. [Google Scholar] [CrossRef] [Green Version]
  18. Sommer, I.; Teufer, B.; Szelag, M.; Nussbaumer-Streit, B.; Titscher, V.; Klerings, I.; Gartlehner, G. The performance of anthropometric tools to determine obesity: A systematic review and meta-analysis. Sci. Rep. 2020, 10, 12699. [Google Scholar] [CrossRef]
  19. Powell-Wiley, T.M.; Poirier, P.; Burke, L.E.; Despres, J.P.; Gordon-Larsen, P.; Lavie, C.J.; Lear, S.A.; Ndumele, C.E.; Neeland, I.J.; Sanders, P.; et al. Obesity and Cardiovascular Disease: A Scientific Statement from the American Heart Association. Circulation 2021, 143, e984–e1010. [Google Scholar] [CrossRef]
  20. Bray, G.A.; Kim, K.K.; Wilding, J.P.H.; World Obesity, F. Obesity: A chronic relapsing progressive disease process. A position statement of the World Obesity Federation. Obes. Rev. 2017, 18, 715–723. [Google Scholar] [CrossRef] [Green Version]
  21. Prospective Studies, C.; Whitlock, G.; Lewington, S.; Sherliker, P.; Clarke, R.; Emberson, J.; Halsey, J.; Qizilbash, N.; Collins, R.; Peto, R. Body-mass index and cause-specific mortality in 900,000 adults: Collaborative analyses of 57 prospective studies. Lancet 2009, 373, 1083–1096. [Google Scholar] [CrossRef] [Green Version]
  22. Kelly, T.; Yang, W.; Chen, C.S.; Reynolds, K.; He, J. Global burden of obesity in 2005 and projections to 2030. Int. J. Obes. 2008, 32, 1431–1437. [Google Scholar] [CrossRef] [Green Version]
  23. Di Cesare, M.; Soric, M.; Bovet, P.; Miranda, J.J.; Bhutta, Z.; Stevens, G.A.; Laxmaiah, A.; Kengne, A.P.; Bentham, J. The epidemiological burden of obesity in childhood: A worldwide epidemic requiring urgent action. BMC Med. 2019, 17, 212. [Google Scholar] [CrossRef] [Green Version]
  24. Djekic, D.; Lindgren, M.; Aberg, N.D.; Aberg, M.; Fengsrud, E.; Poci, D.; Adiels, M.; Rosengren, A. Body Mass Index in Adolescence and Long-Term Risk of Early Incident Atrial Fibrillation and Subsequent Mortality, Heart Failure, and Ischemic Stroke. J. Am. Heart Assoc. 2022, 11, e025984. [Google Scholar] [CrossRef]
  25. Tedrow, U.B.; Conen, D.; Ridker, P.M.; Cook, N.R.; Koplan, B.A.; Manson, J.E.; Buring, J.E.; Albert, C.M. The long- and short-term impact of elevated body mass index on the risk of new atrial fibrillation the WHS (women’s health study). J. Am. Coll. Cardiol. 2010, 55, 2319–2327. [Google Scholar] [CrossRef] [Green Version]
  26. Wang, T.J.; Parise, H.; Levy, D.; D’Agostino, R.B., Sr.; Wolf, P.A.; Vasan, R.S.; Benjamin, E.J. Obesity and the risk of new-onset atrial fibrillation. JAMA 2004, 292, 2471–2477. [Google Scholar] [CrossRef] [Green Version]
  27. Wong, C.X.; Sullivan, T.; Sun, M.T.; Mahajan, R.; Pathak, R.K.; Middeldorp, M.; Twomey, D.; Ganesan, A.N.; Rangnekar, G.; Roberts-Thomson, K.C.; et al. Obesity and the Risk of Incident, Post-Operative, and Post-Ablation Atrial Fibrillation: A Meta-Analysis of 626,603 Individuals in 51 Studies. JACC Clin. Electrophysiol. 2015, 1, 139–152. [Google Scholar] [CrossRef]
  28. Olivia, C.; Hastie, C.; Farshid, A. Adherence to guidelines regarding anticoagulation and risk factors for progression of atrial fibrillation in a nurse-led clinic. Intern. Med. J. 2021, 51, 1136–1142. [Google Scholar] [CrossRef]
  29. Tsang, T.S.; Barnes, M.E.; Miyasaka, Y.; Cha, S.S.; Bailey, K.R.; Verzosa, G.C.; Seward, J.B.; Gersh, B.J. Obesity as a risk factor for the progression of paroxysmal to permanent atrial fibrillation: A longitudinal cohort study of 21 years. Eur. Heart J. 2008, 29, 2227–2233. [Google Scholar] [CrossRef]
  30. Chilukuri, K.; Dalal, D.; Gadrey, S.; Marine, J.E.; Macpherson, E.; Henrikson, C.A.; Cheng, A.; Nazarian, S.; Sinha, S.; Spragg, D.; et al. A prospective study evaluating the role of obesity and obstructive sleep apnea for outcomes after catheter ablation of atrial fibrillation. J. Cardiovasc. Electrophysiol. 2010, 21, 521–525. [Google Scholar] [CrossRef]
  31. Guglin, M.; Maradia, K.; Chen, R.; Curtis, A.B. Relation of obesity to recurrence rate and burden of atrial fibrillation. Am. J. Cardiol. 2011, 107, 579–582. [Google Scholar] [CrossRef] [PubMed]
  32. Chatterjee, N.A.; Giulianini, F.; Geelhoed, B.; Lunetta, K.L.; Misialek, J.R.; Niemeijer, M.N.; Rienstra, M.; Rose, L.M.; Smith, A.V.; Arking, D.E.; et al. Genetic Obesity and the Risk of Atrial Fibrillation: Causal Estimates from Mendelian Randomization. Circulation 2017, 135, 741–754. [Google Scholar] [CrossRef] [Green Version]
  33. Larsson, S.C.; Back, M.; Rees, J.M.B.; Mason, A.M.; Burgess, S. Body mass index and body composition in relation to 14 cardiovascular conditions in UK Biobank: A Mendelian randomization study. Eur. Heart J. 2020, 41, 221–226. [Google Scholar] [CrossRef] [Green Version]
  34. Chen, W.; Yao, D.; Yan, H.; Wang, M.; Pan, Y. Genetically predicted childhood obesity and adult atrial fibrillation: A mendelian randomization study. Nutr. Metab. Cardiovasc. Dis. 2022, 32, 1019–1026. [Google Scholar] [CrossRef]
  35. Zhou, Y.; Zha, L.; Pan, S. The Risk of Atrial Fibrillation Increases with Earlier Onset of Obesity: A Mendelian Randomization Study. Int. J. Med. Sci. 2022, 19, 1388–1398. [Google Scholar] [CrossRef]
  36. Nattel, S.; Shiroshita-Takeshita, A.; Brundel, B.J.; Rivard, L. Mechanisms of atrial fibrillation: Lessons from animal models. Prog. Cardiovasc. Dis. 2005, 48, 9–28. [Google Scholar] [CrossRef]
  37. Bosch, R.F.; Nattel, S. Cellular electrophysiology of atrial fibrillation. Cardiovasc. Res. 2002, 54, 259–269. [Google Scholar] [CrossRef]
  38. Sanchez-Quintana, D.; Lopez-Minguez, J.R.; Pizarro, G.; Murillo, M.; Cabrera, J.A. Triggers and anatomical substrates in the genesis and perpetuation of atrial fibrillation. Curr. Cardiol. Rev. 2012, 8, 310–326. [Google Scholar] [CrossRef] [Green Version]
  39. Nattel, S.; Heijman, J.; Zhou, L.; Dobrev, D. Molecular Basis of Atrial Fibrillation Pathophysiology and Therapy: A Translational Perspective. Circ. Res. 2020, 127, 51–72. [Google Scholar] [CrossRef]
  40. Comtois, P.; Kneller, J.; Nattel, S. Of circles and spirals: Bridging the gap between the leading circle and spiral wave concepts of cardiac reentry. Europace 2005, 7 (Suppl. S2), 10–20. [Google Scholar] [CrossRef]
  41. Pandit, S.V.; Jalife, J. Rotors and the dynamics of cardiac fibrillation. Circ. Res. 2013, 112, 849–862. [Google Scholar] [CrossRef] [PubMed]
  42. Chait, A.; den Hartigh, L.J. Adipose Tissue Distribution, Inflammation and Its Metabolic Consequences, Including Diabetes and Cardiovascular Disease. Front. Cardiovasc. Med. 2020, 7, 22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Ibrahim, M.M. Subcutaneous and visceral adipose tissue: Structural and functional differences. Obes. Rev. 2010, 11, 11–18. [Google Scholar] [CrossRef]
  44. Arner, P. Catecholamine-induced lipolysis in obesity. Int. J. Obes. Relat. Metab. Disord. 1999, 23 (Suppl. S1), 10–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Mulya, A.; Kirwan, J.P. Brown and Beige Adipose Tissue: Therapy for Obesity and Its Comorbidities? Endocrinol. Metab. Clin. N. Am. 2016, 45, 605–621. [Google Scholar] [CrossRef] [Green Version]
  46. Sun, K.; Kusminski, C.M.; Scherer, P.E. Adipose tissue remodeling and obesity. J. Clin. Investig. 2011, 121, 2094–2101. [Google Scholar] [CrossRef] [Green Version]
  47. Xu, H.; Barnes, G.T.; Yang, Q.; Tan, G.; Yang, D.; Chou, C.J.; Sole, J.; Nichols, A.; Ross, J.S.; Tartaglia, L.A.; et al. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J. Clin. Investig. 2003, 112, 1821–1830. [Google Scholar] [CrossRef]
  48. Weisberg, S.P.; McCann, D.; Desai, M.; Rosenbaum, M.; Leibel, R.L.; Ferrante, A.W., Jr. Obesity is associated with macrophage accumulation in adipose tissue. J. Clin. Investig. 2003, 112, 1796–1808. [Google Scholar] [CrossRef]
  49. Ruiz-Ojeda, F.J.; Mendez-Gutierrez, A.; Aguilera, C.M.; Plaza-Diaz, J. Extracellular Matrix Remodeling of Adipose Tissue in Obesity and Metabolic Diseases. Int. J. Mol. Sci. 2019, 2, 4888. [Google Scholar] [CrossRef] [Green Version]
  50. Poirier, P.; Giles, T.D.; Bray, G.A.; Hong, Y.; Stern, J.S.; Pi-Sunyer, F.X.; Eckel, R.H. Obesity and cardiovascular disease: Pathophysiology, evaluation, and effect of weight loss. Arterioscler. Thromb. Vasc. Biol. 2006, 26, 968–976. [Google Scholar] [CrossRef]
  51. Chen, Y.C.; Voskoboinik, A.; Gerche, A.; Marwick, T.H.; McMullen, J.R. Prevention of Pathological Atrial Remodeling and Atrial Fibrillation: JACC State-of-the-Art Review. J. Am. Coll. Cardiol. 2021, 77, 2846–2864. [Google Scholar] [CrossRef] [PubMed]
  52. Garvey, J.F.; Pengo, M.F.; Drakatos, P.; Kent, B.D. Epidemiological aspects of obstructive sleep apnea. J. Thorac. Dis. 2015, 7, 920–929. [Google Scholar] [CrossRef] [PubMed]
  53. Gami, A.S.; Pressman, G.; Caples, S.M.; Kanagala, R.; Gard, J.J.; Davison, D.E.; Malouf, J.F.; Ammash, N.M.; Friedman, P.A.; Somers, V.K. Association of atrial fibrillation and obstructive sleep apnea. Circulation 2004, 110, 364–367. [Google Scholar] [CrossRef] [PubMed]
  54. Latina, J.M.; Estes, N.A., 3rd; Garlitski, A.C. The Relationship between Obstructive Sleep Apnea and Atrial Fibrillation: A Complex Interplay. Pulm. Med. 2013, 2013, 621736. [Google Scholar] [CrossRef] [PubMed]
  55. Tavares, L.; Lador, A.; Valderrabano, M. Sleep Apnea and Atrial Fibrillation: Role of the Cardiac Autonomic Nervous System. Methodist DeBakey Cardiovasc. J. 2021, 17, 49–52. [Google Scholar] [CrossRef]
  56. Agbaedeng, T.A.; Zacharia, A.L.; Iroga, P.E.; Rathnasekara, V.M.; Munawar, D.A.; Bursill, C.; Noubiap, J.J. Associations between adipokines and atrial fibrillation: A systematic review and meta-analysis. Nutr. Metab. Cardiovasc. Dis. 2022, 32, 853–862. [Google Scholar] [CrossRef] [PubMed]
  57. Villarreal-Molina, M.T.; Antuna-Puente, B. Adiponectin: Anti-inflammatory and cardioprotective effects. Biochimie 2012, 94, 2143–2149. [Google Scholar] [CrossRef]
  58. Shimabukuro, M.; Hirata, Y.; Tabata, M.; Dagvasumberel, M.; Sato, H.; Kurobe, H.; Fukuda, D.; Soeki, T.; Kitagawa, T.; Takanashi, S.; et al. Epicardial adipose tissue volume and adipocytokine imbalance are strongly linked to human coronary atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2013, 33, 1077–1084. [Google Scholar] [CrossRef] [Green Version]
  59. Zhu, T.; Wang, Z.; Wang, S.; Hu, W.; Chen, H.; Xie, J.; Wang, M.; Ma, K.; Jiang, H. Association between Serum Adiponectin and Atrial Fibrillation: A Case-Control Study Stratified by Age and Gender. Cardiol. Res. Pract. 2021, 2021, 6633948. [Google Scholar] [CrossRef]
  60. Peller, M.; Kaplon-Cieslicka, A.; Rosiak, M.; Tyminska, A.; Ozieranski, K.; Eyileten, C.; Kondracka, A.; Mirowska-Guzel, D.; Opolski, G.; Postula, M.; et al. Are adipokines associated with atrial fibrillation in type 2 diabetes? Endokrynol. Pol. 2020, 71, 34–41. [Google Scholar] [CrossRef] [Green Version]
  61. Anaszewicz, M.; Wawrzenczyk, A.; Czerniak, B.; Banas, W.; Socha, E.; Lis, K.; Zbikowska-Gotz, M.; Bartuzi, Z.; Budzynski, J. Leptin, adiponectin, tumor necrosis factor alpha, and irisin concentrations as factors linking obesity with the risk of atrial fibrillation among inpatients with cardiovascular diseases. Kardiol. Pol. 2019, 77, 1055–1061. [Google Scholar] [CrossRef] [Green Version]
  62. Macheret, F.; Bartz, T.M.; Djousse, L.; Ix, J.H.; Mukamal, K.J.; Zieman, S.J.; Siscovick, D.S.; Tracy, R.P.; Heckbert, S.R.; Psaty, B.M.; et al. Higher circulating adiponectin levels are associated with increased risk of atrial fibrillation in older adults. Heart 2015, 101, 1368–1374. [Google Scholar] [CrossRef] [PubMed]
  63. Fukui, A.; Ikebe-Ebata, Y.; Kondo, H.; Saito, S.; Aoki, K.; Fukunaga, N.; Shinohara, T.; Masaki, T.; Teshima, Y.; Takahashi, N. Hyperleptinemia Exacerbates High-Fat Diet-Mediated Atrial Fibrosis and Fibrillation. J. Cardiovasc. Electrophysiol. 2017, 28, 702–710. [Google Scholar] [CrossRef]
  64. Klok, M.D.; Jakobsdottir, S.; Drent, M.L. The role of leptin and ghrelin in the regulation of food intake and body weight in humans: A review. Obes. Rev. 2007, 8, 21–34. [Google Scholar] [CrossRef] [PubMed]
  65. Lv, W.; Zhang, L.; Cheng, X.; Wang, H.; Qin, W.; Zhou, X.; Tang, B. Apelin Inhibits Angiotensin II-Induced Atrial Fibrosis and Atrial Fibrillation via TGF-beta1/Smad2/alpha-SMA Pathway. Front. Physiol. 2020, 11, 583570. [Google Scholar] [CrossRef]
  66. Xu, S.; Zhang, J.; Xu, Y.L.; Wu, H.B.; Xue, X.D.; Wang, H.S. Relationship between Angiotensin Converting Enzyme, Apelin, and New-Onset Atrial Fibrillation after Off-Pump Coronary Artery Bypass Grafting. BioMed Res. Int. 2017, 2017, 7951793. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Falcone, C.; Buzzi, M.P.; D’Angelo, A.; Schirinzi, S.; Falcone, R.; Rordorf, R.; Capettini, A.C.; Landolina, M.; Storti, C.; Pelissero, G. Apelin plasma levels predict arrhythmia recurrence in patients with persistent atrial fibrillation. Int. J. Immunopathol. Pharmacol. 2010, 23, 917–925. [Google Scholar] [CrossRef]
  68. Wang, Y.Z.; Fan, J.; Zhong, B.; Xu, Q. Apelin: A novel prognostic predictor for atrial fibrillation recurrence after pulmonary vein isolation. Medicine 2018, 97, e12580. [Google Scholar] [CrossRef] [PubMed]
  69. Zaki, M.; Kamal, S.; Ezzat, W.; Hassan, N.; Yousef, W.; Ryad, H.; Mohamed, R.; Youness, E.; Basha, W.; Elhosary, Y. Serum apelin levels and metabolic risk markers in obese women. J. Genet. Eng. Biotechnol. 2017, 15, 423–429. [Google Scholar] [CrossRef]
  70. Krist, J.; Wieder, K.; Kloting, N.; Oberbach, A.; Kralisch, S.; Wiesner, T.; Schon, M.R.; Gartner, D.; Dietrich, A.; Shang, E.; et al. Effects of weight loss and exercise on apelin serum concentrations and adipose tissue expression in human obesity. Obes. Facts 2013, 6, 57–69. [Google Scholar] [CrossRef]
  71. Lopaschuk, G.D.; Ussher, J.R.; Folmes, C.D.; Jaswal, J.S.; Stanley, W.C. Myocardial fatty acid metabolism in health and disease. Physiol. Rev. 2010, 90, 207–258. [Google Scholar] [CrossRef]
  72. Murray, A.J.; Panagia, M.; Hauton, D.; Gibbons, G.F.; Clarke, K. Plasma free fatty acids and peroxisome proliferator-activated receptor alpha in the control of myocardial uncoupling protein levels. Diabetes 2005, 54, 3496–3502. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Cole, M.A.; Murray, A.J.; Cochlin, L.E.; Heather, L.C.; McAleese, S.; Knight, N.S.; Sutton, E.; Jamil, A.A.; Parassol, N.; Clarke, K. A high fat diet increases mitochondrial fatty acid oxidation and uncoupling to decrease efficiency in rat heart. Basic Res. Cardiol. 2011, 106, 447–457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Zhang, Y.; Yang, S.; Fu, J.; Liu, A.; Liu, D.; Cao, S. Inhibition of endoplasmic reticulum stress prevents high-fat diet mediated atrial fibrosis and fibrillation. J. Cell. Mol. Med. 2020, 24, 13660–13668. [Google Scholar] [CrossRef] [PubMed]
  75. Smith, H.L.; Willius, F.A. Adiposity of the heart: A clinical study of one hundred and thirty six obese patients. Ann. Intern. Med. 1933, 52, 911–931. [Google Scholar] [CrossRef]
  76. Obokata, M.; Reddy, Y.N.V.; Pislaru, S.V.; Melenovsky, V.; Borlaug, B.A. Evidence Supporting the Existence of a Distinct Obese Phenotype of Heart Failure with Preserved Ejection Fraction. Circulation 2017, 136, 6–19. [Google Scholar] [CrossRef] [PubMed]
  77. Thanassoulis, G.; Massaro, J.M.; O’Donnell, C.J.; Hoffmann, U.; Levy, D.; Ellinor, P.T.; Wang, T.J.; Schnabel, R.B.; Vasan, R.S.; Fox, C.S.; et al. Pericardial fat is associated with prevalent atrial fibrillation: The Framingham Heart Study. Circ. Arrhythm. Electrophysiol. 2010, 3, 345–350. [Google Scholar] [CrossRef] [Green Version]
  78. Friedman, D.J.; Wang, N.; Meigs, J.B.; Hoffmann, U.; Massaro, J.M.; Fox, C.S.; Magnani, J.W. Pericardial fat is associated with atrial conduction: The Framingham Heart Study. J. Am. Heart Assoc. 2014, 3, e000477. [Google Scholar] [CrossRef] [Green Version]
  79. Nakamori, S.; Nezafat, M.; Ngo, L.H.; Manning, W.J.; Nezafat, R. Left Atrial Epicardial Fat Volume Is Associated with Atrial Fibrillation: A Prospective Cardiovascular Magnetic Resonance 3D Dixon Study. J. Am. Heart Assoc. 2018, 7, e008232. [Google Scholar] [CrossRef]
  80. Wong, C.X.; Sun, M.T.; Odutayo, A.; Emdin, C.A.; Mahajan, R.; Lau, D.H.; Pathak, R.K.; Wong, D.T.; Selvanayagam, J.B.; Sanders, P.; et al. Associations of Epicardial, Abdominal, and Overall Adiposity with Atrial Fibrillation. Circ. Arrhythm. Electrophysiol. 2016, 9, e004378. [Google Scholar] [CrossRef] [Green Version]
  81. Mahajan, R.; Nelson, A.; Pathak, R.K.; Middeldorp, M.E.; Wong, C.X.; Twomey, D.J.; Carbone, A.; Teo, K.; Agbaedeng, T.; Linz, D.; et al. Electroanatomical Remodeling of the Atria in Obesity: Impact of Adjacent Epicardial Fat. JACC Clin. Electrophysiol. 2018, 4, 1529–1540. [Google Scholar] [CrossRef]
  82. Wong, C.X.; Ganesan, A.N.; Selvanayagam, J.B. Epicardial fat and atrial fibrillation: Current evidence, potential mechanisms, clinical implications, and future directions. Eur. Heart J. 2017, 38, 1294–1302. [Google Scholar] [CrossRef] [Green Version]
  83. Haemers, P.; Hamdi, H.; Guedj, K.; Suffee, N.; Farahmand, P.; Popovic, N.; Claus, P.; LePrince, P.; Nicoletti, A.; Jalife, J.; 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. [Google Scholar] [CrossRef] [Green Version]
  84. Platonov, P.G.; Mitrofanova, L.B.; Orshanskaya, V.; Ho, S.Y. Structural abnormalities in atrial walls are associated with presence and persistency of atrial fibrillation but not with age. J. Am. Coll. Cardiol. 2011, 58, 2225–2232. [Google Scholar] [CrossRef] [Green Version]
  85. Tereshchenko, L.G.; Rizzi, P.; Mewton, N.; Volpe, G.J.; Murthy, S.; Strauss, D.G.; Liu, C.Y.; Marchlinski, F.E.; Spooner, P.; Berger, R.D.; et al. Infiltrated atrial fat characterizes underlying atrial fibrillation substrate in patients at risk as defined by the ARIC atrial fibrillation risk score. Int. J. Cardiol. 2014, 172, 196–201. [Google Scholar] [CrossRef] [Green Version]
  86. Nalliah, C.J.; Bell, J.R.; Raaijmakers, A.J.A.; Waddell, H.M.; Wells, S.P.; Bernasochi, G.B.; Montgomery, M.K.; Binny, S.; Watts, T.; Joshi, S.B.; et al. Epicardial Adipose Tissue Accumulation Confers Atrial Conduction Abnormality. J. Am. Coll. Cardiol. 2020, 76, 1197–1211. [Google Scholar] [CrossRef] [PubMed]
  87. Shan, J.; Xie, W.; Betzenhauser, M.; Reiken, S.; Chen, B.X.; Wronska, A.; Marks, A.R. Calcium leak through ryanodine receptors leads to atrial fibrillation in 3 mouse models of catecholaminergic polymorphic ventricular tachycardia. Circ. Res. 2012, 111, 708–717. [Google Scholar] [CrossRef]
  88. Karam, B.S.; Chavez-Moreno, A.; Koh, W.; Akar, J.G.; Akar, F.G. Oxidative stress and inflammation as central mediators of atrial fibrillation in obesity and diabetes. Cardiovasc. Diabetol. 2017, 16, 120. [Google Scholar] [CrossRef] [PubMed]
  89. Xie, W.; Santulli, G.; Reiken, S.R.; Yuan, Q.; Osborne, B.W.; Chen, B.X.; Marks, A.R. Mitochondrial oxidative stress promotes atrial fibrillation. Sci. Rep. 2015, 5, 11427. [Google Scholar] [CrossRef] [Green Version]
  90. Fang, G.; Cao, W.; Chen, L.; Song, S.; Li, Y.; Yuan, J.; Fei, Y.; Ge, Z.; Chen, Y.; Zhou, L.; et al. Cadherin-11 deficiency mitigates high-fat diet-induced inflammatory atrial remodeling and vulnerability to atrial fibrillation. J. Cell. Physiol. 2021, 236, 5725–5741. [Google Scholar] [CrossRef] [PubMed]
  91. Scott, L., Jr.; Fender, A.C.; Saljic, A.; Li, L.; Chen, X.; Wang, X.; Linz, D.; Lang, J.; Hohl, M.; Twomey, D.; et al. NLRP3 inflammasome is a key driver of obesity-induced atrial arrhythmias. Cardiovasc. Res. 2021, 117, 1746–1759. [Google Scholar] [CrossRef] [PubMed]
  92. Otsuka, N.; Okumura, Y.; Arai, M.; Kurokawa, S.; Nagashima, K.; Watanabe, R.; Wakamatsu, Y.; Yagyu, S.; Ohkubo, K.; Nakai, T.; et al. Effect of obesity and epicardial fat/fatty infiltration on electrical and structural remodeling associated with atrial fibrillation in a novel canine model of obesity and atrial fibrillation: A comparative study. J. Cardiovasc. Electrophysiol. 2021, 32, 889–899. [Google Scholar] [CrossRef] [PubMed]
  93. Okumura, Y.; Watanabe, I.; Nagashima, K.; Sonoda, K.; Sasaki, N.; Kogawa, R.; Takahashi, K.; Iso, K.; Ohkubo, K.; Nakai, T.; et al. Effects of a high-fat diet on the electrical properties of porcine atria. J. Arrhythm. 2015, 31, 352–358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Abed, H.S.; Samuel, C.S.; Lau, D.H.; Kelly, D.J.; Royce, S.G.; Alasady, M.; Mahajan, R.; Kuklik, P.; Zhang, Y.; Brooks, A.G.; et al. Obesity results in progressive atrial structural and electrical remodeling: Implications for atrial fibrillation. Heart Rhythm. 2013, 10, 90–100. [Google Scholar] [CrossRef] [PubMed]
  95. Mahajan, R.; Lau, D.H.; Brooks, A.G.; Shipp, N.J.; Manavis, J.; Wood, J.P.; Finnie, J.W.; Samuel, C.S.; Royce, S.G.; Twomey, D.J.; et al. Electrophysiological, Electroanatomical, and Structural Remodeling of the Atria as Consequences of Sustained Obesity. J Am. Coll. Cardiol. 2015, 66, 1–11. [Google Scholar] [CrossRef]
  96. Hohl, M.; Lau, D.H.; Muller, A.; Elliott, A.D.; Linz, B.; Mahajan, R.; Hendriks, J.M.L.; Bohm, M.; Schotten, U.; Sanders, P.; et al. Concomitant Obesity and Metabolic Syndrome Add to the Atrial Arrhythmogenic Phenotype in Male Hypertensive Rats. J. Am. Heart Assoc. 2017, 6, e006717. [Google Scholar] [CrossRef]
  97. Row, S.; Liu, Y.; Alimperti, S.; Agarwal, S.K.; Andreadis, S.T. Cadherin-11 is a novel regulator of extracellular matrix synthesis and tissue mechanics. J. Cell Sci. 2016, 129, 2950–2961. [Google Scholar] [CrossRef] [Green Version]
  98. Chang, S.K.; Kohlgruber, A.C.; Mizoguchi, F.; Michelet, X.; Wolf, B.J.; Wei, K.; Lee, P.Y.; Lynch, L.; Duquette, D.; Ceperuelo-Mallafre, V.; et al. Stromal cell cadherin-11 regulates adipose tissue inflammation and diabetes. J. Clin. Investig. 2017, 127, 3300–3312. [Google Scholar] [CrossRef] [Green Version]
  99. Shuai, W.; Kong, B.; Fu, H.; Shen, C.; Jiang, X.; Huang, H. MD1 Deficiency Promotes Inflammatory Atrial Remodelling Induced by High-Fat Diets. Can. J. Cardiol. 2019, 35, 208–216. [Google Scholar] [CrossRef]
  100. Bapat, A.; Li, G.; Xiao, L.; Yeri, A.; Hulsmans, M.; Grune, J.; Yamazoe, M.; Schloss, M.J.; Iwamoto, Y.; Tedeschi, J.; et al. Genetic inhibition of serum glucocorticoid kinase 1 prevents obesity-related atrial fibrillation. JCI Insight 2022, 7, e160885. [Google Scholar] [CrossRef]
  101. Schram-Serban, C.; Heida, A.; Roos-Serote, M.C.; Knops, P.; Kik, C.; Brundel, B.; Bogers, A.; de Groot, N.M.S. Heterogeneity in Conduction Underlies Obesity-Related Atrial Fibrillation Vulnerability. Circ. Arrhythm. Electrophysiol. 2020, 13, e008161. [Google Scholar] [CrossRef] [PubMed]
  102. McCauley, M.D.; Hong, L.; Sridhar, A.; Menon, A.; Perike, S.; Zhang, M.; da Silva, I.B.; Yan, J.; Bonini, M.G.; Ai, X.; et al. Ion Channel and Structural Remodeling in Obesity-Mediated Atrial Fibrillation. Circ. Arrhythm. Electrophysiol. 2020, 13, e008296. [Google Scholar] [CrossRef] [PubMed]
  103. Munger, T.M.; Dong, Y.X.; Masaki, M.; Oh, J.K.; Mankad, S.V.; Borlaug, B.A.; Asirvatham, S.J.; Shen, W.K.; Lee, H.C.; Bielinski, S.J.; et al. Electrophysiological and hemodynamic characteristics associated with obesity in patients with atrial fibrillation. J. Am. Coll. Cardiol. 2012, 60, 851–860. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Martinez-Mateu, L.; Saiz, J.; Aromolaran, A.S. Differential Modulation of I(K) and I(Ca,L) Channels in High-Fat Diet-Induced Obese Guinea Pig Atria. Front. Physiol. 2019, 10, 1212. [Google Scholar] [CrossRef] [PubMed]
  105. Zhang, F.; Hartnett, S.; Sample, A.; Schnack, S.; Li, Y. High fat diet induced alterations of atrial electrical activities in mice. Am. J. Cardiovasc. Dis. 2016, 6, 1–9. [Google Scholar] [PubMed]
  106. Pathak, R.K.; Middeldorp, M.E.; Meredith, M.; Mehta, A.B.; Mahajan, R.; Wong, C.X.; Twomey, D.; Elliott, A.D.; Kalman, J.M.; Abhayaratna, W.P.; 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, 2159–2169. [Google Scholar] [CrossRef]
  107. Abed, H.S.; Wittert, G.A.; Leong, D.P.; Shirazi, M.G.; Bahrami, B.; Middeldorp, M.E.; Lorimer, M.F.; Lau, D.H.; Antic, N.A.; Brooks, A.G.; 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, 2050–2060. [Google Scholar] [CrossRef]
  108. Middeldorp, M.E.; Pathak, R.K.; Meredith, M.; Mehta, A.B.; Elliott, A.D.; Mahajan, R.; Twomey, D.; Gallagher, C.; Hendriks, J.M.L.; Linz, D.; et al. PREVEntion and regReSsive Effect of weight-loss and risk factor modification on Atrial Fibrillation: The REVERSE-AF study. Europace 2018, 20, 1929–1935. [Google Scholar] [CrossRef]
  109. Donnellan, E.; Wazni, O.M.; Kanj, M.; Baranowski, B.; Cremer, P.; Harb, S.; McCarthy, C.P.; McEvoy, J.W.; Elshazly, M.B.; Aagaard, P.; et al. Association between pre-ablation bariatric surgery and atrial fibrillation recurrence in morbidly obese patients undergoing atrial fibrillation ablation. Europace 2019, 21, 1476–1483. [Google Scholar] [CrossRef] [PubMed]
  110. Zhao, H.; Li, X.; Yu, P.; Liu, M.; Ma, J.; Wang, J.; Zhu, W.; Liu, X. Association between weight loss and outcomes in patients undergoing atrial fibrillation ablation: A systematic review and dose-response meta-analysis. Nutr. Metab. 2023, 20, 5. [Google Scholar] [CrossRef]
  111. Mahajan, R.; Lau, D.H.; Brooks, A.G.; Shipp, N.J.; Wood, J.P.M.; Manavis, J.; Samuel, C.S.; Patel, K.P.; Finnie, J.W.; Alasady, M.; et al. Atrial Fibrillation and Obesity: Reverse Remodeling of Atrial Substrate with Weight Reduction. JACC Clin. Electrophysiol. 2021, 7, 630–641. [Google Scholar] [CrossRef] [PubMed]
  112. Menezes Junior, A.D.S.; Ferreira, L.C.; Barbosa, L.J.V.; Silva, D.M.E.; Saddi, V.A.; Silva, A. Circulating MicroRNAs as Specific Biomarkers in Atrial Fibrillation: A Meta-Analysis. Noncoding RNA 2023, 9, 13. [Google Scholar] [CrossRef] [PubMed]
  113. Zhang, L.; Wang, X.; Huang, C. A narrative review of non-coding RNAs in atrial fibrillation: Potential therapeutic targets and molecular mechanisms. Ann. Transl. Med. 2021, 9, 1486. [Google Scholar] [CrossRef] [PubMed]
  114. Turagam, M.K.; Mirza, M.; Werner, P.H.; Sra, J.; Kress, D.C.; Tajik, A.J.; Jahangir, A. Circulating Biomarkers Predictive of Postoperative Atrial Fibrillation. Cardiol. Rev. 2016, 24, 76–87. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Boyalla, V.; Harling, L.; Snell, A.; Kralj-Hans, I.; Barradas-Pires, A.; Haldar, S.; Khan, H.R.; Cleland, J.G.F.; Athanasiou, T.; Harding, S.E.; et al. Biomarkers as predictors of recurrence of atrial fibrillation post ablation: An updated and expanded systematic review and meta-analysis. Clin. Res. Cardiol. 2022, 111, 680–691. [Google Scholar] [CrossRef] [PubMed]
  116. Lu, C.; Liu, C.; Mei, D.; Yu, M.; Bai, J.; Bao, X.; Wang, M.; Fu, K.; Yi, X.; Ge, W.; et al. Comprehensive metabolomic characterization of atrial fibrillation. Front. Cardiovasc. Med. 2022, 9, 911845. [Google Scholar] [CrossRef] [PubMed]
  117. Alonso, A.; Yu, B.; Sun, Y.V.; Chen, L.Y.; Loehr, L.R.; O’Neal, W.T.; Soliman, E.Z.; Boerwinkle, E. Serum Metabolomics and Incidence of Atrial Fibrillation (from the Atherosclerosis Risk in Communities Study). Am. J. Cardiol. 2019, 123, 1955–1961. [Google Scholar] [CrossRef]
  118. Hsu, J.C.; Darden, D.; Du, C.; Marine, J.E.; Nichols, S.; Marcus, G.M.; Natale, A.; Noseworthy, P.A.; Selzman, K.A.; Varosy, P.; et al. Initial Findings from the National Cardiovascular Data Registry of Atrial Fibrillation Ablation Procedures. J. Am. Coll. Cardiol. 2023, 81, 867–878. [Google Scholar] [CrossRef]
  119. Colombo, M.; Raposo, G.; Thery, C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu. Rev. Cell Dev. Biol. 2014, 30, 255–289. [Google Scholar] [CrossRef]
  120. Procyk, G.; Bilicki, D.; Balsam, P.; Lodzinski, P.; Grabowski, M.; Gasecka, A. Extracellular Vesicles in Atrial Fibrillation-State of the Art. Int. J. Mol. Sci. 2022, 23, 7591. [Google Scholar] [CrossRef]
  121. Huang, S.; Deng, Y.; Xu, J.; Liu, J.; Liu, L.; Fan, C. The Role of Exosomes and Their Cargos in the Mechanism, Diagnosis, and Treatment of Atrial Fibrillation. Front. Cardiovasc. Med. 2021, 8, 712828. [Google Scholar] [CrossRef] [PubMed]
  122. Shaihov-Teper, O.; Ram, E.; Ballan, N.; Brzezinski, R.Y.; Naftali-Shani, N.; Masoud, R.; Ziv, T.; Lewis, N.; Schary, Y.; Levin-Kotler, L.P.; et al. Extracellular Vesicles from Epicardial Fat Facilitate Atrial Fibrillation. Circulation 2021, 143, 2475–2493. [Google Scholar] [CrossRef] [PubMed]
  123. Thomou, T.; Mori, M.A.; Dreyfuss, J.M.; Konishi, M.; Sakaguchi, M.; Wolfrum, C.; Rao, T.N.; Winnay, J.N.; Garcia-Martin, R.; Grinspoon, S.K.; et al. Adipose-derived circulating miRNAs regulate gene expression in other tissues. Nature 2017, 542, 450–455. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Phu, T.A.; Ng, M.; Vu, N.K.; Bouchareychas, L.; Raffai, R.L. IL-4 polarized human macrophage exosomes control cardiometabolic inflammation and diabetes in obesity. Mol. Ther. 2022, 30, 2274–2297. [Google Scholar] [CrossRef]
  125. Pardo, F.; Villalobos-Labra, R.; Sobrevia, B.; Toledo, F.; Sobrevia, L. Extracellular vesicles in obesity and diabetes mellitus. Mol. Asp. Med. 2018, 60, 81–91. [Google Scholar] [CrossRef]
  126. Koeck, E.S.; Iordanskaia, T.; Sevilla, S.; Ferrante, S.C.; Hubal, M.J.; Freishtat, R.J.; Nadler, E.P. Adipocyte exosomes induce transforming growth factor beta pathway dysregulation in hepatocytes: A novel paradigm for obesity-related liver disease. J. Surg. Res. 2014, 192, 268–275. [Google Scholar] [CrossRef]
  127. Ying, W.; Riopel, M.; Bandyopadhyay, G.; Dong, Y.; Birmingham, A.; Seo, J.B.; Ofrecio, J.M.; Wollam, J.; Hernandez-Carretero, A.; Fu, W.; et al. Adipose Tissue Macrophage-Derived Exosomal miRNAs Can Modulate In Vivo and In Vitro Insulin Sensitivity. Cell 2017, 171, 372–384.e312. [Google Scholar] [CrossRef] [Green Version]
  128. Blanco, E.; Shen, H.; Ferrari, M. Principles of nanoparticle design for overcoming biological barriers to drug delivery. Nat. Biotechnol. 2015, 33, 941–951. [Google Scholar] [CrossRef]
  129. Shah, S.J.; Kitzman, D.W.; Borlaug, B.A.; van Heerebeek, L.; Zile, M.R.; Kass, D.A.; Paulus, W.J. Phenotype-Specific Treatment of Heart Failure with Preserved Ejection Fraction: A Multiorgan Roadmap. Circulation 2016, 134, 73–90. [Google Scholar] [CrossRef] [Green Version]
  130. Perone, F.; Pingitore, A.; Conte, E.; Halasz, G.; Ambrosetti, M.; Peruzzi, M.; Cavarretta, E. Obesity and Cardiovascular Risk: Systematic Intervention Is the Key for Prevention. Healthcare 2023, 11, 902. [Google Scholar] [CrossRef]
Figure 1. Schematic of potential mechanisms underlying adiposity-related atrial fibrillation. Obesity leads to both electrical and anatomical remodeling of atrial tissue. These changes lead to increased AF triggers and create vulnerable substrate that is more prone to perpetuation of AF.
Figure 1. Schematic of potential mechanisms underlying adiposity-related atrial fibrillation. Obesity leads to both electrical and anatomical remodeling of atrial tissue. These changes lead to increased AF triggers and create vulnerable substrate that is more prone to perpetuation of AF.
Jcdd 10 00323 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Limpitikul, W.B.; Das, S. Obesity-Related Atrial Fibrillation: Cardiac Manifestation of a Systemic Disease. J. Cardiovasc. Dev. Dis. 2023, 10, 323. https://doi.org/10.3390/jcdd10080323

AMA Style

Limpitikul WB, Das S. Obesity-Related Atrial Fibrillation: Cardiac Manifestation of a Systemic Disease. Journal of Cardiovascular Development and Disease. 2023; 10(8):323. https://doi.org/10.3390/jcdd10080323

Chicago/Turabian Style

Limpitikul, Worawan B., and Saumya Das. 2023. "Obesity-Related Atrial Fibrillation: Cardiac Manifestation of a Systemic Disease" Journal of Cardiovascular Development and Disease 10, no. 8: 323. https://doi.org/10.3390/jcdd10080323

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

Article Metrics

Back to TopTop