Abstract Atrial fibrillation (AF) is the most prevalent arrhythmia in clinical practice, and obesity serves as a significant risk factor for its development. The underlying mechanisms of obesity-related AF remain intricate and have yet to be fully elucidated. We have identified FPR2 as a potential hub gene involved in obesity-related AF through comprehensive analysis of four transcriptome datasets from AF patients and one transcriptome dataset from obese individuals, and its expression is up-regulated in both AF and obese individuals. Interestingly, ANXA1, the endogenous ligand of FPR2, was found to exhibit differential expression with AF and obesity. Specifically, it was observed to be down-regulated in AF patients but up-regulated in obese individuals. The susceptibility to AF in obese mice induced by high-fat diet (HFD) was increased following with the FPR2 blocker Boc-2.The administration of exogenous ANXA1 active peptide chain Ac2-26 can mitigate the susceptibility to AF in obese mice by attenuating atrial fibrosis, lipid deposition, oxidative stress injury, and myocardial cell apoptosis. However, this protective effect against AF susceptibility is reversed by AAV9-shAMPK-mediated AMPK specific knockdown in the myocardium. The vitro experiments demonstrated that silencing ANXA1 exacerbated lipid deposition, oxidative stress injury, and apoptosis induced by palmitic acid (PA) in cardiomyocytes. Additionally, Ac2-26 effectively mitigated myocardial lipid deposition, oxidative stress injury, and apoptosis induced by PA. These effects were impeded by FPR2 inhibitors Boc-2 and WRW4. The main mechanism involves the activation of AMPK by ANXA1 through FPR2 in order to enhance fatty acid oxidation in cardiomyocytes, thereby ultimately leading to a reduction in lipid accumulation and associated lipotoxicity. Our findings demonstrate that the ANXA1-FPR2 axis plays a protective role in obesity-associated AF by alleviating metabolic stress in the atria of obese mice, thereby emphasizing its potential as a promising therapeutic target for AF. Supplementary Information The online version contains supplementary material available at 10.1186/s12933-024-02545-z. Keywords: Atrial fibrillation, Obesity, ANXA1-FPR2, AMP-activated protein kinase, Lipotoxicity Introduction Atrial fibrillation (AF) is the most common sustained arrhythmia that impairs quality of life and increases the risk for stroke, dementia, heart failure, and sudden cardiac death [[54]1]. Because of the multiformity of potential contributors and the limited understanding of the molecular mechanisms driving AF and its progression, the current rhythm control strategies is inadequate in some patients [[55]2]. The heart is the greatest energy-demanding organ, with 60 to 90% of its energy supplied by mitochondrial oxidation of long-chain fatty acids (FAs) and the remainder by glucose, lactic acid and ketone bodies [[56]3]. Metabolic activity is closely related to arrhythmias by affecting excitation-contraction coupling and ion channel/pump function [[57]4]. Impaired cellular energetic status not only predisposes to atrial arrhythmias, but atrial rhythm disturbances also influence metabolic activity [[58]5]. The rapid rates of electrical and contractile activity during AF will affect atrial energy demands, circulation and oxygen supply, and change the balance between metabolic demand and supply, which is metabolic stress to atria and a driving force for atrial electrical and structural remodeling [[59]3, [60]5–[61]7]. A growing body of evidence suggests that correcting cellular metabolism deficiencies may represent a novel and promising antiarrhythmic strategy [[62]5, [63]8]. Obesity is a well-established risk factor for AF and the prevalence of which is increasing globally [[64]9–[65]11]. Although growing evidences have demonstrated that weight management is effective for primary and secondary AF prevention, the pathophysiological mechanisms linking obesity and AF remain unclear [[66]12, [67]13]. There are pathological metabolic abnormalities in cardiomyocytes in the condition of obesity, characterized by impaired glucose metabolism, increased beta-oxidation of FAs, lipid accumulation, defective mitochondrial oxidative capacity, and increased mitochondrial reactive oxygen species (ROS), which contribute to atrial remodeling and the triggering and maintenance of AF [[68]8, [69]11, [70]14, [71]15]. Our recent studies have shown that improving metabolic abnormalities of cardiomyocytes in obese mice induced by high-fat diet (HFD) can ameliorate the arrhythmogenic substrate and AF susceptibility [[72]6, [73]15, [74]16]. Therefore a comprehensive understanding of the interrelated mechanism of metabolic and arrhythmogenic substrate promises to identify new molecular targets for the treatment of obesity-induced AF. The formyl peptide receptor 2 (FPR2) belongs to the G protein-coupled receptor (GPCR) family; however, unlike typical GPCRs, it exhibits a remarkable ability to recognize diverse ligands with distinct structural characteristics, encompassing formyl peptides, non-formyl peptides, lipids, small molecules, and proteins [[75]17]. Human FPR2 and its mouse homolog FPR2 are highly expressed in a variety of cells, including cardiomyocytes, and interact with peptide and lipid ligands to modulate inflammatory response, cytokine expression and lipid metabolism [[76]17–[77]20]. The latest research findings indicate that resolvin-D1, one of the ligands of FPR2, exhibits potential in preventing AF caused by left ventricular dysfunction. The underlying mechanism suggests that it may exert its effects through the inhibition of myocardial tissue inflammation via the FPR2 receptor [[78]21, [79]22]. These results highlight the therapeutic prospects of targeting the FPR2 receptor for AF treatment. Annexin A1 (ANXA1) is a member of the annexin superfamily and an endogenous ligand of FPR2. ANXA1-FPR2 axis can alleviate the corresponding organ dysfunction by improving metabolic disturbance in different pathological conditions [[80]19, [81]20, [82]23]. However, the role of ANXA1-FPR2 in obesity-induced atrial metabolic disorders and its effect on obesity-related AF susceptibility remain unclear. In this study, we applied bioinformatics tools to execute conjoint analysis of AF and obesity transcriptome datasets with the goal of identifying key molecules involved in the pathological process of obesity-related AF. The results of bioinformatics analysis suggested that ANXA1-FPR2 axis may perform crucially in the pathology of obesity-induced AF. Therefore, using a mouse model of obesity induced by HFD, we further explored the role of ANXA1-FPR2 axis on obesity-induced AF susceptibility and elucidated its underlying mechanism. The experimental design flow is illustrated in (Supplemental Fig. [83]1A). Materials and methods Retrieve the transcriptome datasets of AF and obesity in gene expression omnibus (GEO) The transcriptome datasets about AF and obesity were retrieved from the GEO Datasets. Four AF related atrial transcriptome datasets screened in our previous study were used for analysis, one of which was excluded because it was not on the same sequencing platform as the other datasets [[84]24]. We take “obesity” and “atrium” as keywords for the retrieval in GEO Datasets. The “Homo sapiens” and “Expression profiling by array or Expression profiling by high throughput sequencing” were as filter conditions for “Organism” and “Study type” respectively. The selection criteria for transcriptome datasets of obesity were as follows: (1) tissue samples of atria or appendage obtained from obese and matched controls without AF, (2) datasets were mRNA microarray or high-throughput sequencing results, and (3) the number of samples in each group was not less than three. According to the above screening criteria, an obesity-related atrial transcriptome dataset, [85]GSE159612, was included in the study [[86]25]. The detail information of these five GEO datasets were listed in (Supplemental Table [87]1). Data processing and differentially expressed genes (DEGs) analysis The raw data (CEL file) of microarrays related to AF was processed as previously described [[88]24]. Prior to integrative meta-analysis, individual datasets were normalized by log2 transformation and R-mediated mean, followed by quantile normalization. The meta-analysis was performed using NetworkAnalyst [[89]26], a web-based tool for statistical integration and visualization. The processed and normalized datasets were uploaded to the “multiple gene expression data” input area in the web interface option of multiple gene expression data. They were then subjected to well-established ComBat procedures for removing study-specific batch effects. Principal component analysis (PCA) was employed to visualize and compare the clustering patterns of samples with and without batch effect adjustment, in order to evaluate the effectiveness of batch effect removal. The DEGs were identified through Fisher’s method (p <.05), fixed effects model (p <.001), and Vote Counting (Votes number < 2 and p <.05). Subsequently, the robust DEGs between the AF and control groups were identified by intersecting the DEGs obtained from each model algorithm. The “Define Custom Signature” tool in NetworkAnalyst was utilized to generate a heatmap visualization of the top 10 up- and downregulated genes. The merged data for meta-analysis using NetworkAnalyst is lised in (Supplemental Table [90]2). The gene expression matrix obtained from high-throughput sequencing of atrial tissue in individuals with obesity was downloaded. Inter-group differences were assessed using the limma algorithm, and original p values were adjusted using the Benjamini-Hochberg (BH) method. Gene expression fold changes (FC) were computed by subtracting the mean of normalized log2-based expression levels of the respective control groups from each subject’s normalized log2-based expression levels DEGs were filtered using an adjusted p <.05 and|log2 FC| > 0.5. Common DEGs between AF and obesity were identified. DEGs that were up-regulated or down-regulated in both AF and obesity groups were defined as obesity-related AF DEGs. Functional enrichment analysis of DEGs Metascape ([91]https://metascape.org) was utilized to conduct Gene Ontology (GO) functional analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis [[92]27]. The cutoff criterion was set at p <.01. Construction of protein–protein interaction (PPI) network and hub gene screening STRING database ([93]http://string-db.org) was used to establish the PPI networks [[94]28]. The hub genes were identified through the use of the CytoHuba plugin within Cytoscape software (version 3.8.0), with the degree algorithm being employed to identify these hub genes in this study [[95]29, [96]30]. Gene set expression analyses Gene set enrichment analyses (GSEAs) were conducted using the Molecular Signature database (MSigDB) “c2.cp.kegg.v7.2.symbols.gmt” gene set [[97]31] to evaluate the enrichment of canonical pathways between FPR2 high-expression and low-expression groups, utilizing Gene Set Enrichment Analyses software (Java version 4.0) [[98]32]. Signaling pathways with a normalized p <.05 and a false discovery rate (FDR) q < 0.25 were deemed significantly enriched. Animals and experimental design Male C57BL/6J mice (6 weeks old) were purchased from Xi’an Jiaotong University (Xi’an, China). The animals were housed in a controlled environment with specific conditions (temperature: 22 ± 2 °C; relative humidity: 50% ± 10%; light cycle: 12-hour light/dark cycle with lights on at 7:00 a.m.) and provided ad libitum access to food and water. All experiments were performed in strict accordance with the recommendations of the Guide for the National Institutes of Health Guidelines for the Use of Laboratory Animals and approved by the Animal Care Committee of Xi’an Jiaotong University (The study received ethical approval from the Ethics Committee of Xi’an Jiaotong University, with an assigned approval number: 2022-922). After a one-week acclimatization period, a total of sixty mice were randomly assigned to four groups (n = 15 per group): Standard diet (STD; 10% fat, 70% carbohydrate, 20% protein; Cat. no. D12450B; Research Diets Inc.), STD and Ac2-26 (STD + Ac2-26), HFD (HFD; 60% fat, 20% carbohydrate, 20% protein; Cat. no. D12492; Research Diets Inc.), and HFD + Ac2-26 (HFD + Ac2-26). A mouse model of obesity-induced AF was established by feeding an HFD for 10 weeks when obesity and AF susceptibility changed significantly in the HFD mice compared to the STD mice. The mice were intraperitoneally injected with Ac2-26 (1 mg/kg; Topscience, Shanghai, China), an ANXA1 mimetic peptide, or vehicle for a duration of 10 weeks [[99]33]. At the end of the experiment, electrophysiological examination, echocardiography, and intraperitoneal glucose tolerance testing (IPGTT) were performed on each group before tissue sampling. After an overnight fast, atrial tissues and blood samples were collected from euthanized mice for further analysis. Recombinant viral vector construction and injection The Adeno-associated virus (AAV) particles were procured from GenePharma Co., Ltd (Shanghai, China). Short hairpin sequences were synthesized as previously reported [[100]34], and cloned into the AAV9 vector (AAV9-shAMPK 5′-ATGATGTCAGATGGTGAATTT AAGTTCTCTAAACCTACCATCTGACAT-3′ and AAV9-shNC: 5′-ACGACGTCAGCT GGTGCA GT AGTTCTCTACATGCACCAGCTGACGTCGT-3′). After 8 weeks of high-fat diet feeding, AVV9 (1.7 × 10^13 vg/mL) was administered via intravenous injection through the tail vein. After 4 weeks of intervention with AAV9 and Ac2-26, cardiac electrophysiological examination, echocardiography and IPGTT were performed. Cell isolation, culture and treatment The neonatal rat cardiomyocytes (NRCMs) were isolated from the hearts of male neonatal Sprague-Dawley rats aged 1 to 3 days old. In brief, atrial tissue was surgically excised and subsequently digested with collagenase II (0.1%; Solarbio, Beijing, China) to obtain a cell suspension. NRCMs were then isolated through differential adherence and verified under a microscope. The isolated cells were then cultured in 6-well plates at 37 °C with Dulbecco’s Modified Eagle Medium (DMEM) supplemented with 10% Fetal Bovine Serum (FBS) and 1% penicillin/streptomycin solution, while maintaining a CO[2] concentration of 5%. At 80% confluence, the cells were stimulated with palmitic acid (PA) at a final concentration of 200 µM for 24 h to simulate the effects of lipid overload [[101]15]. Cells were pre-treated with FPR2 antagonists WRW4 (10 µM, Topscience) or Boc-2 (10 µM, Topscience) prior to being treated with 0.3 µM Ac2-26 (Topscience) [[102]35]. Compound C (CC, 0.5 µM; Sigma-Aldrich) was added to inhibit AMPK activation. The NRCMs were transfected with siRNAs targeting rat ANXA1 (GenePharm, Shanghai, China) using Entranster^TM-R4000 siRNA Transfection Reagent (Engreen Biosystem, Beijing, China) according to the manufacturer’s instructions. The sequence of ANXA1 siRNA is 5’-CAAGAUCUAACCAGCAAAUTT-3’. Electrophysiological and echocardiography examinations AF inducibility was evaluated through programmed transesophageal electrical stimulation, as previously described with minor modifications [[103]24]. Briefly, after inducing anesthesia with intraperitoneal injection of sodium pentobarbital (60 mg/kg), a continuous surface lead electrocardiogram (ECG) was recorded using a physiological signal-acquisition system (RM6240; Chengdu Instrument Factory). AF was then induced through high-frequency burst pacing (pulse width 1 ms; 2× threshold current; 30, 35, and 40 Hz, respectively) using a programmable electrical stimulator (VCS3001; MappingLab Ltd.). AF was defined as a period of rapid and irregular atrial rhythm lasting for at least 1 s on ECG. Noninvasive transthoracic echocardiography was performed using a Vevo 2100 Imaging System (VisualSonics Inc.) to discern differences in cardiac structure and function between groups. All images were subsequently processed with Vevolab 3.1 software (VisualSonics). IPGTT The glucose homeostasis was assessed by IPGTT. Overnight fasted mice were intraperitoneally injected with D-glucose (2 g/kg·BW), and blood glucose levels were measured at 0, 15, 30, 60, 90 and 120 min after glucose loading by the ACCU-CHEK Performa Glucometer (Roche Diagnostics, Indianapolis, United States) using tail-vein blood. The glucose tolerance was determined by calculating the area under the curve (AUC) using GraphPad Prism 8.0 software (GraphPad Software, San Diego, USA). Detection of biomarkers in serum and tissue samples Biomarkers were quantified using commercially available kits in accordance with the manufacturer’s instructions, including fasted nonesterified fatty acids (NEFA) (Solarbio), insulin (Cusabio, Wuhan, China), malondialdehyde (MDA) (Beyotime Institute of Biotechnology, Shanghai, China), and superoxide dismutase (SOD) (Beyotime Institute of Biotechnology). Analysis of histological staining and fluorescence Atrial tissue samples were fixed in 4% paraformaldehyde overnight, subsequently embedded in paraffin and sectioned into slices that were 5 μm thick. Atrial sections were stained with Masson’s trichrome, selected antibodies (ANXA1; 1:200; Santa Cruz Biotechnology, California, United States and FPR2; 1:200; Santa Cruz Biotechnology), and TUNEL (in situ cell death detection kit, TMR red; Roche Diagnostics). Moreover, frozen Sect. (10 μm thickness) of atria samples were prepared and stained with oil red O. The sections were examined and captured under a light microscope (Nikon, Melville, New York, United States) or a fluorescence microscope (Leica, Bensheim, Germany), followed by further analysis using ImageJ software (National Institutes of Health). Quantitative real-time PCR (RT-qPCR) analysis Total RNA was extracted from fresh atrial tissue using TRIzol (AG, Changsha, China) following the manufacturer’s instructions. Subsequently, single-stranded cDNA was synthesized with the assistance of Evo M-MLV RT Kit (AG). The SYBR green chimeric fluorescence method (Accurate Biology) was employed for the RT-qPCR reaction on a CFX96™ Real-Time PCR System (Bio-Rad, Hercules, CA, United States). The 2^−△△Ct comparative method was used to quantify results which were then normalized by β-actin. Primer sequences are listed in (Supplemental Table [104]3). Western blot Atrial protein lysates were extracted and quantified using a bicinchoninic acid (BCA) protein assay The proteins underwent sodium dodecyl sulfate polyacrylamide gel electrophoresis and were subsequently transferred onto a polyvinylidene difluoride membrane, which was then incubated with primary antibodies targeting ANXA1 (1:1000; ZEN-BIOSCIENCE, Chengdu, China), FPR2 (1:1000; Santa Cruz Biotechnology), Bax (1:1000; ZEN-BIOSCIENCE), Bcl2 (1:1000; ZEN-BIOSCIENCE), cytochrome complex (Cyt C; 1:1000; Santa Cruz Biotechnology), caspase3 (1:1000; Cell Signaling Technology/CST), phosphorylated c-Jun N-terminal kinase (p-JNK; 1:1000; CST), t-JNK (1:1000; Proteintech), Acetyl-CoA carboxylase (Acc; 1:1000; Proteintech), phosphorylated Acc (p-Acc; 1:1000; CST), AMP-activated protein kinase (AMPK; 1:1000; CST), phosphorylated AMPK (p‐AMPK; Thr172; 1:1000; CST), peroxisome proliferator–activated receptor alpha (PPARα; 1:1000; Santa Cruz Biotechnology), carnitine palmitoylransferase-1B (CPT1B; 1:1000; Proteintech), and Glyceraldehyde-3-phosphate dehydrogenase (GAPDH; 1:1000; Proteintech). Subsequently, the membranes were incubated with secondary antibodies for 1–2 h. The resulting bands were detected using an ECL Kit (Millipore) and imaged with a Tanon imaging system (Tanon). Densitometry analysis was performed using Quantity One software (Bio-Rad) to quantify the results. Statistical analysis The data were presented as mean ± SEM for animal or cell experiments, and all statistical analyses were conducted using GraphPad Prism software (version 8.0). The Kolmogorov-Smirnov normality test was performed to assess the distribution of the data. If the data followed a normal distribution, unpaired two-tailed Student t-tests were used to compare two groups, while One-way ANOVA or Two-way ANOVA with Bonferroni correction was employed for comparing multiple groups. In case of non-normal distribution, the Kruskal-Wallis test was utilized for between-group comparisons. A significance level of p <.05 was considered statistically significant. Results The hub gene FPR2 may be closely associated with obesity-related AF Between the AF and control groups, a total of 3323 DEGs were identified using the Fixed Effect Model algorithm in the Meta-analysis module (Supplemental Table [105]4), while 5003 DEGs were detected by Fisher’s method algorithm model (Supplemental Table [106]4). The Vote Counting algorithm model screening yielded a set of 3013 DEGs (Supplemental Table [107]4). We obtained a robust list of 1083 DEGs by taking the intersection of these sets (Fig. [108]1A, Supplemental Table [109]4), and (Fig. [110]1C) shows the top 10 up- and downregulated genes. Differential analysis identified 7394 DEGs between the obesity and control groups (Fig. [111]1B, Supplemental Table [112]5). Subsequently, we screened for 133 DEGs with synchronous expression trends in both AF and obesity patients by intersecting the DEGs from these two groups (Supplemental Table [113]6). Of these, 132 were significantly up-regulated while only one was down-regulated in both AF and obesity (Fig. [114]1D). Through the online analysis tool Metascape, functional enrichment analysis of DEGs with synchronous expression trends in AF and obesity revealed their significant involvement in regulating inflammatory response, apoptosis, and GPCR downstream signaling pathways (Fig. [115]1E). Fig. 1. [116]Fig. 1 [117]Open in a new tab Identification and validation of hub genes in obesity-related AF. A The DEGs were identified through Fisher’s method (p <.05), fixed effects model (p <.001), and Vote Counting (Votes number < 2 and p <.05). Subsequently, the robust DEGs between the AF and control groups were identified by intersecting the DEGs obtained from each model algorithm. B The volcano plot display DEGs in obesity patients and the control group. C The heat map displays the top 10 significantly up-regulated and down-regulated DEGs between patients with AF and controls. D 133 DEGs with the same expression trend in AF and obesity patients. E Construction of PPI network and F hub gene screening. G, H Representative sections and quantification of FPR2 using immunofluorescence staining in the atrium of obese mice. I, J Representative sections and quantification of ANXA1 using immunofluorescence staining in the atrium of obese mice. K The concentration of ANXA1 in the serum of mice in each group. Data are shown as mean ± SEM. *p <.05; *** p <.001 between indicated groups. n = 5 for immunofluorescence staining analysis. The scale bar = 50 μm. AF, atrial fibrillation; DEGs, differentially expressed genes; PPI, protein–protein interaction The DEGs were utilized to construct a PPI network, and the hub gene of the network was identified as FPR2 due to its high degree of connectivity (Fig. [118]1F). The expression of FPR2 is significantly upregulated in the atria of both AF and obese patients. A receiver operating characteristic (ROC) curve was constructed using FPR2 gene expression from integrated AF and control transcript datasets, revealing that FPR2 expression has good diagnostic value for AF (AUC 0.758, 95%CI 0.635–0.871) (Supplemental Fig. [119]1B). Additionally, AF patients were stratified into a low FPR2 expression group and a high FPR2 expression group based on the median value of FPR2 gene expression for further single-gene GSEA enrichment analysis. The results indicated that the high FPR2 expression group was primarily enriched in lymphocyte differentiation, extracellular matrix (ECM) receptor interaction, adipocytokine signaling pathway, lipid metabolism, and PPAR signaling pathway (Supplemental Fig. [120]1C-D). The findings imply that FPR2 may participate in the pathophysiological mechanisms underlying atrial inflammation and metabolism in patients with AF. Validation of FPR2 and its endogenous ligand ANXA1 The expression levels of FPR2 were investigated in a mouse model of HFD-induced obesity. Immunofluorescence and Western blot results revealed significant upregulation of FPR2 expression in the atria of HFD-induced obese mice compared to the control group (Fig. [121]1G-H, Supplemental Fig. [122]1E-F), which was consistent with the result of our bioinformatics analysis. We conducted further analysis on ANXA1, the endogenous ligand of FPR2, and observed no significant difference in its expression level within the integrated AF database (Supplemental Fig. [123]1G). However, in another high-throughput sequencing dataset for AF ([124]GSE69890), we discovered a notable down-regulation of ANXA1 expression in the atria of AF patients compared to that of the control group (Supplemental Fig. [125]1H, Supplemental Table [126]7). We further conducted a measurement of serum ANXA1 concentrations in patients with AF and matched controls, enrolling 30 AF patients and 26 matched controls through screening (The study received ethical approval from the Ethics Committee of the Second Affiliated Hospital of Xi’an Jiaotong University, with an assigned approval number: 2021197). The basic information is presented in (Supplemental Table [127]8). Our findings indicate that serum ANXA1 levels were significantly lower in AF patients compared to the control group (Supplemental Fig. [128]1I). However, in the obesity-related dataset [129]GSE159612, ANXA1 expression in the atria of obese patients was found to be significantly increased compared to that of the control group (Supplemental Fig. [130]1J). Moreover, both atrial ANXA1 expression and serum ANXA1 concentration were significantly upregulated in HFD-induced obese mice when compared to the control group (Fig. [131]1I-K, Supplemental Fig. [132]1E, K). Ac2-26 attenuates obesity-induced AF susceptibility and insulin resistance To investigate the potential involvement of the ANXA1-FPR2 axis in the pathogenesis of obesity-induced AF, we initially administered Boc-2, an inhibitor of FPR2, to obese mice for a 10 weeks (Supplemental Fig. [133]2A). The findings revealed that while there were no significant disparities in the susceptibility and duration of AF between the obese mice treated with Boc and those treated without it, the intervention with Boc-2 exhibited an ability to augment both the susceptibility and duration of AF (Supplemental Fig. [134]2B-D). Furthermore, Masson staining indicated that Boc-2 could exacerbate atrial fibrosis in obese mice (Supplemental Fig. [135]2E-F). The subsequent investigation focused on evaluating the impact of an additional 10-week supplementation of Ac2-26, an active peptide chain of ANXA1, on the susceptibility to AF in mice with diet-induced obesity (Fig. [136]2A). The baseline characteristics of the mice in each group are summarized in (Table [137]1). HFD mice exhibited progressive weight gain over time, whereas treatment with Ac2-26 resulted in a reduction of body weight specifically in obese mice but not lean counterparts (Fig. [138]2B-C). The AF susceptibility in obese mice was significantly higher than controls, and this phenomenon was significantly reduced with Ac2-26 (Fig. [139]2D-F). Fig. 2. [140]Fig. 2 [141]Open in a new tab The administration of Ac2-26 improves insulin resistance and reduces susceptibility to AF in obese mice induced by HFD. A Experimental design of this study. Mice were randomly divided into four groups: STD group, STD + Ac2-26 group, HFD group, and HFD + Ac2-26 group. B Weight gain curve and C the change of body weight. D Representative ECGs showing AF induction by transesophageal burst pacing. E AF frequency and F AF duration were assessed between the groups. G Fasting blood glucose, H random blood glucose, and I fasting serum insulin among groups. J HOMA-IR (insulin resistance index) were calculated. K GTT and L calculated AUC among groups. Data are shown as mean ± SEM. *p <.05; **p <.01; *** p <.001 between indicated groups. n = 6–14 for animal experiments. AF, atrial fibrillation; AUC, area under the curve; HFD, high‐fat diet; HOMA‐IR, homeostasis model assessment of insulin resistance; GTT, glucose tolerance test Table 1. Physical characteristics, echocardiographic, electrophysiological parameters, and metabolism-related indicators in mice from STD, STD + Ac2-26, HFD, and HFD + Ac2-26 groups Parameter STD (n = 8–11) STD + Ac2-26 (n = 8–12) HFD (n = 9–14) HFD + Ac2-26 (n = 9–14) Physical characteristics BW (g) 28.16 ± 0.36* 26.40 ± 0.40 40.66 ± 1.15*^# 33.43 ± 1.27^# AW (mg) 14.22 ± 1.23 13.15 ± 1.16 15.75 ± 1.42 14.17 ± 1.13 HW (mg) 143.01 ± 2.75 133.91 ± 4.85 147.14 ± 3.48 141.35 ± 2.59 HW/BW (mg/g) 5.08 ± 0.10* 5.07 ± 0.17 3.67 ± 0.17*^# 4.31 ± 0.20^# BL (cm) 10.51 ± 0.06 10.45 ± 0.12 10.58 ± 0.18 10.85 ± 0.05 HW/BL (mg/cm) 13.61 ± 0.26 12.78 ± 0.36 13.92 ± 0.35 13.02 ± 0.26 Echocardiography IVSd (mm) 0.89 ± 0.05 0.86 ± 0.06 0.96 ± 0.06 0.83 ± 0.06 IVSs (mm) 1.33 ± 0.05 1.27 ± 0.06 1.45 ± 0.07 1.20 ± 0.06 LVIDd (mm) 3.44 ± 0.13 3.22 ± 0.16 3.22 ± 0.10 3.38 ± 0.11 LVIDs (mm) 2.24 ± 0.12 2.12 ± 0.16 2.07 ± 0.13 2.19 ± 0.15 LVPWd (mm) 0.93 ± 0.02 0.92 ± 0.03 1.01 ± 0.05^# 0.81 ± 0.04^# LVPWs (mm) 1.30 ± 0.06 1.29 ± 0.07 1.43 ± 0.06 1.27 ± 0.07 LVEF (%) 65.95 ± 2.40 65.55 ± 3.294 66.20 ± 3.21 65.44 ± 3.89 LVFS (%) 34.89 ± 1.78 34.62 ± 2.44 35.41 ± 2.37 35.75 ± 2.98 Electrophysiology P (ms) 20.09 ± 0.57 20.00 ± 0.84 21.58 ± 0.74^# 19.63 ± 0.50^# PR (ms) 37.87 ± 1.06* 42.08 ± 1.21 43.35 ± 1.33* 41.67 ± 1.37 QRS (ms) 23.12 ± 1.82 23.47 ± 2.13 24.86 ± 0.56 20.82 ± 1.08 QT (ms) 65.36 ± 1.32 66.94 ± 1.97 68.05 ± 1.42 65.14 ± 1.01 SACT (ms) 20.12 ± 1.13 20.10 ± 3.38 24.67 ± 6.29 21.50 ± 2.10 AVERP140 (ms) 66.89 ± 2.60 66.11 ± 1.624 63.50 ± 4.22 64.50 ± 3.76 Metabolism-related indicators TG (mmol/l) 0.96 ± 0.05* 0.95 ± 0.05 1.24 ± 0.07*^# 0.97 ± 0.05^# NEFA (mmol/l) 0.52 ± 0.05* 0.49 ± 0.08 0.77 ± 0.06*^# 0.55 ± 0.05^# MDA (µmol/mg prot) 8.60 ± 0.58* 8.22 ± 0.48 12.44 ± 0.98*^# 9.77 ± 0.45^# SOD (U/mg prot) 520.70 ± 0.58* 564.27 ± 0.82 455.80 ± 5.96*^# 501.67 ± 1.83^# [142]Open in a new tab AVERP, atrioventricular node effective refractory period; AW, atria weight; BL, body length; BW, body weight; HFD, high-fat diet; HW, heart weight; IVSd, interventricular septal thickness at end-diastole; IVSs, interventricular septal thickness at end-systole; LVIDd, left ventricular internal diameter at end-diastole; LVIDs, left ventricular internal diameter at end-systole; LVPWd, left ventricular posterior wall thickness at end-diastole; LVPWs, left ventricular posterior wall thickness at end-systole; LVEF, left ventricular ejection fraction; LVFS, left ventricular fractional shortening; NEFA, nonesterified fatty acids; SACT, sinoatrial conduction time; TG, triglycerides. All data are expressed as mean ± SEM and n represents number of animals in each group. *p <.05, STD versus HFD; ^#p <.05, HFD versus HFD + Ac2-26 To investigate whether metabolic disorders, particularly insulin resistance, contribute to the Ac2-26-induced reduction in AF susceptibility in obese mice, we assessed the degree of insulin sensitivity among groups using measures such as fasting blood glucose (FBG), fasting serum insulins (FINs), GTT and homeostasis model assessment of insulin resistance (HOMA-IR). The HFD mice exhibited apparent insulin resistance, as evidenced by elevated FBG, FINs, HOMA-IR values, and impaired glucose tolerance compared to the controls (Fig. [143]2G-L). In mice treated with Ac2-26, the detrimental effects of HFD on insulin sensitivity were mitigated, as evidenced by a reduction in FBG and FINs, an improvement in HOMA-IR score and glucose tolerance (Fig. [144]2G-L). The findings suggest that Ac2-26 may mitigate HFD-induced AF susceptibility by ameliorating metabolic dysfunction in obese mice. AMPK mediates Ac2-26 to reduce obesity-induced AF susceptibility Previous studies have demonstrated the significant involvement of AMPK in AF. Therefore, we conducted an evaluation to investigate whether Ac2-26 could mitigate AF susceptibility in obese mice by activating AMPK through FPR2. Specifically, AAV9-shAMPK was employed to specifically knock down AMPK expression in heart (Supplemental Fig. [145]2G-I), followed by a 4-week intervention with Ac2-26. The baseline characteristics of each group of mice are presented in (Supplemental Table [146]9). No notable differences were observed between the two groups regarding body weight, fasting glucose levels, and IPGTT results (Fig. [147]3A-E). However, after one month of Ac2-26 treatment, a remarkable reduction in both AF susceptibility and duration was observed in the AAV-shNC group compared to AAV9-shAMPK group (Fig. [148]3F-H). Furthermore, the administration of Ac2-26 for a duration of one month resulted in a significant reduction in atrial diameter and atrial fibrosis within the shNC group, as compared to the shAMPK group (Fig. [149]3I-L). The findings suggest that the activation of AMPK by Ac2-26, independent of weight loss and improvement in systemic insulin resistance factors, plays a crucial role in reducing susceptibility to AF in obese mice. Fig. 3. [150]Fig. 3 [151]Open in a new tab AMPK is a key target for Ac2-26 to improve AF susceptibility in obese mice. A Weight gain curve and B the change of body weight. Fasting blood glucose (C), GTT (D) and (E) calculated AUC among groups. F Representative ECGs showing AF induction by transesophageal burst pacing. G AF frequency and H AF duration were assessed between the groups. I-L Echocardiography and Masson staining showed that Ac2-26 did not improve atrial enlargement and fibrosis in AAV9-shAMPK group compared with AAV9-shNC group. Data are shown as mean ± SEM. *p <.05 between indicated groups. n = 3–10 for animal experiments. The scale bar = 50 μm. AF, atrial fibrillation; AAV, Adeno-associated virus; AUC, area under the curve; HFD, high-fat diet; GTT, glucose tolerance test; LAD, left atrial dimension Ac2-26 attenuates obesity-induced atrial remodeling Since electrical and structural remodeling are crucial in the initiation and maintenance of AF, we further investigated whether HFD could impact atrial electrical and structural remodeling, as well as the role of Ac2-26. The baseline Surface ECG (Lead II) indicated that HFD prolonged the P-wave duration in obese mice, which was mitigated by Ac2-26 treatment (Table [152]1). However, there were no significant changes observed in atrioventricular node effective refractory period (AVERP) and sinoatrial conduction time (SACT) between Ac2-26-treated and untreated mice (Table [153]1). Next, we evaluated the transcriptional profiles of pivotal ion channels implicated in AF pathogenesis. HFD augmented Cav1.2, Cav1.3, Ryanodine receptor 2 (RyR2), Kv3.1, Kr/hERG and KATP/Kir6.2 gene expression while Ac2-26 intervention upregulated Nav1.5, Cav1.2 and Cav1.3 but downregulated Kr/hERG in obese mice (Fig. [154]4A-C). The typical atrial action potential morphology with the corresponding AF-related ion channel transcriptional profile is depicted in (Fig. [155]4D). Due to the insignificant impact of Ac2-26 on the up-regulated RyR2 in obese mice, we proceeded to investigate potential post-translational modifications of RyR2. The Western blot analysis revealed that treatment with Ac2-26 significantly reduced the phosphorylation level of atrial RyR2 in HFD-induced obese mice (Fig. [156]4I-J). Fig. 4. [157]Fig. 4 [158]Open in a new tab Effect of Ac2-26 on atrial remodeling in obese mice. A-C Transcriptional profile of AF-related sodium channel (Nav1.5), calcium channels (Cav1.2, Cav1.3 and RyR2), and potassium channels (Kv4.3, Kv1.5, Kv3.1, Kr/hERG, Ks/Kv7.1, KACH/Kir3.4, Kir2.1, and KATP/Kir6.2) among groups (n = 4). D Typical shape of an atrial action potential showing principle currents and the corresponding subunits (ion channels). E-H The echocardiography and Masson staining results revealed left atrial enlargement and a significant increase in fibrosis in obese mice, whereas Ac2-26 exhibited a notable improvement in structural remodeling (n = 3–9). I, J The administration of Ac2-26 resulted in a significant reduction in the phosphorylation level of RyR2 in obese mice (n = 4). Data are shown as mean ± SEM. *p <.05; **p <.01; *** p <.001 between indicated groups. The scale bar = 50 μm. HFD, high‐fat diet; RyR2, ryanodine receptor 2; LAD, left atrial dimension Furthermore, we investigated the atrial structural remodeling. The results of echocardiography and Masson staining demonstrated that HFD significantly increased atrial diameter and myocardial fibrosis compared to the control group, while Ac2-26 intervention notably reduced both parameters in obese mice induced by HFD (Fig. [159]4E-H). These findings suggest that Ac2-26 can ameliorate atrial electrical and structural remodeling in HFD-induced obesity. Ac2-26 alleviated myocardial lipid accumulation and toxicity by promoting lipid metabolism Further investigation in myocardial tissue revealed a significant increase in atrial myocyte apoptosis, as evidenced by TUNEL staining, in obese mice (Fig. [160]5A-B). Western blot analysis demonstrated that HFD promoted the expression of pro-apoptotic proteins Cyt C and cleaved caspase3/caspase3, up-regulated JNK phosphorylation levels, and reduced Bcl2/Bax levels, thereby exerting a pro-apoptotic effect. However, Ac2-26 significantly inhibited these effects and played an anti-apoptotic role in HFD-induced obese mice (Fig. [161]5C-G). Furthermore, HFD resulted in an elevation of MDA levels within the atria while simultaneously reducing SOD concentrations. Ac2-26 exhibits an antioxidative stress effect in the atrium of HFD-induced obese mice by reducing MDA levels and up-regulating SOD concentration (Table [162]1). Fig. 5. [163]Fig. 5 [164]Open in a new tab The promotion of fatty acid oxidation by Ac2-26 may potentially mitigate lipotoxicity in atrial myocytes of obese mice. A, B The tunel staining analysis revealed that Ac2-26 significantly attenuated apoptosis in atrial myocytes of obese mice (n = 3). C-G The Western blot analysis demonstrated that Ac2-26 significantly attenuated the expression of pro-apoptotic protein molecules in obese mice (n = 4). H, I Representative sections and quantification of lipid accumulation using Oil Red O staining (n = 3). J-L Transcriptional profile of, glucose metabolism-related genes (GLUT1, GLUT4, HK2, PFKM, PKM2, and PDK4), fatty acid metabolism‐related genes (CD36, FABPpm, FABP3, and CPT1B) and metabolism regulatory genes (HIF1α, PGC1α, FOXO1, PPARα, PPARγ and PPARδ) among groups (n = 4). M-Q Representative images and Western blot analysis of Acc, AMPK, CPT1B and PPARα among groups (n = 4). Data are shown as mean ± SEM. *p <.05; **p <.01; *** p <.001 between indicated groups. The scale bar = 50 μm. AMPK, AMP‐activated protein kinase; Acc, Acetyl-CoA carboxylase; CD36, fatty acid translocase; CPT1B, carnitine palmitoylransferase-1B; FABP-pm, plasma membrane fatty acid-binding protein; FABP3, fatty acid binding protein 3; FOXO1, forkhead box protein O1, GLUT, glucose transporter; HFD, high‐fat diet; HIF1α, hypoxia inducible factor-1α; HK2, hexokinase2; PDK4, pyruvate dehydrogenase kinase 4; PFKM, phosphofructokinase; PGC1α, peroxisome proliferator-activated receptor γ coactivator1α; PKM2, pyruvate kinase isozyme type M2; PPAR, peroxisome proliferators-activated receptor Our previous study has indicated that HFD-induced obesity in mice leads to abnormal lipid deposition in the myocardium, resulting in myocardial lipid toxicity and contributing to the pathogenesis of AF. Based on previous bioinformatics analysis, it has been suggested that FPR2 may play a role in lipid metabolism. Therefore, we conducted further investigations to determine whether Ac2-26 affects lipid metabolism in myocardial tissue. HFD mice showed an increased tendency in serum NEFA and TG levels, and Ac2-26 treatment decreased it in obese mice but not lean mice (Table [165]1). Furthermore, Oil Red O staining and transmission electron microscope revealed that HFD increased lipid accumulation in the atria, while Ac2-26 treatment alleviated lipid accumulation in HFD mice but not in lean mice (Fig. [166]5H-I and Supplemental Fig. [167]2J-K). Thereafter, we evaluated the transcription and translation profiles of the metabolism-related genes. We observed that treatment with Ac2-26 resulted in increased transcription of genes related to glucose metabolism (pyruvate dehydrogenase kinase 4, PDK4), fatty acid metabolism (fatty acid translocase, CD36; plasma membrane fatty acid binding protein, FABPpm; CPT1B) and metabolic regulation (PPARα) in obese mice (Fig. [168]5J-L). Additionally, HFD was found to down-regulate the phosphorylation of Acc and AMPK, while Ac2-26 up-regulated their phosphorylation (Fig. [169]5M-O). No significant effect of HFD on the protein levels of CPT1B and PPARα was observed, but Ac2-26 treatment significantly upregulated their expression in the atria of obese mice (Fig. [170]5M, P-Q). ANXA1 knockdown aggravates PA-induced lipotoxicity in NRCMs To further investigate the impact of ANXA1 on lipid metabolism and lipid toxicity in NRCMs, siRNA was employed to knockdown ANXA1 expression in NRCMs (Fig. [171]6A-C). Subsequently, the cells were treated with 200 µM PA for 24 h to simulate a high-fat environment (Fig. [172]6D). Compared to the bovine serum albumin (BSA) control group, PA intervention significantly increased cardiomyocyte apoptosis, promoted the production of ROS and MDA, and decreased mitochondrial membrane potential and SOD concentration in cardiomyocytes (Fig. [173]6E-L). Knockdown of ANXA1 further exacerbated ROS and MDA production, reduced SOD activity and mitochondrial membrane potential, and aggravated cardiomyocyte apoptosis (Fig. [174]6E-L). Fig. 6. [175]Fig. 6 [176]Open in a new tab ANXA1 knockdown aggravates PA-induced lipotoxicity in cardiomyocytes. A-C Determination of ANXA1 knockdown efficiency by RT-qPCR and Western blot (n = 4). D Experimental design of cell experiments. E, F Representative images and statistical analysis of flow cytometry-detected apoptosis level (n = 3). (G, H) Representative images and analysis of the subcellular localization of the oxidation products of DHE (n = 100). I, J Representative images and analysis of the mitochondrial membrane potential detected by JC-1 staining (red/green) (n = 100). (K-L) Cellular MDA and SOD concentrations among the groups (n = 5). M-T Representative images and Western blot analysis of t-RyR2, Acc, Ampk, CPT1B, PPARα and caspase3 among the groups (n = 4). Data are shown as mean ± SEM. *p <.05; **p <.01; *** p <.001 between indicated groups. The scale bar = 50 μm. AMPK, AMP‐activated protein kinase; Acc, Acetyl-CoA carboxylase; BSA, bovine serum albumin; CPT1B, carnitine palmitoylransferase-1B; DHE, dihydroethidium; MDA, malondialdehyde; PA, palmitic acid; PPAR, peroxisome proliferators-activated receptors; RyR2, ryanodine receptor 2; RT‐qPCR, quantitative reverse transcription‐PCR; SOD, superoxide dismutase The Western blot results demonstrated that PA significantly attenuated AMPK phosphorylation and increased the ratio of cleaved caspase3/caspase3 in cardiomyocytes, while no significant effect was observed on the expression of t-RyR2, Acc, CPT1B and PPARα (Fig. [177]6M-T). Knockdown of ANXA1 further suppressed AMPK phosphorylation in PA-treated cardiomyocytes, downregulated PPARα and CPT1B protein expression levels in the PA group, and upregulated t-RyR2 and Acc protein expression levels in both BSA and PA groups (Fig. [178]6M-T). In summary, ANXA1 knockdown in cardiomyocytes impeded fatty acid catabolism and exacerbated oxidative stress and lipotoxicity induced by PA. ANXA1 attenuates PA-induced lipotoxicity via FPR2 To further explore the function and mechanism of ANXA1 on cardiomyocytes, we conducted in vitro experiments utilizing Ac2-26 in conjunction with FPR2 inhibitors Boc-2 and WRW4 (Fig. [179]7A). Our findings revealed that high concentrations of Ac2-26 (> 30 µM) exhibited cytotoxicity towards NRCMs, whereas concentrations at 0.3 µM and 3 µM did not elicit this adverse effect. To minimize the potential side effects of Ac2-26, a concentration of 0.3µM was utilized, almost consistent with prior literature [[180]35, [181]36]. Ac2-26 significantly attenuated PA-induced cardiomyocyte apoptosis by down-regulating ROS and MDA production, while up-regulating SOD concentration and mitochondrial membrane potential. However, these effects were attenuated when FPR2 was blocked with the antagonist Boc-2 or WRW4 (Fig. [182]7B-I). Fig. 7. [183]Fig. 7 [184]Open in a new tab ANXA1 attenuates PA-induced lipotoxicity via FPR2. A Experimental design of cell experiments. B-C Representative images and statistical analysis of flow cytometry-detected apoptosis level (n = 4). D, E Representative images and analysis of the subcellular localization of the oxidation products of DHE (n = 100). F, G Representative images and analysis of the mitochondrial membrane potential detected by JC-1 staining (red/green) (n = 100). H, I Cellular MDA and SOD concentrations among the groups (n = 3). J-Q Representative images and Western blot analysis of Acc, Ampk, CPT1B, PPARα, caspase3 and RyR2 among the groups (n = 4). Data are shown as mean ± SEM. *p <.05; **p <.01; *** p <.001 between indicated groups. The scale bar = 50 μm. AMPK, AMP-activated protein kinase; Acc, Acetyl-CoA carboxylase; BSA, bovine serum albumin; CPT1B, carnitine palmitoylransferase-1B; DHE, dihydroethidium; MDA, malondialdehyde; PA, palmitic acid; PPAR, peroxisome proliferators-activated receptors; RyR2, ryanodine receptor 2; SOD, superoxide dismutase Similarly, Ac2-26 was found to decrease the ratio of cleaved caspase3/caspase3 and RyR2 phosphorylation induced by PA. Moreover, Ac2-26 was observed to enhance the phosphorylation of AMPK and Acc, as well as increase the expression of PPARα and CPT1B. These effects were reversed by FPR2 blockers (Fig. [185]7J-Q). Therefore, ANXA1 may mitigate oxidative stress and lipotoxicity caused by PA in cardiomyocytes through promoting fatty acid metabolism via FPR2. AMPK serves as a crucial effector molecule in the ANXA1-FPR2 pathway to mitigate cellular lipotoxicity in NRCMs AMPK serves as a pivotal regulatory component in cellular energy metabolism. While PA can attenuate the phosphorylation level of AMPK in cardiomyocytes, ANXA1 is capable of activating it. Therefore, we further investigated whether AMPK was involved in ANXA1-FPR2-induced cardiomyocyte protective effect by inhibiting AMPK activity using AMPK inhibitor Compound C (Fig. [186]8A). We observed that treatment with Compound C significantly attenuated the effects of Ac2-26 on PA-induced oxidative stress, mitochondrial membrane potential, and apoptosis in NRCMs (Fig. [187]8B-I). The Western blot results demonstrated that Compound C counteracted the anti-apoptotic effect of Ac2-26 and elevated the ratio of Cleaved caspase3/caspase3, while also reinstating RyR2 phosphorylation levels (Fig. [188]8J-P). Additionally, Compound C significantly decreased the protein expression of both CPT1B and PPARα as well as the phosphorylation of Acc that was induced by Ac2-26 (Fig. [189]8J-P). Fig. 8. [190]Fig. 8 [191]Open in a new tab AMPK functions as a pivotal effector molecule within the ANXA1-FPR2 signaling pathway. A Experimental design of cell experiments. B-G Representative images and statistical analysis were conducted to assess the levels of apoptosis detected by flow cytometry, as well as the oxidation products of DHE and JC-1 staining (red/green) (n = 4-100). H, I Cellular MDA and SOD concentrations among the groups (n = 3). J-P Representative images and Western blot analysis of Acc, Ampk, CPT1B, PPARα, caspase3 and RyR2 among the groups (n = 4). Q Proposed model of ANXA1-mediated protection against lipid accumulation and lipotoxicity. Data are shown as mean ± SEM. *p <.05; **p <.01; *** p <.001 between indicated groups. The scale bar = 50 μm. AMPK, AMP-activated protein kinase; Acc, Acetyl-CoA carboxylase; BSA, bovine serum albumin; CD36, fatty acid translocase; CPT1B, carnitine palmitoylransferase-1B; DHE, dihydroethidium; FABP-pm, plasma membrane fatty acid-binding protein; HFD, high‐fat diet; MDA, malondialdehyde; PA, palmitic acid; PPAR, peroxisome proliferators-activated receptors; RyR2, ryanodine receptor 2; SOD, superoxide dismutase Discussion In this study, we have identified differential expression of ANXA1 and FPR2 in the atria of patients with AF and obese individuals for the first time. Subsequently, we investigated the role and potential mechanism of ANXA1-FPR2 in obesity-related AF. Specifically, FPR2 was found to be significantly upregulated in the atria of both AF and obese patients, while its endogenous ligand ANXA1 was significantly upregulated in obese patients but downregulated in AF. Supplementation with the active polypeptide Ac2-26 of ANXA1 significantly mitigated the susceptibility to AF in high-fat diet-induced obese mice; however, this protective effect was diminished in obese mice with specific myocardial AMPK knockdown by AAV9-shAMPK. The mechanism study revealed that ANXA1-FPR2 may participate in mitigating myocardial lipotoxicity by activating the downstream AMPK signaling pathway to facilitate fatty acid catabolism in cardiomyocytes (Fig. [192]8Q). ANXA1 and FPR2 are expressed in a variety of cell types, including cardiomyocytes, macrophages, and neutrophils. ANXA1 functions as an endogenous ligand of FPR2 and is released via autocrine and paracrine pathways [[193]37]. The ANXA1-FPR2 axis modulates diverse pathological processes, including metabolism, inflammatory response, cell proliferation, and cell death [[194]19, [195]37–[196]40]. The role of ANXA1-FPR2 in AF has not been previously investigated. After analyzing the transcriptome dataset, we observed a significant upregulation of FPR2 in the atria of AF patients with obesity. Furthermore, FPR2 expression was also significantly increased in HFD-induced obese mice, which is consistent with previous studies [[197]18, [198]19, [199]41]. These findings suggest that FPR2 plays a crucial role in the pathology of AF and obesity-related AF. Interestingly, the expression level of ANXA1, which serves as an endogenous ligand for FPR2, exhibited opposite trends in AF patients and obese patients. This phenomenon has prompted us to conduct further investigations into the role of ANXA1-FPR2 axis in AF, and our findings have indicated that ANXA1-FPR2 exerts a protective effect against obesity-related AF. Although we initially employed the FPR2 blocker Boc-2 to investigate the impact of ANXA1-FPR2 blockade on AF susceptibility in obese mice, no statistically significant difference was observed. However, it did reveal that inhibiting the ANXA1-FPR2 signaling pathway augmented AF susceptibility and further exacerbated atrial fibrosis in obese mice. Therefore, we attribute the lack of statistical significance to a limited sample size. Based on our findings, we postulate that the down-regulation of ANXA1 expression in AF patients may create a conducive environment for the development of AF by exacerbating its pathological progression. This pathological change, in turn, could up-regulate FPR2 expression as a response to ANXA1 deficiency. Our hypothesis is that the reduction of ANXA1 expression in AF patients may be associated with decreased high density lipoprotein levels and impaired plasminogen/plasmin system, both of which can hinder ANXA1 synthesis [[200]42, [201]43]. However, further investigation is required to elucidate the exact mechanism. There is mounting evidence of an interaction between metabolism and AF. During AF episodes, the frequency of atrial electrical and contractile activity increases by 4 to 6 times [[202]44], leading to a significant rise in atrial energy and oxygen consumption that results in metabolic stress on atrial myocytes. Conversely, metabolic dysfunction of these cells can further exacerbate atrial remodeling and provide a substrate for the initiation and perpetuation of AF [[203]4, [204]5, [205]44]. Obesity is an independent risk factor for AF, partially attributable to metabolic disorders in the atria [[206]13]. Previous metabolomics studies have revealed that glucose metabolism is impaired, while fatty acid oxidation is predominant and abnormal lipid deposition occurs in atrial myocytes of obese mice. This phenomenon can be attributed to excessive intake of fatty acids and insufficient β-oxidation [[207]14]. Similarly, our previous study has demonstrated that insufficient fatty acid β-oxidation in the atrium of mice with HFD-induced obesity leads to excessive intracellular lipid accumulation and myocardial lipotoxicity, including oxidative stress, DNA damage, inflammation, and insulin intolerance. The aforementioned factors play a pivotal role in the development of cardiac hypertrophy, fibrosis, gap junction remodeling, and myocardial injury, thereby contributing to the initiation and progression of AF [[208]15]. The present study also revealed notable disturbances in glucose and lipid metabolism in obese mice, including impaired glucose tolerance, atrial tissue lipid accumulation, and oxidative stress injury. However, Ac2-26 was found to enhance the expression of receptors and regulators associated with glucose and lipid metabolism, ameliorate insulin resistance, mitigate atrial lipid accumulation, and alleviate oxidative stress injury in obese mice. Similarly, comparable outcomes were observed in NRCMs, and the impact of Ac2-26 was hindered by FPR2 blockers. Consistent with our findings, previous studies have also demonstrated the potential of ANXA1-FPR2 to enhance mitochondrial function and promote fatty acid β-oxidation. Additionally, therapeutic effects on lipotoxicity in diabetic mice have been observed with the use of ANXA1 mimetic peptide Ac2-26 [[209]19]. Furthermore, upregulation of ANXA1 significantly suppressed cigarette smoke extract-induced inflammation, apoptosis, and oxidative stress in BEAS-2B cells [[210]40]. The presented evidence suggests that the ANXA1-FPR2 axis exerts a protective role in obesity-related AF by augmenting metabolic status, thereby reducing cardiomyocyte lipotoxicity and associated oxidative stress. AMPK is an energy sensor, and its role in AF pathogenesis is attracting more and more attention. The previous studies have demonstrated that the deletion of liver kinase B1 (LKB1-AMPK), an upstream regulator of the AMPK superfamily, leads to spontaneous AF in mice. The findings suggest a strong association between atrial remodeling in AF and metabolic abnormalities as well as oxidative stress, both of which are closely linked to the LKB1-AMPK pathway [[211]45]. It is currently believed that the induction of metabolic stress by AF itself leads to a reduction in ATP levels and an increase in oxidative stress, ultimately resulting in atrial remodeling characterized by electrical abnormalities, systolic dysfunction, and cardiac structural remodeling [[212]7].The activation of AMPK can enhance glycolysis and fatty acid oxidation, thereby augmenting energy production while suppressing energy expenditure to rectify metabolic abnormalities in AF [[213]46, [214]47]. Our discovery that AMPK serves as a crucial downstream effector of the ANXA1-FPR2 signaling axis in combating obesity-related AF aligns with previous findings observed in other disease models [[215]19, [216]40]. Moreover, despite the absence of significant differences in body weight and insulin resistance between AAV9-shNC and AAV9-shAMPK group, treatment with Ac2-26 significantly attenuated susceptibility to AF in AAV9-shNC group compared to the AAV9-shAMPK group. This suggests that Ac2-26’s effect on improving susceptibility to AF in obese mice is not solely reliant on systemic metabolic changes. The activation of AMPK not only promotes Acc phosphorylation, thereby reducing intracellular lipid synthesis, but also activates PPARα and CPT1B, which accelerates fatty acid β-oxidation. The ultimate result was a decrease in lipotoxicity and oxidative stress in cardiomyocytes, as evidenced by a reduction in ROS production, upregulation of mitochondrial membrane potential, and enhancement of apoptosis. The well-established role of oxidative stress in triggering activity and potentially facilitating the transition from paroxysmal AF to persistent AF is widely recognized [[217]45]. Triggered activity arises from an elevated occurrence of spontaneous Ca^2+ release events (Ca^2+ leak) through RyR2 located in the sarcoplasmic reticulum (SR) [[218]48]. The function of RyR2 is increased by oxidative stress, either through direct oxidation of RyR2 or via an oxidation-mediated activation of Ca2+/calmodulin-dependent protein kinase-II (CaMKII), leading to hyperphosphorylation of RyR2 [[219]49]. The phosphorylation level of RyR2 was found to be significantly elevated in our obese mice, and it was notably reduced following Ac2-26 treatment, thereby providing further evidence for the potential protective role of Ac2-26 in AF by mitigating metabolic stress in obese mice. The obese mice exhibit atrial electrical and structural remodeling, characterized by the down-regulation of I[Na] and I[Ca, L] in cardiomyocytes, up-regulation of I[Kur], shortening of action potential duration (APD), and activation of the transforming growth factorβ (TGFβ)-mediated pro-fibrotic signaling pathway in the atria [[220]9, [221]50]. Our findings demonstrated a down-regulation of Nav1.5 ion channel transcriptional levels, responsible for I[Na], in the atria of obese mice; however, no significant difference was observed between the groups. Notably, Ac2-26 exhibited a remarkable ability to significantly up-regulate their expression in the atria of obese mice. The transcriptional levels of hERG and KATP channels were found to be upregulated in the atrium of obese mice. However, Ac2-26 only exhibited a reduction in the transcriptional level of hERG channel without exerting any significant effect on the transcriptional levels of other potassium channels. Additionally, a significant upregulation of Cav1.2 and Cav1.3 transcription levels was observed in obese mice, while Ac2-26 further augmented the expression of both Cav1.2 and Cav1.3. According to the transcriptional levels of ion channels, Ac2-26 appears to have a beneficial effect on prolonging APD in cardiomyocytes. However, since we have not investigated the functionality of ion channels specifically in cardiomyocytes, it is currently impossible to ascertain the precise impact of Ac2-26 on the functional characteristics of APD and ERP in these cells. Therefore, further studies are warranted to elucidate the role of ANXA1 in modulating ion channel function within cardiomyocytes. The previous studies have demonstrated that recombinant ANXA1 has the potential to ameliorate cardiac diastolic dysfunction in immune dysregulation by modulating the phenotypes of fibroblasts and immune cells within the cardiac tissue in animal models of rheumatoid arthritis [[222]38]. We also observed that ANXA1 in obese mice improved atrial fibrosis. However, since our study primarily focuses on myocardial cell metabolism, we did not further investigate the mechanism of myocardial fibrosis. Based on our findings, we hypothesize that ANXA1 may exert its anti-fibrotic effect by augmenting the cardiomyocytes’ anti-apoptotic response. However, further investigation is required to elucidate the precise mechanism underlying ANXA1’s anti-fibrotic action. Conclusions In conclusion, we have identified FPR2 as an obesity-associated AF hub gene through a comprehensive analysis of transcriptome data from AF and obesity patients. Notably, our study is the first to demonstrate that supplementation of FPR2 endogenous ligand Ac2-26 reduces AF susceptibility in obese mice. The detailed mechanism was investigated from the perspective of metabolic homeostasis, showing that Ac2-26 promotes atrial fatty acid metabolism and mitigates oxidative stress and lipotoxicity in obese mice by activating AMPK. Nevertheless, more functional experiments are warranted to provide deeper insights into the underlying mechanism and to support activating ANXA1-FPR2 axis as a clinical treatment strategy for AF in the future. Electronic Supplementary Material Below is the link to the electronic supplementary material. [223]Supplementary Material 1.^ (2MB, jpg) [224]Supplementary Material 2.^ (4.6MB, jpg) [225]Supplementary Material 3.^ (15.4KB, docx) [226]Supplementary Material 4.^ (13.4MB, xlsx) [227]Supplementary Material 5.^ (16.7KB, docx) [228]Supplementary Material 6.^ (415.7KB, xlsx) [229]Supplementary Material 7.^ (513.3KB, xlsx) [230]Supplementary Material 8.^ (10.6KB, xlsx) [231]Supplementary Material 9.^ (954KB, xlsx) [232]Supplementary Material 10.^ (15.1KB, docx) [233]Supplementary Material 11.^ (22.3KB, docx) Author contributions Q. Z., H. W. and X.Q. took responsibility for the study design. P. L., L. W., Y. W., J. F., Y.Z., H. L., B. F., L. J., H. G., Q. W., Y. F., B. F. and X. L. performed the experiments; P.L., L. W. and Y. W. analyzed the data. P. L. and L. W. prepared the manuscript. All the authors participated in drafting the manuscript and approved the final version for publication. Funding This work was supported by the National Natural Science Foundation of China (Grant Number 81870257, 31871172 and 82400379) and Key Research and Development Program of Shaanxi (grant number 2021SF-132). Availability of data and materials No datasets were generated or analysed during the current study. Declarations Competing interests The authors declare no competing interests. Footnotes Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. Peng Liu, Lu Wang and Yixin Wang contributed equallyto this work. Contributor Information Hongtao Wang, Email: wht506@126.com. Xinghua Qin, Email: xinghuaqin@nwpu.edu.cn. Qiangsun Zheng, Email: zhengqiangsun@126.com. References