1
|
Liu P, Wang L, Wang Y, Jin L, Gong H, Fan J, Zhang Y, Li H, Fu B, Wang Q, Fu Y, Fan B, Li X, Wang H, Qin X, Zheng Q. ANXA1-FPR2 axis mitigates the susceptibility to atrial fibrillation in obesity via rescuing AMPK activity in response to lipid overload. Cardiovasc Diabetol 2024; 23:452. [PMID: 39709478 DOI: 10.1186/s12933-024-02545-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 12/11/2024] [Indexed: 12/23/2024] Open
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.
Collapse
MESH Headings
- Animals
- Atrial Fibrillation/genetics
- Atrial Fibrillation/enzymology
- Atrial Fibrillation/prevention & control
- Atrial Fibrillation/metabolism
- Atrial Fibrillation/physiopathology
- Obesity/enzymology
- Obesity/metabolism
- Obesity/genetics
- AMP-Activated Protein Kinases/metabolism
- AMP-Activated Protein Kinases/genetics
- Humans
- Mice, Inbred C57BL
- Disease Models, Animal
- Annexin A1/metabolism
- Annexin A1/genetics
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Myocytes, Cardiac/drug effects
- Signal Transduction
- Male
- Receptors, Formyl Peptide/metabolism
- Receptors, Formyl Peptide/genetics
- Apoptosis/drug effects
- Diet, High-Fat
- Oxidative Stress/drug effects
- Receptors, Lipoxin/metabolism
- Receptors, Lipoxin/genetics
- Fibrosis
- Lipid Metabolism
- Databases, Genetic
- Mice
- Palmitic Acid/pharmacology
Collapse
Affiliation(s)
- Peng Liu
- Department of Cardiology, The Second Affiliate Hospital of Xi'an Jiaotong University, No.157, Xiwu Road, Xincheng District, Xi'an, 710004, Shaanxi, China
| | - Lu Wang
- Department of Endocrinology, The First Affiliate Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yixin Wang
- Department of Cardiology, The Second Affiliate Hospital of Xi'an Jiaotong University, No.157, Xiwu Road, Xincheng District, Xi'an, 710004, Shaanxi, China
| | - Linyan Jin
- Department of Cardiology, The Second Affiliate Hospital of Xi'an Jiaotong University, No.157, Xiwu Road, Xincheng District, Xi'an, 710004, Shaanxi, China
| | - Haoyu Gong
- Department of Cardiology, The Second Affiliate Hospital of Xi'an Jiaotong University, No.157, Xiwu Road, Xincheng District, Xi'an, 710004, Shaanxi, China
| | - Jiali Fan
- Department of Cardiology, The Second Affiliate Hospital of Xi'an Jiaotong University, No.157, Xiwu Road, Xincheng District, Xi'an, 710004, Shaanxi, China
| | - Yudi Zhang
- Department of Cardiology, Sir Run Run Shaw Hospital, Hangzhou, China
| | - Haiquan Li
- Department of Cardiology, The Second Affiliate Hospital of Xi'an Jiaotong University, No.157, Xiwu Road, Xincheng District, Xi'an, 710004, Shaanxi, China
| | - Bowen Fu
- Department of Cardiology, The Second Affiliate Hospital of Xi'an Jiaotong University, No.157, Xiwu Road, Xincheng District, Xi'an, 710004, Shaanxi, China
| | - Qiaozhu Wang
- Department of Cardiology, The Second Affiliate Hospital of Xi'an Jiaotong University, No.157, Xiwu Road, Xincheng District, Xi'an, 710004, Shaanxi, China
| | - Yuping Fu
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Boyuan Fan
- Department of Cardiology, The Second Affiliate Hospital of Xi'an Jiaotong University, No.157, Xiwu Road, Xincheng District, Xi'an, 710004, Shaanxi, China
| | - Xiaoli Li
- Department of Cardiology, The Second Affiliate Hospital of Xi'an Jiaotong University, No.157, Xiwu Road, Xincheng District, Xi'an, 710004, Shaanxi, China
| | - Hongtao Wang
- Department of Cardiology, The Second Affiliate Hospital of Xi'an Jiaotong University, No.157, Xiwu Road, Xincheng District, Xi'an, 710004, Shaanxi, China.
| | - Xinghua Qin
- Xi'an Key Laboratory of Special Medicine and Health Engineering, School of Life Sciences, Northwestern Polytechnical University, No. 127 Youyixi Road, Beilin District, Xi'an, 710072, Shaanxi, China.
| | - Qiangsun Zheng
- Department of Cardiology, The Second Affiliate Hospital of Xi'an Jiaotong University, No.157, Xiwu Road, Xincheng District, Xi'an, 710004, Shaanxi, China.
| |
Collapse
|
2
|
Wijdeveld LFJM, Collinet ACT, Huiskes FG, Brundel BJJM. Metabolomics in atrial fibrillation - A review and meta-analysis of blood, tissue and animal models. J Mol Cell Cardiol 2024; 197:108-124. [PMID: 39476947 DOI: 10.1016/j.yjmcc.2024.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 10/03/2024] [Accepted: 10/18/2024] [Indexed: 11/10/2024]
Abstract
BACKGROUND Atrial fibrillation (AF) is a highly prevalent cardiac arrhythmia associated with severe cardiovascular complications. AF presents a growing global challenge, however, current treatment strategies for AF do not address the underlying pathophysiology. To advance diagnosis and treatment of AF, a deeper understanding of AF root causes is needed. Metabolomics is a fast approach to identify, quantify and analyze metabolites in a given sample, such as human serum or atrial tissue. In the past two decades, metabolomics have enabled research on metabolite biomarkers to predict AF, metabolic features of AF, and testing metabolic mechanisms of AF in animal models. Due to the field's rapid evolution, the methods of AF metabolomics studies have not always been optimal. Metabolomics research has lacked standardization and requires expertise to face methodological challenges. PURPOSE OF THE REVIEW We summarize and meta-analyze metabolomics research on AF in human plasma and serum, atrial tissue, and animal models. We present the current progress on metabolic biomarkers candidates, metabolic features of clinical AF, and the translation of metabolomics findings from animal to human. We additionally discuss strengths and weaknesses of the metabolomics method and highlight opportunities for future AF metabolomics research.
Collapse
Affiliation(s)
- Leonoor F J M Wijdeveld
- Department of Physiology, Amsterdam UMC, Location Vrije Universiteit, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, 1081 HZ Amsterdam, the Netherlands; Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, MA 02142, Cambridge, United States
| | - Amelie C T Collinet
- Department of Physiology, Amsterdam UMC, Location Vrije Universiteit, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, 1081 HZ Amsterdam, the Netherlands
| | - Fabries G Huiskes
- Department of Physiology, Amsterdam UMC, Location Vrije Universiteit, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, 1081 HZ Amsterdam, the Netherlands
| | - Bianca J J M Brundel
- Department of Physiology, Amsterdam UMC, Location Vrije Universiteit, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, 1081 HZ Amsterdam, the Netherlands.
| |
Collapse
|
3
|
Stachteas P, Nasoufidou A, Karagiannidis E, Patoulias D, Karakasis P, Alexiou S, Samaras A, Zormpas G, Stavropoulos G, Tsalikakis D, Kassimis G, Papadopoulos C, Fragakis N. The Role of Sodium Glucose Co-Transporter 2 Inhibitors in Atrial Fibrillation: A Comprehensive Review. J Clin Med 2024; 13:5408. [PMID: 39336895 PMCID: PMC11431971 DOI: 10.3390/jcm13185408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/06/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Atrial fibrillation (AF) is the most prevalent arrhythmia among adults worldwide, frequently co-occurring with comorbidities such as Heart Failure (HF) and Type 2 Diabetes Mellitus (T2DM). This association contributes to increased morbidity and mortality, elevated healthcare costs, and diminished quality of life. Consequently, preventing or delaying the onset and recurrence of AF is crucial for reducing the incidence of complications. Sodium-glucose cotransporter 2 inhibitors (SGLT2is), due to their multifaceted pharmacological actions, have been proposed as potential therapeutic agents in the management of AF. However, current evidence from both animal models and clinical studies remains inconclusive. This narrative literature review aims to provide a comprehensive analysis of existing evidence on the impact of SGLT2is on the prevalence, incidence of new-onset, and recurrence of AF in diabetic populations and patients with HF. Numerous observational studies, predominantly retrospective, suggest a consistent reduction in AF risk with SGLT2is, while randomized controlled trials (RCTs) have yielded mixed results, with some demonstrating benefits and others not reaching statistical significance. The heterogeneity in study outcomes, population characteristics, follow-up duration, and specific SGLT2is used, as well as potential biases, underscore the need for further extensive and rigorous RCTs to establish definitive conclusions and elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Panagiotis Stachteas
- Second Cardiology Department, Medical School, Hippokration General Hospital, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Athina Nasoufidou
- Second Cardiology Department, Medical School, Hippokration General Hospital, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Efstratios Karagiannidis
- Second Cardiology Department, Medical School, Hippokration General Hospital, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Dimitrios Patoulias
- Outpatient Department of Cardiometabolic Medicine, Second Cardiology Department, Medical School, Hippokration General Hospital, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Paschalis Karakasis
- Second Cardiology Department, Medical School, Hippokration General Hospital, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Sophia Alexiou
- Second Cardiology Department, Medical School, Hippokration General Hospital, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Athanasios Samaras
- Second Cardiology Department, Medical School, Hippokration General Hospital, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Georgios Zormpas
- Second Cardiology Department, Medical School, Hippokration General Hospital, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - George Stavropoulos
- Second Cardiology Department, Medical School, Hippokration General Hospital, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Dimitrios Tsalikakis
- Department of Electrical and Computer Engineering, University of Western Macedonia, 50100 Kozani, Greece
| | - George Kassimis
- Second Cardiology Department, Medical School, Hippokration General Hospital, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Christodoulos Papadopoulos
- Third Cardiology Department, Medical School, Hippokration General Hospital, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Nikolaos Fragakis
- Second Cardiology Department, Medical School, Hippokration General Hospital, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Outpatient Department of Cardiometabolic Medicine, Second Cardiology Department, Medical School, Hippokration General Hospital, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| |
Collapse
|
4
|
Toksoy Z, Ma Y, Goedeke L, Li W, Hu X, Wu X, Cacheux M, Liu Y, Akar FG, Shulman GI, Young LH. Role of AMPK in Atrial Metabolic Homeostasis and Substrate Preference. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.29.608789. [PMID: 39257756 PMCID: PMC11383699 DOI: 10.1101/2024.08.29.608789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Atrial fibrillation is the most common clinical arrhythmia and may be due in part to metabolic stress. Atrial specific deletion of the master metabolic sensor, AMP-activated protein kinase (AMPK), induces atrial remodeling culminating in atrial fibrillation in mice, implicating AMPK signaling in the maintenance of atrial electrical and structural homeostasis. However, atrial substrate preference for mitochondrial oxidation and the role of AMPK in regulating atrial metabolism are unknown. Here, using LC-MS/MS methodology combined with infusions of [ 13 C 6 ]glucose and [ 13 C 4 ]β-hydroxybutyrate in conscious mice, we demonstrate that conditional deletion of atrial AMPK catalytic subunits shifts mitochondrial atrial metabolism away from fatty acid oxidation and towards pyruvate oxidation. LC-MS/MS-based quantification of acyl-CoAs demonstrated decreased atrial tissue content of long-chain fatty acyl-CoAs. Proteomic analysis revealed a broad downregulation of proteins responsible for fatty acid uptake (LPL, CD36, FABP3), acylation and oxidation. Atrial AMPK deletion reduced expression of atrial PGC1-α and downstream PGC1-α/PPARα/RXR regulated gene transcripts. In contrast, atrial [ 14 C]2-deoxyglucose uptake and GLUT1 expression increased with fasting in mice with AMPK deletion, while the expression of glycolytic enzymes exhibited heterogenous changes. Thus, these results highlight the crucial homeostatic role of AMPK in the atrium, with loss of atrial AMPK leading to downregulation of the PGC1-α/PPARα pathway and broad metabolic reprogramming with a loss of fatty acid oxidation, which may contribute to atrial remodeling and arrhythmia.
Collapse
|
5
|
Peng H, Yuan J, Wang Z, Mo B, Wang Y, Wang Y, Wang Q. NR4A3 prevents diabetes induced atrial cardiomyopathy by maintaining mitochondrial energy metabolism and reducing oxidative stress. EBioMedicine 2024; 106:105268. [PMID: 39098108 PMCID: PMC11334830 DOI: 10.1016/j.ebiom.2024.105268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 07/18/2024] [Accepted: 07/23/2024] [Indexed: 08/06/2024] Open
Abstract
BACKGROUND Atrial cardiomyopathy (ACM) is responsible for atrial fibrillation (AF) and thromboembolic events. Diabetes mellitus (DM) is an important risk factor for ACM. However, the potential mechanism between ACM and DM remains elusive. METHODS Atrial tissue samples were obtained from patients diagnosed with AF or sinus rhythm (SR) to assess alterations in NR4A3 expression, and then two distinct animal models were generated by subjecting Nr4a3-/- mice and WT mice to a high-fat diet (HFD) and Streptozotocin (STZ), while db/db mice were administered AAV9-Nr4a3 or AAV9-ctrl. Subsequently, in vivo and in vitro experiments were conducted to assess the impact of NR4A3 on diabetes-induced atrial remodeling through electrophysiological, biological, and histological analyses. RNA sequencing (RNA-seq) and metabolomics analysis were employed to unravel the downstream mechanisms. FINDINGS The expression of NR4A3 was significantly decreased in atrial tissues of both AF patients and diabetic mice compared to their respective control groups. NR4A3 deficiency exacerbated atrial hypertrophy and atrial fibrosis, and increased susceptibility to pacing-induced AF. Conversely, overexpression of NR4A3 alleviated atrial structural remodeling and reduced AF induction rate. Mechanistically, we confirmed that NR4A3 improves mitochondrial energy metabolism and reduces oxidative stress injury by preserving the transcriptional expression of Sdha, thereby exerting a protective influence on atrial remodeling induced by diabetes. INTERPRETATION Our data confirm that NR4A3 plays a protective role in atrial remodeling caused by diabetes, so it may be a new target for treating ACM. FUNDING This study was supported by the major research program of National Natural Science Foundation of China (NSFC) No: 82370316 (to Q-S. W.), No. 81974041 (to Y-P. W.), and No. 82270447 (to Y-P. W.) and Fundation of Shanghai Hospital Development Center (No. SHDC2022CRD044 to Q-S. W.).
Collapse
Affiliation(s)
- Hong Peng
- Department of Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Jiali Yuan
- Department of Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Zhengshuai Wang
- Department of Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Binfeng Mo
- Department of Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yihui Wang
- The Department of Radiology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yuepeng Wang
- Department of Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Qunshan Wang
- Department of Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
6
|
Tang X, Wang J, Ouyang X, Chen Q, Dong R, Luo Y, Zhong J, Huang Z, Peng L, Xie X, Zhu J, Zheng Z, Li S. Coronary Sinus Metabolite 12,13-diHOME Is a Novel Biomarker for Left Atrial Remodeling in Patients With Atrial Fibrillation. Circ Arrhythm Electrophysiol 2024; 17:e012486. [PMID: 38690652 DOI: 10.1161/circep.123.012486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 03/22/2024] [Indexed: 05/02/2024]
Abstract
BACKGROUND 12,13-dihydroxy-9Z-octadecenoic acid (12,13-diHOME) has shown potential in protecting against heart disease, but its relationship with atrial fibrillation (AF) remains unknown. METHODS Coronary sinus (CS) and femoral vein blood samplings were synchronously collected from AF and non-AF subjects (paroxysmal supraventricular tachycardia or idiopathic premature ventricular complexes) who underwent catheter ablation. First, untargeted metabolomic profiling was performed in a discovery cohort (including 12 AF and 12 non-AF subjects) to identify the most promising CS or femoral vein metabolite. Then, the selected metabolite was further measured in a validation cohort (including 119 AF and 103 non-AF subjects) to confirm its relationship with left atrium remodeling and 1-year postablation recurrence of AF. Finally, the biological function of the selected metabolite was validated in a rapid-paced cultured HL-1 atrial cardiomyocytes model. RESULTS Metabolomic analysis identified CS 12,13-diHOME as the most pronounced change metabolite correlated with left atrium remodeling in the discovery cohort. In the validation cohort, CS 12,13-diHOME was significantly lower in patients with AF than non-AF controls (84.32±20.13 versus 96.24±23.56 pg/mL; P<0.01), and associated with worse structural, functional, and electrical remodeling of left atrium. Multivariable regression analyses further demonstrated that decreased CS 12,13-diHOME was an independent predictor of 1-year postablation recurrence of AF (odds ratio, 0.754 [95% CI, 0.648-0.920]; P=0.005). Biological function validations showed that 12,13-diHOME treatment significantly protect the cell viability, improved the expression of MHC (myosin heavy chain) and Cav1.2 (L-type calcium channel α1c), and attenuated mitochondrial damage in the rapid-paced cultured HL-1 cardiomyocytes model. CONCLUSIONS CS metabolite 12,13-diHOME is decreased in patients with AF and can serve as a novel biomarker for left atrium remodeling.
Collapse
Affiliation(s)
- Xixiang Tang
- VIP Medical Service Center (X.T.), the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiafu Wang
- Department of Cardiovascular Medicine (J.W., X.O., Q.C., R.D., Y.L., Z.H., L.P., X.X., J.Z., Z.Z., S.L.), the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaolan Ouyang
- Department of Cardiovascular Medicine (J.W., X.O., Q.C., R.D., Y.L., Z.H., L.P., X.X., J.Z., Z.Z., S.L.), the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qian Chen
- Department of Cardiovascular Medicine (J.W., X.O., Q.C., R.D., Y.L., Z.H., L.P., X.X., J.Z., Z.Z., S.L.), the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ruimin Dong
- Department of Cardiovascular Medicine (J.W., X.O., Q.C., R.D., Y.L., Z.H., L.P., X.X., J.Z., Z.Z., S.L.), the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yanting Luo
- Department of Cardiovascular Medicine (J.W., X.O., Q.C., R.D., Y.L., Z.H., L.P., X.X., J.Z., Z.Z., S.L.), the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Junlin Zhong
- Department of Cardiovascular Medicine (J.W., X.O., Q.C., R.D., Y.L., Z.H., L.P., X.X., J.Z., Z.Z., S.L.), the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhuoshan Huang
- Department of Cardiovascular Medicine (J.W., X.O., Q.C., R.D., Y.L., Z.H., L.P., X.X., J.Z., Z.Z., S.L.), the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Long Peng
- Department of Cardiovascular Medicine (J.W., X.O., Q.C., R.D., Y.L., Z.H., L.P., X.X., J.Z., Z.Z., S.L.), the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xujing Xie
- Department of Cardiovascular Medicine (J.W., X.O., Q.C., R.D., Y.L., Z.H., L.P., X.X., J.Z., Z.Z., S.L.), the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jieming Zhu
- Department of Ultrasonography (J.Z.), the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhenda Zheng
- Department of Cardiovascular Medicine (J.W., X.O., Q.C., R.D., Y.L., Z.H., L.P., X.X., J.Z., Z.Z., S.L.), the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Suhua Li
- Department of Cardiovascular Medicine (J.W., X.O., Q.C., R.D., Y.L., Z.H., L.P., X.X., J.Z., Z.Z., S.L.), the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
7
|
Cao Y, Cui L, Tuo S, Liu H, Cui S. Resveratrol mediates mitochondrial function through the sirtuin 3 pathway to improve abnormal metabolic remodeling in atrial fibrillation. Eur J Histochem 2024; 68:4004. [PMID: 38656259 PMCID: PMC11064893 DOI: 10.4081/ejh.2024.4004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 04/09/2024] [Indexed: 04/26/2024] Open
Abstract
This study investigated the impact of resveratrol on abnormal metabolic remodeling in atrial fibrillation (AF) and explored potential molecular mechanisms. An AF cell model was established by high-frequency electrical stimulation of HL-1 atrial muscle cells. Resveratrol concentrations were optimized using CCK-8 and flow cytometry. AF-induced increases in ROS and mitochondrial calcium, along with decreased adenosine triphosphate (ATP) and mitochondrial membrane potential, were observed. Resveratrol mitigated these changes and maintained normal mitochondrial morphology. Moreover, resveratrol acted through the SIRT3-dependent pathway, as evidenced by its ability to suppress AF-induced acetylation of key metabolic enzymes. SIRT3 overexpression controls acetylation modifications, suggesting its regulatory role. In conclusion, resveratrol's SIRT3-dependent pathway intervenes in AF-induced mitochondrial dysfunction, presenting a potential therapeutic avenue for AF-related metabolic disorders. This study sheds light on the role of resveratrol in mitigating AF-induced mitochondrial remodeling and highlights its potential as a novel treatment for AF.
Collapse
Affiliation(s)
- Yuejuan Cao
- Department of Cardiology, Tianjin Union Medical Center, Tianjin.
| | - Li Cui
- Department of Cardiology, Tianjin Union Medical Center, Tianjin.
| | - Shaoyong Tuo
- Department of Vascular Surgery, Tianjin Union Medical Center, Tianjin.
| | - Hongze Liu
- Department of Cardiology, Tianjin Union Medical Center, Tianjin.
| | - Shaonan Cui
- Department of Cardiology, Tianjin Union Medical Center, Tianjin.
| |
Collapse
|
8
|
Tubeeckx MRL, De Keulenaer GW, Heidbuchel H, Segers VFM. Pathophysiology and clinical relevance of atrial myopathy. Basic Res Cardiol 2024; 119:215-242. [PMID: 38472506 DOI: 10.1007/s00395-024-01038-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 02/01/2024] [Accepted: 02/02/2024] [Indexed: 03/14/2024]
Abstract
Atrial myopathy is a condition that consists of electrical, structural, contractile, and autonomic remodeling of the atria and is the substrate for development of atrial fibrillation, the most common arrhythmia. Pathophysiologic mechanisms driving atrial myopathy are inflammation, oxidative stress, atrial stretch, and neurohormonal signals, e.g., angiotensin-II and aldosterone. These mechanisms initiate the structural and functional remodeling of the atrial myocardium. Novel therapeutic strategies are being developed that target the pathophysiologic mechanisms of atrial myopathy. In this review, we will discuss the pathophysiology of atrial myopathy, as well as diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Michiel R L Tubeeckx
- Laboratory of Physiopharmacology, Universiteitsplein 1, Building T (2nd Floor), 2610, Antwerp, Belgium.
| | - Gilles W De Keulenaer
- Laboratory of Physiopharmacology, Universiteitsplein 1, Building T (2nd Floor), 2610, Antwerp, Belgium
- Department of Cardiology, ZNA Middelheim Hospital Antwerp, Antwerp, Belgium
| | - Hein Heidbuchel
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, University Hospital Antwerp, Antwerp, Belgium
| | - Vincent F M Segers
- Laboratory of Physiopharmacology, Universiteitsplein 1, Building T (2nd Floor), 2610, Antwerp, Belgium
- Department of Cardiology, University Hospital Antwerp, Antwerp, Belgium
| |
Collapse
|
9
|
Huang Y, Lin Q, Zhou Y, Zhu J, Ma Y, Wu K, Ning Z, Zhang Z, Liu N, Li M, Liu Y, Tu T, Liu Q. Amino acid profile alteration in age-related atrial fibrillation. J Transl Med 2024; 22:259. [PMID: 38461346 PMCID: PMC10925006 DOI: 10.1186/s12967-024-05028-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 02/24/2024] [Indexed: 03/11/2024] Open
Abstract
BACKGROUND Amino acids (AAs) are one of the primary metabolic substrates for cardiac work. The correlation between AAs and both atrial fibrillation (AF) and aging has been documented. However, the relationship between AAs and age-related AF remains unclear. METHODS Initially, the plasma AA levels of persistent AF patients and control subjects were assessed, and the correlations between AA levels, age, and other clinical indicators were explored. Subsequently, the age-related AF mouse model was constructed and the untargeted myocardial metabolomics was conducted to detect the level of AAs and related metabolites. Additionally, the gut microbiota composition associated with age-related AF was detected by a 16S rDNA amplicon sequencing analysis on mouse fecal samples. RESULTS Higher circulation levels of lysine (Student's t-test, P = 0.001), tyrosine (P = 0.002), glutamic acid (P = 0.008), methionine (P = 0.008), and isoleucine (P = 0.014), while a lower level of glycine (P = 0.003) were observed in persistent AF patients. The feature AAs identified by machine learning algorithms were glutamic acid and methionine. The association between AAs and age differs between AF and control subjects. Distinct patterns of AA metabolic profiles were observed in the myocardial metabolites of aged AF mice. Aged AF mice had lower levels of Betaine, L-histidine, L-alanine, L-arginine, L-Pyroglutamic acid, and L-Citrulline compared with adult AF mice. Aged AF mice also presented a different gut microbiota pattern, and its functional prediction analysis showed AA metabolism alteration. CONCLUSION This study provided a comprehensive network of AA disturbances in age-related AF from multiple dimensions, including plasma, myocardium, and gut microbiota. Disturbances of AAs may serve as AF biomarkers, and restoring their homeostasis may have potential benefits for the management of age-related AF.
Collapse
Affiliation(s)
- Yunying Huang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, 410011, Hunan, People's Republic of China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, 410011, Hunan, People's Republic of China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, 410011, Hunan, People's Republic of China
| | - Qiuzhen Lin
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, 410011, Hunan, People's Republic of China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, 410011, Hunan, People's Republic of China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, 410011, Hunan, People's Republic of China
| | - Yong Zhou
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, 410011, Hunan, People's Republic of China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, 410011, Hunan, People's Republic of China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, 410011, Hunan, People's Republic of China
| | - Jiayi Zhu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, 410011, Hunan, People's Republic of China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, 410011, Hunan, People's Republic of China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, 410011, Hunan, People's Republic of China
| | - Yingxu Ma
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, 410011, Hunan, People's Republic of China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, 410011, Hunan, People's Republic of China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, 410011, Hunan, People's Republic of China
| | - Keke Wu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, 410011, Hunan, People's Republic of China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, 410011, Hunan, People's Republic of China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, 410011, Hunan, People's Republic of China
| | - Zuodong Ning
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, 410011, Hunan, People's Republic of China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, 410011, Hunan, People's Republic of China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, 410011, Hunan, People's Republic of China
| | - Zixi Zhang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, 410011, Hunan, People's Republic of China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, 410011, Hunan, People's Republic of China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, 410011, Hunan, People's Republic of China
| | - Na Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, 410011, Hunan, People's Republic of China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, 410011, Hunan, People's Republic of China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, 410011, Hunan, People's Republic of China
| | - Mohan Li
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, 410011, Hunan, People's Republic of China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, 410011, Hunan, People's Republic of China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, 410011, Hunan, People's Republic of China
- Department of Geriatrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100730, People's Republic of China
| | - Yaozhong Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, 410011, Hunan, People's Republic of China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, 410011, Hunan, People's Republic of China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, 410011, Hunan, People's Republic of China
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, USA
| | - Tao Tu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China.
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, 410011, Hunan, People's Republic of China.
- Cardiovascular Disease Research Center of Hunan Province, Changsha, 410011, Hunan, People's Republic of China.
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, 410011, Hunan, People's Republic of China.
| | - Qiming Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, 410011, Hunan, People's Republic of China.
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, 410011, Hunan, People's Republic of China.
- Cardiovascular Disease Research Center of Hunan Province, Changsha, 410011, Hunan, People's Republic of China.
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, 410011, Hunan, People's Republic of China.
| |
Collapse
|
10
|
Abdel-Reheim MA, Nomier Y, Zaki MB, Abulsoud AI, Mohammed OA, Rashad AA, Oraby MA, Elballal MS, Tabaa MME, Elazazy O, Abd-Elmawla MA, El-Dakroury WA, Abdel Mageed SS, Abdelmaksoud NM, Elrebehy MA, Helal GK, Doghish AS. Unveiling the regulatory role of miRNAs in stroke pathophysiology and diagnosis. Pathol Res Pract 2024; 253:155085. [PMID: 38183822 DOI: 10.1016/j.prp.2023.155085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 12/28/2023] [Accepted: 12/30/2023] [Indexed: 01/08/2024]
Abstract
Stroke, a major global cause of mortality, leads to a range of problems for those who survive. Besides its brutal events, stroke also tends to have a characteristic of recurrence, making it a complex disease involving intricate regulatory networks. One of the major cellular regulators is the non-coding RNAs (ncRNA), specifically microRNAs (miRNAs), thus the possible functions of miRNAs in the pathogenesis of stroke are discussed as well as the possibility of using miRNA-based therapeutic approaches. Firstly, the molecular mechanisms by which miRNAs regulate vital physiological processes, including synaptic plasticity, oxidative stress, apoptosis, and the integrity of the blood-brain barrier (BBB) are reviewed. The miRNA indirectly impacts stroke outcomes by regulating BBB function and angiogenesis through the targeting of transcription factors and angiogenic factors. In addition, the tendency for some miRNAs to be upregulated in response to hypoxia, which is a prevalent phenomenon in stroke and various neurological disorders, highlights the possibility that it controls hypoxia-inducible factor (HIF) signaling and angiogenesis, thereby influencing the integrity of the BBB as examples of the discussed mechanisms. Furthermore, this review explores the potential therapeutic targets that miRNAs may offer for stroke recovery and highlights their promising capacity to alleviate post-stroke complications. This review provides researchers and clinicians with valuable resources since it attempts to decipher the complex network of miRNA-mediated mechanisms in stroke. Additionally, the review addresses the interplay between miRNAs and stroke risk factors as well as clinical applications of miRNAs as diagnostic and prognostic markers.
Collapse
Affiliation(s)
- Mustafa Ahmed Abdel-Reheim
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef 62521, Egypt.
| | - Yousra Nomier
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and health sciences, Sultan Qaboos University, Muscat, Oman
| | - Mohamed Bakr Zaki
- Biochemistry, Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Menoufia 32897, Egypt
| | - Ahmed I Abulsoud
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt; Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia
| | - Ahmed A Rashad
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Mamdouh A Oraby
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Mohammed S Elballal
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Manar Mohammed El Tabaa
- Pharmacology & Environmental Toxicology, Environmental Studies & Research Institute (ESRI), University of Sadat City, Sadat City 32897, Menoufia, Egypt
| | - Ola Elazazy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Mai A Abd-Elmawla
- Biochemistry, Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Walaa A El-Dakroury
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | | | - Mahmoud A Elrebehy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Gouda Kamel Helal
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo 11231, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt.
| |
Collapse
|
11
|
Krause J, Nickel A, Madsen A, Aitken-Buck HM, Stoter AMS, Schrapers J, Ojeda F, Geiger K, Kern M, Kohlhaas M, Bertero E, Hofmockel P, Hübner F, Assum I, Heinig M, Müller C, Hansen A, Krause T, Park DD, Just S, Aïssi D, Börnigen D, Lindner D, Friedrich N, Alhussini K, Bening C, Schnabel RB, Karakas M, Iacoviello L, Salomaa V, Linneberg A, Tunstall-Pedoe H, Kuulasmaa K, Kirchhof P, Blankenberg S, Christ T, Eschenhagen T, Lamberts RR, Maack C, Stenzig J, Zeller T. An arrhythmogenic metabolite in atrial fibrillation. J Transl Med 2023; 21:566. [PMID: 37620858 PMCID: PMC10464005 DOI: 10.1186/s12967-023-04420-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 08/07/2023] [Indexed: 08/26/2023] Open
Abstract
BACKGROUND Long-chain acyl-carnitines (ACs) are potential arrhythmogenic metabolites. Their role in atrial fibrillation (AF) remains incompletely understood. Using a systems medicine approach, we assessed the contribution of C18:1AC to AF by analysing its in vitro effects on cardiac electrophysiology and metabolism, and translated our findings into the human setting. METHODS AND RESULTS Human iPSC-derived engineered heart tissue was exposed to C18:1AC. A biphasic effect on contractile force was observed: short exposure enhanced contractile force, but elicited spontaneous contractions and impaired Ca2+ handling. Continuous exposure provoked an impairment of contractile force. In human atrial mitochondria from AF individuals, C18:1AC inhibited respiration. In a population-based cohort as well as a cohort of patients, high C18:1AC serum concentrations were associated with the incidence and prevalence of AF. CONCLUSION Our data provide evidence for an arrhythmogenic potential of the metabolite C18:1AC. The metabolite interferes with mitochondrial metabolism, thereby contributing to contractile dysfunction and shows predictive potential as novel circulating biomarker for risk of AF.
Collapse
Affiliation(s)
- Julia Krause
- University Center of Cardiovascular Science, Department of Cardiology, University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Alexander Nickel
- Comprehensive Heart Failure Center, University Clinic Würzburg, Würzburg, Germany
| | - Alexandra Madsen
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hamish M Aitken-Buck
- Department of Physiology, HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - A M Stella Stoter
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jessica Schrapers
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Francisco Ojeda
- Department of Cardiology, University Heart and Vascular Center Hamburg, Hamburg, Germany
| | - Kira Geiger
- Comprehensive Heart Failure Center, University Clinic Würzburg, Würzburg, Germany
| | - Melanie Kern
- Comprehensive Heart Failure Center, University Clinic Würzburg, Würzburg, Germany
| | - Michael Kohlhaas
- Comprehensive Heart Failure Center, University Clinic Würzburg, Würzburg, Germany
| | - Edoardo Bertero
- Comprehensive Heart Failure Center, University Clinic Würzburg, Würzburg, Germany
| | - Patrick Hofmockel
- Comprehensive Heart Failure Center, University Clinic Würzburg, Würzburg, Germany
| | - Florian Hübner
- Institute of Food Chemistry, University of Münster, Münster, Germany
| | - Ines Assum
- Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
- Department of Informatics, Technical University Munich, Munich, Germany
| | - Matthias Heinig
- Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
- Department of Informatics, Technical University Munich, Munich, Germany
| | - Christian Müller
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
- Department of Cardiology, University Heart and Vascular Center Hamburg, Hamburg, Germany
| | - Arne Hansen
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias Krause
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Deung-Dae Park
- Molecular Cardiology, Department of Internal Medicine II, University of Ulm, Ulm, Germany
| | - Steffen Just
- Molecular Cardiology, Department of Internal Medicine II, University of Ulm, Ulm, Germany
| | - Dylan Aïssi
- Department of Cardiology, University Heart and Vascular Center Hamburg, Hamburg, Germany
| | - Daniela Börnigen
- Department of Cardiology, University Heart and Vascular Center Hamburg, Hamburg, Germany
| | - Diana Lindner
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
- Department of Cardiology, University Heart and Vascular Center Hamburg, Hamburg, Germany
- Department of Cardiology and Angiology, Faculty of Medicine, University Heart Center Freiburg-Bad Krozingen, Medical Center - University of Freiburg, University of Freiburg, 79106, Freiburg, Germany
| | - Nele Friedrich
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Khaled Alhussini
- Department of Thoracic and Cardiovascular Surgery, University Clinic Würzburg, Würzburg, Germany
| | - Constanze Bening
- Department of Thoracic and Cardiovascular Surgery, University Clinic Würzburg, Würzburg, Germany
| | - Renate B Schnabel
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
- Department of Cardiology, University Heart and Vascular Center Hamburg, Hamburg, Germany
| | - Mahir Karakas
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Licia Iacoviello
- Department of Epidemiology and Prevention, IRCCS Neuromed, Pozzilli, Italy
- Department of Medicine and Surgery, Research Center in Epidemiology and Preventive Medicine (EPIMED), University of Insubria, Varese, Italy
| | - Veikko Salomaa
- Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Allan Linneberg
- Center for Clinical Research and Prevention, Bispebjerg and Frederiksberg Hospital, Capital Region of Denmark, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hugh Tunstall-Pedoe
- Cardiovascular Epidemiology Unit, Institute of Cardiovascular Research, University of Dundee, Dundee, UK
| | - Kari Kuulasmaa
- Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Paulus Kirchhof
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
- Department of Cardiology, University Heart and Vascular Center Hamburg, Hamburg, Germany
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Stefan Blankenberg
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
- Department of Cardiology, University Heart and Vascular Center Hamburg, Hamburg, Germany
| | - Torsten Christ
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Eschenhagen
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Regis R Lamberts
- Department of Physiology, HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Christoph Maack
- Comprehensive Heart Failure Center, University Clinic Würzburg, Würzburg, Germany
| | - Justus Stenzig
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tanja Zeller
- University Center of Cardiovascular Science, Department of Cardiology, University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany.
| |
Collapse
|
12
|
Chakraborty P, Farhat K, Po SS, Armoundas AA, Stavrakis S. Autonomic Nervous System and Cardiac Metabolism: Links Between Autonomic and Metabolic Remodeling in Atrial Fibrillation. JACC Clin Electrophysiol 2023:S2405-500X(23)00117-2. [PMID: 37086229 DOI: 10.1016/j.jacep.2023.02.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/27/2023] [Accepted: 02/16/2023] [Indexed: 04/23/2023]
Abstract
Simultaneous activation of the sympathetic and parasympathetic nervous systems is crucial for the initiation of paroxysmal atrial fibrillation (AF). However, unbalanced activation of the sympathetic system is characteristic of autonomic remodeling in long-standing persistent AF. Moreover, the adrenergic activation-induced metabolic derangements provide a milieu for acute AF and promote the transition from the paroxysmal to the persistent phase of AF. On the other hand, cholinergic activation ameliorates the maladaptive metabolic remodeling in the face of metabolic challenges. Selective inhibition of the sympathetic system and restoration of the balance of the cholinergic system by neuromodulation is emerging as a novel nonpharmacologic strategy for managing AF. This review explores the link between cardiac autonomic and metabolic remodeling and the potential roles of different autonomic modulation strategies on atrial metabolic aberrations in AF.
Collapse
Affiliation(s)
- Praloy Chakraborty
- Cardiovascular Section, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Kassem Farhat
- Cardiovascular Section, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Sunny S Po
- Cardiovascular Section, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Antonis A Armoundas
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA; Broad Institute, Massachusetts Institute of Technology, Boston, Massachusetts, USA
| | - Stavros Stavrakis
- Cardiovascular Section, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA.
| |
Collapse
|
13
|
Impact of catheter ablation and subsequent recurrence of atrial fibrillation on glucose status in patients undergoing continuous glucose monitoring. Sci Rep 2023; 13:4299. [PMID: 36922617 PMCID: PMC10017667 DOI: 10.1038/s41598-023-31139-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 03/07/2023] [Indexed: 03/18/2023] Open
Abstract
Although glucose metabolism and atrial fibrillation (AF) have complex interrelationships, the impact of catheter ablation of AF on glucose status has not been well evaluated. Continuous glucose monitoring (CGM) with a FreeStyle Libre Pro (Abbott) was performed for 48 h pre-procedure, during the procedure, and for 72 h post-procedure in 58 non-diabetes mellitus (DM) patients with symptomatic AF and 20 patients with supraventricular or ventricular arrhythmias as a control group. All ablation procedures including pulmonary vein isolation were performed successfully. Glucose levels during procedures consistently increased in the AF and control groups (83.1 ± 16.1 to 110.0 ± 20.5 mg/dL and 83.3 ± 14.7 to 98.6 ± 16.3 mg/dL, respectively, P < 0.001 for both), and Δ glucose levels (max minus min/procedure) were greater in the AF group than control group (P < 0.001). There was a trend toward higher mean glucose levels at 72 h after the procedures compared with those before the procedures in both the AF and control groups (from 103.4 ± 15.6 to 106.1 ± 13.0 mg/dL, P = 0.063 and from 100.2 ± 17.1 to 102.9 ± 16.9 mg/dL, P = 0.052). An acute increase in glucose level at the time of early AF recurrence (N = 9, 15.5%) could be detected by simultaneous CGM and ECG monitoring (89.7 ± 18.0 to 108.3 ± 30.5 mg/dL, P = 0.001). In conclusion, although AF ablation caused a statistically significant increase in the glucose levels during the procedures, it did not result in a pathologically significant change after ablation in non-DM patients. Simultaneous post-procedure CGM and ECG monitoring alerted us to possible acute increases in glucose levels at the onset of AF recurrence.
Collapse
|
14
|
Long non-coding RNA and circular RNA: new perspectives for molecular pathophysiology of atrial fibrillation. Mol Biol Rep 2023; 50:2835-2845. [PMID: 36596997 DOI: 10.1007/s11033-022-08216-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 12/15/2022] [Indexed: 01/05/2023]
Abstract
Many studies have demonstrated the association of atrial fibrillation (AF) with endogenous genetic regulatory mechanisms. These interactions could advance the understanding of the AF pathophysiological process, supporting the search for early biomarkers to improve diagnosis and disease monitoring. Among the endogenous genetic regulatory mechanisms, long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) have gained special attention, and studies have demonstrated their involvement in AF development and other AF-related diseases such as coronary artery disease and cardiomyopathy. This review describes the main experimental results reported by studies that analyzed the expression of lncRNAs and circRNAs in AF associated with miRNA or mRNA. The search was conducted in PubMed public database using the terms "lncRNA and atrial fibrillation" or "long ncRNA and atrial fibrillation" or "long non-coding RNA and atrial fibrillation" or "circular RNA and atrial fibrillation" or "circRNA and atrial fibrillation". There was no overlapping of lncRNA or circRNA among the studies, attributed to the different sample types, methods, species, and patient classification evaluated in these studies. Although the regulatory mechanisms in which these molecules are involved are not yet well understood, the studies analyzed show their importance in the pathophysiological process of AF, supporting the idea that lncRNAs and circRNAs are involved in miRNA or mRNA regulation in the molecular mechanism of this disease.
Collapse
|
15
|
Lage R, Cebro-Márquez M, Vilar-Sánchez ME, González-Melchor L, García-Seara J, Martínez-Sande JL, Fernández-López XA, Aragón-Herrera A, Martínez-Monzonís MA, González-Juanatey JR, Rodríguez-Mañero M, Moscoso I. Circulating miR-451a Expression May Predict Recurrence in Atrial Fibrillation Patients after Catheter Pulmonary Vein Ablation. Cells 2023; 12:cells12040638. [PMID: 36831306 PMCID: PMC9953933 DOI: 10.3390/cells12040638] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/07/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
Atrial fibrillation is the most prevalent tachyarrhythmia in clinical practice, with very high cardiovascular morbidity and mortality with a high-cost impact in health systems. Currently, it is one of the main causes of stroke and subsequent heart failure and sudden death. miRNAs mediate in several processes involved in cardiovascular disease, including fibrosis and electrical and structural remodeling. Several studies suggest a key role of miRNAs in the course and maintenance of atrial fibrillation. In our study, we aimed to identify the differential expression of circulating miRNAs and their predictive value as biomarkers of recurrence in atrial fibrillation patients undergoing catheter pulmonary vein ablation. To this effect, 42 atrial fibrillation patients were recruited for catheter ablation. We measured the expression of 84 miRNAs in non-recurrent and recurrent groups (45.2%), both in plasma from peripheral and left atrium blood. Expression analysis showed that miRNA-451a is downregulated in recurrent patients. Receiver operating characteristic curve analysis showed that miR-451a in left atrium plasma could predict atrial fibrillation recurrence after pulmonary vein isolation. In addition, atrial fibrillation recurrence is positively associated with the increment of scar percentage. Our data suggest that miRNA-451a expression plays an important role in AF recurrence by controlling fibrosis and progression.
Collapse
Affiliation(s)
- Ricardo Lage
- Cardiology Group, Centre for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - María Cebro-Márquez
- Cardiology Group, Centre for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Marta E. Vilar-Sánchez
- Cardiology Group, Centre for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - Laila González-Melchor
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - Javier García-Seara
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - José Luis Martínez-Sande
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Xesús Alberte Fernández-López
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - Alana Aragón-Herrera
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - María Amparo Martínez-Monzonís
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - José Ramón González-Juanatey
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Moisés Rodríguez-Mañero
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
- Correspondence: (M.R.-M.); (I.M.); Tel.: +0034-88181-5409 (I.M.)
| | - Isabel Moscoso
- Cardiology Group, Centre for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
- Correspondence: (M.R.-M.); (I.M.); Tel.: +0034-88181-5409 (I.M.)
| |
Collapse
|
16
|
Altieri DI, Etzion Y, Anderson HD. Cannabinoid receptor agonist attenuates angiotensin II-induced enlargement and mitochondrial dysfunction in rat atrial cardiomyocytes. Front Pharmacol 2023; 14:1142583. [PMID: 37113758 PMCID: PMC10126395 DOI: 10.3389/fphar.2023.1142583] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/13/2023] [Indexed: 04/29/2023] Open
Abstract
Pathological remodeling of atrial tissue renders the atria more prone to arrhythmia upon arrival of electrical triggers. Activation of the renin-angiotensin system is an important factor that contributes to atrial remodeling, which may result in atrial hypertrophy and prolongation of P-wave duration. In addition, atrial cardiomyocytes are electrically coupled via gap junctions, and electrical remodeling of connexins may result in dysfunction of coordinated wave propagation within the atria. Currently, there is a lack of effective therapeutic strategies that target atrial remodeling. We previously proposed that cannabinoid receptors (CBR) may have cardioprotective qualities. CB13 is a dual cannabinoid receptor agonist that activates AMPK signaling in ventricular cardiomyocytes. We reported that CB13 attenuates tachypacing-induced shortening of atrial refractoriness and inhibition of AMPK signaling in the rat atria. Here, we evaluated the effects of CB13 on neonatal atrial rat cardiomyocytes (NRAM) stimulated by angiotensin II (AngII) in terms of atrial myocyte enlargement and mitochondrial function. CB13 inhibited AngII-induced enhancement of atrial myocyte surface area in an AMPK-dependent manner. CB13 also inhibited mitochondrial membrane potential deterioration in the same context. However, AngII and CB13 did not affect mitochondrial permeability transition pore opening. We further demonstrate that CB13 increased Cx43 compared to AngII-treated neonatal rat atrial myocytes. Overall, our results support the notion that CBR activation promotes atrial AMPK activation, and prevents myocyte enlargement (an indicator that suggests pathological hypertrophy), mitochondrial depolarization and Cx43 destabilization. Therefore, peripheral CBR activation should be further tested as a novel treatment strategy in the context of atrial remodeling.
Collapse
Affiliation(s)
- Danielle I. Altieri
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Canadian Centre for Agri-Food Research in Health and Medicine (CCARM), Albrechtsen Research Centre, St Boniface Hospital, Winnipeg, MB, Canada
| | - Yoram Etzion
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Hope D. Anderson
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Canadian Centre for Agri-Food Research in Health and Medicine (CCARM), Albrechtsen Research Centre, St Boniface Hospital, Winnipeg, MB, Canada
- *Correspondence: Hope D. Anderson,
| |
Collapse
|
17
|
Lkhagva B, Lee TW, Lin YK, Chen YC, Chung CC, Higa S, Chen YJ. Disturbed Cardiac Metabolism Triggers Atrial Arrhythmogenesis in Diabetes Mellitus: Energy Substrate Alternate as a Potential Therapeutic Intervention. Cells 2022; 11:cells11182915. [PMID: 36139490 PMCID: PMC9497243 DOI: 10.3390/cells11182915] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/10/2022] [Accepted: 09/16/2022] [Indexed: 11/20/2022] Open
Abstract
Atrial fibrillation (AF) is the most common type of sustained arrhythmia in diabetes mellitus (DM). Its morbidity and mortality rates are high, and its prevalence will increase as the population ages. Despite expanding knowledge on the pathophysiological mechanisms of AF, current pharmacological interventions remain unsatisfactory; therefore, novel findings on the underlying mechanism are required. A growing body of evidence suggests that an altered energy metabolism is closely related to atrial arrhythmogenesis, and this finding engenders novel insights into the pathogenesis of the pathophysiology of AF. In this review, we provide comprehensive information on the mechanistic insights into the cardiac energy metabolic changes, altered substrate oxidation rates, and mitochondrial dysfunctions involved in atrial arrhythmogenesis, and suggest a promising advanced new therapeutic approach to treat patients with AF.
Collapse
Affiliation(s)
- Baigalmaa Lkhagva
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Ting-Wei Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
| | - Yung-Kuo Lin
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Yao-Chang Chen
- Department of Biomedical Engineering, National Defense Medical Center, Taipei 11490, Taiwan
| | - Cheng-Chih Chung
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Satoshi Higa
- Cardiac Electrophysiology and Pacing Laboratory, Division of Cardiovascular Medicine, Makiminato Central Hospital, Okinawa 901-2131, Japan
| | - Yi-Jen Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Cardiovascular Research Center, Wan-Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
- Correspondence:
| |
Collapse
|
18
|
Qin X, Zhang Y, Zheng Q. Metabolic Inflexibility as a Pathogenic Basis for Atrial Fibrillation. Int J Mol Sci 2022; 23:ijms23158291. [PMID: 35955426 PMCID: PMC9368187 DOI: 10.3390/ijms23158291] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 12/10/2022] Open
Abstract
Atrial fibrillation (AF), the most common sustained arrhythmia, is closely intertwined with metabolic abnormalities. Recently, a metabolic paradox in AF pathogenesis has been suggested: under different forms of pathogenesis, the metabolic balance shifts either towards (e.g., obesity and diabetes) or away from (e.g., aging, heart failure, and hypertension) fatty acid oxidation, yet they all increase the risk of AF. This has raised the urgent need for a general consensus regarding the metabolic changes that predispose patients to AF. “Metabolic flexibility” aptly describes switches between substrates (fatty acids, glucose, amino acids, and ketones) in response to various energy stresses depending on availability and requirements. AF, characterized by irregular high-frequency excitation and the contraction of the atria, is an energy challenge and triggers a metabolic switch from preferential fatty acid utilization to glucose metabolism to increase the efficiency of ATP produced in relation to oxygen consumed. Therefore, the heart needs metabolic flexibility. In this review, we will briefly discuss (1) the current understanding of cardiac metabolic flexibility with an emphasis on the specificity of atrial metabolic characteristics; (2) metabolic heterogeneity among AF pathogenesis and metabolic inflexibility as a common pathological basis for AF; and (3) the substrate-metabolism mechanism underlying metabolic inflexibility in AF pathogenesis.
Collapse
Affiliation(s)
- Xinghua Qin
- Xi’an Key Laboratory of Special Medicine and Health Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China;
| | - Yudi Zhang
- Department of Cardiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China;
| | - Qiangsun Zheng
- Department of Cardiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China;
- Correspondence: or
| |
Collapse
|
19
|
Karamichalakis N, Kolovos V, Paraskevaidis I, Tsougos E. A New Hope: Sodium-Glucose Cotransporter-2 Inhibition to Prevent Atrial Fibrillation. J Cardiovasc Dev Dis 2022; 9:jcdd9080236. [PMID: 35893226 PMCID: PMC9331782 DOI: 10.3390/jcdd9080236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 07/17/2022] [Accepted: 07/22/2022] [Indexed: 11/16/2022] Open
Abstract
Atrial arrhythmias are common in patients with diabetes mellitus (DM), and despite recent advances in pharmaceutical and invasive treatments, atrial fibrillation (AF) and atrial flutter (AFl) are still associated with substantial mortality and morbidity. Clinical trial data imply a protective effect of sodium-glucose cotransporter-2 inhibitors (SGLT2is) on the occurrence of AF and AFl. This review summarizes the state of knowledge regarding DM-mediated mechanisms responsible for AF genesis and recurrence but also discusses the recent data from experimental studies, published trials and metanalyses.
Collapse
|
20
|
Rafaqat S, Rafaqat S, Rafaqat S. Pathophysiological aspects of insulin resistance in Atrial Fibrillation: novel therapeutic approaches. INTERNATIONAL JOURNAL OF ARRHYTHMIA 2022. [DOI: 10.1186/s42444-021-00057-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Abstract
Background
Insulin resistance is associated with metabolic disorders including diabetes, obesity, hypertension, and inflammation which are the risk factors for Atrial Fibrillation. Many studies have reported that type 2 diabetes and AF are related and also their prevalence is increasing globally. Moreover, insulin resistance begins the type 2 diabetes.
Main body
This review explains the pathophysiological aspects of insulin resistance in AF patients and discusses the drugs that are used to manage insulin resistance including Biguanides (metformin), thiazolidinediones (TZDs) [Pioglitazone, rosiglitazone], Sodium-glucose cotransporter 2 (SGLT2) inhibitors, Concentrated Insulin Products, Dipeptidyl peptidase-4 (DPP-4) Inhibitors, Glucagon-like peptide 1 (GLP-1) receptor Agonists, Pramlintide, Sulfonylureas, Meglitinides, α-Glucosidase Inhibitors, Colesevelam, Bromocriptine. This review will highlight a few major drugs that played a significant role in AF patients. For this purpose, many databases were used for reviewing the literature and keywords are used such as Insulin Resistance, Pathophysiology, Atrial Fibrillation, and Drugs.
Conclusion
This review article concludes that insulin resistance is related to AF. It also provides an outlook on the recent pathophysiological aspects of insulin resistance in AF; however, more studies are needed to clarify the management of insulin resistance in AF patients to prevent the development of type 2 diabetes.
Collapse
|
21
|
Zhang Y, Fu Y, Jiang T, Liu B, Sun H, Zhang Y, Fan B, Li X, Qin X, Zheng Q. Enhancing Fatty Acids Oxidation via L-Carnitine Attenuates Obesity-Related Atrial Fibrillation and Structural Remodeling by Activating AMPK Signaling and Alleviating Cardiac Lipotoxicity. Front Pharmacol 2021; 12:771940. [PMID: 34899326 PMCID: PMC8662783 DOI: 10.3389/fphar.2021.771940] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/12/2021] [Indexed: 12/28/2022] Open
Abstract
Atrial fibrillation (AF) is the most common sustained cardiac arrhythmia in clinical setting. Its pathogenesis was associated with metabolic disorder, especially defective fatty acids oxidation (FAO). However, whether promoting FAO could prevent AF occurrence and development remains elusive. In this study, we established a mouse model of obesity-related AF through high-fat diet (HFD) feeding, and used l-carnitine (LCA, 150 mg/kg⋅BW/d), an endogenous cofactor of carnitine palmitoyl-transferase-1B (CPT1B; the rate-limiting enzyme of FAO) to investigate whether FAO promotion can attenuate the AF susceptibility in obesity. All mice underwent electrophysiological assessment for atrial vulnerability, and echocardiography, histology and molecular evaluation for AF substrates and underlying mechanisms, which were further validated by pharmacological experiments in vitro. HFD-induced obese mice increased AF vulnerability and exhibited apparent atrial structural remodeling, including left atrial dilation, cardiomyocyte hypertrophy, connexin-43 remodeling and fibrosis. Pathologically, HFD apparently leads to defective cardiac FAO and subsequent lipotoxicity, thereby evoking a set of pathological reactions including oxidative stress, DNA damage, inflammation, and insulin resistance. Enhancing FAO via LCA attenuated lipotoxicity and lipotoxicity-induced pathological changes in the atria of obese mice, resulting in restored structural remodeling and ameliorated AF susceptibility. Mechanistically, LCA activated AMPK/PGC1α signaling both in vivo and in vitro, and pharmacological inhibition of AMPK via Compound C attenuated LCA-induced cardio-protection in palmitate-treated primary atrial cardiomyocytes. Taken together, our results demonstrated that FAO promotion via LCA attenuated obesity-mediated AF and structural remodeling by activating AMPK signaling and alleviating atrial lipotoxicity. Thus, enhancing FAO may be a potential therapeutic target for AF.
Collapse
Affiliation(s)
- Yudi Zhang
- The Second Affiliate Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yuping Fu
- The Second Affiliate Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Tiannan Jiang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Binghua Liu
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Hongke Sun
- The Second Affiliate Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ying Zhang
- The Second Affiliate Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Boyuan Fan
- The Second Affiliate Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaoli Li
- The Second Affiliate Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xinghua Qin
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Qiangsun Zheng
- The Second Affiliate Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
22
|
Bulló M, Papandreou C, García-Gavilán J, Ruiz-Canela M, Li J, Guasch-Ferré M, Toledo E, Clish C, Corella D, Estruch R, Ros E, Fitó M, Lee CH, Pierce K, Razquin C, Arós F, Serra-Majem L, Liang L, Martínez-González MA, Hu FB, Salas-Salvadó J. Tricarboxylic acid cycle related-metabolites and risk of atrial fibrillation and heart failure. Metabolism 2021; 125:154915. [PMID: 34678258 PMCID: PMC9206868 DOI: 10.1016/j.metabol.2021.154915] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/28/2021] [Accepted: 10/14/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND Tricarboxylic acid (TCA) cycle deregulation may predispose to cardiovascular diseases, but the role of TCA cycle-related metabolites in the development of atrial fibrillation (AF) and heart failure (HF) remains unexplored. This study sought to investigate the association of TCA cycle-related metabolites with risk of AF and HF. METHODS We used two nested case-control studies within the PREDIMED study. During a mean follow-up for about 10 years, 512 AF and 334 HF incident cases matched by age (±5 years), sex and recruitment center to 616 controls and 433 controls, respectively, were included in this study. Baseline plasma levels of citrate, aconitate, isocitrate, succinate, malate and d/l-2-hydroxyglutarate were measured with liquid chromatography-tandem mass spectrometry. Multivariable conditional logistic regression models were fitted to estimate odds ratios (OR) and 95% confidence intervals (95% CI) for metabolites and the risk of AF or HF. Potential confounders included smoking, family history of premature coronary heart disease, physical activity, alcohol intake, body mass index, intervention groups, dyslipidemia, hypertension, type 2 diabetes and medication use. RESULTS Comparing extreme quartiles of metabolites, elevated levels of succinate, malate, citrate and d/l-2-hydroxyglutarate were associated with a higher risk of AF [ORQ4 vs. Q1 (95% CI): 1.80 (1.21-2.67), 2.13 (1.45-3.13), 1.87 (1.25-2.81) and 1.95 (1.31-2.90), respectively]. One SD increase in aconitate was directly associated with AF risk [OR (95% CI): 1.16 (1.01-1.34)]. The corresponding ORs (95% CI) for HF comparing extreme quartiles of malate, aconitate, isocitrate and d/l-2-hydroxyglutarate were 2.15 (1.29-3.56), 2.16 (1.25-3.72), 2.63 (1.56-4.44) and 1.82 (1.10-3.04), respectively. These associations were confirmed in an internal validation, except for aconitate and AF. CONCLUSION These findings underscore the potential role of the TCA cycle in the pathogenesis of cardiac outcomes.
Collapse
Affiliation(s)
- Mònica Bulló
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Unitat de Nutrició Humana, Reus, Spain; Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; University Hospital of Sant Joan de Reus, Nutrition Unit, Reus, Spain
| | - Christopher Papandreou
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Unitat de Nutrició Humana, Reus, Spain; Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; University Hospital of Sant Joan de Reus, Nutrition Unit, Reus, Spain
| | - Jesus García-Gavilán
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Unitat de Nutrició Humana, Reus, Spain; Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; University Hospital of Sant Joan de Reus, Nutrition Unit, Reus, Spain
| | - Miguel Ruiz-Canela
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; University of Navarra, Department of Preventive Medicine and Public Health, Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Navarra, Spain
| | - Jun Li
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Marta Guasch-Ferré
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Channing Division for Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, MA, USA
| | - Estefanía Toledo
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; University of Navarra, Department of Preventive Medicine and Public Health, Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Navarra, Spain
| | - Clary Clish
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Dolores Corella
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; Department of Preventive Medicine, University of Valencia, Valencia, Spain
| | - Ramon Estruch
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; Department of Internal Medicine, Department of Endocrinology and Nutrition Institut d'Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Emilio Ros
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; Lipid Clinic, Department of Endocrinology and Nutrition Institut d'Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Montserrat Fitó
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; Cardiovascular and Nutrition Research Group, Institut de Recerca Hospital del Mar, Barcelona, Spain
| | - Chih-Hao Lee
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Molecular Metabolism (C.-H.L.), Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Kerry Pierce
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Cristina Razquin
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; University of Navarra, Department of Preventive Medicine and Public Health, Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Navarra, Spain
| | - Fernando Arós
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; Department of Cardiology, University Hospital of Alava, Vitoria, Spain
| | - Lluís Serra-Majem
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; Institute of Health Sciences IUNICS, University of Balearic Islands and Hospital Son Espases, Palma de Mallorca, Spain
| | - Liming Liang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Statistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Miguel A Martínez-González
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; University of Navarra, Department of Preventive Medicine and Public Health, Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Navarra, Spain; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Channing Division for Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Statistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jordi Salas-Salvadó
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Unitat de Nutrició Humana, Reus, Spain; Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; University Hospital of Sant Joan de Reus, Nutrition Unit, Reus, Spain.
| |
Collapse
|
23
|
Zhong W, Yang H, Wang Y, Yang Y, Guo C, Wang C, Ji Q. Proteomic profiles of patients with atrial fibrillation provide candidate biomarkers for diagnosis. Int J Cardiol 2021; 344:205-212. [PMID: 34592249 DOI: 10.1016/j.ijcard.2021.09.047] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/13/2021] [Accepted: 09/22/2021] [Indexed: 01/22/2023]
Abstract
BACKGROUND Atrial fibrillation (AF) is the most common cardiac arrhythmia worldwide with an increasing risk of heart failure, stroke, and thromboembolic events. Currently distinct pathophysiological mechanisms during AF development and valuable biomarkers for AF management remain unknown. METHODS In this study, we collected peripheral plasma samples from 18 non-valvular AF patients and 10 controls. A LC-MS/MS-DIA-based quantitative proteomic analysis, as well as bioinformatic analysis, was performed for discovery of differentially expressed proteins (DEPs) and dysregulated pathways. Next, we utilized enzyme-linked immunosorbent assay (ELISA) to validate selected DEPs and assessed their abilities for discrimination in another group of 20 AF patients and 10 controls. RESULTS The plasma proteome provided evidence for vital roles of abnormal inflammation, hemostasis, tissue remodeling and metabolism in AF patients. Differences between paroxysmal and persistent AF mainly lie in proteins or pathways related to hemostasis and cardiac remodeling. In addition, we successfully validated up-regulated proteins of platelet factor 4 variant 1 (PF4V1), thrombospondin-1 (THBS1), platelet basic protein (PBP) and fructose-bisphosphate aldolase A (ALDOA) in another group, which could make discrimination between AF patients and controls. CONCLUSIONS Our pilot proteomic study provided candidate biomarkers of PF4V1, THBS1, PBP and ALODA with the hope of application in AF management. And we showed the differences in pathophysiological mechanisms between paroxysmal and persistent AF. They mainly focused on pathways of hemostasis and cardiac remodeling, which could be a valuable clue for further research on AF progression.
Collapse
Affiliation(s)
- WeiYi Zhong
- Shanghai Medical College, Fudan University, Shanghai 200032, China..
| | - Hui Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - YuLin Wang
- Department of Cardiac Surgery, Zhongshan Hospital Fudan University, Shanghai 200032, China..
| | - Ye Yang
- Department of Cardiac Surgery, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - ChangFa Guo
- Department of Cardiac Surgery, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - ChunSheng Wang
- Department of Cardiac Surgery, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Qiang Ji
- Department of Cardiac Surgery, Zhongshan Hospital Fudan University, Shanghai 200032, China..
| |
Collapse
|
24
|
Leblanc FJA, Lettre G, Nattel S. Understanding the molecular events preceding and leading to atrial fibrillation. Heart Rhythm 2021; 18:2126-2127. [PMID: 34508877 DOI: 10.1016/j.hrthm.2021.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 09/02/2021] [Indexed: 11/28/2022]
Affiliation(s)
- Francis J A Leblanc
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada; Research Center, Montreal Heart Institute, Montreal, Quebec, Canada
| | - Guillaume Lettre
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada; Research Center, Montreal Heart Institute, Montreal, Quebec, Canada
| | - Stanley Nattel
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada; Research Center, Montreal Heart Institute, Montreal, Quebec, Canada; Institute of Pharmacology, West German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen, Essen, Germany; IHU Liryc and Fondation Bordeaux Université, Bordeaux, France.
| |
Collapse
|
25
|
The Role of Mitochondrial Dysfunction in Atrial Fibrillation: Translation to Druggable Target and Biomarker Discovery. Int J Mol Sci 2021; 22:ijms22168463. [PMID: 34445167 PMCID: PMC8395135 DOI: 10.3390/ijms22168463] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/02/2021] [Accepted: 08/03/2021] [Indexed: 01/02/2023] Open
Abstract
Atrial fibrillation (AF) is the most prevalent and progressive cardiac arrhythmia worldwide and is associated with serious complications such as heart failure and ischemic stroke. Current treatment modalities attenuate AF symptoms and are only moderately effective in halting the arrhythmia. Therefore, there is an urgent need to dissect molecular mechanisms that drive AF. As AF is characterized by a rapid atrial activation rate, which requires a high energy metabolism, a role of mitochondrial dysfunction in AF pathophysiology is plausible. It is well known that mitochondria play a central role in cardiomyocyte function, as they produce energy to support the mechanical and electrical function of the heart. Details on the molecular mechanisms underlying mitochondrial dysfunction are increasingly being uncovered as a contributing factor in the loss of cardiomyocyte function and AF. Considering the high prevalence of AF, investigating the role of mitochondrial impairment in AF may guide the path towards new therapeutic and diagnostic targets. In this review, the latest evidence on the role of mitochondria dysfunction in AF is presented. We highlight the key modulators of mitochondrial dysfunction that drive AF and discuss whether they represent potential targets for therapeutic interventions and diagnostics in clinical AF.
Collapse
|
26
|
Mitochondrial Dysfunction in Atrial Fibrillation-Mechanisms and Pharmacological Interventions. J Clin Med 2021; 10:jcm10112385. [PMID: 34071563 PMCID: PMC8199309 DOI: 10.3390/jcm10112385] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/20/2021] [Accepted: 05/25/2021] [Indexed: 12/22/2022] Open
Abstract
Despite the enormous progress in the treatment of atrial fibrillation, mainly with the use of invasive techniques, many questions remain unanswered regarding the pathomechanism of the arrhythmia and its prevention methods. The development of atrial fibrillation requires functional changes in the myocardium that result from disturbed ionic fluxes and altered electrophysiology of the cardiomyocyte. Electrical instability and electrical remodeling underlying the arrhythmia may result from a cellular energy deficit and oxidative stress, which are caused by mitochondrial dysfunction. The significance of mitochondrial dysfunction in the pathogenesis of atrial fibrillation remains not fully elucidated; however, it is emphasized by the reduction of atrial fibrillation burden after therapeutic interventions improving the mitochondrial welfare. This review summarizes the mechanisms of mitochondrial dysfunction related to atrial fibrillation and current pharmacological treatment options targeting mitochondria to prevent or improve the outcome of atrial fibrillation.
Collapse
|
27
|
Zhang Y, Xie Y, Lv W, Hu C, Xu T, Liu X, Zhang R, Xu G, Xia Y, Zhao X. A high throughput lipidomics method and its application in atrial fibrillation based on 96-well plate pretreatment and liquid chromatography-mass spectrometry. J Chromatogr A 2021; 1651:462271. [PMID: 34102397 DOI: 10.1016/j.chroma.2021.462271] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/14/2021] [Accepted: 05/18/2021] [Indexed: 11/27/2022]
Abstract
Successful applications of lipidomics in clinic need study large-scale samples, and the bottlenecks are in throughput and robustness of the lipid analytical method. Here, we report an untargeted lipidomics method by combining high throughput pretreatment in the 96-well plate with ultra-high performance liquid chromatography coupled to quadrupole time-of-flight tandem mass spectrometry. The developed method was validated to have satisfactory analytical characteristics in terms of linearity, repeatability and extraction recovery. It can be used to handle 96 samples simultaneously in 25 min and detect 441 lipids in plasma sample. Storage stability investigation on lipid extracts provided an operable procedure for large-scale sample analysis and demonstrated most lipids were stable in autosampler at 10 °C within 36 h and at -80 °C within 72 h after the pretreatment. To prove the usefulness, the method was employed to investigate abnormal plasma lipidome related to atrial fibrillation. A biomarker panel with the area under the curve (AUC) values of 0.831 and 0.745 was achieved in the discovery and external validation sets, respectively. These results showed that the developed method is applicable for large-scale biological sample handling and lipid analysis of plasma.
Collapse
Affiliation(s)
- Yuqing Zhang
- Zhang Dayu School of Chemistry, Dalian University of Technology, Dalian 116024, P. R. China; CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, P. R. China
| | - Yunpeng Xie
- The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Wangjie Lv
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, P. R. China
| | - Chunxiu Hu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, P. R. China
| | - Tianrun Xu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, P. R. China
| | - Xinyu Liu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, P. R. China
| | - Rongfeng Zhang
- The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Guowang Xu
- Zhang Dayu School of Chemistry, Dalian University of Technology, Dalian 116024, P. R. China; CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, P. R. China
| | - Yunlong Xia
- The First Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Xinjie Zhao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, P. R. China.
| |
Collapse
|
28
|
Vrachatis DA, Papathanasiou KA, Kossyvakis C, Giotaki SG, Raisakis K, Iliodromitis KE, Reimers B, Stefanini GG, Cleman M, Sianos G, Lansky A, Deftereos SG, Giannopoulos G. Atrial fibrillation risk in patients suffering from type I diabetes mellitus. A review of clinical and experimental evidence. Diabetes Res Clin Pract 2021; 174:108724. [PMID: 33647332 DOI: 10.1016/j.diabres.2021.108724] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 01/31/2021] [Accepted: 02/09/2021] [Indexed: 12/12/2022]
Abstract
Atrial fibrillation (AF) and diabetes mellitus (DM) are commonly encountered in clinical practice. Although, the long term macrovascular and microvascular sequela of DM are well validated, the association between the less prevalent type 1 DM (T1DM) and atrial arrhythmogenesis is poorly understood. In the present review we highlight the current experimental and clinical data addressing this complex interaction. Animal studies support that T1DM, characterized by insulin deficiency and glycemic variability, impairs phosphatidylinositol 3‑kinase (PI3K)/protein kinase B signaling pathway. This pathway holds a central role in atrial electrical and structural remodeling responsible for arrhythmia initiation and maintenance. The molecular ''footprint'' of T1DM in atrial myocytes seems to involve a state of increased oxidative stress, impaired glucose transportation, ionic channel dysregulation and eventually fibrosis. On the contrary only a few clinical studies have examined the role of T1DM as an independent risk factor for AF development, and are discussed here. Further research is needed to solidify the real magnitude of this association and to investigate the clinical implications of PI3K molecular signaling pathway in atrial fibrillation management.
Collapse
Affiliation(s)
- Dimitrios A Vrachatis
- Humanitas Clinical and Research Center IRCCS, Milan, Italy; National and Kapodistrian University of Athens, Athens, Greece
| | | | | | | | | | | | | | | | - Michael Cleman
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - George Sianos
- Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Alexandra Lansky
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | | | | |
Collapse
|
29
|
KLF15 Loss-of-Function Mutation Underlying Atrial Fibrillation as well as Ventricular Arrhythmias and Cardiomyopathy. Genes (Basel) 2021; 12:genes12030408. [PMID: 33809104 PMCID: PMC8001991 DOI: 10.3390/genes12030408] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 02/27/2021] [Accepted: 03/09/2021] [Indexed: 12/04/2022] Open
Abstract
Atrial fibrillation (AF) represents the most common type of clinical cardiac arrhythmia and substantially increases the risks of cerebral stroke, heart failure and death. Accumulating evidence has convincingly demonstrated the strong genetic basis of AF, and an increasing number of pathogenic variations in over 50 genes have been causally linked to AF. Nevertheless, AF is of pronounced genetic heterogeneity, and the genetic determinants underpinning AF in most patients remain obscure. In the current investigation, a Chinese pedigree with AF as well as ventricular arrhythmias and hypertrophic cardiomyopathy was recruited. Whole exome sequencing and bioinformatic analysis of the available family members were conducted, and a novel heterozygous variation in the KLF15 gene (encoding Krüppel-like factor 15, a transcription factor critical for cardiac electrophysiology and structural remodeling), NM_014079.4: c.685A>T; p.(Lys229*), was identified. The variation was verified by Sanger sequencing and segregated with autosomal dominant AF in the family with complete penetrance. The variation was absent from 300 unrelated healthy subjects used as controls. In functional assays using a dual-luciferase assay system, mutant KLF15 showed neither transcriptional activation of the KChIP2 promoter nor transcriptional inhibition of the CTGF promoter, alone or in the presence of TGFB1, a key player in the pathogenesis of arrhythmias and cardiomyopathies. The findings indicate KLF15 as a new causative gene responsible for AF as well as ventricular arrhythmias and hypertrophic cardiomyopathy, and they provide novel insight into the molecular mechanisms underlying cardiac arrhythmias and hypertrophic cardiomyopathy.
Collapse
|
30
|
Tseng CH. Metformin Use Is Associated With a Lower Incidence of Hospitalization for Atrial Fibrillation in Patients With Type 2 Diabetes Mellitus. Front Med (Lausanne) 2021; 7:592901. [PMID: 33693008 PMCID: PMC7937645 DOI: 10.3389/fmed.2020.592901] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 12/03/2020] [Indexed: 12/19/2022] Open
Abstract
Background: The effect of metformin on the risk of atrial fibrillation (AF) requires confirmation. This retrospective cohort study compared the incidence of hospitalization for AF in ever and never users of metformin. Methods: Patients with newly diagnosed type 2 diabetes mellitus during 1999–2005 were enrolled from Taiwan's National Health Insurance database. Analyses were conducted in both an unmatched cohort of 173,398 ever users and 21,666 never users and in a propensity score-matched cohort of 21,662 pairs of ever and never users. They were free from a diagnosis of AF before January 1, 2006 and were followed up until December 31, 2011. Hazard ratios were estimated by Cox regression incorporated with the inverse probability of treatment weighting using the propensity score. Results: A total of 303 ever users and 86 never users in the unmatched cohort and 56 ever users and 86 never users in the matched cohort developed hospitalization for AF during follow-up. The respective incidence rates were 37.72 and 92.45 per 100,000 person-years in the unmatched cohort and were 56.98 and 92.46 per 100,000 person-years in the matched cohort. The hazard ratio for ever vs. never users was 0.405 (95% confidence interval: 0.319–0.515) in the unmatched cohort and 0.617 (0.441–0.864) in the matched cohort. Hazard ratios for the tertiles of cumulative duration of metformin therapy vs. never users showed a dose-response effect. The findings were consistent in sensitivity analyses. Conclusion: Metformin use is associated with a lower risk of hospitalization for AF in patients with type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Chin-Hsiao Tseng
- Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan.,Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Division of Environmental Health and Occupational Medicine of the National Health Research Institutes, Zhunan, Taiwan
| |
Collapse
|
31
|
Lee TW, Lee TI, Lin YK, Chen YC, Kao YH, Chen YJ. Effect of antidiabetic drugs on the risk of atrial fibrillation: mechanistic insights from clinical evidence and translational studies. Cell Mol Life Sci 2021; 78:923-934. [PMID: 32965513 PMCID: PMC11072414 DOI: 10.1007/s00018-020-03648-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 08/18/2020] [Accepted: 09/12/2020] [Indexed: 12/13/2022]
Abstract
Diabetes mellitus (DM) is an independent risk factor for atrial fibrillation (AF), which is the most common sustained arrhythmia and is associated with substantial morbidity and mortality. Advanced glycation end product and its receptor activation, cardiac energy dysmetabolism, structural and electrical remodeling, and autonomic dysfunction are implicated in AF pathophysiology in diabetic hearts. Antidiabetic drugs have been demonstrated to possess therapeutic potential for AF. However, clinical investigations of AF in patients with DM have been scant and inconclusive. This article provides a comprehensive review of research findings on the association between DM and AF and critically analyzes the effect of different pharmacological classes of antidiabetic drugs on AF.
Collapse
Affiliation(s)
- Ting-Wei Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Ting-I Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Department of General Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yung-Kuo Lin
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yao-Chang Chen
- Department of Biomedical Engineering, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Hsun Kao
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yi-Jen Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
- Cardiovascular Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
32
|
Mason FE, Pronto JRD, Alhussini K, Maack C, Voigt N. Cellular and mitochondrial mechanisms of atrial fibrillation. Basic Res Cardiol 2020; 115:72. [PMID: 33258071 PMCID: PMC7704501 DOI: 10.1007/s00395-020-00827-7] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 10/26/2020] [Indexed: 11/06/2022]
Abstract
The molecular mechanisms underlying atrial fibrillation (AF), the most common form of arrhythmia, are poorly understood and therefore target-specific treatment options remain an unmet clinical need. Excitation–contraction coupling in cardiac myocytes requires high amounts of adenosine triphosphate (ATP), which is replenished by oxidative phosphorylation in mitochondria. Calcium (Ca2+) is a key regulator of mitochondrial function by stimulating the Krebs cycle, which produces nicotinamide adenine dinucleotide for ATP production at the electron transport chain and nicotinamide adenine dinucleotide phosphate for the elimination of reactive oxygen species (ROS). While it is now well established that mitochondrial dysfunction plays an important role in the pathophysiology of heart failure, this has been less investigated in atrial myocytes in AF. Considering the high prevalence of AF, investigating the role of mitochondria in this disease may guide the path towards new therapeutic targets. In this review, we discuss the importance of mitochondrial Ca2+ handling in regulating ATP production and mitochondrial ROS emission and how alterations, particularly in these aspects of mitochondrial activity, may play a role in AF. In addition to describing research advances, we highlight areas in which further studies are required to elucidate the role of mitochondria in AF.
Collapse
Affiliation(s)
- Fleur E Mason
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Julius Ryan D Pronto
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Khaled Alhussini
- Department of Thoracic and Cardiovascular Surgery, University Clinic Würzburg, Würzburg, Germany
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center Würzburg, University Clinic Würzburg, Am Schwarzenberg 15, 97078, Würzburg, Germany. .,Department of Internal Medicine I, University Clinic Würzburg, Am Schwarzenberg 15, 97078, Würzburg, Germany.
| | - Niels Voigt
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany. .,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany. .,Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany.
| |
Collapse
|
33
|
Brown SM, Larsen NK, Thankam FG, Agrawal DK. Fetal cardiomyocyte phenotype, ketone body metabolism, and mitochondrial dysfunction in the pathology of atrial fibrillation. Mol Cell Biochem 2020; 476:1165-1178. [PMID: 33188453 DOI: 10.1007/s11010-020-03980-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 11/06/2020] [Indexed: 10/23/2022]
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia diagnosed in clinical practice. Even though hypertension, congestive heart failure, pulmonary disease, and coronary artery disease are the potential risk factors for AF, the underlying molecular pathology is largely unknown. The reversion of the mature cardiomyocytes to fetal phenotype, impaired ketone body metabolism, mitochondrial dysfunction, and the cellular effect of reactive oxygen species (ROS) are the major underlying biochemical events associated with the molecular pathology of AF. On this background, the present manuscript sheds light into these biochemical events in regard to the metabolic derangements in cardiomyocyte leading to AF, especially with respect to structural, contractile, and electrophysiological properties. In addition, the article critically reviews the current understanding, potential demerits, and translational strategies in the management of AF.
Collapse
Affiliation(s)
- Sean M Brown
- Creighton University School of Medicine, Omaha, NE, 68178, USA
| | | | - Finosh G Thankam
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766, USA.
| |
Collapse
|
34
|
Wang Y, Morishima M, Li D, Takahashi N, Saikawa T, Nattel S, Ono K. Binge Alcohol Exposure Triggers Atrial Fibrillation Through T-Type Ca 2+ Channel Upregulation via Protein Kinase C (PKC) / Glycogen Synthesis Kinase 3β (GSK3β) / Nuclear Factor of Activated T-Cells (NFAT) Signaling - An Experimental Account of Holiday Heart Syndrome. Circ J 2020; 84:1931-1940. [PMID: 33028764 DOI: 10.1253/circj.cj-20-0288] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2024]
Abstract
BACKGROUND The association between binge alcohol ingestion and atrial fibrillation (AF), often termed "holiday heart syndrome", has long been recognized. However, the underlying cellular and molecular mechanisms are unknown. METHODS AND RESULTS An experimental model of binge alcohol-induced AF was developed to elucidate the mechanisms linking acute ethanol exposure to changes in ion channel transcription and AF susceptibility. AF-susceptibility during transesophageal electrical stimulation was enhanced 8 h after, but not immediately or 24 h after, acute alcohol intake. T-type calcium channel (TCC) blockade and calcineurin inhibition diminished the AF-promoting effect of ethanol. Long-term (8-24 h) exposure to ethanol augmented TCC isoform-expression (Cav3.1 and Cav3.2) and currents in cardiomyocytes, accompanied by upregulation of the transcription factors, Csx/Nkx2.5 and nuclear factor of activated T-cells (NFAT), in the nucleus, and of phospho-glycogen synthesis kinase 3β (GSK3β) in the cytosol. Inhibition of protein kinase C (PKC) during the 7- to 8-h period following ethanol exposure attenuated susceptibility to AF, whereas acute exposure did not. GSK3β inhibition itself upregulated TCC expression and increased AF susceptibility. CONCLUSIONS The present study results suggest a crucial role for TCC upregulation in the AF substrate following binge alcohol-drinking, resulting from ethanol-induced PKC-activation that hyperphosphorylates GSK3β to cause enhanced calcineurin-NFAT-Csx/Nkx2.5 signaling. These observations elucidate for the first time the potential mechanisms underlying the clinically well-recognized, but mechanistically enigmatic, "holiday heart syndrome".
Collapse
Affiliation(s)
- Yan Wang
- Department of Pathophysiology, Oita University School of Medicine
- Department of Clinical Examination and Diagnostics, Oita University School of Medicine
| | - Masaki Morishima
- Department of Pathophysiology, Oita University School of Medicine
| | - Dan Li
- Department of Pathophysiology, Oita University School of Medicine
| | - Naohiko Takahashi
- Department of Clinical Examination and Diagnostics, Oita University School of Medicine
| | - Tetsunori Saikawa
- Department of Clinical Examination and Diagnostics, Oita University School of Medicine
| | - Stanley Nattel
- Montreal Heart Institute Research Center, University of Montreal
| | - Katsushige Ono
- Department of Pathophysiology, Oita University School of Medicine
- Department of Clinical Examination and Diagnostics, Oita University School of Medicine
| |
Collapse
|
35
|
Brown SM, Larsen NK, Thankam FG, Agrawal DK. Regulatory role of cardiomyocyte metabolism via AMPK activation in modulating atrial structural, contractile, and electrical properties following atrial fibrillation. Can J Physiol Pharmacol 2020; 99:36-41. [PMID: 33049144 DOI: 10.1139/cjpp-2020-0313] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The number of patients diagnosed with atrial fibrillation (AF) has been rising due to increased incidence, enhanced detection methods, and greater survival rates following diagnosis. Due to this increase, AF is now the most commonly diagnosed arrhythmia in clinical practice. AF is characterized by irregular, high-frequency contractions of atrial myocytes that lead to turbulent blood flow and the potential for thrombus formation, stroke, or heart failure. These high-frequency contractions of the atrial myocytes cause an imbalance between metabolic supply and demand. Although advances have been made in understanding the pathophysiology of AF, the etiology and underlying pathogenic mechanism remain unknown. However, recent evidence suggests that cardiomyocyte metabolism involving 5' AMP-activated protein kinase (AMPK) activation is altered in patients with AF. Here, we critically reviewed the current understanding of AMPK activation in AF and how it could affect structural, contractile, and electrophysiological cellular properties in the pathogenesis of AF.
Collapse
Affiliation(s)
- Sean M Brown
- Creighton University School of Medicine, Omaha, NE 68178, USA
| | | | - Finosh G Thankam
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| |
Collapse
|
36
|
Aitken-Buck HM, Krause J, Zeller T, Jones PP, Lamberts RR. Long-Chain Acylcarnitines and Cardiac Excitation-Contraction Coupling: Links to Arrhythmias. Front Physiol 2020; 11:577856. [PMID: 33041874 PMCID: PMC7518131 DOI: 10.3389/fphys.2020.577856] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 08/25/2020] [Indexed: 12/31/2022] Open
Abstract
A growing number of metabolomic studies have associated high circulating levels of the amphiphilic fatty acid metabolites, long-chain acylcarnitines (LCACs), with cardiovascular disease (CVD) risk. These studies show that plasma LCAC levels can be correlated with the stage and severity of CVD and with indices of cardiac hypertrophy and ventricular function. Complementing these recent clinical associations is an extensive body of basic research that stems mostly from the twentieth century. These works, performed in cardiomyocyte and multicellular preparations from animal and cell models, highlight stereotypical derangements in cardiac electrophysiology induced by exogenous LCAC treatment that promote arrhythmic muscle behavior. In many cases, this is coupled with acute inotropic modulation; however, whether LCACs increase or decrease contractility is inconclusive. Linked to the electromechanical alterations induced by LCAC exposure is an array of effects on cardiac excitation-contraction coupling mechanisms that overload the cardiomyocyte cytosol with Na+ and Ca2+ ions. The aim of this review is to revisit this age-old literature and collate it with recent findings to provide a pathophysiological context for the growing body of metabolomic association studies that link circulating LCACs with CVD.
Collapse
Affiliation(s)
- Hamish M Aitken-Buck
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Julia Krause
- University Heart and Vascular Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Hamburg, Hamburg, Germany
| | - Tanja Zeller
- University Heart and Vascular Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Hamburg, Hamburg, Germany
| | - Peter P Jones
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Regis R Lamberts
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
37
|
Soattin L, Lubberding AF, Bentzen BH, Christ T, Jespersen T. Inhibition of Adenosine Pathway Alters Atrial Electrophysiology and Prevents Atrial Fibrillation. Front Physiol 2020; 11:493. [PMID: 32595514 PMCID: PMC7304385 DOI: 10.3389/fphys.2020.00493] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 04/23/2020] [Indexed: 01/13/2023] Open
Abstract
Background Adenosine leads to atrial action potential (AP) shortening through activation of adenosine 1 receptors (A1-R) and subsequent opening of G-protein-coupled inwardly rectifying K+ channels. Extracellular production of adenosine is drastically increased during stress and ischemia. Objective The aim of this study was to address whether the pharmacological blockade of endogenous production of adenosine and of its signaling prevents atrial fibrillation (AF). Methods The role of A1-R activation on atrial action potential duration, refractoriness, and AF vulnerability was investigated in rat isolated beating heart preparations (Langendorff) with an A1-R agonist [2-chloro-N6-cyclopentyladenosine (CCPA), 50 nM] and antagonist [1-butyl-3-(3-hydroxypropyl)-8-(3-noradamantyl)xanthine (PSB36), 40 nM]. Furthermore, to interfere with the endogenous adenosine release, the ecto-5′-nucleotidase (CD73) inhibitor was applied [5′-(α,β-methylene) diphosphate sodium salt (AMPCP), 500 μM]. Isolated trabeculae from human right atrial appendages (hRAAs) were used for comparison. Results As expected, CCPA shortened AP duration at 90% of repolarization (APD90) and effective refractory period (ERP) in rat atria. PSB36 prolonged APD90 and ERP in rat atria, and CD73 inhibition with AMPCP prolonged ERP in rats, confirming that endogenously produced amount of adenosine is sufficiently high to alter atrial electrophysiology. In human atrial appendages, CCPA shortened APD90, while PSB36 prolonged it. Rat hearts treated with CCPA are prone to AF. In contrast, PSB36 and AMPCP prevented AF events and reduced AF duration (vehicle, 11.5 ± 2.6 s; CCPA, 40.6 ± 16.1 s; PSB36, 6.5 ± 3.7 s; AMPCP, 3.0 ± 1.4 s; P < 0.0001). Conclusion A1-R activation by intrinsic adenosine release alters atrial electrophysiology and promotes AF. Inhibition of adenosine pathway protects atria from arrhythmic events.
Collapse
Affiliation(s)
- Luca Soattin
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anniek Frederike Lubberding
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bo Hjorth Bentzen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Torsten Christ
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Thomas Jespersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
38
|
Arginine derivatives in atrial fibrillation progression phenotypes. J Mol Med (Berl) 2020; 98:999-1008. [PMID: 32504111 PMCID: PMC8556202 DOI: 10.1007/s00109-020-01932-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/20/2020] [Accepted: 05/27/2020] [Indexed: 10/24/2022]
Abstract
Arginine, homoarginine (hArg), asymmetric dimethylarginine (ADMA), and symmetric dimethylarginine (SDMA) affect nitric oxide metabolism and altered concentrations are associated with cardiovascular morbidity and mortality. We analyzed these metabolites using liquid chromatography-tandem mass spectrometry in patients with atrial fibrillation (AF) (n = 241) with a focus on heart rhythm at blood withdrawal, AF progression phenotypes, and successful sinus rhythm (SR) restoration (n = 22). AF progression phenotypes were defined as paroxysmal AF with/without low voltage areas (LVA) and persistent AF with/without LVA. While arginine, ADMA, and hArg were within reference limits for healthy controls, SDMA was higher in the AF cohort (0.57 ± 0.12 vs. 0.53 μmol/L (97.5th percentile in reference cohort)). SR restoration in AF patients resulted in normalization of SDMA concentrations (0.465 ± 0.082 vs. 0.570 ± 0.134 μmol/L at baseline, p < 0.001). Patients with AF at the time of blood sampling had significantly lower hArg (1.65 ± 0.51 vs. 1.85 ± 0.60 μmol/L, p = 0.006) and higher ADMA concentrations (0.526 ± 0.08 vs. 0.477 ± 0.08 μmol/L, p < 0.001) compared with AF patients in SR. hArg concentrations were lower in patients with advanced AF progression phenotypes (persistent AF with LVA (p = 0.046)) independent of heart rhythm at blood sampling. Summarizing, arginine metabolism imbalance is associated with AF in general and AF progression and may contribute to associated risk. KEY MESSAGES: • Heart rhythm at blood withdrawal affects ADMA and hArg level in AF patients. • SDMA is higher in AF patients. • SDMA levels normalize after sinus rhythm restoration. • hArg levels decrease in advanced AF progression phenotypes.
Collapse
|
39
|
Kirschner Peretz N, Segal S, Yaniv Y. May the Force Not Be With You During Culture: Eliminating Mechano-Associated Feedback During Culture Preserves Cultured Atrial and Pacemaker Cell Functions. Front Physiol 2020; 11:163. [PMID: 32265724 PMCID: PMC7100534 DOI: 10.3389/fphys.2020.00163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 02/12/2020] [Indexed: 01/24/2023] Open
Abstract
Cultured cardiomyocytes have been shown to possess significant potential as a model for characterization of mechano-Ca2+, mechano-electric, and mechano-metabolic feedbacks in the heart. However, the majority of cultured cardiomyocytes exhibit impaired electrical, mechanical, biochemical, and metabolic functions. More specifically, the cells do not beat spontaneously (pacemaker cells) or beat at a rate far lower than their physiological counterparts and self-oscillate (atrial and ventricular cells) in culture. Thus, efforts are being invested in ensuring that cultured cardiomyocytes maintain the shape and function of freshly isolated cells. Elimination of contraction during culture has been shown to preserve the mechano-Ca2+, mechano-electric, and mechano-metabolic feedback loops of cultured cells. This review focuses on pacemaker cells, which reside in the sinoatrial node (SAN) and generate regular heartbeat through the initiation of the heart’s electrical, metabolic, and biochemical activities. In parallel, it places emphasis on atrial cells, which are responsible for bridging the electrical conductance from the SAN to the ventricle. The review provides a summary of the main mechanisms responsible for mechano-electrical, Ca2+, and metabolic feedback in pacemaker and atrial cells and of culture methods existing for both cell types. The work concludes with an explanation of how the elimination of mechano-electrical, mechano-Ca2+, and mechano-metabolic feedbacks during culture results in sustained cultured cell function.
Collapse
Affiliation(s)
- Noa Kirschner Peretz
- Biomedical Engineering Faculty, Technion Israel Institute of Technology, Haifa, Israel
| | - Sofia Segal
- Biomedical Engineering Faculty, Technion Israel Institute of Technology, Haifa, Israel
| | - Yael Yaniv
- Biomedical Engineering Faculty, Technion Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
40
|
Linking cellular energy state to atrial fibrillation pathogenesis: Potential role of adenosine monophosphate-activated protein kinase. Heart Rhythm 2020; 17:1398-1404. [PMID: 32268208 DOI: 10.1016/j.hrthm.2020.03.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 03/28/2020] [Indexed: 01/01/2023]
Abstract
Adenosine monophosphate-activated protein kinase (AMPK) is the cellular stress-sensing molecule. Apart from maintaining cellular energy balance, AMPK controls expression and regulation of ion channels and ion transporters, including cytosolic Ca2+ handling proteins. Emerging evidence suggests that metabolic impairment plays a crucial role in the pathogenesis of atrial fibrillation. AMPK activation is thought to be protective by preventing metabolic stress, favorably modulating membrane electrophysiology including cytosolic Ca2+ dynamics; preventing cellular growth; and hypertrophic remodeling. This review considers current concepts and evidence from clinical and experimental studies regarding the role of AMPK in atrial fibrillation.
Collapse
|
41
|
Abstract
OBJECTIVE This study investigated the potential microRNAs (miRNAs) having a diagnostic value in atrial fibrillation (AF). METHODS The miRNA and mRNA expression profiles of atrial tissue from healthy individuals and patients with AF were downloaded from the Gene Expression Omnibus database. Differentially expressed miRNAs/mRNAs (DEMis/DEMs) were identified in patients with AF. Furthermore, an interaction network between DEMis and DMEs was constructed. The biological processes, molecular functions, and signaling pathways of DEMs were enriched. Then, the diagnostic values of candidate DECs among healthy individuals and patients with AF were preliminarily evaluated in the GSE101586, GSEE101684, and GSE112214 datasets. RESULTS Twenty DEMis were identified in patients with AF, including seven upregulated and 13 downregulated DEMis. Furthermore, 2,307 DEMs were identified in patients with AF. In the DEMi-DEM interaction network, downregulated miR-193b and upregulated miR-16 interacted with the most targeted DEMs, which interacted with 72 and 65 targeted DEMs, respectively. The targeted DEMs were significantly enriched in biological functions including apoptosis and the PI3K-Akt, mTOR, Hippo, HIF-1, and ErbB signaling pathways. Four of the 20 DEMis (i.e., miR-490-3p, miR-630, miR-146b-5p, and miR-367) had a potential value to distinguish patients with AF from healthy individuals in the GSE68475, GSE70887, and GSE28954 datasets. The area under the curve values for those four DEMis were 0.751, 0.719, 0.709, and 0.7, respectively. CONCLUSION DEMis might play key roles in AF progression through the mTOR and Hippo signaling pathways. miR-409-3p, miR-630, miR-146b-5p, and miR-367 had a potential diagnostic value to discriminate patients with AF from healthy controls in this study.
Collapse
|
42
|
Scholz B, Schulte JS, Hamer S, Himmler K, Pluteanu F, Seidl MD, Stein J, Wardelmann E, Hammer E, Völker U, Müller FU. HDAC (Histone Deacetylase) Inhibitor Valproic Acid Attenuates Atrial Remodeling and Delays the Onset of Atrial Fibrillation in Mice. Circ Arrhythm Electrophysiol 2019; 12:e007071. [PMID: 30879335 PMCID: PMC6426346 DOI: 10.1161/circep.118.007071] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Supplemental Digital Content is available in the text. Background: A structural, electrical and metabolic atrial remodeling is central in the development of atrial fibrillation (AF) contributing to its initiation and perpetuation. In the heart, HDACs (histone deacetylases) control remodeling associated processes like hypertrophy, fibrosis, and energy metabolism. Here, we analyzed, whether the HDAC class I/IIa inhibitor valproic acid (VPA) is able to attenuate atrial remodeling in CREM-IbΔC-X (cAMP responsive element modulator isoform IbΔC-X) transgenic mice, a mouse model of extensive atrial remodeling with age-dependent progression from spontaneous atrial ectopy to paroxysmal and finally long-lasting AF. Methods: VPA was administered for 7 or 25 weeks to transgenic and control mice. Atria were analyzed macroscopically and using widefield and electron microscopy. Action potentials were recorded from atrial cardiomyocytes using patch-clamp technique. ECG recordings documented the onset of AF. A proteome analysis with consecutive pathway mapping identified VPA-mediated proteomic changes and related pathways. Results: VPA attenuated many components of atrial remodeling that are present in transgenic mice, animal AF models, and human AF. VPA significantly (P<0.05) reduced atrial dilatation, cardiomyocyte enlargement, atrial fibrosis, and the disorganization of myocyte’s ultrastructure. It significantly reduced the occurrence of atrial thrombi, reversed action potential alterations, and finally delayed the onset of AF by 4 to 8 weeks. Increased histone H4-acetylation in atria from VPA-treated transgenic mice verified effective in vivo HDAC inhibition. Cardiomyocyte-specific genetic inactivation of HDAC2 in transgenic mice attenuated the ultrastructural disorganization of myocytes comparable to VPA. Finally, VPA restrained dysregulation of proteins in transgenic mice that are involved in a multitude of AF relevant pathways like oxidative phosphorylation or RhoA (Ras homolog gene family, member A) signaling and disease functions like cardiac fibrosis and apoptosis of muscle cells. Conclusions: Our results suggest that VPA, clinically available, well-tolerated, and prescribed to many patients for years, has the therapeutic potential to delay the development of atrial remodeling and the onset of AF in patients at risk.
Collapse
Affiliation(s)
- Beatrix Scholz
- Institute of Pharmacology and Toxicology, University of Münster, Germany (B.S., J.S.S., S.H., K.H., F.P., M.D.S., J.S., F.U.M.)
| | - Jan Sebastian Schulte
- Institute of Pharmacology and Toxicology, University of Münster, Germany (B.S., J.S.S., S.H., K.H., F.P., M.D.S., J.S., F.U.M.)
| | - Sabine Hamer
- Institute of Pharmacology and Toxicology, University of Münster, Germany (B.S., J.S.S., S.H., K.H., F.P., M.D.S., J.S., F.U.M.)
| | - Kirsten Himmler
- Institute of Pharmacology and Toxicology, University of Münster, Germany (B.S., J.S.S., S.H., K.H., F.P., M.D.S., J.S., F.U.M.)
| | - Florentina Pluteanu
- Institute of Pharmacology and Toxicology, University of Münster, Germany (B.S., J.S.S., S.H., K.H., F.P., M.D.S., J.S., F.U.M.)
| | - Matthias Dodo Seidl
- Institute of Pharmacology and Toxicology, University of Münster, Germany (B.S., J.S.S., S.H., K.H., F.P., M.D.S., J.S., F.U.M.)
| | - Juliane Stein
- Institute of Pharmacology and Toxicology, University of Münster, Germany (B.S., J.S.S., S.H., K.H., F.P., M.D.S., J.S., F.U.M.)
| | - Eva Wardelmann
- Gerhard-Domagk-Institute of Pathology, University Hospital Münster, Germany (E.W.)
| | - Elke Hammer
- Interfaculty Institute of Genetics und Functional Genomics, University Medicine Greifswald, Germany (E.H., U.V.).,DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Germany (E.H., U.V.)
| | - Uwe Völker
- Interfaculty Institute of Genetics und Functional Genomics, University Medicine Greifswald, Germany (E.H., U.V.).,DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Germany (E.H., U.V.)
| | - Frank Ulrich Müller
- Institute of Pharmacology and Toxicology, University of Münster, Germany (B.S., J.S.S., S.H., K.H., F.P., M.D.S., J.S., F.U.M.)
| |
Collapse
|
43
|
Li W, Wang L, Wu Y, Yuan Z, Zhou J. Weighted gene co‑expression network analysis to identify key modules and hub genes associated with atrial fibrillation. Int J Mol Med 2019; 45:401-416. [PMID: 31894294 PMCID: PMC6984797 DOI: 10.3892/ijmm.2019.4416] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 11/08/2019] [Indexed: 12/15/2022] Open
Abstract
Atrial fibrillation (AF) is the most common form of cardiac arrhythmia and significantly increases the risks of morbidity, mortality and health care expenditure; however, treatment for AF remains unsatisfactory due to the complicated and incompletely understood underlying mechanisms. In the present study, weighted gene co‑expression network analysis (WGCNA) was conducted to identify key modules and hub genes to determine their potential associations with AF. WGCNA was performed in an AF dataset GSE79768 obtained from the Gene Expression Omnibus, which contained data from paired left and right atria in cardiac patients with persistent AF or sinus rhythm. Differentially expressed gene (DEG) analysis was used to supplement and validate the results of WGCNA. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses were also performed. Green and magenta modules were identified as the most critical modules associated with AF, from which 6 hub genes, acetyl‑CoA Acetyltransferase 1, death domain‑containing protein CRADD, gypsy retrotransposon integrase 1, FTX transcript, XIST regulator, transcription elongation factor A like 2 and minichromosome maintenance complex component 3 associated protein, were hypothesized to serve key roles in the pathophysiology of AF due to their increased intramodular connectivity. Functional enrichment analysis results demonstrated that the green module was associated with energy metabolism, and the magenta module may be associated with the Hippo pathway and contain multiple interactive pathways associated with apoptosis and inflammation. In addition, the blue module was identified to be an important regulatory module in AF with a higher specificity for the left atria, the genes of which were primarily correlated with complement, coagulation and extracellular matrix formation. These results suggest that may improve understanding of the underlying mechanisms of AF, and assist in identifying biomarkers and potential therapeutic targets for treating patients with AF.
Collapse
Affiliation(s)
- Wenyuan Li
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Lijun Wang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yue Wu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Zuyi Yuan
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Juan Zhou
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
44
|
Ciaccio EJ, Wan EY, Saluja DS, Acharya UR, Peters NS, Garan H. Addressing challenges of quantitative methodologies and event interpretation in the study of atrial fibrillation. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2019; 178:113-122. [PMID: 31416540 PMCID: PMC6748794 DOI: 10.1016/j.cmpb.2019.06.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 05/21/2019] [Accepted: 06/14/2019] [Indexed: 05/06/2023]
Abstract
Atrial fibrillation (AF) is the commonest arrhythmia, yet the mechanisms of its onset and persistence are incompletely known. Although techniques for quantitative assessment have been investigated, there have been few attempts to integrate this information to advance disease treatment protocols. In this review, key quantitative methods for AF analysis are described, and suggestions are provided for the coordination of the available information, and to develop foci and directions for future research efforts. Quantitative biologists may have an interest in this topic in order to develop machine learning and tools for arrhythmia characterization, but they may perhaps have a minimal background in the clinical methodology and in the types of observed events and mechanistic hypotheses that have thus far been developed. We attempt to address these issues via exploration of the published literature. Although no new data is presented in this review, examples are shown of current lines of investigation, and in particular, how electrogram analysis and whole-chamber quantitative modeling of the left atrium may be useful to characterize fibrillatory patterns of activity, so as to propose avenues for more efficacious acquisition and interpretation of AF data.
Collapse
Affiliation(s)
- Edward J Ciaccio
- Department of Medicine - Division of Cardiology, Columbia University Medical Center, New York, NY, USA; ElectroCardioMaths Programme, Imperial Centre for Cardiac Engineering, Imperial College London, London, UK.
| | - Elaine Y Wan
- Department of Medicine - Division of Cardiology, Columbia University Medical Center, New York, NY, USA
| | - Deepak S Saluja
- Department of Medicine - Division of Cardiology, Columbia University Medical Center, New York, NY, USA
| | - U Rajendra Acharya
- Department of Electronics and Computer Engineering, Ngee Ann Polytechnic, Singapore
| | - Nicholas S Peters
- ElectroCardioMaths Programme, Imperial Centre for Cardiac Engineering, Imperial College London, London, UK
| | - Hasan Garan
- Department of Medicine - Division of Cardiology, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
45
|
Zhou J, Sun L, Chen L, Liu S, Zhong L, Cui M. Comprehensive metabolomic and proteomic analyses reveal candidate biomarkers and related metabolic networks in atrial fibrillation. Metabolomics 2019; 15:96. [PMID: 31227919 DOI: 10.1007/s11306-019-1557-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 06/15/2019] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Atrial fibrillation (AF) is an abnormal heart rhythm characterized by an irregular beating of the atria and is associated with an increased risk of heart failure, dementia, and stroke. Currently, the perturbation of plasma content due to AF disease onset is not well known. OBJECTIVES To investigate dysregulated molecules in blood plasma of untreated AF patients, with the goal of identifying biomarkers for disease screening and pathological studies. METHODS LC-MS based untargeted metabolomics, lipidomics and proteomics analyses were performed to find candidate biomarkers. A targeted quantification assay and an ELISA were performed to validate the results of the omics analyses. RESULTS We found that 24 metabolites, 16 lipids and 16 proteins were significantly dysregulated in AF patients. Pathway enrichment analysis showed that the purine metabolic pathway and fatty acid metabolism were perturbed by AF onset. FA 20:2 and FA 22:4 show great linear correlational relationship with the left atrial area and could be considered for AF disease stage monitoring or prognosis evaluation. CONCLUSION we used a comprehensive multiple-omics strategy to systematically investigate the dysregulated molecules in the plasma of AF patients, thereby revealing potential biomarkers for diagnosis and providing information for pathological studies.
Collapse
Affiliation(s)
- Juntuo Zhou
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing, 100083, China
| | - Lijie Sun
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, No. 49, Hua Yuan North Rd, Hai Dian District, Beijing, 100191, China
| | - Liwen Chen
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, No. 49, Hua Yuan North Rd, Hai Dian District, Beijing, 100191, China
| | - Shuwang Liu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, No. 49, Hua Yuan North Rd, Hai Dian District, Beijing, 100191, China
| | - Lijun Zhong
- Center of Medical and Health Analysis, Peking University Health Science Center, Beijing, 100191, China.
| | - Ming Cui
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education; Beijing Key Laboratory of Cardiovascular Receptors Research, No. 49, Hua Yuan North Rd, Hai Dian District, Beijing, 100191, China.
| |
Collapse
|
46
|
Chadda KR, Edling CE, Valli H, Ahmad S, Huang CLH, Jeevaratnam K. Gene and Protein Expression Profile of Selected Molecular Targets Mediating Electrophysiological Function in Pgc-1α Deficient Murine Atria. Int J Mol Sci 2018; 19:ijms19113450. [PMID: 30400228 PMCID: PMC6274828 DOI: 10.3390/ijms19113450] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 10/25/2018] [Accepted: 10/29/2018] [Indexed: 12/27/2022] Open
Abstract
Increases in the prevalence of obesity, insulin resistance, and metabolic syndrome has led to the increase of atrial fibrillation (AF) cases in the developed world. These AF risk factors are associated with mitochondrial dysfunction, previously modelled using peroxisome proliferator activated receptor-γ (PPARγ) coactivator-1 (Pgc-1)-deficient murine cardiac models. We explored gene and protein expression profiles of selected molecular targets related to electrophysiological function in murine Pgc-1α−/− atria. qPCR analysis surveyed genes related to Na+-K+-ATPase, K+ conductance, hyperpolarisation-activated cyclic nucleotide-gated (Hcn), Na+ channels, Ca2+ channels, and indicators for adrenergic and cholinergic receptor modulation. Western blot analysis for molecular targets specific to conduction velocity (Nav1.5 channel and gap junctions) was performed. Transcription profiles revealed downregulation of molecules related to Na+-K+-ATPase transport, Hcn-dependent pacemaker function, Na+ channel-dependent action potential activation and propagation, Ca2+ current generation, calsequestrin-2 dependent Ca2+ homeostasis, and adrenergic α1D dependent protection from hypertrophic change. Nav1.5 channel protein expression but not gap junction expression was reduced in Pgc-1α−/− atria compared to WT. Nav1.5 reduction reflects corresponding reduction in its gene expression profile. These changes, as well as the underlying Pgc-1α−/− alteration, suggest potential pharmacological targets directed towards either upstream PGC-1 signalling mechanisms or downstream ion channel changes.
Collapse
Affiliation(s)
- Karan R Chadda
- Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7AL, UK.
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK.
| | - Charlotte E Edling
- Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7AL, UK.
| | - Haseeb Valli
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK.
| | - Shiraz Ahmad
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK.
| | - Christopher L-H Huang
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK.
- Department of Biochemistry, Hopkins Building, University of Cambridge, Cambridge CB2 1QW, UK.
| | - Kamalan Jeevaratnam
- Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7AL, UK.
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK.
- School of Medicine, Perdana University-Royal College of Surgeons Ireland, Serdang 43400, Malaysia.
| |
Collapse
|
47
|
Liu Y, Shi Q, Ma Y, Liu Q. The role of immune cells in atrial fibrillation. J Mol Cell Cardiol 2018; 123:198-208. [DOI: 10.1016/j.yjmcc.2018.09.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 09/19/2018] [Accepted: 09/20/2018] [Indexed: 12/23/2022]
|
48
|
Resveratrol: an effective pharmacological agent to prevent inflammation-induced atrial fibrillation? NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2018; 391:1163-1167. [PMID: 30238135 DOI: 10.1007/s00210-018-1566-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 09/11/2018] [Indexed: 10/28/2022]
|
49
|
Affiliation(s)
- Jordi Heijman
- From the Department of Cardiology, Cardiovascular Research Institute Maastricht, Faculty of Health, Medicine, and Life Sciences, Maastricht University, The Netherlands (J.H.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Canada (J.-B.G., S.N.); University Hospital of Saint-Étienne, University Jean Monnet, Saint-Étienne, France (J.-B.G.); Institute of Pharmacology, West German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen (D.D., S.N.); and
| | - Jean-Baptiste Guichard
- From the Department of Cardiology, Cardiovascular Research Institute Maastricht, Faculty of Health, Medicine, and Life Sciences, Maastricht University, The Netherlands (J.H.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Canada (J.-B.G., S.N.); University Hospital of Saint-Étienne, University Jean Monnet, Saint-Étienne, France (J.-B.G.); Institute of Pharmacology, West German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen (D.D., S.N.); and
| | - Dobromir Dobrev
- From the Department of Cardiology, Cardiovascular Research Institute Maastricht, Faculty of Health, Medicine, and Life Sciences, Maastricht University, The Netherlands (J.H.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Canada (J.-B.G., S.N.); University Hospital of Saint-Étienne, University Jean Monnet, Saint-Étienne, France (J.-B.G.); Institute of Pharmacology, West German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen (D.D., S.N.); and
| | - Stanley Nattel
- From the Department of Cardiology, Cardiovascular Research Institute Maastricht, Faculty of Health, Medicine, and Life Sciences, Maastricht University, The Netherlands (J.H.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Canada (J.-B.G., S.N.); University Hospital of Saint-Étienne, University Jean Monnet, Saint-Étienne, France (J.-B.G.); Institute of Pharmacology, West German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen (D.D., S.N.); and
| |
Collapse
|