1
|
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
|
2
|
Pagonas N, Mueller R, Weiland L, Jaensch M, Dammermann W, Seibert FS, Hillmeister P, Buschmann I, Christ M, Ritter O, Westhoff TH, Sasko B, Kelesidis T. Oxidized high-density lipoprotein associates with atrial fibrillation. Heart Rhythm 2024; 21:362-369. [PMID: 38040404 PMCID: PMC11073573 DOI: 10.1016/j.hrthm.2023.11.024] [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: 08/17/2023] [Revised: 11/07/2023] [Accepted: 11/24/2023] [Indexed: 12/03/2023]
Abstract
BACKGROUND Atrial fibrillation (AF) is the most common heart arrhythmia and considered to be a progressive chronic disease associated with increased morbidity and mortality. Recent data suggest a link between inflammation, oxidative stress, and AF, although the underlying mechanisms are not fully understood. Because oxidized lipoproteins cause structural damage and electrophysiologic changes in cardiomyocytes, it is feasible that the transformation of atheroprotective high-density lipoprotein (HDL) into dysfunctional HDL contributes to the development of AF. OBJECTIVE The purpose of this study was to determine whether a reduced antioxidant function of HDL is associated with the presence of AF. METHODS In this multicenter cross-sectional cohort study, we assessed HDL function in sera of 1206 participants. Patients were divided into groups according to the presence of AF (n = 233) or no AF (n = 973). A validated cell-free biochemical assay was used to determine reduced HDL antioxidant function as assessed by increased normalized HDL lipid peroxide content (nHDLox). RESULTS Participants with AF had a 9% higher mean relative nHDLox compared to persons without AF (P = .025). nHDLox was strongly associated with AF in all models of logistic regression, including the analysis adjusted for age, sex, and risk factors for AF (all P ≤.01). CONCLUSION Reduced antioxidant HDL function is associated with the presence of AF, which supports growing evidence that impaired lipoprotein function is linked to electrophysiological changes in cardiomyocytes. nHDLox is one of several contributors to the initiation and perpetuation of AF.
Collapse
Affiliation(s)
- Nikolaos Pagonas
- Department of Cardiology, University Hospital Ruppin-Brandenburg, Medical School Theodor Fontane, Neuruppin, Germany; Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology Cottbus-Senftenberg, The (MHB) Theodor Fontane and the University of Potsdam, Potsdam, Germany.
| | - Rhea Mueller
- Department of Cardiology, University Medical Center Brandenburg an der Havel, Medical School Theodor Fontane, Brandenburg an der Havel, Germany
| | - Linda Weiland
- Department of Cardiology, University Hospital Ruppin-Brandenburg, Medical School Theodor Fontane, Neuruppin, Germany
| | - Monique Jaensch
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology Cottbus-Senftenberg, The (MHB) Theodor Fontane and the University of Potsdam, Potsdam, Germany; Department of Cardiology, University Medical Center Brandenburg an der Havel, Medical School Theodor Fontane, Brandenburg an der Havel, Germany
| | - Werner Dammermann
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology Cottbus-Senftenberg, The (MHB) Theodor Fontane and the University of Potsdam, Potsdam, Germany; Center for Internal Medicine II, University Medical Center Brandenburg an der Havel, Medical School Theodor Fontane, Brandenburg an der Havel, Germany
| | - Felix S Seibert
- Medical Department I, Marien Hospital Herne, Ruhr-University of Bochum, Herne, Germany
| | - Philipp Hillmeister
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology Cottbus-Senftenberg, The (MHB) Theodor Fontane and the University of Potsdam, Potsdam, Germany; Department of Angiology, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
| | - Ivo Buschmann
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology Cottbus-Senftenberg, The (MHB) Theodor Fontane and the University of Potsdam, Potsdam, Germany; Department of Angiology, Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
| | - Martin Christ
- Department of Cardiology, Knappschaftskrankenhaus Bottrop, Academic Teaching Hospital, University Duisburg-Essen, Bottrop, Germany
| | - Oliver Ritter
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology Cottbus-Senftenberg, The (MHB) Theodor Fontane and the University of Potsdam, Potsdam, Germany; Department of Cardiology, University Medical Center Brandenburg an der Havel, Medical School Theodor Fontane, Brandenburg an der Havel, Germany
| | - Timm H Westhoff
- Medical Department I, Marien Hospital Herne, Ruhr-University of Bochum, Herne, Germany
| | - Benjamin Sasko
- Department of Cardiology, University Medical Center Brandenburg an der Havel, Medical School Theodor Fontane, Brandenburg an der Havel, Germany; Medical Department II, Marien Hospital Herne, Ruhr-University of Bochum, Herne, Germany
| | - Theodoros Kelesidis
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, UT Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
3
|
Peng Y, Su P, Zhao L. Long noncoding RNA and messenger RNA profiling in epicardial adipose tissue of patients with new-onset postoperative atrial fibrillation after coronary artery bypass grafting. Eur J Med Res 2024; 29:134. [PMID: 38368363 PMCID: PMC10874008 DOI: 10.1186/s40001-024-01721-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 02/09/2024] [Indexed: 02/19/2024] Open
Abstract
BACKGROUND Postoperative atrial fibrillation (POAF) constitutes a significant complication following coronary artery bypass graft surgery (CABG), potentially linked to epicardial adipose tissue (EAT). This investigation seeks to elucidate the association between POAF and EAT at the genetic level. METHODS EAT and clinical data from patients undergoing CABG were systematically acquired, adhering to established inclusion and exclusion criteria. Patients were categorized into POAF and Non-POAF groups based on the presence or absence of POAF. High-throughput sequencing data of EAT were subjected to differential expression analysis and gene function assessment. A random selection of long noncoding RNAs (lncRNAs) underwent quantitative real-time polymerase chain reaction (qRT-PCR) for validation of the high-throughput sequencing findings. Coexpression analysis was employed to elucidate the interactions between lncRNAs and messenger RNAs (mRNAs). RESULTS RNA sequencing yielded a total of 69,685 transcripts (37,740 coding and 31,945 noncoding sequences), representing 16,920 genes. Within this dataset, 38 mRNAs and 12 lncRNAs exhibited differential expression between the POAF and Non-POAF groups (P < 0.05, fold change > 1.5). The qRT-PCR results for lncRNAs corroborated the sequencing findings (P < 0.01). Functional enrichment analysis of genes and the coexpression network indicated that these differentially expressed RNAs were primarily implicated in processes such as cell growth, differentiation, signal transduction, as well as influencing tissue fibrosis and ion transmembrane transport. CONCLUSIONS This study unveils a potential association between myocardial fibrosis and ion channels co-regulated by mRNAs and lncRNAs, closely linked to the emergence of new-onset POAF, after accounting for clinical risk factors. This discovery holds promise for further advances in clinical and fundamental research.
Collapse
Affiliation(s)
- Yuanshu Peng
- Heart Center & Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Pixiong Su
- Heart Center & Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China.
| | - Lei Zhao
- Heart Center & Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China.
| |
Collapse
|
4
|
Bialobroda J, Bouazizi K, Ponnaiah M, Kachenoura N, Charpentier E, Zarai M, Clement K, Andreelli F, Aron-Wisnewsky J, Hatem SN, Redheuil A. The epicardial adipose tissue confined in the atrioventricular groove can be used to assess atrial adipose tissue and atrial dysfunction in cardiac magnetic resonance imaging. EUROPEAN HEART JOURNAL. IMAGING METHODS AND PRACTICE 2024; 2:qyae057. [PMID: 39224099 PMCID: PMC11367945 DOI: 10.1093/ehjimp/qyae057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/17/2024] [Indexed: 09/04/2024]
Abstract
Aims The growing interest in epicardial adipose tissue (EAT) as a biomarker of atrial fibrillation is limited by the difficulties in isolating EAT from other paracardial adipose tissues. We tested the feasibility and value of measuring the pure EAT contained in the atrioventricular groove (GEAT) using cardiovascular magnetic resonance (CMR) imaging in patients with distinct metabolic disorders. Methods and results CMR was performed on 100 patients from the MetaCardis cohort: obese (n = 18), metabolic syndrome (MSD) (n = 25), type-2 diabetes (T2D) (n = 42), and age- and gender-matched healthy controls (n = 15). GEAT volume measured from long-axis views was obtained in all patients with a strong correlation between GEAT and atrial EAT (r = 0.95; P < 0.0001). GEAT volume was higher in the three groups of patients with metabolic disorders and highest in the MSD group compared with controls. GEAT volume, as well as body mass and body fat, allowed obese, T2D, and MSD patients to be distinguished from controls. GEAT T1 relaxation and peak longitudinal left atrial (LA) strain in CMR were decreased in T2D patients. Logistic regression and random forest machine learning methods were used to create an algorithm combining GEAT volume, GEAT T1, and peak LA strain to identify T2D patients from other groups with an area under curve (AUC) of 0.81 (Se: 77%, Spe: 80%; 95% confidence interval 0.72-0.91, P < 0.0001). Conclusion Atrioventricular groove adipose tissue characteristics measured during routine CMR can be used as a proxy of atrial EAT and integrated in a multi-parametric CMR biomarker for early identification of atrial cardiomyopathy.
Collapse
Affiliation(s)
- Jonathan Bialobroda
- Institute of Cardiology, Foundation for Innovation in Cardiometabolism and Nutrition—ICAN, INSERM UMRS 1166, Sorbonne Université, AP-HP Pitié-Salpêtrière University Hospital, 47-83, Boulevard de l’Hôpital, 75013 Paris, France
| | - Khaoula Bouazizi
- Laboratoire d’Imagerie Biomédicale, CNRS, INSERM UMR 1146, Sorbonne Université, Paris, France
- Foundation for Innovation in Cardiometabolism and Nutrition (IHU-ICAN, ANR-10-IAHU-05), 47-83, Boulevard de l’Hôpital, 75013 Paris, France
| | - Maharajah Ponnaiah
- Foundation for Innovation in Cardiometabolism and Nutrition (IHU-ICAN, ANR-10-IAHU-05), 47-83, Boulevard de l’Hôpital, 75013 Paris, France
| | - Nadjia Kachenoura
- Laboratoire d’Imagerie Biomédicale, CNRS, INSERM UMR 1146, Sorbonne Université, Paris, France
| | - Etienne Charpentier
- Institute of Cardiometabolism and Nutrition—ICAN, INSERM UMR 1146, Laboratoire d’Imagerie Biomédicale, Unité D’Imagerie Cardiovasculaire et Thoracique, Sorbonne Université, AP-HP Hôpital Pitié-Salpêtrière, Paris, France
| | - Mohamed Zarai
- Institute of Cardiometabolism and Nutrition—ICAN, INSERM UMR 1146, Laboratoire d’Imagerie Biomédicale, Unité D’Imagerie Cardiovasculaire et Thoracique, Sorbonne Université, AP-HP Hôpital Pitié-Salpêtrière, Paris, France
| | - Karine Clement
- Nutrition and Obesity: Systemic Approaches, NutriOmics, Sorbonne Université, INSERM, Paris, France
- Department of Nutrition, Sorbonne Université, Assistance Publique- Hôpitaux de Paris, AP-HP, Pitié-Salpêtrière University Hospital, 47-83 Boulevard de l'Hôpital, 75013 Paris, France
| | - Fabrizio Andreelli
- Nutrition and Obesity: Systemic Approaches, NutriOmics, Sorbonne Université, INSERM, Paris, France
- Department of Diabetology, AP-HP Pitié-Salpêtrière University Hospital, Sorbonne Université, Paris, France
| | - Judith Aron-Wisnewsky
- Nutrition and Obesity: Systemic Approaches, NutriOmics, Sorbonne Université, INSERM, Paris, France
- Department of Nutrition, Sorbonne Université, Assistance Publique- Hôpitaux de Paris, AP-HP, Pitié-Salpêtrière University Hospital, 47-83 Boulevard de l'Hôpital, 75013 Paris, France
| | - Stéphane N Hatem
- Institute of Cardiology, Foundation for Innovation in Cardiometabolism and Nutrition—ICAN, INSERM UMRS 1166, Sorbonne Université, AP-HP Pitié-Salpêtrière University Hospital, 47-83, Boulevard de l’Hôpital, 75013 Paris, France
- Foundation for Innovation in Cardiometabolism and Nutrition (IHU-ICAN, ANR-10-IAHU-05), 47-83, Boulevard de l’Hôpital, 75013 Paris, France
| | - Alban Redheuil
- Institute of Cardiometabolism and Nutrition—ICAN, INSERM UMR 1146, Laboratoire d’Imagerie Biomédicale, Unité D’Imagerie Cardiovasculaire et Thoracique, Sorbonne Université, AP-HP Hôpital Pitié-Salpêtrière, Paris, France
| |
Collapse
|
5
|
Atzemian N, Dovrolis N, Ragia G, Portokallidou K, Kolios G, Manolopoulos VG. Beyond the Rhythm: In Silico Identification of Key Genes and Therapeutic Targets in Atrial Fibrillation. Biomedicines 2023; 11:2632. [PMID: 37893006 PMCID: PMC10604372 DOI: 10.3390/biomedicines11102632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/21/2023] [Accepted: 09/22/2023] [Indexed: 10/29/2023] Open
Abstract
Atrial fibrillation (AF) is a prevalent cardiac arrhythmia worldwide and is characterized by a high risk of thromboembolism, ischemic stroke, and fatality. The precise molecular mechanisms of AF pathogenesis remain unclear. The purpose of this study was to use bioinformatics tools to identify novel key genes in AF, provide deeper insights into the molecular pathogenesis of AF, and uncover potential therapeutic targets. Four publicly available raw RNA-Seq datasets obtained through the ENA Browser, as well as proteomic analysis results, both derived from atrial tissues, were used in this analysis. Differential gene expression analysis was performed and cross-validated with proteomics results to identify common genes/proteins between them. A functional enrichment pathway analysis was performed. Cross-validation analysis revealed five differentially expressed genes, namely FGL2, IGFBP5, NNMT, PLA2G2A, and TNC, in patients with AF compared with those with sinus rhythm (SR). These genes play crucial roles in various cardiovascular functions and may be part of the molecular signature of AF. Furthermore, functional enrichment analysis revealed several pathways related to the extracellular matrix, inflammation, and structural remodeling. This study highlighted five key genes that constitute promising candidates for further experimental exploration as biomarkers as well as therapeutic targets for AF.
Collapse
Affiliation(s)
- Natalia Atzemian
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (N.A.); (G.R.); (K.P.); (G.K.)
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
| | - Nikolas Dovrolis
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (N.A.); (G.R.); (K.P.); (G.K.)
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
| | - Georgia Ragia
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (N.A.); (G.R.); (K.P.); (G.K.)
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
| | - Konstantina Portokallidou
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (N.A.); (G.R.); (K.P.); (G.K.)
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
| | - George Kolios
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (N.A.); (G.R.); (K.P.); (G.K.)
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
| | - Vangelis G. Manolopoulos
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (N.A.); (G.R.); (K.P.); (G.K.)
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
- Clinical Pharmacology Unit, Academic General Hospital of Alexandroupolis, 68100 Alexandroupolis, Greece
| |
Collapse
|
6
|
Huiskes FG, Creemers EE, Brundel BJJM. Dissecting the Molecular Mechanisms Driving Electropathology in Atrial Fibrillation: Deployment of RNA Sequencing and Transcriptomic Analyses. Cells 2023; 12:2242. [PMID: 37759465 PMCID: PMC10526291 DOI: 10.3390/cells12182242] [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: 07/15/2023] [Revised: 08/30/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
Despite many efforts to treat atrial fibrillation (AF), the most common progressive and age-related cardiac tachyarrhythmia in the Western world, the efficacy is still suboptimal. A plausible reason for this is that current treatments are not directed at underlying molecular root causes that drive electrical conduction disorders and AF (i.e., electropathology). Insights into AF-induced transcriptomic alterations may aid in a deeper understanding of electropathology. Specifically, RNA sequencing (RNA-seq) facilitates transcriptomic analyses and discovery of differences in gene expression profiles between patient groups. In the last decade, various RNA-seq studies have been conducted in atrial tissue samples of patients with AF versus controls in sinus rhythm. Identified differentially expressed molecular pathways so far include pathways related to mechanotransduction, ECM remodeling, ion channel signaling, and structural tissue organization through developmental and inflammatory signaling pathways. In this review, we provide an overview of the available human AF RNA-seq studies and highlight the molecular pathways identified. Additionally, a comparison is made between human RNA-seq findings with findings from experimental AF model systems and we discuss contrasting findings. Finally, we elaborate on new exciting RNA-seq approaches, including single-nucleotide variants, spatial transcriptomics and profiling of different populations of total RNA, small RNA and long non-coding RNA.
Collapse
Affiliation(s)
- Fabries G. Huiskes
- Department of Physiology, Amsterdam UMC, Location Vrije Universiteit, VUmc, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, 1081 HZ, Amsterdam, The Netherlands;
- Department of Experimental Cardiology, Amsterdam UMC, Location AMC, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, 1105 AZ Amsterdam, The Netherlands;
| | - Esther E. Creemers
- Department of Experimental Cardiology, Amsterdam UMC, Location AMC, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, 1105 AZ Amsterdam, The Netherlands;
| | - Bianca J. J. M. Brundel
- Department of Physiology, Amsterdam UMC, Location Vrije Universiteit, VUmc, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, 1081 HZ, Amsterdam, The Netherlands;
| |
Collapse
|
7
|
Gawałko M, Saljic A, Li N, Abu-Taha I, Jespersen T, Linz D, Nattel S, Heijman J, Fender A, Dobrev D. Adiposity-associated atrial fibrillation: molecular determinants, mechanisms, and clinical significance. Cardiovasc Res 2023; 119:614-630. [PMID: 35689487 PMCID: PMC10409902 DOI: 10.1093/cvr/cvac093] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/22/2022] [Accepted: 03/23/2022] [Indexed: 12/12/2022] Open
Abstract
Obesity is an important contributing factor to the pathophysiology of atrial fibrillation (AF) and its complications by causing systemic changes, such as altered haemodynamic, increased sympathetic tone, and low-grade chronic inflammatory state. In addition, adipose tissue is a metabolically active organ that comprises various types of fat deposits with discrete composition and localization that show distinct functions. Fatty tissue differentially affects the evolution of AF, with highly secretory active visceral fat surrounding the heart generally having a more potent influence than the rather inert subcutaneous fat. A variety of proinflammatory, profibrotic, and vasoconstrictive mediators are secreted by adipose tissue, particularly originating from cardiac fat, that promote atrial remodelling and increase the susceptibility to AF. In this review, we address the role of obesity-related factors and in particular specific adipose tissue depots in driving AF risk. We discuss the distinct effects of key secreted adipokines from different adipose tissue depots and their participation in cardiac remodelling. The possible mechanistic basis and molecular determinants of adiposity-related AF are discussed, and finally, we highlight important gaps in current knowledge, areas requiring future investigation, and implications for clinical management.
Collapse
Affiliation(s)
- Monika Gawałko
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany
- 1st Department of Cardiology, Medical University of Warsaw, Banacha 1A, 02-197 Warsaw, Poland
- Department of Cardiology, Maastricht University Medical Centre and Cardiovascular Research Institute Maastricht, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Arnela Saljic
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Na Li
- Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
- Cardiovascular Research Institute, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Issam Abu-Taha
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany
| | - Thomas Jespersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Dominik Linz
- Department of Cardiology, Maastricht University Medical Centre and Cardiovascular Research Institute Maastricht, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
- Centre for Heart Rhythm Disorders, Royal Adelaide Hospital, University of Adelaide, Port Road, SA 5000 Adelaide, Australia
- Department of Cardiology, Radboud University Medical Centre, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Stanley Nattel
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany
- Medicine and Research Center, Montréal Heart Institute and University de Montréal, 3655 Promenade Sir William Osler, Montreal, QC H3G 1Y6, Canada
- IHU LIRYC Institute, Avenue du Haut Lévêque, 33600 Pessac, Bordeaux, France
| | - Jordi Heijman
- Department of Cardiology, Maastricht University Medical Centre and Cardiovascular Research Institute Maastricht, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Anke Fender
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
- Medicine and Research Center, Montréal Heart Institute and University de Montréal, 3655 Promenade Sir William Osler, Montreal, QC H3G 1Y6, Canada
| |
Collapse
|
8
|
Goette A. Does epicardial fat modify the effect of catheter ablation in patients with atrial fibrillation? Int J Cardiol 2023:S0167-5273(23)00473-4. [PMID: 37028713 DOI: 10.1016/j.ijcard.2023.03.064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 03/28/2023] [Indexed: 04/09/2023]
Affiliation(s)
- Andreas Goette
- Department of Cardiology and Intensive Care Medicine, St. Vincenz Hospital, Paderborn, Germany.
| |
Collapse
|
9
|
Takahashi N, Abe I, Kira S, Ishii Y. Role of epicardial adipose tissue in human atrial fibrillation. J Arrhythm 2023; 39:93-110. [PMID: 37021018 PMCID: PMC10068928 DOI: 10.1002/joa3.12825] [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: 12/27/2022] [Revised: 01/19/2023] [Accepted: 01/28/2023] [Indexed: 02/22/2023] Open
Abstract
A recent meta-analysis among which four reports were conducted in Japan demonstrated that epicardial adipose tissue (EAT) is closely associated with an increased risk of atrial fibrillation (AF) recurrence after catheter ablation. We previously investigated the role of EAT in AF in humans. Left atrial (LA) appendage samples were obtained from AF patients during cardiovascular surgery. Histologically, the severity of fibrotic EAT remodeling was associated with LA myocardial fibrosis. Total collagen in the LA myocardium (i.e., LA myocardial fibrosis) was positively correlated with proinflammatory and profibrotic cytokines/chemokines, including interleukin-6, monocyte chemoattractant protein-1, and tumor necrosis factor-α, in EAT. Human peri-LA EAT and abdominal subcutaneous adipose tissue (SAT) were obtained by autopsy. EAT- or SAT-derived conditioned medium was applied to the rat LA epicardial surface using an organo-culture system. EAT-conditioned medium induced atrial fibrosis in organo-cultured rat atrium. The profibrotic effect of EAT was greater than that of SAT. The fibrotic area of the organo-cultured rat atrium treated with EAT from patients with AF was greater than in patients without AF. Treatment with human recombinant angiopoietin-like protein 2 (Angptl2) induced fibrosis in organo-cultured rat atrium, which was suppressed by concomitant treatment with anti-Angptl2 antibody. Finally, we attempted to detect fibrotic EAT remodeling on computed tomography (CT) images, which demonstrated that the percent change in EAT fat attenuation was positively correlated with EAT fibrosis. Based on these findings, we conclude that the percent change in EAT fat attenuation determined using CT non-invasively detects EAT remodeling.
Collapse
Affiliation(s)
- Naohiko Takahashi
- Department of Cardiology and Clinical ExaminationOita University Faculty of MedicineOitaJapan
| | - Ichitaro Abe
- Department of Cardiology and Clinical ExaminationOita University Faculty of MedicineOitaJapan
| | - Shintaro Kira
- Department of Cardiology and Clinical ExaminationOita University Faculty of MedicineOitaJapan
| | - Yumi Ishii
- Department of Cardiology and Clinical ExaminationOita University Faculty of MedicineOitaJapan
| |
Collapse
|
10
|
Sang C, Hu X, Zhang D, Shao Y, Qiu B, Li C, Li F, Zhang C, Wang Z, Chen M. The predictive value of left atrium epicardial adipose tissue on recurrence after catheter ablation in patients with different types of atrial fibrillation. Int J Cardiol 2023; 379:33-39. [PMID: 36893857 DOI: 10.1016/j.ijcard.2023.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 02/28/2023] [Accepted: 03/05/2023] [Indexed: 03/09/2023]
Abstract
BACKGROUND A growing body of evidence supports that the left atrium epicardial adipose tissue (LA-EAT) is related to the occurrence and recurrence of atrial fibrillation (AF). The relationship between LA-EAT and the recurrence after radiofrequency catheter ablation (RFCA) in patients with different types of AF is still unclear. This study aims to evaluate the predictive value of LA-EAT on the recurrence of AF after RFCA in patients with different types of AF. METHODS 301 AF patients who underwent RFCA for the first time were divided into the paroxysmal atrial fibrillation (PAF) group (n = 181) and the persistent atrial fibrillation (PersAF) group(n = 120), which were followed up at 3, 6, and 12 months. All patients underwent left atrial computed tomography angiography (CTA) examination before the operation, and LA-EAT was measured using software (Advantage Workstation4.6, GE, USA). RESULTS After a median follow-up of 10.7 months, 73/301 patients (24.25%) had a recurrence of AF, including 43 /120(35.83%) patients with PersAF and 30/181(16.57%) patients with PAF. In multivariable Cox regression analysis, LA-EAT volume (OR = 1.053;95%CI: 1.024-1.083, p < 0.001), attenuation (OR = 0.949;95%CI:0.911-0.988, p = 0.012) and left atrial diameter (LAD) (OR = 1.063;95%CI:1.002-1.127,p = 0.043) were independent risk factors for recurrence in patients with PersAF but not in patients with PAF. CONCLUSION LA-EAT volume and attenuation are independent risk factors for recurrence after RFCA in patients with PersAF.
Collapse
Affiliation(s)
- Chuanyi Sang
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Xiaoqin Hu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Dongdong Zhang
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Yameng Shao
- Department of Cardiology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang 471000, China
| | - Bowen Qiu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Chengzong Li
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Fei Li
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Chaoqun Zhang
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China.
| | - Zhirong Wang
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Minglong Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
11
|
Suffee N, Baptista E, Piquereau J, Ponnaiah M, Doisne N, Ichou F, Lhomme M, Pichard C, Galand V, Mougenot N, Dilanian G, Lucats L, Balse E, Mericskay M, Le Goff W, Hatem SN. Impacts of a high-fat diet on the metabolic profile and the phenotype of atrial myocardium in mice. Cardiovasc Res 2022; 118:3126-3139. [PMID: 34971360 DOI: 10.1093/cvr/cvab367] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 12/27/2021] [Indexed: 12/16/2022] Open
Abstract
AIMS Obesity, diabetes, and metabolic syndromes are risk factors of atrial fibrillation (AF). We tested the hypothesis that metabolic disorders have a direct impact on the atria favouring the formation of the substrate of AF. METHODS AND RESULTS Untargeted metabolomic and lipidomic analysis was used to investigate the consequences of a prolonged high-fat diet (HFD) on mouse atria. Atrial properties were characterized by measuring mitochondria respiration in saponin-permeabilized trabeculae, by recording action potential (AP) with glass microelectrodes in trabeculae and ionic currents in myocytes using the perforated configuration of patch clamp technique and by several immuno-histological and biochemical approaches. After 16 weeks of HFD, obesogenic mice showed a vulnerability to AF. The atrial myocardium acquired an adipogenic and inflammatory phenotypes. Metabolomic and lipidomic analysis revealed a profound transformation of atrial energy metabolism with a predominance of long-chain lipid accumulation and beta-oxidation activation in the obese mice. Mitochondria respiration showed an increased use of palmitoyl-CoA as energy substrate. APs were short duration and sensitive to the K-ATP-dependent channel inhibitor, whereas K-ATP current was enhanced in isolated atrial myocytes of obese mouse. CONCLUSION HFD transforms energy metabolism, causes fat accumulation, and induces electrical remodelling of the atrial myocardium of mice that become vulnerable to AF.
Collapse
Affiliation(s)
- Nadine Suffee
- INSERM UMRS1166, ICAN-Institute of Cardiometabolism and Nutrition, Sorbonne University, Paris, France
| | - Elodie Baptista
- INSERM UMRS1166, ICAN-Institute of Cardiometabolism and Nutrition, Sorbonne University, Paris, France
| | - Jérôme Piquereau
- ICANalytics, Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
| | - Maharajah Ponnaiah
- ICANalytics, Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
| | - Nicolas Doisne
- INSERM UMRS1166, ICAN-Institute of Cardiometabolism and Nutrition, Sorbonne University, Paris, France
| | - Farid Ichou
- ICANalytics, Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
| | - Marie Lhomme
- Paris-Saclay University, Inserm UMRS 1180 Signaling and Cardiovascular Pathophysiology, Châtenay-Malabry, France
| | - Camille Pichard
- INSERM UMRS1166, ICAN-Institute of Cardiometabolism and Nutrition, Sorbonne University, Paris, France
| | - Vincent Galand
- INSERM UMRS1166, ICAN-Institute of Cardiometabolism and Nutrition, Sorbonne University, Paris, France
| | - Nathalie Mougenot
- INSERM UMR_S28, Faculté de médecine Sorbonne University, Paris, France
| | - Gilles Dilanian
- INSERM UMRS1166, ICAN-Institute of Cardiometabolism and Nutrition, Sorbonne University, Paris, France
| | - Laurence Lucats
- Sanofi-Aventis R&D, Cardiovascular and Metabolism Research, Chilly-Mazarin, France
| | - Elise Balse
- INSERM UMRS1166, ICAN-Institute of Cardiometabolism and Nutrition, Sorbonne University, Paris, France
| | - Mathias Mericskay
- Paris-Saclay University, Inserm UMRS 1180 Signaling and Cardiovascular Pathophysiology, Châtenay-Malabry, France
| | - Wilfried Le Goff
- INSERM UMRS1166, ICAN-Institute of Cardiometabolism and Nutrition, Sorbonne University, Paris, France
| | - Stéphane N Hatem
- INSERM UMRS1166, ICAN-Institute of Cardiometabolism and Nutrition, Sorbonne University, Institute of Cardiology, Pitié-Salpêtrière Hospital, Paris, France
| |
Collapse
|
12
|
Schnabel RB, Marinelli EA, Arbelo E, Boriani G, Boveda S, Buckley CM, Camm AJ, Casadei B, Chua W, Dagres N, de Melis M, Desteghe L, Diederichsen SZ, Duncker D, Eckardt L, Eisert C, Engler D, Fabritz L, Freedman B, Gillet L, Goette A, Guasch E, Svendsen JH, Hatem SN, Haeusler KG, Healey JS, Heidbuchel H, Hindricks G, Hobbs FDR, Hübner T, Kotecha D, Krekler M, Leclercq C, Lewalter T, Lin H, Linz D, Lip GYH, Løchen ML, Lucassen W, Malaczynska-Rajpold K, Massberg S, Merino JL, Meyer R, Mont L, Myers MC, Neubeck L, Niiranen T, Oeff M, Oldgren J, Potpara TS, Psaroudakis G, Pürerfellner H, Ravens U, Rienstra M, Rivard L, Scherr D, Schotten U, Shah D, Sinner MF, Smolnik R, Steinbeck G, Steven D, Svennberg E, Thomas D, True Hills M, van Gelder IC, Vardar B, Palà E, Wakili R, Wegscheider K, Wieloch M, Willems S, Witt H, Ziegler A, Daniel Zink M, Kirchhof P. Early diagnosis and better rhythm management to improve outcomes in patients with atrial fibrillation: the 8th AFNET/EHRA consensus conference. Europace 2022; 25:6-27. [PMID: 35894842 PMCID: PMC9907557 DOI: 10.1093/europace/euac062] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
Despite marked progress in the management of atrial fibrillation (AF), detecting AF remains difficult and AF-related complications cause unacceptable morbidity and mortality even on optimal current therapy. This document summarizes the key outcomes of the 8th AFNET/EHRA Consensus Conference of the Atrial Fibrillation NETwork (AFNET) and the European Heart Rhythm Association (EHRA). Eighty-three international experts met in Hamburg for 2 days in October 2021. Results of the interdisciplinary, hybrid discussions in breakout groups and the plenary based on recently published and unpublished observations are summarized in this consensus paper to support improved care for patients with AF by guiding prevention, individualized management, and research strategies. The main outcomes are (i) new evidence supports a simple, scalable, and pragmatic population-based AF screening pathway; (ii) rhythm management is evolving from therapy aimed at improving symptoms to an integrated domain in the prevention of AF-related outcomes, especially in patients with recently diagnosed AF; (iii) improved characterization of atrial cardiomyopathy may help to identify patients in need for therapy; (iv) standardized assessment of cognitive function in patients with AF could lead to improvement in patient outcomes; and (v) artificial intelligence (AI) can support all of the above aims, but requires advanced interdisciplinary knowledge and collaboration as well as a better medico-legal framework. Implementation of new evidence-based approaches to AF screening and rhythm management can improve outcomes in patients with AF. Additional benefits are possible with further efforts to identify and target atrial cardiomyopathy and cognitive impairment, which can be facilitated by AI.
Collapse
Affiliation(s)
- Renate B Schnabel
- Atrial Fibrillation Network (AFNET), Muenster, Germany,Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,German Centre for Cardiovascular Research (DZHK) partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | | | - Elena Arbelo
- Arrhythmia Section, Cardiology Department, Hospital Clinic, Universitat de Barcelona, Barcelona, Spain,IDIBAPS, Institut d'Investigació August Pi i Sunyer, Barcelona, Spain,CIBERCV, Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain
| | - Giuseppe Boriani
- Cardiology Division, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Polyclinic of Modena, Modena, Italy
| | - Serge Boveda
- Cardiology—Heart Rhythm Management Department, Clinique Pasteur, 45 Avenue de Lombez, 31076 Toulouse, France,Universiteit Ziekenhuis, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | | | - A John Camm
- Cardiology Clinical Academic Group, Molecular and Clinical Sciences Institute, St. George's University of London, London, UK
| | - Barbara Casadei
- RDM, Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Winnie Chua
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Nikolaos Dagres
- Department of Electrophysiology, Heart Center Leipzig at University of Leipzig, Leipzig, Germany
| | - Mirko de Melis
- Medtronic Bakken Research Center, Maastricht, The Netherlands
| | - Lien Desteghe
- Research Group Cardiovascular Diseases, University of Antwerp, Antwerp, Belgium,Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium,Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium,Heart Centre Hasselt, Jessa Hospital, Hasselt, Belgium
| | - Søren Zöga Diederichsen
- Department of Cardiology, Copenhagen University Hospital—Rigshospitalet, Copenhagen, Denmark
| | - David Duncker
- Hannover Heart Rhythm Center, Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Lars Eckardt
- Atrial Fibrillation Network (AFNET), Muenster, Germany,Division of Electrophysiology, Department of Cardiology and Angiology, Münster, Germany
| | | | - Daniel Engler
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,German Centre for Cardiovascular Research (DZHK) partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Larissa Fabritz
- Atrial Fibrillation Network (AFNET), Muenster, Germany,Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,German Centre for Cardiovascular Research (DZHK) partner site Hamburg/Kiel/Lübeck, Hamburg, Germany,Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK,University Center of Cardiovascular Science Hamburg, Hamburg, Germany
| | - Ben Freedman
- Heart Research Institute, The University of Sydney, Sydney, Australia
| | | | - Andreas Goette
- Atrial Fibrillation Network (AFNET), Muenster, Germany,St Vincenz Hospital, Paderborn, Germany
| | - Eduard Guasch
- Arrhythmia Section, Cardiology Department, Hospital Clinic, Universitat de Barcelona, Barcelona, Spain,IDIBAPS, Institut d'Investigació August Pi i Sunyer, Barcelona, Spain,CIBERCV, Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain
| | - Jesper Hastrup Svendsen
- Department of Cardiology, Copenhagen University Hospital—Rigshospitalet, Copenhagen, Denmark,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Karl Georg Haeusler
- Atrial Fibrillation Network (AFNET), Muenster, Germany,Department of Neurology, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Jeff S Healey
- Population Health Research Institute, McMaster University Hamilton, ON, Canada
| | - Hein Heidbuchel
- Research Group Cardiovascular Diseases, University of Antwerp, Antwerp, Belgium,Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
| | - Gerhard Hindricks
- Atrial Fibrillation Network (AFNET), Muenster, Germany,Department of Electrophysiology, Heart Center Leipzig at University of Leipzig, Leipzig, Germany
| | | | | | - Dipak Kotecha
- University of Birmingham & University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | | | | | - Thorsten Lewalter
- Atrial Fibrillation Network (AFNET), Muenster, Germany,Hospital Munich South, Department of Cardiology, Munich, Germany,Department of Cardiology, University of Bonn, Bonn, Germany
| | - Honghuang Lin
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Dominik Linz
- Department of Cardiology, Maastricht University Medical Center and Cardiovascular Research Institute Maastricht, Maastricht, The Netherlands,Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Maja Lisa Løchen
- Department of Community Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| | - Wim Lucassen
- Amsterdam UMC (location AMC), Department General Practice, Amsterdam, The Netherlands
| | | | - Steffen Massberg
- Department of Cardiology, University Hospital, LMU Munich, Munich, Germany,German Centre for Cardiovascular Research (DZHK), partner site: Munich Heart Alliance, Munich, Germany
| | - Jose L Merino
- Arrhythmia & Robotic EP Unit, La Paz University Hospital, IDIPAZ, Madrid, Spain
| | | | - Lluıs Mont
- Arrhythmia Section, Cardiology Department, Hospital Clinic, Universitat de Barcelona, Barcelona, Spain,IDIBAPS, Institut d'Investigació August Pi i Sunyer, Barcelona, Spain,CIBERCV, Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Madrid, Spain
| | | | - Lis Neubeck
- Arrhythmia & Robotic EP Unit, La Paz University Hospital, IDIPAZ, Madrid, Spain
| | - Teemu Niiranen
- Medtronic, Dublin, Ireland,Centre for Cardiovascular Health Edinburgh Napier University, Edinburgh, UK
| | - Michael Oeff
- Atrial Fibrillation Network (AFNET), Muenster, Germany
| | - Jonas Oldgren
- University of Turku and Turku University Hospital, Turku, Finland
| | | | - George Psaroudakis
- Uppsala Clinical Research Center and Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Helmut Pürerfellner
- School of Medicine, Belgrade University, Cardiology Clinic, University Clinical Centre of Serbia, Belgrade, Serbia
| | - Ursula Ravens
- Atrial Fibrillation Network (AFNET), Muenster, Germany,Bayer AG, Leverkusen, Germany
| | - Michiel Rienstra
- Ordensklinikum Linz, Elisabethinen, Cardiological Department, Linz, Austria
| | - Lena Rivard
- Institute of Experimental Cardiovascular Medicine, University Hospital Freiburg, Freiburg, Germany
| | - Daniel Scherr
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ulrich Schotten
- Atrial Fibrillation Network (AFNET), Muenster, Germany,Montreal Heart Institute, University of Montreal, Montreal, Canada
| | - Dipen Shah
- Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Moritz F Sinner
- Atrial Fibrillation Network (AFNET), Muenster, Germany,Amsterdam UMC (location AMC), Department General Practice, Amsterdam, The Netherlands,Royal Brompton Hospital, London, UK
| | | | - Gerhard Steinbeck
- Atrial Fibrillation Network (AFNET), Muenster, Germany,MUMC+, Maastricht, The Netherlands
| | - Daniel Steven
- Atrial Fibrillation Network (AFNET), Muenster, Germany,University Hospital of Geneva, Cardiac Electrophysiology Unit, Geneva, Switzerland
| | - Emma Svennberg
- Center for Cardiology at Clinic Starnberg, Starnberg, Germany
| | - Dierk Thomas
- Atrial Fibrillation Network (AFNET), Muenster, Germany,University Hospital Cologne, Heart Center, Department of Electrophysiology, Cologne, Germany,Karolinska Institutet, Department of Medicine Huddinge, Karolinska University Hospital, Stockholm, Sweden,Department of Cardiology, Medical University Hospital, Heidelberg, Germany
| | - Mellanie True Hills
- HCR (Heidelberg Center for Heart Rhythm Disorders), Medical University Hospital Heidelberg, Heidelberg, Germany
| | - Isabelle C van Gelder
- DZHK (German Center for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Burcu Vardar
- Uppsala Clinical Research Center and Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Elena Palà
- StopAfib.org, American Foundation for Women’s Health, Decatur, TX, USA
| | - Reza Wakili
- Atrial Fibrillation Network (AFNET), Muenster, Germany,Department of Cardiology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Karl Wegscheider
- Atrial Fibrillation Network (AFNET), Muenster, Germany,German Centre for Cardiovascular Research (DZHK) partner site Hamburg/Kiel/Lübeck, Hamburg, Germany,Neurovascular Research Laboratory, Vall d’Hebron Institute of Research (VHIR), Autonomous University of Barcelona, Barcelona, Spain
| | - Mattias Wieloch
- Department of Cardiology and Vascular Medicine, Westgerman Heart and Vascular Center, University of Duisburg-Essen, Essen, Germany,Institute of Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Germany
| | - Stephan Willems
- Atrial Fibrillation Network (AFNET), Muenster, Germany,German Centre for Cardiovascular Research (DZHK) partner site Hamburg/Kiel/Lübeck, Hamburg, Germany,Department of Coagulation Disorders, Skane University Hospital, Lund University, Malmö, Sweden
| | | | | | - Matthias Daniel Zink
- Asklepios Hospital St Georg, Department of Cardiology and Internal Intensive Care Medicine, Faculty of Medicine, Semmelweis University Campus Hamburg, Hamburg, Germany
| | - Paulus Kirchhof
- Corresponding author. Tel: +49 40 7410 52438; Fax: +49 40 7410 55862. E-mail address:
| |
Collapse
|
13
|
Jagirdar G, Elsner M, Scharf C, Simm S, Borucki K, Peter D, Lalk M, Methling K, Linnebacher M, Krohn M, Wolke C, Lendeckel U. Re-Expression of Tafazzin Isoforms in TAZ-Deficient C6 Glioma Cells Restores Cardiolipin Composition but Not Proliferation Rate and Alterations in Gene Expression. Front Genet 2022; 13:931017. [PMID: 35957687 PMCID: PMC9358009 DOI: 10.3389/fgene.2022.931017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/13/2022] [Indexed: 11/18/2022] Open
Abstract
Tafazzin—an acyltransferase—is involved in cardiolipin (CL) remodeling. CL is associated with mitochondrial function, structure and more recently with cell proliferation. Various tafazzin isoforms exist in humans. The role of these isoforms in cardiolipin remodeling is unknown. Aim of this study was to investigate if specific isoforms like Δ5 can restore the wild type phenotype with respect to CL composition, cellular proliferation and gene expression profile. In addition, we aimed to determine the molecular mechanism by which tafazzin can modulate gene expression by applying promoter analysis and (Ingenuity Pathway Analyis) IPA to genes regulated by TAZ-deficiency. Expression of Δ5 and rat full length TAZ in C6-TAZ- cells could fully restore CL composition and—as proven for Δ5—this is naturally associated with restoration of mitochondrial respiration. A similar restoration of CL-composition could not be observed after re-expression of an enzymatically dead full-length rat TAZ (H69L; TAZMut). Re-expression of only rat full length TAZ could restore proliferation rate. Surprisingly, the Δ5 variant failed to restore wild-type proliferation. Further, as expected, re-expression of the TAZMut variant completely failed to reverse the gene expression changes, whereas re-expression of the TAZ-FL variant largely did so and the Δ5 variant to somewhat less extent. Very likely TAZ-deficiency provokes substantial long-lasting changes in cellular lipid metabolism which contribute to changes in proliferation and gene expression, and are not or only very slowly reversible.
Collapse
Affiliation(s)
- Gayatri Jagirdar
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, University of Greifswald, Greifswald, Germany
| | - Matthias Elsner
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Christian Scharf
- Department of Otorhinolaryngology, Head, and Neck Surgery, University Medicine Greifswald, Greifswald, Germany
| | - Stefan Simm
- Institute of Bioinformatics, University Medicine Greifswald, Greifswald, Germany
| | - Katrin Borucki
- Institute of Clinical Chemistry, Department of Pathobiochemistry, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Daniela Peter
- Institute of Clinical Chemistry, Department of Pathobiochemistry, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Michael Lalk
- Institute of Biochemistry, University of Greifswald, Greifswald, Germany
| | - Karen Methling
- Institute of Biochemistry, University of Greifswald, Greifswald, Germany
| | - Michael Linnebacher
- Department of General Surgery, Molecular Oncology, and Immunotherapy, Rostock University Medical Center, Rostock, Germany
| | - Mathias Krohn
- Department of General Surgery, Molecular Oncology, and Immunotherapy, Rostock University Medical Center, Rostock, Germany
| | - Carmen Wolke
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, University of Greifswald, Greifswald, Germany
| | - Uwe Lendeckel
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, University of Greifswald, Greifswald, Germany
- *Correspondence: Uwe Lendeckel,
| |
Collapse
|
14
|
Banerjee S, Prabhu Basrur N, Rai PS. Omics technologies in personalized combination therapy for cardiovascular diseases: challenges and opportunities. Per Med 2021; 18:595-611. [PMID: 34689602 DOI: 10.2217/pme-2021-0087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The primary purpose of 'omics' technologies is to understand the intricacy of genomics, proteomics, metabolomics and other molecular mechanisms to reveal the complex traits of human diseases. The significant use of omics technologies and their applications in medicine gear up the study of the pathogenesis of several disorders. The detection of biomarkers in the early onset of diseases is challenging; still, omics can discover novel molecular mechanisms and biomarkers. In this review, the different types of omics and their technologies are explicated and aimed to provide their emerging applications in cardiovascular precision medicine. These technologies significantly impact optimizing medical treatment for individuals to reach a higher level in precision medicine.
Collapse
Affiliation(s)
- Saradindu Banerjee
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Navya Prabhu Basrur
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Padmalatha S Rai
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| |
Collapse
|
15
|
Slipczuk L, Castagna F, Schonberger A, Novogrodsky E, Dey D, Jorde UP, Levsky JM, Di Biase L, Garcia MJ. Incidence of new-onset atrial fibrillation in COVID-19 is associated with increased epicardial adipose tissue. J Interv Card Electrophysiol 2021; 64:383-391. [PMID: 34231098 PMCID: PMC8260236 DOI: 10.1007/s10840-021-01029-4] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 06/24/2021] [Indexed: 12/15/2022]
Abstract
PURPOSE Coronary artery calcium (CAC) and epicardial adipose tissue (EAT) can predict AF in the general population. We aimed to determine if CAC and EAT measured by computed tomographic (CT) scanning can predict new-onset AF in patients admitted with COVID-19 disease. METHODS We performed a retrospective, post hoc analysis of all patients admitted to Montefiore Medical Center with a confirmed COVID-19 diagnosis from March 1st to June 23rd, 2020, who had a non-contrast CT of the chest within 5 years prior to admission. We determined ordinal CAC scores and quantified the EAT volume and examined their relationship with inpatient mortality. RESULTS A total of 379 patients were analyzed. There were 16 events of new-onset AF (4.22%). Patients who developed AF during the index admission were more likely to be male (75 vs 47%, p < 0.001) and had higher EAT (129.5 [76.3-197.3] vs 91.0 [60.0-129.0] ml, p = 0.049). There were no differences on age (68 [56-71] vs 68 [58-76] years; p = 0.712), BMI (28.5 [25.3-30.8] vs 26.9 [23.1-31.8] kg/m2; p = 0.283), ordinal CAC score (3 [1-6] vs 2 [0-4]; p = 0.482), or prevalence of diabetes (56.3 vs 60.1%; p = 0.761), hypertension (75.0 vs 87.3%, p = 0.153), or coronary artery disease (50.0 vs 39.4%, p = 0.396). Patients with new-onset AF had worse clinical outcomes (death/intubation/vasopressors) (87.5 vs 44.1%; p = 0.001). CONCLUSION Increased EAT measured by non-contrast chest CT identifies patients hospitalized with COVID-19 at higher risk of developing new-onset AF. Patients with new-onset AF have worse clinical outcomes.
Collapse
Affiliation(s)
- Leandro Slipczuk
- Cardiology Division, Montefiore Medical Center, 111 E 210th, Bronx, NY, 10467, USA. .,Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Francesco Castagna
- Cardiology Division, Montefiore Medical Center, 111 E 210th, Bronx, NY, 10467, USA
| | | | | | - Damini Dey
- Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ulrich P Jorde
- Cardiology Division, Montefiore Medical Center, 111 E 210th, Bronx, NY, 10467, USA.,Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jeffrey M Levsky
- Albert Einstein College of Medicine, Bronx, NY, USA.,Radiology Division, Montefiore Medical Center, Bronx, NY, USA
| | - Luigi Di Biase
- Cardiology Division, Montefiore Medical Center, 111 E 210th, Bronx, NY, 10467, USA.,Albert Einstein College of Medicine, Bronx, NY, USA
| | - Mario J Garcia
- Cardiology Division, Montefiore Medical Center, 111 E 210th, Bronx, NY, 10467, USA.,Albert Einstein College of Medicine, Bronx, NY, USA.,Radiology Division, Montefiore Medical Center, Bronx, NY, USA
| |
Collapse
|
16
|
Chen YC, Voskoboinik A, Gerche AL, Marwick TH, McMullen JR. Prevention of Pathological Atrial Remodeling and Atrial Fibrillation: JACC State-of-the-Art Review. J Am Coll Cardiol 2021; 77:2846-2864. [PMID: 34082914 DOI: 10.1016/j.jacc.2021.04.012] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 04/07/2021] [Indexed: 12/29/2022]
Abstract
Atrial enlargement in response to pathological stimuli (e.g., hypertension, mitral valve disease) and physiological stimuli (exercise, pregnancy) can be comparable in magnitude, but the diseased enlarged atria is associated with complications such as atrial fibrillation (AF), whereas physiological atrial enlargement is not. Pathological atrial enlargement and AF is also observed in a small percentage of athletes undergoing extreme/intense endurance sport and pregnant women with preeclampsia. Differences between physiological and pathological atrial enlargement and underlying mechanisms are poorly understood. This review describes human and animal studies characterizing atrial enlargement under physiological and pathological conditions and highlights key knowledge gaps and clinical challenges, including: 1) the limited ability of atria to reverse remodel; and 2) distinguishing physiological and pathological enlargement via imaging/biomarkers. Finally, this review discusses how targeting distinct molecular mechanisms underlying physiological and pathological atrial enlargement could provide new therapeutic and diagnostic strategies for preventing or reversing atrial enlargement and AF.
Collapse
Affiliation(s)
- Yi Ching Chen
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Aleksandr Voskoboinik
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Heart Center, Alfred Hospital, Melbourne, Victoria, Australia; Department of Cardiology, Western Health, Melbourne, Victoria, Australia; Monash University, Melbourne, Victoria, Australia
| | - Andre La Gerche
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Department of Cardiometabolic Health, The University of Melbourne, Melbourne, Victoria, Australia; National Centre for Sports Cardiology, St. Vincent's Hospital Melbourne, Fitzroy, Victoria, Australia
| | - Thomas H Marwick
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Heart Center, Alfred Hospital, Melbourne, Victoria, Australia; Department of Cardiology, Western Health, Melbourne, Victoria, Australia; Monash University, Melbourne, Victoria, Australia; Department of Cardiometabolic Health, The University of Melbourne, Melbourne, Victoria, Australia.
| | - Julie R McMullen
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Monash University, Melbourne, Victoria, Australia; Department of Cardiometabolic Health, The University of Melbourne, Melbourne, Victoria, Australia; Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia.
| |
Collapse
|
17
|
Abstract
The Wnt signaling pathway regulates physiological processes such as cell proliferation and differentiation, cell fate decisions, and stem cell maintenance and, thus, plays essential roles in embryonic development, but also in adult tissue homeostasis and repair. The Wnt signaling pathway has been associated with heart development and repair and has been shown to be crucially involved in proliferation and differentiation of progenitor cells into cardiomyocytes. The investigation of the role of the Wnt signaling pathway and the regulation of its expression/activity in atrial fibrillation has only just begun. The present minireview (I) provides original data regarding the expression of Wnt signaling components in atrial tissue of patients with atrial fibrillation or sinus rhythm and (II) summarizes the current state of knowledge of the regulation of Wnt signaling components' expression/activity and the contribution of the various levels of the Wnt signal transduction pathway to the processes of the development, maintenance, and progression of atrial fibrillation.
Collapse
Affiliation(s)
- Carmen Wolke
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Greifswald D-17475, Germany
| | - Elmer Antileo
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Greifswald D-17475, Germany
| | - Uwe Lendeckel
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Greifswald D-17475, Germany
| |
Collapse
|
18
|
Nalliah CJ, Bell JR, Raaijmakers AJA, Waddell HM, Wells SP, Bernasochi GB, Montgomery MK, Binny S, Watts T, Joshi SB, Lui E, Sim CB, Larobina M, O'Keefe M, Goldblatt J, Royse A, Lee G, Porrello ER, Watt MJ, Kistler PM, Sanders P, Delbridge LMD, Kalman JM. Epicardial Adipose Tissue Accumulation Confers Atrial Conduction Abnormality. J Am Coll Cardiol 2021; 76:1197-1211. [PMID: 32883413 DOI: 10.1016/j.jacc.2020.07.017] [Citation(s) in RCA: 119] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/02/2020] [Accepted: 07/09/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND Clinical studies have reported that epicardial adipose tissue (EpAT) accumulation associates with the progression of atrial fibrillation (AF) pathology and adversely affects AF management. The role of local cardiac EpAT deposition in disease progression is unclear, and the electrophysiological, cellular, and molecular mechanisms involved remain poorly defined. OBJECTIVES The purpose of this study was to identify the underlying mechanisms by which EpAT influences the atrial substrate for AF. METHODS Patients without AF undergoing coronary artery bypass surgery were recruited. Computed tomography and high-density epicardial electrophysiological mapping of the anterior right atrium were utilized to quantify EpAT volumes and to assess association with the electrophysiological substrate in situ. Excised right atrial appendages were analyzed histologically to characterize EpAT infiltration, fibrosis, and gap junction localization. Co-culture experiments were used to evaluate the paracrine effects of EpAT on cardiomyocyte electrophysiology. Proteomic analyses were applied to identify molecular mediators of cellular electrophysiological disturbance. RESULTS Higher local EpAT volume clinically correlated with slowed conduction, greater electrogram fractionation, increased fibrosis, and lateralization of cardiomyocyte connexin-40. In addition, atrial conduction heterogeneity was increased with more extensive myocardial EpAT infiltration. Cardiomyocyte culture studies using multielectrode arrays showed that cardiac adipose tissue-secreted factors slowed conduction velocity and contained proteins with capacity to disrupt intermyocyte electromechanical integrity. CONCLUSIONS These findings indicate that atrial pathophysiology is critically dependent on local EpAT accumulation and infiltration. In addition to myocardial architecture disruption, this effect can be attributed to an EpAT-cardiomyocyte paracrine axis. The focal adhesion group proteins are identified as new disease candidates potentially contributing to arrhythmogenic atrial substrate.
Collapse
Affiliation(s)
- Chrishan J Nalliah
- Department of Cardiology, Royal Melbourne Hospital, Melbourne, Victoria, Australia; Department of Medicine and Radiology, University of Melbourne, Melbourne, Victoria, Australia
| | - James R Bell
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia; Department of Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | | | - Helen M Waddell
- Department of Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Simon P Wells
- Department of Physiology, University of Melbourne, Melbourne, Victoria, Australia; Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Gabriel B Bernasochi
- Department of Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | | | - Simon Binny
- Department of Cardiology, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Troy Watts
- Department of Cardiology, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Subodh B Joshi
- Department of Cardiology, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Elaine Lui
- Department of Radiology, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Choon Boon Sim
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Marco Larobina
- Department of Cardiothoracic Surgery, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Michael O'Keefe
- Department of Cardiothoracic Surgery, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - John Goldblatt
- Department of Cardiothoracic Surgery, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Alistair Royse
- Department of Cardiothoracic Surgery, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Geoffrey Lee
- Department of Cardiology, Royal Melbourne Hospital, Melbourne, Victoria, Australia; Department of Medicine and Radiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Enzo R Porrello
- Department of Physiology, University of Melbourne, Melbourne, Victoria, Australia; Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Matthew J Watt
- Department of Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Peter M Kistler
- Department of Cardiology, The Alfred Hospital, Melbourne, Victoria, Australia
| | - Prashanthan Sanders
- Australia Centre for Heart Rhythm Disorders (CHRD), South Australian Health and Medical Research Institute (SAHMRI), University of Adelaide and Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Lea M D Delbridge
- Department of Physiology, University of Melbourne, Melbourne, Victoria, Australia.
| | - Jonathan M Kalman
- Department of Cardiology, Royal Melbourne Hospital, Melbourne, Victoria, Australia; Department of Medicine and Radiology, University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
19
|
Insight into atrial fibrillation through analysis of the coding transcriptome in humans. Biophys Rev 2020; 12:817-826. [PMID: 32666467 DOI: 10.1007/s12551-020-00735-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/08/2020] [Indexed: 12/11/2022] Open
Abstract
Atrial fibrillation is the most common sustained cardiac arrhythmia in humans, and its prevalence continues to increase because of the aging of the world population. Much still needs to be learned about the molecular pathways involved in the development and the persistence of the disease. Analysis of the transcriptome of cardiac tissue has provided valuable insight into diverse aspects of atrial remodeling, in particular concerning electrical remodeling-related to ion channels-and structural remodeling identified by dysregulation of processes linked to inflammation, fibrosis, oxidative stress, and thrombogenesis. The huge amount of data produced by these studies now represents a valuable source for the identification of novel potential therapeutic targets. In addition, the shift from cardiac tissue to peripheral blood as a substrate for transcriptome analysis revealed this strategy as a promising tool for improved diagnosis and therefore better patient care.
Collapse
|
20
|
Zhao Z, Li R, Wang X, Li J, Yuan M, Liu E, Liu T, Li G. Attenuation of atrial remodeling by aliskiren via affecting oxidative stress, inflammation and PI3K/Akt signaling pathway. Cardiovasc Drugs Ther 2020; 35:587-598. [PMID: 32462265 DOI: 10.1007/s10557-020-07002-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
INTRODUCTION Atrial fibrillation (AF) is the most common type of arrhythmia. Atrial remodeling is a major factor to the AF substrate. The purpose of the study is to explore whether aliskiren (ALS) has a cardioprotective effect and its potential molecular mechanisms on atrial remodeling. METHODS In acute experiments, dogs were randomly assigned to Sham, Paced and Paced+aliskiren (10 mg kg-1) (Paced+ALS) groups, with 7 dogs in each group. Rapid atrial pacing (RAP) was maintained at 600 bpm for 2 h for paced and Paced+ALS groups and atrial effective refractory periods (AERPs), inducibility of AF (AFi) and average duration time (ADT) were measured. In chronic experiments, there were 5 groups: Sham, Sham+ALS, Paced, Paced+ALS and Paced+ALS+PI3K antagonist wortmannin (WM) (70 μg kg-1 day-1). RAP at 500 beats/min was maintained for 2 weeks. Inflammation and oxidative stress indicators were measured by ELISA assay, echocardiogram and pathology were used to assess atrial structural remodeling, phosphatidylinositol 3-hydroxy kinase/protein kinase B (PI3K/Akt) signaling pathways were studied by RT-PCR and western blotting to evaluate whether the cardioprotective effect of ALS works through PI3K/Akt signaling pathway. RESULTS The electrophysiological changes were observed after 2-h pacing. The AERP shortened with increased AFi and ADT, which was attenuated by ALS (P < 0.05). After pacing for 2 weeks, oxidative stress and inflammation markers in the Paced group were significantly higher than those in the Sham group (P < 0.01) and were reduced by ALS treatment (P < 0.01). The reduced level of antioxidant enzymes caused by RAP was also found to be elevated in ALS-treated group (P < 0.01). The results of pathology and echocardiography showed that RAP can cause atrial enlargement, fibrosis (P < 0.01), and were attenuated in ALS treatment group. The PI3K/Akt signaling pathway were downregulated induced by RAP. ALS could upregulate the PI3K/Akt pathway expression (P < 0.05). Furthermore, the cardioprotective effects in structural remodeling of ALS were suppressed by WM. CONCLUSIONS ALS may offer cardioprotection in RAP-induced atrial remodeling, which may partly be ascribed to its anti-inflammatory and anti-oxidative stress action and the regulation of PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Zhiqiang Zhao
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Ruiling Li
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Xinghua Wang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Jian Li
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Meng Yuan
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Enzhao Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Tong Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Guangping Li
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, 300211, China.
| |
Collapse
|
21
|
Li B, Po SS, Zhang B, Bai F, Li J, Qin F, Liu N, Sun C, Xiao Y, Tu T, Zhou S, Liu Q. Metformin regulates adiponectin signalling in epicardial adipose tissue and reduces atrial fibrillation vulnerability. J Cell Mol Med 2020; 24:7751-7766. [PMID: 32441464 PMCID: PMC7348162 DOI: 10.1111/jcmm.15407] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 03/31/2020] [Accepted: 04/22/2020] [Indexed: 12/26/2022] Open
Abstract
Epicardial adipose tissue (EAT) remodelling is closely related to the pathogenesis of atrial fibrillation (AF). We investigated whether metformin (MET) prevents AF‐dependent EAT remodelling and AF vulnerability in dogs. A canine AF model was developed by 6‐week rapid atrial pacing (RAP), and electrophysiological parameters were measured. Effective refractory periods (ERP) were decreased in the left and right atrial appendages as well as in the left atrium (LA) and right atrium (RA). MET attenuated the RAP‐induced increase in ERP dispersion, cumulative window of vulnerability, AF inducibility and AF duration. RAP increased reactive oxygen species (ROS) production and nuclear factor kappa‐B (NF‐κB) phosphorylation; up‐regulated interleukin‐6 (IL‐6), tumour necrosis factor‐α (TNF‐α) and transforming growth factor‐β1 (TGF‐β1) levels in LA and EAT; decreased peroxisome proliferator‐activated receptor gamma (PPARγ) and adiponectin (APN) expression in EAT and was accompanied by atrial fibrosis and adipose infiltration. MET reversed these alterations. In vitro, lipopolysaccharide (LPS) exposure increased IL‐6, TNF‐α and TGF‐β1 expression and decreased PPARγ/APN expression in 3T3‐L1 adipocytes, which were all reversed after MET administration. Indirect coculture of HL‐1 cells with LPS‐stimulated 3T3‐L1 conditioned medium (CM) significantly increased IL‐6, TNF‐α and TGF‐β1 expression and decreased SERCA2a and p‐PLN expression, while LPS + MET CM and APN treatment alleviated the inflammatory response and sarcoplasmic reticulum Ca2+ handling dysfunction. MET attenuated the RAP‐induced increase in AF vulnerability, remodelling of atria and EAT adipokines production profiles. APN may play a key role in the prevention of AF‐dependent EAT remodelling and AF vulnerability by MET.
Collapse
Affiliation(s)
- Biao Li
- Department of Cardiology/Cardiac Catheterization Lab, Second Xiangya Hospital, Central South University, Changsha City, Hunan Province, China
| | - Sunny S Po
- Heart Rhythm Institute and Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Baojian Zhang
- Department of Cardiology/Cardiac Catheterization Lab, Second Xiangya Hospital, Central South University, Changsha City, Hunan Province, China.,Department of Cardiology, the Affiliated Chinese Medicine Hospital of Xinjiang Medical University, Urumqi City, Xinjiang Province, China
| | - Fan Bai
- Department of Cardiology/Cardiac Catheterization Lab, Second Xiangya Hospital, Central South University, Changsha City, Hunan Province, China
| | - Jiayi Li
- Department of Cardiology/Cardiac Catheterization Lab, Second Xiangya Hospital, Central South University, Changsha City, Hunan Province, China
| | - Fen Qin
- Department of Cardiology/Cardiac Catheterization Lab, Second Xiangya Hospital, Central South University, Changsha City, Hunan Province, China
| | - Na Liu
- Department of Cardiology/Cardiac Catheterization Lab, Second Xiangya Hospital, Central South University, Changsha City, Hunan Province, China
| | - Chao Sun
- Department of Cardiology/Cardiac Catheterization Lab, Second Xiangya Hospital, Central South University, Changsha City, Hunan Province, China
| | - Yichao Xiao
- Department of Cardiology/Cardiac Catheterization Lab, Second Xiangya Hospital, Central South University, Changsha City, Hunan Province, China
| | - Tao Tu
- Department of Cardiology/Cardiac Catheterization Lab, Second Xiangya Hospital, Central South University, Changsha City, Hunan Province, China
| | - Shenghua Zhou
- Department of Cardiology/Cardiac Catheterization Lab, Second Xiangya Hospital, Central South University, Changsha City, Hunan Province, China
| | - Qiming Liu
- Department of Cardiology/Cardiac Catheterization Lab, Second Xiangya Hospital, Central South University, Changsha City, Hunan Province, China
| |
Collapse
|
22
|
Sapelnikov OV, Kulikov AA, Cherkashin DI, Grishin IR, Nikolaeva OA, Akchurin RS. Atrial Fibrillation: Development Mechanisms, Approaches and Prospects of Therapy. RATIONAL PHARMACOTHERAPY IN CARDIOLOGY 2020. [DOI: 10.20996/1819-6446-2020-02-15] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Atrial fibrillation (AF) is the most common arrhythmia. It is diagnosed in more than 33 million people worldwide and is the leading cause of hospitalization for arrhythmias. AF is characterized by fast and irregular atrial activation without discrete P-waves at a surface electrocardiography. AF pathophysiological mechanisms are very complex and include the dynamic interaction between arrhythmia substrate and triggers. Consequently, the clinical search for effective therapeutic targets should include the entire process of the onset and progression of the disease: from the first paroxysms to the development of a stable permanent form of AF.
Collapse
|
23
|
Zhou M, Wang H, Chen J, Zhao L. Epicardial adipose tissue and atrial fibrillation: Possible mechanisms, potential therapies, and future directions. PACING AND CLINICAL ELECTROPHYSIOLOGY: PACE 2019; 43:133-145. [DOI: 10.1111/pace.13825] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 10/03/2019] [Accepted: 10/22/2019] [Indexed: 12/21/2022]
Affiliation(s)
- Mengmeng Zhou
- Department of Cardiology, Shanghai Chest HospitalShanghai Jiao Tong University Shanghai China
| | - Hao Wang
- Department of Cardiology, Shanghai Chest HospitalShanghai Jiao Tong University Shanghai China
| | - Jindong Chen
- Department of Cardiology, Shanghai Chest HospitalShanghai Jiao Tong University Shanghai China
| | - Liang Zhao
- Department of Cardiology, Shanghai Chest HospitalShanghai Jiao Tong University Shanghai China
| |
Collapse
|
24
|
Bai F, Liu Y, Tu T, Li B, Xiao Y, Ma Y, Qin F, Xie J, Zhou S, Liu Q. Metformin regulates lipid metabolism in a canine model of atrial fibrillation through AMPK/PPAR-α/VLCAD pathway. Lipids Health Dis 2019; 18:109. [PMID: 31077199 PMCID: PMC6511207 DOI: 10.1186/s12944-019-1059-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 04/29/2019] [Indexed: 12/20/2022] Open
Abstract
Background Atrial lipid metabolic remodeling is critical for the process of atrial fibrillation (AF). Abnormal Fatty acid (FA) metabolism in cardiomyocytes is involved in the pathogenesis of AF. MET (Metformin), an AMPK (AMP-activated protein kinase) activator, has been found to be associated with a decreased risk of AF in patients with type 2 diabetes. However, the specific mechanism remains unknown. Methods Fifteen mongrel dogs were divided into three groups: SR, ARP (pacing with 800 beats/min for 6 h), ARP plus MET (treated with MET (100 mg/kg/day) for two weeks before pacing). We assessed metabolic factors, speed limiting enzymes circulating biochemical metabolites (substrates and products), atrial electrophysiology and accumulation of lipid droplets. Results The expression of AMPK increased in the ARP group and significantly increased in the MET+ARP group comparing to the SR group. In the ARP group, the expressions of PPARα、PGC-1α and VLCAD were down-regulated, while the concentration of free fatty acid and triglyceride and the lipid deposition in LAA (left atrial appendage) increased. Moreover, AERP and AERPd have also been found abnormally in this process. Pretreatment with MET before receiving ARP reversed the alterations aforementioned. Conclusions The FA metabolism in LAA is altered in the ARP group, mainly characterized by the abnormal expression of the rate-limiting enzyme. Metformin reduces lipid accumulation and promotes β-oxidation of FA in AF models partially through AMPK/PPAR-α/VLCAD pathway. Our study indicates that MET may inhibit the FA lipid metabolic remodeling in AF.
Collapse
Affiliation(s)
- Fan Bai
- Department of Cardiology/Cardiac Catheterization Lab, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China
| | - Yaozhong Liu
- Department of Cardiology/Cardiac Catheterization Lab, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China
| | - Tao Tu
- Department of Cardiology/Cardiac Catheterization Lab, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China
| | - Biao Li
- Department of Cardiology/Cardiac Catheterization Lab, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China
| | - Yichao Xiao
- Department of Cardiology/Cardiac Catheterization Lab, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China
| | - Yingxu Ma
- Department of Cardiology/Cardiac Catheterization Lab, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China
| | - Fen Qin
- Department of Cardiology/Cardiac Catheterization Lab, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China
| | - Jing Xie
- Department of Cardiology/Cardiac Catheterization Lab, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China
| | - Shenghua Zhou
- Department of Cardiology/Cardiac Catheterization Lab, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China
| | - Qiming Liu
- Department of Cardiology/Cardiac Catheterization Lab, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China.
| |
Collapse
|
25
|
Vroomen M, Olsthoorn JR, Maesen B, L’Espoir V, La Meir M, Das M, Maessen JG, Crijns HJGM, Verheule S, Pison L. Quantification of epicardial adipose tissue in patients undergoing hybrid ablation for atrial fibrillation. Eur J Cardiothorac Surg 2019; 56:79-86. [DOI: 10.1093/ejcts/ezy472] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 12/02/2018] [Accepted: 12/13/2018] [Indexed: 01/10/2023] Open
Abstract
Abstract
OBJECTIVES
Epicardial adipose tissue volume (EAT-V) has been linked to atrial fibrillation (AF) recurrences after catheter ablation. We retrospectively studied the association between atrial EAT-V and outcome after hybrid AF ablation (epicardial surgical and endocardial catheter ablation).
METHODS
On preoperative cardiac computed tomography angiography scans, the left atrium and right atrium were manually delineated using the open source ImageJ. With custom-made automated software, the number of pixels in the regions of interest on each slice was calculated. On the basis of the Hounsfield units, pixel size and slice thickness, EAT-V was computed and normalized in relation to the body surface area (BSA) and the myocardial tissue volume.
RESULTS
Eighty-five patients were included. Left atrial and right atrial EAT-V normalized to BSA were not significantly different between paroxysmal and persistent AF [0.84 (0.51–1.50) vs 0.81 (0.57–1.18), 1.74 (1.02–2.56) vs 1.55 (1.26–2.18), all P = 0.9], neither between the acute conduction block and no acute conduction block in the epicardial box lesion [0.92 (0.55–1.39) vs 0.72 (0.55–1.24), P = 0.5, right atrium not applicable], nor between the sinus rhythm and arrhythmia recurrence after 12 months [0.88 (0.55–1.48) vs 0.63 (0.47–1.10), 1.61 (1.11–2.50) vs 1.55 (1.20–2.20), all P > 0.1]. Left atrial EAT-V normalized to myocardial tissue volume was not different between the groups.
CONCLUSIONS
This study could neither confirm that EAT-V was predictive of recurrence of supraventricular arrhythmias in patients undergoing a hybrid AF ablation, nor that EAT-V was different between patients with paroxysmal AF and persistent and long-standing persistent AF. This suggests that EAT-V might not affect the outcome in surgical ablation procedures and therefore should not influence preoperative or intraoperative decision-making.
Collapse
Affiliation(s)
- Mindy Vroomen
- Department of Cardiology, Maastricht University Medical Center, Maastricht, Netherlands
- Cardiovascular Research Institute Maastricht, Maastricht, Netherlands
| | - Jules R Olsthoorn
- Department of Cardiothoracic Surgery, Maastricht University Medical Center, Maastricht, Netherlands
| | - Bart Maesen
- Cardiovascular Research Institute Maastricht, Maastricht, Netherlands
- Department of Cardiothoracic Surgery, Maastricht University Medical Center, Maastricht, Netherlands
| | - Vladimir L’Espoir
- Department of Cardiothoracic Surgery, Maastricht University Medical Center, Maastricht, Netherlands
| | - Mark La Meir
- Department of Cardiothoracic Surgery, Maastricht University Medical Center, Maastricht, Netherlands
| | - Marco Das
- Cardiovascular Research Institute Maastricht, Maastricht, Netherlands
- Department of Radiology, Maastricht University Medical Center, Maastricht, Netherlands
| | - Jos G Maessen
- Cardiovascular Research Institute Maastricht, Maastricht, Netherlands
- Department of Cardiothoracic Surgery, Maastricht University Medical Center, Maastricht, Netherlands
| | - Harry J G M Crijns
- Department of Cardiology, Maastricht University Medical Center, Maastricht, Netherlands
- Cardiovascular Research Institute Maastricht, Maastricht, Netherlands
| | - Sander Verheule
- Cardiovascular Research Institute Maastricht, Maastricht, Netherlands
- Department of Physiology, Maastricht University Medical Center, Maastricht, Netherlands
| | - Laurent Pison
- Department of Cardiology, Maastricht University Medical Center, Maastricht, Netherlands
- Cardiovascular Research Institute Maastricht, Maastricht, Netherlands
| |
Collapse
|
26
|
Pabon MA, Manocha K, Cheung JW, Lo JC. Linking Arrhythmias and Adipocytes: Insights, Mechanisms, and Future Directions. Front Physiol 2018; 9:1752. [PMID: 30568603 PMCID: PMC6290087 DOI: 10.3389/fphys.2018.01752] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 11/20/2018] [Indexed: 01/14/2023] Open
Abstract
Obesity and atrial fibrillation have risen to epidemic levels worldwide and may continue to grow over the next decades. Emerging evidence suggests that obesity promotes atrial and ventricular arrhythmias. This has led to trials employing various strategies with the ultimate goal of decreasing the atrial arrhythmic burden in obese patients. The effectiveness of these interventions remains to be determined. Obesity is defined by the expansion of adipose mass, making adipocytes a prime candidate to mediate the pro-arrhythmogenic effects of obesity. The molecular mechanisms linking obesity and adipocytes to increased arrhythmogenicity in both the atria and ventricles remain poorly understood. In this focused review, we highlight areas of potential molecular interplay between adipocytes and cardiomyocytes. The effects of adipocytes may be direct, local or remote. Direct effect refers to adipocyte or fatty infiltration of the atrial and ventricular myocardium itself, possibly causing increased dispersion of normal myocardial electrical signals and fibrotic substrate of adipocytes that promote reentry or adipocytes serving as a direct source of aberrant signals. Local effects may originate from nearby adipose depots, specifically epicardial adipose tissue (EAT) and pericardial adipose tissue, which may play a role in the secretion of adipokines and chemokines that can incite inflammation given the direct contact and disrupt the conduction system. Adipocytes can also have a remote effect on the myocardium arising from their systemic secretion of adipokines, cytokines and metabolites. These factors may lead to mitochondrial dysfunction, oxidative stress, autophagy, mitophagy, autonomic dysfunction, and cardiomyocyte death to ultimately produce a pro-arrhythmogenic state. By better understanding the molecular mechanisms connecting dysfunctional adipocytes and arrhythmias, novel therapies may be developed to sever the link between obesity and arrhythmias.
Collapse
Affiliation(s)
- Maria A Pabon
- Joan and Sanford I. Weill, Department of Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY, United States
| | - Kevin Manocha
- Division of Cardiology, Department of Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY, United States
| | - Jim W Cheung
- Division of Cardiology, Department of Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY, United States
| | - James C Lo
- Division of Cardiology, Department of Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY, United States.,Metabolic Health Center, Weill Cornell Medicine, New York, NY, United States.,Department of Pharmacology, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
27
|
Association of fibrotic remodeling and cytokines/chemokines content in epicardial adipose tissue with atrial myocardial fibrosis in patients with atrial fibrillation. Heart Rhythm 2018; 15:1717-1727. [DOI: 10.1016/j.hrthm.2018.06.025] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Indexed: 01/06/2023]
|
28
|
Goudis CA, Vasileiadis IE, Liu T. Epicardial adipose tissue and atrial fibrillation: pathophysiological mechanisms, clinical implications, and potential therapies. Curr Med Res Opin 2018; 34:1933-1943. [PMID: 29625530 DOI: 10.1080/03007995.2018.1462786] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Atrial fibrillation (AF) is the most common arrhythmia in clinical practice and is associated with increased cardiovascular morbidity and mortality. Epicardial adipose tissue (EAT) serves as a biologically active organ with important endocrine and inflammatory function. Review An accumulating body of evidence suggests that EAT is associated with the initiation, perpetuation, and recurrence of AF, but the precise role of EAT in AF pathogenesis is not completely elucidated. Pathophysiological mechanisms involve adipocyte infiltration, profibrotic and pro-inflammatory paracrine effects, oxidative stress, neural mechanisms, and genetic factors. CONCLUSIONS Notably, EAT accumulation seems to be associated with stroke and adverse cardiovascular outcomes in AF. Weight loss, specific medications and ablation of ganglionated plexi (GP) seem to be potential therapies in this setting.
Collapse
Affiliation(s)
- Christos A Goudis
- a Department of Cardiology , Serres General Hospital , Serres , Greece
| | | | - Tong Liu
- c Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular disease, Department of Cardiology , Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University , Tianjin , PR China
| |
Collapse
|
29
|
Pandit SV, Anumonwo J, Jalife J. Atrial Fibrillation Susceptibility in Obesity: An Excess Adiposity and Fibrosis Complicity? Circ Res 2018; 118:1468-1471. [PMID: 27174946 DOI: 10.1161/circresaha.116.308686] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Sandeep V Pandit
- Center for Arrhythmia Research, Department of Internal Medicine-Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - Justus Anumonwo
- Center for Arrhythmia Research, Department of Internal Medicine-Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - José Jalife
- Center for Arrhythmia Research, Department of Internal Medicine-Cardiology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
30
|
Calvo D, Filgueiras-Rama D, Jalife J. Mechanisms and Drug Development in Atrial Fibrillation. Pharmacol Rev 2018; 70:505-525. [PMID: 29921647 PMCID: PMC6010660 DOI: 10.1124/pr.117.014183] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Atrial fibrillation is a highly prevalent cardiac arrhythmia and the most important cause of embolic stroke. Although genetic studies have identified an increasing assembly of AF-related genes, the impact of these genetic discoveries is yet to be realized. In addition, despite more than a century of research and speculation, the molecular and cellular mechanisms underlying AF have not been established, and therapy for AF, particularly persistent AF, remains suboptimal. Current antiarrhythmic drugs are associated with a significant rate of adverse events, particularly proarrhythmia, which may explain why many highly symptomatic AF patients are not receiving any rhythm control therapy. This review focuses on recent advances in AF research, including its epidemiology, genetics, and pathophysiological mechanisms. We then discuss the status of antiarrhythmic drug therapy for AF today, reviewing molecular mechanisms, and the possible clinical use of some of the new atrial-selective antifibrillatory agents, as well as drugs that target atrial remodeling, inflammation and fibrosis, which are being tested as upstream therapies to prevent AF perpetuation. Altogether, the objective is to highlight the magnitude and endemic dimension of AF, which requires a significant effort to develop new and effective antiarrhythmic drugs, but also improve AF prevention and treatment of risk factors that are associated with AF complications.
Collapse
Affiliation(s)
- David Calvo
- Department of Cardiology, Arrhythmia Unit, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain (D.C.); Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (D.F.-R., J.J.); Department of Cardiology, Arrhythmia Unit, Hospital Clínico Universitario San Carlos, Madrid, Spain (D.F.-R.); Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain (D.F.-R., J.J.); and Center for Arrhythmia Research, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan (J.J.)
| | - David Filgueiras-Rama
- Department of Cardiology, Arrhythmia Unit, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain (D.C.); Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (D.F.-R., J.J.); Department of Cardiology, Arrhythmia Unit, Hospital Clínico Universitario San Carlos, Madrid, Spain (D.F.-R.); Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain (D.F.-R., J.J.); and Center for Arrhythmia Research, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan (J.J.)
| | - José Jalife
- Department of Cardiology, Arrhythmia Unit, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain (D.C.); Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (D.F.-R., J.J.); Department of Cardiology, Arrhythmia Unit, Hospital Clínico Universitario San Carlos, Madrid, Spain (D.F.-R.); Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain (D.F.-R., J.J.); and Center for Arrhythmia Research, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan (J.J.)
| |
Collapse
|
31
|
Chilukoti RK, Lendeckel J, Darm K, Bukowska A, Goette A, Sühling M, Utpatel K, Peters B, Homuth G, Völker U, Wolke C, Scharf C, Lendeckel U. Integration of "omics" techniques: Dronedarone affects cardiac remodeling in the infarction border zone. Exp Biol Med (Maywood) 2018; 243:895-910. [PMID: 30105952 PMCID: PMC6108048 DOI: 10.1177/1535370218788517] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 06/18/2018] [Indexed: 01/15/2023] Open
Abstract
Dronedarone improves microvascular flow during atrial fibrillation and reduces the infarct size in acute models of myocardial infarction. However, dronedarone might be harmful in patients with recent decompensated heart failure and increases mortality in patients with permanent atrial fibrillation. A pathophysiological explanation for these discrepant data is lacking. This study investigated the effects of dronedarone on gene and protein expression in the infarcted area and border zone in pigs subjected to anterior ischemia/reperfusion myocardial infarction. The ischemia/reperfusion myocardial infarction was induced in 16 pigs. Eight pigs were treated with dronedarone for 28 days after myocardial infarction, the remaining pigs served as control. Microarray-based transcriptome profiling and 2D-DIGE-based proteome analysis were used to assess the effects of dronedarone on left ventricular gene expression in healthy (LV), infarcted (MI), and border zone tissue. Selected targets were validated by RT-qPCR or immunoblot analyses, with special emphasize given to the transcriptome/proteome overlap. Combined "omics" analysis was performed to identify most significant disease and function charts affected by dronedarone and to establish an integrated network. The levels of 879 (BZ) or 7 (MI) transcripts and 51 (LV) or 15 (BZ) proteins were significantly altered by dronedarone, pointing to a substantial efficacy of dronedarone in the border zone. Transcriptome and proteome data indicate that dronedarone influences post-infarction remodeling processes and identify matricellular proteins as major targets of dronedarone in this setting. This finding is fully supported by the disease and function charts as well as by the integrated network established by combined "omics". Dronedarone therapy alters myocardial gene expression after acute myocardial infarction with pronounced effects in the border zone. Dronedarone promotes infarct healing via regulation of periostin and might contribute to the limitation of its expansion as well as cardiac rupture. Thus, there are no experimental hints that dronedarone per se has direct harmful effects after MI in ventricular tissue. Impact statement Dronedarone reduced the infarct size in models of acute myocardial infarction (MI). Here, we show that dronedarone attenuates many of the substantial changes in gene expression that are provoked by acute myocardial infarction (AMI) in pigs. Dronedarone modifies the expression of gene panels related to post-infarction cardiac healing and remodeling processes and, most remarkably, this occurs predominantly in the infarction border-zone and much less so in the vital or infarcted myocardium. Combined "omics" identified matricellular proteins and ECM as major dronedarone-regulated targets and emphasizes their relevance for Disease Charts and Tox Function Charts associated with tissue remodeling and cellular movement. The results demonstrate dronedarone's capability of regulating cardiac repair and remodeling processes specifically in the infarction border zone and identify underlying mechanisms and pathways that might be employed in future therapeutic strategies to improve long-term cardiac tissue function and stability.
Collapse
Affiliation(s)
- Ravi K Chilukoti
- Institute of Medical Biochemistry and Molecular Biology,
University Medicine Greifswald, Greifswald D-17475, Germany
| | - Josefine Lendeckel
- Institute of Medical Biochemistry and Molecular Biology,
University Medicine Greifswald, Greifswald D-17475, Germany
| | - Katrin Darm
- Department of Otorhinolaryngology, Head and Neck Surgery,
University Medicine Greifswald, Greifswald D-17475, Germany
| | - Alicja Bukowska
- Working Group: Molecular Electrophysiology, Otto-von-Guericke
University, University Hospital Magdeburg, Magdeburg D-39120, Germany
| | - Andreas Goette
- Working Group: Molecular Electrophysiology, Otto-von-Guericke
University, University Hospital Magdeburg, Magdeburg D-39120, Germany
- Department of Cardiology and Intensive Care Medicine, St.
Vincenz-Hospital, Paderborn D-33098, Germany
| | - Marc Sühling
- Institute of Medical Biochemistry and Molecular Biology,
University Medicine Greifswald, Greifswald D-17475, Germany
| | - Kirsten Utpatel
- Department of Pathology, University Medicine Greifswald,
Greifswald D-17475, Germany
| | - Barbara Peters
- Institute of Physiology, University Medicine Greifswald,
Karlsburg D-17495, Germany
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics,
University Medicine Greifswald, D-17475 Greifswald, Germany
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics,
University Medicine Greifswald, D-17475 Greifswald, Germany
| | - Carmen Wolke
- Institute of Medical Biochemistry and Molecular Biology,
University Medicine Greifswald, Greifswald D-17475, Germany
| | - Christian Scharf
- Department of Otorhinolaryngology, Head and Neck Surgery,
University Medicine Greifswald, Greifswald D-17475, Germany
| | - Uwe Lendeckel
- Institute of Medical Biochemistry and Molecular Biology,
University Medicine Greifswald, Greifswald D-17475, Germany
| |
Collapse
|
32
|
Wong CX, Ganesan AN, Selvanayagam JB. Epicardial fat and atrial fibrillation: current evidence, potential mechanisms, clinical implications, and future directions. Eur Heart J 2018; 38:1294-1302. [PMID: 26935271 DOI: 10.1093/eurheartj/ehw045] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 01/25/2016] [Indexed: 12/17/2022] Open
Abstract
Obesity is increasingly recognized as a major modifiable determinant of atrial fibrillation (AF). Although body mass index and other clinical measures are useful indications of general adiposity, much recent interest has focused on epicardial fat, a distinct adipose tissue depot that can be readily assessed using non-invasive imaging techniques. A growing body of data from epidemiological and clinical studies has demonstrated that epicardial fat is consistently associated with the presence, severity, and recurrence of AF across a range of clinical settings. Evidence from basic science and translational studies has also suggested that arrhythmogenic mechanisms may involve adipocyte infiltration, pro-fibrotic, and pro-inflammatory paracrine effects, oxidative stress, and other pathways. Despite these advances, however, significant uncertainty exists and many questions remain unanswered. In this article, we review our present understanding of epicardial fat, including its classification and quantification, existing evidence implicating its role in AF, potential mechanisms, implications for clinicians, and future directions for research.
Collapse
Affiliation(s)
- Christopher X Wong
- Clinical Trial Service Unit and Epidemiological Studies Unit, University of Oxford, Oxford, UK
| | - Anand N Ganesan
- Department of Cardiology, Flinders Medical Centre & Flinders University, Flinders Drive, Bedford Park, Adelaide, South Australia 5043, Australia
| | - Joseph B Selvanayagam
- Department of Cardiology, Flinders Medical Centre & Flinders University, Flinders Drive, Bedford Park, Adelaide, South Australia 5043, Australia
| |
Collapse
|
33
|
Anumonwo JMB, Herron T. Fatty Infiltration of the Myocardium and Arrhythmogenesis: Potential Cellular and Molecular Mechanisms. Front Physiol 2018; 9:2. [PMID: 29403390 PMCID: PMC5786512 DOI: 10.3389/fphys.2018.00002] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 01/03/2018] [Indexed: 11/13/2022] Open
Abstract
Anatomical evidence in several species shows highly heterogeneous fat distribution in the atrial and ventricular myocardium. Atrial appendages have fat deposits, and more so on the posterior left atrium. Although such fat distributions are considered normal, fatty infiltration is regarded arrhythmogenic, and various cardiac pathophysiological conditions show excess myocardial fat deposits, especially in the epicardium. Hypotheses have been presented for the physiological and pathophysiological roles of epicardial fat, however this issue is poorly understood. Therefore, this mini-review will focus on epicardial fat distribution and the (patho)-physiological implications of this distribution. Potential molecular mechanisms that may drive structural and electrical myocardial remodeling attendant to fatty infiltration of the heart are also reviewed.
Collapse
Affiliation(s)
- Justus M B Anumonwo
- Department of Internal Medicine (Cardiovascular Medicine), Center for Arrythmia Research, University of Michigan, Ann Arbor, MI, United States
| | - Todd Herron
- Department of Internal Medicine (Cardiovascular Medicine), Center for Arrythmia Research, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
34
|
Proteomics and transcriptomics in atrial fibrillation. Herzschrittmacherther Elektrophysiol 2018; 29:70-75. [PMID: 29318371 DOI: 10.1007/s00399-017-0551-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 12/12/2017] [Indexed: 01/08/2023]
Abstract
Atrial fibrillation (AF) is the most common tachyarrhythmia. AF, due to substantial remodeling processes initiated in the atria, is a typically self-sustaining and progressive disease. Atrial remodeling has been intensively investigated at the molecular level in recent decades. Although the application of "omics" technologies has already significantly contributed to our current understanding of the pathophysiology of AF, the complexity of the latter and the large heterogeneity of AF patients remained a major limitation. With the advent of novel "omics" and by applying integrative approaches, it will be possible to extract more information and push boundaries. The present review will summarize the contribution of transcriptomics and proteomics to our understanding of the pathophysiology of AF.
Collapse
|
35
|
Rietdorf K, MacQueen H. Investigating interactions between epicardial adipose tissue and cardiac myocytes: what can we learn from different approaches? Br J Pharmacol 2017; 174:3542-3560. [PMID: 27882550 PMCID: PMC5610165 DOI: 10.1111/bph.13678] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 11/14/2016] [Accepted: 11/18/2016] [Indexed: 01/08/2023] Open
Abstract
Heart disease is a major cause of morbidity and mortality throughout the world. Some cardiovascular conditions can be modulated by lifestyle factors such as increased exercise or a healthier diet, but many require surgical or pharmacological interventions for their management. More targeted and less invasive therapies would be beneficial. Recently, it has become apparent that epicardial adipose tissue plays an important role in normal and pathological cardiac function, and it is now the focus of considerable research. Epicardial adipose tissue can be studied by imaging of various kinds, and these approaches have yielded much useful information. However, at a molecular level, it is more difficult to study as it is relatively scarce in animal models and, for practical and ethical reasons, not always available in sufficient quantities from patients. What is needed is a robust model system in which the interactions between epicardial adipocytes and cardiac myocytes can be studied, and physiologically relevant manipulations performed. There are drawbacks to conventional culture methods, not least the difficulty of culturing both cardiac myocytes and adipocytes, each of which has special requirements. We discuss the benefits of a three-dimensional co-culture model in which in vivo interactions can be replicated. LINKED ARTICLES This article is part of a themed section on Molecular Mechanisms Regulating Perivascular Adipose Tissue - Potential Pharmacological Targets? To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.20/issuetoc.
Collapse
Affiliation(s)
- Katja Rietdorf
- School of Life, Health and Chemical SciencesThe Open UniversityMilton KeynesUK
| | - Hilary MacQueen
- School of Life, Health and Chemical SciencesThe Open UniversityMilton KeynesUK
| |
Collapse
|
36
|
Kim JS, Shin SY, Kang JH, Yong HS, Na JO, Choi CU, Kim SH, Kim EJ, Rha SW, Park CG, Seo HS, Oh DJ, Hwang C, Kim YH, Lim HE. Influence of Sex on the Association Between Epicardial Adipose Tissue and Left Atrial Transport Function in Patients With Atrial Fibrillation: A Multislice Computed Tomography Study. J Am Heart Assoc 2017; 6:JAHA.117.006077. [PMID: 28778939 PMCID: PMC5586448 DOI: 10.1161/jaha.117.006077] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
BACKGROUND Epicardial adipose tissue (EAT) is known to play an important role in atrial fibrillation substrate remodeling; however, the influence of sex on the association between EAT and left atrial (LA) transport function has not been elucidated. METHODS AND RESULTS Of the 514 patients who underwent an index atrial fibrillation ablation procedure, 123 postmenopausal women with no history of hormone replacement therapy and 123 men who were matched for age, body mass index, type of atrial fibrillation, and CHADS2 score were enrolled. Before the procedure, LA volume, LA emptying fraction, and EAT volume were assessed using multislice computed tomography. Blood samples were obtained from a coronary sinus for analysis of serum adiponectin level before the ablation procedure. There were no differences in baseline demographics and laboratory findings between sexes. Compared with men, women had significantly less total EAT (P<0.001) and higher serum adiponectin levels (P=0.022) but higher proportions of periatrial EAT to total EAT volume (P/T EAT ratio, P<0.001), lower LA emptying fraction (P=0.042), and lower LA voltage (P=0.034). The ratio of periatrial to total EAT volume correlated significantly with LA emptying fraction and LA voltage in both sexes, whereas total EAT volume and serum adiponectin level did not. On multivariate analysis, increased LA volume and higher periatrial:total EAT volume ratio were independent predictors of decreased LA emptying fraction in both sexes. CONCLUSIONS Compared with matched men, postmenopausal women with atrial fibrillation had higher periatrial adiposity, which was independently correlated with decreased LA voltage and LA transport function.
Collapse
Affiliation(s)
- Jin-Seok Kim
- Division of Cardiology, Cardiovascular Center, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | - Seung Yong Shin
- Division of Cardiology, Heart Research Institute, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Jun Hyuk Kang
- Division of Cardiology, Cardiovascular Center, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | - Hwan Seok Yong
- Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | - Jin Oh Na
- Division of Cardiology, Cardiovascular Center, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | - Cheol Ung Choi
- Division of Cardiology, Cardiovascular Center, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | - Seong Hwan Kim
- Division of Cardiology, Cardiovascular Center, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | - Eung Ju Kim
- Division of Cardiology, Cardiovascular Center, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | - Seung-Woon Rha
- Division of Cardiology, Cardiovascular Center, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | - Chang Gyu Park
- Division of Cardiology, Cardiovascular Center, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | - Hong Seog Seo
- Division of Cardiology, Cardiovascular Center, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | - Dong Joo Oh
- Division of Cardiology, Cardiovascular Center, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | - Chun Hwang
- Division of Cardiology, Utah Valley Regional Medical Center, Provo, UT
| | - Young-Hoon Kim
- Division of Cardiology, Cardiovascular Center, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | - Hong Euy Lim
- Division of Cardiology, Cardiovascular Center, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
37
|
Abstract
Myocardial injury, mechanical stress, neurohormonal activation, inflammation, and/or aging all lead to cardiac remodeling, which is responsible for cardiac dysfunction and arrhythmogenesis. Of the key histological components of cardiac remodeling, fibrosis either in the form of interstitial, patchy, or dense scars, constitutes a key histological substrate of arrhythmias. Here we discuss current research findings focusing on the role of fibrosis, in arrhythmogenesis. Numerous studies have convincingly shown that patchy or interstitial fibrosis interferes with myocardial electrophysiology by slowing down action potential propagation, initiating reentry, promoting after-depolarizations, and increasing ectopic automaticity. Meanwhile, there has been increasing appreciation of direct involvement of myofibroblasts, the activated form of fibroblasts, in arrhythmogenesis. Myofibroblasts undergo phenotypic changes with expression of gap-junctions and ion channels thereby forming direct electrical coupling with cardiomyocytes, which potentially results in profound disturbances of electrophysiology. There is strong evidence that systemic and regional inflammatory processes contribute to fibrogenesis (i.e., structural remodeling) and dysfunction of ion channels and Ca2+ homeostasis (i.e., electrical remodeling). Recognizing the pivotal role of fibrosis in the arrhythmogenesis has promoted clinical research on characterizing fibrosis by means of cardiac imaging or fibrosis biomarkers for clinical stratification of patients at higher risk of lethal arrhythmia, as well as preclinical research on the development of antifibrotic therapies. At the end of this review, we discuss remaining key questions in this area and propose new research approaches. © 2017 American Physiological Society. Compr Physiol 7:1009-1049, 2017.
Collapse
Affiliation(s)
- My-Nhan Nguyen
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Central Clinical School, Monash University, Melbourne, Australia
| | - Helen Kiriazis
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Xiao-Ming Gao
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Central Clinical School, Monash University, Melbourne, Australia
| | - Xiao-Jun Du
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Central Clinical School, Monash University, Melbourne, Australia
| |
Collapse
|
38
|
Antonopoulos AS, Antoniades C. The role of epicardial adipose tissue in cardiac biology: classic concepts and emerging roles. J Physiol 2017; 595:3907-3917. [PMID: 28191635 DOI: 10.1113/jp273049] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 01/05/2017] [Indexed: 12/22/2022] Open
Abstract
Classic concepts about the role of epicardial adipose tissue (EpAT) in heart physiology include its role in cardiac metabolism, mechanical protection of coronaries, innervation and possibly cryoprotection of the heart too. Nevertheless, recent evidence has revealed that epicardial adipose tissue regulates multiple aspects of cardiac biology including myocardial redox state, intracellular Ca2+ cycling, the electrophysiological and contractile properties of cardiomyocytes, cardiac fibrosis as well as coronary atherosclerosis progression. Moreover, it is now understood that the communication between EpAT and the heart is regulated by complex bidirectional pathways, since not only do adipokines regulate cardiac function, but also the heart affects EpAT biology via paracrine 'reverse' signalling. Such complex interactions as well as epicardial fat accumulation as a consequence of cardiac disease and epicardium to adipocyte differentiation should be taken into account by the clinical studies investigating EpAT as a risk marker and its potential as a therapeutic target against cardiovascular disease. Further in-depth exploration of the molecular mechanisms regulating the cross-talk between the heart and EpAT is expected to enhance our understanding regarding the role of the latter in cardiac physiology and relevant disease mechanisms.
Collapse
|
39
|
Atrial natriuretic peptide regulates adipose tissue accumulation in adult atria. Proc Natl Acad Sci U S A 2017; 114:E771-E780. [PMID: 28096344 DOI: 10.1073/pnas.1610968114] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The abundance of epicardial adipose tissue (EAT) is associated with atrial fibrillation (AF), the most frequent cardiac arrhythmia. However, both the origin and the factors involved in EAT expansion are unknown. Here, we found that adult human atrial epicardial cells were highly adipogenic through an epithelial-mesenchymal transition both in vitro and in vivo. In a genetic lineage tracing the WT1CreERT2+/-RosatdT+/- mouse model subjected to a high-fat diet, adipocytes of atrial EAT derived from a subset of epicardial progenitors. Atrial myocardium secretome induces the adipogenic differentiation of adult mesenchymal epicardium-derived cells by modulating the balance between mesenchymal Wingless-type Mouse Mammary Tumor Virus integration site family, member 10B (Wnt10b)/β-catenin and adipogenic ERK/MAPK signaling pathways. The adipogenic property of the atrial secretome was enhanced in AF patients. The atrial natriuretic peptide secreted by atrial myocytes is a major adipogenic factor operating at a low concentration by binding to its natriuretic peptide receptor A (NPRA) receptor and, in turn, by activating a cGMP-dependent pathway. Hence, our data indicate cross-talk between EAT expansion and mechanical function of the atrial myocardium.
Collapse
|
40
|
Hayakawa N, Yamane T, Arias-Loza AP, Shinaji T, Wakabayashi H, Lapa C, Werner RA, Javadi MS, Pelzer T, Higuchi T. Impact of tissue photon attenuation in small animal cardiac PET imaging. Int J Cardiol 2017; 227:257-260. [PMID: 27839818 DOI: 10.1016/j.ijcard.2016.11.119] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 11/06/2016] [Indexed: 12/17/2022]
Abstract
OBJECTIVES Tissue photon attenuation is one of the essential artifacts requiring correction in clinical cardiac positron emission tomography (PET) imaging. However, due to small body size its impact on diagnostic accuracy in small rodents is considered to be limited or even ignorable. The present cardiac PET study compares lean and obese rats to determine the influence of tissue attenuation on quantitative assessment as well as regional tracer distribution. METHODS A dedicated small animal PET system equipped with a 57Co rotating source for transmission was used. To assess the impact of tissue attenuation in rats with different body sizes, cardiac 18F-FDG -PET studies for Zucker diabetic fatty rats (obese rats) and Zucker lean rats (lean rats) were performed. The radiotracer activity reduction by attenuation was compared between the two groups. Regional tracer distribution calculated with and without attenuation correction was also assessed. RESULTS The chest diameter was significantly longer in obese than in lean rats (5.6±0.3cm in obese and 4.5±0.2cm in lean rats, p<0.0001). Whereas the activity reduction by attenuation was significantly greater in obese than in lean rats (44.1±2.5% and 5.1±3.1%, p<0.0001), the regional variation of tissue attenuation among the ventricular walls was minimal in both lean (p=0.73) and obese rats (p=0.65). CONCLUSION Attenuation correction is indispensable for accurate comparison of cardiac tracer activity between animals with different body size, whereas it can be omitted for evaluation of regional tracer distribution.
Collapse
Affiliation(s)
- Nobuyuki Hayakawa
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany; Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Tomohiko Yamane
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany; Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Anahi-Paula Arias-Loza
- Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany; Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Tetsuya Shinaji
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany; Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Hiroshi Wakabayashi
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany; Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Constantin Lapa
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Rudolf A Werner
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany; Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Mehrbod S Javadi
- Division of Nuclear Medicine, Russell H. Morgan Department of Radiology, Johns Hopkins University, Baltimore, MD, United States
| | - Theo Pelzer
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Takahiro Higuchi
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany; Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany.
| |
Collapse
|
41
|
Kusayama T, Furusho H, Kashiwagi H, Kato T, Murai H, Usui S, Kaneko S, Takamura M. Inflammation of left atrial epicardial adipose tissue is associated with paroxysmal atrial fibrillation. J Cardiol 2016; 68:406-411. [DOI: 10.1016/j.jjcc.2015.11.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 10/20/2015] [Accepted: 11/04/2015] [Indexed: 01/01/2023]
|
42
|
Goette A, Kalman JM, Aguinaga L, Akar J, Cabrera JA, Chen SA, Chugh SS, Corradi D, D'Avila A, Dobrev D, Fenelon G, Gonzalez M, Hatem SN, Helm R, Hindricks G, Ho SY, Hoit B, Jalife J, Kim YH, Lip GYH, Ma CS, Marcus GM, Murray K, Nogami A, Sanders P, Uribe W, Van Wagoner DR, Nattel S. EHRA/HRS/APHRS/SOLAECE expert consensus on Atrial cardiomyopathies: Definition, characterisation, and clinical implication. J Arrhythm 2016; 32:247-78. [PMID: 27588148 PMCID: PMC4996910 DOI: 10.1016/j.joa.2016.05.002] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Andreas Goette
- Departement of Cardiology and Intensive Care Medicine, St. Vincenz-Hospital Paderborn, Working Group: Molecular Electrophysiology, University Hospital Magdeburg, Germany
| | - Jonathan M Kalman
- University of Melbourne, Royal Melbourne Hospital, Melbourne, VIC, Australia
| | | | | | | | | | - Sumeet S Chugh
- The Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | | | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | | | - Mario Gonzalez
- Penn State Heart and Vascular Institute, Penn State University, Hershey, PA, USA
| | - Stephane N Hatem
- Department of Cardiology, Assistance Publique - Hô pitaux de Paris, Pitié-Salpêtrière Hospital, Sorbonne University, INSERM UMR_S1166, Institute of Cardiometabolism and Nutrition-ICAN, Paris, France
| | - Robert Helm
- Boston University School of Medicine, Boston Medical Center, Boston, MA, USA
| | | | - Siew Yen Ho
- Royal Brompton Hospital and Imperial College London, London, UK
| | - Brian Hoit
- UH Case Medical Center, Cleveland, OH, USA
| | | | | | | | | | | | | | | | - Prashanthan Sanders
- Centre for Heart Rhythm Disorders, South Australian Health and Medical Research Institute, University of Adelaide and Royal Adelaide Hospital, Adelaide, Australia
| | - William Uribe
- Electrophysiology Deparment at Centros Especializados de San Vicente Fundació n and Clínica CES. Universidad CES, Universidad Pontificia Bolivariana (UPB), Medellin, Colombia
| | | | - Stanley Nattel
- Université de Montréal, Montreal Heart Institute Research Center and McGill University, Montreal, Quebec, Canada; Institute of Pharmacology, West German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen, Essen, Germany
| | | | | |
Collapse
|
43
|
Goette A, Kalman JM, Aguinaga L, Akar J, Cabrera JA, Chen SA, Chugh SS, Corradi D, D'Avila A, Dobrev D, Fenelon G, Gonzalez M, Hatem SN, Helm R, Hindricks G, Ho SY, Hoit B, Jalife J, Kim YH, Lip GYH, Ma CS, Marcus GM, Murray K, Nogami A, Sanders P, Uribe W, Van Wagoner DR, Nattel S. EHRA/HRS/APHRS/SOLAECE expert consensus on atrial cardiomyopathies: definition, characterization, and clinical implication. Europace 2016; 18:1455-1490. [PMID: 27402624 DOI: 10.1093/europace/euw161] [Citation(s) in RCA: 462] [Impact Index Per Article: 57.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Andreas Goette
- Departement of Cardiology and Intensive Care Medicine, St. Vincenz-Hospital Paderborn, Working Group: Molecular Electrophysiology, University Hospital Magdeburg, Germany
| | - Jonathan M Kalman
- University of Melbourne, Royal Melbourne Hospital, Melbourne, VIC, Australia
| | | | | | | | | | - Sumeet S Chugh
- The Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | | | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | | | - Mario Gonzalez
- Penn State Heart and Vascular Institute, Penn State University, Hershey, PA, USA
| | - Stephane N Hatem
- Department of Cardiology, Assistance Publique - Hôpitaux de Paris, Pitié-Salpêtrière Hospital; Sorbonne University; INSERM UMR_S1166; Institute of Cardiometabolism and Nutrition-ICAN, Paris, France
| | - Robert Helm
- Boston University School of Medicine, Boston Medical Center, Boston, MA, USA
| | | | - Siew Yen Ho
- Royal Brompton Hospital and Imperial College London, London, UK
| | - Brian Hoit
- UH Case Medical Center, Cleveland, OH, USA
| | | | | | | | | | | | | | | | - Prashanthan Sanders
- Centre for Heart Rhythm Disorders, South Australian Health and Medical Research Institute, University of Adelaide and Royal Adelaide Hospital, Adelaide, Australia
| | - William Uribe
- Electrophysiology Deparment at Centros Especializados de San Vicente Fundación and Clínica CES. Universidad CES, Universidad Pontificia Bolivariana (UPB), Medellin, Colombia
| | | | - Stanley Nattel
- Université de Montréal, Montreal Heart Institute Research Center and McGill University, Montreal, Quebec, Canada .,Institute of Pharmacology, West German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen, Essen, Germany
| | | |
Collapse
|
44
|
EHRA/HRS/APHRS/SOLAECE expert consensus on atrial cardiomyopathies: Definition, characterization, and clinical implication. Heart Rhythm 2016; 14:e3-e40. [PMID: 27320515 DOI: 10.1016/j.hrthm.2016.05.028] [Citation(s) in RCA: 214] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Indexed: 12/21/2022]
|
45
|
Liu GZ, Hou TT, Yuan Y, Hang PZ, Zhao JJ, Sun L, Zhao GQ, Zhao J, Dong JM, Wang XB, Shi H, Liu YW, Zhou JH, Dong ZX, Liu Y, Zhan CC, Li Y, Li WM. Fenofibrate inhibits atrial metabolic remodelling in atrial fibrillation through PPAR-α/sirtuin 1/PGC-1α pathway. Br J Pharmacol 2016; 173:1095-109. [PMID: 26787506 DOI: 10.1111/bph.13438] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 01/09/2016] [Accepted: 01/13/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE Atrial metabolic remodelling is critical for the process of atrial fibrillation (AF). The PPAR-α/sirtuin 1 /PPAR co-activator α (PGC-1α) pathway plays an important role in maintaining energy metabolism. However, the effect of the PPAR-α agonist fenofibrate on AF is unclear. Therefore, the aim of this study was to determine the effect of fenofibrate on atrial metabolic remodelling in AF and explore its possible mechanisms of action. EXPERIMENTAL APPROACH The expression of metabolic proteins was examined in the left atria of AF patients. Thirty-two rabbits were divided into sham, AF (pacing with 600 beats·min(-1) for 1 week), fenofibrate treated (pretreated with fenofibrate before pacing) and fenofibrate alone treated (for 2 weeks) groups. HL-1 cells were subjected to rapid pacing in the presence or absence of fenofibrate, the PPAR-α antagonist GW6471 or sirtuin 1-specific inhibitor EX527. Metabolic factors, circulating biochemical metabolites, atrial electrophysiology, adenine nucleotide levels and accumulation of glycogen and lipid droplets were assessed. KEY RESULTS The PPAR-α/sirtuin 1/PGC-1α pathway was significantly inhibited in AF patients and in the rabbit/HL-1 cell models, resulting in a reduction of key downstream metabolic factors; this effect was significantly restored by fenofibrate. Fenofibrate prevented the alterations in circulating biochemical metabolites, reduced the level of adenine nucleotides and accumulation of glycogen and lipid droplets, reversed the shortened atrial effective refractory period and increased risk of AF. CONCLUSION AND IMPLICATIONS Fenofibrate inhibited atrial metabolic remodelling in AF by regulating the PPAR-α/sirtuin 1/PGC-1α pathway. The present study may provide a novel therapeutic strategy for AF.
Collapse
Affiliation(s)
- Guang-Zhong Liu
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Ting-Ting Hou
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yue Yuan
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Peng-Zhou Hang
- Institute of Clinical Pharmacology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Jing-Jing Zhao
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Li Sun
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Guan-Qi Zhao
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Jing Zhao
- Key Laboratory of Cardiac Diseases and Heart, Failure of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Jing-Mei Dong
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xiao-Bing Wang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Hang Shi
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yong-Wu Liu
- Centre for Drug Safety Evaluation, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China
| | - Jing-Hua Zhou
- Department of Morphology, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China
| | - Zeng-Xiang Dong
- Key Laboratory of Cardiac Diseases and Heart, Failure of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yang Liu
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Cheng-Chuang Zhan
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yue Li
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China.,Key Laboratory of Cardiac Diseases and Heart, Failure of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Wei-Min Li
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China.,Key Laboratory of Cardiac Diseases and Heart, Failure of Harbin Medical University, Harbin, Heilongjiang Province, China
| |
Collapse
|
46
|
|
47
|
Chirumbolo S. Commentary: Heart Fat Infiltration in Subjects With and Without Coronary Artery Disease. Front Cardiovasc Med 2016; 3:2. [PMID: 26870737 PMCID: PMC4740777 DOI: 10.3389/fcvm.2016.00002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 01/11/2016] [Indexed: 11/13/2022] Open
|
48
|
Hatem SN, Redheuil A, Gandjbakhch E. Cardiac adipose tissue and atrial fibrillation: the perils of adiposity. Cardiovasc Res 2016; 109:502-9. [PMID: 26790475 DOI: 10.1093/cvr/cvw001] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Accepted: 12/09/2015] [Indexed: 12/25/2022] Open
Abstract
The amount of adipose tissue that accumulates around the atria is associated with the risk, persistence, and severity of atrial fibrillation (AF). A strong body of clinical and experimental evidence indicates that this relationship is not an epiphenomenon but is the result of complex crosstalk between the adipose tissue and the neighbouring atrial myocardium. For instance, epicardial adipose tissue is a major source of adipokines, inflammatory cytokines, or reactive oxidative species, which can contribute to the fibrotic remodelling of the atrial myocardium. Fibro-fatty infiltrations of the subepicardium could also contribute to the functional disorganization of the atrial myocardium. The observation that obesity is associated with distinct structural and functional remodelling of the atria has opened new perspectives of treating AF substrate with aggressive risk factor management. Advances in cardiac imaging should lead to an improved ability to visualize myocardial fat depositions and to localize AF substrates.
Collapse
Affiliation(s)
- Stéphane N Hatem
- Sorbonne University, Faculté de médicine, Assistance Publique-Hôpitaux de Paris, GH Pitié-Salpêtrière Hospital, INSERM UMR_S1166, Cardiology Department, Institute of Cardiometabolism and Nutrition-ICAN, 91, boulevard de l'hôpital, 75013 Paris, France
| | - Alban Redheuil
- Sorbonne Universités, Université Pierre et Marie Curie UPMC, Laboratoire d'imagerie biomédicale INSERM UMR_S1146, Cardiovascular Imaging Department, ICAN Imaging Core Lab, Paris, France
| | - Estelle Gandjbakhch
- Sorbonne University, Faculté de médicine, Assistance Publique-Hôpitaux de Paris, GH Pitié-Salpêtrière Hospital, INSERM UMR_S1166, Cardiology Department, Institute of Cardiometabolism and Nutrition-ICAN, 91, boulevard de l'hôpital, 75013 Paris, France
| |
Collapse
|
49
|
Fabritz L, Guasch E, Antoniades C, Bardinet I, Benninger G, Betts TR, Brand E, Breithardt G, Bucklar-Suchankova G, Camm AJ, Cartlidge D, Casadei B, Chua WWL, Crijns HJGM, Deeks J, Hatem S, Hidden-Lucet F, Kääb S, Maniadakis N, Martin S, Mont L, Reinecke H, Sinner MF, Schotten U, Southwood T, Stoll M, Vardas P, Wakili R, West A, Ziegler A, Kirchhof P. Defining the major health modifiers causing atrial fibrillation: a roadmap to underpin personalized prevention and treatment. Nat Rev Cardiol 2015; 13:230-7. [DOI: 10.1038/nrcardio.2015.194] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
50
|
Haemers P, Hamdi H, Guedj K, Suffee N, Farahmand P, Popovic N, Claus P, LePrince P, Nicoletti A, Jalife J, Wolke C, Lendeckel U, Jaïs P, Willems R, Hatem SN. Atrial fibrillation is associated with the fibrotic remodelling of adipose tissue in the subepicardium of human and sheep atria. Eur Heart J 2015; 38:53-61. [DOI: 10.1093/eurheartj/ehv625] [Citation(s) in RCA: 154] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 09/12/2015] [Accepted: 10/27/2015] [Indexed: 11/15/2022] Open
|