1
|
Palermi A, Molinari LV, Ricci F, Gallina S, Renda G. Practical guidance for management of atrial fibrillation in sports cardiology. Curr Probl Cardiol 2025; 50:102995. [PMID: 39890043 DOI: 10.1016/j.cpcardiol.2025.102995] [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: 01/17/2025] [Accepted: 01/28/2025] [Indexed: 02/03/2025]
Abstract
IMPORTANCE Atrial fibrillation (AF), the most prevalent sustained arrhythmia, portends higher risk of cardiovascular morbidity and mortality, and is associated with quantifiable impairment in quality of life. While physical activity is widely recognized for its cardiovascular benefits, recent evidence challenges its role in the development of AF. OBSERVATIONS Emerging data suggest a U-shaped relationship between physical activity and AF risk. Mild to moderate exercise appears protective, whereas prolonged, high-intensity activity is associated with an increased risk, possibly driven by cardiac remodeling, autonomic alterations, and atrial substrate changes typical of the so-called athlete's heart. This relationship is further modulated by other factors, including genetic predisposition, acquired cardiac conditions, and stimulant use. With the growing participation of aging populations in sports, the clinical management of AF in athletes presents unique challenges. Pharmacological strategies for rhythm and rate control may conflict with performance goals, and the use of anticoagulants must be carefully balanced against the risk of traumatic bleeding in contact sports. Recent evidence supports strategies such as short-term anticoagulant withdrawal to mitigate hemorrhagic risk while maintaining thromboembolic protection. A personalized approach, incorporating shared decision-making, is essential, particularly for elite athletes. CONCLUSIONS AND RELEVANCE While regular physical activity generally confers cardiovascular benefits, prolonged intense exercise bouts may paradoxically increase the risk of AF. The evaluation and management of AF in athletes require a tailored, multidisciplinary approach that accounts for the distinct needs of this population.
Collapse
Affiliation(s)
- Andrea Palermi
- Department of Neuroscience, Imaging and Clinical Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti 66100, Italy
| | - Lorenzo Vilhelm Molinari
- Department of Neuroscience, Imaging and Clinical Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti 66100, Italy
| | - Fabrizio Ricci
- Department of Neuroscience, Imaging and Clinical Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti 66100, Italy; Heart Department, SS. Annunziata Hospital, University Cardiology Division, Chieti, Italy; Institute for Advanced Biomedical Technologies, G. D'Annunzio University of Chieti-Pescara, Chieti 66100, Italy; Department of Clinical Sciences, Lund University, Jan Waldenströms gata 35, Malmö 214 28, Sweden
| | - Sabina Gallina
- Department of Neuroscience, Imaging and Clinical Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti 66100, Italy; Heart Department, SS. Annunziata Hospital, University Cardiology Division, Chieti, Italy
| | - Giulia Renda
- Department of Neuroscience, Imaging and Clinical Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti 66100, Italy; Heart Department, SS. Annunziata Hospital, University Cardiology Division, Chieti, Italy.
| |
Collapse
|
2
|
Lewis J, Bentley RF, Connelly KA, Dorian P, Goodman JM. Are Subjective Reports of Exercise Intensity Accurate in Recreational Athletes? Can J Cardiol 2025; 41:531-541. [PMID: 39522870 DOI: 10.1016/j.cjca.2024.11.008] [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: 07/30/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Quantifying exercise intensity accurately is crucial for understanding links between cumulative exercise and cardiovascular outcomes. Exercise burden, the integral of intensity and duration is often estimated from subjective self-reports which have uncertain accuracy. METHODS We studied 40 endurance athletes (EAs) 41 to 69 years of age with > 10 years of training history during a scripted outdoor 42-km cycling training session. Heart rate and power output (watts) were continuously measured. Reports of perceived exertion (RPE) using a word (RPEWord) and numerical Borg scale (RPEBorg) were obtained during and 30 minutes postride and were related to cardiac (heart rate) and metabolic (metabolic equivalent [MET] per minute) exercise endpoints. RESULTS RPEs were highly variable, underestimating objective metrics of exercise intensity. Poor agreement was observed between either scale reported 30 minutes after exercise relative to heart rate: exercise RPEBorg vs mean exercise heart rate and % heart ratepeak (both rs = 0.29; P = 0.07), with no agreement between either scale vs other objective endpoints. Agreement between RPEBorg and RPEWord was good during exercise (rs = 0.86; 95% confidence interval (CI), 0.75- 0.92; P = 0.001), but diminished postride (rs = 0.54; 95% CI, 0.28-0.73; P = 0.001). Different cardiac and metabolic profiles during exercise and a contrast between metabolic and cardiac burden was greater in less fit individuals as they accrued greater cardiac (14,039 ± 2649 vs 11,784 ± 1132 heart rate per minute; P < 0.01) but lower metabolic (808 ± 59 vs 858 ± 61 MET per minute; P < 0.05) burden vs fitter EA. CONCLUSIONS Caution is advised in interpreting MET per minute and heart rate burden estimated from self-reports. Objective measurements of exercise intensity are required for detailed assessment of the risks and benefits of long-term exercise.
Collapse
Affiliation(s)
- Jennifer Lewis
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, Ontario, Canada
| | - Robert F Bentley
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, Ontario, Canada
| | - Kim A Connelly
- Keenan Research Centre for Biomedical Science, Division of Cardiology, St Michael's Hospital, Unity Health, Toronto, Ontario, Canada and University of Toronto and Heart and Stroke/Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Ontario, Canada
| | - Paul Dorian
- Keenan Research Centre for Biomedical Science, Division of Cardiology, St Michael's Hospital, Unity Health, Toronto, Ontario, Canada and University of Toronto and Heart and Stroke/Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Ontario, Canada
| | - Jack M Goodman
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, Ontario, Canada and Division of Cardiology, Mount Sinai Hospital, University of Toronto and Heart and Stroke/Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Ontario, Canada.
| |
Collapse
|
3
|
Bass-Stringer S, Bernardo BC, Yildiz GS, Matsumoto A, Kiriazis H, Harmawan CA, Tai CMK, Chooi R, Bottrell L, Ezeani M, Donner DG, D'Elia AA, Ooi JYY, Mellett NA, Luo J, Masterman EI, Janssens K, Olshansky G, Howden EJ, Cross JH, Hagemeyer CE, Lin RCY, Thomas CJ, Magor GW, Perkins AC, Marwick TH, Kawakami H, Meikle PJ, Greening DW, Weeks KL, La Gerche A, Tham YK, McMullen JR. Reduced PI3K(p110α) induces atrial myopathy, and PI3K-related lipids are dysregulated in athletes with atrial fibrillation. JOURNAL OF SPORT AND HEALTH SCIENCE 2025; 14:101023. [PMID: 39826614 PMCID: PMC11978378 DOI: 10.1016/j.jshs.2025.101023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 11/11/2024] [Accepted: 12/10/2024] [Indexed: 01/22/2025]
Abstract
BACKGROUND Elucidating mechanisms underlying atrial myopathy, which predisposes individuals to atrial fibrillation (AF), will be critical for preventing/treating AF. In a serendipitous discovery, we identified atrial enlargement, fibrosis, and thrombi in mice with reduced phosphoinositide 3-kinase (PI3K) in cardiomyocytes. PI3K(p110α) is elevated in the heart with exercise and is critical for exercise-induced ventricular enlargement and protection, but the role in the atria was unknown. Physical inactivity and extreme endurance exercise can increase AF risk. Therefore, our objective was to investigate whether too little and/or too much PI3K alone induces cardiac pathology. METHODS New cardiomyocyte-specific transgenic mice with increased or decreased PI3K(p110α) activity were generated. Multi-omics was conducted in mouse atrial tissue, and lipidomics in human plasma. RESULTS Elevated PI3K led to an increase in heart size with preserved/enhanced function. Reduced PI3K led to atrial dysfunction, fibrosis, arrhythmia, increased susceptibility to atrial enlargement and thrombi, and dysregulation of monosialodihexosylganglioside (GM3), a lipid that regulates insulin-like growth factor-1 (IGF1)-PI3K signaling. Proteomic profiling identified distinct signatures and signaling networks across atria with varying degrees of dysfunction, enlargement, and thrombi, including commonalities with the human AF proteome. PI3K-related lipids were dysregulated in plasma from athletes with AF. CONCLUSION PI3K(p110α) is a critical regulator of atrial biology and function in mice. This work provides a proteomic resource of candidates for further validation as potential new drug targets and biomarkers for atrial myopathy. Further investigation of PI3K-related lipids as markers for identifying individuals at risk of AF is warranted. Dysregulation of PI3K may contribute to the association between increased cardiac risk with physical inactivity and extreme endurance exercise.
Collapse
Affiliation(s)
- Sebastian Bass-Stringer
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC 3086, Australia
| | - Bianca C Bernardo
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC 3800, Australia; Department of Paediatrics, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Gunes S Yildiz
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Aya Matsumoto
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Helen Kiriazis
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC 3010, Australia
| | | | - Celeste M K Tai
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Roger Chooi
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Lauren Bottrell
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Martin Ezeani
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Daniel G Donner
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Aascha A D'Elia
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Jenny Y Y Ooi
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | | | - Jieting Luo
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Emma I Masterman
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Kristel Janssens
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Gavriel Olshansky
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Erin J Howden
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Jonathon H Cross
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Christoph E Hagemeyer
- Australian Centre for Blood Diseases, Monash University, Clayton, VIC 3800, Australia
| | - Ruby C Y Lin
- School of Medical Sciences, University of NSW, Sydney, NSW 2052, Australia; Westmead Institute for Medical Research, Sydney, NSW 2145, Australia
| | - Colleen J Thomas
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC 3086, Australia; Centre for Cardiovascular Biology and Disease Research, La Trobe University, Bundoora, VIC 3086, Australia
| | - Graham W Magor
- Australian Centre for Blood Diseases, Monash University, Clayton, VIC 3800, Australia
| | - Andrew C Perkins
- Australian Centre for Blood Diseases, Monash University, Clayton, VIC 3800, Australia
| | - Thomas H Marwick
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC 3010, Australia; Menzies Institute for Medical Research, University of TAS, Hobart, TAS 7000, Australia; Department of Cardiology, Royal Hobart Hospital, Hobart, TAS 7001, Australia
| | - Hiroshi Kawakami
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Department of Cardiology, Pulmonology, Hypertension, and Nephrology, Ehime University Graduate School of Medicine, Toon 791-0295, Japan
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC 3010, Australia; Centre for Cardiovascular Biology and Disease Research, La Trobe University, Bundoora, VIC 3086, Australia
| | - David W Greening
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC 3010, Australia; Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC 3086, Australia
| | - Kate L Weeks
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC 3800, Australia; Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC 3010, Australia; Department of Anatomy & Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - André La Gerche
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Yow Keat Tham
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC 3800, Australia; Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC 3010, Australia; Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC 3086, Australia
| | - Julie R McMullen
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC 3086, Australia; Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC 3800, Australia; Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC 3010, Australia; Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC 3086, Australia; Department of Physiology and Department of Medicine Alfred Hospital, Monash University, Clayton, VIC 3800, Australia; Heart Research Institute, Newtown, NSW 2042, Australia.
| |
Collapse
|
4
|
Ostojic M, Ostojic M, Petrovic O, Nedeljkovic-Arsenovic O, Perone F, Banovic M, Stojmenovic T, Stojmenovic D, Giga V, Beleslin B, Nedeljkovic I. Endurance Sports and Atrial Fibrillation: A Puzzling Conundrum. J Clin Med 2024; 13:7691. [PMID: 39768614 PMCID: PMC11677941 DOI: 10.3390/jcm13247691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/09/2024] [Accepted: 11/17/2024] [Indexed: 01/11/2025] Open
Abstract
The confirmed benefits of regular moderate exercise on cardiovascular health have positioned athletes as an illustration of well-being. However, concerns have arisen regarding the potential predisposition to arrhythmias in individuals engaged in prolonged strenuous exercise. Atrial fibrillation (AF), the most common heart arrhythmia, is typically associated with age-related risks but has been documented in otherwise healthy young and middle-aged endurance athletes. The mechanism responsible for AF involves atrial remodeling, fibrosis, inflammation, and alterations in autonomic tone, all of which intersect with the demands of endurance sports, cumulative training hours, and competitive participation. This unique lifestyle requires a tailored therapeutic approach, often favoring radiofrequency ablation as the preferred treatment. As the number of professional and non-professional athletes engaging in high-level daily sports activities rises, awareness of AF within this demographic becomes imperative. This review delivers the etiology, pathophysiology, and therapeutic considerations surrounding AF in endurance sports.
Collapse
Affiliation(s)
- Marina Ostojic
- Cardiology Clinic, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (M.O.); (O.P.); (M.B.); (V.G.); (B.B.); (I.N.)
- School of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Mladen Ostojic
- Cardiology Clinic, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (M.O.); (O.P.); (M.B.); (V.G.); (B.B.); (I.N.)
| | - Olga Petrovic
- Cardiology Clinic, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (M.O.); (O.P.); (M.B.); (V.G.); (B.B.); (I.N.)
- School of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Olga Nedeljkovic-Arsenovic
- School of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
- Radiology and MRI Department, University Clinical Center of Serbia, 11000 Belgrade, Serbia
| | - Francesco Perone
- Cardiac Rehabilitation Unit, Rehabilitation Clinic “Villa delleMagnolie”, 81020 Castel Morrone, Italy;
| | - Marko Banovic
- Cardiology Clinic, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (M.O.); (O.P.); (M.B.); (V.G.); (B.B.); (I.N.)
- School of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Tamara Stojmenovic
- Faculty of Physical Culture and Sports Management, Singidunum University, 11000 Belgrade, Serbia;
| | - Dragutin Stojmenovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Vojislav Giga
- Cardiology Clinic, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (M.O.); (O.P.); (M.B.); (V.G.); (B.B.); (I.N.)
- School of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Branko Beleslin
- Cardiology Clinic, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (M.O.); (O.P.); (M.B.); (V.G.); (B.B.); (I.N.)
- School of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Ivana Nedeljkovic
- Cardiology Clinic, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (M.O.); (O.P.); (M.B.); (V.G.); (B.B.); (I.N.)
- School of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| |
Collapse
|
5
|
D’Ambrosio P, Claessen G, Kistler PM, Heidbuchel H, Kalman JM, La Gerche A. Ventricular arrhythmias in association with athletic cardiac remodelling. Europace 2024; 26:euae279. [PMID: 39499658 PMCID: PMC11641426 DOI: 10.1093/europace/euae279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/04/2024] [Accepted: 10/28/2024] [Indexed: 11/07/2024] Open
Abstract
Athletes are predisposed to atrial arrhythmias but the association between intense endurance exercise training, ventricular arrhythmias (VAs), and sudden cardiac death is less well established. Thus, it is unclear whether the 'athlete's heart' promotes specific arrhythmias or whether it represents a more general pro-arrhythmogenic phenotype. Whilst direct causality has not been established, it appears possible that repeated exposure to high-intensity endurance exercise in some athletes contributes to formation of pro-arrhythmic cardiac phenotypes that underlie VAs. Theories regarding potential mechanisms for exercise-induced VAs include repeated bouts of myocardial inflammation and stretch-induced cellular remodelling. Small animal models provide some insights, but larger animal and human data are sparse. The current clinical approach to VAs in athletes is to differentiate those with and without structural or electrical heart disease. However, if the athlete's heart involves a degree of pro-arrhythmogenic remodelling, then this may not be such a simple dichotomy. Questions are posed by athletes with VAs in combination with extreme remodelling. Some markers, such as scar on magnetic resonance imaging, may point towards a less benign phenotype but are also quite common in ostensibly healthy athletes. Other clinical and invasive electrophysiology features may be helpful in identifying the at-risk athlete. This review seeks to discuss the association between athletic training and VAs. We will discuss the potential mechanisms, clinical significance, and approach to the management of athletes with VAs.
Collapse
MESH Headings
- Humans
- Ventricular Remodeling
- Athletes
- Cardiomegaly, Exercise-Induced
- Death, Sudden, Cardiac/prevention & control
- Death, Sudden, Cardiac/etiology
- Animals
- Arrhythmias, Cardiac/physiopathology
- Arrhythmias, Cardiac/diagnosis
- Arrhythmias, Cardiac/therapy
- Arrhythmias, Cardiac/etiology
- Risk Factors
- Tachycardia, Ventricular/physiopathology
- Tachycardia, Ventricular/etiology
- Tachycardia, Ventricular/diagnosis
Collapse
Affiliation(s)
- Paolo D’Ambrosio
- Department of Medicine, The University of Melbourne, Grattan St, Parkville, VIC 3010, Australia
- Heart Exercise & Research Trials (HEART) Lab, St Vincent’s Institute, 9 Princes St, Fitzroy, VIC 3065, Australia
- Department of Cardiology, The Royal Melbourne Hospital, 300 Grattan St, Parkville, VIC 3052, Australia
| | - Guido Claessen
- Faculty of Medicine and Life Sciences, LCRC, UHasselt, Biomedical Research Institute, Diepenbeek, Belgium
- Hartcentrum Hasselt, Jessa Ziekenhuis, Belgium
- Department of Cardiovascular Sciences, KU Leuven, Belgium
| | - Peter M Kistler
- Department of Medicine, The University of Melbourne, Grattan St, Parkville, VIC 3010, Australia
- Department of Cardiology, The Alfred Hospital, Melbourne, VIC, Australia
- Department of Medicine, Monash University, Clayton, VIC, Australia
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Hein Heidbuchel
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
| | - Jonathan M Kalman
- Department of Medicine, The University of Melbourne, Grattan St, Parkville, VIC 3010, Australia
- Department of Cardiology, The Royal Melbourne Hospital, 300 Grattan St, Parkville, VIC 3052, Australia
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - André La Gerche
- Department of Medicine, The University of Melbourne, Grattan St, Parkville, VIC 3010, Australia
- Heart Exercise & Research Trials (HEART) Lab, St Vincent’s Institute, 9 Princes St, Fitzroy, VIC 3065, Australia
- Department of Cardiology, St Vincent’s Hospital, Fitzroy, VIC, Australia
- HEART Lab, Victor Chang Cardiovascular Research Institute, Darlinghurst, NSW, Australia
| |
Collapse
|
6
|
Wang Y, Wang Y, Xu D. Effects of different exercise methods and intensities on the incidence and prognosis of atrial fibrillation. Trends Cardiovasc Med 2024; 34:510-515. [PMID: 38216075 DOI: 10.1016/j.tcm.2024.01.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 12/21/2023] [Accepted: 01/06/2024] [Indexed: 01/14/2024]
Abstract
Atrial fibrillation (AF), the most common sustained arrhythmia in clinical practice, exhibits a higher risk of cardiovascular adverse events. Exercise plays a crucial role in AF prevention, but the effects of different exercise types and doses are inconclusive. This review aims to comprehensively explore the most recent evidence and possible mechanisms of diverse exercise modalities concerning AF incidence and therapeutic outcomes. Multiple studies underscore the efficacy of moderate-intensity continuous training (MICT) in reducing AF incidence and symptom burden, rendering it the currently favored exercise therapy for AF patients. High-intensity interval training (HIIT) shows promise, potentially surpassing MICT, especially in reducing age-related AF susceptibility and improving symptoms and exercise capacity. Conversely, prolonged high-intensity endurance exercise exacerbates AF risk due to excessive exercise volume, with potential mechanisms encompassing irreversible atrial remodeling, heightened inflammation, and increased vagal tone. In summation, MICT is a secure strategy for populations in mitigating the risk associated with AF incidence and secondary cardiovascular events and should be encouraged. Also, it is recommended to initiate large-scale clinical intervention trials encompassing a variety of exercise types to delineate the optimal exercise prescription for cardiovascular patients, including those afflicted with AF.
Collapse
Affiliation(s)
- Yurong Wang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Ying Wang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Department of Cardiovascular Medicine, Yueyang Central Hospital, China
| | - Danyan Xu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
7
|
Kourek C, Briasoulis A, Tsougos E, Paraskevaidis I. Atrial Fibrillation in Elite Athletes: A Comprehensive Review of the Literature. J Cardiovasc Dev Dis 2024; 11:315. [PMID: 39452285 PMCID: PMC11508555 DOI: 10.3390/jcdd11100315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/26/2024] Open
Abstract
Although the benefits of exercise training have been shown repeatedly in many studies, its relationship with the occurrence of atrial fibrillation (AF) in competitive athletes still remains controversial. In the present review, we sought to demonstrate a comprehensive report of the incidence, pathophysiology, and therapeutic approaches to AF in elite athletes. A 2 to 10 times higher frequency of AF has been shown in many studies in high-intensity endurance athletes compared to individuals who do not exercise. Moreover, a U-shaped relationship between male elite athletes and AF is demonstrated through this finding, while the type and the years of physical activity seem to relate to AF development. A strong correlation seems to exist among the type of exercise (endurance sports), age (>55 years), gender (males), and the time of exercise training, all contributing to an increased risk of AF. The pathophysiology of AF still remains unclear; however, several theories suggest that complex mechanisms are involved, such as bi-atrial dilatation, pulmonary vein stretching, cardiac inflammation, fibrosis, and increased vagal tone. Elite athletes with AF require a comprehensive clinical evaluation and risk factor optimization, similar to the approach taken for nonathletes. Although anticoagulation and rate or rhythm control are cornerstones of AF management, there are still no specific guidelines for elite athletes.
Collapse
Affiliation(s)
- Christos Kourek
- Department of Cardiology, 417 Army Share Fund Hospital of Athens (NIMTS), 11521 Athens, Greece;
| | - Alexandros Briasoulis
- Medical School of Athens, National and Kapodistrian University of Athens, 15772 Athens, Greece;
| | - Elias Tsougos
- Department of Cardiology, Hygeia Hospital, 15123 Athens, Greece;
| | - Ioannis Paraskevaidis
- Medical School of Athens, National and Kapodistrian University of Athens, 15772 Athens, Greece;
- Department of Cardiology, Hygeia Hospital, 15123 Athens, Greece;
| |
Collapse
|
8
|
Lampert R, Chung EH, Ackerman MJ, Arroyo AR, Darden D, Deo R, Dolan J, Etheridge SP, Gray BR, Harmon KG, James CA, Kim JH, Krahn AD, La Gerche A, Link MS, MacIntyre C, Mont L, Salerno JC, Shah MJ. 2024 HRS expert consensus statement on arrhythmias in the athlete: Evaluation, treatment, and return to play. Heart Rhythm 2024; 21:e151-e252. [PMID: 38763377 DOI: 10.1016/j.hrthm.2024.05.018] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 05/09/2024] [Indexed: 05/21/2024]
Abstract
Youth and adult participation in sports continues to increase, and athletes may be diagnosed with potentially arrhythmogenic cardiac conditions. This international multidisciplinary document is intended to guide electrophysiologists, sports cardiologists, and associated health care team members in the diagnosis, treatment, and management of arrhythmic conditions in the athlete with the goal of facilitating return to sport and avoiding the harm caused by restriction. Expert, disease-specific risk assessment in the context of athlete symptoms and diagnoses is emphasized throughout the document. After appropriate risk assessment, management of arrhythmias geared toward return to play when possible is addressed. Other topics include shared decision-making and emergency action planning. The goal of this document is to provide evidence-based recommendations impacting all areas in the care of athletes with arrhythmic conditions. Areas in need of further study are also discussed.
Collapse
Affiliation(s)
- Rachel Lampert
- Yale University School of Medicine, New Haven, Connecticut
| | - Eugene H Chung
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | | | | | | | - Rajat Deo
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Joe Dolan
- University of Utah, Salt Lake City, Utah
| | | | - Belinda R Gray
- University of Sydney, Camperdown, New South Wales, Australia
| | | | | | | | - Andrew D Krahn
- University of British Columbia, Vancouver, British Columbia, Canada
| | - Andre La Gerche
- Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Mark S Link
- UT Southwestern Medical Center, Dallas, Texas
| | | | - Lluis Mont
- Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Jack C Salerno
- University of Washington School of Medicine, Seattle, Washington
| | - Maully J Shah
- Childrens Hospital of Philadelphia, Philadelphia, Pennsylvania
| |
Collapse
|
9
|
Xu B, Xu Y, Ren W, Meng S, Hong T, Cao Z, Xiao X, Guo X, Yu L, Zhao J, Wang H. S-Methyl-L-cysteine targeting MsrA attenuates Ang II-induced oxidative stress and atrial remodeling via the p38 MAPK signaling pathway. Food Funct 2024; 15:9165-9175. [PMID: 39157962 DOI: 10.1039/d4fo03078h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
Atrial fibrillation (AF) is the most prevalent sustained tachyarrhythmia in patients with cardiovascular diseases. Recently, it has been discovered that oxidative stress is an important contributor to AF. Therefore, antioxidant therapies for AF have great potential for clinical applications. Methionine, a sulfur-containing amino acid residue other than cysteine, is recognized as a functional redox switch, which could be rescued from the reversible oxidation of methionine sulfoxide by methionine sulfoxide reductase A (MsrA). S-Methyl-L-cysteine (SMLC), a natural analogue of Met, which is abundantly found in garlic and cabbage, could substitute for Met oxidations and mediate MsrA to scavenge free radicals. However, whether SMLC alleviates AF is unclear. This study aims to clarify the effects of SMLC on AF and elucidate the underlying pharmacological and molecular mechanisms. In vivo, SMLC (70, 140 and 280 mg kg-1 day-1) was orally administered to mice for 4 weeks with angiotensin II (Ang II) by subcutaneous infusion using osmotic pumps to induce AF. Ang II significantly prompted high AF susceptibility and atrial remodeling characterized by oxidative stress, conductive dysfunction and fibrosis. SMLC played a remarkable protective role in Ang II-induced atrial remodeling dose-dependently. Moreover, RNA sequencing was performed on atrial tissues to identify the differentially expressed mRNA, which was to screen out MSRA, CAMK2 and MAPK signaling pathways. Western blots confirmed that Ang II-induced downregulation of MsrA and upregulation of oxidized CaMKII (ox-CaMKII) and p38 MAPK could be reversed in a concentration-dependent manner by SMLC. To investigate the underlying mechanisms, HL-1 cells (mouse atria-derived cardiomyocytes) treated with Ang II were used for an in vitro model. SMLC alleviated Ang II-induced cytotoxicity, mitochondrial damage and oxidative stress. Additionally, knockdown MsrA could attenuate the protective effects of SMLC, which were eliminated by the p38 MAPK inhibitor SB203580. In summary, the present study demonstrates that SMLC protects against atrial remodeling in AF by inhibiting oxidative stress through the mediation of the MsrA/p38 MAPK signaling pathway.
Collapse
Affiliation(s)
- Beibei Xu
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theatre Command, Shenyang 110016, China.
- Postgraduate College, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yinli Xu
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theatre Command, Shenyang 110016, China.
| | - Wenpu Ren
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theatre Command, Shenyang 110016, China.
- Postgraduate College, Liaoning University of Traditional Chinese Medicine, Shenyang 110847, China
| | - Shan Meng
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theatre Command, Shenyang 110016, China.
- Postgraduate College, Jinzhou Medical University, Jinzhou 121001, China
| | - Tao Hong
- Postgraduate College, Dalian Medical University, Dalian 116044, China
- Pediatric Surgery Ward, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen 518052, China
| | - Zijun Cao
- Postgraduate College, Liaoning University of Traditional Chinese Medicine, Shenyang 110847, China
| | - Xiong Xiao
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theatre Command, Shenyang 110016, China.
- Postgraduate College, China Medical University, Shenyang 110122, China
| | - Xiaodong Guo
- Postgraduate College, Dalian Medical University, Dalian 116044, China
| | - Liming Yu
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theatre Command, Shenyang 110016, China.
| | - Jikai Zhao
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theatre Command, Shenyang 110016, China.
| | - Huishan Wang
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theatre Command, Shenyang 110016, China.
| |
Collapse
|
10
|
Chen Y, Wijekoon S, Matsumoto A, Luo J, Kiriazis H, Masterman E, Yildiz G, Cross J, Parslow A, Chooi R, Sadoshima J, Greening D, Weeks K, McMullen J. Distinct functional and molecular profiles between physiological and pathological atrial enlargement offer potential new therapeutic opportunities for atrial fibrillation. Clin Sci (Lond) 2024; 138:941-962. [PMID: 39018488 PMCID: PMC11292366 DOI: 10.1042/cs20240178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 07/17/2024] [Accepted: 07/17/2024] [Indexed: 07/19/2024]
Abstract
Atrial fibrillation (AF) remains challenging to prevent and treat. A key feature of AF is atrial enlargement. However, not all atrial enlargement progresses to AF. Atrial enlargement in response to physiological stimuli such as exercise is typically benign and reversible. Understanding the differences in atrial function and molecular profile underpinning pathological and physiological atrial remodelling will be critical for identifying new strategies for AF. The discovery of molecular mechanisms responsible for pathological and physiological ventricular hypertrophy has uncovered new drug targets for heart failure. Studies in the atria have been limited in comparison. Here, we characterised mouse atria from (1) a pathological model (cardiomyocyte-specific transgenic (Tg) that develops dilated cardiomyopathy [DCM] and AF due to reduced protective signalling [PI3K]; DCM-dnPI3K), and (2) a physiological model (cardiomyocyte-specific Tg with an enlarged heart due to increased insulin-like growth factor 1 receptor; IGF1R). Both models presented with an increase in atrial mass, but displayed distinct functional, cellular, histological and molecular phenotypes. Atrial enlargement in the DCM-dnPI3K Tg, but not IGF1R Tg, was associated with atrial dysfunction, fibrosis and a heart failure gene expression pattern. Atrial proteomics identified protein networks related to cardiac contractility, sarcomere assembly, metabolism, mitochondria, and extracellular matrix which were differentially regulated in the models; many co-identified in atrial proteomics data sets from human AF. In summary, physiological and pathological atrial enlargement are associated with distinct features, and the proteomic dataset provides a resource to study potential new regulators of atrial biology and function, drug targets and biomarkers for AF.
Collapse
MESH Headings
- Atrial Fibrillation/physiopathology
- Atrial Fibrillation/metabolism
- Atrial Fibrillation/genetics
- Animals
- Heart Atria/metabolism
- Heart Atria/physiopathology
- Heart Atria/pathology
- Mice, Transgenic
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Atrial Remodeling
- Receptor, IGF Type 1/metabolism
- Receptor, IGF Type 1/genetics
- Cardiomyopathy, Dilated/physiopathology
- Cardiomyopathy, Dilated/genetics
- Cardiomyopathy, Dilated/metabolism
- Cardiomyopathy, Dilated/pathology
- Disease Models, Animal
- Fibrosis
- Mice
- Humans
- Signal Transduction
- Phosphatidylinositol 3-Kinases/metabolism
- Heart Failure/physiopathology
- Heart Failure/genetics
- Heart Failure/metabolism
- Heart Failure/pathology
Collapse
Affiliation(s)
- Yi Ching Chen
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Diabetes, Central Clinical School, Monash University, Clayton, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Melbourne, Victoria, Australia
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia
| | - Seka Wijekoon
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Aya Matsumoto
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Jieting Luo
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Helen Kiriazis
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Emma Masterman
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Gunes Yildiz
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Jonathon Cross
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Adam C. Parslow
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Melbourne, Victoria, Australia
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia
| | - Roger Chooi
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, NJ, U.S.A
| | - David W. Greening
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Melbourne, Victoria, Australia
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia
| | - Kate L. Weeks
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Diabetes, Central Clinical School, Monash University, Clayton, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Julie R. McMullen
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Diabetes, Central Clinical School, Monash University, Clayton, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Melbourne, Victoria, Australia
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia
- Monash Alfred Baker Centre for Cardiovascular Research, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
11
|
Nath LC, Saljic A, Buhl R, Elliott A, La Gerche A, Ye C, Schmidt Royal H, Lundgren Virklund K, Agbaedeng TA, Stent A, Franklin S. Histological evaluation of cardiac remodelling in equine athletes. Sci Rep 2024; 14:16709. [PMID: 39030282 PMCID: PMC11271503 DOI: 10.1038/s41598-024-67621-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 07/14/2024] [Indexed: 07/21/2024] Open
Abstract
Approximately 1-2 per 100,000 young athletes die from sudden cardiac death (SCD) and extreme exercise may be associated with myocardial scar and arrhythmias. Racehorses have a high prevalence of atrial fibrillation (AF) and SCD but the presence of myocardial scar and inflammation has not been evaluated. Cardiac tissues from the left (LAA) and right (RAA) atrial appendages, left ventricular anterior (LVAPM) and posterior (LVPPM) papillary muscles, and right side of the interventricular septum (IVS-R) were harvested from racehorses with sudden cardiac death (SCD, n = 16) or other fatal injuries (OFI, n = 17), constituting the athletic group (ATH, n = 33), and compared to sedentary horses (SED, n = 10). Horses in the ATH group had myocyte hypertrophy at all sites; increased fibrosis at all sites other than the LAA; increased fibroblast infiltration but a reduction in the overall extracellular matrix (ECM) volume in the RAA, LVAPM, and IVS-R compared to SED horses. In this horse model, athletic conditioning was associated with myocyte hypertrophy and a reduction in ECM. There was an excess of fibrocyte infiltration and focal fibrosis that was not present in non-athletic horses, raising the possibility of an exercise-induced pro-fibrotic substrate.
Collapse
Affiliation(s)
- L C Nath
- University of Adelaide, Adelaide, Australia.
| | - A Saljic
- University of Copenhagen, Copenhagen, Denmark
| | - R Buhl
- University of Copenhagen, Copenhagen, Denmark
| | - A Elliott
- University of Adelaide, Adelaide, Australia
| | - A La Gerche
- St Vincents Institute Medical Research, Fitzroy, Australia
| | - C Ye
- University of Copenhagen, Copenhagen, Denmark
| | | | | | | | - A Stent
- University of Melbourne, Parkville, Australia
| | - S Franklin
- University of Adelaide, Adelaide, Australia
| |
Collapse
|
12
|
Murphy MB, Yang Z, Subati T, Farber-Eger E, Kim K, Blackwell DJ, Fleming MR, Stark JM, Van Amburg JC, Woodall KK, Van Beusecum JP, Agrawal V, Smart CD, Pitzer A, Atkinson JB, Fogo AB, Bastarache JA, Kirabo A, Wells QS, Madhur MS, Barnett JV, Murray KT. LNK/SH2B3 loss of function increases susceptibility to murine and human atrial fibrillation. Cardiovasc Res 2024; 120:899-913. [PMID: 38377486 PMCID: PMC11218690 DOI: 10.1093/cvr/cvae036] [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: 04/28/2023] [Revised: 08/31/2023] [Accepted: 10/07/2023] [Indexed: 02/22/2024] Open
Abstract
AIMS The lymphocyte adaptor protein (LNK) is a negative regulator of cytokine and growth factor signalling. The rs3184504 variant in SH2B3 reduces LNK function and is linked to cardiovascular, inflammatory, and haematologic disorders, including stroke. In mice, deletion of Lnk causes inflammation and oxidative stress. We hypothesized that Lnk-/- mice are susceptible to atrial fibrillation (AF) and that rs3184504 is associated with AF and AF-related stroke in humans. During inflammation, reactive lipid dicarbonyls are the major components of oxidative injury, and we further hypothesized that these mediators are critical drivers of the AF substrate in Lnk-/- mice. METHODS AND RESULTS Lnk-/- or wild-type (WT) mice were treated with vehicle or 2-hydroxybenzylamine (2-HOBA), a dicarbonyl scavenger, for 3 months. Compared with WT, Lnk-/- mice displayed increased AF duration that was prevented by 2-HOBA. In the Lnk-/- atria, action potentials were prolonged with reduced transient outward K+ current, increased late Na+ current, and reduced peak Na+ current, pro-arrhythmic effects that were inhibited by 2-HOBA. Mitochondrial dysfunction, especially for Complex I, was evident in Lnk-/- atria, while scavenging lipid dicarbonyls prevented this abnormality. Tumour necrosis factor-α (TNF-α) and interleukin-1 beta (IL-1β) were elevated in Lnk-/- plasma and atrial tissue, respectively, both of which caused electrical and bioenergetic remodelling in vitro. Inhibition of soluble TNF-α prevented electrical remodelling and AF susceptibility, while IL-1β inhibition improved mitochondrial respiration but had no effect on AF susceptibility. In a large database of genotyped patients, rs3184504 was associated with AF, as well as AF-related stroke. CONCLUSION These findings identify a novel role for LNK in the pathophysiology of AF in both experimental mice and humans. Moreover, reactive lipid dicarbonyls are critical to the inflammatory AF substrate in Lnk-/- mice and mediate the pro-arrhythmic effects of pro-inflammatory cytokines, primarily through electrical remodelling.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Male
- Action Potentials/drug effects
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Atrial Fibrillation/metabolism
- Atrial Fibrillation/physiopathology
- Atrial Fibrillation/genetics
- Benzylamines/pharmacology
- Disease Models, Animal
- Genetic Predisposition to Disease
- Heart Rate/drug effects
- Inflammation Mediators/metabolism
- Interleukin-1beta/metabolism
- Interleukin-1beta/genetics
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Mitochondria, Heart/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/pathology
- Oxidative Stress/drug effects
- Phenotype
- Signal Transduction
- Tumor Necrosis Factor-alpha/metabolism
- Tumor Necrosis Factor-alpha/genetics
Collapse
Affiliation(s)
- Matthew B Murphy
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Zhenjiang Yang
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Tuerdi Subati
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Eric Farber-Eger
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
| | - Kyungsoo Kim
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Daniel J Blackwell
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Matthew R Fleming
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
| | - Joshua M Stark
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Joseph C Van Amburg
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Kaylen K Woodall
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Justin P Van Beusecum
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Vineet Agrawal
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
| | - Charles D Smart
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Ashley Pitzer
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - James B Atkinson
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, 1161 21 Avenue South, Nashville, TN 37232, USA
| | - Agnes B Fogo
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, 1161 21 Avenue South, Nashville, TN 37232, USA
| | - Julie A Bastarache
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Quinn S Wells
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, 2525 West End Avenue, Nashville, TN 37203, USA
| | - Meena S Madhur
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Joey V Barnett
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Katherine T Murray
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| |
Collapse
|
13
|
Jain H, Odat RM, Goyal A, Jain J, Dey D, Ahmed M, Wasir AS, Passey S, Gole S. Association between psoriasis and atrial fibrillation: A Systematic review and meta-analysis. Curr Probl Cardiol 2024; 49:102538. [PMID: 38521291 DOI: 10.1016/j.cpcardiol.2024.102538] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 03/25/2024]
Abstract
INTRODUCTION Psoriasis is a prevalent inflammatory skin condition characterized by erythematous plaques with scaling. Recent research has demonstrated an increased risk of cardiovascular diseases in patients with psoriasis; however, current evidence on atrial fibrillation (AF) risk in psoriasis is limited. MATERIALS AND METHODS A systematic literature search was performed on major bibliographic databases to retrieve studies that evaluated AF risk in patients with psoriasis. The DerSimonian and Laird random effects model was used to pool the hazard ratios (HR) with 95 % confidence intervals (CI). Subgroup analysis was conducted by dividing the patients into mild and severe psoriasis groups. Publication bias was assessed by visual inspection and Egger's regression test. Statistical significance was set at p < 0.05. RESULTS Seven studies were included, with 10,974,668 participants (1,94,230 in the psoriasis group and 10,780,439 in the control group). Patients with psoriasis had a significantly higher risk of AF [Pooled HR: 1.28; 95 % CI: 1.20, 1.36; p < 0.00001]. In subgroup analysis, patients with severe psoriasis [HR: 1.32; 95 % CI: 1.23, 1.42; p < 0.00001] demonstrated a slightly higher risk of AF, although statistically insignificant (p = 0.17), than the mild psoriasis group [HR: 1.21; 95 % CI: 1.10, 1.33; p < 0.0001]. Egger's regression test showed no statistically significant publication bias (p = 0.24). CONCLUSION Our analysis demonstrated that patients with psoriasis are at a significantly higher risk of AF and hence should be closely monitored for AF. Further large-scale and multicenter randomized trials are warranted to validate the robustness of our findings.
Collapse
Affiliation(s)
- Hritvik Jain
- Department of Internal Medicine, All India Institute of Medical Sciences (AIIMS), Jodhpur, India.
| | - Ramez M Odat
- Department of Internal Medicine, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Aman Goyal
- Department of Internal Medicine, Seth GS Medical College and KEM Hospital, Mumbai, India
| | - Jyoti Jain
- Department of Internal Medicine, All India Institute of Medical Sciences (AIIMS), Jodhpur, India
| | - Debankur Dey
- Department of Internal Medicine, Medical College Kolkata, Kolkata, West Bengal, India
| | - Mushood Ahmed
- Department of Internal Medicine, Rawalpindi Medical University, Rawalpindi, Pakistan
| | - Amanpreet Singh Wasir
- Department of Internal Medicine, Bharati Vidyapeeth (Deemed to be) University Medical College, Pune, Maharashtra, India
| | - Siddhant Passey
- Department of Internal Medicine, University of Connecticut Health Center, CT, USA
| | - Shrey Gole
- Department of Immunology and Rheumatology, Stanford University, CA, USA
| |
Collapse
|
14
|
Wu S, Yuan C, Chen Z, Gao Y, Guo X, Chen R, Dai Y, Chen K. Genetically predicted systemic inflammation and the risk of atrial fibrillation: A bidirectional two-sample Mendelian randomization study. IJC HEART & VASCULATURE 2024; 52:101422. [PMID: 38756452 PMCID: PMC11096748 DOI: 10.1016/j.ijcha.2024.101422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 05/02/2024] [Accepted: 05/03/2024] [Indexed: 05/18/2024]
Abstract
Background Systemic inflammation has been proposed to be associated with the incidence of atrial fibrillation (AF), but whether it is a cause or a consequence of AF remains uncertain. We sought to explore the causal associations between systemic inflammation and AF using bidirectional Mendelian randomization (MR) analysis. Methods Independent genetic variants strongly associated with AF were selected as instrumental variables from the largest genome-wide association study (GWAS) with up to 1,030,836 individuals. Regarding inflammation traits, genetic associations with 41 inflammatory cytokines and 5 inflammatory biomarkers were obtained from their corresponding GWASs databases. Effect estimates were primarily evaluated using the inverse-variance weighted (IVW) method, supplemented by sensitivity analyses using MR-Egger, weighted median, and MR-PRESSO methods. Results In our initial MR analyses, we observed suggestive associations of genetically predicted interleukin-17 (IL-17), interleukin-2 receptor subunit alpha (IL-2rα), and procalcitonin (PCT) with AF. One standard deviation (SD) increase in IL-17, IL-2rα, and PCT caused an increase in AF risk by 6.3 % (OR 1.063, 95 %CI 1.011---1.118, p = 0.018), 4.9 % (OR 1.049, 95 %CI 1.007---1.094, p = 0.023) and 3.4 % (OR 1.034, 95 %CI 1.005---1.064, p = 0.022), respectively. Furthermore, our reverse MR analyses indicated that genetically predicted AF contributed to a suggestive increase in the levels of macrophage inflammatory protein-1β (MIP1β) (β 0.055, 95 %CI 0.006 to 0.103, p = 0.028), while a decrease in the levels of fibrinogen (Fbg) (β -0.091, 95 %CI -0.140 to -0.041, p < 0.001), which remained significant after multiple test correction. Conclusions Our MR study identified several inflammatory biomarkers with suggestive causal associations regarding the upstream and downstream regulation of AF occurrence, offering new insights for therapeutic exploitation of AF. Further research is required to validate the underlying link between systemic inflammation and AF in larger cohorts.
Collapse
Affiliation(s)
- Sijin Wu
- Arrhythmia Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Chenxi Yuan
- Department of Epidemiology, Key Laboratory of Cardiovascular Epidemiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, China
| | - Zhongli Chen
- Arrhythmia Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yuan Gao
- Arrhythmia Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiaogang Guo
- Arrhythmia Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ruohan Chen
- Arrhythmia Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yan Dai
- Arrhythmia Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Keping Chen
- Arrhythmia Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
15
|
Ninni S, Algalarrondo V, Brette F, Lemesle G, Fauconnier J. Left atrial cardiomyopathy: Pathophysiological insights, assessment methods and clinical implications. Arch Cardiovasc Dis 2024; 117:283-296. [PMID: 38490844 DOI: 10.1016/j.acvd.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 02/01/2024] [Accepted: 02/06/2024] [Indexed: 03/17/2024]
Abstract
Atrial cardiomyopathy is defined as any complex of structural, architectural, contractile or electrophysiological changes affecting atria, with the potential to produce clinically relevant manifestations. Most of our knowledge about the mechanistic aspects of atrial cardiomyopathy is derived from studies investigating animal models of atrial fibrillation and atrial tissue samples obtained from individuals who have a history of atrial fibrillation. Several noninvasive tools have been reported to characterize atrial cardiomyopathy in patients, which may be relevant for predicting the risk of incident atrial fibrillation and its related outcomes, such as stroke. Here, we provide an overview of the pathophysiological mechanisms involved in atrial cardiomyopathy, and discuss the complex interplay of these mechanisms, including aging, left atrial pressure overload, metabolic disorders and genetic factors. We discuss clinical tools currently available to characterize atrial cardiomyopathy, including electrocardiograms, cardiac imaging and serum biomarkers. Finally, we discuss the clinical impact of atrial cardiomyopathy, and its potential role for predicting atrial fibrillation, stroke, heart failure and dementia. Overall, this review aims to highlight the critical need for a clinically relevant definition of atrial cardiomyopathy to improve treatment strategies.
Collapse
Affiliation(s)
- Sandro Ninni
- CHU de Lille, Université de Lille, 59000 Lille, France.
| | - Vincent Algalarrondo
- Department of Cardiology, Bichat University Hospital, AP-HP, 75018 Paris, France
| | - Fabien Brette
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34093 Montpellier, France
| | | | - Jérémy Fauconnier
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34093 Montpellier, France
| |
Collapse
|
16
|
Dorian D, Gustafson D, Quinn R, Bentley RF, Dorian P, Goodman JM, Fish JE, Connelly KA. Exercise-Dependent Modulation of Immunological Response Pathways in Endurance Athletes With and Without Atrial Fibrillation. J Am Heart Assoc 2024; 13:e033640. [PMID: 38497478 PMCID: PMC11009995 DOI: 10.1161/jaha.123.033640] [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: 11/20/2023] [Accepted: 01/12/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND Atrial fibrillation (AF) is a common arrhythmia characterized by uncoordinated atrial electrical activity. Lone AF occurs in the absence of traditional risk factors and is frequently observed in male endurance athletes, who face a 2- to 5-fold higher risk of AF compared with healthy, moderately active males. Our understanding of how endurance exercise contributes to the pathophysiology of lone AF remains limited. This study aimed to characterize the circulating protein fluctuations during high-intensity exercise as well as explore potential biomarkers of exercise-associated AF. METHODS AND RESULTS A prospective cohort of 12 male endurance cyclists between the ages of 40 and 65 years, 6 of whom had a history of exercise-associated AF, were recruited to participate using a convenience sampling method. The circulating proteome was subsequently analyzed using multiplex immunoassays and aptamer-based proteomics before, during, and after an acute high-intensity endurance exercise bout to assess temporality and identify potential markers of AF. The endurance exercise bout resulted in significant alterations to proteins involved in immune modulation (eg, growth/differentiation factor 15), skeletal muscle metabolism (eg, α-actinin-2), cell death (eg, histones), and inflammation (eg, interleukin-6). Subjects with AF differed from those without, displaying modulation of proteins previously known to have associations with incident AF (eg, C-reactive protein, insulin-like growth factor-1, and angiopoietin-2), and also with proteins having no previous association (eg, tapasin-related protein and α2-Heremans-Schmid glycoprotein). CONCLUSIONS These findings provide insights into the proteomic response to acute intense exercise, provide mechanistic insights into the pathophysiology behind AF in athletes, and identify targets for future study and validation.
Collapse
Affiliation(s)
- David Dorian
- Department of Medicine, Division of CardiologyUniversity of TorontoTorontoOntarioCanada
| | - Dakota Gustafson
- Department of Laboratory Medicine & PathobiologyUniversity of TorontoTorontoOntarioCanada
- Toronto General Hospital Research InstituteUniversity Health NetworkTorontoOntarioCanada
- Faculty of Health SciencesQueen’s UniversityKingstonOntarioCanada
| | - Ryan Quinn
- Division of CardiologyLi Ka Shing Knowledge Institute of St. Michael’s HospitalTorontoOntarioCanada
| | - Robert F. Bentley
- Faculty of Kinesiology and Physical EducationUniversity of TorontoTorontoOntarioCanada
| | - Paul Dorian
- Department of Medicine, Division of CardiologyUniversity of TorontoTorontoOntarioCanada
- Division of CardiologyLi Ka Shing Knowledge Institute of St. Michael’s HospitalTorontoOntarioCanada
- Department of MedicineUniversity of TorontoTorontoOntarioCanada
- Keenan Research Centre for Biomedical ScienceSt Michael’s Hospital, University of TorontoTorontoOntarioCanada
- Department of PhysiologyUniversity of TorontoTorontoOntarioCanada
- Heart and Stroke Richard Lewar Centre for Research ExcellenceUniversity of TorontoTorontoOntarioCanada
| | - Jack M. Goodman
- Faculty of Kinesiology and Physical EducationUniversity of TorontoTorontoOntarioCanada
- Heart and Stroke Richard Lewar Centre for Research ExcellenceUniversity of TorontoTorontoOntarioCanada
- Division of CardiologySinai Health/University Health NetworkTorontoOntarioCanada
| | - Jason E. Fish
- Department of Laboratory Medicine & PathobiologyUniversity of TorontoTorontoOntarioCanada
- Toronto General Hospital Research InstituteUniversity Health NetworkTorontoOntarioCanada
- Peter Munk Cardiac CentreUniversity Health NetworkTorontoOntarioCanada
| | - Kim A. Connelly
- Department of Medicine, Division of CardiologyUniversity of TorontoTorontoOntarioCanada
- Division of CardiologyLi Ka Shing Knowledge Institute of St. Michael’s HospitalTorontoOntarioCanada
- Department of MedicineUniversity of TorontoTorontoOntarioCanada
- Keenan Research Centre for Biomedical ScienceSt Michael’s Hospital, University of TorontoTorontoOntarioCanada
- Department of PhysiologyUniversity of TorontoTorontoOntarioCanada
- Heart and Stroke Richard Lewar Centre for Research ExcellenceUniversity of TorontoTorontoOntarioCanada
| |
Collapse
|
17
|
Lei M, Salvage SC, Jackson AP, Huang CLH. Cardiac arrhythmogenesis: roles of ion channels and their functional modification. Front Physiol 2024; 15:1342761. [PMID: 38505707 PMCID: PMC10949183 DOI: 10.3389/fphys.2024.1342761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 01/22/2024] [Indexed: 03/21/2024] Open
Abstract
Cardiac arrhythmias cause significant morbidity and mortality and pose a major public health problem. They arise from disruptions in the normally orderly propagation of cardiac electrophysiological activation and recovery through successive cardiomyocytes in the heart. They reflect abnormalities in automaticity, initiation, conduction, or recovery in cardiomyocyte excitation. The latter properties are dependent on surface membrane electrophysiological mechanisms underlying the cardiac action potential. Their disruption results from spatial or temporal instabilities and heterogeneities in the generation and propagation of cellular excitation. These arise from abnormal function in their underlying surface membrane, ion channels, and transporters, as well as the interactions between them. The latter, in turn, form common regulatory targets for the hierarchical network of diverse signaling mechanisms reviewed here. In addition to direct molecular-level pharmacological or physiological actions on these surface membrane biomolecules, accessory, adhesion, signal transduction, and cytoskeletal anchoring proteins modify both their properties and localization. At the cellular level of excitation-contraction coupling processes, Ca2+ homeostatic and phosphorylation processes affect channel activity and membrane excitability directly or through intermediate signaling. Systems-level autonomic cellular signaling exerts both acute channel and longer-term actions on channel expression. Further upstream intermediaries from metabolic changes modulate the channels both themselves and through modifying Ca2+ homeostasis. Finally, longer-term organ-level inflammatory and structural changes, such as fibrotic and hypertrophic remodeling, similarly can influence all these physiological processes with potential pro-arrhythmic consequences. These normal physiological processes may target either individual or groups of ionic channel species and alter with particular pathological conditions. They are also potentially alterable by direct pharmacological action, or effects on longer-term targets modifying protein or cofactor structure, expression, or localization. Their participating specific biomolecules, often clarified in experimental genetically modified models, thus constitute potential therapeutic targets. The insights clarified by the physiological and pharmacological framework outlined here provide a basis for a recent modernized drug classification. Together, they offer a translational framework for current drug understanding. This would facilitate future mechanistically directed therapeutic advances, for which a number of examples are considered here. The latter are potentially useful for treating cardiac, in particular arrhythmic, disease.
Collapse
Affiliation(s)
- Ming Lei
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Samantha C. Salvage
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Antony P. Jackson
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Christopher L.-H. Huang
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
- Physiological Laboratory, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
18
|
Ye L, Yuan J, Zhu S, Ji S, Dai J. Swimming exercise reverses transcriptomic changes in aging mouse lens. BMC Med Genomics 2024; 17:67. [PMID: 38439070 PMCID: PMC10913554 DOI: 10.1186/s12920-024-01839-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 02/28/2024] [Indexed: 03/06/2024] Open
Abstract
BACKGROUND The benefits of physical activity for the overall well-being of elderly individuals are well-established, the precise mechanisms through which exercise improves pathological changes in the aging lens have yet to be fully understood. METHODS 3-month-old C57BL/6J mice comprised young sedentary (YS) group, while aging mice (18-month-old) were divided into aging sedentary (AS) group and aging exercising (AE) group. Mice in AE groups underwent sequential stages of swimming exercise. H&E staining was employed to observe alterations in lens morphology. RNA-seq analysis was utilized to examine transcriptomic changes. Furthermore, qPCR and immunohistochemistry were employed for validation of the results. RESULTS AE group showed alleviation of histopathological aging changes in AS group. By GSEA analysis of the transcriptomic changes, swimming exercise significantly downregulated approximately half of the pathways that underwent alterations upon aging, where notable improvements were 'calcium signaling pathway', 'neuroactive ligand receptor interaction' and 'cell adhesion molecules'. Furthermore, we revealed a total of 92 differentially expressed genes between the YS and AS groups, of which 10 genes were observed to be mitigated by swimming exercise. The result of qPCR was in consistent with the transcriptome data. We conducted immunohistochemical analysis on Ciart, which was of particular interest due to its dual association as a common aging gene and its significant responsiveness to exercise. The Protein-protein Interaction network of Ciart showed the involvement of the regulation of Rorb and Sptbn5 during the process. CONCLUSION The known benefits of exercise could extend to the aging lens and support further investigation into the specific roles of Ciart-related pathways in aging lens.
Collapse
Affiliation(s)
- Lin Ye
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Jiayue Yuan
- Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shijie Zhu
- School of Medicine, Tongji University, Shanghai, China
| | - Shunmei Ji
- Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jinhui Dai
- Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
19
|
Huang M, Huiskes FG, de Groot NMS, Brundel BJJM. The Role of Immune Cells Driving Electropathology and Atrial Fibrillation. Cells 2024; 13:311. [PMID: 38391924 PMCID: PMC10886649 DOI: 10.3390/cells13040311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/02/2024] [Accepted: 02/04/2024] [Indexed: 02/24/2024] Open
Abstract
Atrial fibrillation (AF) is the most common progressive cardiac arrhythmia worldwide and entails serious complications including stroke and heart failure. Despite decades of clinical research, the current treatment of AF is suboptimal. This is due to a lack of knowledge on the mechanistic root causes of AF. Prevailing theories indicate a key role for molecular and structural changes in driving electrical conduction abnormalities in the atria and as such triggering AF. Emerging evidence indicates the role of the altered atrial and systemic immune landscape in driving this so-called electropathology. Immune cells and immune markers play a central role in immune remodeling by exhibiting dual facets. While the activation and recruitment of immune cells contribute to maintaining atrial stability, the excessive activation and pronounced expression of immune markers can foster AF. This review delineates shifts in cardiac composition and the distribution of immune cells in the context of cardiac health and disease, especially AF. A comprehensive exploration of the functions of diverse immune cell types in AF and other cardiac diseases is essential to unravel the intricacies of immune remodeling. Usltimately, we delve into clinical evidence showcasing immune modifications in both the atrial and systemic domains among AF patients, aiming to elucidate immune markers for therapy and diagnostics.
Collapse
Affiliation(s)
- Mingxin Huang
- Department of Physiology, Amsterdam UMC, Location Vrije Universiteit, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, 1081 HZ Amsterdam, The Netherlands; (M.H.); (F.G.H.)
- Department of Cardiology, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands;
| | - Fabries G. Huiskes
- Department of Physiology, Amsterdam UMC, Location Vrije Universiteit, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, 1081 HZ Amsterdam, The Netherlands; (M.H.); (F.G.H.)
| | | | - Bianca J. J. M. Brundel
- Department of Physiology, Amsterdam UMC, Location Vrije Universiteit, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, 1081 HZ Amsterdam, The Netherlands; (M.H.); (F.G.H.)
| |
Collapse
|
20
|
Chen C, Wei L, He W, Zhang Y, Xiao J, Lu Y, Wang F, Zhu X. Associations of severe liver diseases with cataract using data from UK Biobank: a prospective cohort study. EClinicalMedicine 2024; 68:102424. [PMID: 38304745 PMCID: PMC10831806 DOI: 10.1016/j.eclinm.2024.102424] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/23/2023] [Accepted: 01/03/2024] [Indexed: 02/03/2024] Open
Abstract
Background Liver disease is linked to series of extrahepatic multisystem manifestations. However, little is known about the associations between liver and eye diseases, especially cataract, the global leading cause of blindness. We aimed to investigate whether severe liver diseases, including non-alcoholic fatty liver disease (NAFLD), alcoholic liver disease (ALD), viral hepatitis, and liver fibrosis and cirrhosis, were associated with an increased risk of the cataract. Methods A total of 326,558 participants without cataract at baseline enrolled in the UK Biobank between 2006 and 2010 were included in this prospective study. The exposures of interest were severe liver diseases (defined as hospital admission), including NAFLD, ALD, viral hepatitis and liver fibrosis and cirrhosis. The outcome was incident cataract. Cox proportional hazards models were used to estimate the hazard ratios (HRs) and 95% confidence intervals (CIs). Each liver disease was first treated as a binary time-varying variable to investigate its association with cataract, and then was treated as a ternary time-varying variable to examine the recent (liver disease within 0-5 years) vs. long-term (liver disease > 5 years) state associations with the risk of cataract. Findings After a median follow-up of 13.3 years (interquartile range, 12.5-14.0 years), 37,064 individuals were documented as developing cataract. Higher risk of cataract was found in those with severe NAFLD (HR, 1.47; 95% CI, 1.33-1.61), ALD (HR, 1.57; 95% CI, 1.28-1.94) and liver fibrosis and cirrhosis (HR, 1.58; 95% CI, 1.35-1.85), but not in individuals with viral hepatitis when exposure was treated as a binary time-varying variable (P = 0.13). When treating exposure as a ternary time-varying variable, an association between recently diagnosed viral hepatitis and cataract was also observed (HR, 1.55; 95% CI, 1.07-2.23). Results from the combined model suggested they were independent risk factors for incident cataract. No substantial changes were found in further sensitivity analyses. Interpretation Severe liver diseases, including NAFLD, ALD, liver fibrosis and cirrhosis and recently diagnosed viral hepatitis, were associated with cataract. The revelation of liver-eye connection suggests the importance of ophthalmic care in the management of liver disease, and the intervention precedence of patients with liver disease in the early screening and diagnosis of cataract. Funding National Natural Science Foundation of China, Science and Technology Innovation Action Plan of Shanghai Science and Technology Commission, Clinical Research Plan of Shanghai Shenkang Hospital Development Center, Shanghai Municipal Key Clinical Specialty Program, the Guangdong Basic and Applied Basic Research Foundation and Shenzhen Science and Technology Program.
Collapse
Affiliation(s)
- Chao Chen
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
- NHC Key Laboratory of Myopia, Fudan University, Shanghai 200031, China
- Key Laboratory of Myopia, Chinese Academy of Medical Science, Shanghai 200031, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China
| | - Ling Wei
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
- NHC Key Laboratory of Myopia, Fudan University, Shanghai 200031, China
- Key Laboratory of Myopia, Chinese Academy of Medical Science, Shanghai 200031, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China
| | - Wenwen He
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
- NHC Key Laboratory of Myopia, Fudan University, Shanghai 200031, China
- Key Laboratory of Myopia, Chinese Academy of Medical Science, Shanghai 200031, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China
| | - Ye Zhang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
- NHC Key Laboratory of Myopia, Fudan University, Shanghai 200031, China
- Key Laboratory of Myopia, Chinese Academy of Medical Science, Shanghai 200031, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China
| | - Jia Xiao
- Changsha Aier Eye Hospital, Changsha, Hunan Province 410015, China
- Aier Eye Institute, Changsha, Hunan Province 410015, China
- Shandong Provincial Key Laboratory for Clinical Research of Liver Diseases, Qingdao Hospital, University of Health and Rehabilitation Sciences, Qingdao 266001, China
| | - Yi Lu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
- NHC Key Laboratory of Myopia, Fudan University, Shanghai 200031, China
- Key Laboratory of Myopia, Chinese Academy of Medical Science, Shanghai 200031, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China
| | - Fei Wang
- Division of Gastroenterology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
| | - Xiangjia Zhu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
- NHC Key Laboratory of Myopia, Fudan University, Shanghai 200031, China
- Key Laboratory of Myopia, Chinese Academy of Medical Science, Shanghai 200031, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China
| |
Collapse
|
21
|
Gorman RA, Yakobov S, Polidovitch N, Debi R, Sanfrancesco VC, Hood DA, Lakin R, Backx PH. The effects of daily dose of intense exercise on cardiac responses and atrial fibrillation. J Physiol 2024; 602:569-596. [PMID: 38319954 DOI: 10.1113/jp285697] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 12/08/2023] [Indexed: 02/08/2024] Open
Abstract
Atrial fibrillation (AF) is a supraventricular tachyarrhythmia that is strongly associated with cardiovascular (CV) disease and sedentary lifestyles. Despite the benefits of exercise on overall health, AF incidence in high-level endurance athletes rivals that of CV disease patients, suggesting a J-shaped relationship with AF. To investigate the dependence of AF vulnerability on exercise, we varied daily swim durations (120, 180 or 240 min day-1 ) in 7-week-old male CD1 mice. We assessed mice after performing equivalent amounts of cumulative work during swimming (i.e. ∼700 L O2 kg-1 ), as determined from O2 consumption rates (V ̇ O 2 ${\dot V_{{{\mathrm{O}}_2}}}$ ). The meanV ̇ O 2 ${\dot V_{{{\mathrm{O}}_2}}}$ during exercise increased progressively throughout the training period and was indistinguishable between the swim groups. Consistent with similar improvements in aerobic conditioning induced by swimming, skeletal muscle mitochondria content increased (P = 0.027) indistinguishably between exercise groups. Physiological ventricular remodelling, characterized by mild hypertrophy and left ventricular dilatation, was also similar between exercised mice without evidence of ventricular arrhythmia inducibility. By contrast, prolongation of daily swim durations caused progressive and vagal-dependent heart rate reductions (P = 0.008), as well as increased (P = 0.005) AF vulnerability. As expected, vagal inhibition prolonged (P = 0.013) atrial refractoriness, leading to reduced AF vulnerability, although still inducible in the 180 and 240 min swim groups. Accordingly, daily swim dose progressively increased atrial hypertrophy (P = 0.003), fibrosis (P < 0.001) and macrophage accumulation (P = 0.006) without differentially affecting the ventricular tissue properties. Thus, increasing daily exercise duration drives progressively adverse atrial-specific remodelling and vagal-dependent AF vulnerability despite robust and beneficial aerobic conditioning and physiological remodelling of ventricles and skeletal muscle. KEY POINTS: Previous studies have suggested that a J-shaped dose-response relationship exists between physical activity and cardiovascular health outcomes, with moderate exercise providing protection against many cardiovascular disease conditions, whereas chronic endurance exercise can promote atrial fibrillation (AF). We found that AF vulnerability increased alongside elevated atrial hypertrophy, fibrosis and inflammation as daily swim exercise durations in mice were prolonged (i.e. ≥180 min day-1 for 6 weeks). The MET-h week-1 (based on O2 measurements during swimming) needed to induce increased AF vulnerability mirrored the levels linked to AF in athletes. These adverse atria effects associated with excessive daily exercise occurred despite improved aerobic conditioning, skeletal muscle adaptation and physiological ventricular remodelling. We suggest that atrial-specific changes observed with exercise arise from excessive elevations in venous filling pressures during prolonged exercise bouts, which we argue has implications for all AF patients because elevated atrial pressures occur in most cardiovascular disease conditions as well as ageing which are linked to AF.
Collapse
Affiliation(s)
- Renée A Gorman
- Department of Biology, York University, Toronto, ON, Canada
- Muscle Health Research Centre, York University, Toronto, ON, Canada
| | - Simona Yakobov
- Department of Biology, York University, Toronto, ON, Canada
- Muscle Health Research Centre, York University, Toronto, ON, Canada
| | | | - Ryan Debi
- Department of Biology, York University, Toronto, ON, Canada
| | - Victoria C Sanfrancesco
- Muscle Health Research Centre, York University, Toronto, ON, Canada
- Department of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - David A Hood
- Muscle Health Research Centre, York University, Toronto, ON, Canada
- Department of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - Robert Lakin
- Department of Biology, York University, Toronto, ON, Canada
- Muscle Health Research Centre, York University, Toronto, ON, Canada
| | - Peter H Backx
- Department of Biology, York University, Toronto, ON, Canada
- Muscle Health Research Centre, York University, Toronto, ON, Canada
| |
Collapse
|
22
|
Kordi N, Sanaei M, Akraminia P, Yavari S, Saydi A, Abadi FK, Heydari N, Jung F, Karami S. PANoptosis and cardiovascular disease: The preventive role of exercise training. Clin Hemorheol Microcirc 2024; 88:499-512. [PMID: 39269827 DOI: 10.3233/ch-242396] [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] [Indexed: 09/15/2024]
Abstract
Regulated cell death, including pyroptosis, apoptosis, and necroptosis, is vital for the body's defense system. Recent research suggests that these three types of cell death are interconnected, giving rise to a new concept called PANoptosis. PANoptosis has been linked to various diseases, making it crucial to comprehend its mechanism for effective treatments. PANoptosis is controlled by upstream receptors and molecular signals, which form polymeric complexes known as PANoptosomes. Cell death combines necroptosis, apoptosis, and pyroptosis and cannot be fully explained by any of these processes alone. Understanding pyroptosis, apoptosis, and necroptosis is essential for understanding PANoptosis. Physical exercise has been shown to suppress pyroptotic, apoptotic, and necroptotic signaling pathways by reducing inflammatory factors, proapoptotic factors, and necroptotic factors such as caspases and TNF-alpha. This ultimately leads to a decrease in cardiac structural remodeling. The beneficial effects of exercise on cardiovascular health may be attributed to its ability to inhibit these cell death pathways.
Collapse
Affiliation(s)
- Negin Kordi
- Department of Exercise Physiology, Faculty of Sport Sciences, Razi University, Kermanshah, Iran
| | | | - Peyman Akraminia
- Department of Sports Physiology, Faculty of Physical Education and Sports Sciences, Islamic Azad University, South Tehran Branch, Iran
| | - Sajad Yavari
- Department of Exercise Physiology, Faculty of Sport Sciences, Razi University, Kermanshah, Iran
| | - Ali Saydi
- Department of Exercise Physiology, Faculty of Sport Sciences, Razi University, Kermanshah, Iran
| | - Fatemeh Khamis Abadi
- Department of Sport Physiology, Faculty of Human Sciences, Islamic Azad University, Borujerd, Iran
| | - Naser Heydari
- Faculty of Physical Education and Sport Sciences, Shahid Rajaee Teacher Training University, Tehran, Iran
| | - Friedrich Jung
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | - Sajad Karami
- Faculty of Physical Education and Sport Sciences, Shahid Rajaee Teacher Training University, Tehran, Iran
| |
Collapse
|
23
|
Lakin R, Polidovitch N, Yang S, Parikh M, Liu X, Debi R, Gao X, Chen W, Guzman C, Yakobov S, Izaddoustdar F, Wauchop M, Lei Q, Xu W, Nedospasov SA, Christoffels VM, Backx PH. Cardiomyocyte and endothelial cells play distinct roles in the tumour necrosis factor (TNF)-dependent atrial responses and increased atrial fibrillation vulnerability induced by endurance exercise training in mice. Cardiovasc Res 2023; 119:2607-2622. [PMID: 37713664 PMCID: PMC10730243 DOI: 10.1093/cvr/cvad144] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 06/22/2023] [Accepted: 07/18/2023] [Indexed: 09/17/2023] Open
Abstract
AIMS Endurance exercise is associated with an increased risk of atrial fibrillation (AF). We previously established that adverse atrial remodelling and AF susceptibility induced by intense exercise in mice require the mechanosensitive and pro-inflammatory cytokine tumour necrosis factor (TNF). The cellular and mechanistic basis for these TNF-mediated effects is unknown. METHODS AND RESULTS We studied the impact of Tnf excision, in either atrial cardiomyocytes or endothelial cells (using Cre-recombinase expression controlled by Nppa or Tie2 promoters, respectively), on the cardiac responses to six weeks of intense swim exercise training. TNF ablation, in either cell type, had no impact on the changes in heart rate, autonomic tone, or left ventricular structure and function induced by exercise training. Tnf excision in atrial cardiomyocytes did, however, prevent atrial hypertrophy, fibrosis, and macrophage infiltration as well as conduction slowing and increased AF susceptibility arising from exercise training. In contrast, endothelial-specific excision only reduced the training-induced atrial hypertrophy. Consistent with these cell-specific effects of Tnf excision, inducing TNF loss from atrial cardiomyocytes prevented activation of p38MAPKinase, a strain-dependent downstream mediator of TNF signalling, without affecting the atrial stretch as assessed by atrial pressures induced by exercise. Despite TNF's established role in innate immune responses and inflammation, neither acute nor chronic exercise training caused measurable NLRP3 inflammasome activation. CONCLUSIONS Our findings demonstrate that adverse atrial remodelling and AF vulnerability induced by intense exercise require TNF in atrial cardiomyocytes whereas the impact of endothelial-derived TNF is limited to hypertrophy modulation. The implications of the cell autonomous effects of TNF and crosstalk between cells in the atria are discussed.
Collapse
Affiliation(s)
- Robert Lakin
- Department of Biology, York University, 354 & 357 Farquharson Building, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
| | - Nazari Polidovitch
- Department of Biology, York University, 354 & 357 Farquharson Building, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
| | - Sibao Yang
- Department of Biology, York University, 354 & 357 Farquharson Building, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130022, China
| | - Mihir Parikh
- Department of Biology, York University, 354 & 357 Farquharson Building, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
| | - Xueyan Liu
- Department of Biology, York University, 354 & 357 Farquharson Building, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130022, China
| | - Ryan Debi
- Department of Biology, York University, 354 & 357 Farquharson Building, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
| | - Xiaodong Gao
- Department of Biology, York University, 354 & 357 Farquharson Building, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
| | - Wenliang Chen
- Department of Biology, York University, 354 & 357 Farquharson Building, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
| | - Camilo Guzman
- Department of Biology, York University, 354 & 357 Farquharson Building, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
| | - Simona Yakobov
- Department of Biology, York University, 354 & 357 Farquharson Building, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
| | - Farzad Izaddoustdar
- Department of Physiology, University of Toronto, Toronto, ON M5S 3E2, Canada
| | - Marianne Wauchop
- Department of Physiology, University of Toronto, Toronto, ON M5S 3E2, Canada
| | - Qian Lei
- Department of Anesthesiology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Weimin Xu
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130022, China
| | - Sergei A Nedospasov
- Laboratory of Molecular Mechanisms of Immunity, Engelhardt Institute of Molecular Biology, Moscow 119991, Russia
- Division of Immunobiology and Biomedicine, Sirius University of Science and Technology, Sirius 354349, Russia
| | - Vincent M Christoffels
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Amsterdam, The Netherlands
| | - Peter H Backx
- Department of Biology, York University, 354 & 357 Farquharson Building, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
| |
Collapse
|
24
|
Jiang J, Ni L, Zhang X, Chatterjee E, Lehmann HI, Li G, Xiao J. Keeping the Heart Healthy: The Role of Exercise in Cardiac Repair and Regeneration. Antioxid Redox Signal 2023; 39:1088-1107. [PMID: 37132606 DOI: 10.1089/ars.2023.0301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Significance: Heart failure is often accompanied by a decrease in the number of cardiomyocytes. Although the adult mammalian hearts have limited regenerative capacity, the rate of regeneration is extremely low and decreases with age. Exercise is an effective means to improve cardiovascular function and prevent cardiovascular diseases. However, the molecular mechanisms of how exercise acts on cardiomyocytes are still not fully elucidated. Therefore, it is important to explore the role of exercise in cardiomyocytes and cardiac regeneration. Recent Advances: Recent advances have shown that the effects of exercise on cardiomyocytes are critical for cardiac repair and regeneration. Exercise can induce cardiomyocyte growth by increasing the size and number. It can induce physiological cardiomyocyte hypertrophy, inhibit cardiomyocyte apoptosis, and promote cardiomyocyte proliferation. In this review, we have discussed the molecular mechanisms and recent studies of exercise-induced cardiac regeneration, with a focus on its effects on cardiomyocytes. Critical Issues: There is no effective way to promote cardiac regeneration. Moderate exercise can keep the heart healthy by encouraging adult cardiomyocytes to survive and regenerate. Therefore, exercise could be a promising tool for stimulating the regenerative capability of the heart and keeping the heart healthy. Future Directions: Although exercise is an important measure to promote cardiomyocyte growth and subsequent cardiac regeneration, more studies are needed on how to do beneficial exercise and what factors are involved in cardiac repair and regeneration. Thus, it is important to clarify the mechanisms, pathways, and other critical factors involved in the exercise-mediated cardiac repair and regeneration. Antioxid. Redox Signal. 39, 1088-1107.
Collapse
Affiliation(s)
- Jizong Jiang
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, China
| | - Lingyan Ni
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, China
| | - Xinxin Zhang
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, China
| | - Emeli Chatterjee
- Cardiovascular Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - H Immo Lehmann
- Cardiovascular Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Guoping Li
- Cardiovascular Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, China
| |
Collapse
|
25
|
Wu K, Zhu J, Ma Y, Zhou Y, Lin Q, Tu T, Liu Q. Exploring immune related gene signatures and mechanisms linking non alcoholic fatty liver disease to atrial fibrillation through transcriptome data analysis. Sci Rep 2023; 13:17548. [PMID: 37845390 PMCID: PMC10579333 DOI: 10.1038/s41598-023-44884-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 10/12/2023] [Indexed: 10/18/2023] Open
Abstract
Atrial fibrillation (AF) and related cardiovascular complications pose a heavy burden to patients and society. Mounting evidence suggests a close association between nonalcoholic fatty liver disease (NAFLD) and AF. NAFLD and AF transcriptomic datasets were obtained from GEO database and analyzed using several bioinformatics approaches. We established a NAFLD-AF associated gene diagnostic signature (NAGDS) using protein-protein interaction analysis and machine learning, which was further quantified through RT-qPCR. Potential miRNA targeting NAGDS were predicted. Gene modules highly correlated with NAFLD liver pathology or AF occurrence were identified by WGCNA. Enrichment analysis of the overlapped genes from key module revealed that T-cell activation plays essential roles in NAFLD and AF, which was further confirmed by immune infiltration. Furthermore, an integrated SVM-RFE and LASSO algorithm was used to identify CCL4, CD48, ITGB2, and RNASE6 as NAGDS, all of which were found to be upregulated in NAFLD and AF mouse tissues. Patients with higher NAGDS showed augmented T cell and macrophage immunity, more advanced liver pathological characteristics, and prolonged AF duration. Additionally, hsa-miR-26a-5p played a central role in the regulation of NAGDS. Our findings highlight the central role of T-cell immune response in linking NAFLD to AF, and established an accurate NAGDS diagnostic model, which could serve as potential targets for immunoregulatory therapy.
Collapse
Affiliation(s)
- Keke Wu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Road, Changsha, 410011, Hunan, People's Republic of China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, People's Republic of China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, People's Republic of China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, 410011, Hunan, People's Republic of China
| | - Jiayi Zhu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Road, Changsha, 410011, Hunan, People's Republic of China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, People's Republic of China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, People's Republic of China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, 410011, Hunan, People's Republic of China
| | - Yingxu Ma
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Road, Changsha, 410011, Hunan, People's Republic of China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, People's Republic of China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, People's Republic of China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, 410011, Hunan, People's Republic of China
| | - Yong Zhou
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Road, Changsha, 410011, Hunan, People's Republic of China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, People's Republic of China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, People's Republic of China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, 410011, Hunan, People's Republic of China
| | - Qiuzhen Lin
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Road, Changsha, 410011, Hunan, People's Republic of China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, People's Republic of China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, People's Republic of China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, 410011, Hunan, People's Republic of China
| | - Tao Tu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Road, Changsha, 410011, Hunan, People's Republic of China.
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, People's Republic of China.
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, People's Republic of China.
- Cardiovascular Disease Research Center of Hunan Province, Changsha, 410011, Hunan, People's Republic of China.
| | - Qiming Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Road, Changsha, 410011, Hunan, People's Republic of China.
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, People's Republic of China.
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, People's Republic of China.
- Cardiovascular Disease Research Center of Hunan Province, Changsha, 410011, Hunan, People's Republic of China.
| |
Collapse
|
26
|
Zhang MJ, Gyberg DJ, Healy CL, Zhang N, Liu H, Dudley SC, O’Connell TD. Atrial Myopathy Quantified by Speckle-tracking Echocardiography in Mice. Circ Cardiovasc Imaging 2023; 16:e015735. [PMID: 37795649 PMCID: PMC10591948 DOI: 10.1161/circimaging.123.015735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 08/17/2023] [Indexed: 10/06/2023]
Abstract
BACKGROUND Emerging evidence suggests that atrial myopathy may be the underlying pathophysiology that explains adverse cardiovascular outcomes in heart failure (HF) and atrial fibrillation. Lower left atrial (LA) function (strain) is a key biomarker of atrial myopathy, but murine LA strain has not been described, thus limiting translational investigation. Therefore, the objective of this study was to characterize LA function by speckle-tracking echocardiography in mouse models of atrial myopathy. METHODS We used 3 models of atrial myopathy in wild-type male and female C57Bl6/J mice: (1) aged 16 to 17 months, (2) Ang II (angiotensin II) infusion, and (3) high-fat diet+Nω-nitro-L-arginine methyl ester (HF with preserved ejection fraction, HFpEF). LA reservoir, conduit, and contractile strain were measured using speckle-tracking echocardiography from a modified parasternal long-axis window. Left ventricular systolic and diastolic function, and global longitudinal strain were also measured. Transesophageal rapid atrial pacing was used to induce atrial fibrillation. RESULTS LA reservoir, conduit, and contractile strain were significantly reduced in aged, Ang II and HFpEF mice compared with young controls. There were no sex-based interactions. Left ventricular diastolic function and global longitudinal strain were lower in aged, Ang II and HFpEF, but left ventricular ejection fraction was unchanged. Atrial fibrillation inducibility was low in young mice (5%), moderately higher in aged mice (20%), and high in Ang II (75%) and HFpEF (83%) mice. CONCLUSIONS Using speckle-tracking echocardiography, we observed reduced LA function in established mouse models of atrial myopathy with concurrent atrial fibrillation inducibility, thus providing the field with a timely and clinically relevant platform for understanding the pathophysiology and discovery of novel treatment targets for atrial myopathy.
Collapse
Affiliation(s)
- Michael J. Zhang
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN
- Department of Medicine, Cardiovascular Division, University of Minnesota, Minneapolis, MN
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN
| | - Dylan J. Gyberg
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN
| | - Chastity L. Healy
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN
| | - Naixin Zhang
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN
| | - Hong Liu
- Department of Medicine, Cardiovascular Division, University of Minnesota, Minneapolis, MN
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN
| | - Samuel C. Dudley
- Department of Medicine, Cardiovascular Division, University of Minnesota, Minneapolis, MN
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN
| | - Timothy D. O’Connell
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN
| |
Collapse
|
27
|
Elliott AD, Ariyaratnam J, Howden EJ, La Gerche A, Sanders P. Influence of exercise training on the left atrium: implications for atrial fibrillation, heart failure, and stroke. Am J Physiol Heart Circ Physiol 2023; 325:H822-H836. [PMID: 37505470 DOI: 10.1152/ajpheart.00322.2023] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/24/2023] [Accepted: 07/24/2023] [Indexed: 07/29/2023]
Abstract
The left atrium (LA) plays a critical role in receiving pulmonary venous return and modulating left ventricular (LV) filling. With the onset of exercise, LA function contributes to the augmentation in stroke volume. Due to the growing focus on atrial imaging, there is now evidence that structural remodeling and dysfunction of the LA is associated with adverse outcomes including incident cardiovascular disease. In patients with established disease, pathological changes in atrial structure and function are associated with exercise intolerance, increased hospital admissions and mortality, independent of left ventricular function. Exercise training is widely recommended in patients with cardiovascular disease to improve patient outcomes and maintain functional capacity. There are widely documented changes in LV function with exercise, yet less attention has been given to the LA. In this review, we first describe LA physiology at rest and during exercise, before exploring its association with cardiac disease outcomes including atrial fibrillation, heart failure, and stroke. The adaptation of the LA to short- and longer-term exercise training is evaluated through review of longitudinal studies of exercise training in healthy participants free of cardiovascular disease and athletes. We then consider the changes in LA structure and function among patients with established disease, where adverse atrial remodeling may be implicated in the disease process. Finally, we consider important future directions for assessment of atrial structure and function using novel imaging modalities, in response to acute and chronic exercise.
Collapse
Affiliation(s)
- Adrian D Elliott
- Center for Heart Rhythm Disorders, University of Adelaide, South Australian Health and Medical Research Institute and Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Jonathan Ariyaratnam
- Center for Heart Rhythm Disorders, University of Adelaide, South Australian Health and Medical Research Institute and Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Erin J Howden
- Clinical Research Domain, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Andre La Gerche
- Clinical Research Domain, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Prashanthan Sanders
- Center for Heart Rhythm Disorders, University of Adelaide, South Australian Health and Medical Research Institute and Royal Adelaide Hospital, Adelaide, South Australia, Australia
| |
Collapse
|
28
|
Carstensen H, Nissen SD, Saljic A, Hesselkilde EM, van Hunnik A, Hohl M, Sattler SM, Fløgstad C, Hopster-Iversen C, Verheule S, Böhm M, Schotten U, Jespersen T, Buhl R. Long-Term Training Increases Atrial Fibrillation Sustainability in Standardbred Racehorses. J Cardiovasc Transl Res 2023; 16:1205-1219. [PMID: 37014465 PMCID: PMC10615936 DOI: 10.1007/s12265-023-10378-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 03/14/2023] [Indexed: 04/05/2023]
Abstract
Atrial fibrillation (AF) is more prevalent in athletes, and currently, the mechanisms are not fully understood. Atrial fibrillation inducibility and stability was investigated in trained and untrained Standardbred racehorses. The horses underwent echocardiography for evaluation of atrial size. High-density mapping during AF was performed, and the presence of structural remodeling, as well as the expression of inflammatory and pro-inflammatory markers in the atria, was studied. Atrial fibrillation sustained significantly longer after tachypacing in the trained horses, whereas no difference in AF inducibility was found. The untrained horses displayed a significant difference in the AF complexity when comparing right and left atria, whereas such difference was not observed in the trained animals. No evidence of increased structural remodeling or inflammation could be identified. Left atrial dimensions were not significantly increased. The increased AF sustainability in trained horses was not related to fibrosis or inflammation as seen in other animal exercise models.
Collapse
Affiliation(s)
- Helena Carstensen
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Højbakkegaard Allé 5, 2630, Taastrup, Denmark
| | - Sarah Dalgas Nissen
- Laboratory of Cardiac Physiology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark.
| | - Arnela Saljic
- Laboratory of Cardiac Physiology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Eva Melis Hesselkilde
- Laboratory of Cardiac Physiology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Arne van Hunnik
- Department of Physiology, Maastricht University, Maastricht, Netherlands
| | - Mathias Hohl
- Department of Internal Medicine III, University Hospital, Saarland University, Homburg, Saar, Germany
| | - Stefan Michael Sattler
- Laboratory of Cardiac Physiology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
- Department of Cardiology, Herlev and Gentofte University Hospital, Herlev, Denmark
| | - Cecilie Fløgstad
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Højbakkegaard Allé 5, 2630, Taastrup, Denmark
| | - Charlotte Hopster-Iversen
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Højbakkegaard Allé 5, 2630, Taastrup, Denmark
| | - Sander Verheule
- Department of Physiology, Maastricht University, Maastricht, Netherlands
| | - Michael Böhm
- Department of Internal Medicine III, University Hospital, Saarland University, Homburg, Saar, Germany
| | - Ulrich Schotten
- Department of Physiology, Maastricht University, Maastricht, Netherlands
| | - Thomas Jespersen
- Laboratory of Cardiac Physiology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Rikke Buhl
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Højbakkegaard Allé 5, 2630, Taastrup, Denmark
| |
Collapse
|
29
|
Meza-Ramos A, Alcarraz A, Lazo-Rodriguez M, Sangüesa G, Banon-Maneus E, Rovira J, Ramirez-Bajo MJ, Sitges M, Mont L, Ventura-Aguiar P, Batlle M, Guasch E. High-Intensity Exercise Promotes Deleterious Cardiovascular Remodeling in a High-Cardiovascular-Risk Model: A Role for Oxidative Stress. Antioxidants (Basel) 2023; 12:1462. [PMID: 37508000 PMCID: PMC10376780 DOI: 10.3390/antiox12071462] [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: 04/23/2023] [Revised: 07/01/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023] Open
Abstract
Although the benefits of moderate exercise in patients at high cardiovascular risk are well established, the effects of strenuous exercise remain unknown. We aimed to study the impact of strenuous exercise in a very high cardiovascular risk model. Nephrectomized aged Zucker obese rats were trained at a moderate (MOD) or high (INT) intensity or were kept sedentary (SED) for 10 weeks. Subsequently, echocardiography and ex vivo vascular reactivity assays were performed, and blood, aortas, perivascular adipose tissue (PVAT), and left ventricles (LVs) were harvested. An improved risk profile consisting of decreased body weight and improved response to a glucose tolerance test was noted in the trained groups. Vascular reactivity experiments in the descending thoracic aorta demonstrated increased endothelial NO release in the MOD group but not in the INT group, compared with SED; the free radical scavenger TEMPOL improved endothelial function in INT rats to a similar level as MOD. An imbalance in the expression of oxidative stress-related genes toward a pro-oxidant environment was observed in the PVAT of INT rats. In the heart, INT training promoted eccentric hypertrophy and a mild reduction in ejection fraction. Obesity was associated with LV fibrosis and a transition toward β-myosin heavy chain and the N2Ba titin isoform. Exercise reverted the myosin imbalance, but only MOD reduced the predominance of the N2Ba titin isoform. In conclusion, moderate exercise yields the most intense cardiovascular benefits in a high-cardiovascular-risk animal model, while intense training partially reverts them.
Collapse
Affiliation(s)
- Aline Meza-Ramos
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Medicine Department, Universitat de Barcelona, 08036 Barcelona, Spain
- Consejo Nacional de Ciencia y Tecnología (CONACyT), Ciudad de México 03940, Mexico
| | - Anna Alcarraz
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Medicine Department, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Marta Lazo-Rodriguez
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), 08036 Barcelona, Spain
| | - Gemma Sangüesa
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Elisenda Banon-Maneus
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), 08036 Barcelona, Spain
| | - Jordi Rovira
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), 08036 Barcelona, Spain
| | - Maria Jose Ramirez-Bajo
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), 08036 Barcelona, Spain
| | - Marta Sitges
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Medicine Department, Universitat de Barcelona, 08036 Barcelona, Spain
- Cardiovascular Institute, Clínic Barcelona, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Lluís Mont
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Medicine Department, Universitat de Barcelona, 08036 Barcelona, Spain
- Cardiovascular Institute, Clínic Barcelona, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Pedro Ventura-Aguiar
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), 08036 Barcelona, Spain
- Department of Nephrology and Kidney Transplantation, Clínic Barcelona, 08036 Barcelona, Spain
| | - Montserrat Batlle
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Eduard Guasch
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Medicine Department, Universitat de Barcelona, 08036 Barcelona, Spain
- Cardiovascular Institute, Clínic Barcelona, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| |
Collapse
|
30
|
Narasimhan B, Gandhi K, Moras E, Wu L, Da Wariboko A, Aronow W. Experimental drugs for supraventricular tachycardia: an analysis of early phase clinical trials. Expert Opin Investig Drugs 2023; 32:825-838. [PMID: 37728554 DOI: 10.1080/13543784.2023.2259309] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 09/12/2023] [Indexed: 09/21/2023]
Abstract
INTRODUCTION Supraventricular tachycardias (SVT) are a diverse group of commonly encountered arrhythmias arising at or above the atrioventricular (AV) node. Conventional anti-arrhythmic medications are restricted by extensive side-effect profiles and limited efficacy. Catheter ablation has emerged as a first-line therapy for many arrhythmias but is not a suitable option for all patients. This has prompted the exploration of novel pharmacological approaches targeting specific molecular mechanisms of SVT. AREAS COVERED This review article aims to summarize recent advancements in pharmacological therapeutics for SVT and their clinical implications. The understanding of molecular mechanisms underlying these arrhythmias, particularly atrial fibrillation, has opened up new possibilities for targeted interventions. Beyond the manipulation of ion channels and membrane potentials, pharmacotherapy now focuses on upstream targets such as inflammation, oxidative stress, and structural remodeling. This review strives to provide a comprehensive overview of recent advancements in pharmacological therapeutics directed at the management of SVT. We begin by providing a brief summary of the mechanisms and management of commonly encountered SVT before delving into individual agents, which in turn are stratified based on their molecular treatment targets. EXPERT OPINION The evolving landscape of pharmacologic therapy offers hope for more personalized and tailored interventions in the management of SVT.
Collapse
Affiliation(s)
- Bharat Narasimhan
- DeBakey Cardiovascular Institute, Houston Methodist, Houston, TX, USA
| | - Kruti Gandhi
- Department of Internal Medicine, Icahn School of Medicine, Mount Sinai, New York, NY, USA
| | - Errol Moras
- Department of Internal Medicine, Icahn School of Medicine, Mount Sinai, New York, NY, USA
| | - Lingling Wu
- Department of Medicine, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Akanibo Da Wariboko
- Department of Internal Medicine, Icahn School of Medicine, Mount Sinai, New York, NY, USA
| | - Wilbert Aronow
- Department of Cardiology, Westchester Medical Center, Valhalla, NY, USA
| |
Collapse
|
31
|
Hu D, Barajas-Martinez H, Zhang ZH, Duan HY, Zhao QY, Bao MW, Du YM, Burashnikov A, Monasky MM, Pappone C, Huang CX, Antzelevitch C, Jiang H. Advances in basic and translational research in atrial fibrillation. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220174. [PMID: 37122214 PMCID: PMC10150218 DOI: 10.1098/rstb.2022.0174] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/08/2023] [Indexed: 05/02/2023] Open
Abstract
Atrial fibrillation (AF) is a very common cardiac arrhythmia with an estimated prevalence of 33.5 million patients globally. It is associated with an increased risk of death, stroke and peripheral embolism. Although genetic studies have identified a growing number of genes associated with AF, the definitive impact of these genetic findings is yet to be established. Several mechanisms, including electrical, structural and neural remodelling of atrial tissue, have been proposed to contribute to the development of AF. Despite over a century of exploration, the molecular and cellular mechanisms underlying AF have not been fully established. Current antiarrhythmic drugs are associated with a significant rate of adverse events and management of AF using ablation is not optimal, especially in cases of persistent AF. This review discusses recent advances in our understanding and management of AF, including new concepts of epidemiology, genetics and pathophysiological mechanisms. We review the current status of antiarrhythmic drug therapy for AF, new potential agents, as well as mechanism-based AF ablation. This article is part of the theme issue 'The heartbeat: its molecular basis and physiological mechanisms'.
Collapse
Affiliation(s)
- Dan Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, People's Republic of China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, People's Republic of China
| | - Hector Barajas-Martinez
- Lankenau Institute for Medical Research, and Lankenau Heart Institute, Wynnwood, PA 19096, USA
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19104, USA
| | - Zhong-He Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, People's Republic of China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, People's Republic of China
| | - Hong-Yi Duan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, People's Republic of China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, People's Republic of China
| | - Qing-Yan Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, People's Republic of China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, People's Republic of China
| | - Ming-Wei Bao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, People's Republic of China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, People's Republic of China
| | - Yi-Mei Du
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| | - Alexander Burashnikov
- Lankenau Institute for Medical Research, and Lankenau Heart Institute, Wynnwood, PA 19096, USA
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19104, USA
| | - Michelle M. Monasky
- Arrhythmology Department, IRCCS Policlinico San Donato, San Donato Milanese, Milan 20097, Italy
| | - Carlo Pappone
- Arrhythmology Department, IRCCS Policlinico San Donato, San Donato Milanese, Milan 20097, Italy
- Vita-Salute San Raffaele University, Milan 20132, Italy
- Institute of Molecular and Translational Cardiology (IMTC), San Donato Milanese, Milan 20097, Italy
| | - Cong-Xin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, People's Republic of China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, People's Republic of China
| | - Charles Antzelevitch
- Lankenau Institute for Medical Research, and Lankenau Heart Institute, Wynnwood, PA 19096, USA
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19104, USA
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, People's Republic of China
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, People's Republic of China
| |
Collapse
|
32
|
Sang W, Wang L, Yan X, Sun H, Han Y, Wang F, Tang B, Li Y. Establishment of Risk Model and Analysis of Immunoinfiltration Based on Mitophagy-Related Associated Genes in Atrial Fibrillation. J Inflamm Res 2023; 16:2561-2583. [PMID: 37346800 PMCID: PMC10281282 DOI: 10.2147/jir.s415410] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 06/13/2023] [Indexed: 06/23/2023] Open
Abstract
Objective Atrial fibrillation (AF) is a common tachyarrhythmia whose pathogenesis remains elusive. In the present study, we aimed to investigate the pathological mechanism of mitophagy and immunoinfiltration in AF. Methods First, we identified differentially expressed mitophagy-related genes (DEMRGs) based on the GSE79768 and GSE115574 datasets, subjecting them to functional enrichment analysis. STRING, TRRUST, miRNet, miRwalk, and Cytoscape were used to explore the potential regulatory roles of downstream signaling pathways. Subsequently, the random forest method was used to construct the AF risk model, and the DEMRGs most correlated with AF risk were determined by combining the Gini index. ssGSEA algorithm, NMF algorithm, and unsupervised clustering were used to subdivide AF molecular types. We then studied the characteristics of mitophagy- and immune infiltration-related genes in AF. Ultimately, we detected the expression of key genes in canine atrial tissues and HL-1 cells by immunofluorescence and Western blot. Results Mitophagy and immune infiltration were significantly enriched and activated in AF samples. Thirty-seven DEMRGs were screened, of which MAPK1, VDAC1, MAPK14, and MTERF3 were most associated with AF risk. The risk model based on these could identify patients at a high risk of AF. The infiltration of immunocells such as mast cells and neutrophils was significantly different among AF types. Finally, expression verification indicated that the expression trend of four key genes in canine atrial muscle tissue and HL-1 cells was consistent. Conclusion We found that mitophagy may participate in AF progression through immune activation. In addition, the AF risk prediction model composed of VDAC1, MAPK1, MAPK14, and MTERF3 has a good AF prediction performance, which provides new ideas for the study of AF pathogenesis and potential therapeutic targets.
Collapse
Affiliation(s)
- Wanyue Sang
- Cardiac Pacing and Electrophysiology Department, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
| | - Lu Wang
- Cardiac Pacing and Electrophysiology Department, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
| | - Xiaoji Yan
- Department of Emergency, Sir Run Run Shaw Hospital Affiliated to Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Huaxin Sun
- Cardiac Pacing and Electrophysiology Department, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
| | - Yafan Han
- Cardiac Pacing and Electrophysiology Department, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
| | - Feifei Wang
- Cardiac Pacing and Electrophysiology Department, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
| | - Baopeng Tang
- Cardiac Pacing and Electrophysiology Department, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
| | - Yaodong Li
- Cardiac Pacing and Electrophysiology Department, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
| |
Collapse
|
33
|
Tatangelo M, Rebecchi M, Sgueglia M, Colella A, Crescenzi C, Panattoni G, Ciampi P, Lanza O, Canali E, Calò L. The Complex but Fascinating Relationship between Sport and Atrial Fibrillation: From Pathophysiology to the Clinical Scenario. J Cardiovasc Dev Dis 2023; 10:255. [PMID: 37367420 DOI: 10.3390/jcdd10060255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/04/2023] [Accepted: 06/07/2023] [Indexed: 06/28/2023] Open
Abstract
Atrial fibrillation (AF) is the most common cause of hospital admission among all arrhythmias in the general population. Moreover, AF represents the most common arrhythmia in the athletic population as well. The complex but fascinating relationship between sport and atrial fibrillation has not yet been fully clarified. Although the benefits of moderate physical activity in controlling cardiovascular risk factors and in reducing the risk of atrial fibrillation have been widely demonstrated, some concerns have been raised about the potential adverse effects of physical activity. Endurance activity in middle-aged men athletes appears to increase the risk of AF. Several different physiopathological mechanisms may explain the increased risk of AF in endurance athletes, including the imbalance of the autonomic nervous system, changes in left atrial size and function and presence of atrial fibrosis. The goal of this article is to review the epidemiology, pathophysiology and clinical management for AF in athletes, including pharmacological and electrophysiological strategies.
Collapse
Affiliation(s)
- Mario Tatangelo
- Division of Cardiology, Policlinico Casilino, Via Casilina 1049, 00169 Rome, Italy
| | - Marco Rebecchi
- Division of Cardiology, Policlinico Casilino, Via Casilina 1049, 00169 Rome, Italy
| | - Marianna Sgueglia
- Division of Cardiology, Policlinico Casilino, Via Casilina 1049, 00169 Rome, Italy
| | - Alessandra Colella
- Division of Cardiology, Policlinico Casilino, Via Casilina 1049, 00169 Rome, Italy
- BIND Department, University of Palermo, Piazza Marina, 61, 90133 Palermo, Italy
| | - Cinzia Crescenzi
- Division of Cardiology, Policlinico Casilino, Via Casilina 1049, 00169 Rome, Italy
| | - Germana Panattoni
- Division of Cardiology, Policlinico Casilino, Via Casilina 1049, 00169 Rome, Italy
| | - Pellegrino Ciampi
- Division of Cardiology, Policlinico Casilino, Via Casilina 1049, 00169 Rome, Italy
| | - Oreste Lanza
- Division of Cardiology, Policlinico Casilino, Via Casilina 1049, 00169 Rome, Italy
| | - Emanuele Canali
- Division of Cardiology, Policlinico Casilino, Via Casilina 1049, 00169 Rome, Italy
| | - Leonardo Calò
- Division of Cardiology, Policlinico Casilino, Via Casilina 1049, 00169 Rome, Italy
| |
Collapse
|
34
|
Sun X, Li L, Sun M, Hou S, Li Z, Li P, Liu M, Hua S. Evaluation of Left Ventricular Systolic Function Using Layer-Specific Strain in Rats Performing Endurance Exercise: A Pilot Study. ULTRASOUND IN MEDICINE & BIOLOGY 2023; 49:1395-1400. [PMID: 36878830 DOI: 10.1016/j.ultrasmedbio.2023.01.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/30/2022] [Accepted: 01/20/2023] [Indexed: 05/11/2023]
Abstract
OBJECTIVE The functional characteristics of exercise-induced myocardial hypertrophy were studied in a rat model in conjunction with ultrasound layered strain technique to investigate the hidden changes in the heart brought about by exercise. METHODS Forty specific pathogen free (SPF) adult Sprague-Dawley rats were selected and randomly divided into two groups of 20 exercise and 20 control rats. The longitudinal and circumferential strain parameters were measured using the ultrasonic stratified strain technique. The differences between the two groups and the predictive effect of stratified strain parameters on left ventricular systolic function were analyzed. RESULTS The exercise group had significantly higher global endocardial myocardial longitudinal strain (GLSendo), global mid-myocardial global longitudinal strain (GLSmid) and global endocardial myocardial global longitudinal strain (GCSendo) values than the control group (p < 0.05). Even though global mid-myocardial circumferential strain (GCSmid) and global epicardial myocardial circumferential strain (GCSepi) were higher in the exercise group than in the control group, statistical significance was not reached (p > 0.05). Conventional echocardiography parameters were well correlated with GLSendo, GLSmid, and GCSendo (p < 0.05). GLSendo was the best predictor of left ventricular myocardial contractile performance in athletes determined using the receiver operating characteristic curve, with an area under the curve of 0.97, sensitivity of 95% and specificity of 90%. CONCLUSION Rats performing endurance exercise exhibited subclinical changes in the heart after prolonged high-intensity exercise. A stratified strain parameter, GLSendo, played an important role in the evaluation of LV systolic performance in exercising rats.
Collapse
Affiliation(s)
- Xinxin Sun
- Department of Ultrasound, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lijin Li
- Department of Ultrasound, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mengjiao Sun
- Department of Ultrasound, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - SuYun Hou
- Department of Ultrasound, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhen Li
- Department of Ultrasound, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Pengge Li
- Department of Ultrasound, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mengmeng Liu
- Department of Ultrasound, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shaohua Hua
- Department of Ultrasound, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
35
|
Markandeya YS, Gregorich ZR, Feng L, Ramchandran V, O' Hara T, Vaidyanathan R, Mansfield C, Keefe AM, Beglinger CJ, Best JM, Kalscheur MM, Lea MR, Hacker TA, Gorelik J, Trayanova NA, Eckhardt LL, Makielski JC, Balijepalli RC, Kamp TJ. Caveolin-3 and Caveolae regulate ventricular repolarization. J Mol Cell Cardiol 2023; 177:38-49. [PMID: 36842733 PMCID: PMC10065933 DOI: 10.1016/j.yjmcc.2023.02.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 02/03/2023] [Accepted: 02/21/2023] [Indexed: 02/28/2023]
Abstract
RATIONALE Flask-shaped invaginations of the cardiomyocyte sarcolemma called caveolae require the structural protein caveolin-3 (Cav-3) and host a variety of ion channels, transporters, and signaling molecules. Reduced Cav-3 expression has been reported in models of heart failure, and variants in CAV3 have been associated with the inherited long-QT arrhythmia syndrome. Yet, it remains unclear whether alterations in Cav-3 levels alone are sufficient to drive aberrant repolarization and increased arrhythmia risk. OBJECTIVE To determine the impact of cardiac-specific Cav-3 ablation on the electrophysiological properties of the adult mouse heart. METHODS AND RESULTS Cardiac-specific, inducible Cav3 homozygous knockout (Cav-3KO) mice demonstrated a marked reduction in Cav-3 expression by Western blot and loss of caveolae by electron microscopy. However, there was no change in macroscopic cardiac structure or contractile function. The QTc interval was increased in Cav-3KO mice, and there was an increased propensity for ventricular arrhythmias. Ventricular myocytes isolated from Cav-3KO mice exhibited a prolonged action potential duration (APD) that was due to reductions in outward potassium currents (Ito, Iss) and changes in inward currents including slowed inactivation of ICa,L and increased INa,L. Mathematical modeling demonstrated that the changes in the studied ionic currents were adequate to explain the prolongation of the mouse ventricular action potential. Results from human iPSC-derived cardiomyocytes showed that shRNA knockdown of Cav-3 similarly prolonged APD. CONCLUSION We demonstrate that Cav-3 and caveolae regulate cardiac repolarization and arrhythmia risk via the integrated modulation of multiple ionic currents.
Collapse
Affiliation(s)
- Yogananda S Markandeya
- Cellular and Molecular Arrhythmia Research Program, Department of Medicine, University of Wisconsin Madison, WI, USA; National Institute of Mental Health and Neuroscience, Bengaluru, India
| | - Zachery R Gregorich
- Cellular and Molecular Arrhythmia Research Program, Department of Medicine, University of Wisconsin Madison, WI, USA
| | - Li Feng
- Department of Cardiology, Beijing Anzhen Hospital, Captial Medical University, National Clinical Research Center for Cardiovascular Diseases, Beijing, China
| | - Vignesh Ramchandran
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Thomas O' Hara
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Ravi Vaidyanathan
- Cellular and Molecular Arrhythmia Research Program, Department of Medicine, University of Wisconsin Madison, WI, USA
| | - Catherine Mansfield
- National Heart and Lung Institute, Imperial College London, ICTEM, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - Alexis M Keefe
- Cellular and Molecular Arrhythmia Research Program, Department of Medicine, University of Wisconsin Madison, WI, USA
| | - Carl J Beglinger
- Cellular and Molecular Arrhythmia Research Program, Department of Medicine, University of Wisconsin Madison, WI, USA
| | - Jabe M Best
- Cellular and Molecular Arrhythmia Research Program, Department of Medicine, University of Wisconsin Madison, WI, USA
| | - Matthew M Kalscheur
- Cellular and Molecular Arrhythmia Research Program, Department of Medicine, University of Wisconsin Madison, WI, USA
| | - Martin R Lea
- Cellular and Molecular Arrhythmia Research Program, Department of Medicine, University of Wisconsin Madison, WI, USA
| | - Timothy A Hacker
- Cellular and Molecular Arrhythmia Research Program, Department of Medicine, University of Wisconsin Madison, WI, USA
| | - Julia Gorelik
- National Heart and Lung Institute, Imperial College London, ICTEM, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - Natalia A Trayanova
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Lee L Eckhardt
- Cellular and Molecular Arrhythmia Research Program, Department of Medicine, University of Wisconsin Madison, WI, USA
| | - Jonathan C Makielski
- Cellular and Molecular Arrhythmia Research Program, Department of Medicine, University of Wisconsin Madison, WI, USA
| | - Ravi C Balijepalli
- Cellular and Molecular Arrhythmia Research Program, Department of Medicine, University of Wisconsin Madison, WI, USA
| | - Timothy J Kamp
- Cellular and Molecular Arrhythmia Research Program, Department of Medicine, University of Wisconsin Madison, WI, USA.
| |
Collapse
|
36
|
Dobrev D, Heijman J, Hiram R, Li N, Nattel S. Inflammatory signalling in atrial cardiomyocytes: a novel unifying principle in atrial fibrillation pathophysiology. Nat Rev Cardiol 2023; 20:145-167. [PMID: 36109633 PMCID: PMC9477170 DOI: 10.1038/s41569-022-00759-w] [Citation(s) in RCA: 132] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/26/2022] [Indexed: 02/08/2023]
Abstract
Inflammation has been implicated in atrial fibrillation (AF), a very common and clinically significant cardiac rhythm disturbance, but its precise role remains poorly understood. Work performed over the past 5 years suggests that atrial cardiomyocytes have inflammatory signalling machinery - in particular, components of the NLRP3 (NACHT-, LRR- and pyrin domain-containing 3) inflammasome - that is activated in animal models and patients with AF. Furthermore, work in animal models suggests that NLRP3 inflammasome activation in atrial cardiomyocytes might be a sufficient and necessary condition for AF occurrence. In this Review, we evaluate the evidence for the role and pathophysiological significance of cardiomyocyte NLRP3 signalling in AF. We first summarize the evidence for a role of inflammation in AF and review the biochemical properties of the NLRP3 inflammasome, as defined primarily in studies of classic inflammation. We then briefly consider the broader evidence for a role of inflammatory signalling in heart disease, particularly conditions that predispose individuals to develop AF. We provide a detailed discussion of the available information about atrial cardiomyocyte NLRP3 inflammasome signalling in AF and related conditions and evaluate the possibility that similar signalling might be important in non-myocyte cardiac cells. We then review the evidence on the role of active resolution of inflammation and its potential importance in suppressing AF-related inflammatory signalling. Finally, we consider the therapeutic potential and broader implications of this new knowledge and highlight crucial questions to be addressed in future research.
Collapse
Affiliation(s)
- Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Duisburg, Germany
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Canada
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Jordi Heijman
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Faculty of Health, Medicine, and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Roddy Hiram
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Canada
| | - Na Li
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Section of Cardiovascular Research, Baylor College of Medicine, Houston, TX, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Stanley Nattel
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Duisburg, Germany.
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Canada.
- IHU LIRYC and Fondation Bordeaux Université, Bordeaux, France.
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
37
|
Polyák A, Topal L, Zombori-Tóth N, Tóth N, Prorok J, Kohajda Z, Déri S, Demeter-Haludka V, Hegyi P, Venglovecz V, Ágoston G, Husti Z, Gazdag P, Szlovák J, Árpádffy-Lovas T, Naveed M, Sarusi A, Jost N, Virág L, Nagy N, Baczkó I, Farkas AS, Varró A. Cardiac electrophysiological remodeling associated with enhanced arrhythmia susceptibility in a canine model of elite exercise. eLife 2023; 12:80710. [PMID: 36815557 PMCID: PMC10014074 DOI: 10.7554/elife.80710] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 02/16/2023] [Indexed: 02/24/2023] Open
Abstract
The health benefits of regular physical exercise are well known. Even so, there is increasing evidence that the exercise regimes of elite athletes can evoke cardiac arrhythmias including ventricular fibrillation and even sudden cardiac death (SCD). The mechanism of exercise-induced arrhythmia and SCD is poorly understood. Here, we show that chronic training in a canine model (12 sedentary and 12 trained dogs) that mimics the regime of elite athletes induces electrophysiological remodeling (measured by ECG, patch-clamp, and immunocytochemical techniques) resulting in increases of both the trigger and the substrate for ventricular arrhythmias. Thus, 4 months sustained training lengthened ventricular repolarization (QTc: 237.1±3.4 ms vs. 213.6±2.8 ms, n=12; APD90: 472.8±29.6 ms vs. 370.1±32.7 ms, n=29 vs. 25), decreased transient outward potassium current (6.4±0.5 pA/pF vs. 8.8±0.9 pA/pF at 50 mV, n=54 vs. 42), and increased the short-term variability of repolarization (29.5±3.8 ms vs. 17.5±4.0 ms, n=27 vs. 18). Left ventricular fibrosis and HCN4 protein expression were also enhanced. These changes were associated with enhanced ectopic activity (number of escape beats from 0/hr to 29.7±20.3/hr) in vivo and arrhythmia susceptibility (elicited ventricular fibrillation: 3 of 10 sedentary dogs vs. 6 of 10 trained dogs). Our findings provide in vivo, cellular electrophysiological and molecular biological evidence for the enhanced susceptibility to ventricular arrhythmia in an experimental large animal model of endurance training.
Collapse
Affiliation(s)
- Alexandra Polyák
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
| | - Leila Topal
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
| | - Noémi Zombori-Tóth
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
| | - Noémi Tóth
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
| | - János Prorok
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
- ELKH-SZTE Research Group for Cardiovascular Pharmacology, Eötvös Loránd Research NetworkSzegedHungary
| | - Zsófia Kohajda
- ELKH-SZTE Research Group for Cardiovascular Pharmacology, Eötvös Loránd Research NetworkSzegedHungary
| | - Szilvia Déri
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
| | | | - Péter Hegyi
- Centre for Translational Medicine and Institute of Pancreatic Diseases, Semmelweis UniversityBudapestHungary
- Institute for Translational Medicine, Medical School, University of PécsPécsHungary
- Translational Pancreatology Research Group, Interdisciplinary Centre of Excellence for Research Development and Innovation, University of SzegedSzegedHungary
| | - Viktória Venglovecz
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
| | - Gergely Ágoston
- Institute of Family Medicine, University of SzegedSzegedHungary
| | - Zoltán Husti
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
| | - Péter Gazdag
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
| | - Jozefina Szlovák
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
| | - Tamás Árpádffy-Lovas
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
| | - Muhammad Naveed
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
| | - Annamária Sarusi
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
| | - Norbert Jost
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
- ELKH-SZTE Research Group for Cardiovascular Pharmacology, Eötvös Loránd Research NetworkSzegedHungary
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of SzegedSzegedHungary
| | - László Virág
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of SzegedSzegedHungary
| | - Norbert Nagy
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
- ELKH-SZTE Research Group for Cardiovascular Pharmacology, Eötvös Loránd Research NetworkSzegedHungary
| | - István Baczkó
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of SzegedSzegedHungary
| | - Attila S Farkas
- Department of Internal Medicine, Cardiology ward, University of SzegedSzegedHungary
| | - András Varró
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
- ELKH-SZTE Research Group for Cardiovascular Pharmacology, Eötvös Loránd Research NetworkSzegedHungary
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of SzegedSzegedHungary
| |
Collapse
|
38
|
Abstract
The global prevalence of atrial fibrillation (AF) has increased substantially over the past three decades and is currently approximately 60 million cases. Incident AF and its clinical consequences are largely the result of risk factors that can be modified by lifestyle changes. In this Review, we provide evidence that the lifetime risk of AF is modified not only by sex and race but also through the clinical risk factor and comorbidity burden of individual patients. We begin by summarizing the epidemiology of AF, focusing on non-modifiable and modifiable risk factors, as well as targets and strategies for the primary prevention of AF. Furthermore, we evaluate the role of modifiable risk factors in the secondary prevention of AF as well as the potential effects of risk factor interventions on the frequency and severity of subsequent AF episodes. We end the Review by proposing strategies that require evaluation as well as global policy changes that are needed for the prevention of incident AF and the management of recurrent episodes in patients already affected by AF.
Collapse
|
39
|
Murphy MB, Kannankeril PJ, Murray KT. Overview of programmed electrical stimulation to assess atrial fibrillation susceptibility in mice. Front Physiol 2023; 14:1149023. [PMID: 37113690 PMCID: PMC10126433 DOI: 10.3389/fphys.2023.1149023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/31/2023] [Indexed: 04/29/2023] Open
Abstract
Atrial fibrillation (AF) is the most common human arrhythmia and is associated with increased risk of stroke, dementia, heart failure, and death. Among several animal models that have been used to investigate the molecular determinants of AF, mouse models have become the most prevalent due to low cost, ease of genetic manipulation, and similarity to human disease. Programmed electrical stimulation (PES) using intracardiac or transesophageal atrial pacing is used to induce AF as most mouse models do not develop spontaneous AF. However, there is a lack of standardized methodology resulting in numerous PES protocols in the literature that differ with respect to multiple parameters, including pacing protocol and duration, stimulus amplitude, pulse width, and even the definition of AF. Given this complexity, the selection of the appropriate atrial pacing protocol for a specific model has been arbitrary. Herein we review the development of intracardiac and transesophageal PES, including commonly used protocols, selected experimental models, and advantages and disadvantages of both techniques. We also emphasize detection of artifactual AF induction due to unintended parasympathetic stimulation, which should be excluded from results. We recommend that the optimal pacing protocol to elicit an AF phenotype should be individualized to the specific model of genetic or acquired risk factors, with an analysis using several definitions of AF as an endpoint.
Collapse
|
40
|
Inflammatory cytokines differ between patients with high versus low CHA2DS2-VASc scores in sinus rhythm-a possible mechanism for adverse cardiovascular events. INTERNATIONAL JOURNAL OF CARDIOLOGY. CARDIOVASCULAR RISK AND PREVENTION 2022; 15:200155. [PMID: 36573192 PMCID: PMC9789347 DOI: 10.1016/j.ijcrp.2022.200155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022]
Abstract
Background The CHA2DS2-VASc score was shown to predict systemic thromboembolism and mortality in certain groups of patients in sinus rhythm (SR). Previous data showed that patients in SR with high CHA2DS2-VASc score have higher plasma levels of inflammatory markers such as sP-selectin and C-reactive protein. We further investigated this group. Methods Blood samples were collected from consecutive patients in SR. Plasma was extracted and stored at -80 °C. Concentrations of a panel of soluble markers IL-1β, IL-6, IL-8, IL-10, TNF-α and VEGF were measured by Magnetic Luminex Performance Assay. The PLF4 cytokine blood level was measured by ELISA. Results 66 patients were enrolled (age 53 ± 18 years, 60% women). Patients with high CHA2DS2-VASc scores (n = 23) had significantly higher median IQR concentrations of TNF-α [10.34 (8.55,14.92) vs. 7.69 (6.06, 9.85) pg/ml, p = 0.009] and a trend towards higher levels of IL-1β [0.59 (0.4,0.8) vs. 0.44 (0.31, 0.62) pg/ml, p = 0.07] and IL-8 [5.92 (4.5,9.4) vs. 5.04 (3.63, 6.04) pg/ml, p = 0.07], compared to the group with low scores (n = 43). Median IQR concentrations of VEGF, IL-6, IL-10 and PF4 did not significantly differ between the CHA2DS2-VASc score groups. Conclusion Patients in SR with high versus low CHA2DS2-VASc scores have high plasma concentrations of systemic inflammation cytokines. The already proven high levels of sP-selectin, that promotes release of inflammatory cytokines from leukocytes, is in line with these results. This pro-inflammatory state in patients with high CHA2DS2-VASc scores, may explain the higher rate of adverse cardiovascular events associated with elevated CHA2DS2-VASc score even without atrial fibrillation.
Collapse
|
41
|
La Gerche A, Wasfy MM, Brosnan MJ, Claessen G, Fatkin D, Heidbuchel H, Baggish AL, Kovacic JC. The Athlete's Heart-Challenges and Controversies: JACC Focus Seminar 4/4. J Am Coll Cardiol 2022; 80:1346-1362. [PMID: 36075838 DOI: 10.1016/j.jacc.2022.07.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 07/18/2022] [Accepted: 07/21/2022] [Indexed: 12/11/2022]
Abstract
Regular exercise promotes structural, functional, and electrical remodeling of the heart, often referred to as the "athlete's heart," with intense endurance sports being associated with the greatest degree of cardiac remodeling. However, the extremes of exercise-induced cardiac remodeling are potentially associated with uncommon side effects. Atrial fibrillation is more common among endurance athletes and there is speculation that other arrhythmias may also be more prevalent. It is yet to be determined whether this arrhythmic susceptibility is a result of extreme exercise remodeling, genetic predisposition, or other factors. Gender may have the greatest influence on the cardiac response to exercise, but there has been far too little research directed at understanding differences in the sportsman's vs sportswoman's heart. Here in part 4 of a 4-part seminar series, the controversies and ambiguities regarding the athlete's heart, and in particular, its arrhythmic predisposition, genetic, and gender influences are reviewed in depth.
Collapse
Affiliation(s)
- Andre La Gerche
- Clinical Research Domain, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; National Centre for Sports Cardiology, Fitzroy, Victoria, Australia; Cardiology Department, St Vincent's Hospital Melbourne, Fitzroy, Victoria, Australia.
| | - Meagan M Wasfy
- Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts, USA; Cardiovascular Performance Program, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Maria J Brosnan
- National Centre for Sports Cardiology, Fitzroy, Victoria, Australia; Cardiology Department, St Vincent's Hospital Melbourne, Fitzroy, Victoria, Australia
| | - Guido Claessen
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium; Department of Cardiovascular Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Diane Fatkin
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, New South Wales, Australia; Cardiology Department, St Vincent's Hospital, Darlinghurst, New South Wales, Australia
| | - Hein Heidbuchel
- Antwerp University Hospital, Department of Cardiology, Antwerp, Belgium; Cardiovascular Sciences, Antwerp University, Antwerp, Belgium
| | - Aaron L Baggish
- Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts, USA; Cardiovascular Performance Program, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jason C Kovacic
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, New South Wales, Australia; Cardiology Department, St Vincent's Hospital, Darlinghurst, New South Wales, Australia; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| |
Collapse
|
42
|
Sung DJ, Jeon YK, Choi J, Kim B, Golpasandi S, Park SW, Oh SB, Bae YM. Protective effect of low-intensity treadmill exercise against acetylcholine-calcium chloride-induced atrial fibrillation in mice. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2022; 26:313-323. [PMID: 36039732 PMCID: PMC9437371 DOI: 10.4196/kjpp.2022.26.5.313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 06/15/2023]
Abstract
Atrial fibrillation (AF) is the most common supraventricular arrhythmia, and it corresponds highly with exercise intensity. Here, we induced AF in mice using acetylcholine (ACh)-CaCl2 for 7 days and aimed to determine the appropriate exercise intensity (no, low, moderate, high) to protect against AF by running the mice at different intensities for 4 weeks before the AF induction by ACh-CaCl2. We examined the AF-induced atrial remodeling using electrocardiogram, patch-clamp, and immunohistochemistry. After the AF induction, heart rate, % increase of heart rate, and heart weight/body weight ratio were significantly higher in all the four AF groups than in the normal control; highest in the high-ex AF and lowest in the low-ex (lower than the no-ex AF), which indicates that low-ex treated the AF. Consistent with these changes, G protein-gated inwardly rectifying K+ currents, which were induced by ACh, increased in an exercise intensity-dependent manner and were lower in the low-ex AF than the no-ex AF. The peak level of Ca2+ current (at 0 mV) increased also in an exercise intensity-dependent manner and the inactivation time constants were shorter in all AF groups except for the low-ex AF group, in which the time constant was similar to that of the control. Finally, action potential duration was shorter in all the four AF groups than in the normal control; shortest in the high-ex AF and longest in the low-ex AF. Taken together, we conclude that low-intensity exercise protects the heart from AF, whereas high-intensity exercise might exacerbate AF.
Collapse
Affiliation(s)
- Dong-Jun Sung
- Department of Sport and Health Studies, College of Biomedical and Health Science, Konkuk University, Chungju 27478, Korea
- Sports Convergence Institute, Chungju 27478, Korea
- Center for Metabolic Diseases, Konkuk University, Chungju 27478, Korea
| | - Yong-Kyun Jeon
- Department of Physical Education at the Graduate School of Education, Dankook University, Yongin 16890, Korea
| | - Jaeil Choi
- Department of Physical Education at the Graduate School of Education, Dankook University, Yongin 16890, Korea
| | - Bokyung Kim
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 27478, Korea
| | - Shadi Golpasandi
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 27478, Korea
| | - Sang Woong Park
- Department of Emergency Medical Services, College of Health Sciences, Eulji University, Seongam 13135, Korea
| | - Seung-Bum Oh
- Department of Sport and Health Studies, College of Biomedical and Health Science, Konkuk University, Chungju 27478, Korea
| | - Young Min Bae
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 27478, Korea
| |
Collapse
|
43
|
Yao Y, Yang M, Liu D, Zhao Q. Immune remodeling and atrial fibrillation. Front Physiol 2022; 13:927221. [PMID: 35936905 PMCID: PMC9355726 DOI: 10.3389/fphys.2022.927221] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 06/30/2022] [Indexed: 11/13/2022] Open
Abstract
Atrial fibrillation (AF) is a highly prevalent arrhythmia that causes high morbidity and mortality. However, the underlying mechanism of AF has not been fully elucidated. Recent research has suggested that, during AF, the immune system changes considerably and interacts with the environment and cells involved in the initiation and maintenance of AF. This may provide a new direction for research and therapeutic strategies for AF. In this review, we elaborate the concept of immune remodeling based on available data in AF. Then, we highlight the complex relationships between immune remodeling and atrial electrical, structural and neural remodeling while also pointing out some research gaps in these field. Finally, we discuss several potential immunomodulatory treatments for AF. Although the heterogeneity of existing evidence makes it ambiguous to extrapolate immunomodulatory treatments for AF into the clinical practice, immune remodeling is still an evolving concept in AF pathophysiology and further studies within this field are likely to provide effective therapies for AF.
Collapse
Affiliation(s)
- Yajun Yao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Mei Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Dishiwen Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Qingyan Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
- *Correspondence: Qingyan Zhao,
| |
Collapse
|
44
|
Chu Y, Yu F, Wu Y, Yang J, Shi J, Ye T, Han D, Wang X. Identification of genes and key pathways underlying the pathophysiological association between nonalcoholic fatty liver disease and atrial fibrillation. BMC Med Genomics 2022; 15:150. [PMID: 35790963 PMCID: PMC9258143 DOI: 10.1186/s12920-022-01300-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 06/27/2022] [Indexed: 11/15/2022] Open
Abstract
Background Atrial fibrillation (AF) is one of the most prevalent sustained cardiac arrhythmias. The latest studies have revealed a tight correlation between nonalcoholic fatty liver disease (NAFLD) and AF. However, the exact molecular mechanisms underlying the association between NAFLD and AF remain unclear. The current research aimed to expound the genes and signaling pathways that are related to the mechanisms underlying the association between these two diseases. Materials and methods NAFLD- and AF- related differentially expressed genes (DEGs) were identified via bioinformatic analysis of the Gene Expression Omnibus (GEO) datasets GSE63067 and GSE79768, respectively. Further enrichment analysis of Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG), the construction of a protein–protein interaction (PPI) network, the identification of significant hub genes, and receiver operator characteristic curve analysis were conducted. The gene-disease interactions were analyzed using the Comparative Toxicogenomics Database. In addition, the hub genes were validated by quantitative Real-Time PCR (qRT-PCR) in NAFLD cell model. Results A total of 45 co-expressed differentially expressed genes (co-DEGs) were identified between the NAFLD/AF and healthy control individuals. GO and KEGG pathway analyses revealed that the co-DEGs were mostly enriched in neutrophil activation involved in the immune response and cytokine-cytokine receptor interactions. Moreover, eight hub genes were selected owing to their high degree of connectivity and upregulation in both the NAFLD and AF datasets. These genes included CCR2, PTPRC, CXCR2, MNDA, S100A9, NCF2, S100A12, and S100A8. Conclusions In summary, we conducted the gene differential expression analysis, functional enrichment analysis, and PPI analysis of DEGs in AF and NAFLD, which provides novel insights into the identification of potential biomarkers and valuable therapeutic leads for AF and NAFLD. Supplementary Information The online version contains supplementary material available at 10.1186/s12920-022-01300-1.
Collapse
|
45
|
Rao P, Belanger MJ, Robbins JM. Exercise, Physical Activity, and Cardiometabolic Health: Insights into the Prevention and Treatment of Cardiometabolic Diseases. Cardiol Rev 2022; 30:167-178. [PMID: 34560712 PMCID: PMC8920940 DOI: 10.1097/crd.0000000000000416] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Physical activity (PA) and exercise are widely recognized as essential components of primary and secondary cardiovascular disease (CVD) prevention efforts and are emphasized in the health promotion guidelines of numerous professional societies and committees. The protean benefits of PA and exercise extend across the spectrum of CVD, and include the improvement and reduction of risk factors and events for atherosclerotic CVD (ASCVD), cardiometabolic disease, heart failure, and atrial fibrillation (AF), respectively. Here, we highlight recent insights into the salutary effects of PA and exercise on the primary and secondary prevention of ASCVD, including their beneficial effects on both traditional and nontraditional risk mediators; exercise "prescriptions" for ASCVD; the role of PA regular exercise in the prevention and treatment of heart failure; and the relationships between, PA, exercise, and AF. While our understanding of the relationship between exercise and CVD has evolved considerably, several key questions remain including the association between extreme volumes of exercise and subclinical ASCVD and its risk; high-intensity exercise and resistance (strength) training as complementary modalities to continuous aerobic exercise; and dose- and intensity-dependent associations between exercise and AF. Recent advances in molecular profiling technologies (ie, genomics, transcriptomics, proteomics, and metabolomics) have begun to shed light on interindividual variation in cardiometabolic responses to PA and exercise and may provide new opportunities for clinical prediction in addition to mechanistic insights.
Collapse
Affiliation(s)
- Prashant Rao
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA
- Cardiovascular Research Center, Beth Israel Deaconess Medical Center, Boston, MA
| | | | - Jeremy M. Robbins
- Cardiovascular Research Center, Beth Israel Deaconess Medical Center, Boston, MA
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| |
Collapse
|
46
|
Mechanisms of Quercetin against atrial fibrillation explored by network pharmacology combined with molecular docking and experimental validation. Sci Rep 2022; 12:9777. [PMID: 35697725 PMCID: PMC9192746 DOI: 10.1038/s41598-022-13911-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 05/30/2022] [Indexed: 01/19/2023] Open
Abstract
Atrial fibrillation (AF) is a common atrial arrhythmia for which there is no specific therapeutic drug. Quercetin (Que) has been used to treat cardiovascular diseases such as arrhythmias. In this study, we explored the mechanism of action of Que in AF using network pharmacology and molecular docking. The chemical structure of Que was obtained from Pubchem. TCMSP, Swiss Target Prediction, Drugbank, STITCH, Pharmmapper, CTD, GeneCards, DISGENET and TTD were used to obtain drug component targets and AF-related genes, and extract AF and normal tissue by GEO database differentially expressed genes by GEO database. The top targets were IL6, VEGFA, JUN, MMP9 and EGFR, and Que for AF treatment might involve the role of AGE-RAGE signaling pathway in diabetic complications, MAPK signaling pathway and IL-17 signaling pathway. Molecular docking showed that Que binds strongly to key targets and is differentially expressed in AF. In vivo results showed that Que significantly reduced the duration of AF fibrillation and improved atrial remodeling, reduced p-MAPK protein expression, and inhibited the progression of AF. Combining network pharmacology and molecular docking approaches with in vivo studies advance our understanding of the intensive mechanisms of Quercetin, and provide the targeted basis for clinical Atrial fibrillation treatment.
Collapse
|
47
|
Inflammasome Signaling in Atrial Fibrillation. J Am Coll Cardiol 2022; 79:2349-2366. [DOI: 10.1016/j.jacc.2022.03.379] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/29/2022] [Accepted: 03/30/2022] [Indexed: 12/26/2022]
|
48
|
Effect of Alpha-Lipoic Acid on Rat Ventricles and Atria under LPS-Induced Oxidative Stress. Antioxidants (Basel) 2022; 11:antiox11040734. [PMID: 35453419 PMCID: PMC9024801 DOI: 10.3390/antiox11040734] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/02/2022] [Accepted: 04/06/2022] [Indexed: 02/01/2023] Open
Abstract
Alpha-lipoic acid (α-LA) is a disulfide compound and one of the most effective antioxidants. Many studies have indicated positive effects of α-LA in the prevention of pathologic conditions mediated by oxidative stress, such as cardiovascular diseases. However, the therapeutic potential of α-LA for the heart has not been explored with regards to the ventricles and atria. The aim of our study was to evaluate the effects of α-LA on oxidative stress parameters and inflammation in the ventricles and atria of the heart in rats under LPS-induced oxidative stress. Wistar rats were divided into 4 groups: I—control (received 2 doses of 0.2 mL of 0.9% NaCl i.v., 0.5 h apart); II—α-LA (received 0.2 mL of 0.9% NaCl and 0.5 h later received α-LA 60 mg/kg b.w. i.v.); III—lipopolysaccharide (LPS) (received 0.2 mL of 0.9% NaCl and 0.5 h later received LPS 30 mg/kg b.w. i.v.); and IV—LPS + LA (received LPS 30 mg/kg b.w. i.v. and 0.5 h later received α-LA 60 mg/kg b.w. i.v.). Five hours later, the rats were euthanized. The hearts were surgically removed and weighed to estimate heart edema. The ventricular and atrium tissue was isolated to measure levels of TNF-α, IL-6, superoxide dismutase (SOD), thiobarbituric acid reactive substances (TBARS), hydrogen peroxide (H2O2), total sulfhydryl groups (-SH), total glutathione (tGSH), reduced glutathione (GSH), glutathione disulfide (GSSG), and the GSH/GSSG ratio. LPS significantly increased TNF-α, IL-6, TBARS, and H2O2 levels and decreased SOD, -SH groups, tGSH, the GSH/GSSG ratio, and GSH levels in rat ventricles and atria while α-LA administered after the injection of LPS significantly decreased TNF-α, IL-6, TBARS, and H2O2 levels. α-LA also increased SOD and -SH group levels and ameliorated the glutathione redox status when compared to the LPS group. Our data suggest that α-LA administration 30 min after LPS infusion may effectively prevent inflammation and oxidative stress in the ventricles and atria.
Collapse
|
49
|
Emerging Antiarrhythmic Drugs for Atrial Fibrillation. Int J Mol Sci 2022; 23:ijms23084096. [PMID: 35456912 PMCID: PMC9029767 DOI: 10.3390/ijms23084096] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/28/2022] [Accepted: 04/01/2022] [Indexed: 12/19/2022] Open
Abstract
Atrial fibrillation (AF), the most common cardiac arrhythmia worldwide, is driven by complex mechanisms that differ between subgroups of patients. This complexity is apparent from the different forms in which AF presents itself (post-operative, paroxysmal and persistent), each with heterogeneous patterns and variable progression. Our current understanding of the mechanisms responsible for initiation, maintenance and progression of the different forms of AF has increased significantly in recent years. Nevertheless, antiarrhythmic drugs for the management of AF have not been developed based on the underlying arrhythmia mechanisms and none of the currently used drugs were specifically developed to target AF. With the increased knowledge on the mechanisms underlying different forms of AF, new opportunities for developing more effective and safer AF therapies are emerging. In this review, we provide an overview of potential novel antiarrhythmic approaches based on the underlying mechanisms of AF, focusing both on the development of novel antiarrhythmic agents and on the possibility of repurposing already marketed drugs. In addition, we discuss the opportunity of targeting some of the key players involved in the underlying AF mechanisms, such as ryanodine receptor type-2 (RyR2) channels and atrial-selective K+-currents (IK2P and ISK) for antiarrhythmic therapy. In addition, we highlight the opportunities for targeting components of inflammatory signaling (e.g., the NLRP3-inflammasome) and upstream mechanisms targeting fibroblast function to prevent structural remodeling and progression of AF. Finally, we critically appraise emerging antiarrhythmic drug principles and future directions for antiarrhythmic drug development, as well as their potential for improving AF management.
Collapse
|
50
|
Bressi E, Rebecchi M, Sgueglia M, Crescenzi C, Panattoni G, Martino A, Casalese A, Sangiorgi C, Politano A, Cicogna F, Fagagnini A, Grieco D, DE Ruvo E, Calò L. Atrial fibrillation and sport: need for monitoring. Minerva Cardiol Angiol 2022; 70:594-605. [PMID: 35343173 DOI: 10.23736/s2724-5683.22.05842-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Historically, regular exercise contributed to reduce the arrhythmic burden and improve cardiovascular outcomes in the general population. However, a heightened risk of Atrial Fibrillation (AF) seems to occur mainly amongst endurance athletes. The exact mechanisms are not fully elucidated, but dynamic interactions between electroanatomical changes induced by exercise, the autonomic system, variable triggers, along individual genetic predisposition are the main contributors to AF development in athletes. The type and training load of sports are also crucial in determining the arrhythmogenic milieu predisposing to AF insurgence and perpetuation. Moreover, a sex difference seems to influence an increased risk of AF only in men undergoing strenuous exercise, whereas women appear protected even during more vigorous training. In the absence of solid evidence, the advent of modern technologies could help to monitor and deep investigate the peculiar aspects of AF in these athletes. This review aims to describe the pathophysiology, diagnosis, and management of AF in athletes, shedding light on possible future strategies to face AF in this population.
Collapse
Affiliation(s)
- Edoardo Bressi
- Department of Cardiology, Policlinico Casilino of Rome, Rome, Italy -
| | - Marco Rebecchi
- Department of Cardiology, Policlinico Casilino of Rome, Rome, Italy
| | | | - Cinzia Crescenzi
- Department of Cardiology, Policlinico Casilino of Rome, Rome, Italy
| | | | | | | | - Catia Sangiorgi
- Department of Cardiology, Policlinico Casilino of Rome, Rome, Italy
| | | | | | | | - Domenico Grieco
- Department of Cardiology, Policlinico Casilino of Rome, Rome, Italy
| | | | - Leonardo Calò
- Department of Cardiology, Policlinico Casilino of Rome, Rome, Italy
| |
Collapse
|