1
|
Schmidt C, Wiedmann F. Disease mechanism and novel drug therapies for atrial fibrillation. MED GENET-BERLIN 2025; 37:147-154. [PMID: 40207039 PMCID: PMC11976400 DOI: 10.1515/medgen-2025-2005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
Atrial fibrillation (AF), the most common sustained cardiac arrhythmia, affects over 3 % of adults globally, increasing risks for stroke, heart failure, and cognitive decline. Early rhythm control shows promise in improving AF prognosis, and catheter ablation remains an effective, safe option, especially for paroxysmal AF. However, high recurrence rates with antiarrhythmic drugs and ablation persist, particularly in cases of persistent AF. Emerging research on molecular mechanisms has led to innovative therapeutic strategies targeting these pathways, offering hope for more effective AF management. This review explores recent insights into the complex pathophysiology of AF, with a particular focus on ion channel dysfunction, calcium mishandling, oxidative stress, and fibrosis. It further considers how these factors will inspire new therapeutic options.
Collapse
Affiliation(s)
- Constanze Schmidt
- Medical University Hospital HeidelbergDepartment of CardiologyIm Neuenheimer Feld 41069120HeidelbergGermany
| | - Felix Wiedmann
- Medical University Hospital HeidelbergDepartment of CardiologyIm Neuenheimer Feld 41069120HeidelbergGermany
| |
Collapse
|
2
|
Chen C, Wang G, Zou Q, Xiong K, Chen Z, Shao B, Liu Y, Xie D, Ji Y. m 6A reader YTHDF2 governs the onset of atrial fibrillation by modulating Cacna1c translation. SCIENCE CHINA. LIFE SCIENCES 2025; 68:706-721. [PMID: 39432207 DOI: 10.1007/s11427-024-2674-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/02/2024] [Indexed: 10/22/2024]
Abstract
Atrial fibrillation (AF) is the most common arrhythmia, which is tightly associated with the abnormal expression and function of ion channels in the atrial cardiomyocytes. N6-methyladenosine (m6A), a widespread chemical modification in eukaryotic mRNA, is known to play a significant regulatory role in the pathogenesis of heart disease. However, the significance of m6A regulatory proteins in the onset of AF remains unclear. Here, we demonstrate that the m6A reader protein YTHDF2 regulates atrial electrical remodeling and AF onset by modulating the Cav1.2 expression. Firstly, YTHDF2 expression was selectively upregulated in rat atrial cardiomyocytes with AF. Secondly, YTHDF2 knockout reduced AF susceptibility in mice. Thirdly, the knockout of YTHDF2 increased Cav1.2 protein levels in an m6A-in-dependent manner, ultimately prolonging the atrial myocardial refractory period, a critical electrophysiological substrate for the onset of AF. Fourthly, the N-terminal domain of YTHDF2 was identified as critical for Cacna1c mRNA translation regulation. Overall, our findings unveil that YTHDF2 can alter Cav1.2 protein expression in an m6A-independent manner, thereby facilitating the onset of AF. Our study suggests that YTHDF2 may be a potential intervention target for AF.
Collapse
Affiliation(s)
- Chuansheng Chen
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Guanghua Wang
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Department of Pathology and Pathophysiology, School of Medicine, Tongji University, Shanghai, 200092, China
- Department of Cardiology, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Qicheng Zou
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Department of Cardiology, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Ke Xiong
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Department of Cardiology, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Zhiwen Chen
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Department of Pathology and Pathophysiology, School of Medicine, Tongji University, Shanghai, 200092, China
- Department of Cardiology, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Beihua Shao
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Department of Pathology and Pathophysiology, School of Medicine, Tongji University, Shanghai, 200092, China
- Department of Cardiology, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Yi Liu
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Department of Cardiology, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Duanyang Xie
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
- Department of Pathology and Pathophysiology, School of Medicine, Tongji University, Shanghai, 200092, China.
- Department of Cardiology, School of Medicine, Tongji University, Shanghai, 200120, China.
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| | - Yong Ji
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
3
|
Saljic A, Heijman J, Dobrev D. From Atrial Small-conductance Calcium-activated Potassium Channels to New Antiarrhythmics. Eur Cardiol 2024; 19:e26. [PMID: 39872420 PMCID: PMC11770539 DOI: 10.15420/ecr.2024.41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 10/27/2024] [Indexed: 01/30/2025] Open
Abstract
Despite significant advances in its management, AF remains a major healthcare burden affecting millions of individuals. Rhythm control with antiarrhythmic drugs or catheter ablation has been shown to improve symptoms and outcomes in AF patients, but current treatment options have limited efficacy and/or significant side-effects. Novel mechanism-based approaches could potentially be more effective, enabling improved therapeutic strategies for managing AF. Small-conductance calcium-activated potassium (SK or KCa2.x) channels encoded by KCNN1-3 have recently gathered interest as novel antiarrhythmic targets with potential atrial-predominant effects. Here, the molecular composition of smallconductance calcium-activated potassium channels and their complex regulation in AF as the basis for understanding the distinct mechanism of action of pore-blockers (apamin, UCL1684, ICAGEN) and modulators of calcium-dependent activation (NS8593, AP14145, AP30663) are summarised. Furthermore, the preclinical and early clinical evidence for the role of small-conductance calcium-activated potassium channel inhibitors in the treatment of AF are reviewed.
Collapse
Affiliation(s)
- Arnela Saljic
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of CopenhagenCopenhagen, Denmark
- Institute of Pharmacology, West German Heart and Vascular Center, University of Duisburg-EssenEssen, Germany
| | - Jordi Heijman
- Gottfried Schatz Research Centre, Division of Medical Physics & Biophysics, Medical University of GrazGraz, Austria
- Department of Cardiology, Maastricht University Medical Centre and Cardiovascular Research Institute Maastricht, Maastricht UniversityMaastricht, the Netherlands
| | - Dobromir Dobrev
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of CopenhagenCopenhagen, Denmark
- Departments of Medicine and Research Centre, Montreal Heart Institute and Université de MontréalMontreal, Canada
- Department of Integrative Physiology, Baylor College of MedicineHouston, TX, US
| |
Collapse
|
4
|
Zhang X, Wu Y, Smith CER, Louch WE, Morotti S, Dobrev D, Grandi E, Ni H. Enhanced Ca 2+-Driven Arrhythmogenic Events in Female Patients With Atrial Fibrillation: Insights From Computational Modeling. JACC Clin Electrophysiol 2024; 10:2371-2391. [PMID: 39340505 PMCID: PMC11602355 DOI: 10.1016/j.jacep.2024.07.020] [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: 04/22/2024] [Revised: 07/10/2024] [Accepted: 07/29/2024] [Indexed: 09/30/2024]
Abstract
BACKGROUND Substantial sex-based differences have been reported in atrial fibrillation (AF), but the underlying mechanisms are poorly understood. OBJECTIVES This study sought to gain a mechanistic understanding of Ca2+-handling disturbances and Ca2+-driven arrhythmogenic events in male vs female atrial cardiomyocytes and establish their responses to Ca2+-targeted interventions. METHODS We integrated reported sex differences and AF-associated changes (ie, expression and phosphorylation of Ca2+-handling proteins, cardiomyocyte ultrastructural characteristics, and dimensions) into our human atrial cardiomyocyte model that couples electrophysiology with spatially detailed Ca2+-handling processes. Sex-specific responses of atrial cardiomyocytes to arrhythmia-provoking protocols and Ca2+-targeted interventions were evaluated. RESULTS Simulated quiescent cardiomyocytes showed increased incidence of Ca2+ sparks in female vs male myocytes in AF, in agreement with previous experimental reports. Additionally, our female model exhibited elevated propensity to develop pacing-induced spontaneous Ca2+ releases (SCRs) and augmented beat-to-beat variability in action potential (AP)-elicited Ca2+ transients compared with the male model. Sensitivity analysis uncovered distinct arrhythmogenic contributions of each component involved in sex and/or AF alterations. Specifically, increased ryanodine receptor phosphorylation emerged as the major SCR contributor in female AF cardiomyocytes, whereas reduced L-type Ca2+ current was protective against SCRs for male AF cardiomyocytes. Furthermore, simulated Ca2+-targeted interventions identified potential strategies (eg, t-tubule restoration, and inhibition of ryanodine receptor and sarcoplasmic/endoplasmic reticulum Ca2⁺-ATPase) to attenuate Ca2+-driven arrhythmogenic events in women, and revealed enhanced efficacy when applied in combination. CONCLUSIONS Sex-specific modeling uncovers increased Ca2+-driven arrhythmogenic events in female vs male atria in AF, and suggests combined Ca2+-targeted interventions are promising therapeutic approaches in women.
Collapse
Affiliation(s)
- Xianwei Zhang
- Department of Pharmacology, University of California-Davis, Davis, California, USA. https://twitter.com/xianweizhang1
| | - Yixuan Wu
- Department of Pharmacology, University of California-Davis, Davis, California, USA
| | - Charlotte E R Smith
- Department of Pharmacology, University of California-Davis, Davis, California, USA. https://twitter.com/Char_Smith3
| | - William E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway. https://twitter.com/IEMRLouch
| | - Stefano Morotti
- Department of Pharmacology, University of California-Davis, Davis, California, USA. https://twitter.com/MorottiLab
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany; Montréal Heart Institute, Université de Montréal, Montréal, Québec, Canada; Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas, USA. https://twitter.com/dr_dobrev
| | - Eleonora Grandi
- Department of Pharmacology, University of California-Davis, Davis, California, USA.
| | - Haibo Ni
- Department of Pharmacology, University of California-Davis, Davis, California, USA.
| |
Collapse
|
5
|
Mo W, Donahue JK. Gene therapy for atrial fibrillation. J Mol Cell Cardiol 2024; 196:84-93. [PMID: 39270930 PMCID: PMC11534567 DOI: 10.1016/j.yjmcc.2024.09.004] [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: 11/09/2023] [Revised: 08/19/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024]
Abstract
Atrial fibrillation (AF) is the most common sustained arrhythmia in adults. Current limitations of pharmacological and ablative therapies motivate the development of novel therapies as next generation treatments for AF. The arrhythmia mechanisms creating and sustaining AF are key elements in the development of this novel treatment. Gene therapy provides a useful platform that allows us to regulate the mechanisms of interest using a suitable transgene(s), vector, and delivery method. Effective gene therapy strategies in the literature have targeted maladaptive electrical or structural remodeling that increase vulnerability to AF. In this review, we will summarize key elements of gene therapy for AF, including molecular targets, gene transfer vectors, atrial gene delivery and preclinical efficacy and toxicity testing. Recent advances and challenges in the field will be also discussed.
Collapse
Affiliation(s)
- Weilan Mo
- From the Division of Cardiology, University of Massachusetts Medical School, Worcester, MA, United States of America
| | - J Kevin Donahue
- From the Division of Cardiology, University of Massachusetts Medical School, Worcester, MA, United States of America.
| |
Collapse
|
6
|
Qin X, Jin L, Gong H, Zheng Q. Electro-metabolic coupling in atrial fibrillation: A deeper understanding of the metabolic driver. Biomed Pharmacother 2024; 180:117536. [PMID: 39378681 DOI: 10.1016/j.biopha.2024.117536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/26/2024] [Accepted: 10/04/2024] [Indexed: 10/10/2024] Open
Abstract
Atrial fibrillation (AF), the most common sustained heart rhythm abnormality, disrupts the normal link between electrical activity and atrial muscle contraction; this disruption is termed "excitation-contraction uncoupling". It weakens atrial contractions and contributes to the development and persistence of AF. In addition to electrical dysfunction, AF is increasingly recognized as a metabolic disorder. Metabolic remodeling may reportedly precede electrophysiological, contractile, and structural changes in AF. Both clinical observations and experimental studies have underscored the critical importance of metabolic homeostasis, and its disturbance is considered a key initial factor in the development of AF. Research in this field has progressed, and a consensus has emerged that metabolic status (energy flux) and electrophysiological signaling (ion flux) are interactively regulated, highlighting the concept of "electro-metabolic coupling." Their uncoupling or decompensation constitutes a common pathological basis of AF. Despite growing recognition of the importance of metabolic balance, the role of electro-metabolic coupling in AF remains unclear. Thus, this review aimed to discuss 1) a comprehensive understanding of electro-metabolic alterations post-AF, 2) the pivotal role of metabolic homeostasis in AF pathogenesis, and 3) the mutual regulation of electro-metabolic signaling, along with potential therapeutic strategies targeting these imbalances.
Collapse
Affiliation(s)
- Xinghua Qin
- Xi'an Key Laboratory of Special Medicine and Health Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China.
| | - Lingyan Jin
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Haoyu Gong
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Qiangsun Zheng
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| |
Collapse
|
7
|
Bode D, Pronto JRD, Schiattarella GG, Voigt N. Metabolic remodelling in atrial fibrillation: manifestations, mechanisms and clinical implications. Nat Rev Cardiol 2024; 21:682-700. [PMID: 38816507 DOI: 10.1038/s41569-024-01038-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/22/2024] [Indexed: 06/01/2024]
Abstract
Atrial fibrillation (AF) is a continually growing health-care burden that often presents together with metabolic disorders, including diabetes mellitus and obesity. Current treatments often fall short of preventing AF and its adverse outcomes. Accumulating evidence suggests that metabolic disturbances can promote the development of AF through structural and electrophysiological remodelling, but the underlying mechanisms that predispose an individual to AF are aetiology-dependent, thus emphasizing the need for tailored therapeutic strategies to treat AF that target an individual's metabolic profile. AF itself can induce changes in glucose, lipid and ketone metabolism, mitochondrial function and myofibrillar energetics (as part of a process referred to as 'metabolic remodelling'), which can all contribute to atrial dysfunction. In this Review, we discuss our current understanding of AF in the setting of metabolic disorders, as well as changes in atrial metabolism that are relevant to the development of AF. We also describe the potential of available and emerging treatment strategies to target metabolic remodelling in the setting of AF and highlight key questions and challenges that need to be addressed to improve outcomes in these patients.
Collapse
Affiliation(s)
- David Bode
- Max Rubner Center for Cardiovascular Metabolic Renal Research (MRC), Deutsches Herzzentrum der Charité (DHZC), Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Julius Ryan D Pronto
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Gabriele G Schiattarella
- Max Rubner Center for Cardiovascular Metabolic Renal Research (MRC), Deutsches Herzzentrum der Charité (DHZC), Charité - Universitätsmedizin Berlin, Berlin, Germany.
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.
- Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy.
| | - Niels Voigt
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Göttingen, Germany.
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany.
- Cluster of Excellence 'Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells' (MBExC), University of Göttingen, Göttingen, Germany.
| |
Collapse
|
8
|
Wang XC, Zhou Y, Chen HX, Hou HT, He GW, Yang Q. ER stress modulates Kv1.5 channels via PERK branch in HL-1 atrial myocytes: Relevance to atrial arrhythmogenesis and the effect of tetramethylpyrazine. Heliyon 2024; 10:e37767. [PMID: 39318794 PMCID: PMC11420496 DOI: 10.1016/j.heliyon.2024.e37767] [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: 04/19/2024] [Revised: 09/05/2024] [Accepted: 09/09/2024] [Indexed: 09/26/2024] Open
Abstract
Endoplasmic reticulum (ER) stress is implicated in cardiac arrhythmia whereas the associated mechanisms remain inadequately understood. Kv1.5 channels are essential for atrial repolarization. Whether ER stress affects Kv1.5 channels is unknown. This study aimed to elucidate the response of Kv1.5 channels to ER stress by clarifying the unfolded protein response (UPR) branch responsible for the channel modulation. In addition, the effect of tetramethylpyrazine (TMP) on Kv1.5 channels was studied. Patch clamp and western-blot results revealed that exposure of HL-1 atrial myocytes to ER stress inducer tunicamycin upregulates Kv1.5 expression, increases Kv1.5 channel current (I Kur ) (14.91 ± 1.11 vs. 6.11 ± 1.31 pA/pF, P < 0.001), and shortened action potential duration (APD) (APD90: 82.79 ± 5.25 vs.121.11 ± 6.72 ms, P < 0.01), which could be reverted by ER stress inhibitors. Blockade of the PERK branch while not IRE1 and ATF6 branches of UPR downregulated Kv1.5 expression, accompanied by a decreased I Kur (9.03 ± 0.99 pA/pF) and a prolonged APD90 (113.69 ± 4.41 ms) (P < 0.01). PERK-mediated increases of Kv1.5 expression and I Kur were also observed in HL-1 cells incubated with thapsigargin. TMP suppressed the enhancement of I Kur (10.52 ± 0.97 vs. 17.52 ± 2.25 pA/pF, P < 0.05), prevented the shortening of APD (APD90: 110.16 ± 5.36 vs. 84.84 ± 4.58 ms, P < 0.05), and inhibited the upregulation of Kv1.5 triggered by ER stress. Our study suggests that ER stress induces upregulation and activation of Kv1.5 channels in atrial myocytes through the PERK branch of UPR. TMP prevents Kv1.5 upregulation/activation and the resultant APD shortening by inhibiting ER stress. These results may shed light on the mechanisms of atrial arrhythmogenesis and the antiarrhythmic effect of the traditional Chinese herb TMP.
Collapse
Affiliation(s)
- Xiang-Chong Wang
- Institute of Cardiovascular Diseases & Department of Cardiac Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College & Tianjin University, Tianjin, 300457, China
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin, China
- Department of Pharmacology, Hebei Higher Education Institute Applied Technology Research Center on TCM Formula Preparation, Hebei International Cooperation Center for Ion channel Function and Innovative Traditional Chinese Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050091, China
- School of Medicine, Nankai University, Tianjin, 300457, China
| | - Yang Zhou
- Institute of Cardiovascular Diseases & Department of Cardiac Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College & Tianjin University, Tianjin, 300457, China
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin, China
| | - Huan-Xin Chen
- Institute of Cardiovascular Diseases & Department of Cardiac Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College & Tianjin University, Tianjin, 300457, China
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin, China
| | - Hai-Tao Hou
- Institute of Cardiovascular Diseases & Department of Cardiac Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College & Tianjin University, Tianjin, 300457, China
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin, China
| | - Guo-Wei He
- Institute of Cardiovascular Diseases & Department of Cardiac Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College & Tianjin University, Tianjin, 300457, China
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin, China
| | - Qin Yang
- Institute of Cardiovascular Diseases & Department of Cardiac Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College & Tianjin University, Tianjin, 300457, China
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin, China
| |
Collapse
|
9
|
Mazhar F, Bartolucci C, Regazzoni F, Paci M, Dedè L, Quarteroni A, Corsi C, Severi S. A detailed mathematical model of the human atrial cardiomyocyte: integration of electrophysiology and cardiomechanics. J Physiol 2024; 602:4543-4583. [PMID: 37641426 DOI: 10.1113/jp283974] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 08/02/2023] [Indexed: 08/31/2023] Open
Abstract
Mechano-electric regulations (MER) play an important role in the maintenance of cardiac performance. Mechano-calcium and mechano-electric feedback (MCF and MEF) pathways adjust the cardiomyocyte contractile force according to mechanical perturbations and affects electro-mechanical coupling. MER integrates all these regulations in one unit resulting in a complex phenomenon. Computational modelling is a useful tool to accelerate the mechanistic understanding of complex experimental phenomena. We have developed a novel model that integrates the MER loop for human atrial cardiomyocytes with proper consideration of feedforward and feedback pathways. The model couples a modified version of the action potential (AP) Koivumäki model with the contraction model by Quarteroni group. The model simulates iso-sarcometric and isometric twitches and the feedback effects on AP and Ca2+-handling. The model showed a biphasic response of Ca2+ transient (CaT) peak to increasing pacing rates and highlights the possible mechanisms involved. The model has shown a shift of the threshold for AP and CaT alternans from 4.6 to 4 Hz under post-operative atrial fibrillation, induced by depressed SERCA activity. The alternans incidence was dependent on a chain of mechanisms including RyRs availability time, MCF coupling, CaMKII phosphorylation, and the stretch levels. As a result, the model predicted a 10% slowdown of conduction velocity for a 20% stretch, suggesting a role of stretch in creation of substrate formation for atrial fibrillation. Overall, we conclude that the developed model provides a physiological CaT followed by a physiological twitch. This model can open pathways for the future studies of human atrial electromechanics. KEY POINTS: With the availability of human atrial cellular data, interest in atrial-specific model integration has been enhanced. We have developed a detailed mathematical model of human atrial cardiomyocytes including the mechano-electric regulatory loop. The model has gone through calibration and evaluation phases against a wide collection of available human in-vitro data. The usefulness of the model for analysing clinical problems has been preliminaryly tested by simulating the increased incidence of Ca2+ transient and action potential alternans at high rates in post-operative atrial fibrillation condition. The model determines the possible role of mechano-electric feedback in alternans incidence, which can increase vulnerability to atrial arrhythmias by varying stretch levels. We found that our physiologically accurate description of Ca2+ handling can reproduce many experimental phenomena and can help to gain insights into the underlying pathophysiological mechanisms.
Collapse
Affiliation(s)
- Fazeelat Mazhar
- Department of Electrical, Electronic and Information Engineering 'Guglielmo Marconi', University of Bologna, Cesena, Italy
| | - Chiara Bartolucci
- Department of Electrical, Electronic and Information Engineering 'Guglielmo Marconi', University of Bologna, Cesena, Italy
| | | | - Michelangelo Paci
- Department of Electrical, Electronic and Information Engineering 'Guglielmo Marconi', University of Bologna, Cesena, Italy
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Luca Dedè
- MOX - Dipartimento di Matematica, Politecnico di Milano, Milan, Italy
| | - Alfio Quarteroni
- MOX - Dipartimento di Matematica, Politecnico di Milano, Milan, Italy
- Mathematics Institute, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Cristiana Corsi
- Department of Electrical, Electronic and Information Engineering 'Guglielmo Marconi', University of Bologna, Cesena, Italy
| | - Stefano Severi
- Department of Electrical, Electronic and Information Engineering 'Guglielmo Marconi', University of Bologna, Cesena, Italy
| |
Collapse
|
10
|
Kohajda Z, Corici C, Kristóf A, Virág L, Husti Z, Baczkó I, Sághy L, Varró A, Jost N. The Properties of the Transient Outward, Inward Rectifier and Acetylcholine-Sensitive Potassium Currents in Atrial Myocytes from Dogs in Sinus Rhythm and Experimentally Induced Atrial Fibrillation Dog Models. Pharmaceuticals (Basel) 2024; 17:1138. [PMID: 39338302 PMCID: PMC11434634 DOI: 10.3390/ph17091138] [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: 05/27/2024] [Revised: 07/26/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
AIMS Atrial fibrillation (AF) is the most common chronic/recurrent arrhythmia, which significantly impairs quality of life and increases cardiovascular morbidity and mortality. Therefore, the aim of the present study was to investigate the properties of three repolarizing potassium currents which were shown to contribute to AF-induced electrical remodeling, i.e., the transient outward (Ito), inward rectifier (IK1) and acetylcholine-sensitive (IK,ACh) potassium currents in isolated atrial myocytes obtained from dogs either with sinus rhythm (SR) or following chronic atrial tachypacing (400/min)-induced AF. METHODS Atrial remodeling and AF were induced by chronic (4-6 weeks of) right atrial tachypacing (400/min) in dogs. Transmembrane ionic currents were measured by applying the whole-cell patch-clamp technique at 37 °C. RESULTS The Ito current was slightly downregulated in AF cells when compared with that recorded in SR cells. This downregulation was also associated with slowed inactivation kinetics. The IK1 current was found to be larger in AF cells; however, this upregulation was not statistically significant in the voltage range corresponding with atrial action potential (-80 mV to 0 mV). IK,ACh was activated by the cholinergic agonist carbachol (CCh; 2 µM). In SR, CCh activated a large current either in inward or outward directions. The selective IK,ACh inhibitor tertiapin (10 nM) blocked the outward CCh-induced current by 61%. In atrial cardiomyocytes isolated from dogs with AF, the presence of a constitutively active IK,ACh was observed, blocked by 59% with 10 nM tertiapin. However, in "AF atrial myocytes", CCh activated an additional, significant ligand-dependent and tertiapin-sensitive IK,ACh current. CONCLUSIONS In our dog AF model, Ito unlike in humans was downregulated only in a slight manner. Due to its slow inactivation kinetics, it seems that Ito may play a more significant role in atrial repolarization than in ventricular working muscle myocytes. The presence of the constitutively active IK,ACh in atrial myocytes from AF dogs shows that electrical remodeling truly developed in this model. The IK,ACh current (both ligand-dependent and constitutively active) seems to play a significant role in canine atrial electrical remodeling and may be a promising atrial selective drug target for suppressing AF.
Collapse
Affiliation(s)
- Zsófia Kohajda
- HUN-REN-SZTE Research Group of Cardiovascular Pharmacology, H-6701 Szeged, Hungary
| | - Claudia Corici
- Department of Pharmacology & Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 12, P.O. Box 427, H-6701 Szeged, Hungary (I.B.)
| | - Attila Kristóf
- Department of Pharmacology & Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 12, P.O. Box 427, H-6701 Szeged, Hungary (I.B.)
| | - László Virág
- Department of Pharmacology & Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 12, P.O. Box 427, H-6701 Szeged, Hungary (I.B.)
- Pharmaceutical and Medical Device Developments Competence Centre of the Life Sciences Cluster, Centre of Excellence for Interdisciplinary Research, Development and Innovation, University of Szeged, H-6701 Szeged, Hungary
| | - Zoltán Husti
- Department of Pharmacology & Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 12, P.O. Box 427, H-6701 Szeged, Hungary (I.B.)
| | - István Baczkó
- Department of Pharmacology & Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 12, P.O. Box 427, H-6701 Szeged, Hungary (I.B.)
- Pharmaceutical and Medical Device Developments Competence Centre of the Life Sciences Cluster, Centre of Excellence for Interdisciplinary Research, Development and Innovation, University of Szeged, H-6701 Szeged, Hungary
| | - László Sághy
- Cardiac Electrophysiology Division, Department of Internal Medicine, Albert Szent-Györgyi Medical School, University of Szeged, H-6701 Szeged, Hungary
| | - András Varró
- HUN-REN-SZTE Research Group of Cardiovascular Pharmacology, H-6701 Szeged, Hungary
- Department of Pharmacology & Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 12, P.O. Box 427, H-6701 Szeged, Hungary (I.B.)
- Pharmaceutical and Medical Device Developments Competence Centre of the Life Sciences Cluster, Centre of Excellence for Interdisciplinary Research, Development and Innovation, University of Szeged, H-6701 Szeged, Hungary
| | - Norbert Jost
- HUN-REN-SZTE Research Group of Cardiovascular Pharmacology, H-6701 Szeged, Hungary
- Department of Pharmacology & Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 12, P.O. Box 427, H-6701 Szeged, Hungary (I.B.)
- Pharmaceutical and Medical Device Developments Competence Centre of the Life Sciences Cluster, Centre of Excellence for Interdisciplinary Research, Development and Innovation, University of Szeged, H-6701 Szeged, Hungary
| |
Collapse
|
11
|
Fakuade FE, Hubricht D, Möller V, Sobitov I, Liutkute A, Döring Y, Seibertz F, Gerloff M, Pronto JRD, Haghighi F, Brandenburg S, Alhussini K, Ignatyeva N, Bonhoff Y, Kestel S, El-Essawi A, Jebran AF, Großmann M, Danner BC, Baraki H, Schmidt C, Sossalla S, Kutschka I, Bening C, Maack C, Linke WA, Heijman J, Lehnart SE, Kensah G, Ebert A, Mason FE, Voigt N. Impaired Intracellular Calcium Buffering Contributes to the Arrhythmogenic Substrate in Atrial Myocytes From Patients With Atrial Fibrillation. Circulation 2024; 150:544-559. [PMID: 38910563 PMCID: PMC11319087 DOI: 10.1161/circulationaha.123.066577] [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: 08/01/2023] [Accepted: 05/31/2024] [Indexed: 06/25/2024]
Abstract
BACKGROUND Alterations in the buffering of intracellular Ca2+, for which myofilament proteins play a key role, have been shown to promote cardiac arrhythmia. It is interesting that although studies report atrial myofibrillar degradation in patients with persistent atrial fibrillation (persAF), the intracellular Ca2+ buffering profile in persAF remains obscure. Therefore, we aimed to investigate the intracellular buffering of Ca2+ and its potential arrhythmogenic role in persAF. METHODS Transmembrane Ca2+ fluxes (patch-clamp) and intracellular Ca2+ signaling (fluo-3-acetoxymethyl ester) were recorded simultaneously in myocytes from right atrial biopsies of sinus rhythm (Ctrl) and patients with persAF, alongside human atrial subtype induced pluripotent stem cell-derived cardiac myocytes (iPSC-CMs). Protein levels were quantified by immunoblotting of human atrial tissue and induced pluripotent stem cell-derived cardiac myocytes. Mouse whole heart and atrial electrophysiology were measured on a Langendorff system. RESULTS Cytosolic Ca2+ buffering was decreased in atrial myocytes of patients with persAF because of a depleted amount of Ca2+ buffers. In agreement, protein levels of selected Ca2+ binding myofilament proteins, including cTnC (cardiac troponin C), a major cytosolic Ca2+ buffer, were significantly lower in patients with persAF. Small interfering RNA (siRNA)-mediated knockdown of cTnC (si-cTNC) in atrial iPSC-CM phenocopied the reduced cytosolic Ca2+ buffering observed in persAF. Si-cTnC treated atrial iPSC-CM exhibited a higher predisposition to spontaneous Ca2+ release events and developed action potential alternans at low stimulation frequencies. Last, indirect reduction of cytosolic Ca2+ buffering using blebbistatin in an ex vivo mouse whole heart model increased vulnerability to tachypacing-induced atrial arrhythmia, validating the direct mechanistic link between impaired cytosolic Ca2+ buffering and atrial arrhythmogenesis. CONCLUSIONS Our findings suggest that loss of myofilament proteins, particularly reduced cTnC protein levels, causes diminished cytosolic Ca2+ buffering in persAF, thereby potentiating the occurrence of spontaneous Ca2+ release events and atrial fibrillation susceptibility. Strategies targeting intracellular buffering may represent a promising therapeutic lead in persAF management.
Collapse
Affiliation(s)
- Funsho E. Fakuade
- Cluster of Excellence “Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells” (F.E.F., A.L., F.S., F.H., S.E.L., A.E., N.V.), Georg-August-University Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Dominik Hubricht
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Vanessa Möller
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Izzatullo Sobitov
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Aiste Liutkute
- Cluster of Excellence “Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells” (F.E.F., A.L., F.S., F.H., S.E.L., A.E., N.V.), Georg-August-University Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Yannic Döring
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Fitzwilliam Seibertz
- Cluster of Excellence “Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells” (F.E.F., A.L., F.S., F.H., S.E.L., A.E., N.V.), Georg-August-University Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Marcus Gerloff
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Julius Ryan D. Pronto
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Fereshteh Haghighi
- Cluster of Excellence “Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells” (F.E.F., A.L., F.S., F.H., S.E.L., A.E., N.V.), Georg-August-University Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Thoracic and Cardiovascular Surgery (F.H., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., G.K.), University Medical Center Göttingen, Germany
| | - Sören Brandenburg
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Cardiology and Pneumology (S.B., N.I., W.A.L., S.E.L., A.E.), Heart Research Center Göttingen, University Medical Center Göttingen, Germany
| | - Khaled Alhussini
- Department of Thoracic and Cardiovascular Surgery (K.A., C.B.), University Clinic Würzburg, Germany
- Comprehensive Heart Failure Center Würzburg (K.A., C.B., C.M.), University Clinic Würzburg, Germany
| | - Nadezda Ignatyeva
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Cardiology and Pneumology (S.B., N.I., W.A.L., S.E.L., A.E.), Heart Research Center Göttingen, University Medical Center Göttingen, Germany
| | - Yara Bonhoff
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Stefanie Kestel
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Aschraf El-Essawi
- Cluster of Excellence “Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells” (F.E.F., A.L., F.S., F.H., S.E.L., A.E., N.V.), Georg-August-University Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Thoracic and Cardiovascular Surgery (F.H., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., G.K.), University Medical Center Göttingen, Germany
- Department of Thoracic and Cardiovascular Surgery, Klinikum Braunschweig, Germany (A.E.-E.)
| | - Ahmad Fawad Jebran
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Thoracic and Cardiovascular Surgery (F.H., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., G.K.), University Medical Center Göttingen, Germany
| | - Marius Großmann
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Thoracic and Cardiovascular Surgery (F.H., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., G.K.), University Medical Center Göttingen, Germany
| | - Bernhard C. Danner
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Thoracic and Cardiovascular Surgery (F.H., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., G.K.), University Medical Center Göttingen, Germany
| | - Hassina Baraki
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Thoracic and Cardiovascular Surgery (F.H., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., G.K.), University Medical Center Göttingen, Germany
| | - Constanze Schmidt
- Department of Cardiology, University Hospital Heidelberg, Germany (C.S.)
- German Center for Cardiovascular Research Partner Site Heidelberg/Mannheim, Heidelberg University (C.S.)
| | - Samuel Sossalla
- Department of Cardiology, University Hospital Giessen & Kerckhoff Clinic, Germany (S.S.)
- Department of Cardiology, Bad Nauheim & German Center for Cardiovascular Research Partner Site Rhine-Main, Germany (S.S.)
| | - Ingo Kutschka
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Thoracic and Cardiovascular Surgery (F.H., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., G.K.), University Medical Center Göttingen, Germany
| | - Constanze Bening
- Department of Thoracic and Cardiovascular Surgery (K.A., C.B.), University Clinic Würzburg, Germany
- Comprehensive Heart Failure Center Würzburg (K.A., C.B., C.M.), University Clinic Würzburg, Germany
| | - Christoph Maack
- Comprehensive Heart Failure Center Würzburg (K.A., C.B., C.M.), University Clinic Würzburg, Germany
| | - Wolfgang A. Linke
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Cardiology and Pneumology (S.B., N.I., W.A.L., S.E.L., A.E.), Heart Research Center Göttingen, University Medical Center Göttingen, Germany
- Institute of Physiology II, University of Münster, Germany (W.A.L.)
| | - Jordi Heijman
- Gottfried Schatz Research Center, Division of Medical Physics and Biophysics, Medical University of Graz, Austria (J.H.)
- Department of Cardiology, Maastricht University Medical Centre and Cardiovascular Research Institute Maastricht, Maastricht University, The Netherlands (J.H.)
| | - Stephan E. Lehnart
- Cluster of Excellence “Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells” (F.E.F., A.L., F.S., F.H., S.E.L., A.E., N.V.), Georg-August-University Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Cardiology and Pneumology (S.B., N.I., W.A.L., S.E.L., A.E.), Heart Research Center Göttingen, University Medical Center Göttingen, Germany
| | - George Kensah
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Thoracic and Cardiovascular Surgery (F.H., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., G.K.), University Medical Center Göttingen, Germany
| | - Antje Ebert
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Cardiology and Pneumology (S.B., N.I., W.A.L., S.E.L., A.E.), Heart Research Center Göttingen, University Medical Center Göttingen, Germany
| | - Fleur E. Mason
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Niels Voigt
- Cluster of Excellence “Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells” (F.E.F., A.L., F.S., F.H., S.E.L., A.E., N.V.), Georg-August-University Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| |
Collapse
|
12
|
Medvedev RY, Afolabi SO, Turner DGP, Glukhov AV. Mechanisms of stretch-induced electro-anatomical remodeling and atrial arrhythmogenesis. J Mol Cell Cardiol 2024; 193:11-24. [PMID: 38797242 PMCID: PMC11260238 DOI: 10.1016/j.yjmcc.2024.05.011] [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: 10/31/2023] [Revised: 05/15/2024] [Accepted: 05/21/2024] [Indexed: 05/29/2024]
Abstract
Atrial fibrillation (AF) is the most common cardiac rhythm disorder, often occurring in the setting of atrial distension and elevated myocardialstretch. While various mechano-electrochemical signal transduction pathways have been linked to AF development and progression, the underlying molecular mechanisms remain poorly understood, hampering AF therapies. In this review, we describe different aspects of stretch-induced electro-anatomical remodeling as seen in animal models and in patients with AF. Specifically, we focus on cellular and molecular mechanisms that are responsible for mechano-electrochemical signal transduction and the development of ectopic beats triggering AF from pulmonary veins, the most common source of paroxysmal AF. Furthermore, we describe structural changes caused by stretch occurring before and shortly after the onset of AF as well as during AF progression, contributing to longstanding forms of AF. We also propose mechanical stretch as a new dimension to the concept "AF begets AF", in addition to underlying diseases. Finally, we discuss the mechanisms of these electro-anatomical alterations in a search for potential therapeutic strategies and the development of novel antiarrhythmic drugs targeted at the components of mechano-electrochemical signal transduction not only in cardiac myocytes, but also in cardiac non-myocyte cells.
Collapse
Affiliation(s)
- Roman Y Medvedev
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Saheed O Afolabi
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA; Department of Pharmacology and Therapeutics, University of Ilorin, Ilorin, Nigeria
| | - Daniel G P Turner
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Alexey V Glukhov
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
13
|
Li D, Liu Y, Li C, Zhou Z, Gao K, Bao H, Yang J, Xue G, Yin D, Zhao X, Shen K, Zhang L, Li J, Li C, Song J, Zhao L, Pei Y, Xuan L, Zhang Y, Lu Y, Zhang ZR, Yang B, Li Y, Pan Z. Spexin Diminishes Atrial Fibrillation Vulnerability by Acting on Galanin Receptor 2. Circulation 2024; 150:111-127. [PMID: 38726666 DOI: 10.1161/circulationaha.123.067517] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 04/15/2024] [Indexed: 07/10/2024]
Abstract
BACKGROUND G protein-coupled receptors play a critical role in atrial fibrillation (AF). Spexin is a novel ligand of galanin receptors (GALRs). In this study, we investigated the regulation of spexin and GALRs on AF and the underlying mechanisms. METHODS Global spexin knockout (SPX-KO) and cardiomyocyte-specific GALRs knockout (GALR-cKO) mice underwent burst pacing electrical stimulation. Optical mapping was used to determine atrial conduction velocity and action potential duration. Atrial myocyte action potential duration and inward rectifying K+ current (IK1) were recorded using whole-cell patch clamps. Isolated cardiomyocytes were stained with Fluo-3/AM dye, and intracellular Ca2+ handling was examined by CCD camera. A mouse model of AF was established by Ang-II (angiotensin II) infusion. RESULTS Spexin plasma levels in patients with AF were lower than those in subjects without AF, and knockout of spexin increased AF susceptibility in mice. In the atrium of SPX-KO mice, potassium inwardly rectifying channel subfamily J member 2 (KCNJ2) and sarcolipin (SLN) were upregulated; meanwhile, IK1 current was increased and Ca2+ handling was impaired in isolated atrial myocytes of SPX-KO mice. GALR2-cKO mice, but not GALR1-cKO and GALR3-cKO mice, had a higher incidence of AF, which was associated with higher IK1 current and intracellular Ca2+ overload. The phosphorylation level of CREB (cyclic AMP responsive element binding protein 1) was upregulated in atrial tissues of SPX-KO and GALR2-cKO mice. Chromatin immunoprecipitation confirmed the recruitment of p-CREB to the proximal promoter regions of KCNJ2 and SLN. Finally, spexin treatment suppressed CREB signaling, decreased IK1 current and decreased intracellular Ca2+ overload, which thus reduced the inducibility of AF in Ang-II-infused mice. CONCLUSIONS Spexin reduces atrial fibrillation susceptibility by inhibiting CREB phosphorylation and thus downregulating KCNJ2 and SLN transcription by GALR2 receptor. The spexin/GALR2/CREB signaling pathway represents a novel therapeutic avenue in the development of agents against atrial fibrillation.
Collapse
Affiliation(s)
- Desheng Li
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, International Cooperation Base for Major Cardiovascular Diseases in Cold Regions, China) College of Pharmacy (D.L., Changzhu Li, Z.Z., K.G., H.B., J.Y., K.S., L. Zhang, J.L., Chenhong Li, J.S., L. Zhao, Y.P., L.X., Y.Z., Y. Lu, B.Y., Z.P.), First Affiliated Hospital, Harbin Medical University, China
| | - Yang Liu
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Department of Cardiology (Y. Liu, D.Y., X.Z., Z.-R.Z., Y. Li, Z.P.), First Affiliated Hospital, Harbin Medical University, China
| | - Changzhu Li
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, International Cooperation Base for Major Cardiovascular Diseases in Cold Regions, China) College of Pharmacy (D.L., Changzhu Li, Z.Z., K.G., H.B., J.Y., K.S., L. Zhang, J.L., Chenhong Li, J.S., L. Zhao, Y.P., L.X., Y.Z., Y. Lu, B.Y., Z.P.), First Affiliated Hospital, Harbin Medical University, China
| | - Zhiwen Zhou
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, International Cooperation Base for Major Cardiovascular Diseases in Cold Regions, China) College of Pharmacy (D.L., Changzhu Li, Z.Z., K.G., H.B., J.Y., K.S., L. Zhang, J.L., Chenhong Li, J.S., L. Zhao, Y.P., L.X., Y.Z., Y. Lu, B.Y., Z.P.), First Affiliated Hospital, Harbin Medical University, China
| | - Kangyi Gao
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, International Cooperation Base for Major Cardiovascular Diseases in Cold Regions, China) College of Pharmacy (D.L., Changzhu Li, Z.Z., K.G., H.B., J.Y., K.S., L. Zhang, J.L., Chenhong Li, J.S., L. Zhao, Y.P., L.X., Y.Z., Y. Lu, B.Y., Z.P.), First Affiliated Hospital, Harbin Medical University, China
| | - Hairong Bao
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, International Cooperation Base for Major Cardiovascular Diseases in Cold Regions, China) College of Pharmacy (D.L., Changzhu Li, Z.Z., K.G., H.B., J.Y., K.S., L. Zhang, J.L., Chenhong Li, J.S., L. Zhao, Y.P., L.X., Y.Z., Y. Lu, B.Y., Z.P.), First Affiliated Hospital, Harbin Medical University, China
| | - Jiming Yang
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, International Cooperation Base for Major Cardiovascular Diseases in Cold Regions, China) College of Pharmacy (D.L., Changzhu Li, Z.Z., K.G., H.B., J.Y., K.S., L. Zhang, J.L., Chenhong Li, J.S., L. Zhao, Y.P., L.X., Y.Z., Y. Lu, B.Y., Z.P.), First Affiliated Hospital, Harbin Medical University, China
| | - Genlong Xue
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, China (G.X.)
| | - Dechun Yin
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Department of Cardiology (Y. Liu, D.Y., X.Z., Z.-R.Z., Y. Li, Z.P.), First Affiliated Hospital, Harbin Medical University, China
| | - Xinbo Zhao
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Department of Cardiology (Y. Liu, D.Y., X.Z., Z.-R.Z., Y. Li, Z.P.), First Affiliated Hospital, Harbin Medical University, China
| | - Kewei Shen
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, International Cooperation Base for Major Cardiovascular Diseases in Cold Regions, China) College of Pharmacy (D.L., Changzhu Li, Z.Z., K.G., H.B., J.Y., K.S., L. Zhang, J.L., Chenhong Li, J.S., L. Zhao, Y.P., L.X., Y.Z., Y. Lu, B.Y., Z.P.), First Affiliated Hospital, Harbin Medical University, China
| | - Lingmin Zhang
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, International Cooperation Base for Major Cardiovascular Diseases in Cold Regions, China) College of Pharmacy (D.L., Changzhu Li, Z.Z., K.G., H.B., J.Y., K.S., L. Zhang, J.L., Chenhong Li, J.S., L. Zhao, Y.P., L.X., Y.Z., Y. Lu, B.Y., Z.P.), First Affiliated Hospital, Harbin Medical University, China
| | - Jialiang Li
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, International Cooperation Base for Major Cardiovascular Diseases in Cold Regions, China) College of Pharmacy (D.L., Changzhu Li, Z.Z., K.G., H.B., J.Y., K.S., L. Zhang, J.L., Chenhong Li, J.S., L. Zhao, Y.P., L.X., Y.Z., Y. Lu, B.Y., Z.P.), First Affiliated Hospital, Harbin Medical University, China
| | - Chenhong Li
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, International Cooperation Base for Major Cardiovascular Diseases in Cold Regions, China) College of Pharmacy (D.L., Changzhu Li, Z.Z., K.G., H.B., J.Y., K.S., L. Zhang, J.L., Chenhong Li, J.S., L. Zhao, Y.P., L.X., Y.Z., Y. Lu, B.Y., Z.P.), First Affiliated Hospital, Harbin Medical University, China
| | - Jiahui Song
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, International Cooperation Base for Major Cardiovascular Diseases in Cold Regions, China) College of Pharmacy (D.L., Changzhu Li, Z.Z., K.G., H.B., J.Y., K.S., L. Zhang, J.L., Chenhong Li, J.S., L. Zhao, Y.P., L.X., Y.Z., Y. Lu, B.Y., Z.P.), First Affiliated Hospital, Harbin Medical University, China
| | - Lexin Zhao
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, International Cooperation Base for Major Cardiovascular Diseases in Cold Regions, China) College of Pharmacy (D.L., Changzhu Li, Z.Z., K.G., H.B., J.Y., K.S., L. Zhang, J.L., Chenhong Li, J.S., L. Zhao, Y.P., L.X., Y.Z., Y. Lu, B.Y., Z.P.), First Affiliated Hospital, Harbin Medical University, China
| | - Yao Pei
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, International Cooperation Base for Major Cardiovascular Diseases in Cold Regions, China) College of Pharmacy (D.L., Changzhu Li, Z.Z., K.G., H.B., J.Y., K.S., L. Zhang, J.L., Chenhong Li, J.S., L. Zhao, Y.P., L.X., Y.Z., Y. Lu, B.Y., Z.P.), First Affiliated Hospital, Harbin Medical University, China
| | - Lina Xuan
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, International Cooperation Base for Major Cardiovascular Diseases in Cold Regions, China) College of Pharmacy (D.L., Changzhu Li, Z.Z., K.G., H.B., J.Y., K.S., L. Zhang, J.L., Chenhong Li, J.S., L. Zhao, Y.P., L.X., Y.Z., Y. Lu, B.Y., Z.P.), First Affiliated Hospital, Harbin Medical University, China
| | - Yang Zhang
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, International Cooperation Base for Major Cardiovascular Diseases in Cold Regions, China) College of Pharmacy (D.L., Changzhu Li, Z.Z., K.G., H.B., J.Y., K.S., L. Zhang, J.L., Chenhong Li, J.S., L. Zhao, Y.P., L.X., Y.Z., Y. Lu, B.Y., Z.P.), First Affiliated Hospital, Harbin Medical University, China
| | - Yanjie Lu
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, International Cooperation Base for Major Cardiovascular Diseases in Cold Regions, China) College of Pharmacy (D.L., Changzhu Li, Z.Z., K.G., H.B., J.Y., K.S., L. Zhang, J.L., Chenhong Li, J.S., L. Zhao, Y.P., L.X., Y.Z., Y. Lu, B.Y., Z.P.), First Affiliated Hospital, Harbin Medical University, China
| | - Zhi-Ren Zhang
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Department of Cardiology (Y. Liu, D.Y., X.Z., Z.-R.Z., Y. Li, Z.P.), First Affiliated Hospital, Harbin Medical University, China
- National Health Commission Key Laboratory of Cell Transplantation (Z.-R.Z., Y. Li, Z.P.), First Affiliated Hospital, Harbin Medical University, China
| | - Baofeng Yang
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, International Cooperation Base for Major Cardiovascular Diseases in Cold Regions, China) College of Pharmacy (D.L., Changzhu Li, Z.Z., K.G., H.B., J.Y., K.S., L. Zhang, J.L., Chenhong Li, J.S., L. Zhao, Y.P., L.X., Y.Z., Y. Lu, B.Y., Z.P.), First Affiliated Hospital, Harbin Medical University, China
| | - Yue Li
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Department of Cardiology (Y. Liu, D.Y., X.Z., Z.-R.Z., Y. Li, Z.P.), First Affiliated Hospital, Harbin Medical University, China
- National Health Commission Key Laboratory of Cell Transplantation (Z.-R.Z., Y. Li, Z.P.), First Affiliated Hospital, Harbin Medical University, China
| | - Zhenwei Pan
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, International Cooperation Base for Major Cardiovascular Diseases in Cold Regions, China) College of Pharmacy (D.L., Changzhu Li, Z.Z., K.G., H.B., J.Y., K.S., L. Zhang, J.L., Chenhong Li, J.S., L. Zhao, Y.P., L.X., Y.Z., Y. Lu, B.Y., Z.P.), First Affiliated Hospital, Harbin Medical University, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Department of Cardiology (Y. Liu, D.Y., X.Z., Z.-R.Z., Y. Li, Z.P.), First Affiliated Hospital, Harbin Medical University, China
- National Health Commission Key Laboratory of Cell Transplantation (Z.-R.Z., Y. Li, Z.P.), First Affiliated Hospital, Harbin Medical University, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, 2019 Research Unit 070, Harbin, China (Z.P.)
| |
Collapse
|
14
|
Ayagama T, Charles PD, Bose SJ, Boland B, Priestman DA, Aston D, Berridge G, Fischer R, Cribbs AP, Song Q, Mirams GR, Amponsah K, Heather L, Galione A, Herring N, Kramer H, Capel RA, Platt FM, Schotten U, Verheule S, Burton RA. Compartmentalization proteomics revealed endolysosomal protein network changes in a goat model of atrial fibrillation. iScience 2024; 27:109609. [PMID: 38827406 PMCID: PMC11141153 DOI: 10.1016/j.isci.2024.109609] [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: 08/30/2023] [Revised: 02/07/2024] [Accepted: 03/25/2024] [Indexed: 06/04/2024] Open
Abstract
Endolysosomes (EL) are known for their role in regulating both intracellular trafficking and proteostasis. EL facilitate the elimination of damaged membranes, protein aggregates, membranous organelles and play an important role in calcium signaling. The specific role of EL in cardiac atrial fibrillation (AF) is not well understood. We isolated atrial EL organelles from AF goat biopsies and conducted a comprehensive integrated omics analysis to study the EL-specific proteins and pathways. We also performed electron tomography, protein and enzyme assays on these biopsies. Our results revealed the upregulation of the AMPK pathway and the expression of EL-specific proteins that were not found in whole tissue lysates, including GAA, DYNLRB1, CLTB, SIRT3, CCT2, and muscle-specific HSPB2. We also observed structural anomalies, such as autophagic-vacuole formation, irregularly shaped mitochondria, and glycogen deposition. Our results provide molecular information suggesting EL play a role in AF disease process over extended time frames.
Collapse
Affiliation(s)
- Thamali Ayagama
- Department of Pharmacology, University of Oxford, Oxford, UK
| | | | - Samuel J. Bose
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Barry Boland
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | | | - Daniel Aston
- Department of Anaesthesia and Critical Care, Royal Papworth Hospital NHS Foundation Trust, Papworth Road, Cambridge CB2 0AY, UK
| | | | - Roman Fischer
- Target Discovery Institute, University of Oxford, Oxford, UK
| | - Adam P. Cribbs
- Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Headington OX3 7LD, UK
| | - Qianqian Song
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Gary R. Mirams
- Centre for Mathematical Medicine & Biology, Mathematical Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| | - Kwabena Amponsah
- Centre for Mathematical Medicine & Biology, Mathematical Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| | - Lisa Heather
- Department of Physiology, Anatomy and Genetics, , University of Oxford, South Park Road, Oxford OX1 3PT, UK
| | - Antony Galione
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Neil Herring
- Department of Physiology, Anatomy and Genetics, , University of Oxford, South Park Road, Oxford OX1 3PT, UK
| | - Holger Kramer
- Mass spectrometry Facility, The MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | | | | | - Ulrich Schotten
- Departments of Physiology and Cardiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Sander Verheule
- Departments of Physiology and Cardiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Rebecca A.B. Burton
- Department of Pharmacology, University of Oxford, Oxford, UK
- University of Liverpool, Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, Liverpool, UK
| |
Collapse
|
15
|
Zhang X, Wu Y, Smith C, Louch WE, Morotti S, Dobrev D, Grandi E, Ni H. Enhanced Ca2+-Driven Arrhythmias in Female Patients with Atrial Fibrillation: Insights from Computational Modeling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.04.583217. [PMID: 38496584 PMCID: PMC10942295 DOI: 10.1101/2024.03.04.583217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
BACKGROUND AND AIMS Substantial sex-based differences have been reported in atrial fibrillation (AF), with female patients experiencing worse symptoms, increased complications from drug side effects or ablation, and elevated risk of AF-related stroke and mortality. Recent studies revealed sex-specific alterations in AF-associated Ca2+ dysregulation, whereby female cardiomyocytes more frequently exhibit potentially proarrhythmic Ca2+-driven instabilities compared to male cardiomyocytes. In this study, we aim to gain a mechanistic understanding of the Ca2+-handling disturbances and Ca2+-driven arrhythmogenic events in males vs females and establish their responses to Ca2+-targeted interventions. METHODS AND RESULTS We incorporated known sex differences and AF-associated changes in the expression and phosphorylation of key Ca2+-handling proteins and in ultrastructural properties and dimensions of atrial cardiomyocytes into our recently developed 3D atrial cardiomyocyte model that couples electrophysiology with spatially detailed Ca2+-handling processes. Our simulations of quiescent cardiomyocytes show increased incidence of Ca2+ sparks in female vs male myocytes in AF, in agreement with previous experimental reports. Additionally, our female model exhibited elevated propensity to develop pacing-induced spontaneous Ca2+ releases (SCRs) and augmented beat-to-beat variability in action potential (AP)-elicited Ca2+ transients compared with the male model. Parameter sensitivity analysis uncovered precise arrhythmogenic contributions of each component that was implicated in sex and/or AF alterations. Specifically, increased ryanodine receptor phosphorylation in female AF cardiomyocytes emerged as the major SCR contributor, while reduced L-type Ca2+ current was protective against SCRs for male AF cardiomyocytes. Furthermore, simulations of tentative Ca2+-targeted interventions identified potential strategies to attenuate Ca2+-driven arrhythmogenic events in female atria (e.g., t-tubule restoration, and inhibition of ryanodine receptor and sarcoplasmic/endoplasmic reticulum Ca2+-ATPase), and revealed enhanced efficacy when applied in combination. CONCLUSIONS Our sex-specific computational models of human atrial cardiomyocytes uncover increased propensity to Ca2+-driven arrhythmogenic events in female compared to male atrial cardiomyocytes in AF, and point to combined Ca2+-targeted interventions as promising approaches to treat AF in female patients. Our study establishes that AF treatment may benefit from sex-dependent strategies informed by sex-specific mechanisms.
Collapse
|
16
|
Ye T, Zhou Y, Yang J, Yu F, Song Z, Shi J, Wang L, Huang Z, Yang B, Wang X. P2X7 receptor inhibition prevents atrial fibrillation in rodent models of depression. Europace 2024; 26:euae022. [PMID: 38261756 PMCID: PMC10873709 DOI: 10.1093/europace/euae022] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 12/07/2023] [Indexed: 01/25/2024] Open
Abstract
AIMS Depression, the most prevalent psychiatric disorder, is associated with the occurrence and development of atrial fibrillation (AF). P2X7 receptor (P2X7R) activation participates in the development of depression, but little attention has been given to its role in AF. This study was to investigate the effects of P2X7R on AF in depression models. METHODS AND RESULTS Lipopolysaccharide (LPS) and chronic unpredictable stress (CUS) were carried out to induce depression in rodents. Behavioural assessments, atrial electrophysiological parameters, electrocardiogram (ECG) parameters, western blot, and histology were performed. Atrial fibrillation inducibility was increased in both LPS- and CUS-induced depression, along with the up-regulation of P2X7R in atria. CUS facilitated atrial fibrosis. CUS reduced heart rate variability (HRV) and increased the expression of TH and GAP43, representing autonomic dysfunction. Down-regulation of Nav1.5, Cav1.2, Kv1.5, Kv4.3, Cx40, and Cx43 in CUS indicated the abnormalities in ion channels. In addition, the expression levels of TLR4, P65, P-P65, NLRP3, ASC, caspase-1, and IL-1β were elevated in depression models. Pharmacological inhibitor (Brilliant Blue G, BBG) or genetic deficiency of P2X7R significantly mitigated depressive-like behaviours; ameliorated electrophysiological deterioration and autonomic dysfunction; improved ion channel expression and atrial fibrosis; and prevented atrial NLRP3 inflammasome activation in the pathophysiological process of AF in depression models. CONCLUSION LPS or CUS induces AF and promotes P2X7R-dependent activation of NLRP3 inflammasome, whereas pharmacological P2X7R inhibition or P2X7R genetic deficiency prevents atrial remodelling without interrupting normal atrial physiological functions. Our results point to P2X7R as an important factor in the pathology of AF in depression.
Collapse
Affiliation(s)
- Tianxin Ye
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, #79 Qingchun Road, Hangzhou, Zhejiang Province 310003, PR China
| | - Yunping Zhou
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, #79 Qingchun Road, Hangzhou, Zhejiang Province 310003, PR China
| | - Jinxiu Yang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, #79 Qingchun Road, Hangzhou, Zhejiang Province 310003, PR China
| | - Fangcong Yu
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, #79 Qingchun Road, Hangzhou, Zhejiang Province 310003, PR China
| | - Zhuonan Song
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, #79 Qingchun Road, Hangzhou, Zhejiang Province 310003, PR China
| | - Jiaran Shi
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, #79 Qingchun Road, Hangzhou, Zhejiang Province 310003, PR China
| | - Longbo Wang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, #79 Qingchun Road, Hangzhou, Zhejiang Province 310003, PR China
| | - Zhouqing Huang
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, 2 Fuxue Road, Wenzhou, Zhejiang Province 325000, PR China
| | - Bo Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan 430060, PR China
- Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan 430060, PR China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Xingxiang Wang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, #79 Qingchun Road, Hangzhou, Zhejiang Province 310003, PR China
| |
Collapse
|
17
|
Seibertz F, Rubio T, Springer R, Popp F, Ritter M, Liutkute A, Bartelt L, Stelzer L, Haghighi F, Pietras J, Windel H, Pedrosa NDI, Rapedius M, Doering Y, Solano R, Hindmarsh R, Shi R, Tiburcy M, Bruegmann T, Kutschka I, Streckfuss-Bömeke K, Kensah G, Cyganek L, Zimmermann WH, Voigt N. Atrial fibrillation-associated electrical remodelling in human induced pluripotent stem cell-derived atrial cardiomyocytes: a novel pathway for antiarrhythmic therapy development. Cardiovasc Res 2023; 119:2623-2637. [PMID: 37677054 PMCID: PMC10730244 DOI: 10.1093/cvr/cvad143] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 07/18/2023] [Accepted: 08/03/2023] [Indexed: 09/09/2023] Open
Abstract
AIMS Atrial fibrillation (AF) is associated with tachycardia-induced cellular electrophysiology alterations which promote AF chronification and treatment resistance. Development of novel antiarrhythmic therapies is hampered by the absence of scalable experimental human models that reflect AF-associated electrical remodelling. Therefore, we aimed to assess if AF-associated remodelling of cellular electrophysiology can be simulated in human atrial-like cardiomyocytes derived from induced pluripotent stem cells in the presence of retinoic acid (iPSC-aCM), and atrial-engineered human myocardium (aEHM) under short term (24 h) and chronic (7 days) tachypacing (TP). METHODS AND RESULTS First, 24-h electrical pacing at 3 Hz was used to investigate whether AF-associated remodelling in iPSC-aCM and aEHM would ensue. Compared to controls (24 h, 1 Hz pacing) TP-stimulated iPSC-aCM presented classical hallmarks of AF-associated remodelling: (i) decreased L-type Ca2+ current (ICa,L) and (ii) impaired activation of acetylcholine-activated inward-rectifier K+ current (IK,ACh). This resulted in action potential shortening and an absent response to the M-receptor agonist carbachol in both iPSC-aCM and aEHM subjected to TP. Accordingly, mRNA expression of the channel-subunit Kir3.4 was reduced. Selective IK,ACh blockade with tertiapin reduced basal inward-rectifier K+ current only in iPSC-aCM subjected to TP, thereby unmasking an agonist-independent constitutively active IK,ACh. To allow for long-term TP, we developed iPSC-aCM and aEHM expressing the light-gated ion-channel f-Chrimson. The same hallmarks of AF-associated remodelling were observed after optical-TP. In addition, continuous TP (7 days) led to (i) increased amplitude of inward-rectifier K+ current (IK1), (ii) hyperpolarization of the resting membrane potential, (iii) increased action potential-amplitude and upstroke velocity as well as (iv) reversibly impaired contractile function in aEHM. CONCLUSIONS Classical hallmarks of AF-associated remodelling were mimicked through TP of iPSC-aCM and aEHM. The use of the ultrafast f-Chrimson depolarizing ion channel allowed us to model the time-dependence of AF-associated remodelling in vitro for the first time. The observation of electrical remodelling with associated reversible contractile dysfunction offers a novel platform for human-centric discovery of antiarrhythmic therapies.
Collapse
Affiliation(s)
- Fitzwilliam Seibertz
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Cluster of Excellence ‘Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells’ (MBExC), University of Göttingen, Göttingen, Germany
| | - Tony Rubio
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
| | - Robin Springer
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
| | - Fiona Popp
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
| | - Melanie Ritter
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
| | - Aiste Liutkute
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
| | - Lena Bartelt
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
| | - Lea Stelzer
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
| | - Fereshteh Haghighi
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Department of Cardiothoracic and Vascular Surgery, Georg-August-University Göttingen, Göttingen, Germany
| | - Jan Pietras
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Department of Cardiothoracic and Vascular Surgery, Georg-August-University Göttingen, Göttingen, Germany
| | - Hendrik Windel
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Department of Cardiothoracic and Vascular Surgery, Georg-August-University Göttingen, Göttingen, Germany
| | - Núria Díaz i Pedrosa
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
| | | | - Yannic Doering
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
| | - Richard Solano
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Department of Cardiothoracic and Vascular Surgery, Georg-August-University Göttingen, Göttingen, Germany
| | - Robin Hindmarsh
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Georg-August University Göttingen, Germany
| | - Runzhu Shi
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Institute for Cardiovascular Physiology, University Medical Center Göttingen, Göttingen, Germany
| | - Malte Tiburcy
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
| | - Tobias Bruegmann
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Cluster of Excellence ‘Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells’ (MBExC), University of Göttingen, Göttingen, Germany
- Institute for Cardiovascular Physiology, University Medical Center Göttingen, Göttingen, Germany
| | - Ingo Kutschka
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Department of Cardiothoracic and Vascular Surgery, Georg-August-University Göttingen, Göttingen, Germany
| | - Katrin Streckfuss-Bömeke
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Georg-August University Göttingen, Germany
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
| | - George Kensah
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Department of Cardiothoracic and Vascular Surgery, Georg-August-University Göttingen, Göttingen, Germany
| | - Lukas Cyganek
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Cluster of Excellence ‘Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells’ (MBExC), University of Göttingen, Göttingen, Germany
- Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Georg-August University Göttingen, Germany
| | - Wolfram H Zimmermann
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Cluster of Excellence ‘Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells’ (MBExC), University of Göttingen, Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Göttingen, Germany
- Campus-Institute Data Science (CIDAS), University of Göttingen, Göttingen, Germany
| | - Niels Voigt
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany
- Cluster of Excellence ‘Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells’ (MBExC), University of Göttingen, Göttingen, Germany
| |
Collapse
|
18
|
Ávila G. Fluorofenidone enhances cardiac contractility by stimulating CICR and Ca V1.2. Biochem Biophys Res Commun 2023; 681:242-248. [PMID: 37788591 DOI: 10.1016/j.bbrc.2023.09.079] [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: 08/30/2023] [Accepted: 09/25/2023] [Indexed: 10/05/2023]
Abstract
Fluorofenidone (AKF-PD) is a novel pyridone derivative that inhibits fibrosis and inflammation in many tissues. Accordingly, it has been effective in disease models, such as liver failure, nephropathy, and pulmonary fibrosis. However, its potential role in cardiac physiology and pathology has yet to be elucidated. Thus, this paper investigated a possible functional impact of AKF-PD on adult rat cardiac myocytes. Cells were kept in culture for 1-2 days under either control conditions or the presence of AKF-PD (500 μM). They were next examined concerning cell contractility, intracellular Ca2+ homeostasis, and activity of voltage-gated Ca2+ channels. Remarkably, AKF-PD enhanced the percentage of cell shortening and rates of both contraction and relaxation by nearly 100%. A stimulus in Ca2+-induced Ca2+ release (CICR) most likely accounts for these effects because AKF-PD also increased the magnitude of electrically evoked Ca2+ transients. Of note, the compound did not alter the peak value of caffeine-elicited Ca2+ transients, indicating stimulation of CICR at constant sarcoplasmic reticulum Ca2+ load. Since CICR is triggered by the entry of Ca2+ through CaV1.2 (ICa), a possible effect on these Ca2+ channels was also investigated. AKF-PD increased the magnitude of both ICa and maximal macroscopic Ca2+ conductance (Gmax) by about 50%. However, no differences were found in either voltage dependence of inactivation or the amount of maximal immobilization-resistant charge movement (Qmax). Thus, the effect on ICa could be explained by a higher channel's open probability (Po) rather than a greater abundance of channel proteins. Additional data indicate that AKF-PD reduces the rate of Ca2+ extrusion in the presence of caffeine, suggesting inhibition of the Na/Ca exchanger. Overall, these results indicate that AKF-PD upregulates the Po of CaV1.2 and then sequentially enhances ICa, CICR, and contractility. Therefore, the novel compound is also a candidate to be tested in cardiac disease models.
Collapse
Affiliation(s)
- Guillermo Ávila
- Departamento de Bioquímica, Cinvestav-IPN, AP 14-740, México City, 07000, México.
| |
Collapse
|
19
|
Ni H, Morotti S, Zhang X, Dobrev D, Grandi E. Integrative human atrial modelling unravels interactive protein kinase A and Ca2+/calmodulin-dependent protein kinase II signalling as key determinants of atrial arrhythmogenesis. Cardiovasc Res 2023; 119:2294-2311. [PMID: 37523735 PMCID: PMC11318383 DOI: 10.1093/cvr/cvad118] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/18/2023] [Accepted: 06/05/2023] [Indexed: 08/02/2023] Open
Abstract
AIMS Atrial fibrillation (AF), the most prevalent clinical arrhythmia, is associated with atrial remodelling manifesting as acute and chronic alterations in expression, function, and regulation of atrial electrophysiological and Ca2+-handling processes. These AF-induced modifications crosstalk and propagate across spatial scales creating a complex pathophysiological network, which renders AF resistant to existing pharmacotherapies that predominantly target transmembrane ion channels. Developing innovative therapeutic strategies requires a systems approach to disentangle quantitatively the pro-arrhythmic contributions of individual AF-induced alterations. METHODS AND RESULTS Here, we built a novel computational framework for simulating electrophysiology and Ca2+-handling in human atrial cardiomyocytes and tissues, and their regulation by key upstream signalling pathways [i.e. protein kinase A (PKA), and Ca2+/calmodulin-dependent protein kinase II (CaMKII)] involved in AF-pathogenesis. Populations of atrial cardiomyocyte models were constructed to determine the influence of subcellular ionic processes, signalling components, and regulatory networks on atrial arrhythmogenesis. Our results reveal a novel synergistic crosstalk between PKA and CaMKII that promotes atrial cardiomyocyte electrical instability and arrhythmogenic triggered activity. Simulations of heterogeneous tissue demonstrate that this cellular triggered activity is further amplified by CaMKII- and PKA-dependent alterations of tissue properties, further exacerbating atrial arrhythmogenesis. CONCLUSIONS Our analysis reveals potential mechanisms by which the stress-associated adaptive changes turn into maladaptive pro-arrhythmic triggers at the cellular and tissue levels and identifies potential anti-AF targets. Collectively, our integrative approach is powerful and instrumental to assemble and reconcile existing knowledge into a systems network for identifying novel anti-AF targets and innovative approaches moving beyond the traditional ion channel-based strategy.
Collapse
Affiliation(s)
- Haibo Ni
- Department of Pharmacology, University of California Davis,
451 Health Sciences Drive, Davis, CA 95616, USA
| | - Stefano Morotti
- Department of Pharmacology, University of California Davis,
451 Health Sciences Drive, Davis, CA 95616, USA
| | - Xianwei Zhang
- Department of Pharmacology, University of California Davis,
451 Health Sciences Drive, Davis, CA 95616, USA
| | - Dobromir Dobrev
- Institute of Pharmacology, Faculty of Medicine, University
Duisburg-Essen, Essen, Germany
- Department of Medicine and Research Center, Montreal Heart Institute and
Université de Montréal, Montréal, Canada
- Department of Molecular Physiology and Biophysics, Baylor College of
Medicine, Houston, TX, USA
| | - Eleonora Grandi
- Department of Pharmacology, University of California Davis,
451 Health Sciences Drive, Davis, CA 95616, USA
| |
Collapse
|
20
|
Zhan Y, Yue H, Zhao X, Tang J, Wu Z. Colchicine in atrial fibrillation: are old trees in bloom? Front Physiol 2023; 14:1260774. [PMID: 37916222 PMCID: PMC10616799 DOI: 10.3389/fphys.2023.1260774] [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: 07/18/2023] [Accepted: 09/26/2023] [Indexed: 11/03/2023] Open
Abstract
Colchicine is a widely used drug that was originally used to treat gout and rheumatic diseases. In recent years, colchicine has shown high potential in the cardiovascular field. Atrial fibrillation (AF) is a cardiovascular disease with a high incidence. One of the most frequent complications following cardiovascular surgery is postoperative atrial fibrillation (POAF), which affects patient health and disease burden. This article reviews the research status of colchicine in AF and summarizes the relevant progress.
Collapse
Affiliation(s)
- Yujia Zhan
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Honghua Yue
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xueshan Zhao
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Juan Tang
- Acupuncture and Moxibustion School of Teaching, Hospital of Chengdu, University of Traditional Chinese Medicine, Tianjin, China
- Key Laboratory of Emergency and Trauma, Ministry of Education, Hainan Medical University, Haikou, China
| | - Zhong Wu
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
21
|
Zhang X, Smith CER, Morotti S, Edwards AG, Sato D, Louch WE, Ni H, Grandi E. Mechanisms of spontaneous Ca 2+ release-mediated arrhythmia in a novel 3D human atrial myocyte model: II. Ca 2+ -handling protein variation. J Physiol 2023; 601:2685-2710. [PMID: 36114707 PMCID: PMC10017376 DOI: 10.1113/jp283602] [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: 07/18/2022] [Accepted: 09/02/2022] [Indexed: 11/08/2022] Open
Abstract
Disruption of the transverse-axial tubule system (TATS) in diseases such as heart failure and atrial fibrillation occurs in combination with changes in the expression and distribution of key Ca2+ -handling proteins. Together this ultrastructural and ionic remodelling is associated with aberrant Ca2+ cycling and electrophysiological instabilities that underlie arrhythmic activity. However, due to the concurrent changes in TATs and Ca2+ -handling protein expression and localization that occur in disease it is difficult to distinguish their individual contributions to the arrhythmogenic state. To investigate this, we applied our novel 3D human atrial myocyte model with spatially detailed Ca2+ diffusion and TATS to investigate the isolated and interactive effects of changes in expression and localization of key Ca2+ -handling proteins and variable TATS density on Ca2+ -handling abnormality driven membrane instabilities. We show that modulating the expression and distribution of the sodium-calcium exchanger, ryanodine receptors and the sarcoplasmic reticulum (SR) Ca2+ buffer calsequestrin have varying pro- and anti-arrhythmic effects depending on the balance of opposing influences on SR Ca2+ leak-load and Ca2+ -voltage relationships. Interestingly, the impact of protein remodelling on Ca2+ -driven proarrhythmic behaviour varied dramatically depending on TATS density, with intermediately tubulated cells being more severely affected compared to detubulated and densely tubulated myocytes. This work provides novel mechanistic insight into the distinct and interactive consequences of TATS and Ca2+ -handling protein remodelling that underlies dysfunctional Ca2+ cycling and electrophysiological instability in disease. KEY POINTS: In our companion paper we developed a 3D human atrial myocyte model, coupling electrophysiology and Ca2+ handling with subcellular spatial details governed by the transverse-axial tubule system (TATS). Here we utilize this model to mechanistically examine the impact of TATS loss and changes in the expression and distribution of key Ca2+ -handling proteins known to be remodelled in disease on Ca2+ homeostasis and electrophysiological stability. We demonstrate that varying the expression and localization of these proteins has variable pro- and anti-arrhythmic effects with outcomes displaying dependence on TATS density. Whereas detubulated myocytes typically appear unaffected and densely tubulated cells seem protected, the arrhythmogenic effects of Ca2+ handling protein remodelling are profound in intermediately tubulated cells. Our work shows the interaction between TATS and Ca2+ -handling protein remodelling that underlies the Ca2+ -driven proarrhythmic behaviour observed in atrial fibrillation and may help to predict the effects of antiarrhythmic strategies at varying stages of ultrastructural remodelling.
Collapse
Affiliation(s)
- Xianwei Zhang
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | | | - Stefano Morotti
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | | | - Daisuke Sato
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - William E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Haibo Ni
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - Eleonora Grandi
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| |
Collapse
|
22
|
Zhang X, Ni H, Morotti S, Smith C, Sato D, Louch W, Edwards A, Grandi E. Mechanisms of spontaneous Ca 2+ release-mediated arrhythmia in a novel 3D human atrial myocyte model: I. Transverse-axial tubule variation. J Physiol 2023; 601:2655-2683. [PMID: 36094888 PMCID: PMC10008525 DOI: 10.1113/jp283363] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 09/02/2022] [Indexed: 11/08/2022] Open
Abstract
Intracellular calcium (Ca2+ ) cycling is tightly regulated in the healthy heart ensuring effective contraction. This is achieved by transverse (t)-tubule membrane invaginations that facilitate close coupling of key Ca2+ -handling proteins such as the L-type Ca2+ channel and Na+ -Ca2+ exchanger (NCX) on the cell surface with ryanodine receptors (RyRs) on the intracellular Ca2+ store. Although less abundant and regular than in the ventricle, t-tubules also exist in atrial myocytes as a network of transverse invaginations with axial extensions known as the transverse-axial tubule system (TATS). In heart failure and atrial fibrillation, there is TATS remodelling that is associated with aberrant Ca2+ -handling and Ca2+ -induced arrhythmic activity; however, the mechanism underlying this is not fully understood. To address this, we developed a novel 3D human atrial myocyte model that couples electrophysiology and Ca2+ -handling with variable TATS organization and density. We extensively parameterized and validated our model against experimental data to build a robust tool examining TATS regulation of subcellular Ca2+ release. We found that varying TATS density and thus the localization of key Ca2+ -handling proteins has profound effects on Ca2+ handling. Following TATS loss, there is reduced NCX that results in increased cleft Ca2+ concentration through decreased Ca2+ extrusion. This elevated Ca2+ increases RyR open probability causing spontaneous Ca2+ releases and the promotion of arrhythmogenic waves (especially in the cell interior) leading to voltage instabilities through delayed afterdepolarizations. In summary, the present study demonstrates a mechanistic link between TATS remodelling and Ca2+ -driven proarrhythmic behaviour that probably reflects the arrhythmogenic state observed in disease. KEY POINTS: Transverse-axial tubule systems (TATS) modulate Ca2+ handling and excitation-contraction coupling in atrial myocytes, with TATS remodelling in heart failure and atrial fibrillation being associated with altered Ca2+ cycling and subsequent arrhythmogenesis. To investigate the poorly understood mechanisms linking TATS variation and spontaneous Ca2+ release, we built, parameterized and validated a 3D human atrial myocyte model coupling electrophysiology and spatially-detailed subcellular Ca2+ handling governed by the TATS. Simulated TATS loss causes diastolic Ca2+ and voltage instabilities through reduced Na+ -Ca2+ exchanger-mediated Ca2+ removal, cleft Ca2+ accumulation and increased ryanodine receptor open probability, resulting in spontaneous Ca2+ release and promotion of arrhythmogenic waves and delayed afterdepolarizations. At fast electrical rates typical of atrial tachycardia/fibrillation, spontaneous Ca2+ releases are larger and more frequent in the cell interior than at the periphery. Our work provides mechanistic insight into how atrial TATS remodelling can lead to Ca2+ -driven instabilities that may ultimately contribute to the arrhythmogenic state in disease.
Collapse
Affiliation(s)
- X. Zhang
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - H. Ni
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - S. Morotti
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - C.E.R. Smith
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - D. Sato
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - W.E. Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo Norway
| | - A.G. Edwards
- Department of Pharmacology, University of California Davis, Davis, CA, USA
- Simula Research Laboratory, Lysaker, Norway
| | - E. Grandi
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| |
Collapse
|
23
|
Schulz C, Lemoine MD, Mearini G, Koivumäki J, Sani J, Schwedhelm E, Kirchhof P, Ghalawinji A, Stoll M, Hansen A, Eschenhagen T, Christ T. PITX2 Knockout Induces Key Findings of Electrical Remodeling as Seen in Persistent Atrial Fibrillation. Circ Arrhythm Electrophysiol 2023; 16:e011602. [PMID: 36763906 DOI: 10.1161/circep.122.011602] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
BACKGROUND Electrical remodeling in human persistent atrial fibrillation is believed to result from rapid electrical activation of the atria, but underlying genetic causes may contribute. Indeed, common gene variants in an enhancer region close to PITX2 (paired-like homeodomain transcription factor 2) are strongly associated with atrial fibrillation, but the mechanism behind this association remains unknown. This study evaluated the consequences of PITX2 deletion (PITX2-/-) in human induced pluripotent stem cell-derived atrial cardiomyocytes. METHODS CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/clustered regularly interspaced short palindromic repeat-associated 9) was used to delete PITX2 in a healthy human iPSC line that served as isogenic control. Human induced pluripotent stem cell-derived atrial cardiomyocytes were differentiated with unfiltered retinoic acid and cultured in atrial engineered heart tissue. Force and action potential were measured in atrial engineered heart tissues. Single human induced pluripotent stem cell-derived atrial cardiomyocytes were isolated from atrial engineered heart tissue for ion current measurements. RESULTS PITX2-/- atrial engineered heart tissue beats slightly slower than isogenic control without irregularity. Force was lower in PITX2-/- than in isogenic control (0.053±0.015 versus 0.131±0.017 mN, n=28/3 versus n=28/4, PITX2-/- versus isogenic control; P<0.0001), accompanied by lower expression of CACNA1C and lower L-type Ca2+ current density. Early repolarization was weaker (action potential duration at 20% repolarization; 45.5±13.2 versus 8.6±5.3 ms, n=18/3 versus n=12/4, PITX2-/- versus isogenic control; P<0.0001), and maximum diastolic potential was more negative (-78.3±3.1 versus -69.7±0.6 mV, n=18/3 versus n=12/4, PITX2-/- versus isogenic control; P=0.001), despite normal inward rectifier currents (both IK1 and IK,ACh) and carbachol-induced shortening of action potential duration. CONCLUSIONS Complete PITX2 deficiency in human induced pluripotent stem cell-derived atrial cardiomyocytes recapitulates some findings of electrical remodeling of atrial fibrillation in the absence of fast beating, indicating that these abnormalities could be primary consequences of lower PITX2 levels.
Collapse
Affiliation(s)
- Carl Schulz
- Institute of Experimental Pharmacology and Toxicology (C.S., M.D.L., G.M., J.S., A.H., T.E., T.C.), University Medical Center Hamburg-Eppendorf, Germany
- German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck (C.S., M.D.L., G.M., J.S., E.S., P.K.)
| | - Marc D Lemoine
- Institute of Experimental Pharmacology and Toxicology (C.S., M.D.L., G.M., J.S., A.H., T.E., T.C.), University Medical Center Hamburg-Eppendorf, Germany
- German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck (C.S., M.D.L., G.M., J.S., E.S., P.K.)
- Department of Cardiology, University Heart and Vascular Center, Hamburg, Germany (M.D.L., A.H., P.K., T.E., T.C.)
| | - Giulia Mearini
- Institute of Experimental Pharmacology and Toxicology (C.S., M.D.L., G.M., J.S., A.H., T.E., T.C.), University Medical Center Hamburg-Eppendorf, Germany
- German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck (C.S., M.D.L., G.M., J.S., E.S., P.K.)
- DiNAQOR AG, Pfäffikon, Switzerland (G.M., P.K.)
| | - Jussi Koivumäki
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Finland (J.K.)
| | - Jascha Sani
- Institute of Experimental Pharmacology and Toxicology (C.S., M.D.L., G.M., J.S., A.H., T.E., T.C.), University Medical Center Hamburg-Eppendorf, Germany
- German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck (C.S., M.D.L., G.M., J.S., E.S., P.K.)
| | - Edzard Schwedhelm
- Institute of Clinical Pharmacology and Toxicology (E.S.), University Medical Center Hamburg-Eppendorf, Germany
- German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck (C.S., M.D.L., G.M., J.S., E.S., P.K.)
| | - Paulus Kirchhof
- German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck (C.S., M.D.L., G.M., J.S., E.S., P.K.)
- Department of Cardiology, University Heart and Vascular Center, Hamburg, Germany (M.D.L., A.H., P.K., T.E., T.C.)
- DiNAQOR AG, Pfäffikon, Switzerland (G.M., P.K.)
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, United Kingdom (P.K.)
| | - Amer Ghalawinji
- Division of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Germany (A.G., M.S.)
| | - Monika Stoll
- Division of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Germany (A.G., M.S.)
- Department of Biochemistry, CARIM School for Cardiovascular Sciences, Maastricht University, the Netherlands (M.S.)
| | - Arne Hansen
- Institute of Experimental Pharmacology and Toxicology (C.S., M.D.L., G.M., J.S., A.H., T.E., T.C.), University Medical Center Hamburg-Eppendorf, Germany
- Department of Cardiology, University Heart and Vascular Center, Hamburg, Germany (M.D.L., A.H., P.K., T.E., T.C.)
| | - Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology (C.S., M.D.L., G.M., J.S., A.H., T.E., T.C.), University Medical Center Hamburg-Eppendorf, Germany
- Department of Cardiology, University Heart and Vascular Center, Hamburg, Germany (M.D.L., A.H., P.K., T.E., T.C.)
| | - Torsten Christ
- Institute of Experimental Pharmacology and Toxicology (C.S., M.D.L., G.M., J.S., A.H., T.E., T.C.), University Medical Center Hamburg-Eppendorf, Germany
- Department of Cardiology, University Heart and Vascular Center, Hamburg, Germany (M.D.L., A.H., P.K., T.E., T.C.)
| |
Collapse
|
24
|
Dong Y, Zhai Z, Wang J, Xia Z, Xia Z, Zhu B, Dong Q, Li Q, Li J. Angiotensin receptor-neprilysin inhibitor delays progression from paroxysmal to persistent atrial fibrillation. Sci Rep 2023; 13:3140. [PMID: 36823222 PMCID: PMC9950488 DOI: 10.1038/s41598-023-30349-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Progression from paroxysmal to persistent atrial fibrillation (AF) is linked to adverse clinical outcomes. The present study sought to clarify whether angiotensin receptor-neprilysin inhibitor (ARNI) can delay AF progression. A retrospective cohort study was conducted on consecutive patients with paroxysmal AF admitted at the Second Affiliated Hospital of Nanchang University between January 2017 and January 2022. The risk of AF progression from paroxysmal to persistent was compared between paroxysmal patients treated with ARNI and those who received an angiotensin receptor blocker (ARB). Seven-day Holter monitoring was performed to identify persistent AF. Propensity-score matched analysis was performed to compare the two groups. Cox-regression was used to estimate the hazard ratio (HR) for AF progression events. A total of 1083 patients were screened, and 113 patients in the ARB group and 57 patients in the ARNI group were eligible for analysis. Before propensity-score matching, the ARNI therapy was associated with a lower risk of AF progression than the ARB therapy (HR 0.34; 95% confidence interval [CI] 0.14-0.81; P = 0.015) after a median follow-up of 705 (interquartile range [IQR] 512 to 895) days. Among 170 patients, 47 ARNI-treated patients were successfully matched to 47 ARB-treated patients. After a median follow-up of 724 (541-929) days, compared to ARB, ARNI significantly reduced the risk of AF progression (HR 0.32; 95% CI 0.12-0.88; P = 0.016). ARNI may be superior to ARB in reducing the risk of progression from paroxysmal to persistent AF.
Collapse
Affiliation(s)
- Youzheng Dong
- grid.412455.30000 0004 1756 5980Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, No.1 of Minde Road, Nanchang, 330006 China
| | - Zhenyu Zhai
- grid.412455.30000 0004 1756 5980Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, No.1 of Minde Road, Nanchang, 330006 China
| | - Jihong Wang
- grid.412455.30000 0004 1756 5980Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, No.1 of Minde Road, Nanchang, 330006 China
| | - Zhen Xia
- grid.412455.30000 0004 1756 5980Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, No.1 of Minde Road, Nanchang, 330006 China
| | - Zirong Xia
- grid.412455.30000 0004 1756 5980Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, No.1 of Minde Road, Nanchang, 330006 China
| | - Bo Zhu
- grid.412455.30000 0004 1756 5980Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, No.1 of Minde Road, Nanchang, 330006 China
| | - Quanbing Dong
- grid.412455.30000 0004 1756 5980Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, No.1 of Minde Road, Nanchang, 330006 China
| | - Qing Li
- grid.412455.30000 0004 1756 5980Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, No.1 of Minde Road, Nanchang, 330006 China
| | - Juxiang Li
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, No.1 of Minde Road, Nanchang, 330006, China.
| |
Collapse
|
25
|
Chen Y, Ouyang T, Yin Y, Fang C, Tang CE, Luo J, Luo F. Analysis of infiltrated immune cells in left atriums from patients with atrial fibrillation and identification of circRNA biomarkers for postoperative atrial fibrillation. Front Genet 2022; 13:1003366. [PMID: 36568366 PMCID: PMC9780452 DOI: 10.3389/fgene.2022.1003366] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 11/22/2022] [Indexed: 12/14/2022] Open
Abstract
Background: Atrial fibrillation (AF) increases the risk of stroke and heart failure. Postoperative AF (POAF) increases the risk of mortality after cardiac surgery. This study aims to explore mechanisms underlying AF, analyze infiltration of immune cells in left atrium (LA) from patients with AF, and identify potential circular RNA (circRNA) biomarkers for POAF. Methods: Raw data of GSE797689, GSE115574, and GSE97455 were downloaded and processed. AF-related gene co-expression network was constructed using weighted gene correlation network analysis and enrichment analysis of genes in relevant module was conducted. Gene set enrichment analysis (GSEA) and gene set variation analysis (GSVA) were applied to investigate pathways significantly enriched in AF group. Infiltration of immune cells was analyzed using single-sample GSEA. Differentially expressed genes (DEGs) between patients with or without AF were identified and competing endogenous RNA (ceRNA) networks of DEGs were constructed. To screen biomarkers for POAF, differentially expressed circRNAs (DEcircRNAs) between patients with or without POAF were identified. Intersection between DEcircRNAs and circRNAs in ceRNA networks of DEGs were extracted and circRNAs in the intersection were further screened using support vector machine, random forest, and neural network to identify biomarkers for POAF. Results: Three modules were found to be relevant with AF and enrichment analysis indicated that genes in these modules were enriched in synthesis of extracellular matrix and inflammatory response. The results of GSEA and GSVA suggested that inflammatory response-related pathways were significantly enriched in AF group. Immune cells like macrophages, mast cells, and neutrophils were significantly infiltrated in LA tissues from patients with AF. The expression levels of immune genes such as CHGB, HLA-DRA, LYZ, IGKV1-17 and TYROBP were significantly upregulated in patients with AF, which were correlated with infiltration of immune cells. ceRNA networks of DEGs were constructed and has_circ_0006314 and hsa_circ_0055387 were found to have potential predictive values for POAF. Conclusion: Synthesis of extracellular matrix and inflammatory response were main processes involved in development and progression of AF. Infiltration of immune cells was significantly different between patients with or without AF. Has_circ_0006314 and hsa_circ_0055387 were found to have potential predictive values for POAF.
Collapse
Affiliation(s)
- Yubin Chen
- Department of Cardiac Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Tianyu Ouyang
- Department of Cardiac Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Yue Yin
- Department of Cardiac Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Cheng Fang
- Department of Cardiac Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Can-E Tang
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China,The Institute of Medical Science Research, Xiangya Hospital, Central South University, Changsha, China
| | - Jingmin Luo
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, China,*Correspondence: Jingmin Luo, ; Fanyan Luo,
| | - Fanyan Luo
- Department of Cardiac Surgery, Xiangya Hospital, Central South University, Changsha, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China,*Correspondence: Jingmin Luo, ; Fanyan Luo,
| |
Collapse
|
26
|
Adenosine and Adenosine Receptors: Advances in Atrial Fibrillation. Biomedicines 2022; 10:biomedicines10112963. [PMID: 36428533 PMCID: PMC9687155 DOI: 10.3390/biomedicines10112963] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/07/2022] [Accepted: 11/14/2022] [Indexed: 11/19/2022] Open
Abstract
Atrial fibrillation (AF) is the most common arrhythmia in the world. Because the key to developing innovative therapies that limit the onset and the progression of AF is to fully understand the underlying molecular mechanisms of AF, the aim of the present narrative review is to report the most recent advances in the potential role of the adenosinergic system in the pathophysiology of AF. After a comprehensive approach describing adenosinergic system signaling and the mechanisms of the initiation and maintenance of AF, we address the interactions of the adenosinergic system's signaling with AF. Indeed, adenosine release can activate four G-coupled membrane receptors, named A1, A2A, A2B and A3. Activation of the A2A receptors can promote the occurrence of delayed depolarization, while activation of the A1 receptors can shorten the action potential's duration and induce the resting membrane's potential hyperpolarization, which promote pulmonary vein firing, stabilize the AF rotors and allow for functional reentry. Moreover, the A2B receptors have been associated with atrial fibrosis homeostasis. Finally, the adenosinergic system can modulate the autonomous nervous system and is associated with AF risk factors. A question remains regarding adenosine release and the adenosine receptors' activation and whether this would be a cause or consequence of AF.
Collapse
|
27
|
Saxena P, Myles RC, Smith GL, Workman AJ. Adrenoceptor sub-type involvement in Ca 2+ current stimulation by noradrenaline in human and rabbit atrial myocytes. Pflugers Arch 2022; 474:1311-1321. [PMID: 36131146 DOI: 10.1007/s00424-022-02746-z] [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: 05/23/2022] [Revised: 08/17/2022] [Accepted: 09/02/2022] [Indexed: 10/14/2022]
Abstract
Atrial fibrillation (AF) from elevated adrenergic activity may involve increased atrial L-type Ca2+ current (ICaL) by noradrenaline (NA). However, the contribution of the adrenoceptor (AR) sub-types to such ICaL-increase is poorly understood, particularly in human. We therefore investigated effects of various broad-action and sub-type-specific α- and β-AR antagonists on NA-stimulated atrial ICaL. ICaL was recorded by whole-cell-patch clamp at 37 °C in myocytes isolated enzymatically from atrial tissues from consenting patients undergoing elective cardiac surgery and from rabbits. NA markedly increased human atrial ICaL, maximally by ~ 2.5-fold, with EC75 310 nM. Propranolol (β1 + β2-AR antagonist, 0.2 microM) substantially decreased NA (310 nM)-stimulated ICaL, in human and rabbit. Phentolamine (α1 + α2-AR antagonist, 1 microM) also decreased NA-stimulated ICaL. CGP20712A (β1-AR antagonist, 0.3 microM) and prazosin (α1-AR antagonist, 0.5 microM) each decreased NA-stimulated ICaL in both species. ICI118551 (β2-AR antagonist, 0.1 microM), in the presence of NA + CGP20712A, had no significant effect on ICaL in human atrial myocytes, but increased it in rabbit. Yohimbine (α2-AR antagonist, 10 microM), with NA + prazosin, had no significant effect on human or rabbit ICaL. Stimulation of atrial ICaL by NA is mediated, based on AR sub-type antagonist responses, mainly by activating β1- and α1-ARs in both human and rabbit, with a β2-inhibitory contribution evident in rabbit, and negligible α2 involvement in either species. This improved understanding of AR sub-type contributions to noradrenergic activation of atrial ICaL could help inform future potential optimisation of pharmacological AR-antagonism strategies for inhibiting adrenergic AF.
Collapse
Affiliation(s)
- Priyanka Saxena
- Institute of Cardiovascular & Medical Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, 126 University Place, Glasgow, G12 8TA, UK
| | - Rachel C Myles
- Institute of Cardiovascular & Medical Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, 126 University Place, Glasgow, G12 8TA, UK
| | - Godfrey L Smith
- Institute of Cardiovascular & Medical Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, 126 University Place, Glasgow, G12 8TA, UK
| | - Antony J Workman
- Institute of Cardiovascular & Medical Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, 126 University Place, Glasgow, G12 8TA, UK.
| |
Collapse
|
28
|
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
|
29
|
Effects of Sacubitril/Valsartan on the Expression of CaMKII/Cav1.2 in Atrial Fibrillation Stimulation Rabbit Model. BIOMED RESEARCH INTERNATIONAL 2022. [DOI: 10.1155/2022/5832543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Background and Objective. Atrial fibrillation (AF) is linked to high morbidity and death rates throughout the world due to limited therapeutic options and thus presents a major challenge to the developed and developing countries. In this study, we aim to investigate the influence of sacubitril/valsartan (sac/val) treatment on the calmodulin-dependent protein kinase II (CaMKII)/Cav1.2 expression in AF models. Methods. Overall, 18 rabbits were randomly divided into control, pacing (600 beats/min), and pacing+sac/val groups. The rabbits in the pacing+sac/val cohort received oral sac/val (10 mg/kg twice daily) across the 21-day investigation period. After three weeks, the atrial effective refractory period (AERP) and AF induction rate were compared. HL-1 cultures were exposed to fast pacing (24 h) with and without LBQ657 (active sacubitril form)/valsartan. Western blots were used for detecting Cav1.2 and CaMKII expression within atrial muscles of the rabbits and HL-1 cultures of AF model. Results. In comparison to the sham cohort, the AF induction rate was markedly increased together with AERP reduction within pacing cohort. Such changes were markedly rescued through sac/val treatment in pacing+sac/val cohort. The proteomic expression profiles of CaMKII and Cav1.2 showed that the CaMKII expression was markedly upregulated, while Cav1.2 expression was downregulated in the pacing cohort. Importantly, these effects were absent in pacing+sac/val cohort. Conclusion. Results of this study show that sac/val treatment regulates the expression of CaMKII/Cav1.2 and could alter this pathway in atrial rapid electrical stimulation models. Therefore, this investigation could contribute to a novel strategy in AF therapeutics in clinical settings.
Collapse
|
30
|
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia despite substantial efforts to understand the pathophysiology of the condition and develop improved treatments. Identifying the underlying causative mechanisms of AF in individual patients is difficult and the efficacy of current therapies is suboptimal. Consequently, the incidence of AF is steadily rising and there is a pressing need for novel therapies. Research has revealed that defects in specific molecular pathways underlie AF pathogenesis, resulting in electrical conduction disorders that drive AF. The severity of this so-called electropathology correlates with the stage of AF disease progression and determines the response to AF treatment. Therefore, unravelling the molecular mechanisms underlying electropathology is expected to fuel the development of innovative personalized diagnostic tools and mechanism-based therapies. Moreover, the co-creation of AF studies with patients to implement novel diagnostic tools and therapies is a prerequisite for successful personalized AF management. Currently, various treatment modalities targeting AF-related electropathology, including lifestyle changes, pharmaceutical and nutraceutical therapy, substrate-based ablative therapy, and neuromodulation, are available to maintain sinus rhythm and might offer a novel holistic strategy to treat AF.
Collapse
Affiliation(s)
- Bianca J J M Brundel
- Department of Physiology, Amsterdam University Medical Centers, VU Universiteit, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands.
| | - Xun Ai
- Department of Physiology and Cell Biology, College of Medicine/Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | | | - Myrthe F Kuipers
- AFIPonline.org, Atrial Fibrillation Innovation Platform, Amsterdam, Netherlands
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | | |
Collapse
|
31
|
Sergienko NM, Donner DG, Delbridge LMD, McMullen JR, Weeks KL. Protein phosphatase 2A in the healthy and failing heart: New insights and therapeutic opportunities. Cell Signal 2021; 91:110213. [PMID: 34902541 DOI: 10.1016/j.cellsig.2021.110213] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 12/02/2021] [Accepted: 12/07/2021] [Indexed: 02/06/2023]
Abstract
Protein phosphatases have emerged as critical regulators of phosphoprotein homeostasis in settings of health and disease. Protein phosphatase 2A (PP2A) encompasses a large subfamily of enzymes that remove phosphate groups from serine/threonine residues within phosphoproteins. The heterogeneity in PP2A structure, which arises from the grouping of different catalytic, scaffolding and regulatory subunit isoforms, creates distinct populations of catalytically active enzymes (i.e. holoenzymes) that localise to different parts of the cell. This structural complexity, combined with other regulatory mechanisms, such as interaction of PP2A heterotrimers with accessory proteins and post-translational modification of the catalytic and/or regulatory subunits, enables PP2A holoenzymes to target phosphoprotein substrates in a highly specific manner. In this review, we summarise the roles of PP2A in cardiac physiology and disease. PP2A modulates numerous processes that are vital for heart function including calcium handling, contractility, β-adrenergic signalling, metabolism and transcription. Dysregulation of PP2A has been observed in human cardiac disease settings, including heart failure and atrial fibrillation. Efforts are underway, particularly in the cancer field, to develop therapeutics targeting PP2A activity. The development of small molecule activators of PP2A (SMAPs) and other compounds that selectively target specific PP2A holoenzymes (e.g. PP2A/B56α and PP2A/B56ε) will improve understanding of the function of different PP2A species in the heart, and may lead to the development of therapeutics for normalising aberrant protein phosphorylation in settings of cardiac remodelling and dysfunction.
Collapse
Affiliation(s)
- Nicola M Sergienko
- Baker Heart and Diabetes Institute, Melbourne VIC 3004, Australia; Central Clinical School, Monash University, Clayton VIC 3800, Australia
| | - Daniel G Donner
- Baker Heart and Diabetes Institute, Melbourne VIC 3004, Australia; Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville VIC 3010, Australia
| | - Lea M D Delbridge
- Department of Anatomy and Physiology, The University of Melbourne, Parkville VIC 3010, Australia
| | - Julie R McMullen
- Baker Heart and Diabetes Institute, Melbourne VIC 3004, Australia; Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville VIC 3010, Australia; Department of Physiology and Department of Medicine Alfred Hospital, Monash University, Clayton VIC 3800, 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.
| | - Kate L Weeks
- Baker Heart and Diabetes Institute, Melbourne VIC 3004, Australia; Department of Anatomy and Physiology, The University of Melbourne, Parkville VIC 3010, Australia; Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville VIC 3010, Australia; Department of Diabetes, Central Clinical School, Monash University, Clayton VIC 3800, Australia.
| |
Collapse
|
32
|
Function and regulation of phosphatase 1 in healthy and diseased heart. Cell Signal 2021; 90:110203. [PMID: 34822978 DOI: 10.1016/j.cellsig.2021.110203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 11/12/2021] [Accepted: 11/18/2021] [Indexed: 12/12/2022]
Abstract
Reversible phosphorylation of ion channels and calcium-handling proteins provides precise post-translational regulation of cardiac excitation and contractility. Serine/threonine phosphatases govern dephosphorylation of the majority of cardiac proteins. Accordingly, dysfunction of this regulation contributes to the development and progression of heart failure and atrial fibrillation. On the molecular level, these changes include alterations in the expression level and phosphorylation status of Ca2+ handling and excitation-contraction coupling proteins provoked by dysregulation of phosphatases. The serine/threonine protein phosphatase PP1 is one a major player in the regulation of cardiac excitation-contraction coupling. PP1 essentially impacts on cardiac physiology and pathophysiology via interactions with the cardiac ion channels Cav1.2, NKA, NCX and KCNQ1, sarcoplasmic reticulum-bound Ca2+ handling proteins such as RyR2, SERCA and PLB as well as the contractile proteins MLC2, TnI and MyBP-C. PP1 itself but also PP1-regulatory proteins like inhibitor-1, inhibitor-2 and heat-shock protein 20 are dysregulated in cardiac disease. Therefore, they represent interesting targets to gain more insights in heart pathophysiology and to identify new treatment strategies for patients with heart failure or atrial fibrillation. We describe the genetic and holoenzymatic structure of PP1 and review its role in the heart and cardiac disease. Finally, we highlight the importance of the PP1 regulatory proteins for disease manifestation, provide an overview of genetic models to study the role of PP1 for the development of heart failure and atrial fibrillation and discuss possibilities of pharmacological interventions.
Collapse
|
33
|
Reinhardt F, Beneke K, Pavlidou NG, Conradi L, Reichenspurner H, Hove-Madsen L, Molina CE. Abnormal Calcium Handling in Atrial Fibrillation Is Linked to Changes in Cyclic AMP Dependent Signaling. Cells 2021; 10:cells10113042. [PMID: 34831263 PMCID: PMC8616167 DOI: 10.3390/cells10113042] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/22/2021] [Accepted: 11/02/2021] [Indexed: 01/03/2023] Open
Abstract
Both, the decreased L-type Ca2+ current (ICa,L) density and increased spontaneous Ca2+ release from the sarcoplasmic reticulum (SR), have been associated with atrial fibrillation (AF). In this study, we tested the hypothesis that remodeling of 3′,5′-cyclic adenosine monophosphate (cAMP)-dependent protein kinase A (PKA) signaling is linked to these compartment-specific changes (up- or down-regulation) in Ca2+-handling. Perforated patch-clamp experiments were performed in atrial myocytes from 53 patients with AF and 104 patients in sinus rhythm (Ctl). A significantly higher frequency of transient inward currents (ITI) activated by spontaneous Ca2+ release was confirmed in myocytes from AF patients. Next, inhibition of PKA by H-89 promoted a stronger effect on the ITI frequency in these myocytes compared to myocytes from Ctl patients (7.6-fold vs. 2.5-fold reduction), while the β-agonist isoproterenol (ISO) caused a greater increase in Ctl patients (5.5-fold vs. 2.1-fold). ICa,L density was larger in myocytes from Ctl patients at baseline (p < 0.05). However, the effect of ISO on ICa,L density was only slightly stronger in AF than in Ctl myocytes (3.6-fold vs. 2.7-fold). Interestingly, a significant reduction of ICa,L and Ca2+ sparks was observed upon Ca2+/Calmodulin-dependent protein kinase II inhibition by KN-93, but this inhibition had no effect on ITI. Fluorescence resonance energy transfer (FRET) experiments showed that although AF promoted cytosolic desensitization to β-adrenergic stimulation, ISO increased cAMP to similar levels in both groups of patients in the L-type Ca2+ channel and ryanodine receptor compartments. Basal cAMP signaling also showed compartment-specific regulation by phosphodiesterases in atrial myocytes from 44 Ctl and 43 AF patients. Our results suggest that AF is associated with opposite changes in compartmentalized PKA/cAMP-dependent regulation of ICa,L (down-regulation) and ITI (up-regulation).
Collapse
Affiliation(s)
- Franziska Reinhardt
- Department of Cardiovascular Surgery, University Heart & Vascular Center Hamburg UKE, 20251 Hamburg, Germany; (F.R.); (L.C.); (H.R.)
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20251 Hamburg, Germany; (K.B.); (N.G.P.)
| | - Kira Beneke
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20251 Hamburg, Germany; (K.B.); (N.G.P.)
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf (UKE), 20251 Hamburg, Germany
| | - Nefeli Grammatica Pavlidou
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20251 Hamburg, Germany; (K.B.); (N.G.P.)
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf (UKE), 20251 Hamburg, Germany
| | - Lenard Conradi
- Department of Cardiovascular Surgery, University Heart & Vascular Center Hamburg UKE, 20251 Hamburg, Germany; (F.R.); (L.C.); (H.R.)
| | - Hermann Reichenspurner
- Department of Cardiovascular Surgery, University Heart & Vascular Center Hamburg UKE, 20251 Hamburg, Germany; (F.R.); (L.C.); (H.R.)
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf (UKE), 20251 Hamburg, Germany
| | - Leif Hove-Madsen
- Biomedical Research Institute Barcelona, IIBB-CSIC and IIB Sant Pau, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain;
| | - Cristina E. Molina
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20251 Hamburg, Germany; (K.B.); (N.G.P.)
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf (UKE), 20251 Hamburg, Germany
- Correspondence: ; Tel.: +49-407-4105-7095
| |
Collapse
|
34
|
Xu D, Murakoshi N, Tajiri K, Duo F, Okabe Y, Murakata Y, Yuan Z, Li S, Aonuma K, Song Z, Shimoda Y, Mori H, Sato A, Nogami A, Aonuma K, Ieda M. Xanthine oxidase inhibitor febuxostat reduces atrial fibrillation susceptibility by inhibition of oxidized CaMKII in Dahl salt-sensitive rats. Clin Sci (Lond) 2021; 135:2409-2422. [PMID: 34386810 DOI: 10.1042/cs20210405] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 08/06/2021] [Accepted: 08/13/2021] [Indexed: 11/17/2022]
Abstract
Oxidative stress could be a possible mechanism and a therapeutic target of atrial fibrillation (AF). However, the effects of the xanthine oxidase (XO) inhibition for AF remain to be fully elucidated. We investigated the effects of a novel XO inhibitor febuxostat on AF compared with allopurinol in hypertension rat model. Five-week-old Dahl salt-sensitive rats were fed either low-salt (LS) (0.3% NaCl) or high-salt (HS) (8% NaCl) diet. After 4 weeks of diet, HS diet rats were divided into three groups: orally administered to vehicle (HS-C), febuxostat (5 mg/kg/day) (HS-F), or allopurinol (50 mg/kg/day) (HS-A). After 4 weeks of treatment, systolic blood pressure (SBP) was significantly higher in HS-C than LS, and it was slightly but significantly decreased by treatment with each XO inhibitor. AF duration was significantly prolonged in HS-C compared with LS, and significantly suppressed in both HS-F and HS-A (LS; 5.8 ± 3.5 s, HS-C; 33.9 ± 23.7 s, HS-F; 15.0 ± 14.1 s, HS-A; 20.1 ± 11.9 s: P<0.05). Ca2+ spark frequency was obviously increased in HS-C rats and reduced in the XO inhibitor-treated rats, especially in HS-F group. Western blotting revealed that the atrial expression levels of Met281/282-oxidized Ca2+/Calmodulin-dependent kinase II (CaMKII) and Ser2814-phosphorylated ryanodine receptor 2 were significantly increased in HS-C, and those were suppressed in HS-F and HS-A. Decreased expression of gap junction protein connexin 40 in HS-C was partially restored by treatment with each XO inhibitor. In conclusion, XO inhibitor febuxostat, as well as allopurinol, could reduce hypertension-related increase in AF perpetuation by restoring Ca2+ handling and gap junction.
Collapse
Affiliation(s)
- DongZhu Xu
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Nobuyuki Murakoshi
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kazuko Tajiri
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Feng Duo
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yuta Okabe
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yoshiko Murakata
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Zixun Yuan
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Siqi Li
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kazuhiro Aonuma
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Zonghu Song
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yuzuno Shimoda
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Haruka Mori
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Akira Sato
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Akihiko Nogami
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kazutaka Aonuma
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Masaki Ieda
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
35
|
Ai X, Yan J, Pogwizd SM. Serine-threonine protein phosphatase regulation of Cx43 dephosphorylation in arrhythmogenic disorders. Cell Signal 2021; 86:110070. [PMID: 34217833 PMCID: PMC8963383 DOI: 10.1016/j.cellsig.2021.110070] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 06/11/2021] [Accepted: 06/29/2021] [Indexed: 12/11/2022]
Abstract
Regulation of cell-to-cell communication in the heart by the gap junction protein Connexin43 (Cx43) involves modulation of Cx43 phosphorylation state by protein kinases, and dephosphorylation by protein phosphatases. Dephosphorylation of Cx43 has been associated with impaired intercellular coupling and enhanced arrhythmogenesis in various pathologic states. While there has been extensive study of the protein kinases acting on Cx43, there has been limited studies of the protein phosphatases that may underlie Cx43 dephosphorylation. The focus of this review is to introduce serine-threonine protein phosphatase regulation of Cx43 phosphorylation state and cell-to-cell communication, and its impact on arrhythmogenesis in the setting of chronic heart failure and myocardial ischemia, as well as on atrial fibrillation. We also discuss the therapeutic potential of modulating protein phosphatases to treat arrhythmias in these clinical settings.
Collapse
Affiliation(s)
- Xun Ai
- Department of Physiology & Biophysics, Rush University, Chicago, IL, United States of America
| | - Jiajie Yan
- Department of Physiology & Biophysics, Rush University, Chicago, IL, United States of America
| | - Steven M Pogwizd
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States of America.
| |
Collapse
|
36
|
Kaplan AD, Joca HC, Boyman L, Greiser M. Calcium Signaling Silencing in Atrial Fibrillation: Implications for Atrial Sodium Homeostasis. Int J Mol Sci 2021; 22:10513. [PMID: 34638854 PMCID: PMC8508839 DOI: 10.3390/ijms221910513] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 09/14/2021] [Accepted: 09/14/2021] [Indexed: 12/19/2022] Open
Abstract
Atrial fibrillation (AF) is the most common type of cardiac arrhythmia, affecting more than 33 million people worldwide. Despite important advances in therapy, AF's incidence remains high, and treatment often results in recurrence of the arrhythmia. A better understanding of the cellular and molecular changes that (1) trigger AF and (2) occur after the onset of AF will help to identify novel therapeutic targets. Over the past 20 years, a large body of research has shown that intracellular Ca2+ handling is dramatically altered in AF. While some of these changes are arrhythmogenic, other changes counteract cellular arrhythmogenic mechanisms (Calcium Signaling Silencing). The intracellular Na+ concentration ([Na+])i is a key regulator of intracellular Ca2+ handling in cardiac myocytes. Despite its importance in the regulation of intracellular Ca2+ handling, little is known about [Na+]i, its regulation, and how it might be changed in AF. Previous work suggests that there might be increases in the late component of the atrial Na+ current (INa,L) in AF, suggesting that [Na+]i levels might be high in AF. Indeed, a pharmacological blockade of INa,L has been suggested as a treatment for AF. Here, we review calcium signaling silencing and changes in intracellular Na+ homeostasis during AF. We summarize the proposed arrhythmogenic mechanisms associated with increases in INa,L during AF and discuss the evidence from clinical trials that have tested the pharmacological INa,L blocker ranolazine in the treatment of AF.
Collapse
Affiliation(s)
- Aaron D. Kaplan
- Center for Biomedical Engineering and Technology, Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.D.K.); (H.C.J.); (L.B.)
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Humberto C. Joca
- Center for Biomedical Engineering and Technology, Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.D.K.); (H.C.J.); (L.B.)
| | - Liron Boyman
- Center for Biomedical Engineering and Technology, Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.D.K.); (H.C.J.); (L.B.)
| | - Maura Greiser
- Center for Biomedical Engineering and Technology, Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.D.K.); (H.C.J.); (L.B.)
| |
Collapse
|
37
|
Papathanasiou KA, Giotaki SG, Vrachatis DA, Siasos G, Lambadiari V, Iliodromitis KE, Kossyvakis C, Kaoukis A, Raisakis K, Deftereos G, Papaioannou TG, Giannopoulos G, Avramides D, Deftereos SG. Molecular Insights in Atrial Fibrillation Pathogenesis and Therapeutics: A Narrative Review. Diagnostics (Basel) 2021; 11:diagnostics11091584. [PMID: 34573926 PMCID: PMC8470040 DOI: 10.3390/diagnostics11091584] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 08/29/2021] [Accepted: 08/30/2021] [Indexed: 12/15/2022] Open
Abstract
The prevalence of atrial fibrillation (AF) is bound to increase globally in the following years, affecting the quality of life of millions of people, increasing mortality and morbidity, and beleaguering health care systems. Increasingly effective therapeutic options against AF are the constantly evolving electroanatomic substrate mapping systems of the left atrium (LA) and ablation catheter technologies. Yet, a prerequisite for better long-term success rates is the understanding of AF pathogenesis and maintenance. LA electrical and anatomical remodeling remains in the epicenter of current research for novel diagnostic and treatment modalities. On a molecular level, electrical remodeling lies on impaired calcium handling, enhanced inwardly rectifying potassium currents, and gap junction perturbations. In addition, a wide array of profibrotic stimuli activates fibroblast to an increased extracellular matrix turnover via various intermediaries. Concomitant dysregulation of the autonomic nervous system and the humoral function of increased epicardial adipose tissue (EAT) are established mediators in the pathophysiology of AF. Local atrial lymphomononuclear cells infiltrate and increased inflammasome activity accelerate and perpetuate arrhythmia substrate. Finally, impaired intracellular protein metabolism, excessive oxidative stress, and mitochondrial dysfunction deplete atrial cardiomyocyte ATP and promote arrhythmogenesis. These overlapping cellular and molecular alterations hinder us from distinguishing the cause from the effect in AF pathogenesis. Yet, a plethora of therapeutic modalities target these molecular perturbations and hold promise in combating the AF burden. Namely, atrial selective ion channel inhibitors, AF gene therapy, anti-fibrotic agents, AF drug repurposing, immunomodulators, and indirect cardiac neuromodulation are discussed here.
Collapse
Affiliation(s)
- Konstantinos A. Papathanasiou
- Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (S.G.G.); (D.A.V.); (G.S.); (V.L.); (T.G.P.)
| | - Sotiria G. Giotaki
- Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (S.G.G.); (D.A.V.); (G.S.); (V.L.); (T.G.P.)
| | - Dimitrios A. Vrachatis
- Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (S.G.G.); (D.A.V.); (G.S.); (V.L.); (T.G.P.)
| | - Gerasimos Siasos
- Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (S.G.G.); (D.A.V.); (G.S.); (V.L.); (T.G.P.)
| | - Vaia Lambadiari
- Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (S.G.G.); (D.A.V.); (G.S.); (V.L.); (T.G.P.)
| | | | - Charalampos Kossyvakis
- Department of Cardiology, “G. Gennimatas” General Hospital of Athens, 11527 Athens, Greece; (C.K.); (A.K.); (K.R.); (G.D.); (D.A.)
| | - Andreas Kaoukis
- Department of Cardiology, “G. Gennimatas” General Hospital of Athens, 11527 Athens, Greece; (C.K.); (A.K.); (K.R.); (G.D.); (D.A.)
| | - Konstantinos Raisakis
- Department of Cardiology, “G. Gennimatas” General Hospital of Athens, 11527 Athens, Greece; (C.K.); (A.K.); (K.R.); (G.D.); (D.A.)
| | - Gerasimos Deftereos
- Department of Cardiology, “G. Gennimatas” General Hospital of Athens, 11527 Athens, Greece; (C.K.); (A.K.); (K.R.); (G.D.); (D.A.)
| | - Theodore G. Papaioannou
- Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (S.G.G.); (D.A.V.); (G.S.); (V.L.); (T.G.P.)
| | | | - Dimitrios Avramides
- Department of Cardiology, “G. Gennimatas” General Hospital of Athens, 11527 Athens, Greece; (C.K.); (A.K.); (K.R.); (G.D.); (D.A.)
| | - Spyridon G. Deftereos
- Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (S.G.G.); (D.A.V.); (G.S.); (V.L.); (T.G.P.)
- Correspondence: ; Tel.: +30-21-0583-2355
| |
Collapse
|
38
|
Jakob D, Klesen A, Darkow E, Kari FA, Beyersdorf F, Kohl P, Ravens U, Peyronnet R. Heterogeneity and Remodeling of Ion Currents in Cultured Right Atrial Fibroblasts From Patients With Sinus Rhythm or Atrial Fibrillation. Front Physiol 2021; 12:673891. [PMID: 34149453 PMCID: PMC8209389 DOI: 10.3389/fphys.2021.673891] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/19/2021] [Indexed: 11/23/2022] Open
Abstract
Cardiac fibroblasts express multiple voltage-dependent ion channels. Even though fibroblasts do not generate action potentials, they may influence cardiac electrophysiology by electrical coupling via gap junctions with cardiomyocytes, and through fibrosis. Here, we investigate the electrophysiological phenotype of cultured fibroblasts from right atrial appendage tissue of patients with sinus rhythm (SR) or atrial fibrillation (AF). Using the patch-clamp technique in whole-cell mode, we observed steady-state outward currents exhibiting either no rectification or inward and/or outward rectification. The distributions of current patterns between fibroblasts from SR and AF patients were not significantly different. In response to depolarizing voltage pulses, we measured transient outward currents with fast and slow activation kinetics, an outward background current, and an inward current with a potential-dependence resembling that of L-type Ca2+ channels. In cell-attached patch-clamp mode, large amplitude, paxilline-sensitive single channel openings were found in ≈65% of SR and ∼38% of AF fibroblasts, suggesting the presence of “big conductance Ca2+-activated K+ (BKCa)” channels. The open probability of BKCa was significantly lower in AF than in SR fibroblasts. When cultured in the presence of paxilline, the shape of fibroblasts became wider and less spindle-like. Our data confirm previous findings on cardiac fibroblast electrophysiology and extend them by illustrating differential channel expression in human atrial fibroblasts from SR and AF tissue.
Collapse
Affiliation(s)
- Dorothee Jakob
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg - Bad Krozingen, Freiburg, Germany.,Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Alexander Klesen
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg - Bad Krozingen, Freiburg, Germany.,Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Elisa Darkow
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg - Bad Krozingen, Freiburg, Germany.,Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Fabian A Kari
- Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Cardiovascular Surgery, University Heart Center Freiburg - Bad Krozingen, Freiburg, Germany
| | - Friedhelm Beyersdorf
- Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Cardiovascular Surgery, University Heart Center Freiburg - Bad Krozingen, Freiburg, Germany
| | - Peter Kohl
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg - Bad Krozingen, Freiburg, Germany.,Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany.,CIBSS Centre for Integrative Biological Signaling Studies, University of Freiburg, Freiburg, Germany
| | - Ursula Ravens
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg - Bad Krozingen, Freiburg, Germany.,Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Rémi Peyronnet
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg - Bad Krozingen, Freiburg, Germany.,Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
39
|
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia, largely associated to morbidity and mortality. Over the past decades, research in appearance and progression of this arrhythmia have turned into significant advances in its management. However, the incidence of AF continues to increase with the aging of the population and many important fundamental and translational underlaying mechanisms remain elusive. Here, we review recent advances in molecular and cellular basis for AF initiation, maintenance and progression. We first provide an overview of the basic molecular and electrophysiological mechanisms that lead and characterize AF. Next, we discuss the upstream regulatory factors conducting the underlying mechanisms which drive electrical and structural AF-associated remodeling, including genetic factors (risk variants associated to AF as transcriptional regulators and genetic changes associated to AF), neurohormonal regulation (i.e., cAMP) and oxidative stress imbalance (cGMP and mitochondrial dysfunction). Finally, we discuss the potential therapeutic implications of those findings, the knowledge gaps and consider future approaches to improve clinical management.
Collapse
|
40
|
Safabakhsh S, Panwar P, Barichello S, Sangha SS, Hanson PJ, Van Petegem F, Laksman Z. THE ROLE OF PHOSPHORYLATION IN ATRIAL FIBRILLATION: A FOCUS ON MASS SPECTROMETRY APPROACHES. Cardiovasc Res 2021; 118:1205-1217. [PMID: 33744917 DOI: 10.1093/cvr/cvab095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 03/16/2021] [Indexed: 11/14/2022] Open
Abstract
Atrial fibrillation (AF) is the most common arrhythmia worldwide. It is associated with significant increases in morbidity in the form of stroke and heart failure, and a doubling in all-cause mortality. The pathophysiology of AF is incompletely understood, and this has contributed to a lack of effective treatments and disease-modifying therapies. An important cellular process that may explain how risk factors give rise to AF includes post-translational modification (PTM) of proteins. As the most commonly occurring PTM, protein phosphorylation is especially relevant. Although many methods exist for studying protein phosphorylation, a common and highly resolute technique is mass spectrometry (MS). This review will discuss recent evidence surrounding the role of protein phosphorylation in the pathogenesis of AF. MS-based technology to study phosphorylation and uses of MS in other areas of medicine such as oncology will also be presented. Based on these data, future goals and experiments will be outlined that utilize MS technology to better understand the role of phosphorylation in AF and elucidate its role in AF pathophysiology. This may ultimately allow for the development of more effective AF therapies.
Collapse
Affiliation(s)
- Sina Safabakhsh
- Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Pankaj Panwar
- AbCellera Biologicals Inc., Vancouver, British Columbia, Canada
| | - Scott Barichello
- Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sarabjit S Sangha
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, 950 West 28th Avenue, Vancouver, British Columbia, Canada.,Molecular Cardiac Physiology Group, Departments of Biomedical Physiology and Kinesiology and Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia, Canada
| | - Paul J Hanson
- UBC Heart Lung Innovation Centre, Vancouver, British Columbia, Canada.,UBC Department of Pathology and Laboratory Medicine, Vancouver, British Columbia, Canada
| | - Filip Van Petegem
- Department of Biochemistry and Molecular Biology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Zachary Laksman
- Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
41
|
Kraft M, Büscher A, Wiedmann F, L’hoste Y, Haefeli WE, Frey N, Katus HA, Schmidt C. Current Drug Treatment Strategies for Atrial Fibrillation and TASK-1 Inhibition as an Emerging Novel Therapy Option. Front Pharmacol 2021; 12:638445. [PMID: 33897427 PMCID: PMC8058608 DOI: 10.3389/fphar.2021.638445] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 01/21/2021] [Indexed: 12/19/2022] Open
Abstract
Atrial fibrillation (AF) is the most common sustained arrhythmia with a prevalence of up to 4% and an upwards trend due to demographic changes. It is associated with an increase in mortality and stroke incidences. While stroke risk can be significantly reduced through anticoagulant therapy, adequate treatment of other AF related symptoms remains an unmet medical need in many cases. Two main treatment strategies are available: rate control that modulates ventricular heart rate and prevents tachymyopathy as well as rhythm control that aims to restore and sustain sinus rhythm. Rate control can be achieved through drugs or ablation of the atrioventricular node, rendering the patient pacemaker-dependent. For rhythm control electrical cardioversion and pharmacological cardioversion can be used. While electrical cardioversion requires fasting and sedation of the patient, antiarrhythmic drugs have other limitations. Most antiarrhythmic drugs carry a risk for pro-arrhythmic effects and are contraindicated in patients with structural heart diseases. Furthermore, catheter ablation of pulmonary veins can be performed with its risk of intraprocedural complications and varying success. In recent years TASK-1 has been introduced as a new target for AF therapy. Upregulation of TASK-1 in AF patients contributes to prolongation of the action potential duration. In a porcine model of AF, TASK-1 inhibition by gene therapy or pharmacological compounds induced cardioversion to sinus rhythm. The DOxapram Conversion TO Sinus rhythm (DOCTOS)-Trial will reveal whether doxapram, a potent TASK-1 inhibitor, can be used for acute cardioversion of persistent and paroxysmal AF in patients, potentially leading to a new treatment option for AF.
Collapse
Affiliation(s)
- Manuel Kraft
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
- HCR, Heidelberg Center for Heart Rhythm Disorders, University of Heidelberg, Heidelberg, Germany
| | - Antonius Büscher
- Clinic for Cardiology II: Electrophysiology, University Hospital Münster, Münster, Germany
| | - Felix Wiedmann
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
- HCR, Heidelberg Center for Heart Rhythm Disorders, University of Heidelberg, Heidelberg, Germany
| | - Yannick L’hoste
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
- HCR, Heidelberg Center for Heart Rhythm Disorders, University of Heidelberg, Heidelberg, Germany
| | - Walter E. Haefeli
- Department of Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, Heidelberg, Germany
| | - Norbert Frey
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
- HCR, Heidelberg Center for Heart Rhythm Disorders, University of Heidelberg, Heidelberg, Germany
| | - Hugo A. Katus
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
- HCR, Heidelberg Center for Heart Rhythm Disorders, University of Heidelberg, Heidelberg, Germany
| | - Constanze Schmidt
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
- HCR, Heidelberg Center for Heart Rhythm Disorders, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
42
|
Garnier A, Bork NI, Jacquet E, Zipfel S, Muñoz-Guijosa C, Baczkó I, Reichenspurner H, Donzeau-Gouge P, Maier LS, Dobrev D, Girdauskas E, Nikolaev VO, Fischmeister R, Molina CE. Mapping genetic changes in the cAMP-signaling cascade in human atria. J Mol Cell Cardiol 2021; 155:10-20. [PMID: 33631188 DOI: 10.1016/j.yjmcc.2021.02.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/04/2021] [Accepted: 02/11/2021] [Indexed: 11/15/2022]
Abstract
AIM To obtain a quantitative expression profile of the main genes involved in the cAMP-signaling cascade in human control atria and in different cardiac pathologies. METHODS AND RESULTS Expression of 48 target genes playing a relevant role in the cAMP-signaling cascade was assessed by RT-qPCR. 113 samples were obtained from right atrial appendages (RAA) of patients in sinus rhythm (SR) with or without atrium dilation, paroxysmal atrial fibrillation (AF), persistent AF or heart failure (HF); and left atrial appendages (LAA) from patients in SR or with AF. Our results show that right and left atrial appendages in donor hearts or from SR patients have similar expression values except for AC7 and PDE2A. Despite the enormous chamber-dependent variability in the gene-expression changes between pathologies, several distinguishable patterns could be identified. PDE8A, PI3Kγ and EPAC2 were upregulated in AF. Different phosphodiesterase (PDE) families showed specific pathology-dependent changes. CONCLUSION By comparing mRNA-expression patterns of the cAMP-signaling cascade related genes in right and left atrial appendages of human hearts and across different pathologies, we show that 1) gene expression is not significantly affected by cardioplegic solution content, 2) it is appropriate to use SR atrial samples as controls, and 3) many genes in the cAMP-signaling cascade are affected in AF and HF but only few of them appear to be chamber (right or left) specific. TOPIC Genetic changes in human diseased atria. TRANSLATIONAL PERSPECTIVE The cyclic AMP signaling pathway is important for atrial function. However, expression patterns of the genes involved in the atria of healthy and diseased hearts are still unclear. We give here a general overview of how different pathologies affect the expression of key genes in the cAMP signaling pathway in human right and left atria appendages. Our study may help identifying new genes of interest as potential therapeutic targets or clinical biomarkers for these pathologies and could serve as a guide in future gene therapy studies.
Collapse
Affiliation(s)
- Anne Garnier
- Université Paris-Saclay, Inserm, UMR-S 1180, Châtenay-Malabry, France
| | - Nadja I Bork
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany; German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Germany
| | - Eric Jacquet
- Université Paris-Saclay, Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Gif-sur-Yvette, France
| | - Svante Zipfel
- Dept. of Cardiovascular Surgery, University Heart Center Hamburg, Germany
| | | | - Istvan Baczkó
- Dept. Pharmacology and Pharmacotherapy, Univ. of Szeged, Hungary
| | | | | | - Lars S Maier
- Dept. Internal Medicine II, University Heart Center, University Hospital Regensburg, Germany
| | - Dobromir Dobrev
- Institute of Pharmacology, West-German Heart and Vascular Center, Faculty of Medicine, University Duisburg-, Essen, Germany
| | - Evaldas Girdauskas
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Germany; Dept. of Cardiovascular Surgery, University Heart Center Hamburg, Germany
| | - Viacheslav O Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany; German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Germany
| | | | - Cristina E Molina
- Université Paris-Saclay, Inserm, UMR-S 1180, Châtenay-Malabry, France; Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany; German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Germany
| |
Collapse
|
43
|
Wang X, Chen X, Dobrev D, Li N. The crosstalk between cardiomyocyte calcium and inflammasome signaling pathways in atrial fibrillation. Pflugers Arch 2021; 473:389-405. [PMID: 33511453 DOI: 10.1007/s00424-021-02515-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/14/2020] [Accepted: 01/04/2021] [Indexed: 02/07/2023]
Abstract
Atrial fibrillation (AF) is the most frequent arrhythmia in adults. The prevalence and incidence of AF is going to increase substantially over the next few decades. Because AF increases the risk of stroke, heart failure, dementia, and others, it severely impacts the quality of life, morbidity, and mortality. Although the pathogenesis of AF is multifaceted and complex, focal ectopic activity and reentry are considered as the fundamental proarrhythmic mechanisms underlying AF development. Over the past 2 decades, large amount of evidence points to the key role of intracellular Ca2+ dysregulation in both initiation and maintenance of AF. More recently, emerging evidence reveal that NLRP3 (NACHT, LRR, PYD domain-containing 3) inflammasome pathway contributes to the substrate of both triggered activity and reentry, ultimately promoting AF. In this article, we review the current state of knowledge on Ca2+ signaling and NLRP3 inflammasome activity in AF. We also discuss the potential crosstalk between these two quintessential contributors to AF promotion.
Collapse
Affiliation(s)
- Xiaolei Wang
- Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, Houston, TX, USA
| | - Xiaohui Chen
- Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, Houston, TX, USA
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Na Li
- Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, Houston, TX, USA. .,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA. .,Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
44
|
Bernardi J, Aromolaran KA, Zhu H, Aromolaran AS. Circadian Mechanisms: Cardiac Ion Channel Remodeling and Arrhythmias. Front Physiol 2021; 11:611860. [PMID: 33519516 PMCID: PMC7841411 DOI: 10.3389/fphys.2020.611860] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 12/18/2020] [Indexed: 12/31/2022] Open
Abstract
Circadian rhythms are involved in many physiological and pathological processes in different tissues, including the heart. Circadian rhythms play a critical role in adverse cardiac function with implications for heart failure and sudden cardiac death, highlighting a significant contribution of circadian mechanisms to normal sinus rhythm in health and disease. Cardiac arrhythmias are a leading cause of morbidity and mortality in patients with heart failure and likely cause ∼250,000 deaths annually in the United States alone; however, the molecular mechanisms are poorly understood. This suggests the need to improve our current understanding of the underlying molecular mechanisms that increase vulnerability to arrhythmias. Obesity and its associated pathologies, including diabetes, have emerged as dangerous disease conditions that predispose to adverse cardiac electrical remodeling leading to fatal arrhythmias. The increasing epidemic of obesity and diabetes suggests vulnerability to arrhythmias will remain high in patients. An important objective would be to identify novel and unappreciated cellular mechanisms or signaling pathways that modulate obesity and/or diabetes. In this review we discuss circadian rhythms control of metabolic and environmental cues, cardiac ion channels, and mechanisms that predispose to supraventricular and ventricular arrhythmias including hormonal signaling and the autonomic nervous system, and how understanding their functional interplay may help to inform the development and optimization of effective clinical and therapeutic interventions with implications for chronotherapy.
Collapse
Affiliation(s)
- Joyce Bernardi
- Masonic Medical Research Institute, Utica, NY, United States
| | | | - Hua Zhu
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | | |
Collapse
|
45
|
Mechanisms underlying pathological Ca 2+ handling in diseases of the heart. Pflugers Arch 2021; 473:331-347. [PMID: 33399957 PMCID: PMC10070045 DOI: 10.1007/s00424-020-02504-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/01/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023]
Abstract
Cardiomyocyte contraction relies on precisely regulated intracellular Ca2+ signaling through various Ca2+ channels and transporters. In this article, we will review the physiological regulation of Ca2+ handling and its role in maintaining normal cardiac rhythm and contractility. We discuss how inherited variants or acquired defects in Ca2+ channel subunits contribute to the development or progression of diseases of the heart. Moreover, we highlight recent insights into the role of protein phosphatase subunits and striated muscle preferentially expressed protein kinase (SPEG) in atrial fibrillation, heart failure, and cardiomyopathies. Finally, this review summarizes current drug therapies and new advances in genome editing as therapeutic strategies for the cardiac diseases caused by aberrant intracellular Ca2+ signaling.
Collapse
|
46
|
Braga TC, de Jesus ICG, Soares KV, Guatimosim S, da Silva Neto L, da-Silva CJ, Modolo LV, Menezes Filho JER, Rhana P, Cruz JS, de Fátima Â. A novel H2S releasing-monastrol hybrid (MADTOH) inhibits L-type calcium channels. NEW J CHEM 2021. [DOI: 10.1039/d0nj04415f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
A new alleged monastrol-H2S releasing hybrid, named MADTOH, was designed based on the structure of monastrol (M) and 5-(4-hydroxyphenyl)-3H-1,2-dithiole-3-thione (ADTOH) and synthesized in 7.8% overall yield.
Collapse
Affiliation(s)
| | - Itamar Couto Guedes de Jesus
- Departamento de Fisiologia e Biofísica
- Instituto de Ciências Biológicas
- Universidade Federal de Minas Gerais
- Belo Horizonte
- Brazil
| | | | - Silvia Guatimosim
- Departamento de Fisiologia e Biofísica
- Instituto de Ciências Biológicas
- Universidade Federal de Minas Gerais
- Belo Horizonte
- Brazil
| | | | - Cristiane Jovelina da-Silva
- Departamento de Botânica
- Instituto de Ciências Biológicas
- Universidade Federal de Minas Gerais
- Belo Horizonte
- Brazil
| | - Luzia Valentina Modolo
- Departamento de Botânica
- Instituto de Ciências Biológicas
- Universidade Federal de Minas Gerais
- Belo Horizonte
- Brazil
| | | | - Paula Rhana
- Departamento de Bioquímica e Imunologia
- Instituto de Ciências Biológicas
- Universidade Federal de Minas Gerais
- Belo Horizonte
- Brazil
| | - Jader Santos Cruz
- Departamento de Bioquímica e Imunologia
- Instituto de Ciências Biológicas
- Universidade Federal de Minas Gerais
- Belo Horizonte
- Brazil
| | - Ângelo de Fátima
- Departamento de Química
- Universidade Federal de Minas Gerais
- Belo Horizonte
- Brazil
| |
Collapse
|
47
|
Wang Y, Morishima M, Li D, Takahashi N, Saikawa T, Nattel S, Ono K. Binge Alcohol Exposure Triggers Atrial Fibrillation Through T-Type Ca 2+ Channel Upregulation via Protein Kinase C (PKC) / Glycogen Synthesis Kinase 3β (GSK3β) / Nuclear Factor of Activated T-Cells (NFAT) Signaling - An Experimental Account of Holiday Heart Syndrome. Circ J 2020; 84:1931-1940. [PMID: 33028764 DOI: 10.1253/circj.cj-20-0288] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2024]
Abstract
BACKGROUND The association between binge alcohol ingestion and atrial fibrillation (AF), often termed "holiday heart syndrome", has long been recognized. However, the underlying cellular and molecular mechanisms are unknown. METHODS AND RESULTS An experimental model of binge alcohol-induced AF was developed to elucidate the mechanisms linking acute ethanol exposure to changes in ion channel transcription and AF susceptibility. AF-susceptibility during transesophageal electrical stimulation was enhanced 8 h after, but not immediately or 24 h after, acute alcohol intake. T-type calcium channel (TCC) blockade and calcineurin inhibition diminished the AF-promoting effect of ethanol. Long-term (8-24 h) exposure to ethanol augmented TCC isoform-expression (Cav3.1 and Cav3.2) and currents in cardiomyocytes, accompanied by upregulation of the transcription factors, Csx/Nkx2.5 and nuclear factor of activated T-cells (NFAT), in the nucleus, and of phospho-glycogen synthesis kinase 3β (GSK3β) in the cytosol. Inhibition of protein kinase C (PKC) during the 7- to 8-h period following ethanol exposure attenuated susceptibility to AF, whereas acute exposure did not. GSK3β inhibition itself upregulated TCC expression and increased AF susceptibility. CONCLUSIONS The present study results suggest a crucial role for TCC upregulation in the AF substrate following binge alcohol-drinking, resulting from ethanol-induced PKC-activation that hyperphosphorylates GSK3β to cause enhanced calcineurin-NFAT-Csx/Nkx2.5 signaling. These observations elucidate for the first time the potential mechanisms underlying the clinically well-recognized, but mechanistically enigmatic, "holiday heart syndrome".
Collapse
Affiliation(s)
- Yan Wang
- Department of Pathophysiology, Oita University School of Medicine
- Department of Clinical Examination and Diagnostics, Oita University School of Medicine
| | - Masaki Morishima
- Department of Pathophysiology, Oita University School of Medicine
| | - Dan Li
- Department of Pathophysiology, Oita University School of Medicine
| | - Naohiko Takahashi
- Department of Clinical Examination and Diagnostics, Oita University School of Medicine
| | - Tetsunori Saikawa
- Department of Clinical Examination and Diagnostics, Oita University School of Medicine
| | - Stanley Nattel
- Montreal Heart Institute Research Center, University of Montreal
| | - Katsushige Ono
- Department of Pathophysiology, Oita University School of Medicine
- Department of Clinical Examination and Diagnostics, Oita University School of Medicine
| |
Collapse
|
48
|
Molecular adaptation to calsequestrin 2 (CASQ2) point mutations leading to catecholaminergic polymorphic ventricular tachycardia (CPVT): comparative analysis of R33Q and D307H mutants. J Muscle Res Cell Motil 2020; 41:251-258. [PMID: 32902830 PMCID: PMC7666291 DOI: 10.1007/s10974-020-09587-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 08/29/2020] [Indexed: 12/16/2022]
Abstract
Homozygous calsequestrin 2 (CASQ2) point mutations leads to catecholaminergic polymorphic ventricular tachycardia: a common pathogenetic feature appears to be the drastic reduction of mutant CASQ2 in spite of normal transcription. Comparative biochemical analysis of R33Q and D307H knock in mutant mice identifies different pathogenetic mechanisms for CASQ2 degradation and different molecular adaptive mechanisms. In particular, each CASQ2 point mutation evokes specific adaptive cellular and molecular processes in each of the four adaptive pathways investigated. Thus, similar clinical phenotypes and identical cellular mechanism for cardiac arrhythmia might imply different molecular adaptive mechanisms.
Collapse
|
49
|
Role of Oxidation-Dependent CaMKII Activation in the Genesis of Abnormal Action Potentials in Atrial Cardiomyocytes: A Simulation Study. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1597012. [PMID: 32685443 PMCID: PMC7327560 DOI: 10.1155/2020/1597012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 05/20/2020] [Accepted: 06/02/2020] [Indexed: 01/04/2023]
Abstract
Atrial fibrillation is a common cardiac arrhythmia with an increasing incidence rate. Particularly for the aging population, understanding the underlying mechanisms of atrial arrhythmia is important in designing clinical treatment. Recently, experiments have shown that atrial arrhythmia is associated with oxidative stress. In this study, an atrial cell model including oxidative-dependent Ca2+/calmodulin- (CaM-) dependent protein kinase II (CaMKII) activation was developed to explore the intrinsic mechanisms of atrial arrhythmia induced by oxidative stress. The simulation results showed that oxidative stress caused early afterdepolarizations (EADs) of action potentials by altering the dynamics of transmembrane currents and intracellular calcium cycling. Oxidative stress gradually elevated the concentration of calcium ions in the cytoplasm by enhancing the L-type Ca2+ current and sarcoplasmic reticulum (SR) calcium release. Owing to increased intracellular calcium concentration, the inward Na+/Ca2+ exchange current was elevated which slowed down the repolarization of the action potential. Thus, the action potential was prolonged and the L-type Ca2+ current was reactivated, resulting in the genesis of EAD. Furthermore, based on the atrial single-cell model, a two-dimensional (2D) ideal tissue model was developed to explore the effect of oxidative stress on the electrical excitation wave conduction in 2D tissue. Simulation results demonstrated that, under oxidative stress conditions, EAD hindered the conduction of electrical excitation and caused an unstable spiral wave, which could disrupt normal cardiac rhythm and cause atrial arrhythmia. This study showed the effects of excess reactive oxygen species on calcium cycling and action potential in atrial myocytes and provided insights regarding atrial arrhythmia induced by oxidative stress.
Collapse
|
50
|
Case Report on: Very Early Afterdepolarizations in HiPSC-Cardiomyocytes-An Artifact by Big Conductance Calcium Activated Potassium Current (I bk,Ca). Cells 2020; 9:cells9010253. [PMID: 31968557 PMCID: PMC7017352 DOI: 10.3390/cells9010253] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 12/16/2019] [Accepted: 01/15/2020] [Indexed: 12/21/2022] Open
Abstract
Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) represent an unlimited source of human CMs that could be a standard tool in drug research. However, there is concern whether hiPSC-CMs express all cardiac ion channels at physiological level and whether they might express non-cardiac ion channels. In a control hiPSC line, we found large, “noisy” outward K+ currents, when we measured outward potassium currents in isolated hiPSC-CMs. Currents were sensitive to iberiotoxin, the selective blocker of big conductance Ca2+-activated K+ current (IBK,Ca). Seven of 16 individual differentiation batches showed a strong initial repolarization in the action potentials (AP) recorded from engineered heart tissue (EHT) followed by very early afterdepolarizations, sometimes even with consecutive oscillations. Iberiotoxin stopped oscillations and normalized AP shape, but had no effect in other EHTs without oscillations or in human left ventricular tissue (LV). Expression levels of the alpha-subunit (KCa1.1) of the BKCa correlated with the presence of oscillations in hiPSC-CMs and was not detectable in LV. Taken together, individual batches of hiPSC-CMs can express sarcolemmal ion channels that are otherwise not found in the human heart, resulting in oscillating afterdepolarizations in the AP. HiPSC-CMs should be screened for expression of non-cardiac ion channels before being applied to drug research.
Collapse
|