51
|
Ronen D, Rokach Y, Abedat S, Qadan A, Daana S, Amir O, Asleh R. Human Gut Microbiota in Cardiovascular Disease. Compr Physiol 2024; 14:5449-5490. [PMID: 39109979 DOI: 10.1002/cphy.c230012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
The gut ecosystem, termed microbiota, is composed of bacteria, archaea, viruses, protozoa, and fungi and is estimated to outnumber human cells. Microbiota can affect the host by multiple mechanisms, including the synthesis of metabolites and toxins, modulating inflammation and interaction with other organisms. Advances in understanding commensal organisms' effect on human conditions have also elucidated the importance of this community for cardiovascular disease (CVD). This effect is driven by both direct CV effects and conditions known to increase CV risk, such as obesity, diabetes mellitus (DM), hypertension, and renal and liver diseases. Cardioactive metabolites, such as trimethylamine N -oxide (TMAO), short-chain fatty acids (SCFA), lipopolysaccharides, bile acids, and uremic toxins, can affect atherosclerosis, platelet activation, and inflammation, resulting in increased CV incidence. Interestingly, this interaction is bidirectional with microbiota affected by multiple host conditions including diet, bile acid secretion, and multiple diseases affecting the gut barrier. This interdependence makes manipulating microbiota an attractive option to reduce CV risk. Indeed, evolving data suggest that the benefits observed from low red meat and Mediterranean diet consumption can be explained, at least partially, by the changes that these diets may have on the gut microbiota. In this article, we depict the current epidemiological and mechanistic understanding of the role of microbiota and CVD. Finally, we discuss the potential therapeutic approaches aimed at manipulating gut microbiota to improve CV outcomes. © 2024 American Physiological Society. Compr Physiol 14:5449-5490, 2024.
Collapse
Affiliation(s)
- Daniel Ronen
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yair Rokach
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Suzan Abedat
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Abed Qadan
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Samar Daana
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Offer Amir
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Rabea Asleh
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
52
|
Gu XH, Li W, Li H, Guo X, He J, Liu Y, Gong J, Huang Y, Zhang B. β-blockades and the risk of atrial fibrillation in patients with cardiovascular diseases. Front Pharmacol 2024; 15:1418465. [PMID: 38983917 PMCID: PMC11232185 DOI: 10.3389/fphar.2024.1418465] [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] [Received: 04/16/2024] [Accepted: 05/30/2024] [Indexed: 07/11/2024] Open
Abstract
Background β-blockers have been widely used in patients with extensive cardiovascular disease (CVD) and have provided benefits. However, they are more likely to cause symptomatic bradycardia, hypotension, or glucose metabolism disorders, which may lead to an increased risk of atrial fibrillation (AF), but evidence is lacking. Aims This study was to analyze the association between the use of β-blockers and the risk of developing AF. Methods This nationwide, prospective cohort study utilized data from the 2013-2020 National Health and Nutrition Examination Survey (NHANES). The patients were stratified into a β-blocker treatment group (n = 2585) and a non-β-blocker treatment group (n = 8525). Univariate and multivariate logistic regression analyses were performed to identify the relationship between β-blockades and the risk of AF. Propensity matching analysis was used to balance patient baseline characteristics and to control for confounders. Results A total of 11,110 subjects were included in this study (mean [SD] age, 59.89 [15.07] years; 5657 [49.7%] males). A total of 111/2585 subjects developed AF in the β-blocker treatment group, and 75/8525 developed AF in the non-β-blocker treatment group (incidence rate, 4.2% vs. 0.8%). Compared with the non-β-blocker group, the β-blocker group had an increased risk of incident AF (aOR, 2.339; 95% CI, 1.614-3.410). Some sensitivity analyses also revealed consistent findings of increased AF risk associated with β-blocker treatment. Conclusion The findings from this study suggest that β-blocker treatment is associated with an increased risk of incident AF and may help physicians select a modest medication for patients while also assessing the risk of AF.
Collapse
Affiliation(s)
- Xun-Hu Gu
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Weichao Li
- Department of Anesthesiology, Affiliated Qingyuan Hospital, Qingyuan People’s Hospital, Guangzhou Medical University, Qingyuan, China
| | - Heng Li
- Department of Anesthesiology, Affiliated Qingyuan Hospital, Qingyuan People’s Hospital, Guangzhou Medical University, Qingyuan, China
| | - Xun Guo
- Department of Anesthesiology, Affiliated Qingyuan Hospital, Qingyuan People’s Hospital, Guangzhou Medical University, Qingyuan, China
| | - Jiang He
- Department of Anesthesiology, Affiliated Qingyuan Hospital, Qingyuan People’s Hospital, Guangzhou Medical University, Qingyuan, China
| | - Yuyan Liu
- Department of Anesthesiology, Affiliated Qingyuan Hospital, Qingyuan People’s Hospital, Guangzhou Medical University, Qingyuan, China
| | - Jianping Gong
- Department of Anesthesiology, Affiliated Qingyuan Hospital, Qingyuan People’s Hospital, Guangzhou Medical University, Qingyuan, China
| | - Yizhou Huang
- Department of Anesthesiology, Affiliated Qingyuan Hospital, Qingyuan People’s Hospital, Guangzhou Medical University, Qingyuan, China
| | - Bin Zhang
- Department of Anesthesiology, Affiliated Qingyuan Hospital, Qingyuan People’s Hospital, Guangzhou Medical University, Qingyuan, China
| |
Collapse
|
53
|
Hegner P, Ofner F, Schaner B, Gugg M, Trum M, Lauerer AM, Maier LS, Arzt M, Lebek S, Wagner S. CaMKIIδ-dependent dysregulation of atrial Na + homeostasis promotes pro-arrhythmic activity in an obstructive sleep apnea mouse model. Front Pharmacol 2024; 15:1411822. [PMID: 38966545 PMCID: PMC11222670 DOI: 10.3389/fphar.2024.1411822] [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] [Received: 04/03/2024] [Accepted: 05/16/2024] [Indexed: 07/06/2024] Open
Abstract
Background Obstructive sleep apnea (OSA) has been linked to various pathologies, including arrhythmias such as atrial fibrillation. Specific treatment options for OSA are mainly limited to symptomatic approaches. We previously showed that increased production of reactive oxygen species (ROS) stimulates late sodium current through the voltage-dependent Na+ channels via Ca2+/calmodulin-dependent protein kinase IIδ (CaMKIIδ), thereby increasing the propensity for arrhythmias. However, the impact on atrial intracellular Na+ homeostasis has never been demonstrated. Moreover, the patients often exhibit a broad range of comorbidities, making it difficult to ascertain the effects of OSA alone. Objective We analyzed the effects of OSA on ROS production, cytosolic Na+ level, and rate of spontaneous arrhythmia in atrial cardiomyocytes isolated from an OSA mouse model free from comorbidities. Methods OSA was induced in C57BL/6 wild-type and CaMKIIδ-knockout mice by polytetrafluorethylene (PTFE) injection into the tongue. After 8 weeks, their atrial cardiomyocytes were analyzed for cytosolic and mitochondrial ROS production via laser-scanning confocal microscopy. Quantifications of the cytosolic Na+ concentration and arrhythmia were performed by epifluorescence microscopy. Results PTFE treatment resulted in increased cytosolic and mitochondrial ROS production. Importantly, the cytosolic Na+ concentration was dramatically increased at various stimulation frequencies in the PTFE-treated mice, while the CaMKIIδ-knockout mice were protected. Accordingly, the rate of spontaneous Ca2+ release events increased in the wild-type PTFE mice while being impeded in the CaMKIIδ-knockout mice. Conclusion Atrial Na+ concentration and propensity for spontaneous Ca2+ release events were higher in an OSA mouse model in a CaMKIIδ-dependent manner, which could have therapeutic implications.
Collapse
Affiliation(s)
- Philipp Hegner
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Florian Ofner
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Benedikt Schaner
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
- Department of Neurology and Clinical Neurophysiology, University Hospital Augsburg, Augsburg, Germany
| | - Mathias Gugg
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Maximilian Trum
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Anna-Maria Lauerer
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Lars Siegfried Maier
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Michael Arzt
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Simon Lebek
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Stefan Wagner
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
54
|
Shen J, Fu H, Ding Y, Yuan Z, Xiang Z, Ding M, Huang M, Peng Y, Li T, Zha K, Ye Q. The role of iron overload and ferroptosis in arrhythmia pathogenesis. IJC HEART & VASCULATURE 2024; 52:101414. [PMID: 38694269 PMCID: PMC11060960 DOI: 10.1016/j.ijcha.2024.101414] [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: 01/21/2024] [Revised: 04/20/2024] [Accepted: 04/23/2024] [Indexed: 05/04/2024]
Abstract
Ferroptosis is a newly discovered form of programmed cell death triggered by intracellular iron overload, which leads to the accumulation of lipid peroxides in various cells. It has been implicated in the pathogenesis and progression of various diseases, including tumors, neurological disorders, and cardiovascular diseases. The intricate mechanism underlying ferroptosis involves an imbalance between the oxidation and antioxidant systems, disturbances in iron metabolism, membrane lipid peroxidation, and dysregulation of amino acid metabolism. We highlight the key molecular mechanisms governing iron overload and ferroptosis, and discuss potential molecular pathways linking ferroptosis with arrhythmias.
Collapse
Affiliation(s)
- Jingsong Shen
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Hengsong Fu
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Yanling Ding
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Ziyang Yuan
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Zeming Xiang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Miao Ding
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Min Huang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Yongquan Peng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Tao Li
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
| | - Kelan Zha
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Qiang Ye
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
55
|
Zhong J, Chen H, Liu Q, Zhou S, Liu Z, Xiao Y. GLP-1 receptor agonists and myocardial metabolism in atrial fibrillation. J Pharm Anal 2024; 14:100917. [PMID: 38799233 PMCID: PMC11127228 DOI: 10.1016/j.jpha.2023.12.007] [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: 07/02/2023] [Revised: 10/15/2023] [Accepted: 12/07/2023] [Indexed: 05/29/2024] Open
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia. Many medical conditions, including hypertension, diabetes, obesity, sleep apnea, and heart failure (HF), increase the risk for AF. Cardiomyocytes have unique metabolic characteristics to maintain adenosine triphosphate production. Significant changes occur in myocardial metabolism in AF. Glucagon-like peptide-1 receptor agonists (GLP-1RAs) have been used to control blood glucose fluctuations and weight in the treatment of type 2 diabetes mellitus (T2DM) and obesity. GLP-1RAs have also been shown to reduce oxidative stress, inflammation, autonomic nervous system modulation, and mitochondrial function. This article reviews the changes in metabolic characteristics in cardiomyocytes in AF. Although the clinical trial outcomes are unsatisfactory, the findings demonstrate that GLP-1 RAs can improve myocardial metabolism in the presence of various risk factors, lowering the incidence of AF.
Collapse
Affiliation(s)
- Jiani Zhong
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Xiangya School of Medicine, Central South University, Changsha, 410008, China
| | - Hang Chen
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Xiangya School of Medicine, Central South University, Changsha, 410008, China
| | - Qiming Liu
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Shenghua Zhou
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Zhenguo Liu
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, School of Medicine, University of Missouri, Columbia, MO, 65211, USA
| | - Yichao Xiao
- Department of Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| |
Collapse
|
56
|
Seitz J, Mohr Durdez T, Lotteau S, Bars C, Pisapia A, Gitenay E, Monteau J, Reist M, Serdi M, Dayot A, Bremondy M, Benadel M, Siame S, Appetiti A, Milpied P, Kalifa J. Artificial intelligence-adjudicated spatiotemporal dispersion: A patient-unique fingerprint of persistent atrial fibrillation. Heart Rhythm 2024; 21:540-552. [PMID: 38215808 DOI: 10.1016/j.hrthm.2024.01.007] [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: 07/05/2023] [Revised: 12/27/2023] [Accepted: 01/06/2024] [Indexed: 01/14/2024]
Abstract
BACKGROUND Spatiotemporal dispersion-guided ablation is a tailored approach for patients in persistent atrial fibrillation (PsAF). The characterization of dispersion extent and distribution and its association with common clinical descriptors of PsAF patients has not been studied. OBJECTIVES Artificial intelligence-adjudicated dispersion extent and distribution (AI-DED) was obtained with a machine/deep learning classifier (VX1 Software, Volta Medical) in PsAF patients undergoing ablation. The purpose of this study was to test the hypothesis that AI-DED is unique to each patient and independent of common procedural and clinical parameters. METHODS In a subanalysis of the Ev-AIFib study (NCT03434964), spatiotemporal dispersion maps were built with VX1 software in 78 consecutive persistent and long-standing PsAF patients. AI-DED was quantified using 2 distinct approaches (visual regional characterization or automated global quantification of AI-DED). RESULTS AI-DED paired-subregion Euclidean distance measurements between 78 patients (average distance 5.07 ± 0.60; min 2.23; max 9.75) demonstrate that AI-DED is a patient-unique characteristic of PsAF. Importantly, both AF type and AF history do not correlate with AI-DED levels (R2 = 0.006, P = .53; and R2 = 0.03, P = .25, respectively). The most extensive AI-DED levels are not associated with poorer procedural (83%, 81%, and 83% of AF termination in low, medium, and high dispersion groups, respectively; P = .954) and long-term (88%, 75%, and 91% of freedom from AF/atrial tachycardia after multiple procedures; P = .517) outcomes. CONCLUSION The atrial distribution and extent of multipolar electrogram spatiotemporal dispersion follow a nonrandom, albeit patient-unique, distribution in PsAF patients. AI-DED may represent a procedure-implementable fingerprint of the PsAF substrate.
Collapse
|
57
|
Burg S, Levi O, Elyagon S, Shapiro S, Murninkas M, Etzion S, Gradwohl G, Makarovsky D, Lichtenstein A, Gordon Y, Attali B, Etzion Y. The SK4 channel allosteric blocker, BA6b9, reduces atrial fibrillation substrate in rats with reduced ejection fraction. PNAS NEXUS 2024; 3:pgae192. [PMID: 38783894 PMCID: PMC11114471 DOI: 10.1093/pnasnexus/pgae192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 04/30/2024] [Indexed: 05/25/2024]
Abstract
Atrial fibrillation (AF), the most common cardiac arrhythmia, is strongly associated with several comorbidities including heart failure (HF). AF in general, and specifically in the context of HF, is progressive in nature and associated with poor clinical outcomes. Current therapies for AF are limited in number and efficacy and do not target the underlying causes of atrial remodeling such as inflammation or fibrosis. We previously identified the calcium-activated SK4 K+ channels, which are preferentially expressed in the atria relative to the ventricles in both rat and human hearts, as attractive druggable target for AF treatment. Here, we examined the ability of BA6b9, a novel allosteric inhibitor of SK4 channels that targets the specific calmodulin-PIP2 binding domain, to alter AF susceptibility and atrial remodeling in a systolic HF rat postmyocardial infarction (post-MI) model. Daily BA6b9 injection (20 mg/kg/day) for 3 weeks starting 1-week post-MI prolonged the atrial effective refractory period, reduced AF induction and duration, and dramatically prevented atrial structural remodeling. In the post-MI left atrium (LA), pronounced upregulation of the SK4 K+ channel was observed, with corresponding increases in collagen deposition, α-SMA levels, and NLRP3 inflammasome expression. Strikingly, BA6b9 treatment reversed these changes while also significantly reducing the lateralization of the atrial connexin Cx43 in the LA of post-MI rats. Our findings indicate that the blockade of SK4 K+ channels using BA6b9 not only favors rhythm control but also remarkably reduces atrial structural remodeling, a property that is highly desirable for novel AF therapies, particularly in patients with comorbid HF.
Collapse
Affiliation(s)
- Shira Burg
- Department of Physiology & Pharmacology, Sackler Faculty of Medicine and Sagol School of Neurosciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Or Levi
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Sigal Elyagon
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Shir Shapiro
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Michael Murninkas
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Sharon Etzion
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Gideon Gradwohl
- Medical Engineering Unit, The Jerusalem College of Technology, Jerusalem 9116001, Israel
| | - Daria Makarovsky
- Inter-Departmental Core Facility, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Alexandra Lichtenstein
- Inter-Departmental Core Facility, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Yaara Gordon
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Bernard Attali
- Department of Physiology & Pharmacology, Sackler Faculty of Medicine and Sagol School of Neurosciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Yoram Etzion
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| |
Collapse
|
58
|
Wang Y, Gu YH, Ren KW, Xie X, Wang SH, Zhu XX, Wang L, Yang XL, Bi HL. Administration of USP7 inhibitor p22077 alleviates Angiotensin II (Ang II)-induced atrial fibrillation in Mice. Hypertens Res 2024; 47:1309-1322. [PMID: 38374239 DOI: 10.1038/s41440-024-01581-2] [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: 07/10/2023] [Revised: 11/21/2023] [Accepted: 12/24/2023] [Indexed: 02/21/2024]
Abstract
Atrial fibrillation (AF), the most common cardiac arrhythmia, is an important contributor to mortality and morbidity. Ubquitin-specific protease 7 (USP7), one of the most abundant ubiquitin-specific proteases (USP), participated in many cellular events, such as cell proliferation, apoptosis, and tumourigenesis. However, its role in AF remains unknown. Here, the mice were treated with Ang II infusion to induce the AF model. Echocardiography was used to measure the atrial diameter. Electrical stimulation was programmed to measure the induction and duration of AF. The changes in atrial remodeling were measured using routine histologic analysis. Here, a significant increase in USP7 expression was observed in Ang II-stimulated atrial cardiomyocytes and atrial tissues, as well as in atrial tissues from patients with AF. The administration of p22077, the inhibitor of USP7, attenuated Ang II-induced inducibility and duration of AF, atrial dilatation, connexin dysfunction, atrial fibrosis, atrial inflammation, and atrial oxidase stress, and then inhibited the progression of AF. Mechanistically, the administration of p22077 alleviated Ang II-induced activation of TGF-β/Smad2, NF-κB/NLRP3, NADPH oxidases (NOX2 and NOX4) signals, the up-regulation of CX43, ox-CaMKII, CaMKII, Kir2.1, and down-regulation of SERCA2a. Together, this study, for the first time, suggests that USP7 is a critical driver of AF and revealing USP7 may present a new target for atrial fibrillation therapeutic strategies.
Collapse
Affiliation(s)
- Yu Wang
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yu-Hui Gu
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Kai-Wen Ren
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xin Xie
- Department of pharmacology, College of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Shi-Hao Wang
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiao-Xue Zhu
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Lei Wang
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiao-Lei Yang
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China.
- Department of Cardiology, First Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Hai-Lian Bi
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China.
| |
Collapse
|
59
|
Lin AE, Bapat AC, Xiao L, Niroula A, Ye J, Wong WJ, Agrawal M, Farady CJ, Boettcher A, Hergott CB, McConkey M, Flores-Bringas P, Shkolnik V, Bick AG, Milan D, Natarajan P, Libby P, Ellinor PT, Ebert BL. Clonal Hematopoiesis of Indeterminate Potential With Loss of Tet2 Enhances Risk for Atrial Fibrillation Through Nlrp3 Inflammasome Activation. Circulation 2024; 149:1419-1434. [PMID: 38357791 PMCID: PMC11058018 DOI: 10.1161/circulationaha.123.065597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 01/10/2024] [Indexed: 02/16/2024]
Abstract
BACKGROUND Clonal hematopoiesis of indeterminate potential (CHIP), a common age-associated phenomenon, associates with increased risk of both hematological malignancy and cardiovascular disease. Although CHIP is known to increase the risk of myocardial infarction and heart failure, the influence of CHIP in cardiac arrhythmias, such as atrial fibrillation (AF), is less explored. METHODS CHIP prevalence was determined in the UK Biobank, and incident AF analysis was stratified by CHIP status and clone size using Cox proportional hazard models. Lethally irradiated mice were transplanted with hematopoietic-specific loss of Tet2, hematopoietic-specific loss of Tet2 and Nlrp3, or wild-type control and fed a Western diet, compounded with or without NLRP3 (NLR [NACHT, LRR {leucine rich repeat}] family pyrin domain containing protein 3) inhibitor, NP3-361, for 6 to 9 weeks. Mice underwent in vivo invasive electrophysiology studies and ex vivo optical mapping. Cardiomyocytes from Ldlr-/- mice with hematopoietic-specific loss of Tet2 or wild-type control and fed a Western diet were isolated to evaluate calcium signaling dynamics and analysis. Cocultures of pluripotent stem cell-derived atrial cardiomyocytes were incubated with Tet2-deficient bone marrow-derived macrophages, wild-type control, or cytokines IL-1β (interleukin 1β) or IL-6 (interleukin 6). RESULTS Analysis of the UK Biobank showed individuals with CHIP, in particular TET2 CHIP, have increased incident AF. Hematopoietic-specific inactivation of Tet2 increases AF propensity in atherogenic and nonatherogenic mouse models and is associated with increased Nlrp3 expression and CaMKII (Ca2+/calmodulin-dependent protein kinase II) activation, with AF susceptibility prevented by inactivation of Nlrp3. Cardiomyocytes isolated from Ldlr-/- mice with hematopoietic inactivation of Tet2 and fed a Western diet have impaired calcium release from the sarcoplasmic reticulum into the cytosol, contributing to atrial arrhythmogenesis. Abnormal sarcoplasmic reticulum calcium release was recapitulated in cocultures of cardiomyocytes with the addition of Tet2-deficient macrophages or cytokines IL-1β or IL-6. CONCLUSIONS We identified a modest association between CHIP, particularly TET2 CHIP, and incident AF in the UK Biobank population. In a mouse model of AF resulting from hematopoietic-specific inactivation of Tet2, we propose altered calcium handling as an arrhythmogenic mechanism, dependent on Nlrp3 inflammasome activation. Our data are in keeping with previous studies of CHIP in cardiovascular disease, and further studies into the therapeutic potential of NLRP3 inhibition for individuals with TET2 CHIP may be warranted.
Collapse
Affiliation(s)
- Amy Erica Lin
- Division of Cardiovascular Medicine, Department of Medicine (A.E.L., P.L.), Brigham and Women’s Hospital, Boston, MA
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA (A.E.L., A.N., M.A., C.B.H., M.M.C., V.S., B.L.E.)
| | - Aneesh C. Bapat
- Cardiovascular Research Center (A.C.B., L.X., J.Y., D.M., P.N., P.T.E.), Massachusetts General Hospital, Boston
- Demoulas Cardiac Arrhythmia Service, Division of Cardiovascular Medicine, Department of Medicine (A.C.B., P.T.E.), Massachusetts General Hospital, Boston
| | - Ling Xiao
- Cardiovascular Research Center (A.C.B., L.X., J.Y., D.M., P.N., P.T.E.), Massachusetts General Hospital, Boston
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge (L.X., A.N., J.Y., P.F.-B., P.N., P.T.E., B.L.E.)
| | - Abhishek Niroula
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA (A.E.L., A.N., M.A., C.B.H., M.M.C., V.S., B.L.E.)
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge (L.X., A.N., J.Y., P.F.-B., P.N., P.T.E., B.L.E.)
- Department of Laboratory Medicine, Lund University, Sweden (A.N.)
- Institute of Biomedicine, SciLifeLab, University of Gothenburg, Sweden (A.N.)
| | - Jiangchuan Ye
- Cardiovascular Research Center (A.C.B., L.X., J.Y., D.M., P.N., P.T.E.), Massachusetts General Hospital, Boston
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge (L.X., A.N., J.Y., P.F.-B., P.N., P.T.E., B.L.E.)
| | - Waihay J. Wong
- Department of Pathology (W.J.W., C.B.H.), Brigham and Women’s Hospital, Boston, MA
| | - Mridul Agrawal
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA (A.E.L., A.N., M.A., C.B.H., M.M.C., V.S., B.L.E.)
| | - Christopher J. Farady
- Novartis Institutes for BioMedical Research Forum 1, Basel, Switzerland (C.J.F., A.B.)
| | - Andreas Boettcher
- Novartis Institutes for BioMedical Research Forum 1, Basel, Switzerland (C.J.F., A.B.)
| | - Christopher B. Hergott
- Department of Pathology (W.J.W., C.B.H.), Brigham and Women’s Hospital, Boston, MA
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA (A.E.L., A.N., M.A., C.B.H., M.M.C., V.S., B.L.E.)
| | - Marie McConkey
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA (A.E.L., A.N., M.A., C.B.H., M.M.C., V.S., B.L.E.)
| | - Patricio Flores-Bringas
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge (L.X., A.N., J.Y., P.F.-B., P.N., P.T.E., B.L.E.)
| | - Veronica Shkolnik
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA (A.E.L., A.N., M.A., C.B.H., M.M.C., V.S., B.L.E.)
| | - Alexander G. Bick
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN (A.G.B.)
| | - David Milan
- Cardiovascular Research Center (A.C.B., L.X., J.Y., D.M., P.N., P.T.E.), Massachusetts General Hospital, Boston
- Leducq Foundation, Boston, MA (D.M.)
| | - Pradeep Natarajan
- Cardiovascular Research Center (A.C.B., L.X., J.Y., D.M., P.N., P.T.E.), Massachusetts General Hospital, Boston
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge (L.X., A.N., J.Y., P.F.-B., P.N., P.T.E., B.L.E.)
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine (A.E.L., P.L.), Brigham and Women’s Hospital, Boston, MA
| | - Patrick T. Ellinor
- Cardiovascular Research Center (A.C.B., L.X., J.Y., D.M., P.N., P.T.E.), Massachusetts General Hospital, Boston
- Demoulas Cardiac Arrhythmia Service, Division of Cardiovascular Medicine, Department of Medicine (A.C.B., P.T.E.), Massachusetts General Hospital, Boston
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge (L.X., A.N., J.Y., P.F.-B., P.N., P.T.E., B.L.E.)
| | - Benjamin L. Ebert
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA (A.E.L., A.N., M.A., C.B.H., M.M.C., V.S., B.L.E.)
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge (L.X., A.N., J.Y., P.F.-B., P.N., P.T.E., B.L.E.)
- Howard Hughes Medical Institute, Boston, MA (B.L.E.)
| |
Collapse
|
60
|
Li L, Li L, Yang D, Nong S, Luo C, Gui C. Analysis of the thickness characteristics of the left atrial posterior wall and its correlation with the low and no voltage areas of the left atrial posterior wall in patients with atrial fibrillation. J Cardiothorac Surg 2024; 19:187. [PMID: 38582871 PMCID: PMC10998308 DOI: 10.1186/s13019-024-02658-2] [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: 07/20/2023] [Accepted: 03/19/2024] [Indexed: 04/08/2024] Open
Abstract
OBJECTIVE To analyze the relationship between the thickness of the left atrial posterior wall and the low and no voltage zones in the left atrial posterior wall in patients with atrial fibrillation (AF). METHODS 61 patients admitted to our cardiology department for AF and radiofrequency ablation of AF from January 1, 2020 to May 30, 2022 were enrolled according to inclusion and exclusion criteria. The atrial wall thickness was measured by CT scan. Baseline data, preoperative cardiac ultrasound data, preoperative biochemical parameters, low voltage zone (fibrotic zone) and no voltage zone (scar zone) in the left atrial posterior wall area, and various parameters of posterior left atrial wall thickness were collected. RESULTS The differences of the thickness between the upper, middle and lower mean levels of the left atrial posterior wall were statistically significant (P = 0.004). The results showed that body mass index was weakly positively correlated with the mean level of total left atrial posterior wall thickness (r = 0.426, P = 0.001) and was statistically significant. The remaining indices were positively or negatively correlated with the mean level of total left atrial posterior wall thickness, but none were statistically significant (P > 0.05). CONCLUSIONS Both left atrial posterior wall low-voltage zone and voltage-free zone were positively correlated with the mean total left atrial posterior wall thickness, and left atrial posterior wall low-voltage zone and voltage-free zone were significantly positively correlated. Body mass index was weakly positively correlated with total left atrial posterior wall thickness.
Collapse
Affiliation(s)
- Longchang Li
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Department of Cardiology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, 530022, Guangxi, People's Republic of China
| | - Lijun Li
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Department of Cardiology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, 530022, Guangxi, People's Republic of China
| | - Dezhi Yang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Department of Cardiology Second Ward, Hechi People's Hospital, Hechi, 547000, Guangxi, People's Republic of China
| | - Shuxiong Nong
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Nanning, 530021, Guangxi, People's Republic of China
- Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning, 530021, Guangxi, People's Republic of China
| | - Cheng Luo
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Nanning, 530021, Guangxi, People's Republic of China
- Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning, 530021, Guangxi, People's Republic of China
| | - Chun Gui
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China.
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Nanning, 530021, Guangxi, People's Republic of China.
- Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning, 530021, Guangxi, People's Republic of China.
| |
Collapse
|
61
|
Li B, Lin M, Wu L. Drug-induced AF: Arrhythmogenic Mechanisms and Management Strategies. Arrhythm Electrophysiol Rev 2024; 13:e06. [PMID: 38706787 PMCID: PMC11066853 DOI: 10.15420/aer.2023.24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 01/12/2024] [Indexed: 05/07/2024] Open
Abstract
AF is a prevalent condition that is associated with various modifiable and unmodifiable risk factors. Drug-induced AF, despite being commonly under-recognised, can be relatively easy to manage. Numerous cardiovascular and non-cardiovascular agents, including catecholaminergic agents, adenosine, anti-tumour agents and others, have been reported to induce AF. However, the mechanisms underlying drug-induced AF are diverse and not fully understood. The complexity of clinical scenarios and insufficient knowledge regarding drug-induced AF have rendered the management of this condition complicated, and current treatment guidelines follow those for other types of AF. Here, we present a review of the epidemiology of drug-induced AF and highlight a range of drugs that can induce or exacerbate AF, along with their molecular and electrophysiological mechanisms. Given the inadequate evidence and lack of attention, further research is crucial to underscore the clinical significance of drug-induced AF, clarify the underlying mechanisms and develop effective treatment strategies for the condition.
Collapse
Affiliation(s)
- Bingxun Li
- Department of Cardiology, Peking University First HospitalBeijing, China
| | - Mingjie Lin
- Department of Cardiology, Qilu Hospital of Shandong University Qingdao BranchQingdao, China
| | - Lin Wu
- Department of Cardiology, Peking University First HospitalBeijing, China
- Key Laboratory of Medical Electrophysiology of the Ministry of Education and Institute of Cardiovascular Research, Southwest Medical UniversityLuzhou, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking UniversityBeijing, China
| |
Collapse
|
62
|
Zhang Z, Vlcek J, Pauly V, Hesse N, Bauer J, Chataut KR, Maderspacher F, Volz LS, Buchberger K, Xia R, Hildebrand B, Kääb S, Schüttler D, Tomsits P, Clauss S. Atrial fibrosis heterogeneity is a risk for atrial fibrillation in pigs with ischaemic heart failure. Eur J Clin Invest 2024; 54:e14137. [PMID: 38012826 DOI: 10.1111/eci.14137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 11/02/2023] [Accepted: 11/18/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND Atrial fibrillation (AF) is the most common arrhythmia and is associated with considerable morbidity and mortality. Ischaemic heart failure (IHF) remains one of the most common causes of AF in clinical practice. However, ischaemia-mediated mechanisms leading to AF are still incompletely understood, and thus, current treatment approaches are limited. To improve our understanding of the pathophysiology, we studied a porcine IHF model. METHODS In pigs, IHF was induced by balloon occlusion of the left anterior descending artery for 90 min. After 30 days of reperfusion, invasive haemodynamic measurements and electrophysiological studies were performed. Masson trichrome and immunofluorescence staining were conducted to assess interstitial fibrosis and myofibroblast activation in different heart regions. RESULTS After 30 days of reperfusion, heart failure with significantly reduced ejection fraction (left anterior obique 30°, 34.78 ± 3.29% [IHF] vs. 62.03 ± 2.36% [control], p < .001; anterior-posterior 0°, 29.16 ± 3.61% vs. 59.54 ± 1.09%, p < .01) was observed. These pigs showed a significantly higher susceptibility to AF (33.90% [IHF] vs. 12.98% [control], p < .05). Histological assessment revealed aggravated fibrosis in atrial appendages but not in atrial free walls in IHF pigs (11.13 ± 1.44% vs. 5.99 ± .86%, p < .01 [LAA], 8.28 ± .56% vs. 6.01 ± .35%, p < .01 [RAA]), which was paralleled by enhanced myofibroblast activation (12.09 ± .65% vs. 9.00 ± .94%, p < .05 [LAA], 14.37 ± .60% vs. 10.30 ± 1.41%, p < .05 [RAA]). Correlation analysis indicated that not fibrosis per se but its cross-regional heterogeneous distribution across the left atrium was associated with AF susceptibility (r = .6344, p < .01). CONCLUSION Our results suggest that left atrial cross-regional fibrosis difference rather than overall fibrosis level is associated with IHF-related AF susceptibility, presumably by establishing local conduction disturbances and heterogeneity.
Collapse
Affiliation(s)
- Zhihao Zhang
- Department of Medicine I, Campus Grosshadern, University Hospital Munich, Ludwig-Maximilians University (LMU), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Julia Vlcek
- Department of Medicine I, Campus Grosshadern, University Hospital Munich, Ludwig-Maximilians University (LMU), Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Valerie Pauly
- Department of Medicine I, Campus Grosshadern, University Hospital Munich, Ludwig-Maximilians University (LMU), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Nora Hesse
- Department of Medicine I, Campus Grosshadern, University Hospital Munich, Ludwig-Maximilians University (LMU), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Julia Bauer
- Department of Medicine I, Campus Grosshadern, University Hospital Munich, Ludwig-Maximilians University (LMU), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Kavi Raj Chataut
- Department of Medicine I, Campus Grosshadern, University Hospital Munich, Ludwig-Maximilians University (LMU), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Florian Maderspacher
- Department of Medicine I, Campus Grosshadern, University Hospital Munich, Ludwig-Maximilians University (LMU), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Lina Sophie Volz
- Department of Medicine I, Campus Grosshadern, University Hospital Munich, Ludwig-Maximilians University (LMU), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Katharina Buchberger
- Department of Medicine I, Campus Grosshadern, University Hospital Munich, Ludwig-Maximilians University (LMU), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Ruibing Xia
- Department of Medicine I, Campus Grosshadern, University Hospital Munich, Ludwig-Maximilians University (LMU), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Bianca Hildebrand
- Department of Medicine I, Campus Grosshadern, University Hospital Munich, Ludwig-Maximilians University (LMU), Munich, Germany
| | - Stefan Kääb
- Department of Medicine I, Campus Grosshadern, University Hospital Munich, Ludwig-Maximilians University (LMU), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
| | - Dominik Schüttler
- Department of Medicine I, Campus Grosshadern, University Hospital Munich, Ludwig-Maximilians University (LMU), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Philipp Tomsits
- Department of Medicine I, Campus Grosshadern, University Hospital Munich, Ludwig-Maximilians University (LMU), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Sebastian Clauss
- Department of Medicine I, Campus Grosshadern, University Hospital Munich, Ludwig-Maximilians University (LMU), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
| |
Collapse
|
63
|
Jin J, Wang Y, Liu Y, Chakrabarti S, Su Z. Cardiac resident macrophages: Spatiotemporal distribution, development, physiological functions, and their translational potential on cardiac diseases. Acta Pharm Sin B 2024; 14:1483-1493. [PMID: 38572111 PMCID: PMC10985034 DOI: 10.1016/j.apsb.2023.12.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/27/2023] [Accepted: 12/29/2023] [Indexed: 04/05/2024] Open
Abstract
Cardiac resident macrophages (CRMs) are the main population of cardiac immune cells. The role of these cells in regeneration, functional remodeling, and repair after cardiac injury is always the focus of research. However, in recent years, their dynamic changes and contributions in physiological states have a significant attention. CRMs have specific phenotypes and functions in different cardiac chambers or locations of the heart and at different stages. They further show specific differentiation and development processes. The present review will summarize the new progress about the spatiotemporal distribution, potential developmental regulation, and their roles in cardiac development and aging as well as the translational potential of CRMs on cardiac diseases. Of course, the research tools for CRMs, their respective advantages and disadvantages, and key issues on CRMs will further be discussed.
Collapse
Affiliation(s)
- Jing Jin
- International Genome Center, Jiangsu University, Zhenjiang 212013, China
- Institute for Medical Immunology, Jiangsu University, Zhenjiang 212013, China
| | - Yurou Wang
- International Genome Center, Jiangsu University, Zhenjiang 212013, China
- Institute for Medical Immunology, Jiangsu University, Zhenjiang 212013, China
| | - Yueqin Liu
- Center Laboratory, the Fourth People's Hospital of Zhenjiang, Zhenjiang 212008, China
| | - Subrata Chakrabarti
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario N6A 5C1, Canada
| | - Zhaoliang Su
- International Genome Center, Jiangsu University, Zhenjiang 212013, China
- Institute for Medical Immunology, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
64
|
Roberts JD, Chalazan B, Andrade JG, Macle L, Nattel S, Tadros R. Clinical Genetic Testing for Atrial Fibrillation: Are We There Yet? Can J Cardiol 2024; 40:540-553. [PMID: 38551553 DOI: 10.1016/j.cjca.2023.11.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 10/17/2023] [Accepted: 11/19/2023] [Indexed: 04/13/2024] Open
Abstract
Important progress has been made toward unravelling the complex genetics underlying atrial fibrillation (AF). Initial studies were aimed to identify monogenic causes; however, it has become increasingly clear that the most common predisposing genetic substrate for AF is polygenic. Despite intensive investigations, there is robust evidence for rare variants for only a limited number of genes and cases. Although the current yield for genetic testing in early onset AF might be modest, there is an increasing appreciation that genetic culprits for potentially life-threatening ventricular cardiomyopathies and channelopathies might initially present with AF. The potential clinical significance of this recognition is highlighted by evidence that suggests that identification of a pathogenic or likely pathogenic rare variant in a patient with early onset AF is associated with an increased risk of death. These findings suggest that it might be warranted to screen patients with early onset AF for these potentially more sinister cardiac conditions. Beyond facilitating the early identification of genetic culprits associated with potentially malignant phenotypes, insight into underlying AF genetic substrates might improve the selection of patients for existing therapies and guide the development of novel ones. Herein, we review the evidence that links genetic factors to AF, then discuss an approach to using genetic testing for early onset AF patients in the present context, and finally consider the potential value of genetic testing in the foreseeable future. Although further work might be necessary before recommending uniform integration of genetic testing in cases of early onset AF, ongoing research increasingly highlights its potential contributions to clinical care.
Collapse
Affiliation(s)
- Jason D Roberts
- Population Health Research Institute, McMaster University, and Hamilton Health Sciences, Hamilton, Ontario, Canada.
| | - Brandon Chalazan
- Division of Biochemical Genetics, Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jason G Andrade
- Centre for Cardiovascular Innovation and Division of Cardiology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Laurent Macle
- Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada
| | - Stanley Nattel
- Department of Medicine and Research Center, Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada
| | - Rafik Tadros
- Cardiovascular Genetics Center, Montreal Heart Institute, Faculty of Medicine, Université de Montréal, Montreal, Québec, Canada
| |
Collapse
|
65
|
Ninni S, Algalarrondo V, Brette F, Lemesle G, Fauconnier J. Left atrial cardiomyopathy: Pathophysiological insights, assessment methods and clinical implications. Arch Cardiovasc Dis 2024; 117:283-296. [PMID: 38490844 DOI: 10.1016/j.acvd.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 02/01/2024] [Accepted: 02/06/2024] [Indexed: 03/17/2024]
Abstract
Atrial cardiomyopathy is defined as any complex of structural, architectural, contractile or electrophysiological changes affecting atria, with the potential to produce clinically relevant manifestations. Most of our knowledge about the mechanistic aspects of atrial cardiomyopathy is derived from studies investigating animal models of atrial fibrillation and atrial tissue samples obtained from individuals who have a history of atrial fibrillation. Several noninvasive tools have been reported to characterize atrial cardiomyopathy in patients, which may be relevant for predicting the risk of incident atrial fibrillation and its related outcomes, such as stroke. Here, we provide an overview of the pathophysiological mechanisms involved in atrial cardiomyopathy, and discuss the complex interplay of these mechanisms, including aging, left atrial pressure overload, metabolic disorders and genetic factors. We discuss clinical tools currently available to characterize atrial cardiomyopathy, including electrocardiograms, cardiac imaging and serum biomarkers. Finally, we discuss the clinical impact of atrial cardiomyopathy, and its potential role for predicting atrial fibrillation, stroke, heart failure and dementia. Overall, this review aims to highlight the critical need for a clinically relevant definition of atrial cardiomyopathy to improve treatment strategies.
Collapse
Affiliation(s)
- Sandro Ninni
- CHU de Lille, Université de Lille, 59000 Lille, France.
| | - Vincent Algalarrondo
- Department of Cardiology, Bichat University Hospital, AP-HP, 75018 Paris, France
| | - Fabien Brette
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34093 Montpellier, France
| | | | - Jérémy Fauconnier
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34093 Montpellier, France
| |
Collapse
|
66
|
Tubeeckx MRL, De Keulenaer GW, Heidbuchel H, Segers VFM. Pathophysiology and clinical relevance of atrial myopathy. Basic Res Cardiol 2024; 119:215-242. [PMID: 38472506 DOI: 10.1007/s00395-024-01038-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 02/01/2024] [Accepted: 02/02/2024] [Indexed: 03/14/2024]
Abstract
Atrial myopathy is a condition that consists of electrical, structural, contractile, and autonomic remodeling of the atria and is the substrate for development of atrial fibrillation, the most common arrhythmia. Pathophysiologic mechanisms driving atrial myopathy are inflammation, oxidative stress, atrial stretch, and neurohormonal signals, e.g., angiotensin-II and aldosterone. These mechanisms initiate the structural and functional remodeling of the atrial myocardium. Novel therapeutic strategies are being developed that target the pathophysiologic mechanisms of atrial myopathy. In this review, we will discuss the pathophysiology of atrial myopathy, as well as diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Michiel R L Tubeeckx
- Laboratory of Physiopharmacology, Universiteitsplein 1, Building T (2nd Floor), 2610, Antwerp, Belgium.
| | - Gilles W De Keulenaer
- Laboratory of Physiopharmacology, Universiteitsplein 1, Building T (2nd Floor), 2610, Antwerp, Belgium
- Department of Cardiology, ZNA Middelheim Hospital Antwerp, Antwerp, Belgium
| | - Hein Heidbuchel
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, University Hospital Antwerp, Antwerp, Belgium
| | - Vincent F M Segers
- Laboratory of Physiopharmacology, Universiteitsplein 1, Building T (2nd Floor), 2610, Antwerp, Belgium
- Department of Cardiology, University Hospital Antwerp, Antwerp, Belgium
| |
Collapse
|
67
|
Zakharov IP, Chomakhidze PS, Kopylov FY, Sultygova EA, Mesitskaya DF, Lyubimova EA, Andreev DA. Determining The Risk of Atrial Fibrillation Paroxysm in Patients With Chronic Heart Failure With Intact and Reduced Ejection Fraction. KARDIOLOGIIA 2024; 64:25-33. [PMID: 38597759 DOI: 10.18087/cardio.2024.3.n2466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/23/2023] [Indexed: 04/11/2024]
Abstract
AIM To determine predictors for the development of atrial fibrillation (AF) in patients with chronic heart failure (CHF) with preserved and reduced ejection fraction by echocardiography (EchoCG) according to an extended protocol with determination of diastolic function and left atrial global strain. MATERIAL AND METHODS Data of 168 patients with stage I-III CHF without a history of AF were analyzed. All patients underwent echocardiography according to an extended protocol with the determination of diastolic dysfunction (DD), left atrial ejection fraction (LA EF), and left atrial global strain (LA GS). Tissue Doppler imaging (TDI) was used to evaluate the early (E) and late (A) LV filling velocity and the early (E') and late (A') diastolic mitral annular velocity. In all patients, Holter ECG monitoring (HM ECG) of heart rhythm was performed for 3 days, and ECG monitoring with telemedicine technologies was performed for 7 days, 3 times a day for 3 minutes. The follow-up period was 3 months or until an AF episode. RESULTS During the study, paroxysmal AF (pAF) was detected in 41 (24.4%) patients using various methods of heart rhythm monitoring. Complaints of palpitations were noted for 10 (24.4%) patients during pAF, which was recorded using a CardioQVARK® device, HM ECG or a 12-lead ECG. In 5 (12.2%) patients, daily ECG monitoring revealed pAF without associated complaints. HM ECG detected 8, 2, 4 (19.5%, 4.8%, and 9.7%) cases during 24, 48 and 72 hours, respectively; a single-channel CardioQVARK® detected 30 (73.2%) cases when used 3 times a day for 7 days. These results showed that AF frequently develops in CHF without accompanying symptoms. The method for detecting pAF with CardioQVARK® showed good results: it was twice more effective than HM ECG and three times more effective than 12-lead ECG. Also, according to ultrasound data, significant changes in the following parameters were noted in patients with AF: LA EF <36% (OR 1.04, 95% CI: 1.02-1.08), p=0.003; LA GS <9.9% (OR 1.16, 95% CI: 1.02-1.38), p<0.001; TDI E med <5.7 cm/s (OR 0.97, 95% CI: 0.94-1.00), p=0.026. Grade 2 DD did not show statistically significant results (OR 1.1, 95% CI: 0.7-1.5, p=0.54). However, it was detected more frequently in patients with AF, in 34% of cases, compared to 29% of cases in patients without AF, which requires further study on a larger patient sample. CONCLUSION Patients with CHF have a high risk of developing pAF (24.4%). 75% of patients with AF do not feel the development of paroxysm. All CHF patients should undergo EchoCG with assessment of LA EF, TDI E med and LA GS to identify a group at risk for the development of AF. Heart rhythm remote monitoring with CardioQVARK® devices can be considered a reliable method for early detection of pAF and timely initiation of anticoagulant therapy in patients with CHF.
Collapse
Affiliation(s)
| | | | | | | | | | | | - D A Andreev
- Sechenov First Moscow State Medical University
| |
Collapse
|
68
|
Hiram R, Xiong F, Naud P, Xiao J, Sosnowski DK, Le Quilliec E, Saljic A, Abu-Taha IH, Kamler M, LeBlanc CA, Al-U’Datt DGF, Sirois MG, Hebert TE, Tanguay JF, Tardif JC, Dobrev D, Nattel S. An inflammation resolution-promoting intervention prevents atrial fibrillation caused by left ventricular dysfunction. Cardiovasc Res 2024; 120:345-359. [PMID: 38091977 PMCID: PMC10981525 DOI: 10.1093/cvr/cvad175] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 10/31/2023] [Accepted: 11/09/2023] [Indexed: 02/24/2024] Open
Abstract
AIMS Recent studies suggest that bioactive mediators called resolvins promote an active resolution of inflammation. Inflammatory signalling is involved in the development of the substrate for atrial fibrillation (AF). The aim of this study is to evaluate the effects of resolvin-D1 on atrial arrhythmogenic remodelling resulting from left ventricular (LV) dysfunction induced by myocardial infarction (MI) in rats. METHODS AND RESULTS MI was produced by left anterior descending coronary artery ligation. Intervention groups received daily intraperitoneal resolvin-D1, beginning before MI surgery (early-RvD1) or Day 7 post-MI (late-RvD1) and continued until Day 21 post-MI. AF vulnerability was evaluated by performing an electrophysiological study. Atrial conduction was analysed by using optical mapping. Fibrosis was quantified by Masson's trichrome staining and gene expression by quantitative polymerase chain reaction and RNA sequencing. Investigators were blinded to group identity. Early-RvD1 significantly reduced MI size (17 ± 6%, vs. 39 ± 6% in vehicle-MI) and preserved LV ejection fraction; these were unaffected by late-RvD1. Transoesophageal pacing induced atrial tachyarrhythmia in 2/18 (11%) sham-operated rats, vs. 18/18 (100%) MI-only rats, in 5/18 (28%, P < 0.001 vs. MI) early-RvD1 MI rats, and in 7/12 (58%, P < 0.01) late-RvD1 MI rats. Atrial conduction velocity significantly decreased post-MI, an effect suppressed by RvD1 treatment. Both early-RvD1 and late-RvD1 limited MI-induced atrial fibrosis and prevented MI-induced increases in the atrial expression of inflammation-related and fibrosis-related biomarkers and pathways. CONCLUSIONS RvD1 suppressed MI-related atrial arrhythmogenic remodelling. Early-RvD1 had MI sparing and atrial remodelling suppressant effects, whereas late-RvD1 attenuated atrial remodelling and AF promotion without ventricular protection, revealing atrial-protective actions unrelated to ventricular function changes. These results point to inflammation resolution-promoting compounds as novel cardio-protective interventions with a particular interest in attenuating AF substrate development.
Collapse
Affiliation(s)
- Roddy Hiram
- Department of Medicine, Montreal Heart Institute (MHI), Université de Montréal, 5000 Belanger Street, Montreal, Quebec, CanadaH1T 1C8
| | - Feng Xiong
- Department of Medicine, Montreal Heart Institute (MHI), Université de Montréal, 5000 Belanger Street, Montreal, Quebec, CanadaH1T 1C8
- Department of Pharmacology and Therapeutics, McGill University, 3655 Prom. Sir William Osler, Montreal, Canada H3G 1Y6
| | - Patrice Naud
- Department of Medicine, Montreal Heart Institute (MHI), Université de Montréal, 5000 Belanger Street, Montreal, Quebec, CanadaH1T 1C8
| | - Jiening Xiao
- Department of Medicine, Montreal Heart Institute (MHI), Université de Montréal, 5000 Belanger Street, Montreal, Quebec, CanadaH1T 1C8
| | - Deanna K Sosnowski
- Department of Medicine, Montreal Heart Institute (MHI), Université de Montréal, 5000 Belanger Street, Montreal, Quebec, CanadaH1T 1C8
- Department of Pharmacology and Therapeutics, McGill University, 3655 Prom. Sir William Osler, Montreal, Canada H3G 1Y6
| | - Ewen Le Quilliec
- Department of Medicine, Montreal Heart Institute (MHI), Université de Montréal, 5000 Belanger Street, Montreal, Quebec, CanadaH1T 1C8
| | - Arnela Saljic
- Institute of Pharmacology, West German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen, Hufelandstr 55, Essen, Germany D-45122
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Norregade 10 P.O. Box 2177, Copenhagen, Denmark
| | - Issam H Abu-Taha
- Institute of Pharmacology, West German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen, Hufelandstr 55, Essen, Germany D-45122
| | - Markus Kamler
- Department of Thoracic and Cardiovascular Surgery, West German Heart and Vascular Center Essen, University Hospital Essen, Hufelanstr 55, Essen, Germany 45122
| | - Charles-Alexandre LeBlanc
- Department of Medicine, Montreal Heart Institute (MHI), Université de Montréal, 5000 Belanger Street, Montreal, Quebec, CanadaH1T 1C8
| | - Doa’a G F Al-U’Datt
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, P.O. Box 3030 Irbid, Jordan 22110
| | - Martin G Sirois
- Department of Medicine, Montreal Heart Institute (MHI), Université de Montréal, 5000 Belanger Street, Montreal, Quebec, CanadaH1T 1C8
| | - Terence E Hebert
- Department of Pharmacology and Therapeutics, McGill University, 3655 Prom. Sir William Osler, Montreal, Canada H3G 1Y6
| | - Jean-François Tanguay
- Department of Medicine, Montreal Heart Institute (MHI), Université de Montréal, 5000 Belanger Street, Montreal, Quebec, CanadaH1T 1C8
| | - Jean-Claude Tardif
- Department of Medicine, Montreal Heart Institute (MHI), Université de Montréal, 5000 Belanger Street, Montreal, Quebec, CanadaH1T 1C8
| | - Dobromir Dobrev
- Department of Medicine, Montreal Heart Institute (MHI), Université de Montréal, 5000 Belanger Street, Montreal, Quebec, CanadaH1T 1C8
- Department of Pharmacology and Therapeutics, McGill University, 3655 Prom. Sir William Osler, Montreal, Canada H3G 1Y6
- Institute of Pharmacology, West German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen, Hufelandstr 55, Essen, Germany D-45122
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, P.O. Box 3030 Irbid, Jordan 22110
| | - Stanley Nattel
- Department of Medicine, Montreal Heart Institute (MHI), Université de Montréal, 5000 Belanger Street, Montreal, Quebec, CanadaH1T 1C8
- Department of Pharmacology and Therapeutics, McGill University, 3655 Prom. Sir William Osler, Montreal, Canada H3G 1Y6
- Institute of Pharmacology, West German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen, Hufelandstr 55, Essen, Germany D-45122
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Norregade 10 P.O. Box 2177, Copenhagen, Denmark
- IHU Liryc and Fondation Bordeaux Université, 166 cours de l'Argonne, Bordeaux, France 33000
| |
Collapse
|
69
|
Tzeis S, Gerstenfeld EP, Kalman J, Saad EB, Sepehri Shamloo A, Andrade JG, Barbhaiya CR, Baykaner T, Boveda S, Calkins H, Chan NY, Chen M, Chen SA, Dagres N, Damiano RJ, De Potter T, Deisenhofer I, Derval N, Di Biase L, Duytschaever M, Dyrda K, Hindricks G, Hocini M, Kim YH, la Meir M, Merino JL, Michaud GF, Natale A, Nault I, Nava S, Nitta T, O’Neill M, Pak HN, Piccini JP, Pürerfellner H, Reichlin T, Saenz LC, Sanders P, Schilling R, Schmidt B, Supple GE, Thomas KL, Tondo C, Verma A, Wan EY. 2024 European Heart Rhythm Association/Heart Rhythm Society/Asia Pacific Heart Rhythm Society/Latin American Heart Rhythm Society expert consensus statement on catheter and surgical ablation of atrial fibrillation. Europace 2024; 26:euae043. [PMID: 38587017 PMCID: PMC11000153 DOI: 10.1093/europace/euae043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 01/16/2024] [Indexed: 04/09/2024] Open
Abstract
In the last three decades, ablation of atrial fibrillation (AF) has become an evidence-based safe and efficacious treatment for managing the most common cardiac arrhythmia. In 2007, the first joint expert consensus document was issued, guiding healthcare professionals involved in catheter or surgical AF ablation. Mounting research evidence and technological advances have resulted in a rapidly changing landscape in the field of catheter and surgical AF ablation, thus stressing the need for regularly updated versions of this partnership which were issued in 2012 and 2017. Seven years after the last consensus, an updated document was considered necessary to define a contemporary framework for selection and management of patients considered for or undergoing catheter or surgical AF ablation. This consensus is a joint effort from collaborating cardiac electrophysiology societies, namely the European Heart Rhythm Association, the Heart Rhythm Society, the Asia Pacific Heart Rhythm Society, and the Latin American Heart Rhythm Society .
Collapse
Affiliation(s)
- Stylianos Tzeis
- Department of Cardiology, Mitera Hospital, 6, Erythrou Stavrou Str., Marousi, Athens, PC 151 23, Greece
| | - Edward P Gerstenfeld
- Section of Cardiac Electrophysiology, University of California, San Francisco, CA, USA
| | - Jonathan Kalman
- Department of Cardiology, Royal Melbourne Hospital, Melbourne, Australia
- Department of Medicine, University of Melbourne and Baker Research Institute, Melbourne, Australia
| | - Eduardo B Saad
- Electrophysiology and Pacing, Hospital Samaritano Botafogo, Rio de Janeiro, Brazil
- Cardiac Arrhythmia Service, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | - Jason G Andrade
- Department of Medicine, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | | | - Tina Baykaner
- Division of Cardiology and Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Serge Boveda
- Heart Rhythm Management Department, Clinique Pasteur, Toulouse, France
- Universiteit Brussel (VUB), Brussels, Belgium
| | - Hugh Calkins
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Ngai-Yin Chan
- Department of Medicine and Geriatrics, Princess Margaret Hospital, Hong Kong Special Administrative Region, China
| | - Minglong Chen
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shih-Ann Chen
- Heart Rhythm Center, Taipei Veterans General Hospital, Taipei, and Cardiovascular Center, Taichung Veterans General Hospital, Taichung, Taiwan
| | | | - Ralph J Damiano
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, Barnes-Jewish Hospital, St. Louis, MO, USA
| | | | - Isabel Deisenhofer
- Department of Electrophysiology, German Heart Center Munich, Technical University of Munich (TUM) School of Medicine and Health, Munich, Germany
| | - Nicolas Derval
- IHU LIRYC, Electrophysiology and Heart Modeling Institute, Cardiac Electrophysiology and Stimulation Department, Fondation Bordeaux Université and Bordeaux University Hospital (CHU), Pessac-Bordeaux, France
| | - Luigi Di Biase
- Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - Katia Dyrda
- Department of Medicine, Montreal Heart Institute, Université de Montréal, Montreal, Canada
| | | | - Meleze Hocini
- IHU LIRYC, Electrophysiology and Heart Modeling Institute, Cardiac Electrophysiology and Stimulation Department, Fondation Bordeaux Université and Bordeaux University Hospital (CHU), Pessac-Bordeaux, France
| | - Young-Hoon Kim
- Division of Cardiology, Korea University College of Medicine and Korea University Medical Center, Seoul, Republic of Korea
| | - Mark la Meir
- Cardiac Surgery Department, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Jose Luis Merino
- La Paz University Hospital, Idipaz, Universidad Autonoma, Madrid, Spain
- Hospital Viamed Santa Elena, Madrid, Spain
| | | | - Andrea Natale
- Texas Cardiac Arrhythmia Institute, St. David’s Medical Center, Austin, TX, USA
- Case Western Reserve University, Cleveland, OH, USA
- Interventional Electrophysiology, Scripps Clinic, San Diego, CA, USA
- Department of Biomedicine and Prevention, Division of Cardiology, University of Tor Vergata, Rome, Italy
| | - Isabelle Nault
- Institut Universitaire de Cardiologie et de Pneumologie de Quebec (IUCPQ), Quebec, Canada
| | - Santiago Nava
- Departamento de Electrocardiología, Instituto Nacional de Cardiología ‘Ignacio Chávez’, Ciudad de México, México
| | - Takashi Nitta
- Department of Cardiovascular Surgery, Nippon Medical School, Tokyo, Japan
| | - Mark O’Neill
- Cardiovascular Directorate, St. Thomas’ Hospital and King’s College, London, UK
| | - Hui-Nam Pak
- Division of Cardiology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | | | | | - Tobias Reichlin
- Department of Cardiology, Inselspital Bern, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Luis Carlos Saenz
- International Arrhythmia Center, Cardioinfantil Foundation, Bogota, Colombia
| | - Prashanthan Sanders
- Centre for Heart Rhythm Disorders, University of Adelaide and Royal Adelaide Hospital, Adelaide, Australia
| | | | - Boris Schmidt
- Cardioangiologisches Centrum Bethanien, Medizinische Klinik III, Agaplesion Markuskrankenhaus, Frankfurt, Germany
| | - Gregory E Supple
- Cardiac Electrophysiology Section, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | - Claudio Tondo
- Department of Clinical Electrophysiology and Cardiac Pacing, Centro Cardiologico Monzino, IRCCS, Milan, Italy
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Atul Verma
- McGill University Health Centre, McGill University, Montreal, Canada
| | - Elaine Y Wan
- Department of Medicine, Division of Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| |
Collapse
|
70
|
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
|
71
|
Ye T, Yang J, Liu Z, Yu Y, Zhang C, Guo Y, Yu F, Zhou Y, Song Z, Shi J, Wang L, Yang B, Wang X. Inhibition of the P2X7 receptor prevents atrial proarrhythmic remodeling in experimental post-operative atrial fibrillation. Int Immunopharmacol 2024; 129:111536. [PMID: 38320354 DOI: 10.1016/j.intimp.2024.111536] [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: 10/18/2023] [Revised: 01/10/2024] [Accepted: 01/10/2024] [Indexed: 02/08/2024]
Abstract
BACKGROUND Post-operative atrial fibrillation (POAF) is a common complication in patients undergoing cardiac surgery. The purinergic receptor P2X7 (P2X7R) is involved in some cardiovascular diseases, whereas its effects on atrial fibrillation (AF) are unclear. OBJECTIVE This study was to assess the effect of P2X7R on atrial arrhythmogenic remodeling in the rat model of sterile pericarditis (SP). METHODS Male Sprague-Dawley (SD) rats were used to induce the SP model. Electrocardiogram, atrial electrophysiological protocol, histology, mRNA sequencing, real-time quantitative PCR, western blot, and Elisa assay were performed. RESULTS SP significantly up-regulated P2X7R expression; increased AF susceptibility; reduced the protein expression of ion channels including Nav1.5, Cav1.2, Kv4.2, Kv4.3, and Kv1.5; caused atrial fibrosis; increased norepinephrine (NE) level in plasma; promoted the production of inflammatory cytokines such as TNF-α, IL-1β, and IL-6; increased the accumulation of immune cells (CD68- and MPO- positive cells); and activated NLRP3 inflammasome signaling pathway. P2X7R antagonist Brilliant Blue G (BBG) mitigated SP-induced alterations. The mRNA sequencing demonstrated that BBG prevented POAF mainly by regulating the immune system. In addition, another selective P2X7R antagonist A740003, and IL-1R antagonist anakinra also reduced AF inducibility in the SP model. CONCLUSIONS P2X7R inhibition prevents SP-induced atrial proarrhythmic remodeling, which is closely associated with the improvement of inflammatory changes, ion channel expression, atrial fibrosis, and sympathetic activation. The findings point to P2X7R inhibition as a promising target for AF (particularly POAF) and perhaps other conditions.
Collapse
Affiliation(s)
- Tianxin Ye
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Jinxiu Yang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Zhangchi Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Yi Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Cui Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Yan Guo
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Fangcong Yu
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Yunping Zhou
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Zhuonan Song
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Jiaran Shi
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Longbo Wang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Bo Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, 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, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
72
|
Hegemann N, Barth L, Döring Y, Voigt N, Grune J. Implications for neutrophils in cardiac arrhythmias. Am J Physiol Heart Circ Physiol 2024; 326:H441-H458. [PMID: 38099844 PMCID: PMC11219058 DOI: 10.1152/ajpheart.00590.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 12/08/2023] [Accepted: 12/13/2023] [Indexed: 02/03/2024]
Abstract
Cardiac arrhythmias commonly occur as a result of aberrant electrical impulse formation or conduction in the myocardium. Frequently discussed triggers include underlying heart diseases such as myocardial ischemia, electrolyte imbalances, or genetic anomalies of ion channels involved in the tightly regulated cardiac action potential. Recently, the role of innate immune cells in the onset of arrhythmic events has been highlighted in numerous studies, correlating leukocyte expansion in the myocardium to increased arrhythmic burden. Here, we aim to call attention to the role of neutrophils in the pathogenesis of cardiac arrhythmias and their expansion during myocardial ischemia and infectious disease manifestation. In addition, we will elucidate molecular mechanisms associated with neutrophil activation and discuss their involvement as direct mediators of arrhythmogenicity.
Collapse
Affiliation(s)
- Niklas Hegemann
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Lukas Barth
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Yannic Döring
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg August University Göttingen, Göttingen, Germany
- German Centre for Cardiovascular Research (DZHK), Göttingen, Germany
| | - Niels Voigt
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg August University Göttingen, Göttingen, Germany
- German Centre for Cardiovascular Research (DZHK), Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Jana Grune
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| |
Collapse
|
73
|
Zhao Y, Ning Y, Lei L, Liu Q, Li M, Lei X, Chen W, Hu Y, Xie T, Luan J, Yang H, Luo G. The relationship between atrial cardiopathy biomarkers and prognosis of patients with acute ischemic stroke after endovascular treatment. Neurotherapeutics 2024; 21:e00327. [PMID: 38320384 PMCID: PMC10963924 DOI: 10.1016/j.neurot.2024.e00327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 01/21/2024] [Indexed: 02/08/2024] Open
Abstract
Thromboembolism is a possible consequence of underlying atrial cardiopathy, which can occur even before the onset of atrial fibrillation. Our objective was to examine the association between biomarkers of atrial cardiopathy and outcomes of acute ischemic stroke (AIS) following endovascular treatment (EVT). We conducted a retrospective study that collected data from patients with AIS who underwent EVT and compared the outcomes between those with and without atrial cardiopathy. Neurological function was assessed using the modified Rankin Scale (mRS), with an mRS score >2 indicating poor function at day 90. Additionally, we evaluated secondary consequences, including symptomatic intracerebral hemorrhage (sICH), early neurological deterioration (END), and malignant cerebral edema (MCE). Our study included 87 patients (77.6 % male; mean age 60.93 ± 12.47 years). Among these patients, 29 (33.3 %) had atrial cardiopathy, while the remaining 58 (66.7 %) did not. In the atrial cardiopathy group, 12 patients (41.4 %) had poor functional outcomes (mRS>2), compared to 19 (32.8 %) in the non-atrial cardiopathy group. We observed sICH in 22 (25.3 %) patients, END in 14 (16.1 %) patients, MCE in 11 (12.6 %) patients, and two (2.3 %) patients who died in the hospital. We found that patients with PTFV1>5000 μV/ms (OR: 8.39, 95 % CI: 1.43-105.95, P = 0.02) and NT-proBNP>250 pg/mL (OR: 5.09, 95 % CI: 1.20-27.63, P = 0.03) had significantly higher risk of END. After adjusting for covariates in the Firth logistic regression, we further found that atrial cardiopathy was significantly associated with END, as revealed by both univariate (OR: 6.31, 95 % CI: 1.42-59.87, P = 0.01) and multivariable firth regression models (Modle 1, OR: 7.10, 95 % CI: 1.57-67.38, P < 0.01; Modle 2, OR: 7.82, 95 % CI: 1.69, 76.36, P < 0.01; Modle 3, OR: 8.59, 95 % CI: 1.72-91.70, P < 0.01). Moreover, we observed that atrial cardiopathy was associated with an increased risk of END in AIS patients with large artery atherosclerosis (LAA) receiving EVT. Therefore, clinicians should consider atrial cardiopathy as a possible underlying cause of AIS in their patients. Further investigation is warranted to elucidate the relationship between atrial cardiopathy and AIS's occurrence, progression, and prognosis.
Collapse
Affiliation(s)
- Yixin Zhao
- Stroke Centre and Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an, 710061, China; Department of Neurology, Huashan Hospital, Fudan University, Shanghai, 200235, China
| | - Yuye Ning
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Lei Lei
- Stroke Centre and Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an, 710061, China
| | - Qin Liu
- Stroke Centre and Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an, 710061, China
| | - Mengmeng Li
- Stroke Centre and Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an, 710061, China
| | - Xiangyu Lei
- Stroke Centre and Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an, 710061, China
| | - Wanying Chen
- Stroke Centre and Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an, 710061, China
| | - Yiting Hu
- Pavlov First Saint Petersburg State Medical University, L'va Tolstogo Str. 6-8, Saint Petersburg, 197022, Russia
| | - Ting Xie
- Stroke Centre and Department of Neurology, Hancheng People's Hospital of Shaanxi Province, Ziyun Dajie, and Huanghe Dajie, Hancheng, 715400, China
| | - Jiaxin Luan
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an, 710061, China
| | - Haoyu Yang
- Pharmacy Department, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an, 710061, China
| | - Guogang Luo
- Stroke Centre and Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an, 710061, China.
| |
Collapse
|
74
|
Yan S, Xu W, Fang N, Li L, Yang N, Zhao X, Hao H, Zhang Y, Liang Q, Wang Z, Duan Y, Zhang S, Gong Y, Li Y. Ibrutinib-induced pulmonary angiotensin-converting enzyme activation promotes atrial fibrillation in rats. iScience 2024; 27:108926. [PMID: 38357670 PMCID: PMC10864204 DOI: 10.1016/j.isci.2024.108926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/25/2023] [Accepted: 01/12/2024] [Indexed: 02/16/2024] Open
Abstract
The molecular mechanism of ibrutinib-induced atrial fibrillation (AF) remains unclear. We here demonstrate that treating rats with ibrutinib for 4 weeks resulted in the development of inducible AF, left atrial enlargement, atrial fibrosis, and downregulation of connexin expression, which were associated with C-terminal Src kinase (CSK) inhibition and Src activation. Ibrutinib upregulated angiotensin-converting enzyme (ACE) protein expression in human pulmonary microvascular endothelial cells (HPMECs) by inhibiting the PI3K-AKT pathway, subsequently increasing circulating angiotensin II (Ang II) levels. However, the expression of ACE and Ang II in the left atria was not affected. Importantly, we observed that perindopril significantly mitigated ibrutinib-induced left atrial remodeling and AF promotion by inhibiting the activation of the ACE and its downstream CSK-Src signaling pathway. These findings indicate that the Ibrutinib-induced activation of the ACE contributes to AF development and could serve as a novel target for potential prevention strategies.
Collapse
Affiliation(s)
- Sen Yan
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Wei Xu
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Ning Fang
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Luyifei Li
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Ning Yang
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Xinbo Zhao
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Hongting Hao
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Yun Zhang
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Qian Liang
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Zhiqi Wang
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Yu Duan
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Song Zhang
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Yongtai Gong
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Yue Li
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Heilongjiang 150001, China
- Key Laboratory of Hepatosplenic Surgery, Harbin Medical University, Ministry of Education, Harbin 150001, China
- Key Laboratory of Cardiac Diseases and Heart Failure, Harbin Medical University, Harbin 150001, China
- Heilongjiang Key Laboratory for Metabolic Disorder & Cancer Related Cardiovascular Diseases, Harbin 150081, China
- Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Harbin, China
| |
Collapse
|
75
|
Portero V, Deng S, Boink GJJ, Zhang GQ, de Vries A, Pijnappels DA. Optoelectronic control of cardiac rhythm: Toward shock-free ambulatory cardioversion of atrial fibrillation. J Intern Med 2024; 295:126-145. [PMID: 37964404 DOI: 10.1111/joim.13744] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
Abstract
Atrial fibrillation (AF) is the most prevalent cardiac arrhythmia, progressive in nature, and known to have a negative impact on mortality, morbidity, and quality of life. Patients requiring acute termination of AF to restore sinus rhythm are subjected to electrical cardioversion, which requires sedation and therefore hospitalization due to pain resulting from the electrical shocks. However, considering the progressive nature of AF and its detrimental effects, there is a clear need for acute out-of-hospital (i.e., ambulatory) cardioversion of AF. In the search for shock-free cardioversion methods to realize such ambulatory therapy, a method referred to as optogenetics has been put forward. Optogenetics enables optical control over the electrical activity of cardiomyocytes by targeted expression of light-activated ion channels or pumps and may therefore serve as a means for cardioversion. First proof-of-principle for such light-induced cardioversion came from in vitro studies, proving optogenetic AF termination to be very effective. Later, these results were confirmed in various rodent models of AF using different transgenes, illumination methods, and protocols, whereas computational studies in the human heart provided additional translational insight. Based on these results and fueled by recent advances in molecular biology, gene therapy, and optoelectronic engineering, a basis is now being formed to explore clinical translations of optoelectronic control of cardiac rhythm. In this review, we discuss the current literature regarding optogenetic cardioversion of AF to restore normal rhythm in a shock-free manner. Moreover, key translational steps will be discussed, both from a biological and technological point of view, to outline a path toward realizing acute shock-free ambulatory termination of AF.
Collapse
Affiliation(s)
- Vincent Portero
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Shanliang Deng
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
- Department of Microelectronics, Delft University of Technology, Delft, The Netherlands
| | - Gerard J J Boink
- Department of Medical Biology, Department of Cardiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Guo Qi Zhang
- Department of Microelectronics, Delft University of Technology, Delft, The Netherlands
| | - Antoine de Vries
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Daniël A Pijnappels
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| |
Collapse
|
76
|
Vinciguerra M, Dobrev D, Nattel S. Atrial fibrillation: pathophysiology, genetic and epigenetic mechanisms. THE LANCET REGIONAL HEALTH. EUROPE 2024; 37:100785. [PMID: 38362554 PMCID: PMC10866930 DOI: 10.1016/j.lanepe.2023.100785] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 09/08/2023] [Accepted: 11/02/2023] [Indexed: 02/17/2024]
Abstract
Atrial fibrillation (AF) is the most common supraventricular arrhythmia affecting up to 1% of the general population. Its prevalence dramatically increases with age and could reach up to ∼10% in the elderly. The management of AF is a complex issue that is object of extensive ongoing basic and clinical research, it depends on its genetic and epigenetic causes, and it varies considerably geographically and also according to the ethnicity. Mechanistically, over the last decade, Genome Wide Association Studies have uncovered over 100 genetic loci associated with AF, and have shown that European ancestry is associated with elevated risk of AF. These AF-associated loci revolve around different types of disturbances, including inflammation, electrical abnormalities, and structural remodeling. Moreover, the discovery of epigenetic regulatory mechanisms, involving non-coding RNAs, DNA methylation and histone modification, has allowed unravelling what modifications reshape the processes leading to arrhythmias. Our review provides a current state of the field regarding the identification and functional characterization of AF-related genetic and epigenetic regulatory networks, including ethnic differences. We discuss clear and emerging connections between genetic regulation and pathophysiological mechanisms of AF.
Collapse
Affiliation(s)
- Manlio Vinciguerra
- Department of Translational Stem Cell Biology, Research Institute, Medical University of Varna, Varna, Bulgaria
- Liverpool Centre for Cardiovascular Science, Faculty of Health, Liverpool John Moores University, Liverpool, United Kingdom
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Duisburg, Germany
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Canada
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Stanley Nattel
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Duisburg, Germany
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Canada
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Faculty of Health, Medicine, and Life Sciences, Maastricht University, Maastricht, Netherlands
- IHU LIRYC and Fondation Bordeaux Université, Bordeaux, France
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
77
|
Chen IS, Yasuda J, Notomi T, Nakamura TY. Licorice metabolite 18β-glycyrrhetinic acid activates G protein-gated inwardly rectifying K + channels. Br J Pharmacol 2024; 181:447-463. [PMID: 37642133 DOI: 10.1111/bph.16228] [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: 02/24/2023] [Revised: 08/16/2023] [Accepted: 08/22/2023] [Indexed: 08/31/2023] Open
Abstract
BACKGROUND AND PURPOSE Licorice (liquorice) is a common food additive and is used in Chinese medicine. Excess licorice intake can induce atrial fibrillation. Patients with atrial fibrillation possess constitutively activated G protein-gated inwardly rectifying K+ (GIRK) channels. Whether licorice affects GIRK channel activity is unknown. We aimed to clarify the effects of licorice ingredients on GIRK current and the mechanism of action. EXPERIMENTAL APPROACH A major component of licorice, glycyrrhizic acid (GA), and its metabolite, 18β-glycyrrhetinic acid (18β-GA), were tested. We performed electrophysiological recordings in Xenopus oocytes to examine the effects of GA and 18β-GA on various GIRK subunits (Kir 3.1-Kir 3.4), mutagenesis analyses to identify the crucial residues for drug action and motion analysis in cultured rat atrial myocytes to clarify effects of 18β-GA on atrial functions. KEY RESULTS GA inhibited Kir 3.1-containing channels, while 18β-GA activated all Kir 3.x subunits. A pore helix residue Phe137 in Kir 3.1 was critical for GA-mediated inhibition, and the corresponding Ser148 in Kir 3.2 was critical for 18β-GA-mediated activation. 18β-GA activated GIRK channel in a Gβγ -independent manner, whereas phosphatidylinositol 4,5-bisphosphate (PIP2 ) was essential for activation. Glu236 located at the cytoplasmic pore of Kir 3.2 appeared to be important to interactions with 18β-GA. In rat atrial myocytes, 18β-GA suppressed spontaneous beating via activation of GIRK channels. CONCLUSION AND IMPLICATIONS GA acts as a novel GIRK inhibitor, and 18β-GA acts as a novel GIRK activator. 18β-GA alters atrial function via activation of GIRK channels. This study elucidates the pharmacological activity of licorice ingredients and provides information for drug design.
Collapse
Affiliation(s)
- I-Shan Chen
- Department of Pharmacology, Faculty of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Jumpei Yasuda
- Department of Pharmacology, Faculty of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Takuya Notomi
- Department of Pharmacology, Faculty of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Tomoe Y Nakamura
- Department of Pharmacology, Faculty of Medicine, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
78
|
Van Wagoner DR. Collagen type V, interstitial fibrosis and the substrate for atrial fibrillation. IJC HEART & VASCULATURE 2024; 50:101356. [PMID: 38419609 PMCID: PMC10899731 DOI: 10.1016/j.ijcha.2024.101356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 03/02/2024]
Affiliation(s)
- David R. Van Wagoner
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, M/S ND50, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| |
Collapse
|
79
|
Tamargo J, Villacastín J, Caballero R, Delpón E. Drug-induced atrial fibrillation. A narrative review of a forgotten adverse effect. Pharmacol Res 2024; 200:107077. [PMID: 38244650 DOI: 10.1016/j.phrs.2024.107077] [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: 10/26/2023] [Revised: 12/22/2023] [Accepted: 01/12/2024] [Indexed: 01/22/2024]
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia and is associated with an increased morbidity and mortality. There is clinical evidence that an increasing number of cardiovascular and non-cardiovascular drugs, mainly anticancer drugs, can induce AF either in patients with or without pre-existing cardiac disorders, but drug-induced AF (DIAF) has not received the attention that it might deserve. In many cases DIAF is asymptomatic and paroxysmal and patients recover sinus rhythm spontaneously, but sometimes, DIAF persists, and it is necessary to perform a cardioversion. Furthermore, DIAF is not mentioned in clinical guidelines on the treatment of AF. The risk of DIAF increases in elderly and in patients treated with polypharmacy and with risk factors and comorbidities that commonly coexist with AF. This is the case of cancer patients. Under these circumstances ascribing causality of DIAF to a given drug often represents a clinical challenge. We review the incidence, the pathophysiological mechanisms, risk factors, clinical relevance, and treatment of DIAF. Because of the limited information presently available, further research is needed to obtain a deeper insight into DIAF. Meanwhile, it is important that clinicians are aware of the problem that DIAF represents, recognize which drugs may cause DIAF, and consider the possibility that a drug may be responsible for a new-onset AF episode.
Collapse
Affiliation(s)
- Juan Tamargo
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón, CIBERCV, 28040 Madrid, Spain
| | - Julián Villacastín
- Hospital Clínico San Carlos, CardioRed1, Universidad Complutense de Madrid, CIBERCV, 28040 Madrid, Spain
| | - Ricardo Caballero
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón, CIBERCV, 28040 Madrid, Spain.
| | - Eva Delpón
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón, CIBERCV, 28040 Madrid, Spain
| |
Collapse
|
80
|
Zhang X, Yin T, Wang Y, Du J, Dou J, Zhang X. Effects of scutellarin on the mechanism of cardiovascular diseases: a review. Front Pharmacol 2024; 14:1329969. [PMID: 38259289 PMCID: PMC10800556 DOI: 10.3389/fphar.2023.1329969] [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] [Received: 10/30/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
Cardiovascular diseases represent a significant worldwide problem, jeopardizing individuals' physical and mental wellbeing as well as their quality of life as a result of their widespread incidence and fatality. With the aging society, the occurrence of Cardiovascular diseases is progressively rising each year. However, although drugs developed for treating Cardiovascular diseases have clear targets and proven efficacy, they still carry certain toxic and side effect risks. Therefore, finding safe, effective, and practical treatment options is crucial. Scutellarin is the primary constituent of Erigeron breviscapus (Vant.) Hand-Mazz. This article aims to establish a theoretical foundation for the creation and use of secure, productive, and logical medications for Scutellarin in curing heart-related illnesses. Additionally, the examination and analysis of the signal pathway and its associated mechanisms with regard to the employment of SCU in treating heart diseases will impart innovative resolving concepts for the treatment and prevention of Cardiovascular diseases.
Collapse
Affiliation(s)
- Xinyu Zhang
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Tong Yin
- First Clinical Medical School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yincang Wang
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jiazhe Du
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jinjin Dou
- Department of Cardiovascular, The First Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xiwu Zhang
- Experimental Training Centre, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
81
|
Rachieru C, Lighezan DF, Petrescu L, Târtea G, Goanță EV, Buzas R, Cirin L, Cozma D. The Significance of Inflammation in Atrial Fibrillation. CURRENT HEALTH SCIENCES JOURNAL 2024; 50:59-66. [PMID: 38846471 PMCID: PMC11151943 DOI: 10.12865/chsj.50.01.08] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 03/27/2024] [Indexed: 06/09/2024]
Abstract
AIM The aim of the study was to assess the inflammatory status in individuals diagnosed with atrial fibrillation (Afi) and establish an association between this status and the clinicopathological features. MATERIAL AND METHODS Our study was conducted retrospectively and initially involved 278 patients. However, after excluding 27 patients, we ultimately ended up with 167 patients who had an inflammatory status and 84 patients who did not have an inflammatory status. These patients were then analyzed. RESULTS Patients who had inflammation showed higher values for the CHA2DS2-VASc and HAS-BLED scores (P= 0.0132 for CHA2DS2-VASc and P= 0.0024 for HAS-BLED). Also, it was observed that patients with associated inflammation exhibited an increase in both the volume and the area of the left atrium. Patients with hypertension had a higher prevalence of inflammation, with heart failure and with ischemic heart disease. It is worth noting that patients with atrial fibrillation and increased inflammatory status exhibited higher rates of stroke (22.75% vs 10.71% in patients without inflammation, odds ratio = 2.455, 95% confidence interval 1.161 to 5.425, p = 0.0253). CONCLUSIONS Our research has demonstrated that patients diagnosed with atrial fibrillation and exhibiting a heightened inflammatory status also present association with other comorbidities, including hypertension, heart failure, ischemic heart disease, and stroke.
Collapse
Affiliation(s)
- Ciprian Rachieru
- Department of Internal Medicine I, Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
- Department of Internal Medicine, Municipal Emergency Hospital, Timisoara, Romania
- Center for Advanced Research in Cardiovascular Pathology and Hemostaseology, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Daniel-Florin Lighezan
- Department of Internal Medicine I, Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
- Department of Internal Medicine, Municipal Emergency Hospital, Timisoara, Romania
- Center for Advanced Research in Cardiovascular Pathology and Hemostaseology, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Lucian Petrescu
- Department of Cardiology, Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
- Department of Cardiology, Institute of Cardiovascular Diseases, Timisoara, Romania
| | - Georgică Târtea
- Department of Physiology, University of Medicine and Pharmacy of Craiova, Romania
- Department of Cardiology, Emergency County Hospital of Craiova, Romania
| | - Emilia Violeta Goanță
- Department of Cardiology, Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
- Department of Cardiology, Emergency County Hospital of Craiova, Romania
| | - Roxana Buzas
- Department of Internal Medicine I, Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
- Department of Internal Medicine, Municipal Emergency Hospital, Timisoara, Romania
- Center for Advanced Research in Cardiovascular Pathology and Hemostaseology, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Liviu Cirin
- Department of Cardiology, Institute of Cardiovascular Diseases, Timisoara, Romania
| | - Dragoș Cozma
- Department of Cardiology, Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
- Department of Cardiology, Institute of Cardiovascular Diseases, Timisoara, Romania
| |
Collapse
|
82
|
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
|
83
|
Ördög B, De Coster T, Dekker SO, Bart CI, Zhang J, Boink GJJ, Bax WH, Deng S, den Ouden BL, de Vries AAF, Pijnappels DA. Opto-electronic feedback control of membrane potential for real-time control of action potentials. CELL REPORTS METHODS 2023; 3:100671. [PMID: 38086387 PMCID: PMC10753386 DOI: 10.1016/j.crmeth.2023.100671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 10/09/2023] [Accepted: 11/19/2023] [Indexed: 12/21/2023]
Abstract
To unlock new research possibilities by acquiring control of action potential (AP) morphologies in excitable cells, we developed an opto-electronic feedback loop-based system integrating cellular electrophysiology, real-time computing, and optogenetic approaches and applied it to monolayers of heart muscle cells. This allowed accurate restoration and preservation of cardiac AP morphologies in the presence of electrical perturbations of different origin in an unsupervised, self-regulatory manner, without any prior knowledge of the disturbance. Moreover, arbitrary AP waveforms could be enforced onto these cells. Collectively, these results set the stage for the refinement and application of opto-electronic control systems to enable in-depth investigation into the regulatory role of membrane potential in health and disease.
Collapse
Affiliation(s)
- Balázs Ördög
- Laboratory of Experimental Cardiology, Department of Cardiology, Heart Lung Center Leiden, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands.
| | - Tim De Coster
- Laboratory of Experimental Cardiology, Department of Cardiology, Heart Lung Center Leiden, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Sven O Dekker
- Laboratory of Experimental Cardiology, Department of Cardiology, Heart Lung Center Leiden, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Cindy I Bart
- Laboratory of Experimental Cardiology, Department of Cardiology, Heart Lung Center Leiden, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Juan Zhang
- Laboratory of Experimental Cardiology, Department of Cardiology, Heart Lung Center Leiden, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Gerard J J Boink
- Amsterdam Cardiovascular Sciences, Department of Cardiology, Amsterdam UMC, Location AMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands; Department of Medical Biology, Amsterdam UMC, Location AMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Wilhelmina H Bax
- Laboratory of Experimental Cardiology, Department of Cardiology, Heart Lung Center Leiden, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Shanliang Deng
- Laboratory of Experimental Cardiology, Department of Cardiology, Heart Lung Center Leiden, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands; Department of Microelectronics, Delft University of Technology, 2628 CD Delft, the Netherlands
| | - Bram L den Ouden
- Laboratory of Experimental Cardiology, Department of Cardiology, Heart Lung Center Leiden, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands; Department of Microelectronics, Delft University of Technology, 2628 CD Delft, the Netherlands
| | - Antoine A F de Vries
- Laboratory of Experimental Cardiology, Department of Cardiology, Heart Lung Center Leiden, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Daniël A Pijnappels
- Laboratory of Experimental Cardiology, Department of Cardiology, Heart Lung Center Leiden, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands.
| |
Collapse
|
84
|
Huang W, Sun H, Tang Y, Luo Y, Liu H. Platelet-to-Lymphocyte Ratio Improves the Predictive Ability of the Risk Score for Atrial Fibrillation Recurrence After Radiofrequency Ablation. J Inflamm Res 2023; 16:6023-6038. [PMID: 38107387 PMCID: PMC10723594 DOI: 10.2147/jir.s440722] [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: 10/12/2023] [Accepted: 12/03/2023] [Indexed: 12/19/2023] Open
Abstract
Purpose To investigate the effect and comprehensive predictive value of the platelet-to-lymphocyte ratio (PLR) for long-term recurrence in patients with atrial fibrillation (AF) post ablation. Patients and Methods We retrospectively analysed 638 consecutive AF patients who underwent ablation, including 302 (47.3%) with paroxysmal AF and 336 (52.7%) with nonparoxysmal AF. Patients were grouped into the recurrence and nonrecurrence groups. Results After a mean follow-up of 15.1±9.3 months, 175 patients (27.4%) with AF had long-term recurrence, including 114 patients (33.9%) with nonparoxysmal AF and 61 patients (20.2%) with paroxysmal AF. In the entire cohort and in patients with nonparoxysmal AF, but not in those with paroxysmal AF, the PLR was significantly higher in the recurrence group than in the nonrecurrence group (P<0.05). After adjusting for the APPLE score, the PLR as a continuous variable independently predicted AF recurrence (hazard ratio [HR], 1.003; 95% confidence interval [CI], 1.001-1.005; P<0.01). The addition of the PLR to the APPLE score improved its predictive ability for recurrence (the C-statistic value increased from 0.645 to 0.675, P=0.02; the net reclassification improvement was 0.221, 95% CI 0.049-0.394, P=0.01; and the integrated discrimination improvement was 0.029, 95% CI 0.013-0.045, P<0.01). For nonparoxysmal AF, the PLR was stratified into tertiles, the PLR independently increased the nonparoxysmal AF recurrence risk after adjusting for multiple confounding factors (HR, 1.393; 95% CI, 1.102-1.762; P<0.01), and the addition of the PLR to the left atrial diameter improved its predictive ability for arrhythmia recurrence (the C-statistic value increased from 0.601 to 0.667, P<0.01). Conclusion The PLR is an independent predictive factor of long-term AF recurrence post ablation after adjusting for the APPLE score and can improve the predictive ability and clinical usefulness of the APPLE score. However, the PLR is an effective predictor of recurrence in patients with nonparoxysmal AF rather than in paroxysmal AF.
Collapse
Affiliation(s)
- Wenchao Huang
- Department of Cardiology, The Third People’s Hospital of Chengdu, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, People’s Republic of China
| | - Huaxin Sun
- Department of Cardiology, The Third People’s Hospital of Chengdu, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, People’s Republic of China
| | - Yan Tang
- Department of Cardiology, The Third People’s Hospital of Chengdu, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, People’s Republic of China
| | - Yan Luo
- Department of Cardiology, The Third People’s Hospital of Chengdu, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, People’s Republic of China
| | - Hanxiong Liu
- Department of Cardiology, The Third People’s Hospital of Chengdu, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, People’s Republic of China
| |
Collapse
|
85
|
Li X, Wu X, Chen X, Peng S, Chen S, Zhou G, Wei Y, Lu X, Zhou C, Ye Y, Li J, Liu S, Xu J. Selective blockade of interleukin 6 trans-signaling depresses atrial fibrillation. Heart Rhythm 2023; 20:1759-1770. [PMID: 37633428 DOI: 10.1016/j.hrthm.2023.08.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 08/15/2023] [Accepted: 08/16/2023] [Indexed: 08/28/2023]
Abstract
BACKGROUND Atrial fibrillation (AF) has been accepted as an inflammatory atrial myopathy. Interleukin 6 (IL-6)-dependent inflammatory signaling pathways take context-dependent effects on cardiovascular diseases. IL-6 trans-signaling is predominantly pro-inflammatory. However, its effect on AF is unclear. OBJECTIVE The purpose of this study was to investigate the role of IL-6 trans-signaling in AF. METHODS Circulating levels of IL-6, soluble IL-6 receptor, and soluble glycoprotein 130 (sgp130) in patients with AF and controls were measured to estimate the activation of IL-6 trans-signaling. A mouse model of AF was established by transverse aortic constriction surgery. Sgp130Fc administration was used for the selective blockade of IL-6 trans-signaling. Studies were conducted to evaluate the effects and underlying mechanisms of sgp130Fc on AF inducibility and atrial conduction abnormalities and structural remodeling. RESULTS In patients, the elevation of IL-6 trans-signaling level was positively associated with AF occurrence. IL-6 trans-signaling activation was recapitulated in the mouse model of AF. In transverse aortic constriction-challenged mice, the selective blockade of IL-6 trans-signaling with sgp130Fc attenuated AF inducibility, which was attributable to the amelioration of slow conduction and conduction heterogeneity induced by atrial dilation, fibrosis, and reduction in connexin 40 and redistribution of connexin 43. Sgp130Fc administration also reduced immune cell infiltration and oxidative stress in the mouse atrium and abrogated IL-6 trans-signaling activation-mediated connexin dysregulation and reactive oxygen species production in atrial myocytes. CONCLUSION IL-6 trans-signaling activation contributes to AF development, and its selective blockade may promise a novel therapeutic strategy.
Collapse
Affiliation(s)
- Xintao Li
- Department of Cardiology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Department of Cardiology, First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu Province, China
| | - Xiaoyu Wu
- Department of Cardiology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoqiang Chen
- Department of Cardiology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shi Peng
- Department of Cardiology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Songwen Chen
- Department of Cardiology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Genqing Zhou
- Department of Cardiology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yong Wei
- Department of Cardiology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaofeng Lu
- Department of Cardiology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Changzuan Zhou
- Department of Cardiology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yutong Ye
- Department of Cardiology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jun Li
- Department of Cardiology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shaowen Liu
- Department of Cardiology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Juan Xu
- Department of Cardiology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
86
|
van den Berg NWE, Kawasaki M, Nariswari FA, Fabrizi B, Neefs J, van der Made I, Wesselink R, van Boven WJP, Driessen AHG, Jongejan A, de Groot JR. MicroRNAs in atrial fibrillation target genes in structural remodelling. Cell Tissue Res 2023; 394:497-514. [PMID: 37833432 DOI: 10.1007/s00441-023-03823-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 08/07/2023] [Indexed: 10/15/2023]
Abstract
We aim to elucidate how miRNAs regulate the mRNA signature of atrial fibrillation (AF), to gain mechanistic insight and identify candidate targets for future therapies. We present combined miRNA-mRNA sequencing using atrial tissues of patient without AF (n = 22), with paroxysmal AF (n = 22) and with persistent AF (n = 20). mRNA sequencing previously uncovered upregulated epithelial to mesenchymal transition, endothelial cell proliferation and extracellular matrix remodelling involving glycoproteins and proteoglycans in AF. MiRNA co-sequencing discovered miRNAs regulating the mRNA expression changes. Key downregulated miRNAs included miR-135b-5p, miR-138-5p, miR-200a-3p, miR-200b-3p and miR-31-5p and key upregulated miRNAs were miR-144-3p, miR-15b-3p, miR-182-5p miR-18b-5p, miR-4306 and miR-206. MiRNA expression levels were negatively correlated with the expression levels of a multitude of predicted target genes. Downregulated miRNAs associated with increased gene expression are involved in upregulated epithelial and endothelial cell migration and glycosaminoglycan biosynthesis. In vitro inhibition of miR-135b-5p and miR-138-5p validated an effect of miRNAs on multiple predicted targets. Altogether, the discovered miRNAs may be explored in further functional studies as potential targets for anti-fibrotic therapies in AF.
Collapse
Affiliation(s)
- Nicoline W E van den Berg
- Amsterdam UMC, University of Amsterdam, Heart Center; Department of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Amsterdam Cardiovascular Sciences, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands.
| | - Makiri Kawasaki
- Amsterdam UMC, University of Amsterdam, Heart Center; Department of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Amsterdam Cardiovascular Sciences, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Fransisca A Nariswari
- Amsterdam UMC, University of Amsterdam, Heart Center; Department of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Amsterdam Cardiovascular Sciences, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Benedetta Fabrizi
- Amsterdam UMC, University of Amsterdam, Heart Center; Department of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Amsterdam Cardiovascular Sciences, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Jolien Neefs
- Amsterdam UMC, University of Amsterdam, Heart Center; Department of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Amsterdam Cardiovascular Sciences, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Ingeborg van der Made
- Amsterdam UMC, University of Amsterdam, Heart Center; Department of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Amsterdam Cardiovascular Sciences, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Robin Wesselink
- Amsterdam UMC, University of Amsterdam, Heart Center; Department of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Amsterdam Cardiovascular Sciences, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Wim Jan P van Boven
- Amsterdam UMC, University of Amsterdam, Heart Center; Department of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Amsterdam Cardiovascular Sciences, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Antoine H G Driessen
- Amsterdam UMC, University of Amsterdam, Heart Center; Department of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Amsterdam Cardiovascular Sciences, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Aldo Jongejan
- Amsterdam UMC, Department of Epidemiology and Data Science, University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Joris R de Groot
- Amsterdam UMC, University of Amsterdam, Heart Center; Department of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Amsterdam Cardiovascular Sciences, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands.
| |
Collapse
|
87
|
Benzoni P, Da Dalt L, Elia N, Popolizio V, Cospito A, Giannetti F, Dell’Era P, Olesen MS, Bucchi A, Baruscotti M, Norata GD, Barbuti A. PITX2 gain-of-function mutation associated with atrial fibrillation alters mitochondrial activity in human iPSC atrial-like cardiomyocytes. Front Physiol 2023; 14:1250951. [PMID: 38028792 PMCID: PMC10679737 DOI: 10.3389/fphys.2023.1250951] [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] [Received: 06/30/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia worldwide; however, the underlying causes of AF initiation are still poorly understood, particularly because currently available models do not allow in distinguishing the initial causes from maladaptive remodeling that induces and perpetuates AF. Lately, the genetic background has been proven to be important in the AF onset. iPSC-derived cardiomyocytes, being patient- and mutation-specific, may help solve this diatribe by showing the initial cell-autonomous changes underlying the development of the disease. Transcription factor paired-like homeodomain 2 (PITX2) has been identified as a key regulator of atrial development/differentiation, and the PITX2 genomic locus has the highest association with paroxysmal AF. PITX2 influences mitochondrial activity, and alterations in either its expression or function have been widely associated with AF. In this work, we investigate the activity of mitochondria in iPSC-derived atrial cardiomyocytes (aCMs) obtained from a young patient (24 years old) with paroxysmal AF, carrying a gain-of-function mutation in PITX2 (rs138163892) and from its isogenic control (CTRL) in which the heterozygous point mutation has been reverted to WT. PITX2 aCMs show a higher mitochondrial content, increased mitochondrial activity, and superoxide production under basal conditions when compared to CTRL aCMs. However, increasing mitochondrial workload by FCCP or β-adrenergic stimulation allows us to unmask mitochondrial defects in PITX2 aCMs, which are incapable of responding efficiently to the higher energy demand, determining ATP deficiency.
Collapse
Affiliation(s)
- Patrizia Benzoni
- The Cell Physiology MiLab, Department Biosciences, Università degli Studi di Milano, Milano, Italy
| | - Lorenzo Da Dalt
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy
| | - Noemi Elia
- The Cell Physiology MiLab, Department Biosciences, Università degli Studi di Milano, Milano, Italy
- Cell Factory, Fondazione IRCCS Ca’Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Vera Popolizio
- The Cell Physiology MiLab, Department Biosciences, Università degli Studi di Milano, Milano, Italy
| | - Alessandro Cospito
- The Cell Physiology MiLab, Department Biosciences, Università degli Studi di Milano, Milano, Italy
| | - Federica Giannetti
- The Cell Physiology MiLab, Department Biosciences, Università degli Studi di Milano, Milano, Italy
- Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Istituto Auxologico Italiano IRCCS, Milano, Italy
| | - Patrizia Dell’Era
- Department of Molecular and Translational Medicine, Università degli Studi di Brescia, Brescia, Italy
| | - Morten S. Olesen
- The Heart Centre, Rigshospitalet, Laboratory for Molecular Cardiology, Department of Cardiology, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Annalisa Bucchi
- The Cell Physiology MiLab, Department Biosciences, Università degli Studi di Milano, Milano, Italy
| | - Mirko Baruscotti
- The Cell Physiology MiLab, Department Biosciences, Università degli Studi di Milano, Milano, Italy
| | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy
| | - Andrea Barbuti
- The Cell Physiology MiLab, Department Biosciences, Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
88
|
Jonmundsson T, Steindorsdottir AE, Austin TR, Frick EA, Axelsson GT, Launer L, Psaty BM, Loureiro J, Orth AP, Aspelund T, Emilsson V, Floyd JS, Jennings L, Gudnason V, Gudmundsdottir V. A proteomic analysis of atrial fibrillation in a prospective longitudinal cohort (AGES-Reykjavik study). Europace 2023; 25:euad320. [PMID: 37967346 PMCID: PMC10685397 DOI: 10.1093/europace/euad320] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/01/2023] [Accepted: 10/06/2023] [Indexed: 11/17/2023] Open
Abstract
AIMS Atrial fibrillation (AF) is associated with high risk of comorbidities and mortality. Our aim was to examine causal and predictive relationships between 4137 serum proteins and incident AF in the prospective population-based Age, Gene/Environment Susceptibility-Reykjavik (AGES-Reykjavik) study. METHODS AND RESULTS The study included 4765 participants, of whom 1172 developed AF. Cox proportional hazards regression models were fitted for 4137 baseline protein measurements adjusting for known risk factors. Protein associations were tested for replication in the Cardiovascular Health Study (CHS). Causal relationships were examined in a bidirectional, two-sample Mendelian randomization analysis. The time-dependent area under the receiver operating characteristic curve (AUC)-statistic was examined as protein levels and an AF-polygenic risk score (PRS) were added to clinical risk models. The proteomic signature of incident AF consisted of 76 proteins, of which 63 (83%) were novel and 29 (38%) were replicated in CHS. The signature included both N-terminal prohormone of brain natriuretic peptide (NT-proBNP)-dependent (e.g. CHST15, ATP1B1, and SVEP1) and independent components (e.g. ASPN, AKR1B, and LAMA1/LAMB1/LAMC1). Nine causal candidates were identified (TAGLN, WARS, CHST15, CHMP3, COL15A1, DUSP13, MANBA, QSOX2, and SRL). The reverse causal analysis suggested that most AF-associated proteins were affected by the genetic liability to AF. N-terminal prohormone of brain natriuretic peptide improved the prediction of incident AF events close to baseline with further improvements gained by the AF-PRS at all time points. CONCLUSION The AF proteomic signature includes biologically relevant proteins, some of which may be causal. It mainly reflects an NT-proBNP-dependent consequence of the genetic liability to AF. N-terminal prohormone of brain natriuretic peptide is a promising marker for incident AF in the short term, but risk assessment incorporating a PRS may improve long-term risk assessment.
Collapse
Affiliation(s)
- Thorarinn Jonmundsson
- Icelandic Heart Association, Holtasmari 1, Kopavogur 201, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik 101, Iceland
| | | | - Thomas R Austin
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Elisabet A Frick
- Icelandic Heart Association, Holtasmari 1, Kopavogur 201, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik 101, Iceland
| | - Gisli T Axelsson
- Icelandic Heart Association, Holtasmari 1, Kopavogur 201, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik 101, Iceland
| | - Lenore Launer
- Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, Bethesda, MD, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Health Systems and Population Health, University of Washington, Seattle, WA, USA
| | | | | | - Thor Aspelund
- Icelandic Heart Association, Holtasmari 1, Kopavogur 201, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik 101, Iceland
| | - Valur Emilsson
- Icelandic Heart Association, Holtasmari 1, Kopavogur 201, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik 101, Iceland
| | - James S Floyd
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | | | - Vilmundur Gudnason
- Icelandic Heart Association, Holtasmari 1, Kopavogur 201, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik 101, Iceland
| | - Valborg Gudmundsdottir
- Icelandic Heart Association, Holtasmari 1, Kopavogur 201, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik 101, Iceland
| |
Collapse
|
89
|
Wang D, Wang X, Yang T, Tian H, Su Y, Wang Q. Long Non-Coding RNA Dancr Affects Myocardial Fibrosis in Atrial Fibrillation Mice via the MicroRNA-146b-5p/Smad5 Axis. ACTA CARDIOLOGICA SINICA 2023; 39:841-853. [PMID: 38022420 PMCID: PMC10646592 DOI: 10.6515/acs.202311_39(6).20230619b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 06/19/2023] [Indexed: 12/01/2023]
Abstract
Objectives Atrial fibrillation (AF) is the most frequent arrhythmia, and myocardial fibrosis (MF) has a close association with atrial remodeling and leads to AF. This study aimed to explore the function of the long non-coding RNA (lncRNA) differentiation antagonizing non-protein coding RNA (Dancr)/microRNA (miR)-146b-5p/Smad5 axis on MF in AF mice. Methods AF mouse models were established. Overexpression Dancr lentivirus was injected into AF mice to increase Dancr expression in myocardial tissues. LncRNA Dancr, miR-146b-5p, and Smad5 expression levels and inflammatory factors (IL-18 and TNF-α) in the myocardial tissues were measured. MF was measured and the expression levels of MF-related genes (COL1A1, α-SMA, and FN1) were detected. In addition, in vitro HL-1 cell rapid pacing models were constructed, and after lncRNA Dancr and miR-146b-5p-related construct transfection, cell viability and cell apoptosis were determined. Results LncRNA Dancr up-regulation ameliorated MF in the AF mice, reduced IL-18 and TNF-α expression levels in myocardial tissues, and decreased COL1A1, α-SMA, and FN1 expression levels. The in vitro HL-1 cell rapid pacing models suggested that miR-146b-5p overexpression reversed the inhibitory effects of lncRNA Dancr overexpression on MF in HL-1 cells, and Smad5 interference reversed the ameliorative effects of miR-146b-5p interference on MF in HL-1 cells. Conclusions LncRNA Dancr can sponge miR-146b-5p to promote Smad5 expression, thereby delaying MF in AF mice.
Collapse
Affiliation(s)
- Dejin Wang
- Department of Cardiology, Zibo Central Hospital, Zibo 255036, Shandong, People's Republic of China
| | - Xiqian Wang
- Department of Cardiology, Zibo Central Hospital, Zibo 255036, Shandong, People's Republic of China
| | - Tianxiao Yang
- Department of Cardiology, Zibo Central Hospital, Zibo 255036, Shandong, People's Republic of China
| | - Hongliang Tian
- Department of Cardiology, Zibo Central Hospital, Zibo 255036, Shandong, People's Republic of China
| | - Yuanzhen Su
- Department of Cardiology, Zibo Central Hospital, Zibo 255036, Shandong, People's Republic of China
| | - Qilei Wang
- Department of Cardiology, Zibo Central Hospital, Zibo 255036, Shandong, People's Republic of China
| |
Collapse
|
90
|
Mucke HAM. Drug Repurposing Patent Applications July-September 2023. Assay Drug Dev Technol 2023; 21:385-391. [PMID: 37948550 DOI: 10.1089/adt.2023.125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2023] Open
|
91
|
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
|
92
|
Zhang J, Guo C. Current progress of ferroptosis in cardiovascular diseases. Front Cardiovasc Med 2023; 10:1259219. [PMID: 37942067 PMCID: PMC10628442 DOI: 10.3389/fcvm.2023.1259219] [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] [Received: 07/15/2023] [Accepted: 09/29/2023] [Indexed: 11/10/2023] Open
Abstract
Ferroptosis, a newly recognized form of nonapoptotic regulated cell death, is characterized by iron-dependent lipid peroxidation. Biological processes, such as iron metabolism, lipid peroxidation, and amino acid metabolism, are involved in the process of ferroptosis. However, the related molecular mechanism of ferroptosis has not yet been completely clarified, and specific and sensitive biomarkers for ferroptosis need to be explored. Recently, studies have revealed that ferroptosis probably causes or exacerbates the progress of cardiovascular diseases, and could be the potential therapeutic target for cardiovascular diseases. In this review, we summarize the molecular mechanisms regulating ferroptosis, inducers or inhibitors of ferroptosis, and the current progresses of ferroptosis in cardiovascular diseases. Furthermore, we discuss the emerging challenges and future perspectives, which may provide novel insights into the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
| | - Caixia Guo
- Cardiovascular Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
93
|
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
|
94
|
Kupusovic J, Weber M, Bruns F, Kessler L, Pesch E, Bohnen J, Dobrev D, Rassaf T, Wakili R, Rischpler C, Siebermair J. PET/CT-identified atrial hypermetabolism is an index of atrial inflammation in patients with atrial fibrillation. J Nucl Cardiol 2023; 30:1761-1772. [PMID: 37592057 DOI: 10.1007/s12350-023-03248-w] [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: 10/28/2022] [Accepted: 01/17/2023] [Indexed: 08/19/2023]
Abstract
BACKGROUND Although atrial inflammation has been implicated in the pathophysiology of atrial fibrillation (AF), the identification of atrial inflammation remains challenging. We aimed to establish a positron emission tomography/computed tomography (PET/CT) protocol with 18Fluor-labeled fluorodeoxyglucose (18F-FDG) for the detection of atrial hypermetabolism as surrogate for inflammation in AF. METHODS We included n = 75 AF and n = 75 non-AF patients undergoing three common PET/CT protocols (n = 25 per group) optimized for the detection of (a) inflammation and (b) malignancy in predefined fasting protocols, and (c) cardiac viability allowing for maximized glucose uptake. 18F-FDG-uptake was analyzed in predefined loci. RESULTS Differences of visual atrial uptake in AF vs non-AF patients were observed in fasting (inflammation [13/25 vs 0/25] and malignancy [10/25 vs 0/25]) protocols while viability protocols showed non-specific uptake in both the groups. In the inflammation protocol, AF patients showed higher uptake in the right atrium [(SUVmax: 2.5 ± .7 vs 2.0 ± .7, P = .01), atrial appendage (SUVmax: 2.4 ± .7 vs 2.0 ± .6, P = .03), and epicardial adipose tissue (SUVmax: 1.4 ± .5 vs 1.1 ± .4, P = .04)]. Malignancy and viability protocols failed to differentiate between AF and non-AF. CONCLUSION Glucose uptake suppression protocols appear suitable in detecting differential atrial 18F-FDG uptake between AF and non-AF patients. Imaging-based assessment of inflammation might help to stratify AF patients offering individualized therapeutic approaches.
Collapse
Affiliation(s)
- J Kupusovic
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany
- Department of Cardiology and Vascular Medicine, University Hospital Frankfurt, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - M Weber
- Department of Nuclear Medicine, University Duisburg-Essen, Essen, Germany
| | - F Bruns
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - L Kessler
- Department of Nuclear Medicine, University Duisburg-Essen, Essen, Germany
| | - E Pesch
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany
| | - J Bohnen
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany
| | - D Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, QC, Canada
| | - T Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany
| | - R Wakili
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany.
- Department of Cardiology and Vascular Medicine, University Hospital Frankfurt, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany.
- German Centre for Cardiovascular Research (DZHK) Partner Site Rhine/Main, Frankfurt, Germany.
| | - C Rischpler
- Department of Nuclear Medicine, University Duisburg-Essen, Essen, Germany
- Department of Nuclear Medicine, Klinikum Stuttgart, Stuttgart, Germany
| | - J Siebermair
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany
- Department of Cardiology, Krankenhaus Göttlicher Heiland GmbH, Vienna, Austria
| |
Collapse
|
95
|
Herrera NT, Zhang X, Ni H, Maleckar MM, Heijman J, Dobrev D, Grandi E, Morotti S. Dual effects of the small-conductance Ca 2+-activated K + current on human atrial electrophysiology and Ca 2+-driven arrhythmogenesis: an in silico study. Am J Physiol Heart Circ Physiol 2023; 325:H896-H908. [PMID: 37624096 PMCID: PMC10659325 DOI: 10.1152/ajpheart.00362.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/11/2023] [Accepted: 08/11/2023] [Indexed: 08/26/2023]
Abstract
By sensing changes in intracellular Ca2+, small-conductance Ca2+-activated K+ (SK) channels dynamically regulate the dynamics of the cardiac action potential (AP) on a beat-to-beat basis. Given their predominance in atria versus ventricles, SK channels are considered a promising atrial-selective pharmacological target against atrial fibrillation (AF), the most common cardiac arrhythmia. However, the precise contribution of SK current (ISK) to atrial arrhythmogenesis is poorly understood, and may potentially involve different mechanisms that depend on species, heart rates, and degree of AF-induced atrial remodeling. Both reduced and enhanced ISK have been linked to AF. Similarly, both SK channel up- and downregulation have been reported in chronic AF (cAF) versus normal sinus rhythm (nSR) patient samples. Here, we use our multiscale modeling framework to obtain mechanistic insights into the contribution of ISK in human atrial cardiomyocyte electrophysiology. We simulate several protocols to quantify how ISK modulation affects the regulation of AP duration (APD), Ca2+ transient, refractoriness, and occurrence of alternans and delayed afterdepolarizations (DADs). Our simulations show that ISK activation shortens the APD and atrial effective refractory period, limits Ca2+ cycling, and slightly increases the propensity for alternans in both nSR and cAF conditions. We also show that increasing ISK counteracts DAD development by enhancing the repolarization force that opposes the Ca2+-dependent depolarization. Taken together, our results suggest that increasing ISK in human atrial cardiomyocytes could promote reentry while protecting against triggered activity. Depending on the leading arrhythmogenic mechanism, ISK inhibition may thus be a beneficial or detrimental anti-AF strategy.NEW & NOTEWORTHY Using our established framework for human atrial myocyte simulations, we investigated the role of the small-conductance Ca2+-activated K+ current (ISK) in the regulation of cell function and the development of Ca2+-driven arrhythmias. We found that ISK inhibition, a promising atrial-selective pharmacological strategy against atrial fibrillation, counteracts the reentry-promoting abbreviation of atrial refractoriness, but renders human atrial myocytes more vulnerable to delayed afterdepolarizations, thus potentially increasing the propensity for ectopic (triggered) activity.
Collapse
Affiliation(s)
- Nathaniel T Herrera
- Department of Pharmacology, University of California Davis, Davis, California, United States
| | - Xianwei Zhang
- Department of Pharmacology, University of California Davis, Davis, California, United States
| | - Haibo Ni
- Department of Pharmacology, University of California Davis, Davis, California, United States
| | - Mary M Maleckar
- Department of Computational Physiology, Simula Research Laboratory, Oslo, Norway
| | - Jordi Heijman
- Department of Cardiology, Faculty of Health, Medicine, and Life Sciences, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Dobromir Dobrev
- Faculty of Medicine, West German Heart and Vascular Center, Institute of Pharmacology, University Duisburg-Essen, Essen, Germany
- Department of Medicine, Montreal Heart Institute and Université de Montréal, Montreal, Quebec, Canada
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas, United States
| | - Eleonora Grandi
- Department of Pharmacology, University of California Davis, Davis, California, United States
| | - Stefano Morotti
- Department of Pharmacology, University of California Davis, Davis, California, United States
| |
Collapse
|
96
|
Shi S, Mao X, Lv J, Wang Y, Zhang X, Shou X, Zhang B, Li Y, Wu H, Song Q, Hu Y. Qi-Po-Sheng-Mai granule ameliorates Ach-CaCl 2 -induced atrial fibrillation by regulating calcium homeostasis in cardiomyocytes. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 119:155017. [PMID: 37597360 DOI: 10.1016/j.phymed.2023.155017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 07/15/2023] [Accepted: 08/06/2023] [Indexed: 08/21/2023]
Abstract
BACKGROUND Atrial fibrillation (AF) is one of the most common arrhythmias encountered in clinical settings. Currently, the pathophysiology of AF remains unclear, which severely limits the effectiveness and safety of medical therapies. The Chinese herbal formula Qi-Po-Sheng-Mai Granule (QPSM) has been widely used in China to treat AF. However, its pharmacological and molecular mechanisms remain unknown. PURPOSE The purpose of this study was to investigate the molecular mechanisms and potential targets of QPSM for AF. STUDY DESIGN AND METHODS The AF model was induced by Ach (66 μg/ml) and CaCl2 (10 mg/kg), and the dose of 0.1 ml/100 g was injected into the tail vein for 5 weeks. QPSM was administered daily at doses of 4.42 and 8.84 g/kg, and amiodarone (0.18 g/kg) was used as the positive control. The effect of QPSM on AF was assessed by electrocardiogram, echocardiography, and histopathological analysis. Then, we employed network pharmacology with single nucleus RNA sequencing (snRNA-Seq) to investigate the molecular mechanisms and potential targets of QPSM for AF. Furthermore, high performance liquid chromatography (HPLC) method was used for component analysis of QPSM, and molecular docking was used to verify the potential targets. Using the IonOptix single cell contraction and ion synchronization test equipment, single myocyte length and calcium ion variations were observed in real time. The expression levels of calcium Transporter-related proteins were detected by western blot and immunohistochemistry. RESULTS Based on an Ach-CaCl2-induced AF model, we found that QPSM treatment significantly reduced atrial electrical remodeling-related markers, such as AF inducibility and duration, and attenuated atrial dilation and fibrosis. Network pharmacology identified 52 active ingredients and 119 potential targets for QPSM in the treatment of AF, and 45 Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were enriched, among which calcium pathway had the greatest impact. Using single nucleus sequencing (snRNA-seq), we identified cardiomyocytes as the most differentially expressed in response to drug treatment, with nine differentially expressed genes enriched in calcium signaling pathways. High performance liquid chromatography and molecular docking confirmed that the core components of QPSM strongly bind to the key factors in the calcium signaling pathway. Additional experiments have shown that QPSM increases calcium transients (CaT) and contractility in the individual cardiomyocyte. This was accomplished by increasing the expression of CACNA1C and SERCA2a and decreasing the expression of CAMK2B and NCX1. CONCLUSION The present study has systematically elucidated the role of QPSM in maintaining calcium homeostasis in cardiomyocytes through the regulation of calcium transporters, which could lead to new drug development ideas for AF.
Collapse
Affiliation(s)
- Shuqing Shi
- Department of Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, 5 Beixiange Street Xicheng District, Beijing 100053, China
| | - Xinxin Mao
- Department of Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, 5 Beixiange Street Xicheng District, Beijing 100053, China
| | - Jiayu Lv
- Department of Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, 5 Beixiange Street Xicheng District, Beijing 100053, China
| | - Yajiao Wang
- Department of Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, 5 Beixiange Street Xicheng District, Beijing 100053, China
| | - Xuesong Zhang
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xintian Shou
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Bingxuan Zhang
- Department of Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, 5 Beixiange Street Xicheng District, Beijing 100053, China
| | - Yumeng Li
- Department of Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, 5 Beixiange Street Xicheng District, Beijing 100053, China
| | - Huaqin Wu
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qingqiao Song
- Department of Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, 5 Beixiange Street Xicheng District, Beijing 100053, China.
| | - Yuanhui Hu
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
97
|
Remme CA, Heijman J, Gomez AM, Zaza A, Odening KE. 25 years of basic and translational science in EP Europace: novel insights into arrhythmia mechanisms and therapeutic strategies. Europace 2023; 25:euad210. [PMID: 37622575 PMCID: PMC10450791 DOI: 10.1093/europace/euad210] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 06/19/2023] [Indexed: 08/26/2023] Open
Abstract
In the last 25 years, EP Europace has published more than 300 basic and translational science articles covering different arrhythmia types (ranging from atrial fibrillation to ventricular tachyarrhythmias), different diseases predisposing to arrhythmia formation (such as genetic arrhythmia disorders and heart failure), and different interventional and pharmacological anti-arrhythmic treatment strategies (ranging from pacing and defibrillation to different ablation approaches and novel drug-therapies). These studies have been conducted in cellular models, small and large animal models, and in the last couple of years increasingly in silico using computational approaches. In sum, these articles have contributed substantially to our pathophysiological understanding of arrhythmia mechanisms and treatment options; many of which have made their way into clinical applications. This review discusses a representative selection of EP Europace manuscripts covering the topics of pacing and ablation, atrial fibrillation, heart failure and pro-arrhythmic ventricular remodelling, ion channel (dys)function and pharmacology, inherited arrhythmia syndromes, and arrhythmogenic cardiomyopathies, highlighting some of the advances of the past 25 years. Given the increasingly recognized complexity and multidisciplinary nature of arrhythmogenesis and continued technological developments, basic and translational electrophysiological research is key advancing the field. EP Europace aims to further increase its contribution to the discovery of arrhythmia mechanisms and the implementation of mechanism-based precision therapy approaches in arrhythmia management.
Collapse
Affiliation(s)
- Carol Ann Remme
- Department of Experimental Cardiology, Amsterdam UMC location University of Amsterdam, Heart Centre, Academic Medical Center, Room K2-104.2, Meibergdreef 11, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam, The Netherlands
| | - Jordi Heijman
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Ana M Gomez
- Signaling and Cardiovascular Pathophysiology, UMR-S 1180, Inserm, Université Paris-Saclay, 91400 Orsay, France
| | - Antonio Zaza
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy
| | - Katja E Odening
- Translational Cardiology, Department of Cardiology and Department of Physiology, Inselspital University Hospital Bern, University of Bern, 3012 Bern, Switzerland
| |
Collapse
|
98
|
Parra-Lucares A, Villa E, Romero-Hernández E, Méndez-Valdés G, Retamal C, Vizcarra G, Henríquez I, Maldonado-Morales EAJ, Grant-Palza JH, Ruíz-Tagle S, Estrada-Bobadilla V, Toro L. Tic-Tac: A Translational Approach in Mechanisms Associated with Irregular Heartbeat and Sinus Rhythm Restoration in Atrial Fibrillation Patients. Int J Mol Sci 2023; 24:12859. [PMID: 37629037 PMCID: PMC10454641 DOI: 10.3390/ijms241612859] [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: 06/01/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
Atrial fibrillation (AF) is a prevalent cardiac condition predominantly affecting older adults, characterized by irregular heartbeat rhythm. The condition often leads to significant disability and increased mortality rates. Traditionally, two therapeutic strategies have been employed for its treatment: heart rate control and rhythm control. Recent clinical studies have emphasized the critical role of early restoration of sinus rhythm in improving patient outcomes. The persistence of the irregular rhythm allows for the progression and structural remodeling of the atria, eventually leading to irreversible stages, as observed clinically when AF becomes permanent. Cardioversion to sinus rhythm alters this progression pattern through mechanisms that are still being studied. In this review, we provide an in-depth analysis of the pathophysiological mechanisms responsible for maintaining AF and how they are modified during sinus rhythm restoration using existing therapeutic strategies at different stages of clinical investigation. Moreover, we explore potential future therapeutic approaches, including the promising prospect of gene therapy.
Collapse
Affiliation(s)
- Alfredo Parra-Lucares
- Critical Care Unit, Department of Medicine, Hospital Clínico Universidad de Chile, Santiago 8380420, Chile
- Cardiovascular Department, Hospital Clínico Universidad de Chile, Santiago 8380420, Chile
| | - Eduardo Villa
- School of Medicine, Faculty of Medicine, Universidad de Chile, Santiago 8380420, Chile
| | | | - Gabriel Méndez-Valdés
- School of Medicine, Faculty of Medicine, Universidad de Chile, Santiago 8380420, Chile
| | - Catalina Retamal
- School of Medicine, Faculty of Medicine, Universidad de Chile, Santiago 8380420, Chile
| | - Geovana Vizcarra
- Division of Internal Medicine, Department of Medicine, Hospital Clínico Universidad de Chile, Santiago 8380420, Chile
| | - Ignacio Henríquez
- School of Medicine, Faculty of Medicine, Universidad de Chile, Santiago 8380420, Chile
| | | | - Juan H. Grant-Palza
- School of Medicine, Faculty of Medicine, Universidad de Chile, Santiago 8380420, Chile
| | - Sofía Ruíz-Tagle
- School of Medicine, Faculty of Medicine, Universidad de Chile, Santiago 8380420, Chile
| | | | - Luis Toro
- Division of Nephrology, Department of Medicine, Hospital Clínico Universidad de Chile, Santiago 8380420, Chile
- Centro de Investigación Clínica Avanzada, Hospital Clínico, Universidad de Chile, Santiago 8380420, Chile
| |
Collapse
|
99
|
Wang X, Huang T, Jia J. Proteome-Wide Mendelian Randomization Analysis Identified Potential Drug Targets for Atrial Fibrillation. J Am Heart Assoc 2023; 12:e029003. [PMID: 37581400 PMCID: PMC10492951 DOI: 10.1161/jaha.122.029003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 06/27/2023] [Indexed: 08/16/2023]
Abstract
Background Finding effective and safe therapeutic drugs for atrial fibrillation (AF) is an important concern for clinicians. Proteome-wide Mendelian randomization analysis provides new ideas for finding potential drug targets. Methods and Results Using a proteome-wide Mendelian randomization approach, we assessed the genetic predictive causality between thousands of proteins and AF risk and found that genetically predicted plasma levels of phosphomevalonate kinase, tumor necrosis factor ligand superfamily member 12, sulfhydryl oxidase 2, interleukin-6 receptor subunit alpha, and low-affinity immunoglobulin gamma Fc region receptor II-b might decrease AF risk, while genetically predicted plasma levels of beta-mannosidase, collagen alpha-1(XV) chain, ANXA4 (annexin A4), COF2 (cofilin-2), and RAB1A (Ras-related protein Rab-1A) might increase AF risk (P<3.4×10-5). By using different Mendelian randomization methods and instrumental variable selection thresholds, we performed sensitivity analyses in 30 scenarios to test the robustness of positive findings. Replication analyses were also performed in independent samples to further avoid false-positive findings. Drugs targeting tumor necrosis factor ligand superfamily member 12, interleukin-6 receptor subunit alpha, low-affinity immunoglobulin gamma Fc region receptor II-b, and annexin A4 are approved or in development. The results of the phenome-wide Mendelian randomization analysis showed that changing the plasma levels of phosphomevalonate kinase, cofilin-2, annexin A4, Ras-related protein Rab-1A, sulfhydryl oxidase 2, and collagen alpha-1(XV) chain did not increase the risk of other diseases while decreasing the risk of AF. Conclusions We found a significant causal association between genetically predicted levels of 10 plasma proteins and AF risk. Four of these proteins have drugs targeting them that are approved or in development, and our results suggest the potential for these drugs to treat AF or cause AF. Sulfhydryl oxidase 2, low-affinity immunoglobulin gamma Fc region receptor II-b, and beta-mannosidase have not been suggested by previous laboratory or epidemiological studies to be associated with AF and may reveal new pathophysiological pathways as well as therapeutic targets for AF.
Collapse
Affiliation(s)
- Xinpei Wang
- Department of Biostatistics, School of Public Health Peking University Beijing China
| | - Tao Huang
- Department of Epidemiology & Biostatistics, School of Public Health Peking University Beijing China
- Center for Intelligent Public Health, Academy for Artificial Intelligence Peking University Beijing China
- Key Laboratory of Molecular Cardiovascular Sciences (Peking University), Ministry of Education Beijing China
| | - Jinzhu Jia
- Department of Biostatistics, School of Public Health Peking University Beijing China
- Center for Statistical Science Peking University Beijing China
| |
Collapse
|
100
|
Gawałko M, Linz D, Dobrev D. Does weight matter in cryoballoon ablation? IJC HEART & VASCULATURE 2023; 47:101251. [PMID: 37576074 PMCID: PMC10422653 DOI: 10.1016/j.ijcha.2023.101251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Affiliation(s)
- Monika Gawałko
- 1st Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Germany
- Department of Cardiology, Maastricht University Medical Centre and Cardiovascular Research Institute Maastricht, Maastricht, the Netherlands
| | - Dominik Linz
- Department of Cardiology, Maastricht University Medical Centre and Cardiovascular Research Institute Maastricht, Maastricht, the Netherlands
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Centre for Heart Rhythm Disorders, Royal Adelaide Hospital, University of Adelaide, Adelaide, Australia
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Germany
- Department of Integrative Physiology, Baylor College of Medicine, Houston, USA
- Department of Medicine and Research Center, Montréal Heart Institute and Université de Montréal, Montréal, Canada
| |
Collapse
|