1
|
Băghină RM, Crișan S, Luca S, Pătru O, Lazăr MA, Văcărescu C, Negru AG, Luca CT, Gaiță D. Association between Inflammation and New-Onset Atrial Fibrillation in Acute Coronary Syndromes. J Clin Med 2024; 13:5088. [PMID: 39274304 PMCID: PMC11396258 DOI: 10.3390/jcm13175088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 09/16/2024] Open
Abstract
Acute coronary syndrome (ACS) is a complex clinical syndrome that encompasses acute myocardial infarction (AMI) and unstable angina (UA). Its underlying mechanism refers to coronary plaque disruption, with consequent platelet aggregation and thrombosis. Inflammation plays an important role in the progression of atherosclerosis by mediating the removal of necrotic tissue following myocardial infarction and shaping the repair processes that are essential for the recovery process after ACS. As a chronic inflammatory disorder, atherosclerosis is characterized by dysfunctional immune inflammation involving interactions between immune (macrophages, T lymphocytes, and monocytes) and vascular cells (endothelial cells and smooth muscle cells). New-onset atrial fibrillation (NOAF) is one of the most common arrhythmic complications in the setting of acute coronary syndromes, especially in the early stages, when the myocardial inflammatory reaction is at its maximum. The main changes in the atrial substrate are due to atrial ischemia and acute infarcts that can be attributed to neurohormonal factors. The high incidence of atrial fibrillation (AF) post-myocardial infarction may be secondary to inflammation. Inflammatory response and immune system cells have been involved in the initiation and development of atrial fibrillation. Several inflammatory indexes, such as C-reactive protein and interleukins, have been demonstrated to be predictive of prognosis in patients with ACS. The cell signaling activation patterns associated with fibrosis, apoptosis, and hypertrophy are forms of cardiac remodeling that occur at the atrial level, predisposing to AF. According to a recent study, the presence of fibrosis and lymphomononuclear infiltration in the atrial tissue was associated with a prior history of AF. However, inflammation may contribute to both the occurrence/maintenance of AF and its thromboembolic complications.
Collapse
Affiliation(s)
- Ruxandra-Maria Băghină
- Cardiology Department, "Victor Babes" University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
- Research Center of the Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
| | - Simina Crișan
- Cardiology Department, "Victor Babes" University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
- Research Center of the Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
| | - Silvia Luca
- Cardiology Department, "Victor Babes" University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
- Research Center of the Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
| | - Oana Pătru
- Cardiology Department, "Victor Babes" University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
- Research Center of the Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
| | - Mihai-Andrei Lazăr
- Cardiology Department, "Victor Babes" University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
- Research Center of the Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
| | - Cristina Văcărescu
- Cardiology Department, "Victor Babes" University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
- Research Center of the Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
| | - Alina Gabriela Negru
- Cardiology Department, "Victor Babes" University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
- Research Center of the Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
| | - Constantin-Tudor Luca
- Cardiology Department, "Victor Babes" University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
- Research Center of the Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
| | - Dan Gaiță
- Cardiology Department, "Victor Babes" University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
- Research Center of the Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
| |
Collapse
|
2
|
Lei M, Salvage SC, Jackson AP, Huang CLH. Cardiac arrhythmogenesis: roles of ion channels and their functional modification. Front Physiol 2024; 15:1342761. [PMID: 38505707 PMCID: PMC10949183 DOI: 10.3389/fphys.2024.1342761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 01/22/2024] [Indexed: 03/21/2024] Open
Abstract
Cardiac arrhythmias cause significant morbidity and mortality and pose a major public health problem. They arise from disruptions in the normally orderly propagation of cardiac electrophysiological activation and recovery through successive cardiomyocytes in the heart. They reflect abnormalities in automaticity, initiation, conduction, or recovery in cardiomyocyte excitation. The latter properties are dependent on surface membrane electrophysiological mechanisms underlying the cardiac action potential. Their disruption results from spatial or temporal instabilities and heterogeneities in the generation and propagation of cellular excitation. These arise from abnormal function in their underlying surface membrane, ion channels, and transporters, as well as the interactions between them. The latter, in turn, form common regulatory targets for the hierarchical network of diverse signaling mechanisms reviewed here. In addition to direct molecular-level pharmacological or physiological actions on these surface membrane biomolecules, accessory, adhesion, signal transduction, and cytoskeletal anchoring proteins modify both their properties and localization. At the cellular level of excitation-contraction coupling processes, Ca2+ homeostatic and phosphorylation processes affect channel activity and membrane excitability directly or through intermediate signaling. Systems-level autonomic cellular signaling exerts both acute channel and longer-term actions on channel expression. Further upstream intermediaries from metabolic changes modulate the channels both themselves and through modifying Ca2+ homeostasis. Finally, longer-term organ-level inflammatory and structural changes, such as fibrotic and hypertrophic remodeling, similarly can influence all these physiological processes with potential pro-arrhythmic consequences. These normal physiological processes may target either individual or groups of ionic channel species and alter with particular pathological conditions. They are also potentially alterable by direct pharmacological action, or effects on longer-term targets modifying protein or cofactor structure, expression, or localization. Their participating specific biomolecules, often clarified in experimental genetically modified models, thus constitute potential therapeutic targets. The insights clarified by the physiological and pharmacological framework outlined here provide a basis for a recent modernized drug classification. Together, they offer a translational framework for current drug understanding. This would facilitate future mechanistically directed therapeutic advances, for which a number of examples are considered here. The latter are potentially useful for treating cardiac, in particular arrhythmic, disease.
Collapse
Affiliation(s)
- Ming Lei
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Samantha C. Salvage
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Antony P. Jackson
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Christopher L.-H. Huang
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
- Physiological Laboratory, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
3
|
Niu J, Zhang M, Liu P, Hua C, Zhong G. Research progress on predicting atrial fibrillation recurrence after radiofrequency ablation based on electrocardiogram-related parameters. J Electrocardiol 2023; 81:146-152. [PMID: 37708737 DOI: 10.1016/j.jelectrocard.2023.08.015] [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: 07/01/2023] [Revised: 08/18/2023] [Accepted: 08/28/2023] [Indexed: 09/16/2023]
Abstract
Atrial fibrillation (AF) is the most common arrhythmia. It is associated with increased stroke risks, thromboembolism, and other complications, which are great life and economic burdens for patients. In recent years, with the maturity of percutaneous catheter radiofrequency ablation (RFA) technology, it has become a first-line therapy for AF. However, some patients still experience AF recurrence (AFR) after RFA, which can cause serious consequences. Therefore, it is critical to identify appropriate parameters that are predictive of prognosis and to be able to translate the parameters easily into the clinical setting. Here, we reviewed possible predicting indicators for AFR, focusing on all the electrocardiogram indicators, such as P wave duration, PR interval and so on. It may provide valuable information for guiding clinical works.
Collapse
Affiliation(s)
- Jiayin Niu
- Heart Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Min Zhang
- Research Ward, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Pengfei Liu
- Heart Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Cuncun Hua
- Heart Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Guangzhen Zhong
- Heart Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China; Research Ward, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
4
|
Kim J, Shanmugasundaram A, Kim DS, Jeong YJ, Kanade PP, Kim ES, Lee BK, Lee DW. Quantitative assessment of cardiomyocyte mechanobiology through high-throughput cantilever-based functional well plate systems. Analyst 2023; 148:5133-5143. [PMID: 37695027 DOI: 10.1039/d3an01286g] [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: 09/12/2023]
Abstract
Proper regulation of the in vitro cell culture environment is essential for disease modelling and drug toxicity screening. The main limitation of well plates used for cell culture is that they cannot accurately maintain energy sources and compounds needed during cell growth. Herein, to understand the importance of perfusion in cardiomyocyte culture, changes in contractile force and heart rate during cardiomyocyte growth are systematically investigated, and the results are compared with those of a perfusion-free system. The proposed perfusion system consists of a Peltier refrigerator, a peristaltic pump, and a functional well plate. A functional well plate with 12 wells is made through injection moulding, with two tubes integrated in the cover for each well to continuously circulate the culture medium. The contractile force of cardiomyocytes growing on the cantilever surface is analysed through changes in cantilever displacement. The maturation of cardiomyocytes is evaluated through fluorescence staining and western blot; cardiomyocytes cultured in the perfusion system show greater maturity than those cultured in a manually replaced culture medium. The pH of the culture medium manually replaced at intervals of 3 days decreases to 6.8, resulting in an abnormal heartbeat, while cardiomyocytes cultured in the perfusion system maintained at pH 7.4 show improved contractility and a uniform heart rate. Two well-known ion channel blockers, verapamil and quinidine, are used to measure changes in the contractile force of cardiomyocytes from the two systems. Cardiomyocytes in the perfusion system show greater stability during drug toxicity screening, proving that the perfusion system provides a better environment for cell growth.
Collapse
Affiliation(s)
- Jongyun Kim
- MEMS and Nanotechnology Laboratory, School of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea.
- Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Arunkumar Shanmugasundaram
- MEMS and Nanotechnology Laboratory, School of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea.
- Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Dong-Su Kim
- MEMS and Nanotechnology Laboratory, School of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea.
- Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju 61186, Republic of Korea
- Green Energy & Nano Technology R&D Group, Korea Institute of Industrial Technology (KITECH), Gwangju, 61012, Republic of Korea
| | - Yun-Jin Jeong
- MEMS and Nanotechnology Laboratory, School of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea.
- Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Pooja P Kanade
- MEMS and Nanotechnology Laboratory, School of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea.
- Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Eung-Sam Kim
- Department of Biological Sciences, Chonnam National University, Gwangju, 61186, Republic of Korea
- Center for Next-Generation Sensor Research and Development, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Bong-Kee Lee
- MEMS and Nanotechnology Laboratory, School of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea.
- Center for Next-Generation Sensor Research and Development, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Dong-Weon Lee
- MEMS and Nanotechnology Laboratory, School of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea.
- Center for Next-Generation Sensor Research and Development, Chonnam National University, Gwangju 61186, Republic of Korea
- Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju 61186, Republic of Korea
| |
Collapse
|
5
|
Oancea AF, Jigoranu RA, Morariu PC, Miftode RS, Trandabat BA, Iov DE, Cojocaru E, Costache II, Baroi LG, Timofte DV, Tanase DM, Floria M. Atrial Fibrillation and Chronic Coronary Ischemia: A Challenging Vicious Circle. Life (Basel) 2023; 13:1370. [PMID: 37374152 DOI: 10.3390/life13061370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/31/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Atrial fibrillation, the most frequent arrhythmia in clinical practice and chronic coronary syndrome, is one of the forms of coronary ischemia to have a strong dual relationship. Atrial fibrillation may accelerate atherosclerosis and may increase oxygen consumption in the myocardium, creating a mismatch between supply and demand, thus promoting the development or worsening of coronary ischemia. Chronic coronary syndrome alters the structure and function of gap junction proteins, affecting the conduction of action potential and leading to ischemic necrosis of cardiomyocytes and their replacement with fibrous tissue, in this way sustaining the focal ectopic activity in atrial myocardium. They have many risk factors in common, such as hypertension, obesity, type 2 diabetes mellitus, and dyslipidemia. It is vital for the prognosis of patients to break this vicious circle by controlling risk factors, drug therapies, of which antithrombotic therapy may sometimes be challenging in terms of prothrombotic and bleeding risk, and interventional therapies (revascularization and catheter ablation).
Collapse
Affiliation(s)
- Alexandru Florinel Oancea
- Department of Internal Medicine, Faculty of Medicine, University of Medicine and Pharmacy Grigore T. Popa, 700115 Iasi, Romania
- Cardiology Clinic, St. Spiridon Emergency Hospital, 700115 Iasi, Romania
| | - Raul Alexandru Jigoranu
- Department of Internal Medicine, Faculty of Medicine, University of Medicine and Pharmacy Grigore T. Popa, 700115 Iasi, Romania
- Cardiology Clinic, St. Spiridon Emergency Hospital, 700115 Iasi, Romania
| | - Paula Cristina Morariu
- Department of Internal Medicine, Faculty of Medicine, University of Medicine and Pharmacy Grigore T. Popa, 700115 Iasi, Romania
- Internal Medicine Clinic, St. Spiridon Emergency Hospital, 700115 Iasi, Romania
| | - Radu-Stefan Miftode
- Department of Internal Medicine, Faculty of Medicine, University of Medicine and Pharmacy Grigore T. Popa, 700115 Iasi, Romania
- Cardiology Clinic, St. Spiridon Emergency Hospital, 700115 Iasi, Romania
| | - Bogdan Andrei Trandabat
- Department of Internal Medicine, Faculty of Medicine, University of Medicine and Pharmacy Grigore T. Popa, 700115 Iasi, Romania
- Cardiology Clinic, St. Spiridon Emergency Hospital, 700115 Iasi, Romania
| | - Diana Elena Iov
- Department of Internal Medicine, Faculty of Medicine, University of Medicine and Pharmacy Grigore T. Popa, 700115 Iasi, Romania
- Internal Medicine Clinic, St. Spiridon Emergency Hospital, 700115 Iasi, Romania
| | - Elena Cojocaru
- Department of Internal Medicine, Faculty of Medicine, University of Medicine and Pharmacy Grigore T. Popa, 700115 Iasi, Romania
- Department of Morphofunctional Sciences-Pathology, Pediatric Hospital, 700115 Iasi, Romania
| | - Irina Iuliana Costache
- Department of Internal Medicine, Faculty of Medicine, University of Medicine and Pharmacy Grigore T. Popa, 700115 Iasi, Romania
- Cardiology Clinic, St. Spiridon Emergency Hospital, 700115 Iasi, Romania
| | - Livia Genoveva Baroi
- Department of Internal Medicine, Faculty of Medicine, University of Medicine and Pharmacy Grigore T. Popa, 700115 Iasi, Romania
- Surgery Clinic, St. Spiridon Emergency Hospital, 700115 Iasi, Romania
| | - Daniel Vasile Timofte
- Department of Internal Medicine, Faculty of Medicine, University of Medicine and Pharmacy Grigore T. Popa, 700115 Iasi, Romania
- Surgery Clinic, St. Spiridon Emergency Hospital, 700115 Iasi, Romania
| | - Daniela Maria Tanase
- Department of Internal Medicine, Faculty of Medicine, University of Medicine and Pharmacy Grigore T. Popa, 700115 Iasi, Romania
- Internal Medicine Clinic, St. Spiridon Emergency Hospital, 700115 Iasi, Romania
| | - Mariana Floria
- Department of Internal Medicine, Faculty of Medicine, University of Medicine and Pharmacy Grigore T. Popa, 700115 Iasi, Romania
- Internal Medicine Clinic, St. Spiridon Emergency Hospital, 700115 Iasi, Romania
| |
Collapse
|
6
|
Ernault AC, Kawasaki M, Fabrizi B, Montañés-Agudo P, Amersfoorth SCM, Al-Shama RFM, Coronel R, De Groot JR. Knockdown of Ift88 in fibroblasts causes extracellular matrix remodeling and decreases conduction velocity in cardiomyocyte monolayers. Front Physiol 2022; 13:1057200. [DOI: 10.3389/fphys.2022.1057200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 11/07/2022] [Indexed: 11/18/2022] Open
Abstract
Background: Atrial fibrosis plays an important role in the development and persistence of atrial fibrillation by promoting reentry. Primary cilia have been identified as a regulator of fibroblasts (FB) activation and extracellular matrix (ECM) deposition. We hypothesized that selective reduction of primary cilia causes increased fibrosis and facilitates reentry.Aim: The aim of this study was to disrupt the formation of primary cilia in FB and examine its consequences on ECM and conduction in a co-culture system of cardiomyocytes (CM) and FB.Materials: Using short interfering RNA (siRNA), we removed primary cilia in neonatal rat ventricular FB by reducing the expression of Ift88 gene required for ciliary assembly. We co-cultured neonatal rat ventricular cardiomyocytes (CM) with FB previously transfected with Ift88 siRNA (siIft88) or negative control siRNA (siNC) for 48 h. We examined the consequences of ciliated fibroblasts reduction on conduction and tissue remodeling by performing electrical mapping, microelectrode, and gene expression measurements.Results: Transfection of FB with siIft88 resulted in a significant 60% and 30% reduction of relative Ift88 expression in FB and CM-FB co-cultures, respectively, compared to siNC. Knockdown of Ift88 significantly increased the expression of ECM genes Fn1, Col1a1 and Ctgf by 38%, 30% and 18%, respectively, in comparison to transfection with siNC. Conduction velocity (CV) was significantly decreased in the siIft88 group in comparison to siNC [11.12 ± 4.27 cm/s (n = 10) vs. 17.00 ± 6.20 (n = 10) respectively, p < 0.05]. The fraction of sites with interelectrode activation block was larger in the siIft88 group than in the siNC group (6.59 × 10−2 ± 8.01 × 10−2 vs. 1.18 × 10−2 ± 3.72 × 10−2 respectively, p < 0.05). We documented spontaneous reentrant arrhythmias in two cultures in the siIft88 group and in none of the siNC group. Action potentials were not significantly different between siNC and siIft88 groups.Conclusion: Disruption of cilia formation by siIft88 causes ECM remodeling and conduction abnormalities. Prevention of cilia loss could be a target for prevention of arrhythmias.
Collapse
|
7
|
Sun W, Li H, Wang Z, Li Q, Wen H, Wu Y, Du J. Elevated tissue inhibitor of metalloproteinase-1 along with left atrium hypertrophy predict atrial fibrillation recurrence after catheter ablation. Front Cardiovasc Med 2022; 9:1010443. [PMID: 36386356 PMCID: PMC9663807 DOI: 10.3389/fcvm.2022.1010443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022] Open
Abstract
This study aimed to establish a model that predicts atrial fibrillation (AF) recurrence after catheter ablation using clinical risk factors and biomarkers. We used a prospective cohort study, including 230 consecutive persistent AF patients successfully treated with catheter ablation from January 2019 to December 2020 in our hospital. AF recurrence was followed-up after catheter ablation, and clinical risk factors and biomarkers for AF recurrence were analyzed. AF recurred after radiofrequency ablation in 72 (31%) patients. Multiple multivariate logistic regression analysis demonstrated that tissue inhibitor of metalloproteinase-1 (TIMP-1) and left atrium diameter (LAd) were closely associated with AF recurrence. The prediction model constructed by combining TIMP-1 and LAd effectively predicted AF recurrence. Additionally, the model’s performance discrimination, accuracy, and calibration were confirmed through internal validation using bootstrap resampling (1,000 times). The model showed good fitting (Hosmer–Lemeshow goodness chi-square 3.76138, p = 0.926) and had a superior discrimination ability (the area under the receiver operation characteristic curve0.917; 95% CI 0.882–0.952). The calibration curve showed good agreement between the predicted probability and the actual probability. Moreover, the decision curve analysis (DCA) showed the clinical useful of the nomogram. In conclusion, our predictive model based on serum TIMP-1 and LAd levels could predict AF recurrence after catheter ablation.
Collapse
Affiliation(s)
- Weiping Sun
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, China
| | - Haiwei Li
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Zefeng Wang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Qin Li
- Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, China
| | - Haichu Wen
- Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, China
| | - Yongquan Wu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- *Correspondence: Yongquan Wu,
| | - Jie Du
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, China
- Jie Du,
| |
Collapse
|
8
|
Mining of Potential Biomarkers and Pathway in Valvular Atrial Fibrillation (VAF) via Systematic Screening of Gene Coexpression Network. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:3645402. [PMID: 36226239 PMCID: PMC9550484 DOI: 10.1155/2022/3645402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/06/2022] [Accepted: 09/19/2022] [Indexed: 11/17/2022]
Abstract
Purpose. We apply the bioinformatics method to excavate the potential genes and therapeutic targets associated with valvular atrial fibrillation (VAF). Methods. The downloaded gene expression files from the gene expression omnibus (GEO) included patients with primary severe mitral regurgitation complicated with sinus or atrial fibrillation rhythm. Subsequently, the differential gene expression in left and right atrium was analyzed by R software. Additionally, weighted correlation network analysis (WGCNA), principal component analysis (PCA), and linear model for microarray data (LIMMA) algorithm were used to determine hub genes. Then, Metascape database, DAVID database, and STRING database were used to annotate and visualize the gene ontology (GO) analysis, KEGG pathway enrichment analysis, and PPI network analysis of differentially expressed genes (DEGs). Finally, the TFs and miRNAs were predicted by using online tools, such as PASTAA and miRDB. Results. 20,484 differentially expressed genes related to atrial fibrillation were obtained through the analysis of left and right atrial tissue samples of GSE115574 gene chip, and 1,009 were with statistical significance, including 45 upregulated genes and 964 downregulated genes. And the hub genes implicated in AF of NPC2, ODC1, SNAP29, LAPTM5, ST8SIA5, and FCGR3B were screened. Finally, the main regulators of targeted candidate biomarkers and microRNAs, EIF5A2, HIF1A, ZIC2, ELF1, and STAT2, were found in this study. Conclusion. These hub genes, NPC2, ODC1, SNAP29, LAPTM5, ST8SIA5, and FCGR3B, are important for the development of VAF, and their enrichment pathways and TFs elucidate the involved molecular mechanisms and assist in the validation of drug targets.
Collapse
|
9
|
Circulating Galectin-3 and Aldosterone for Predicting Atrial Fibrillation Recurrence after Radiofrequency Catheter Ablation. Cardiovasc Ther 2022; 2022:6993904. [PMID: 35692374 PMCID: PMC9151002 DOI: 10.1155/2022/6993904] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/22/2022] [Accepted: 05/06/2022] [Indexed: 12/29/2022] Open
Abstract
Background Circulating galectin-3 (Gal-3) and aldosterone (ALD) are involved in fibrosis and inflammation. However, their potential value as predictors of atrial fibrillation (AF) recurrence after radiofrequency catheter ablation (RFCA) is unknown or controversial. Therefore, the aim of this study was to assess the relationship between baseline Gal-3, ALD levels, and AF recurrence in patients performing RFCA. Methods 153 consecutive patients undergoing RFCA were included. Gal-3 and ALD were measured at baseline. Univariate and multivariate Cox regressions were performed to determine the predictors of AF recurrence. Receiver operating characteristic (ROC) curve and Kaplan-Meier (K-M) curve were used to assess the value of predictors. Results There were 35 (22.88%) cases of AF recurrence after RFCA. The recurrence group had significantly higher preoperative serum levels of Gal-3 and ALD than the nonrecurrence group. Univariate and multivariate analysis showed that Gal-3 (HR = 1.28, 95% CI: 1.04-1.56, p = 0.02) and ALD (OR = 1.02, 95% CI: 1.00-1.03, p < 0.03) were significantly associated with AF recurrence after RFCA. The area under the curve (AUC) of preoperative serum Gal-3, ALD, and 2 combined to predict the recurrence of AF patients after RFCA was 0.636, 0.798, and 0.893, respectively, while sensitivity was 65.32%, 71.69%, and 88.61%, respectively and specificity was 77.46%, 78.53%, and 86.0%, respectively. Patients with Gal-3 above the cutoff value of 14.57 pg/ml had higher frequent AF recurrence than the patients with Gal − 3 ≤ 14.57 pg/ml (35% vs. 12%, p < 0.001) during a follow-up. Meanwhile, patients with ALD above the cutoff value of 243.61 pg/ml also had a higher AF recurrence rate than those with ALD ≤ 243.61 pg/ml (37% vs. 11%, p < 0.001) during a follow-up. The recurrence rate in patients with Gal − 3 > 14.57 pg/ml + ALD > 243.61 pg/ml was higher than that in patients with baseline Gal − 3 > 14.57 pg/ml or ALD > 243.61 pg/ml and patients with Gal − 3 ≤ 14.57 pg/ml + ALD ≤ 243.61 pg/ml (57% vs. 14% vs. 9%, p < 0.01, respectively). Conclusion AF recurrence after RFCA had higher baseline Gal-3 and ALD levels, and higher preoperative circulating Gal-3 and ALD levels were independent predictors of AF recurrence for patients undergoing RFCA, while combination of preoperative Gal-3 and ALD levels has higher prediction accuracy.
Collapse
|
10
|
A Review on Atrial Fibrillation (Computer Simulation and Clinical Perspectives). HEARTS 2022. [DOI: 10.3390/hearts3010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Atrial fibrillation (AF), a heart condition, has been a well-researched topic for the past few decades. This multidisciplinary field of study deals with signal processing, finite element analysis, mathematical modeling, optimization, and clinical procedure. This article is focused on a comprehensive review of journal articles published in the field of AF. Topics from the age-old fundamental concepts to specialized modern techniques involved in today’s AF research are discussed. It was found that a lot of research articles have already been published in modeling and simulation of AF. In comparison to that, the diagnosis and post-operative procedures for AF patients have not yet been totally understood or explored by the researchers. The simulation and modeling of AF have been investigated by many researchers in this field. Cellular model, tissue model, and geometric model among others have been used to simulate AF. Due to a very complex nature, the causes of AF have not been fully perceived to date, but the simulated results are validated with real-life patient data. Many algorithms have been proposed to detect the source of AF in human atria. There are many ablation strategies for AF patients, but the search for more efficient ablation strategies is still going on. AF management for patients with different stages of AF has been discussed in the literature as well but is somehow limited mostly to the patients with persistent AF. The authors hope that this study helps to find existing research gaps in the analysis and the diagnosis of AF.
Collapse
|
11
|
van Schie MS, Starreveld R, Roos-Serote MC, Taverne YJHJ, van Schaagen FRN, Bogers AJJC, de Groot NMS. Classification of sinus rhythm single potential morphology in patients with mitral valve disease. Europace 2021; 22:1509-1519. [PMID: 33033830 PMCID: PMC7544534 DOI: 10.1093/europace/euaa130] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/24/2020] [Accepted: 04/28/2020] [Indexed: 12/31/2022] Open
Abstract
Aims The morphology of unipolar single potentials (SPs) contains information on intra-atrial conduction disorders and possibly the substrate underlying atrial fibrillation (AF). This study examined the impact of AF episodes on features of SP morphology during sinus rhythm (SR) in patients with mitral valve disease. Methods and results Intraoperative epicardial mapping (interelectrode distance 2 mm) of the right and left atrium (RA, LA), Bachmann’s bundle (BB), and pulmonary vein area (PVA) was performed in 67 patients (27 male, 67 ± 11 years) with or without a history of paroxysmal AF (PAF). Unipolar SPs were classified according to their differences in relative R- and S-wave amplitude ratios. A clear predominance of S-waves was observed at BB and the RA in both the no AF and PAF groups (BB 88.8% vs. 85.9%, RA 92.1% vs. 85.1%, respectively). Potential voltages at the RA, BB, and PVA were significantly lower in the PAF group (P < 0.001 for each) and were mainly determined by the size of the S-waves amplitudes. The largest difference in S-wave amplitudes was found at BB; the S-wave amplitude was lower in the PAF group [4.08 (2.45–6.13) mV vs. 2.94 (1.40–4.75) mV; P < 0.001]. In addition, conduction velocity (CV) at BB was lower as well [0.97 (0.70–1.21) m/s vs. 0.89 (0.62–1.16) m/s, P < 0.001]. Conclusion Though excitation of the atria during SR is heterogeneously disrupted, a history of AF is characterized by decreased SP amplitudes at BB due to loss of S-wave amplitudes and decreased CV. This suggests that SP morphology could provide additional information on wavefront propagation.
Collapse
Affiliation(s)
- Mathijs S van Schie
- Department of Cardiology, Unit Translational Electrophysiology, Erasmus Medical Centre, Dr Molewaterplein 40, 3015GD Rotterdam, the Netherlands
| | - Roeliene Starreveld
- Department of Cardiology, Unit Translational Electrophysiology, Erasmus Medical Centre, Dr Molewaterplein 40, 3015GD Rotterdam, the Netherlands
| | - Maarten C Roos-Serote
- Department of Cardiology, Unit Translational Electrophysiology, Erasmus Medical Centre, Dr Molewaterplein 40, 3015GD Rotterdam, the Netherlands
| | - Yannick J H J Taverne
- Department of Cardiothoracic Surgery, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Frank R N van Schaagen
- Department of Cardiothoracic Surgery, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Ad J J C Bogers
- Department of Cardiothoracic Surgery, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Natasja M S de Groot
- Department of Cardiology, Unit Translational Electrophysiology, Erasmus Medical Centre, Dr Molewaterplein 40, 3015GD Rotterdam, the Netherlands
| |
Collapse
|
12
|
Clerx M, Mirams GR, Rogers AJ, Narayan SM, Giles WR. Immediate and Delayed Response of Simulated Human Atrial Myocytes to Clinically-Relevant Hypokalemia. Front Physiol 2021; 12:651162. [PMID: 34122128 PMCID: PMC8188899 DOI: 10.3389/fphys.2021.651162] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/22/2021] [Indexed: 12/18/2022] Open
Abstract
Although plasma electrolyte levels are quickly and precisely regulated in the mammalian cardiovascular system, even small transient changes in K+, Na+, Ca2+, and/or Mg2+ can significantly alter physiological responses in the heart, blood vessels, and intrinsic (intracardiac) autonomic nervous system. We have used mathematical models of the human atrial action potential (AP) to explore the electrophysiological mechanisms that underlie changes in resting potential (Vr) and the AP following decreases in plasma K+, [K+]o, that were selected to mimic clinical hypokalemia. Such changes may be associated with arrhythmias and are commonly encountered in patients (i) in therapy for hypertension and heart failure; (ii) undergoing renal dialysis; (iii) with any disease with acid-base imbalance; or (iv) post-operatively. Our study emphasizes clinically-relevant hypokalemic conditions, corresponding to [K+]o reductions of approximately 1.5 mM from the normal value of 4 to 4.5 mM. We show how the resulting electrophysiological responses in human atrial myocytes progress within two distinct time frames: (i) Immediately after [K+]o is reduced, the K+-sensing mechanism of the background inward rectifier current (IK1) responds. Specifically, its highly non-linear current-voltage relationship changes significantly as judged by the voltage dependence of its region of outward current. This rapidly alters, and sometimes even depolarizes, Vr and can also markedly prolong the final repolarization phase of the AP, thus modulating excitability and refractoriness. (ii) A second much slower electrophysiological response (developing 5-10 minutes after [K+]o is reduced) results from alterations in the intracellular electrolyte balance. A progressive shift in intracellular [Na+]i causes a change in the outward electrogenic current generated by the Na+/K+ pump, thereby modifying Vr and AP repolarization and changing the human atrial electrophysiological substrate. In this study, these two effects were investigated quantitatively, using seven published models of the human atrial AP. This highlighted the important role of IK1 rectification when analyzing both the mechanisms by which [K+]o regulates Vr and how the AP waveform may contribute to "trigger" mechanisms within the proarrhythmic substrate. Our simulations complement and extend previous studies aimed at understanding key factors by which decreases in [K+]o can produce effects that are known to promote atrial arrhythmias in human hearts.
Collapse
Affiliation(s)
- Michael Clerx
- Centre for Mathematical Medicine and Biology, School of Mathematical Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Gary R Mirams
- Centre for Mathematical Medicine and Biology, School of Mathematical Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Albert J Rogers
- Department of Medicine and Cardiovascular Institute, Stanford University, Stanford, CA, United States
| | - Sanjiv M Narayan
- Department of Medicine and Cardiovascular Institute, Stanford University, Stanford, CA, United States
| | - Wayne R Giles
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
13
|
Choi W, Kim HJ, Park SY, Park SJ, Kim JB, Jung SH, Lee JW. The Impact of Left Atrial Reduction During Surgical Ablation of Atrial Fibrillation. Semin Thorac Cardiovasc Surg 2021; 34:537-546. [PMID: 33713828 DOI: 10.1053/j.semtcvs.2021.03.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 03/04/2021] [Indexed: 11/11/2022]
Abstract
Enlarged left atrium (LA) is a risk factor for ablation failure after atrial fibrillation (AF) surgery. It predisposes patients to thromboembolic events, even in successful ablation; therefore, concomitant resection of the LA wall during surgical ablation was introduced. This study examined the clinical impacts of LA reduction in patients undergoing concomitant ablation for AF. This study enrolled 1484 patients with enlarged LA (≥50 mm) who underwent surgical AF ablation during major cardiac surgery between January 2001 and August 2018. Among them, 876 (59%) patients underwent concomitant LA reduction (Reduction group), whereas in the remaining 608 (41%), the LA wall was unresected (Preservation group). The primary outcome of interest was overall stroke. The secondary outcomes were overall mortality, late recurrence of AF, early postoperative complications and postoperative echocardiographic parameters. Outcomes were compared after adjusting baseline characteristics with inverse probability of treatment weighting (IPTW) using propensity score. The median follow-up was 60.1 months. After IPTW adjustment, long-term mortality (P = 0.250) and AF-free rates (P = 0.196) did not significantly differ between groups. However, the Reduction group showed a decreased risk of stroke (hazard ratio 0.54; 95% confidence interval 0.32-0.90; P = 0.018). Early postoperative complications rate such as mortality or reoperation for bleeding, was not significantly different between the 2 groups. The Reduction group showed smaller LA diameter (50.6 ± 8.0 mm vs 53.6 ± 8.9 mm; P < 0.001) on follow-up echocardiography. LA reduction effectively decreased LA size and appeared to decrease the stroke risk in patients with enlarged LA undergoing ablation for AF.
Collapse
Affiliation(s)
- Wooseok Choi
- Department of Thoracic and Cardiovascular Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Ho Jin Kim
- Department of Thoracic and Cardiovascular Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seo Young Park
- Department of Clinical Epidemiology and Biostatistics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seong Jun Park
- Department of Thoracic and Cardiovascular Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Joon Bum Kim
- Department of Thoracic and Cardiovascular Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Sung-Ho Jung
- Department of Thoracic and Cardiovascular Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jae Won Lee
- Department of Thoracic and Cardiovascular Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
14
|
Matteucci F, Maesen B, De Asmundis C, Parise G, Micali LR, Tuijthof G, Gerits P, Vernooy K, Maessen JG, La Meir M, Gelsomino S. New Biparietal Bipolar Catheter Prototype for Hybrid Atrial Fibrillation Ablation. INNOVATIONS-TECHNOLOGY AND TECHNIQUES IN CARDIOTHORACIC AND VASCULAR SURGERY 2021; 16:181-187. [PMID: 33410714 PMCID: PMC8108111 DOI: 10.1177/1556984520981025] [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] [Indexed: 12/04/2022]
Abstract
Objective To evaluate the size and depth of linear lesions by in vitro testing with a custom-made radio frequency biparietal bipolar ablation catheter in a single-stage setting. Methods A custom-made catheter was created to generate linear lesions around the left atrium and pulmonary veins of an ex vivo pig. Two frames were made, 1 epicardial and 1 endocardial. A continuous copper braid electrode and an alignment system consisting of 2 parallel rows of neodymium magnets were embedded in a flexible plastic support. After 24 hours of formalin conservation, samples of the left atrium of a freshly slaughtered pig were sliced in a cryotome, thus obtaining a sequence of 100-µm thick layers extending from the endocardial to the epicardial side. After being digitized through a scanner, these layers were evaluated using morphometric computer software. For each slice, we evaluated the maximum length of the lesions, the maximum epicardial length, the maximum endocardial length, the total area of the lesion, and the total volume. Results Forty transmural lesions from 40 specimens were obtained. The results were the following (the number in parenthesis is the interquartile range in mm): lesion maximum length (LMAX) was 7.297 mm (0.006), epicardial maximum length (LEPI) was 7.291 mm (0.014), and endocardial maximum length was 7.291 mm (0.018). The total area and total volume were 1018.50 ± 36.51 mm2 and 101.85 ± 3.65 mm3, respectively. Conclusions Our prototype showed very promising results. The next step will be to enhance the design for clinical application.
Collapse
Affiliation(s)
- Francesco Matteucci
- 118066 Cardiovascular Research Institute Maastricht - CARIM, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Bart Maesen
- 118066 Cardiovascular Research Institute Maastricht - CARIM, Maastricht University Medical Center, Maastricht, The Netherlands.,5211 Cardiothoracic Department, Maastricht University Hospital, Maastricht, The Netherlands
| | - Carlo De Asmundis
- 60201 Cardiothoracic Department, Brussels University Hospital, Brussels, Belgium
| | - Gianmarco Parise
- 118066 Cardiovascular Research Institute Maastricht - CARIM, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Linda Renata Micali
- 118066 Cardiovascular Research Institute Maastricht - CARIM, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Gabrielle Tuijthof
- 5211 IDEE Engineering, Maastricht University, Maastricht, The Netherlands
| | - Peter Gerits
- 118066 Maastricht Instruments BV, Maastricht, The Netherlands
| | - Kevin Vernooy
- 5211 Cardiothoracic Department, Maastricht University Hospital, Maastricht, The Netherlands
| | - Jos G Maessen
- 118066 Cardiovascular Research Institute Maastricht - CARIM, Maastricht University Medical Center, Maastricht, The Netherlands.,5211 Cardiothoracic Department, Maastricht University Hospital, Maastricht, The Netherlands
| | - Mark La Meir
- 118066 Cardiovascular Research Institute Maastricht - CARIM, Maastricht University Medical Center, Maastricht, The Netherlands.,60201 Cardiothoracic Department, Brussels University Hospital, Brussels, Belgium
| | - Sandro Gelsomino
- 118066 Cardiovascular Research Institute Maastricht - CARIM, Maastricht University Medical Center, Maastricht, The Netherlands.,60201 Cardiothoracic Department, Brussels University Hospital, Brussels, Belgium
| |
Collapse
|
15
|
Kim YM, Lakin R, Zhang H, Liu J, Sachedina A, Singh M, Wilson E, Perez M, Verma S, Quertermous T, Olgin J, Backx PH, Ashley EA. Apelin increases atrial conduction velocity, refractoriness, and prevents inducibility of atrial fibrillation. JCI Insight 2020; 5:126525. [PMID: 32879139 PMCID: PMC7526452 DOI: 10.1172/jci.insight.126525] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 07/22/2020] [Indexed: 11/17/2022] Open
Abstract
Previous studies have shown an association between elevated atrial NADPH-dependent oxidative stress and decreased plasma apelin in patients with atrial fibrillation (AF), though the basis for this relationship is unclear. In the current study, RT-PCR and immunofluorescence studies of human right atrial appendages (RAAs) showed expression of the apelin receptor, APJ, and reduced apelin content in the atria, but not in plasma, of patients with AF versus normal sinus rhythm. Disruption of the apelin gene in mice increased (2.4-fold) NADPH-stimulated superoxide levels and slowed atrial conduction velocities in optical mapping of a Langendorff-perfused isolated heart model, suggesting that apelin levels may influence AF vulnerability. Indeed, in mice with increased AF vulnerability (induced by chronic intense exercise), apelin administration reduced the incidence and duration of induced atrial arrhythmias in association with prolonged atrial refractory periods. Moreover, apelin decreased AF induction in isolated atria from exercised mice while accelerating conduction velocity and increasing action potential durations. At the cellular level, these changes were associated with increased atrial cardiomyocyte sodium currents. These findings support the conclusion that reduced atrial apelin is maladaptive in fibrillating human atrial myocardium and that increasing apelin bioavailability may be a worthwhile therapeutic strategy for treating and preventing AF.
Collapse
Affiliation(s)
- Young M Kim
- Division of Cardiovascular Medicine, Stanford Medicine, Stanford, California, USA
| | - Robert Lakin
- Department of Biology, York University, Toronto, Ontario, Canada.,Division of Cardiology, University Health Network, Toronto, Ontario, Canada
| | - Hao Zhang
- Division of Cardiovascular Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Jack Liu
- Division of Cardiology, University Health Network, Toronto, Ontario, Canada
| | - Ayaaz Sachedina
- Division of Cardiology, University Health Network, Toronto, Ontario, Canada
| | - Maneesh Singh
- Division of Cardiovascular Medicine, Stanford Medicine, Stanford, California, USA
| | - Emily Wilson
- Division of Cardiovascular Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Marco Perez
- Division of Cardiovascular Medicine, Stanford Medicine, Stanford, California, USA
| | - Subodh Verma
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Thomas Quertermous
- Division of Cardiovascular Medicine, Stanford Medicine, Stanford, California, USA
| | - Jeffrey Olgin
- Division of Cardiovascular Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Peter H Backx
- Department of Biology, York University, Toronto, Ontario, Canada.,Division of Cardiology, University Health Network, Toronto, Ontario, Canada
| | - Euan A Ashley
- Division of Cardiovascular Medicine, Stanford Medicine, Stanford, California, USA
| |
Collapse
|
16
|
Xiao M, Zhang M, Bie M, Wang X, Guo J, Xiao H. Galectin-3 Induces Atrial Fibrosis by Activating the TGF-β1/Smad Pathway in Patients with Atrial Fibrillation. Cardiology 2020; 145:446-455. [PMID: 32516780 DOI: 10.1159/000506072] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 01/01/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND Atrial fibrosis plays a critical role in the occurrence and maintenance of atrial fibrillation. The role of TGF-β1 in mediating atrial fibrosis is well documented. The β-galactoside-binding lectin galectin-3 (Gal-3) is mainly produced by macrophages in biological events such as inflammation and angiogenesis. Previous studies have shown that Gal-3 is associated with atrial fibrosis, but the relationship between TGF-β1 and Gal-3 in atrial fibrosis remains unclear. OBJECTIVE To determine whether Gal-3 induces atrial fibrosis and atrial fibrillation by activating the TGF-β1/Smad pathway and whether the expression of Gal-3 is mediated by TGF-β1, which can enable assessing the relationship between Gal-3 and TGF-β1 in atrial fibrosis. METHODS In this study, 30 patients' right atrial appendages were collected and divided into 3 groups: congenital heart disease sinus rhythm group (n = 10, as a control group), rheumatic heart disease sinus rhythm group (n = 10), and rheumatic heart disease atrial fibrillation group (n = 10). Rat atrial fibroblasts were cultured in vitro, and recombinant Gal-3 and recombinant TGF-β1 proteins were added to the cell culture. The expression of Gal-3, TGF-β1, Smad2, and collagen I was detected by Western blotting and quantitative real-time PCR. Atrial tissues were stained with Masson's trichrome stain to evaluate the extent of atrial fibrosis. The expression of Gal-3 and TGF-β1 was detected by immunohistochemical staining and immunofluorescence staining. Gal-3 and TGF-β1 interaction was demonstrated by immunoprecipitation. RESULTS The expression levels of Gal-3, TGF-β1, Smad2, and collagen I were elevated in the rheumatic heart disease atrial fibrillation group compared with the congenital heart disease sinus rhythm group and the rheumatic heart disease sinus rhythm group. In cultured atrial fibroblasts, there is a synergistic interaction between Gal-3 and TGF-β1. Gal-3 stimulated the TGF-β1/Smad pathway, and overexpression of TGF-β1 induced Gal-3 expression. CONCLUSIONS Gal-3 and TGF-β1 interact with each other and stimulate the downstream TGF-β1/Smad pathway. This finding suggests that Gal-3 could be an important factor in TGF-β1-induced fibrosis in atrial fibrillation.
Collapse
Affiliation(s)
- Minghan Xiao
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Meixia Zhang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mengjun Bie
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaowen Wang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jingwen Guo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hua Xiao
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China,
| |
Collapse
|
17
|
Potential causal association of a prolonged PR interval and clinical recurrence of atrial fibrillation after catheter ablation: a Mendelian randomization analysis. J Hum Genet 2020; 65:813-821. [PMID: 32409696 DOI: 10.1038/s10038-020-0774-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 04/26/2020] [Accepted: 04/30/2020] [Indexed: 11/08/2022]
Abstract
A prolonged PR interval predicts atrial fibrillation (AF) recurrence after catheter ablation. We investigated the causal association between the PR interval and AF clinical recurrence by a Mendelian randomization. We prospectively included 1722 patients with AF (73.2% male, 58.6 ± 10.8 years old, 71.3% paroxysmal AF) who underwent catheter ablation into a genome-wide association study (GWAS). We searched for the genetic associations between the PR interval and AF recurrence by analyzing 44 single nucleotide polymorphisms (SNPs) already known to be associated with the PR interval, and investigated the Mendelian randomization. Based on the quartile analysis, the highest quartile of the PR interval was associated with an increased risk of AF recurrence compared with the lowest quartile (Hazard ratio (HR) = 1.91, 95% CI = 1.51-2.42, P = 8.41 × 10-8) during 35.7 ± 28.5 months of follow-up. Among 44 SNPs known to be associated with the PR interval, two SNPs had significant associations with the PR interval (P < 0.001 for each SNP). CAV1 (HR = 1.15, 95% CI = 1.02-1.31, P = 0.024) was associated with clinical recurrence of AF. A Mendelian randomization analysis demonstrated a significant association with CAV1 (HR = 1.04, 95% CI = 1.01-1.07, P = 0.006). A prolonged PR interval was a risk factor for an AF recurrence, and the PR interval had a potentially causal association with an AF clinical recurrence after catheter ablation at the genetic level.
Collapse
|
18
|
Pranata R, Vania R, Raharjo SB. Efficacy and safety of renal denervation in addition to pulmonary vein isolation for atrial fibrillation and hypertension-Systematic review and meta-analysis of randomized controlled trials. J Arrhythm 2020; 36:386-394. [PMID: 32528562 PMCID: PMC7279983 DOI: 10.1002/joa3.12353] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 04/02/2020] [Accepted: 04/10/2020] [Indexed: 12/23/2022] Open
Abstract
Introduction This systematic review and meta‐analysis aimed to assess the latest evidence on the use of renal denervation (RDN) + pulmonary vein isolation (PVI) compared to PVI alone for treating atrial fibrillation (AF) with hypertension. Methods A systematic literature search from several electronic databases was performed up until January 2020. The primary outcome was AF recurrence defined as AF/atrial flutter (AFL)/atrial tachycardia (AT) ≥30 seconds at 12‐month follow‐up and the secondary outcome was procedure‐related complications. Results There were 568 subjects from five studies. AF recurrence was 90/280 (32.1%) in the RDN + PVI group and 142/274 (51.8%) in the PVI group. RDN + PVI was associated with a lower incidence of AF recurrence (RR 0.62 [0.51, 076], P < .001; I2: 0%). Pooled analysis of HR showed that RDN + PVI was associated with reduced AF recurrence (HR 0.51 [0.38, 0.70], P < .001; I2: 0%). Complications were 7/241 (2.9%) in the RDN + PVI group and 8/237 (3.4%) in the PVI group. The rate of complications between the groups was similar (RR 0.87 [0.33, 2.29], P = .77; I2: 0%). In the subgroup analysis of paroxysmal AF, RDN + PVI was shown to reduce AF recurrence (RR 0.64 [0.49, 0.82], P < .001; I2: 0% and HR 0.56 [0.38, 0.82], P = .003; I2: 0%) compared to PVI alone. RDN + PVI has a moderate certainty of evidence in the reducing AF recurrence with an absolute reduction of 197 fewer per 1000 (from 254 fewer to 124 fewer). Conclusion RDN in addition to PVI, is associated with reduced 12‐month AF recurrence and similar procedure‐related complications compared to PVI alone.
Collapse
Affiliation(s)
- Raymond Pranata
- Faculty of MedicineUniversitas Pelita HarapanTangerangIndonesia
| | - Rachel Vania
- Faculty of MedicineUniversitas Pelita HarapanTangerangIndonesia
| | - Sunu Budhi Raharjo
- Department of Cardiology and Vascular MedicineFaculty of Medicine Universitas IndonesiaNational Cardiovascular Center Harapan KitaJakartaIndonesia
| |
Collapse
|
19
|
Coveney S, Clayton RH. Sensitivity and Uncertainty Analysis of Two Human Atrial Cardiac Cell Models Using Gaussian Process Emulators. Front Physiol 2020; 11:364. [PMID: 32390867 PMCID: PMC7191317 DOI: 10.3389/fphys.2020.00364] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 03/30/2020] [Indexed: 12/20/2022] Open
Abstract
Biophysically detailed cardiac cell models reconstruct the action potential and calcium dynamics of cardiac myocytes. They aim to capture the biophysics of current flow through ion channels, pumps, and exchangers in the cell membrane, and are highly detailed. However, the relationship between model parameters and model outputs is difficult to establish because the models are both complex and non-linear. The consequences of uncertainty and variability in model parameters are therefore difficult to determine without undertaking large numbers of model evaluations. The aim of the present study was to demonstrate how sensitivity and uncertainty analysis using Gaussian process emulators can be used for a systematic and quantitive analysis of biophysically detailed cardiac cell models. We selected the Courtemanche and Maleckar models of the human atrial action potential for analysis because these models describe a similar set of currents, with different formulations. In our approach Gaussian processes emulate the main features of the action potential and calcium transient. The emulators were trained with a set of design data comprising samples from parameter space and corresponding model outputs, initially obtained from 300 model evaluations. Variance based sensitivity indices were calculated using the emulators, and first order and total effect indices were calculated for each combination of parameter and output. The differences between the first order and total effect indices indicated that the effect of interactions between parameters was small. A second set of emulators were then trained using a new set of design data with a subset of the model parameters with a sensitivity index of more than 0.1 (10%). This second stage analysis enabled comparison of mechanisms in the two models. The second stage sensitivity indices enabled the relationship between the L-type Ca 2+ current and the action potential plateau to be quantified in each model. Our quantitative analysis predicted that changes in maximum conductance of the ultra-rapid K + channel I Kur would have opposite effects on action potential duration in the two models, and this prediction was confirmed by additional simulations. This study has demonstrated that Gaussian process emulators are an effective tool for sensitivity and uncertainty analysis of biophysically detailed cardiac cell models.
Collapse
Affiliation(s)
| | - Richard H. Clayton
- Insigneo Institute for in-silico Medicine and Department of Computer Science, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
20
|
Bai J, Lu Y, Lo A, Zhao J, Zhang H. PITX2 upregulation increases the risk of chronic atrial fibrillation in a dose-dependent manner by modulating IKs and ICaL -insights from human atrial modelling. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:191. [PMID: 32309338 PMCID: PMC7154416 DOI: 10.21037/atm.2020.01.90] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Functional analysis has shown that the paired-like homeodomain transcription factor 2 (PITX2) overexpression associated with atrial fibrillation (AF) leads to the slow delayed rectifier K+ current (IKs) increase and the L-type Ca2+ current (ICaL) reduction observed in isolated right atrial myocytes from chronic AF (CAF) patients. Through multiscale computational models, this study aimed to investigate the functional impact of the PITX2 overexpression on atrial electrical activity. Methods The well-known Courtemanche-Ramirez-Nattel (CRN) model of human atrial action potentials (APs) was updated to incorporate experimental data on alterations in IKs and ICaL due to the PITX2 overexpression. These cell models for sinus rhythm (SR) and CAF were then incorporated into homogeneous multicellular one-dimensional (1D), two-dimensional (2D), and three-dimensional (3D) tissue models. The proarrhythmic effects of the PITX2 overexpression were quantified with ion current profiles, AP morphology, AP duration (APD) restitution, conduction velocity restitution (CVR), wavelength (WL), vulnerable window (VW) for unidirectional conduction block, and minimal substrate size required to induce re-entry. Dynamic behaviors of spiral waves were characterized by measuring lifespan (LS), tip patterns and dominant frequencies. Results The IKs increase and the ICaL decrease arising from the PITX2 overexpression abbreviated APD and flattened APD restitution (APDR) curves in single cells. It reduced WL and increased CV at high excitation rates at the 1D tissue level. Although it had no effects on VW for initiating spiral waves, it decreased the minimal substrate size necessary to sustain re-entry. It also stabilized and accelerated spiral waves in 2D and 3D tissue models. Conclusions Electrical remodeling (IKs and ICaL) due to the PITX2 overexpression increases susceptibility to AF due to increased tissue vulnerability, abbreviated APD, shortened WL and altered CV, which, in combination, facilitate initiation and maintenance of spiral waves.
Collapse
Affiliation(s)
- Jieyun Bai
- Department of Electronic Engineering, College of Information Science and Technology, Jinan University, Guangzhou 510632, China
| | - Yaosheng Lu
- Department of Electronic Engineering, College of Information Science and Technology, Jinan University, Guangzhou 510632, China
| | - Andy Lo
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Jichao Zhao
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Henggui Zhang
- Biological Physics Group, School of Physics & Astronomy, University of Manchester, Manchester, UK
| |
Collapse
|
21
|
Abstract
Determining optimal treatment strategies for complex arrhythmogenesis in AF is confounded by the lack of consensus regarding the mechanisms causing AF. Studies report different mechanisms for AF, ranging from hierarchical drivers to anarchical multiple activation wavelets. Differences in the assessment of AF mechanisms are likely due to AF being recorded across diverse models using different investigational tools, spatial scales and clinical populations. The authors review different AF mechanisms, including anatomical and functional re-entry, hierarchical drivers and anarchical multiple wavelets. They then describe different cardiac mapping techniques and analysis tools, including activation mapping, phase mapping and fibrosis identification. They explain and review different data challenges, including differences between recording devices in spatial and temporal resolutions, spatial coverage and recording surface, and report clinical outcomes using different data modalities. They suggest future research directions for investigating the mechanisms underlying human AF.
Collapse
Affiliation(s)
- Caroline H Roney
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK.,Imperial Centre for Cardiac Engineering, Imperial College London, London, UK
| | - Andrew L Wit
- Imperial Centre for Cardiac Engineering, Imperial College London, London, UK.,Department of Pharmacology, Columbia University College of Physicians and Surgeons, New York, NY, US
| | - Nicholas S Peters
- Imperial Centre for Cardiac Engineering, Imperial College London, London, UK.,National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
22
|
Sánchez J, Gomez JF, Martinez-Mateu L, Romero L, Saiz J, Trenor B. Heterogeneous Effects of Fibroblast-Myocyte Coupling in Different Regions of the Human Atria Under Conditions of Atrial Fibrillation. Front Physiol 2019; 10:847. [PMID: 31333496 PMCID: PMC6620707 DOI: 10.3389/fphys.2019.00847] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 06/19/2019] [Indexed: 12/19/2022] Open
Abstract
Background: Atrial fibrillation (AF), the most common cardiac arrhythmia, is characterized by alteration of the action potential (AP) propagation. Under persistent AF, myocytes undergo electrophysiological and structural remodeling, which involves fibroblast proliferation and differentiation, modifying the substrate for AP propagation. The aim of this study was to analyze the effects on the AP of fibroblast-myocyte coupling during AF and its propagation in different regions of the atria. Methods: Isolated myocytes were coupled to different numbers of fibroblasts using the established AP models and tissue simulations were performed by randomly distributing fibroblasts. Fibroblast formulations were updated to match recent experimental data. Major ion current conductances of the myocyte model were modified to simulate AP heterogeneity in four different atrial regions (right atrium posterior wall, crista terminalis, left atrium posterior wall, and pulmonary vein) according to experimental and computational studies. Results: The results of the coupled myocyte-fibroblast simulations suggest that a more depolarized membrane potential and higher fibroblast membrane capacitance have a greater impact on AP duration and myocyte maximum depolarization velocity. The number of coupled fibroblasts and the stimulation frequency are determining factors in altering myocyte AP. Strand simulations show that conduction velocity tends to homogenize in all regions, while the left atrium is more likely to be affected by fibroblast and AP propagation block is more likely to occur. The pulmonary vein is the most affected region, even at low fibroblast densities. In 2D sheets with randomly placed fibroblasts, wavebreaks are observed in the low density (10%) central fibrotic zone and when fibroblast density increases (40%) propagation in the fibrotic region is practically blocked. At densities of 10 and 20% the width of the vulnerable window increases with respect to control but is decreased at 40%. Conclusion: Myocyte-fibroblast coupling characteristics heterogeneously affect AP propagation and features in the different atrial zones, and myocytes from the left atria are more sensitive to fibroblast coupling.
Collapse
Affiliation(s)
- Jorge Sánchez
- Centre for Research and Innovation in Bioengineering, Universitat Politècnica de València, Valencia, Spain
| | - Juan F Gomez
- Centre for Research and Innovation in Bioengineering, Universitat Politècnica de València, Valencia, Spain
| | - Laura Martinez-Mateu
- Centre for Research and Innovation in Bioengineering, Universitat Politècnica de València, Valencia, Spain
| | - Lucia Romero
- Centre for Research and Innovation in Bioengineering, Universitat Politècnica de València, Valencia, Spain
| | - Javier Saiz
- Centre for Research and Innovation in Bioengineering, Universitat Politècnica de València, Valencia, Spain
| | - Beatriz Trenor
- Centre for Research and Innovation in Bioengineering, Universitat Politècnica de València, Valencia, Spain
| |
Collapse
|
23
|
Filos D, Tachmatzidis D, Maglaveras N, Vassilikos V, Chouvarda I. Understanding the Beat-to-Beat Variations of P-Waves Morphologies in AF Patients During Sinus Rhythm: A Scoping Review of the Atrial Simulation Studies. Front Physiol 2019; 10:742. [PMID: 31275161 PMCID: PMC6591370 DOI: 10.3389/fphys.2019.00742] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 05/28/2019] [Indexed: 11/13/2022] Open
Abstract
The remarkable advances in high-performance computing and the resulting increase of the computational power have the potential to leverage computational cardiology toward improving our understanding of the pathophysiological mechanisms of arrhythmias, such as Atrial Fibrillation (AF). In AF, a complex interaction between various triggers and the atrial substrate is considered to be the leading cause of AF initiation and perpetuation. In electrocardiography (ECG), P-wave is supposed to reflect atrial depolarization. It has been found that even during sinus rhythm (SR), multiple P-wave morphologies are present in AF patients with a history of AF, suggesting a higher dispersion of the conduction route in this population. In this scoping review, we focused on the mechanisms which modify the electrical substrate of the atria in AF patients, while investigating the existence of computational models that simulate the propagation of the electrical signal through different routes. The adopted review methodology is based on a structured analytical framework which includes the extraction of the keywords based on an initial limited bibliographic search, the extensive literature search and finally the identification of relevant articles based on the reference list of the studies. The leading mechanisms identified were classified according to their scale, spanning from mechanisms in the cell, tissue or organ level, and the produced outputs. The computational modeling approaches for each of the factors that influence the initiation and the perpetuation of AF are presented here to provide a clear overview of the existing literature. Several levels of categorization were adopted while the studies which aim to translate their findings to ECG phenotyping are highlighted. The results denote the availability of multiple models, which are appropriate under specific conditions. However, the consideration of complex scenarios taking into account multiple spatiotemporal scales, personalization of electrophysiological and anatomical models and the reproducibility in terms of ECG phenotyping has only partially been tackled so far.
Collapse
Affiliation(s)
- Dimitrios Filos
- Lab of Computing, Medical Informatics and Biomedical Imaging Technologies, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Nicos Maglaveras
- Lab of Computing, Medical Informatics and Biomedical Imaging Technologies, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Department of Industrial Engineering and Management Sciences, Northwestern University, Evanston, IL, United States
| | - Vassilios Vassilikos
- 3rd Cardiology Department, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ioanna Chouvarda
- Lab of Computing, Medical Informatics and Biomedical Imaging Technologies, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
24
|
Menon A, Hong L, Savio-Galimberti E, Sridhar A, Youn SW, Zhang M, Kor K, Blair M, Kupershmidt S, Darbar D. Electrophysiologic and molecular mechanisms of a frameshift NPPA mutation linked with familial atrial fibrillation. J Mol Cell Cardiol 2019; 132:24-35. [PMID: 31077706 DOI: 10.1016/j.yjmcc.2019.05.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 05/01/2019] [Accepted: 05/03/2019] [Indexed: 11/28/2022]
Abstract
A frameshift (fs) mutation in the natriuretic peptide precursor A (NPPA) gene, encoding a mutant atrial natriuretic peptide (Mut-ANP), has been linked with familial atrial fibrillation (AF) but the underlying mechanisms by which the mutation causes AF remain unclear. We engineered 2 transgenic (TG) mouse lines expressing the wild-type (WT)-NPPA gene (H-WT-NPPA) and the human fs-Mut-NPPA gene (H-fsMut-NPPA) to test the hypothesis that mice overexpressing the human NPPA mutation are more susceptible to AF and elucidate the underlying electrophysiologic and molecular mechanisms. Transthoracic echocardiography and surface electrocardiography (ECG) were performed in H-fsMut-NPPA, H-WT-NPPA, and Non-TG mice. Invasive electrophysiology, immunohistochemistry, Western blotting and patch clamping of membrane potentials were performed. To examine the role of the Mut-ANP in ion channel remodeling, we measured plasma cyclic guanosine monophosphate (cGMP) and cyclic adenosine monophosphate (cAMP) levels and protein kinase A (PKA) activity in the 3 groups of mice. In H-fsMut-NPPA mice mean arterial pressure (MAP) was reduced when compared to H-WT-NPPA and Non-TG mice. Furthermore, injection of synthetic fs-Mut-ANP lowered the MAP in H-WT-NPPA and Non-TG mice while synthetic WT-ANP had no effect on MAP in the 3 groups of mice. ECG characterization revealed significantly prolonged QRS duration in H-fsMut-NPPA mice when compared to the other two groups. Trans-Esophageal (TE) atrial pacing of H-fsMut-NPPA mice showed increased AF burden and AF episodes when compared with H-WT-NPPA or Non-TG mice. The cardiac Na+ (NaV1.5) and Ca2+ (CaV1.2/CaV1.3) channel expression and currents (INa, ICaL) and action potential durations (APD90/APD50/APD20) were significantly reduced in H-fsMut-NPPA mice while the rectifier K+ channel current (IKs) was markedly increased when compared to the other 2 groups of mice. In addition, plasma cGMP levels were only increased in H-fsMut-NPPA mice with a corresponding reduction in plasma cAMP levels and PKA activity. In summary, we showed that mice overexpressing an AF-linked NPPA mutation are more prone to develop AF and this risk is mediated in part by remodeling of the cardiac Na+, Ca2+ and K+ channels creating an electrophysiologic substrate for reentrant AF.
Collapse
Affiliation(s)
- Ambili Menon
- Departments of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Liang Hong
- Departments of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Eleonora Savio-Galimberti
- Department of Biomedical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, PA, United States of America
| | - Arvind Sridhar
- Departments of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Seock-Won Youn
- Departments of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America; Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Meihong Zhang
- Departments of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Kaylen Kor
- Department of Pharmacology, Vanderbilt University Medical Center, United States of America
| | - Marcia Blair
- Department of Pharmacology, Vanderbilt University Medical Center, United States of America
| | - Sabina Kupershmidt
- Department of Nursing, University of South Dakota Sioux Falls, SD, United States of America
| | - Dawood Darbar
- Departments of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America; Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, United States of America; Pharmacology, University of Illinois at Chicago, Chicago, IL, United States of America.
| |
Collapse
|
25
|
Complexity reduction in human atrial modeling using extended Kalman filter. Med Biol Eng Comput 2018; 57:777-794. [PMID: 30402788 DOI: 10.1007/s11517-018-1921-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 10/24/2018] [Indexed: 10/27/2022]
Abstract
Human atrial tissue electrophysiology is modeled upon biophysical details obtained from cellular level measurements. Data collected for this purpose typically represent a unique state of the tissue. As reproducing dynamic cases such as subject-varied and/or disorder-varied electrophysiological properties is in question, such complex models are typically hard to use. Hence, there is a need for simpler yet biophysically accurate and mathematically tractable models to be used for case-specific reproductions and simulations. In this study, a scheme for parameter estimation of a phenomenological cardiac model to match a targeted behavior generated from a complex model is used. Specifically, an algorithm incorporating extended Kalman filter (EKF) into the scheme is proposed. Its performance is then compared to that of particle swarm optimization (PSO) and sequential quadratic programming (SQP), algorithms that have been widely used for parameter optimization. Both robustness and adaptability performance of the algorithms are tested through various designs. For this, reproducing action potential (AP) waveforms of varying remodeling states of atrial fibrillation (AF) at different stimulus protocols was targeted. Also, randomly generated initial parameter sets are included in the tests. In addition, AP duration (APD) restitution curve (RC) is used for a multiscale evaluation of fitting performance. Finally, wavefront propagation on 2D of a selected AF remodeling state using parameter solutions from each of the algorithms is simulated for a qualitative evaluation. In general, PSO yielded superior performances than EKF and SQP with respect to fitting AP waveforms. Considering both AP and APD RC, however, EKF yielded the best accuracies. Also, more accurate spiral wave reentry is obtained with EKF. Overall, EKF algorithm yielded the best performance in robustness and adaptability. Graphical Abstract.
Collapse
|
26
|
Saha M, Roney CH, Bayer JD, Meo M, Cochet H, Dubois R, Vigmond EJ. Wavelength and Fibrosis Affect Phase Singularity Locations During Atrial Fibrillation. Front Physiol 2018; 9:1207. [PMID: 30246796 PMCID: PMC6139329 DOI: 10.3389/fphys.2018.01207] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 08/10/2018] [Indexed: 01/06/2023] Open
Abstract
The mechanisms underlying atrial fibrillation (AF), the most common sustained cardiac rhythm disturbance, remain elusive. Atrial fibrosis plays an important role in the development of AF and rotor dynamics. Both electrical wavelength (WL) and the degree of atrial fibrosis change as AF progresses. However, their combined effect on rotor core location remains unknown. The aim of this study was to analyze the effects of WL change on rotor core location in both fibrotic and non-fibrotic atria. Three patient specific fibrosis distributions (total fibrosis content: 16.6, 22.8, and 19.2%) obtained from clinical imaging data of persistent AF patients were incorporated in a bilayer atrial computational model. Fibrotic effects were modeled as myocyte-fibroblast coupling + conductivity remodeling; structural remodeling; ionic current changes + conductivity remodeling; and combinations of these methods. To change WL, action potential duration (APD) was varied from 120 to 240ms, representing the range of clinically observed AF cycle length, by modifying the inward rectifier potassium current (IK1) conductance between 80 and 140% of the original value. Phase singularities (PSs) were computed to identify rotor core locations. Our results show that IK1 conductance variation resulted in a decrease of APD and WL across the atria. For large WL in the absence of fibrosis, PSs anchored to regions with high APD gradient at the center of the left atrium (LA) anterior wall and near the junctions of the inferior pulmonary veins (PVs) with the LA. Decreasing the WL induced more PSs, whose distribution became less clustered. With fibrosis, PS locations depended on the fibrosis distribution and the fibrosis implementation method. The proportion of PSs in fibrotic areas and along the borders varied with both WL and fibrosis modeling method: for patient one, this was 4.2-14.9% as IK1 varied for the structural remodeling representation, but 12.3-88.4% using the combination of structural remodeling with myocyte-fibroblast coupling. The degree and distribution of fibrosis and the choice of implementation technique had a larger effect on PS locations than the WL variation. Thus, distinguishing the fibrotic mechanisms present in a patient is important for interpreting clinical fibrosis maps to create personalized models.
Collapse
Affiliation(s)
- Mirabeau Saha
- IMB, UMR 5251, University of Bordeaux, Pessac, France
- IHU Liryc, Electrophysiology and Heart Modeling Institute, Fondation Bordeaux University, Pessac, France
| | - Caroline H. Roney
- Department of Biomedical Engineering, King's College London, London, United Kingdom
| | - Jason D. Bayer
- IMB, UMR 5251, University of Bordeaux, Pessac, France
- IHU Liryc, Electrophysiology and Heart Modeling Institute, Fondation Bordeaux University, Pessac, France
| | - Marianna Meo
- IMB, UMR 5251, University of Bordeaux, Pessac, France
- IHU Liryc, Electrophysiology and Heart Modeling Institute, Fondation Bordeaux University, Pessac, France
| | - Hubert Cochet
- IMB, UMR 5251, University of Bordeaux, Pessac, France
- IHU Liryc, Electrophysiology and Heart Modeling Institute, Fondation Bordeaux University, Pessac, France
| | - Remi Dubois
- IMB, UMR 5251, University of Bordeaux, Pessac, France
- IHU Liryc, Electrophysiology and Heart Modeling Institute, Fondation Bordeaux University, Pessac, France
| | - Edward J. Vigmond
- IMB, UMR 5251, University of Bordeaux, Pessac, France
- IHU Liryc, Electrophysiology and Heart Modeling Institute, Fondation Bordeaux University, Pessac, France
| |
Collapse
|
27
|
Zhang X, Jing W. Upregulation of miR‑122 is associated with cardiomyocyte apoptosis in atrial fibrillation. Mol Med Rep 2018; 18:1745-1751. [PMID: 29901138 DOI: 10.3892/mmr.2018.9124] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 01/29/2018] [Indexed: 11/06/2022] Open
Abstract
Atrial fibrillation (AF) is a common cardiac arrhythmia, which is associated with increased cardiovascular morbidity and mortality. microRNA (miRNA/miR)‑122 has been reported to be related with heart diseases, however, the functional role of miR‑122 in atrial fibrillation is unclear. Therefore, the aim of the present study was to investigate the roles of miR‑122 in atrial fibrillation. Male C57BL/6 mice were divided into the following three groups: Control, sham‑operation and AF. Mice in the AF group received transesophageal rapid atrial stimulation for the induction of AF. Cardiomyocytes isolated from mice in the AF group and were transfected with miR‑122 inhibitors. Reverse transcription‑quantitative polymerase chain reaction was used to assess the expression of miR‑122 in cardiomyocytes isolated from mice in the AF, sham‑operation and control groups, and in cells transfected with miR‑122 inhibitors. MTT and TUNEL assays were used to evaluate cardiomyocyte viability and apoptosis, respectively. Western blot analysis was used to assess the expression levels of extracellular signal‑regulated kinase (ERK) and phosphorylated (p)‑ERK, as well as the apoptosis‑associated proteins caspase‑3 and B‑cell lymphoma 2‑like 1 (Bcl‑x). The present results demonstrated that miR‑122 expression in the AF group was significantly increased compared with the sham‑operation and control groups, whereas it was significantly decreased following transfection with the miR‑122 inhibitor. Cardiomyocyte viability was increased and their apoptosis rate was significantly decreased following miR‑122 transfection. In addition, the expression of the anti‑apoptotic protein Bcl‑x was significantly upregulated, whereas the expression of the pro‑apoptotic caspase‑3 was significantly downregulated following miR‑122 inhibition. Furthermore, the p‑ERK/total ERK ratio was significantly increased in the miR‑122 inhibitor group compared with the AF and control groups. The present results suggested that miR‑122 may be implicated in the molecular mechanisms underlying the proliferation and apoptosis of cardiomyocytes in AF.
Collapse
Affiliation(s)
- Xiangqun Zhang
- Department of Emergency, Tianjin Medical University General Hospital, Tianjin 300070, P.R. China
| | - Wenli Jing
- Tianjin Medical College, Tianjin 300222, P.R. China
| |
Collapse
|
28
|
Barichello S, Roberts JD, Backx P, Boyle PM, Laksman Z. Personalizing therapy for atrial fibrillation: the role of stem cell and in silico disease models. Cardiovasc Res 2018; 114:931-943. [DOI: 10.1093/cvr/cvy090] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 04/06/2018] [Indexed: 11/12/2022] Open
Affiliation(s)
- Scott Barichello
- University of British Columbia, 2329 West Mall, Vancouver, BC V6T 1Z4, Canada
| | - Jason D Roberts
- Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine, Western University, London, ON, Canada
| | | | - Patrick M Boyle
- Department of Biomedical Engineering and Institute for Computational Medicine, Johns Hopkins University
| | - Zachary Laksman
- Division of Cardiology, University of British Columbia, 211-1033 Davie Street Vancouver, BC V6E 1M7, Canada
| |
Collapse
|
29
|
Vysotskaya Z, Chidipi B, Rodgers JL, Tang X, Samal E, Kolliputi N, Mohapatra S, Bennett ES, Panguluri SK. Elevated potassium outward currents in hyperoxia treated atrial cardiomyocytes. J Cell Physiol 2017; 233:4317-4326. [PMID: 29139549 DOI: 10.1002/jcp.26263] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 10/09/2017] [Indexed: 12/18/2022]
Abstract
Supplementation of 100% oxygen is a very common intervention in intensive care units (ICU) and critical care centers for patients with dysfunctional lung and lung disorders. Although there is advantage in delivering sufficient levels of oxygen, hyperoxia is reported to be directly associated with increasing in-hospital deaths. Our previous studies reported ventricular and electrical remodeling in hyperoxia treated mouse hearts, and in this article, for the first time, we are investigating the effects of hyperoxia on atrial electrophysiology using whole-cell patch-clamp electrophysiology experiments along with assessment of Kv1.5, Kv4.2, and KChIP2 transcripts and protein profiles using real-time quantitative RT-PCR and Western blotting. Our data showed that induction of hyperoxia for 3 days in mice showed larger outward potassium currents with shorter action potential durations (APD). This increase in current densities is due to significant increase in ultrarapid delayed rectifier outward K+ currents (IKur ) and rapidly activating, rapidly inactivating transient outward K+ current (Ito ) densities. We also observed a significant increase in both transcripts and protein levels of Kv1.5 and KChIP2 in hyperoxia treated atrial cardiomyocytes, whereas no significant change was observed in Kv4.2 transcripts or protein. The data presented here further support our previous findings that hyperoxia induces not only ventricular remodeling, but also atrial electrical remodeling.
Collapse
Affiliation(s)
- Zhanna Vysotskaya
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Bojjibabu Chidipi
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Jennifer L Rodgers
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, Florida
| | - Xiaolan Tang
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Eva Samal
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Narasaiah Kolliputi
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Subhra Mohapatra
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, Florida
| | - Eric S Bennett
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Siva K Panguluri
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, Florida
| |
Collapse
|
30
|
Lin YT, Chang ETY, Eatock J, Galla T, Clayton RH. Mechanisms of stochastic onset and termination of atrial fibrillation studied with a cellular automaton model. J R Soc Interface 2017; 14:rsif.2016.0968. [PMID: 28356539 PMCID: PMC5378131 DOI: 10.1098/rsif.2016.0968] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 03/02/2017] [Indexed: 01/23/2023] Open
Abstract
Mathematical models of cardiac electrical excitation are increasingly complex, with multiscale models seeking to represent and bridge physiological behaviours across temporal and spatial scales. The increasing complexity of these models makes it computationally expensive to both evaluate long term (more than 60 s) behaviour and determine sensitivity of model outputs to inputs. This is particularly relevant in models of atrial fibrillation (AF), where individual episodes last from seconds to days, and interepisode waiting times can be minutes to months. Potential mechanisms of transition between sinus rhythm and AF have been identified but are not well understood, and it is difficult to simulate AF for long periods of time using state-of-the-art models. In this study, we implemented a Moe-type cellular automaton on a novel, topologically equivalent surface geometry of the left atrium. We used the model to simulate stochastic initiation and spontaneous termination of AF, arising from bursts of spontaneous activation near pulmonary veins. The simplified representation of atrial electrical activity reduced computational cost, and so permitted us to investigate AF mechanisms in a probabilistic setting. We computed large numbers (approx. 105) of sample paths of the model, to infer stochastic initiation and termination rates of AF episodes using different model parameters. By generating statistical distributions of model outputs, we demonstrated how to propagate uncertainties of inputs within our microscopic level model up to a macroscopic level. Lastly, we investigated spontaneous termination in the model and found a complex dependence on its past AF trajectory, the mechanism of which merits future investigation.
Collapse
Affiliation(s)
- Yen Ting Lin
- Theoretical Physics Division, School of Physics and Astronomy, University of Manchester, Manchester, UK
| | - Eugene T Y Chang
- Department of Computer Science and INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, UK
| | - Julie Eatock
- Department of Computer Science, Brunel University London, Uxbridge UB8 3PH, UK
| | - Tobias Galla
- Theoretical Physics Division, School of Physics and Astronomy, University of Manchester, Manchester, UK
| | - Richard H Clayton
- Department of Computer Science and INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, UK
| |
Collapse
|
31
|
Valli H, Ahmad S, Chadda KR, Al-Hadithi ABAK, Grace AA, Jeevaratnam K, Huang CLH. Age-dependent atrial arrhythmic phenotype secondary to mitochondrial dysfunction in Pgc-1β deficient murine hearts. Mech Ageing Dev 2017; 167:30-45. [PMID: 28919427 PMCID: PMC5652526 DOI: 10.1016/j.mad.2017.09.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 08/24/2017] [Accepted: 09/13/2017] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Ageing and several age-related chronic conditions including obesity, insulin resistance and hypertension are associated with mitochondrial dysfunction and represent independent risk factors for atrial fibrillation (AF). MATERIALS AND METHODS Atrial arrhythmogenesis was investigated in Langendorff-perfused young (3-4 month) and aged (>12 month), wild type (WT) and peroxisome proliferator activated receptor-γ coactivator-1β deficient (Pgc-1β-/-) murine hearts modeling age-dependent chronic mitochondrial dysfunction during regular pacing and programmed electrical stimulation (PES). RESULTS AND DISCUSSION The Pgc-1β-/- genotype was associated with a pro-arrhythmic phenotype progressing with age. Young and aged Pgc-1β-/- hearts showed compromised maximum action potential (AP) depolarization rates, (dV/dt)max, prolonged AP latencies reflecting slowed action potential (AP) conduction, similar effective refractory periods and baseline action potential durations (APD90) but shortened APD90 in APs in response to extrasystolic stimuli at short stimulation intervals. Electrical properties of APs triggering arrhythmia were similar in WT and Pgc-1β-/- hearts. Pgc-1β-/- hearts showed accelerated age-dependent fibrotic change relative to WT, with young Pgc-1β-/- hearts displaying similar fibrotic change as aged WT, and aged Pgc-1β-/- hearts the greatest fibrotic change. Mitochondrial deficits thus result in an arrhythmic substrate, through slowed AP conduction and altered repolarisation characteristics, arising from alterations in electrophysiological properties and accelerated structural change.
Collapse
Affiliation(s)
- Haseeb Valli
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge, CB2 3EG, United Kingdom
| | - Shiraz Ahmad
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge, CB2 3EG, United Kingdom
| | - Karan R Chadda
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge, CB2 3EG, United Kingdom; Faculty of Health and Medical Sciences, University of Surrey, GU2 7AL, Guildford, Surrey, United Kingdom
| | - Ali B A K Al-Hadithi
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge, CB2 3EG, United Kingdom
| | - Andrew A Grace
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QW, United Kingdom
| | - Kamalan Jeevaratnam
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge, CB2 3EG, United Kingdom; Faculty of Health and Medical Sciences, University of Surrey, GU2 7AL, Guildford, Surrey, United Kingdom; PU-RCSI School of Medicine, Perdana University, 43400, Serdang, Selangor Darul Ehsan, Malaysia
| | - Christopher L-H Huang
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge, CB2 3EG, United Kingdom; Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QW, United Kingdom.
| |
Collapse
|
32
|
Krogh-Madsen T, Christini DJ. Slow [Na +] i dynamics impacts arrhythmogenesis and spiral wave reentry in cardiac myocyte ionic model. CHAOS (WOODBURY, N.Y.) 2017; 27:093907. [PMID: 28964146 DOI: 10.1063/1.4999475] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Accumulation of intracellular Na+ is gaining recognition as an important regulator of cardiac myocyte electrophysiology. The intracellular Na+ concentration can be an important determinant of the cardiac action potential duration, can modulate the tissue-level conduction of excitation waves, and can alter vulnerability to arrhythmias. Mathematical models of cardiac electrophysiology often incorporate a dynamic intracellular Na+ concentration, which changes much more slowly than the remaining variables. We investigated the dependence of several arrhythmogenesis-related factors on [Na+]i in a mathematical model of the human atrial action potential. In cell simulations, we found that [Na+]i accumulation stabilizes the action potential duration to variations in several conductances and that the slow dynamics of [Na+]i impacts bifurcations to pro-arrhythmic afterdepolarizations, causing intermittency between different rhythms. In long-lasting tissue simulations of spiral wave reentry, [Na+]i becomes spatially heterogeneous with a decreased area around the spiral wave rotation center. This heterogeneous region forms a functional anchor, resulting in diminished meandering of the spiral wave. Our findings suggest that slow, physiological, rate-dependent variations in [Na+]i may play complex roles in cellular and tissue-level cardiac dynamics.
Collapse
Affiliation(s)
- Trine Krogh-Madsen
- Greenberg Division of Cardiology, Weill Cornell Medicine, New York, New York 10065, USA; Institute for Computational Biomedicine, Weill Cornell Medicine, New York, New York 10065, USA; and Cardiovascular Research Institute, Weill Cornell Medicine, New York, New York 10065, USA
| | - David J Christini
- Greenberg Division of Cardiology, Weill Cornell Medicine, New York, New York 10065, USA; Institute for Computational Biomedicine, Weill Cornell Medicine, New York, New York 10065, USA; and Cardiovascular Research Institute, Weill Cornell Medicine, New York, New York 10065, USA
| |
Collapse
|
33
|
Deng D, Murphy MJ, Hakim JB, Franceschi WH, Zahid S, Pashakhanloo F, Trayanova NA, Boyle PM. Sensitivity of reentrant driver localization to electrophysiological parameter variability in image-based computational models of persistent atrial fibrillation sustained by a fibrotic substrate. CHAOS (WOODBURY, N.Y.) 2017; 27:093932. [PMID: 28964164 PMCID: PMC5605332 DOI: 10.1063/1.5003340] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 09/04/2017] [Indexed: 05/30/2023]
Abstract
Atrial fibrillation (AF) is the most common sustained cardiac arrhythmia, causing morbidity and mortality in millions worldwide. The atria of patients with persistent AF (PsAF) are characterized by the presence of extensive and distributed atrial fibrosis, which facilitates the formation of persistent reentrant drivers (RDs, i.e., spiral waves), which promote fibrillatory activity. Targeted catheter ablation of RD-harboring tissues has shown promise as a clinical treatment for PsAF, but the outcomes remain sub-par. Personalized computational modeling has been proposed as a means of non-invasively predicting optimal ablation targets in individual PsAF patients, but it remains unclear how RD localization dynamics are influenced by inter-patient variability in the spatial distribution of atrial fibrosis, action potential duration (APD), and conduction velocity (CV). Here, we conduct simulations in computational models of fibrotic atria derived from the clinical imaging of PsAF patients to characterize the sensitivity of RD locations to these three factors. We show that RDs consistently anchor to boundaries between fibrotic and non-fibrotic tissues, as delineated by late gadolinium-enhanced magnetic resonance imaging, but those changes in APD/CV can enhance or attenuate the likelihood that an RD will anchor to a specific site. These findings show that the level of uncertainty present in patient-specific atrial models reconstructed without any invasive measurements (i.e., incorporating each individual's unique distribution of fibrotic tissue from medical imaging alongside an average representation of AF-remodeled electrophysiology) is sufficiently high that a personalized ablation strategy based on targeting simulation-predicted RD trajectories alone may not produce the desired result.
Collapse
Affiliation(s)
- Dongdong Deng
- Institute for Computational Medicine and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Michael J Murphy
- Institute for Computational Medicine and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Joe B Hakim
- Institute for Computational Medicine and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - William H Franceschi
- Institute for Computational Medicine and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Sohail Zahid
- Institute for Computational Medicine and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Farhad Pashakhanloo
- Institute for Computational Medicine and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Natalia A Trayanova
- Institute for Computational Medicine and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Patrick M Boyle
- Institute for Computational Medicine and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
| |
Collapse
|
34
|
Iravanian S, Langberg JJ. Critical phase transitions during ablation of atrial fibrillation. CHAOS (WOODBURY, N.Y.) 2017; 27:093925. [PMID: 28964135 DOI: 10.1063/1.5000350] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Atrial fibrillation (AF) is the most common sustained cardiac arrhythmia with significant morbidity and mortality. Pharmacological agents are not very effective in the management of AF. Therefore, ablation procedures have become the mainstay of AF management. The irregular and seemingly chaotic atrial activity in AF is caused by one or more meandering spiral waves. Previously, we have shown the presence of sudden rhythm organization during ablation of persistent AF. We hypothesize that the observed transitions from a disorganized to an organized rhythm is a critical phase transition. Here, we explore this hypothesis by simulating ablation in an anatomically-correct 3D AF model. In 722 out of 2160 simulated ablation, at least one sudden transition from AF to an organized rhythm (flutter) was noted (33%). They were marked by a sudden decrease in the cycle length entropy and increase in the mean cycle length. At the same time, the number of reentrant wavelets decreased from 2.99 ± 0.06 in AF to 1.76 ± 0.05 during flutter, and the correlation length scale increased from 13.3 ± 1.0 mm to 196.5 ± 86.6 mm (both P < 0.0001). These findings are consistent with the hypothesis that transitions from AF to an anatomical flutter behave as phase transitions in complex non-equilibrium dynamical systems with flutter acting as an absorbing state. Clinically, the facilitation of phase transition should be considered a novel mechanism of ablation and may help to design effective ablation strategies.
Collapse
Affiliation(s)
- Shahriar Iravanian
- Emory University Hospital, 1364 Clifton Road, NE, Ste F-414, Atlanta, Georgia 30322, USA
| | - Jonathan J Langberg
- Emory University Hospital, 1364 Clifton Road, NE, Ste F-414, Atlanta, Georgia 30322, USA
| |
Collapse
|
35
|
Song JS, Wi J, Lee HJ, Hwang M, Lim B, Kim TH, Uhm J, Joung B, Lee M, Seo JW, Pak HN. Role of atrial wall thickness in wave-dynamics of atrial fibrillation. PLoS One 2017; 12:e0182174. [PMID: 28827810 PMCID: PMC5565105 DOI: 10.1371/journal.pone.0182174] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 07/13/2017] [Indexed: 01/13/2023] Open
Abstract
Background/Aims Atrial anatomy and thickness may affect the electrical wave-dynamics of atrial fibrillation (AF). We explored the relationship between left atrial (LA) wall thickness (LAWT) or LA geometry and AF wave-dynamics. Methods We included 15 patients with persistent AF (age, 62.3 ± 11.9 years) who underwent AF catheter ablation. We measured the LAWT, LA endocardial curvature, and SD-curvature (surface bumpiness) from preprocedural computed tomography images. We compared those anatomical characteristics with electrophysiologic parameters such as dominant frequency (DF), Shannon entropy (ShEn), or complex fractionated atrial electrogram (CFAE)-cycle length (CL), calculated from intracardiac bipolar electrograms (300–500 points, 5 s), acquired during ablation procedures. Results 1. LAWT (excluding fat) varied widely among patients, locations, and types of AF. LAWT was inversely correlated with LA volume (r = -0.565, p = 0.028) and positively correlated with SD-curvature (r = 0.272, p < 0.001). 2. LAWT was positively correlated with ShEn (r = 0.233, p < 0.001) and negatively correlated with CFAE-CL (r = -0.107, p = 0.038). 3. In the multivariate linear regression analyses for AF wave-dynamics parameters, DF (β = -0.29 [95% CI -0.44–-0.14], p < 0.001), ShEn (β = 0.19 [95% CI 0.12–0.25], p < 0.001), and CFAE-CL (β = 7.49 [95% CI 0.65–14.34], p = 0.032) were independently associated with LAWT. Conclusion Regional LAWT is associated with LA structural features, and has significant correlations with the wave-dynamics parameters associated with electrical wavebreaks or rotors in patients with persistent AF.
Collapse
Affiliation(s)
- Jun-Seop Song
- Yonsei University Health System, Seoul, Republic of Korea
| | - Jin Wi
- Yonsei University Health System, Seoul, Republic of Korea
| | - Hye-Jeong Lee
- Yonsei University Health System, Seoul, Republic of Korea
| | - Minki Hwang
- Yonsei University Health System, Seoul, Republic of Korea
| | - Byounghyun Lim
- Yonsei University Health System, Seoul, Republic of Korea
| | - Tae-Hoon Kim
- Yonsei University Health System, Seoul, Republic of Korea
| | - Jae‐Sun Uhm
- Yonsei University Health System, Seoul, Republic of Korea
| | - Boyoung Joung
- Yonsei University Health System, Seoul, Republic of Korea
| | | | - Jeong-Wook Seo
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hui-Nam Pak
- Yonsei University Health System, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
36
|
Boyle PM, Zahid S, Trayanova NA. Towards personalized computational modelling of the fibrotic substrate for atrial arrhythmia. Europace 2017; 18:iv136-iv145. [PMID: 28011841 DOI: 10.1093/europace/euw358] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 07/28/2016] [Indexed: 11/13/2022] Open
Abstract
: Atrial arrhythmias involving a fibrotic substrate are an important cause of morbidity and mortality. In many cases, effective treatment of such rhythm disorders is severely hindered by a lack of mechanistic understanding relating features of fibrotic remodelling to dynamics of re-entrant arrhythmia. With the advent of clinical imaging modalities capable of resolving the unique fibrosis spatial pattern present in the atria of each individual patient, a promising new research trajectory has emerged in which personalized computational models are used to analyse mechanistic underpinnings of arrhythmia dynamics based on the distribution of fibrotic tissue. In this review, we first present findings that have yielded a robust and detailed biophysical representation of fibrotic substrate electrophysiological properties. Then, we summarize the results of several recent investigations seeking to use organ-scale models of the fibrotic human atria to derive new insights on mechanisms of arrhythmia perpetuation and to develop novel strategies for model-assisted individualized planning of catheter ablation procedures for atrial arrhythmias.
Collapse
Affiliation(s)
- Patrick M Boyle
- Department of Biomedical Engineering and Institute for Computational Medicine, Johns Hopkins University, 3400 N Charles St, 208 Hackerman Hall, Baltimore, MD 21218, USA
| | - Sohail Zahid
- Department of Biomedical Engineering and Institute for Computational Medicine, Johns Hopkins University, 3400 N Charles St, 208 Hackerman Hall, Baltimore, MD 21218, USA
| | - Natalia A Trayanova
- Department of Biomedical Engineering and Institute for Computational Medicine, Johns Hopkins University, 3400 N Charles St, 208 Hackerman Hall, Baltimore, MD 21218, USA
| |
Collapse
|
37
|
Altered long non-coding RNA expression profile in rabbit atria with atrial fibrillation: TCONS_00075467 modulates atrial electrical remodeling by sponging miR-328 to regulate CACNA1C. J Mol Cell Cardiol 2017; 108:73-85. [PMID: 28546098 DOI: 10.1016/j.yjmcc.2017.05.009] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 05/17/2017] [Accepted: 05/19/2017] [Indexed: 02/01/2023]
Abstract
Electrical remodeling has been reported to play a major role in the initiation and maintenance of atrial fibrillation (AF). Long non-coding RNAs (lncRNAs) have been increasingly recognized as contributors to the pathology of heart diseases. However, the roles and mechanisms of lncRNAs in electrical remodeling during AF remain unknown. In this study, the lncRNA expression profiles of right atria were investigated in AF and non-AF rabbit models by using RNA sequencing technique and validated using quantitative real-time polymerase chain reaction (qRT-PCR). A total of 99,843 putative new lncRNAs were identified, in which 1220 differentially expressed transcripts exhibited >2-fold change. Bioinformatics analysis was conducted to predict the functions and interactions of the aberrantly expressed genes. On the basis of a series of filtering pipelines, one lncRNA, TCONS_00075467, was selected to explore its effects and mechanisms on electrical remodeling. The atrial effective refractory period was shortened in vivo and the L-type calcium current and action potential duration were decreased in vitro by silencing of TCONS_00075467 with lentiviruses. Besides, the expression of miRNA-328 was negatively correlated with TCONS_00075467. We further demonstrated that TCONS_00075467 could sponge miRNA-328 in vitro and in vivo to regulate the downstream protein coding gene CACNA1C. In addition, miRNA-328 could partly reverse the effects of TCONS_00075467 on electrical remodeling. In summary, dysregulated lncRNAs may play important roles in modulating electrical remodeling during AF. Our study may facilitate the mechanism studies of lncRNAs in AF pathogenesis and provide potential therapeutic targets for AF.
Collapse
|
38
|
Multiphysics Modeling of the Atrial Systole under Standard Ablation Strategies. Cardiovasc Eng Technol 2017; 8:205-218. [DOI: 10.1007/s13239-017-0308-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 05/10/2017] [Indexed: 11/26/2022]
|
39
|
Abstract
Cardiac arrhythmias can follow disruption of the normal cellular electrophysiological processes underlying excitable activity and their tissue propagation as coherent wavefronts from the primary sinoatrial node pacemaker, through the atria, conducting structures and ventricular myocardium. These physiological events are driven by interacting, voltage-dependent, processes of activation, inactivation, and recovery in the ion channels present in cardiomyocyte membranes. Generation and conduction of these events are further modulated by intracellular Ca2+ homeostasis, and metabolic and structural change. This review describes experimental studies on murine models for known clinical arrhythmic conditions in which these mechanisms were modified by genetic, physiological, or pharmacological manipulation. These exemplars yielded molecular, physiological, and structural phenotypes often directly translatable to their corresponding clinical conditions, which could be investigated at the molecular, cellular, tissue, organ, and whole animal levels. Arrhythmogenesis could be explored during normal pacing activity, regular stimulation, following imposed extra-stimuli, or during progressively incremented steady pacing frequencies. Arrhythmic substrate was identified with temporal and spatial functional heterogeneities predisposing to reentrant excitation phenomena. These could arise from abnormalities in cardiac pacing function, tissue electrical connectivity, and cellular excitation and recovery. Triggering events during or following recovery from action potential excitation could thereby lead to sustained arrhythmia. These surface membrane processes were modified by alterations in cellular Ca2+ homeostasis and energetics, as well as cellular and tissue structural change. Study of murine systems thus offers major insights into both our understanding of normal cardiac activity and its propagation, and their relationship to mechanisms generating clinical arrhythmias.
Collapse
Affiliation(s)
- Christopher L-H Huang
- Physiological Laboratory and the Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
40
|
Mechanisms of arrhythmogenesis related to calcium-driven alternans in a model of human atrial fibrillation. Sci Rep 2016; 6:36395. [PMID: 27812021 PMCID: PMC5095679 DOI: 10.1038/srep36395] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 10/11/2016] [Indexed: 11/08/2022] Open
Abstract
The occurrence of atrial fibrillation (AF) is associated with progressive changes in the calcium handling system of atrial myocytes. Calcium cycling instability has been implicated as an underlying mechanism of electrical alternans observed in patients who experience AF. However, the extent to which calcium-induced alternation of electrical activity in the atria contributes to arrhythmogenesis is unknown. In this study, we investigated the effects of calcium-driven alternans (CDA) on arrhythmia susceptibility in a biophysically detailed, 3D computer model of the human atria representing electrical and structural remodeling secondary to chronic AF. We found that elevated propensity to CDA rendered the atria vulnerable to ectopy-induced arrhythmia. It also increased the complexity and persistence of arrhythmias induced by fast pacing, with unstable scroll waves meandering and frequently breaking up to produce multiple wavelets. Our results suggest that calcium-induced electrical instability may increase arrhythmia vulnerability and promote increasing disorganization of arrhythmias in the chronic AF-remodeled atria, thus playing an important role in the progression of the disease.
Collapse
|
41
|
Jacquemet V. Lessons from computer simulations of ablation of atrial fibrillation. J Physiol 2016; 594:2417-30. [PMID: 26846178 DOI: 10.1113/jp271660] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 01/28/2016] [Indexed: 11/08/2022] Open
Abstract
This paper reviews the simulations of catheter ablation in computer models of the atria, from the first attempts to the most recent anatomical models. It describes how postulated substrates of atrial fibrillation can be incorporated into mathematical models, how modelling studies can be designed to test ablation strategies, what their current trade-offs and limitations are, and what clinically relevant lessons can be learnt from these simulations. Drawing a parallel between clinical and modelling studies, six ablation targets are considered: pulmonary vein isolation, linear ablation, ectopic foci, complex fractionated atrial electrogram, rotors and ganglionated plexi. The examples presented for each ablation target illustrate a major advantage of computer models, the ability to identify why a therapy is successful or not in a given atrial fibrillation substrate. The integration of pathophysiological data to create detailed models of arrhythmogenic substrates is expected to solidify the understanding of ablation mechanisms and to provide theoretical arguments supporting substrate-specific ablation strategies.
Collapse
Affiliation(s)
- Vincent Jacquemet
- Department of Molecular and Integrative Physiology, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada.,Centre de Recherche, Hôpital du Sacré-Cœur, Montréal, QC, Canada
| |
Collapse
|
42
|
Cheng W, Zhu Y, Wang H. The MAPK pathway is involved in the regulation of rapid pacing-induced ionic channel remodeling in rat atrial myocytes. Mol Med Rep 2016; 13:2677-82. [PMID: 26847818 DOI: 10.3892/mmr.2016.4862] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Accepted: 01/11/2016] [Indexed: 11/06/2022] Open
Abstract
Alterations to the expression L‑type calcium channels (LTCCs) and Kv4.3 potassium channels form the possible basis of atrial electrical remodeling during rapid pacing. The mitogen‑activated protein kinase (MAPK) pathway is affected by increases in cytoplasmic Ca2+, and therefore represents an attractive candidate for the regulation and mediation of Ca2+‑induced ion channel remodeling. The present study aimed to investigate alterations to the ion channel‑MAPK axis, and to determine its influence on ion channel remodeling during atrial fibrillation. Rat atrial myocytes were isolated, cultured, and in vitro rapid pacing was established. Intracellular Ca2+ signals were monitored using the Fluo‑3/AM Ca2+ indicator. Verapamil, PD98058 and SB203580 were added to the culture medium of various groups at specific time‑points. The mRNA expression levels of LTCC‑α1c and Kv4.3 potassium channels were detected by reverse transcription‑polymerase chain reaction. Western blotting was performed to determine the expression levels of channel and signaling proteins. The results demonstrated that fast pacing significantly increased the intracellular Ca2+ concentration in atrial myocytes, whereas treatment with verapamil markedly inhibited this increase. In addition, verapamil significantly antagonized the rapid pacing‑induced activation of extracellular signal‑regulated kinase (ERK) and p38MAPK. These results indicated that the MAPK pathway may have an important role in the opening of LTCCs, and alterations to MAPK molecule expression could affect the expression and remodeling of ion channels.
Collapse
Affiliation(s)
- Wei Cheng
- Department of Cardiothoracic Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Yun Zhu
- Department of Cardiothoracic Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Haidong Wang
- Department of Cardiothoracic Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| |
Collapse
|
43
|
Ferrer A, Sebastián R, Sánchez-Quintana D, Rodríguez JF, Godoy EJ, Martínez L, Saiz J. Detailed Anatomical and Electrophysiological Models of Human Atria and Torso for the Simulation of Atrial Activation. PLoS One 2015; 10:e0141573. [PMID: 26523732 PMCID: PMC4629897 DOI: 10.1371/journal.pone.0141573] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 10/09/2015] [Indexed: 01/24/2023] Open
Abstract
Atrial arrhythmias, and specifically atrial fibrillation (AF), induce rapid and irregular activation patterns that appear on the torso surface as abnormal P-waves in electrocardiograms and body surface potential maps (BSPM). In recent years both P-waves and the BSPM have been used to identify the mechanisms underlying AF, such as localizing ectopic foci or high-frequency rotors. However, the relationship between the activation of the different areas of the atria and the characteristics of the BSPM and P-wave signals are still far from being completely understood. In this work we developed a multi-scale framework, which combines a highly-detailed 3D atrial model and a torso model to study the relationship between atrial activation and surface signals in sinus rhythm. Using this multi scale model, it was revealed that the best places for recording P-waves are the frontal upper right and the frontal and rear left quadrants of the torso. Our results also suggest that only nine regions (of the twenty-one structures in which the atrial surface was divided) make a significant contribution to the BSPM and determine the main P-wave characteristics.
Collapse
Affiliation(s)
- Ana Ferrer
- Centro de Investigación e Innovación en Bioingeniería (Ci2B), Universitat Politècnica de València, Valencia, Spain
- * E-mail:
| | - Rafael Sebastián
- Computational Multiscale Physiology Lab (CoMMLab), Department of Computer Science, Universitat de Valencia, Valencia, Spain
| | - Damián Sánchez-Quintana
- Department of Anatomy and Cell Biology, Faculty of Medicine, Universidad de Extremadura, Badajoz, Spain
| | - José F. Rodríguez
- Applied Mechanics and Bioengineering Group (AMB), Universidad de Zaragoza, Zaragoza, Spain, and Dipartimento di Chimica, Materiali e Ingegneria Chimica “Giulio Natta”, Politecnico di Milano, Milano, Italy
| | - Eduardo J. Godoy
- Centro de Investigación e Innovación en Bioingeniería (Ci2B), Universitat Politècnica de València, Valencia, Spain
| | - Laura Martínez
- Centro de Investigación e Innovación en Bioingeniería (Ci2B), Universitat Politècnica de València, Valencia, Spain
| | - Javier Saiz
- Centro de Investigación e Innovación en Bioingeniería (Ci2B), Universitat Politècnica de València, Valencia, Spain
| |
Collapse
|
44
|
Sun Y, Huang ZY, Wang ZH, Li CP, Meng XL, Zhang YJ, Su F, Ma N. TGF-β1 and TIMP-4 regulate atrial fibrosis in atrial fibrillation secondary to rheumatic heart disease. Mol Cell Biochem 2015; 406:131-8. [PMID: 25971370 DOI: 10.1007/s11010-015-2431-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 05/05/2015] [Indexed: 10/23/2022]
Abstract
To investigate the involvement of transforming growth factor-β1 (TGF-β1) and tissue inhibitor of metalloproteinase 4 (TIMP-4) in influencing the severity of atrial fibrosis in rheumatic heart disease (RHD) patients with atrial fibrillation (AF). The degree of myocardial fibrosis was evaluated using Masson staining. The expression levels of TGF-β1, TIMP-4, matrix metalloproteinase-2 (MMP-2), type I collagen, and type III collagen were estimated by Western blot analysis. Additionally, TGF-β1 and TIMP-4 mRNA levels were quantified by qRT-PCR. The effect of TGF-β1 stimulation on TIMP-4 expression was assessed by in vitro stimulation of freshly isolated human atrial fibroblasts with recombinant human TGF-β1, followed by Western blot analysis to detect changes in TIMP-4 levels. Masson stain revealed that the left atrial diameter and collagen volume fraction were obviously increased in AF patients, compared to sinus rhythm (SR) controls (both P < 0.05). Western blot analysis showed significantly elevated levels of the AF markers MMP-2, type I collagen, and type III collagen in the AF group, in comparison to the SR controls (all P < 0.05). In the AF group, TGF-β1 expression was relatively higher, while TIMP-4 expression was apparently lower than the SR group (all P < 0.05). TIMP-4 expression level showed a negative association with TGF-β1 expression level (r = -0.98, P < 0.01) and TGF-β1 stimulation of atrial fibroblasts led to a sharp decrease in TIMP-4 protein level. Increased TGF-β1 expression and decreased TIMP-4 expression correlated with atrial fibrosis and ECM changes in the atria of RHD patients with AF. Notably, TGF-β1 suppressed TIMP-4 expression, suggesting that selective TGF-β1 inhibitors may be useful therapeutic agents.
Collapse
Affiliation(s)
- Yu Sun
- Cardiovascular Department, Second Affiliated Hospital and Second Clinical Medical College, Fujian Medical University, Zhongshan North Road No.34, Quanzhou, 362000, Fujian Province, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Dong J, Zhao J, Zhang M, Liu G, Wang X, Liu Y, Yang N, Liu Y, Zhao G, Sun J, Tian J, Cheng C, Wei L, Li Y, Li W. β3-Adrenoceptor Impairs Mitochondrial Biogenesis and Energy Metabolism During Rapid Atrial Pacing-Induced Atrial Fibrillation. J Cardiovasc Pharmacol Ther 2015; 21:114-26. [PMID: 26130614 DOI: 10.1177/1074248415590440] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 05/11/2015] [Indexed: 11/17/2022]
Abstract
BACKGROUND The β3-adrenoceptor (β3-AR) is implicated in cardiac remodeling. Since metabolic dysfunction due to loss of mitochondria plays an important role in heart diseases, we examined the effects of β3-AR on mitochondrial biogenesis and energy metabolism in atrial fibrillation (AF). METHODS Atrial fibrillation was created by rapid atrial pacing in adult rabbits. Rabbits were randomly divided into 4 groups: control, pacing (P7), β3-AR antagonist (L748337), and β3-AR agonist (BRL37344) groups. Atrial effective refractory period (AERP) and AF induction rate were measured. Atrial concentrations of adenine nucleotides and phosphocreatine were quantified through high-performance liquid chromatography. Mitochondrial DNA content was determined. Real-time polymerase chain reaction and Western blot were used to examine the expression levels of signaling intermediates related to mitochondrial biogenesis. RESULTS After pacing for 7 days, β3-AR was significantly upregulated, AERP was reduced, and the AF induction rate was increased. The total adenine nucleotides pool was significantly reduced due to the decrease in adenosine triphosphate (ATP). The P7 group showed decreased activity of F0F1-ATPase. Mitochondrial DNA content was decreased and mitochondrial respiratory chain subunits were downregulated after pacing. Furthermore, expression of transcription factors involved in mitochondrial biogenesis, including peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α), nuclear respiratory factor 1 (NRF-1), and mitochondrial transcription factor A (Tfam), was lower in the P7 group in response to β3-AR activation. Further stimulation of β3-AR with BRL37344 exacerbated these effects, together with a significant decrease in the levels of phosphocreatine. In contrast, inhibition of β3-AR with L748337 partially restored mitochondrial biogenesis and energy metabolism of atria in the paced rabbits. CONCLUSION The activation of β3-AR contributes to atrial metabolic remodeling via transcriptional downregulation of PGC-1α/NRF-1/Tfam pathway that are involved in mitochondrial biogenesis, which ultimately perturbs mitochondrial function in rapid pacing-induced AF. The β3-AR is therefore a potential novel therapeutic target for the treatment or prevention of AF.
Collapse
Affiliation(s)
- Jingmei Dong
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jingjing Zhao
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Miaomiao Zhang
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Guangzhong Liu
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaobing Wang
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yixi Liu
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ning Yang
- Ultrasonic Cardiogram Room, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yongwu Liu
- Centre for Drug Safety Evaluation, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Guanqi Zhao
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiayu Sun
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jingpu Tian
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Cheping Cheng
- Department of Cardiology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Lin Wei
- Department of Cardiology, First Hospital of Harbin City, Harbin, China
| | - Yue Li
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Weimin Li
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
46
|
Egom EE, Vella K, Hua R, Jansen HJ, Moghtadaei M, Polina I, Bogachev O, Hurnik R, Mackasey M, Rafferty S, Ray G, Rose RA. Impaired sinoatrial node function and increased susceptibility to atrial fibrillation in mice lacking natriuretic peptide receptor C. J Physiol 2015; 593:1127-46. [PMID: 25641115 DOI: 10.1113/jphysiol.2014.283135] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 12/06/2014] [Indexed: 12/17/2022] Open
Abstract
Natriuretic peptides (NPs) are critical regulators of the cardiovascular system that are currently viewed as possible therapeutic targets for the treatment of heart disease. Recent work demonstrates potent NP effects on cardiac electrophysiology, including in the sinoatrial node (SAN) and atria. NPs elicit their effects via three NP receptors (NPR-A, NPR-B and NPR-C). Among these receptors, NPR-C is poorly understood. Accordingly, the goal of this study was to determine the effects of NPR-C ablation on cardiac structure and arrhythmogenesis. Cardiac structure and function were assessed in wild-type (NPR-C(+/+)) and NPR-C knockout (NPR-C(-/-)) mice using echocardiography, intracardiac programmed stimulation, patch clamping, high-resolution optical mapping, quantitative polymerase chain reaction and histology. These studies demonstrate that NPR-C(-/-) mice display SAN dysfunction, as indicated by a prolongation (30%) of corrected SAN recovery time, as well as an increased susceptibility to atrial fibrillation (6% in NPR-C(+/+) vs. 47% in NPR-C(-/-)). There were no differences in SAN or atrial action potential morphology in NPR-C(-/-) mice; however, increased atrial arrhythmogenesis in NPR-C(-/-) mice was associated with reductions in SAN (20%) and atrial (15%) conduction velocity, as well as increases in expression and deposition of collagen in the atrial myocardium. No differences were seen in ventricular arrhythmogenesis or fibrosis in NPR-C(-/-) mice. This study demonstrates that loss of NPR-C results in SAN dysfunction and increased susceptibility to atrial arrhythmias in association with structural remodelling and fibrosis in the atrial myocardium. These findings indicate a critical protective role for NPR-C in the heart.
Collapse
Affiliation(s)
- Emmanuel E Egom
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Human atrial cell models to analyse haemodialysis-related effects on cardiac electrophysiology: work in progress. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2014; 2014:291598. [PMID: 25587348 PMCID: PMC4284940 DOI: 10.1155/2014/291598] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 11/05/2014] [Accepted: 11/12/2014] [Indexed: 11/25/2022]
Abstract
During haemodialysis (HD) sessions, patients undergo alterations in the extracellular environment, mostly concerning plasma electrolyte concentrations, pH, and volume, together with a modification of sympathovagal balance. All these changes affect cardiac electrophysiology, possibly leading to an increased arrhythmic risk. Computational modeling may help to investigate the impact of HD-related changes on atrial electrophysiology. However, many different human atrial action potential (AP) models are currently available, all validated only with the standard electrolyte concentrations used in experiments. Therefore, they may respond in different ways to the same environmental changes. After an overview on how the computational approach has been used in the past to investigate the effect of HD therapy on cardiac electrophysiology, the aim of this work has been to assess the current state of the art in human atrial AP models, with respect to the HD context. All the published human atrial AP models have been considered and tested for electrolytes, volume changes, and different acetylcholine concentrations. Most of them proved to be reliable for single modifications, but all of them showed some drawbacks. Therefore, there is room for a new human atrial AP model, hopefully able to physiologically reproduce all the HD-related effects. At the moment, work is still in progress in this specific field.
Collapse
|
48
|
Chang KC, Bayer JD, Trayanova NA. Disrupted calcium release as a mechanism for atrial alternans associated with human atrial fibrillation. PLoS Comput Biol 2014; 10:e1004011. [PMID: 25501557 PMCID: PMC4263367 DOI: 10.1371/journal.pcbi.1004011] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 10/29/2014] [Indexed: 12/19/2022] Open
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia, but our knowledge of the arrhythmogenic substrate is incomplete. Alternans, the beat-to-beat alternation in the shape of cardiac electrical signals, typically occurs at fast heart rates and leads to arrhythmia. However, atrial alternans have been observed at slower pacing rates in AF patients than in controls, suggesting that increased vulnerability to arrhythmia in AF patients may be due to the proarrythmic influence of alternans at these slower rates. As such, alternans may present a useful therapeutic target for the treatment and prevention of AF, but the mechanism underlying alternans occurrence in AF patients at heart rates near rest is unknown. The goal of this study was to determine how cellular changes that occur in human AF affect the appearance of alternans at heart rates near rest. To achieve this, we developed a computational model of human atrial tissue incorporating electrophysiological remodeling associated with chronic AF (cAF) and performed parameter sensitivity analysis of ionic model parameters to determine which cellular changes led to alternans. Of the 20 parameters tested, only decreasing the ryanodine receptor (RyR) inactivation rate constant (kiCa) produced action potential duration (APD) alternans seen clinically at slower pacing rates. Using single-cell clamps of voltage, fluxes, and state variables, we determined that alternans onset was Ca2+-driven rather than voltage-driven and occurred as a result of decreased RyR inactivation which led to increased steepness of the sarcoplasmic reticulum (SR) Ca2+ release slope. Iterated map analysis revealed that because SR Ca2+ uptake efficiency was much higher in control atrial cells than in cAF cells, drastic reductions in kiCa were required to produce alternans at comparable pacing rates in control atrial cells. These findings suggest that RyR kinetics may play a critical role in altered Ca2+ homeostasis which drives proarrhythmic APD alternans in patients with AF. Atrial fibrillation is an irregular heart rhythm affecting millions of people worldwide. Effective treatment of this cardiac disorder relies upon our detailed knowledge and understanding of the mechanisms that lead to arrhythmia. Recent clinical observations have suggested that alternans, a phenomenon where the shape of the electrical signal in the heart alternates from beat to beat, may play an important role in this process, but the underlying mechanisms remain unknown. In this study, we use computational models to conduct a detailed examination of the causes and contributors to alternans associated with human atrial fibrillation. We find that in atria remodeled by atrial fibrillation, alternans appears near resting heart rates because several aspects of calcium cycling are disrupted in the atrial cells. In particular, the release and uptake of calcium from the cellular storage compartment, the sarcoplasmic reticulum, becomes imbalanced, leading to alternation in calcium signals from beat to beat. These findings provide important insights into the mechanisms of proarrhythmic alternans in human atrial fibrillation which may be used to develop novel therapeutic targets and treatment strategies in the future.
Collapse
Affiliation(s)
- Kelly C. Chang
- Institute for Computational Medicine, Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Jason D. Bayer
- IHU-LIRYC - L'Institut de RYthmologie et Modélisation Cardiaque, University of Bordeaux, Bordeaux, France
| | - Natalia A. Trayanova
- Institute for Computational Medicine, Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
49
|
Trayanova NA, Boyle PM, Arevalo HJ, Zahid S. Exploring susceptibility to atrial and ventricular arrhythmias resulting from remodeling of the passive electrical properties in the heart: a simulation approach. Front Physiol 2014; 5:435. [PMID: 25429272 PMCID: PMC4228852 DOI: 10.3389/fphys.2014.00435] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 10/24/2014] [Indexed: 12/19/2022] Open
Abstract
Under diseased conditions, remodeling of the cardiac tissue properties (“passive properties”) takes place; these are aspects of electrophysiological behavior that are not associated with active ion transport across cell membranes. Remodeling of the passive electrophysiological properties most often results from structural remodeling, such as gap junction down-regulation and lateralization, fibrotic growth infiltrating the myocardium, or the development of an infarct scar. Such structural remodeling renders atrial or ventricular tissue as a major substrate for arrhythmias. The current review focuses on these aspects of cardiac arrhythmogenesis. Due to the inherent complexity of cardiac arrhythmias, computer simulations have provided means to elucidate interactions pertinent to this spatial scale. Here we review the current state-of-the-art in modeling atrial and ventricular arrhythmogenesis as arising from the disease-induced changes in the passive tissue properties, as well as the contributions these modeling studies have made to our understanding of the mechanisms of arrhythmias in the heart. Because of the rapid advance of structural imaging methodologies in cardiac electrophysiology, we chose to present studies that have used such imaging methodologies to construct geometrically realistic models of cardiac tissue, or the organ itself, where the regional remodeling properties of the myocardium can be represented in a realistic way. We emphasize how the acquired knowledge can be used to pave the way for clinical applications of cardiac organ modeling under the conditions of structural remodeling.
Collapse
Affiliation(s)
- Natalia A Trayanova
- Department of Biomedical Engineering, Institute for Computational Medicine, Johns Hopkins University Baltimore, MD, USA
| | - Patrick M Boyle
- Department of Biomedical Engineering, Institute for Computational Medicine, Johns Hopkins University Baltimore, MD, USA
| | - Hermenegild J Arevalo
- Department of Biomedical Engineering, Institute for Computational Medicine, Johns Hopkins University Baltimore, MD, USA
| | - Sohail Zahid
- Department of Biomedical Engineering, Institute for Computational Medicine, Johns Hopkins University Baltimore, MD, USA
| |
Collapse
|
50
|
Dupraz M, Jacquemet V. Geometrical measurement of cardiac wavelength in reaction-diffusion models. CHAOS (WOODBURY, N.Y.) 2014; 24:033133. [PMID: 25273213 DOI: 10.1063/1.4895811] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
The dynamics of reentrant arrhythmias often consists in multiple wavelets propagating throughout an excitable medium. An arrhythmia can be sustained only if these reentrant waves have a sufficiently short wavelength defined as the distance traveled by the excitation wave during its refractory period. In a uniform medium, wavelength may be estimated as the product of propagation velocity and refractory period (electrophysiological wavelength). In order to accurately measure wavelength in more general substrates relevant to atrial arrhythmias (heterogeneous and anisotropic), we developed a mathematical framework to define geometrical wavelength at each time instant based on the length of streamlines following the propagation velocity field within refractory regions. Two computational methods were implemented: a Lagrangian approach in which a set of streamlines were integrated, and an Eulerian approach in which wavelength was the solution of a partial differential equation. These methods were compared in 1D/2D tissues and in a model of the left atrium. An advantage of geometrical definition of wavelength is that the wavelength of a wavelet can be tracked over time with high temporal resolution and smaller temporal variability in an anisotropic and heterogeneous medium. The results showed that the average electrophysiological wavelength was consistent with geometrical measurements of wavelength. Wavelets were however often shorter than the electrophysiological wavelength due to interactions with boundaries and other wavelets. These tools may help to assess more accurately the relation between substrate properties and wavelet dynamics in computer models.
Collapse
Affiliation(s)
- Marie Dupraz
- Institut de Génie Biomédical, Department of Physiology, Faculty of Medicine, Université de Montréal, Montréal (Québec) H3T 1J4, Canada and Centre de Recherche, Hôpital du Sacré-Coeur de Montréal, Montréal (Québec) H4J 1C5, Canada
| | - Vincent Jacquemet
- Institut de Génie Biomédical, Department of Physiology, Faculty of Medicine, Université de Montréal, Montréal (Québec) H3T 1J4, Canada and Centre de Recherche, Hôpital du Sacré-Coeur de Montréal, Montréal (Québec) H4J 1C5, Canada
| |
Collapse
|