151
|
The Predictive Value of Epicardial Fat Tissue Volume in the Occurrence and Development of Atrial Fibrillation: A Systematic Review and Meta-Analysis. Cardiol Res Pract 2022; 2022:2090309. [PMID: 36213458 PMCID: PMC9537030 DOI: 10.1155/2022/2090309] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/20/2022] [Accepted: 08/26/2022] [Indexed: 11/18/2022] Open
Abstract
Background Atrial fibrillation (AF) is one of the most common arrhythmias in clinical practice. Although fat is currently considered to be a risk factor for AF and a pathogenic link between epicardial fat tissue (EFT) and AF has been speculated, there are currently few clinical studies and literature data domestically or abroad. Objective This study conducted a meta-analysis of observational case series studies to verify the relationship between atrial fibrillation and EFT and to strengthen the predictive value of EFT in the occurrence, development, and postablative recurrence of AF. Methods We conducted a systematic search of the literature in electronic databases until December 2021 and supplemented this through manual searches of individual studies, reviewed articles, and reference lists in conference proceedings. This study conducted a meta-analysis to compare the differences between different populations, such as healthy participants and AF patients, healthy subjects and AF subtype cases, and paroxysmal and persistent AF with AF recurrence and without AF recurrence after ablation. Results Following the retrieval of 828 articles, only 22 articles were selected as research results. Accordingly, the meta-analysis results show that the volume of EFT in AF is greater than that in healthy subjects (MD = 39.34 ml, 95% CI = 27.11, 51.58); persistent AF is greater than paroxysmal AF (MD = 14.37 ml, 95% CI = 7.46, 21.27); and recurrence after ablation is greater than without recurrence (MD = 14.37 ml, 95% CI = 7.46, 21.27). Conclusion The results of this study further confirm the connection between EFT and AF and that EFT has a certain predictive value for the occurrence and development of AF.
Collapse
|
152
|
Jiang F, Zhang W, Lu H, Tan M, Zeng Z, Song Y, Ke X, Lin F. Prediction of herbal medicines based on immune cell infiltration and immune- and ferroptosis-related gene expression levels to treat valvular atrial fibrillation. Front Genet 2022; 13:886860. [PMID: 36246656 PMCID: PMC9554472 DOI: 10.3389/fgene.2022.886860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 09/06/2022] [Indexed: 11/13/2022] Open
Abstract
Inflammatory immune response is apparently one of the determinants of progressive exacerbation of valvular atrial fibrillation(VAF). Ferroptosis, an iron-dependent modality of regulated cell death, is involved in the immune regulation of cardiovascular disease. However, the relevant regulatory mechanisms of immune infiltration and ferroptosis in VAF have been less studied. In the current study, a highly efficient system for screening immunity- and ferroptosis-related biomarkers and immunomodulatory ability of herbal ingredients has been developed with the integration of intelligent data acquisition, data mining, network pharmacology, and computer-assisted target fishing. VAF patients showed higher infiltration of neutrophils and resting stage dendritic cells, while VSR patients showed higher infiltration of follicular helper T cells. In addition, six (e.g., PCSK2) and 47 (e.g., TGFBR1) ImmDEGs and one (SLC38A1) and four (TGFBR1, HMGB1, CAV1, and CD44) FerDEGs were highly expressed in patients with valvular sinus rhythm (VSR) and VAF, respectively. We further identified a core subnetwork containing 34 hub genes, which were intersected with ImmDEGs and FerDEGs to obtain the key gene TGFBR1. Based on TGFBR1, 14 herbs (e.g., Fructus zizyphi jujubae, Semen Juglandis, and Polygonum cuspidatum) and six herbal ingredients (curcumin, curcumine, D-glucose, hexose, oleovitamin A, and resveratrol) were predicted. Finally, TGFBR1 was found to dock well with curcumin and resveratrol, and it was further verified that curcumin and resveratrol could significantly reduce myocardial fibrosis. We believe that herbs rich in curcumin and resveratrol such as Rhizoma curcumae longae and Curcuma kwangsiensis, mitigate myocardial fibrosis to improve VAF by modulating the TGFβ/Smad signaling pathway. This strategy provides a prospective approach systemically characterizing phenotype-target-herbs relationships based on the tissue-specific biological functions in VAF and brings us new insights into the searching lead compounds from Chinese herbs.
Collapse
Affiliation(s)
- Feng Jiang
- Cardiology Department, Affiliated Baoan TCM Hospital, Guangzhou University of Traditional Chinese Medicine, Shenzhen, China
| | - Weiwei Zhang
- Cardiology Department, Affiliated Baoan TCM Hospital, Guangzhou University of Traditional Chinese Medicine, Shenzhen, China
| | - Hongdan Lu
- Cardiology Department, Affiliated Baoan TCM Hospital, Guangzhou University of Traditional Chinese Medicine, Shenzhen, China
| | - Meiling Tan
- Wenhua Community Health Service Center, Shenzhen Luohu Hospital Group, Shenzhen, China
| | - Zhicong Zeng
- Cardiology Department, Affiliated Baoan TCM Hospital, Guangzhou University of Traditional Chinese Medicine, Shenzhen, China
| | - Yinzhi Song
- Cardiology Department, Affiliated Baoan TCM Hospital, Guangzhou University of Traditional Chinese Medicine, Shenzhen, China
| | - Xiao Ke
- Department of Cardiology, Fuwai Hospital, Chinese Academy of Medical Sciences, Shenzhen(Shenzhen Sun Yat-sen Cardiovascular Hospital), Shenzhen, China
- *Correspondence: Fengxia Lin, ; Xiao Ke,
| | - Fengxia Lin
- Cardiology Department, Affiliated Baoan TCM Hospital, Guangzhou University of Traditional Chinese Medicine, Shenzhen, China
- *Correspondence: Fengxia Lin, ; Xiao Ke,
| |
Collapse
|
153
|
Dong C, Li J, Ding W, Ueda R, Xie X, Wu J, Matsuura H, Horie M. Open channel block of Kv1.5 channels by HMQ1611. Front Pharmacol 2022; 13:965086. [PMID: 36188606 PMCID: PMC9524145 DOI: 10.3389/fphar.2022.965086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/26/2022] [Indexed: 11/13/2022] Open
Abstract
Kv1.5 channels conduct the ultra-rapid delayed rectifier potassium current (IKur). Pharmacological blockade of human Kv1.5 (hKv1.5) has been regarded as an effective treatment of re-entrant based atrial fibrillation, because Kv1.5 is highly expressed in human cardiac atria but scarcely in ventricles. The Kv1.5 blockade is also expected to be used in cancer therapeutics since Kv1.5 is overexpressed in some types of human tumors. Here, we investigated the blockade of hKv1.5 channels by HMQ1611, a symmetrical biphenyl derivative. hKv1.5 channels were heterologously expressed in Chinese hamster ovary cells. The effects of HMQ1611 on wild-type and 13 hKv1.5 mutant channels were examined using the whole-cell patch-clamp method, and molecular docking simulation was conducted to predict the docking position of HMQ1611 within Kv1.5 channels. We showed that HMQ1611 reversibly inhibited the hKv1.5 current in a concentration-dependent manner (IC50 = 2.07 μM). HMQ1611 blockade of hKv1.5 current developed with time during depolarizing voltage-clamp steps, and this blockade was also voltage-dependent with a steep increase over the voltage range for channel openings. HMQ1611 inhibition was significantly reduced in the T479A, T480A, V505A, I508A, L510A, V512A, and V516A hKv1.5 mutant channels. Molecular docking analysis predicted that V505, V512, and T480 were involved in the blocking action of HMQ1611 on hKv1.5 channels. These results suggest that HMQ1611 inhibits hKv1.5 currents as an open channel blocker. Amino acid residues located at the base of the selectivity filter (T479 and T480) and in the S6 segment (V505, I508, L510, V512, and V516) of hKv1.5 appear to constitute potential binding sites for HMQ1611.
Collapse
Affiliation(s)
- Chao Dong
- Department of Pharmacology, School of Basic Medical Science, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi’an Jiaotong University, Ministry of Education, Xi’an, Shaanxi, China
- Department of Pharmacy, The First Affiliated Hospital of Xi’an Medical University, Xi’an, China
- Department of Physiology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Jiawei Li
- Department of Pharmacology, School of Basic Medical Science, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi’an Jiaotong University, Ministry of Education, Xi’an, Shaanxi, China
| | - Weiguang Ding
- Department of Physiology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Rika Ueda
- Department of Physiology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Xiaolu Xie
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Jie Wu
- Department of Pharmacology, School of Basic Medical Science, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi’an Jiaotong University, Ministry of Education, Xi’an, Shaanxi, China
- Department of Physiology, Shiga University of Medical Science, Otsu, Shiga, Japan
- Department of Cardiovascular and Respiratory Medicine, Shiga University of Medical Science, Otsu, Shiga, Japan
- *Correspondence: Jie Wu,
| | - Hiroshi Matsuura
- Department of Physiology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Minoru Horie
- Department of Cardiovascular and Respiratory Medicine, Shiga University of Medical Science, Otsu, Shiga, Japan
| |
Collapse
|
154
|
Tu T, Qin F, Bai F, Xiao Y, Ma Y, Li B, Liu N, Zhang B, Sun C, Liao X, Zhou S, Liu Q. Quantitative acetylated proteomics on left atrial appendage tissues revealed atrial energy metabolism and contraction status in patients with valvular heart disease with atrial fibrillation. Front Cardiovasc Med 2022; 9:962036. [PMID: 36176981 PMCID: PMC9513032 DOI: 10.3389/fcvm.2022.962036] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 08/18/2022] [Indexed: 11/21/2022] Open
Abstract
Background Numerous basic studies have demonstrated critical roles of metabolic and contractile remodeling in pathophysiological changes of atrial fibrillation (AF), but acetylation changes underlying atrial remodeling have not been fully elucidated. Quantitative acetylated proteomics enables researchers to identify a comprehensive map of protein alterations responsible for pathological development and progression of AF in the heart of patients. Materials and methods In this study, 18 samples (9 with chronic AF and 9 with sinus rhythm) of left atrial appendage (LAA) tissues were obtained during mitral valve replacement surgery. Changes in the quantitative acetylated proteome between the AF and sinus rhythm (SR) groups were studied by dimethyl labeling, acetylation affinity enrichment, and high-performance liquid chromatography-tandem mass spectrometry analysis. Results We identified a total of 5,007 acetylated sites on 1,330 acetylated proteins, among which 352 acetylated sites on 193 acetylated proteins were differentially expressed between the AF and SR groups by setting a quantification ratio of 1.3 for threshold value and P < 0.05 for significant statistical difference. The bioinformatics analysis showed that the differentially expressed acetylated proteins were mainly involved in energy metabolism and cellular contraction and structure function-related biological processes and pathways. Among 87 differentially expressed energy metabolism acetylated proteins related to the processes of fatty acid, carbohydrate, ketone body metabolism, and oxidative phosphorylation, nearly 87.1% Kac sites were upregulated (148 Kac sites among 170) in the AF group. Besides, generally declining acetylation of cardiac muscle contraction-related proteins (88.9% Kac sites of myosin) was found in the LAA of patients with AF. Immune coprecipitation combined with Western blotting was conducted to validate the differential expression of acetylated proteins. Conclusion Many differentially expressed energy metabolism and cellular contraction acetylated proteins were found in the LAA tissues of patients with chronic AF, and may reflect the impaired ATP production capacity and decreased atrial muscle contractility in the atrium during AF. Thus, acetylation may play an important regulatory role in metabolic and contractile remodeling of the atrium during AF. Moreover, the identified new acetylated sites and proteins may become promising targets for prevention and treatment of AF.
Collapse
Affiliation(s)
- Tao Tu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Fen Qin
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Fen Qin,
| | - Fan Bai
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yichao Xiao
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yingxu Ma
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Biao Li
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Na Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Baojian Zhang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Chao Sun
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xiaobo Liao
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Shenghua Zhou
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Qiming Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha, China
- Qiming Liu,
| |
Collapse
|
155
|
Benito B, García-Elías A, Ois Á, Tajes M, Vallès E, Ble M, Yáñez Bisbe L, Giralt-Steinhauer E, Rodríguez-Campello A, Cladellas Capdevila M, Martí-Almor J, Roquer J, Cuadrado-Godia E. Plasma levels of miRNA-1-3p are associated with subclinical atrial fibrillation in patients with cryptogenic stroke. REVISTA ESPANOLA DE CARDIOLOGIA (ENGLISH ED.) 2022; 75:717-726. [PMID: 35067470 DOI: 10.1016/j.rec.2021.12.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 11/30/2021] [Indexed: 06/14/2023]
Abstract
INTRODUCTION AND OBJECTIVES Identifying biomarkers of subclinical atrial fibrillation (AF) is of most interest in patients with cryptogenic stroke (CrS). We sought to evaluate the circulating microRNA (miRNA) profile of patients with CrS and AF compared with those in persistent sinus rhythm. METHODS Among 64 consecutive patients with CrS under continuous monitoring by a predischarge insertable monitor, 18 patients (9 with AF and 9 in persistent sinus rhythm) were selected for high-throughput determination of 754 miRNAs. Nine patients with concomitant stroke and AF were also screened to improve the yield of miRNA selection. Differentially expressed miRNAs were replicated in an independent cohort (n=46). Biological markers were stratified by the median and included in logistic regression analyses to evaluate their association with AF at 6 and 12 months. RESULTS Eight miRNAs were differentially expressed between patients with and without AF. In the replication cohort, miR-1-3p, a gene regulator involved in cardiac arrhythmogenesis, was the only miRNA to remain significantly higher in patients with CrS and AF vs those in sinus rhythm and showed a modest association with AF burden. High (= above the median) miR-1-3p plasma values, together with a low left atrial ejection fraction, were independently associated with the presence of AF at 6 and 12 months. CONCLUSIONS In this cohort, plasma levels of miR-1-3p were elevated in CrS patients with subsequent AF. Our results preliminarily suggest that miR-1-3p could be a novel biomarker that, together with clinical parameters, could help identify patients with CrS and a high risk of occult AF.
Collapse
Affiliation(s)
- Begoña Benito
- Servicio de Cardiología, Hospital Universitari Vall d'Hebron, Barcelona, Spain; Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain; Departament de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Spain.
| | - Anna García-Elías
- Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Parc de Salut Mar, Barcelona, Spain; Research Center, Montreal Heart Institute, Montreal, Canada
| | - Ángel Ois
- Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Parc de Salut Mar, Barcelona, Spain; Servicio de Neurología, Hospital del Mar, Parc de Salut Mar, Barcelona, Spain
| | - Marta Tajes
- Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Parc de Salut Mar, Barcelona, Spain
| | - Ermengol Vallès
- Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Parc de Salut Mar, Barcelona, Spain; Servicio de Cardiología, Hospital del Mar, Parc de Salut Mar, Barcelona, Spain
| | - Mireia Ble
- Servicio de Cardiología, Hospital del Mar, Parc de Salut Mar, Barcelona, Spain
| | | | - Eva Giralt-Steinhauer
- Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Parc de Salut Mar, Barcelona, Spain; Servicio de Cardiología, Hospital del Mar, Parc de Salut Mar, Barcelona, Spain
| | - Ana Rodríguez-Campello
- Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Parc de Salut Mar, Barcelona, Spain; Servicio de Cardiología, Hospital del Mar, Parc de Salut Mar, Barcelona, Spain
| | - Mercè Cladellas Capdevila
- Departament de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain; Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Parc de Salut Mar, Barcelona, Spain; Servicio de Cardiología, Hospital del Mar, Parc de Salut Mar, Barcelona, Spain
| | - Julio Martí-Almor
- Departament de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain; Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Parc de Salut Mar, Barcelona, Spain; Servicio de Cardiología, Hospital del Mar, Parc de Salut Mar, Barcelona, Spain
| | - Jaume Roquer
- Departament de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain; Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Parc de Salut Mar, Barcelona, Spain; Servicio de Cardiología, Hospital del Mar, Parc de Salut Mar, Barcelona, Spain
| | - Elisa Cuadrado-Godia
- Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Parc de Salut Mar, Barcelona, Spain; Servicio de Cardiología, Hospital del Mar, Parc de Salut Mar, Barcelona, Spain
| |
Collapse
|
156
|
Biendarra‐Tiegs SM, Yechikov S, Shergill B, Brumback B, Takahashi K, Shirure VS, Gonzalez RE, Houshmand L, Zhong D, Weng K, Silva J, Smith TW, Rentschler SL, George SC. An iPS-derived in vitro model of human atrial conduction. Physiol Rep 2022; 10:e15407. [PMID: 36117385 PMCID: PMC9483613 DOI: 10.14814/phy2.15407] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 04/27/2022] [Accepted: 07/14/2022] [Indexed: 11/25/2022] Open
Abstract
Atrial fibrillation (AF) is the most common arrhythmia in the United States, affecting approximately 1 in 10 adults, and its prevalence is expected to rise as the population ages. Treatment options for AF are limited; moreover, the development of new treatments is hindered by limited (1) knowledge regarding human atrial electrophysiological endpoints (e.g., conduction velocity [CV]) and (2) accurate experimental models. Here, we measured the CV and refractory period, and subsequently calculated the conduction wavelength, in vivo (four subjects with AF and four controls), and ex vivo (atrial slices from human hearts). Then, we created an in vitro model of human atrial conduction using induced pluripotent stem (iPS) cells. This model consisted of iPS-derived human atrial cardiomyocytes plated onto a micropatterned linear 1D spiral design of Matrigel. The CV (34-41 cm/s) of the in vitro model was nearly five times faster than 2D controls (7-9 cm/s) and similar to in vivo (40-64 cm/s) and ex vivo (28-51 cm/s) measurements. Our iPS-derived in vitro model recapitulates key features of in vivo atrial conduction and may be a useful methodology to enhance our understanding of AF and model patient-specific disease.
Collapse
Affiliation(s)
| | - Sergey Yechikov
- Department of Biomedical EngineeringUniversity of California, DavisDavisCaliforniaUSA
| | - Bhupinder Shergill
- Department of Biomedical EngineeringUniversity of California, DavisDavisCaliforniaUSA
| | - Brittany Brumback
- Department of Biomedical EngineeringWashington University in St. LouisSt. LouisMissouriUSA
| | - Kentaro Takahashi
- Department of MedicineWashington University in St. LouisSt. LouisMissouriUSA
| | - Venktesh S. Shirure
- Department of Biomedical EngineeringUniversity of California, DavisDavisCaliforniaUSA
| | - Ruth Estelle Gonzalez
- Department of Biomedical EngineeringUniversity of California, DavisDavisCaliforniaUSA
| | - Laura Houshmand
- Department of Biomedical EngineeringUniversity of California, DavisDavisCaliforniaUSA
| | - Denise Zhong
- Department of Biomedical EngineeringUniversity of California, DavisDavisCaliforniaUSA
| | - Kuo‐Chan Weng
- Department of Biomedical EngineeringWashington University in St. LouisSt. LouisMissouriUSA
| | - Jon Silva
- Department of Biomedical EngineeringWashington University in St. LouisSt. LouisMissouriUSA
| | - Timothy W. Smith
- Department of MedicineWashington University in St. LouisSt. LouisMissouriUSA
| | - Stacey L. Rentschler
- Department of MedicineWashington University in St. LouisSt. LouisMissouriUSA
- Department of Developmental BiologyWashington University in St. LouisSt. LouisMissouriUSA
| | - Steven C. George
- Department of Biomedical EngineeringUniversity of California, DavisDavisCaliforniaUSA
| |
Collapse
|
157
|
Bruns F, Dobrev D. Assessment of atrial function by echocardiography: Can this be used to assess the arrhythmogenic atrial substrate? Int J Cardiol 2022; 362:190-191. [PMID: 35659557 DOI: 10.1016/j.ijcard.2022.05.070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 05/30/2022] [Indexed: 11/25/2022]
Affiliation(s)
- Florian Bruns
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, 45147 Essen, Germany; Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Duisburg-Essen, 45147 Essen, Germany
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, 45147 Essen, Germany; Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, USA; Montréal Heart Institute, University de Montréal, Montréal, Canada.
| |
Collapse
|
158
|
Bai J, Lu Y, Wang H, Zhao J. How synergy between mechanistic and statistical models is impacting research in atrial fibrillation. Front Physiol 2022; 13:957604. [PMID: 36111152 PMCID: PMC9468674 DOI: 10.3389/fphys.2022.957604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Atrial fibrillation (AF) with multiple complications, high morbidity and mortality, and low cure rates, has become a global public health problem. Although significant progress has been made in the treatment methods represented by anti-AF drugs and radiofrequency ablation, the therapeutic effect is not as good as expected. The reason is mainly because of our lack of understanding of AF mechanisms. This field has benefited from mechanistic and (or) statistical methodologies. Recent renewed interest in digital twin techniques by synergizing between mechanistic and statistical models has opened new frontiers in AF analysis. In the review, we briefly present findings that gave rise to the AF pathophysiology and current therapeutic modalities. We then summarize the achievements of digital twin technologies in three aspects: understanding AF mechanisms, screening anti-AF drugs and optimizing ablation strategies. Finally, we discuss the challenges that hinder the clinical application of the digital twin heart. With the rapid progress in data reuse and sharing, we expect their application to realize the transition from AF description to response prediction.
Collapse
Affiliation(s)
- Jieyun Bai
- Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Information Technology, Jinan University, Guangzhou, China
- College of Information Science and Technology, Jinan University, Guangzhou, China
| | - Yaosheng Lu
- Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Information Technology, Jinan University, Guangzhou, China
- College of Information Science and Technology, Jinan University, Guangzhou, China
| | - Huijin Wang
- College of Information Science and Technology, Jinan University, Guangzhou, China
| | - Jichao Zhao
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| |
Collapse
|
159
|
Sun M, Chen Z, Song Y, Zhang B, Yang J, Tan H. PLXND1-mediated calcium dyshomeostasis impairs endocardial endothelial autophagy in atrial fibrillation. Front Physiol 2022; 13:960480. [PMID: 36017337 PMCID: PMC9395636 DOI: 10.3389/fphys.2022.960480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 07/14/2022] [Indexed: 11/26/2022] Open
Abstract
Left atrial appendage (LAA) thrombus detachment resulting in intracranial embolism is a major complication of atrial fibrillation (AF). Endocardial endothelial cell (EEC) injury leads to thrombosis, whereas autophagy protects against EEC dysfunction. However, the role and underlying mechanisms of autophagy in EECs during AF have not been elucidated. In this study, we isolated EECs from AF model mice and observed reduced autophagic flux and intracellular calcium concentrations in EECs from AF mice. In addition, we detected an increased expression of the mechanosensitive protein PLXND1 in the cytomembranes of EECs. PLXND1 served as a scaffold protein to bind with ORAI1 and further decreased ORAI1-mediated calcium influx. The decrease in the calcium influx-mediated phosphorylation of CAMK2 is associated with the inhibition of autophagy, which results in EEC dysfunction in AF. Our study demonstrated that the change in PLXND1 expression contributes to intracellular calcium dyshomeostasis, which inhibits autophagy flux and results in EEC dysfunction in AF. This study provides a potential intervention target for EEC dysfunction to prevent and treat intracardiac thrombosis in AF and its complications.
Collapse
Affiliation(s)
- Mengjia Sun
- Institute of Cardiovascular Diseases of PLA, the Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- Department of Cardiology, the Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhen Chen
- Institute of Cardiovascular Diseases of PLA, the Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- Department of Cardiology, the Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yuanbin Song
- Institute of Cardiovascular Diseases of PLA, the Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- Department of Cardiology, the Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Bo Zhang
- Institute of Cardiovascular Diseases of PLA, the Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- Department of Cardiology, the Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jie Yang
- Institute of Cardiovascular Diseases of PLA, the Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- Department of Cardiology, the Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- *Correspondence: Jie Yang, ; Hu Tan,
| | - Hu Tan
- Institute of Cardiovascular Diseases of PLA, the Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- Department of Cardiology, the Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- *Correspondence: Jie Yang, ; Hu Tan,
| |
Collapse
|
160
|
Dong Y, Xiao S, He J, Shi K, Chen S, Liu D, Huang B, Zhai Z, Li J. Angiotensin receptor-neprilysin inhibitor therapy and recurrence of atrial fibrillation after radiofrequency catheter ablation: A propensity-matched cohort study. Front Cardiovasc Med 2022; 9:932780. [PMID: 35990986 PMCID: PMC9386595 DOI: 10.3389/fcvm.2022.932780] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 06/30/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundCompared with conventional medicines, angiotensin receptor-neprilysin inhibitor (ARNI) could further improve the prognosis for multiple cardiovascular diseases, such as heart failure, hypertension, and myocardial infarction. However, the relationship between ARNI therapy and the recurrence of atrial fibrillation (AF) after radiofrequency catheter ablation is currently unknown.MethodsThis study is a retrospective cohort study. Patients with consecutive persistent or paroxysmal AF undergoing first-time radiofrequency ablation were enrolled from February 2018 to October 2021. We compared the risk of AF recurrence in patients with catheter ablation who received ARNI with the risk of AF recurrence in those who received the angiotensin-converting enzyme inhibitor (ACEI). The propensity-score matched analysis was conducted to examine the effectiveness of ARNI. We used a Cox regression model to evaluate AF recurrence events.ResultsAmong 679 eligible patients, 155 patients with ARNI treatment and 155 patients with ACEI treatment were included in the analyses. At a median follow-up of 228 (196–322) days, ARNI as compared with ACEI was associated with a lower risk of AF recurrence [adjusted hazard ratio (HR), 0.39; 95% confidence interval (CI), 0.24–0.63; p < 0.001]. In addition, no interaction was found in the subgroup analysis.ConclusionAngiotensin receptor-neprilysin inhibitor treatment was associated with a decreased risk of AF recurrence after first-time radiofrequency catheter ablation.
Collapse
|
161
|
Xie D, Xiong K, Su X, Wang G, Wang L, Zou Q, Zhang C, Cao Y, Liu Y, Chen YH. Memantine targets glutamate receptors in atrial cardiomyocytes to prevent and treat atrial fibrillation. Cell Discov 2022; 8:76. [PMID: 35918317 PMCID: PMC9345967 DOI: 10.1038/s41421-022-00429-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 06/02/2022] [Indexed: 11/20/2022] Open
Affiliation(s)
- Duanyang Xie
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Tongji University School of Medicine, Shanghai, China
- Institute of Medical Genetics, Tongji University, Shanghai, China
| | - Ke Xiong
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Tongji University School of Medicine, Shanghai, China
- Institute of Medical Genetics, Tongji University, Shanghai, China
| | - Xuling Su
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Tongji University School of Medicine, Shanghai, China
- Institute of Medical Genetics, Tongji University, Shanghai, China
| | - Guanghua Wang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Tongji University School of Medicine, Shanghai, China
- Institute of Medical Genetics, Tongji University, Shanghai, China
| | - Luxin Wang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Tongji University School of Medicine, Shanghai, China
- Institute of Medical Genetics, Tongji University, Shanghai, China
| | - Qicheng Zou
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Tongji University School of Medicine, Shanghai, China
- Institute of Medical Genetics, Tongji University, Shanghai, China
| | - Caihong Zhang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Tongji University School of Medicine, Shanghai, China
- Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Yuting Cao
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Tongji University School of Medicine, Shanghai, China
- Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Yi Liu
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Tongji University School of Medicine, Shanghai, China
- Institute of Medical Genetics, Tongji University, Shanghai, China
| | - Yi-Han Chen
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Tongji University School of Medicine, Shanghai, China.
- Institute of Medical Genetics, Tongji University, Shanghai, China.
- Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai, China.
- Research Units of Origin and Regulation of Heart Rhythm, Chinese Academy of Medical Sciences, Shanghai, China.
| |
Collapse
|
162
|
CaMKII inhibition protects against hyperthyroid arrhythmias and adverse myocardial remodeling. Biochem Biophys Res Commun 2022; 615:136-142. [PMID: 35617800 DOI: 10.1016/j.bbrc.2022.04.082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 04/08/2022] [Accepted: 04/18/2022] [Indexed: 11/24/2022]
Abstract
Hyperthyroidism can potentiate arrhythmias and cardiac hypertrophy, whereas Ca2+/calmodulin-dependent kinase II (CaMKII) promotes maladaptive myocardial remodeling. However, it remains unclear whether CaMKII contributes to the progression of hyperthyroid heart disease (HHD). This study demonstrated that CaMKII inhibition can relieve adverse myocardial remodeling and reduce sinus tachycardia, isoproterenol-induced atrial fibrillation, and ventricular arrhythmias in hyperthyroid mice with preserved heart function. Hyperthyroid cardiac hypertrophy was promoted by CaMKII upregulation-induced HDAC4/MEF2a activation. Briefly, CaMKII inhibition benefits HHD management greatly in mice by preventing arrhythmias and maladaptive remodeling.
Collapse
|
163
|
Zuo K, Fang C, Liu Z, Fu Y, Liu Y, Liu L, Wang Y, Yin X, Liu X, Li J, Zhong J, Chen M, Xu L, Yang X. Commensal microbe-derived SCFA alleviates atrial fibrillation via GPR43/NLRP3 signaling. Int J Biol Sci 2022; 18:4219-4232. [PMID: 35844801 PMCID: PMC9274492 DOI: 10.7150/ijbs.70644] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 06/04/2022] [Indexed: 12/28/2022] Open
Abstract
Rationale: Dysbiotic gut microbiota (GM) and NLRP3 inflammasome are proarrhythmic factors in atrial fibrillation (AF). Herein, whether short-chain fatty acid (SCFA) produced from GM fermentation of dietary fiber serving as invisible mediators is yet unclear. Thus, the current study aimed to determine whether SCFA alleviated from NLRP3 signaling-mediated atrial remodeling protects AF development. Methods: First, a cross-sectional study based on the GC-MS metabolomics was performed to explore the association between fecal SCFA levels and AF traits in a cohort consisted of 48 individuals. Then, a well-established mice model fed diet deficient or enriched in dietary fiber was established to elucidate the pathophysiological role of SCFA involved in AF susceptibility, atrial remodeling, and G-protein-coupled receptor 43 (GPR43)/NLRP3 signaling. Finally, the effects of SCFA were verified on HL-1 cells. Results: Fecal SCFA levels were remarkably reduced in AF patients with a declining trend from paroxysmal to persistent AF. Prolonged P wave duration based on surface ECG and increased left atrial diameter gained from echocardiography was identified in low-fiber diet mice but lost in SCFA-supplemented group. Lack of dietary fiber enhanced susceptibility to AF under burst pacing, whereas SCFA might exert a protective effect. The supplementation of SCFA prevented dietary fiber deficiency-upregulated phosphorylation of calmodulin-dependent protein kinase II and ryanodine receptor 2, the disarray fibrosis, collagen expression, and NLRP3 inflammasome activation in atrial tissue. Finally, the AF protective roles of SCFA were identified through GPR43 mediated deactivation of NLRP3 by GPR43 knockdown in HL-1 cells. Conclusions: SCFA derived from dietary fiber fermentation by gut commensals alleviates AF development via GPR43/NLRP3 signaling.
Collapse
Affiliation(s)
- Kun Zuo
- Heart Center & Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Chen Fang
- Heart Center & Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Zheng Liu
- Heart Center & Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Yuan Fu
- Heart Center & Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Ye Liu
- Heart Center & Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Lifeng Liu
- Heart Center & Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Yuxing Wang
- Heart Center & Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Xiandong Yin
- Heart Center & Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Xiaoqing Liu
- Heart Center & Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Jing Li
- Heart Center & Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Jiuchang Zhong
- Heart Center & Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Mulei Chen
- Heart Center & Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Li Xu
- Heart Center & Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Xinchun Yang
- Heart Center & Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| |
Collapse
|
164
|
Cunha PS, Laranjo S, Heijman J, Oliveira MM. The Atrium in Atrial Fibrillation - A Clinical Review on How to Manage Atrial Fibrotic Substrates. Front Cardiovasc Med 2022; 9:879984. [PMID: 35859594 PMCID: PMC9289204 DOI: 10.3389/fcvm.2022.879984] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 06/03/2022] [Indexed: 12/27/2022] Open
Abstract
Atrial fibrillation (AF) is the most common sustained arrhythmia in the population and is associated with a significant clinical and economic burden. Rigorous assessment of the presence and degree of an atrial arrhythmic substrate is essential for determining treatment options, predicting long-term success after catheter ablation, and as a substrate critical in the pathophysiology of atrial thrombogenesis. Catheter ablation of AF has developed into an essential rhythm-control strategy. Nowadays is one of the most common cardiac ablation procedures performed worldwide, with its success inversely related to the extent of atrial structural disease. Although atrial substrate evaluation remains complex, several diagnostic resources allow for a more comprehensive assessment and quantification of the extent of left atrial structural remodeling and the presence of atrial fibrosis. In this review, we summarize the current knowledge on the pathophysiology, etiology, and electrophysiological aspects of atrial substrates promoting the development of AF. We also describe the risk factors for its development and how to diagnose its presence using imaging, electrocardiograms, and electroanatomic voltage mapping. Finally, we discuss recent data regarding fibrosis biomarkers that could help diagnose atrial fibrotic substrates.
Collapse
Affiliation(s)
- Pedro Silva Cunha
- Arrhythmology, Pacing and Electrophysiology Unit, Cardiology Service, Santa Marta Hospital, Central Lisbon Hospital University Center, Lisbon, Portugal
- Lisbon School of Medicine, Universidade de Lisboa, Lisbon, Portugal
- Comprehensive Health Research Center, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Sérgio Laranjo
- Arrhythmology, Pacing and Electrophysiology Unit, Cardiology Service, Santa Marta Hospital, Central Lisbon Hospital University Center, Lisbon, Portugal
- Lisbon School of Medicine, Universidade de Lisboa, Lisbon, Portugal
- Comprehensive Health Research Center, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Jordi Heijman
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - Mário Martins Oliveira
- Arrhythmology, Pacing and Electrophysiology Unit, Cardiology Service, Santa Marta Hospital, Central Lisbon Hospital University Center, Lisbon, Portugal
- Lisbon School of Medicine, Universidade de Lisboa, Lisbon, Portugal
- Comprehensive Health Research Center, Universidade NOVA de Lisboa, Lisbon, Portugal
| |
Collapse
|
165
|
Saljic A, Grandi E, Dobrev D. TGF-β1-induced endothelial-mesenchymal transition: a potential contributor to fibrotic remodeling in atrial fibrillation? J Clin Invest 2022; 132:e161070. [PMID: 35775488 PMCID: PMC9246376 DOI: 10.1172/jci161070] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia worldwide, with an unmet therapeutic need. Fibrotic remodeling, in which collagen-producing atrial fibroblasts play a crucial role, substantially contributes to arrhythmia promotion and progression. In this issue of the JCI, Lai, Tsai, and co-authors reveal that TGF-β1 promoted endothelial-mesenchymal transition during AF and put forward the notion that, in the adult heart, atrial fibroblasts can originate from different cellular sources. These important findings extend our understanding of the origin, biology, and function of fibroblasts and offer possibilities for therapeutic targeting of fibrosis in AF.
Collapse
Affiliation(s)
- Arnela Saljic
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Eleonora Grandi
- Department of Pharmacology, University of California Davis, Davis, California, USA
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, USA
- Montréal Heart Institute and University de Montréal, Medicine and Research Center, Montréal, Quebec, Canada
| |
Collapse
|
166
|
Yu LM, Dong X, Xu YL, Zhou ZJ, Huang YT, Zhao JK, Xu DY, Xue XD, Zhao QS, Liu T, Yin ZT, Jiang H, Wang HS. Icariin attenuates excessive alcohol consumption-induced susceptibility to atrial fibrillation through SIRT3 signaling. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166483. [DOI: 10.1016/j.bbadis.2022.166483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 06/25/2022] [Accepted: 06/29/2022] [Indexed: 10/17/2022]
|
167
|
Xia R, Tomsits P, Loy S, Zhang Z, Pauly V, Schüttler D, Clauss S. Cardiac Macrophages and Their Effects on Arrhythmogenesis. Front Physiol 2022; 13:900094. [PMID: 35812333 PMCID: PMC9257039 DOI: 10.3389/fphys.2022.900094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 05/30/2022] [Indexed: 12/24/2022] Open
Abstract
Cardiac electrophysiology is a complex system established by a plethora of inward and outward ion currents in cardiomyocytes generating and conducting electrical signals in the heart. However, not only cardiomyocytes but also other cell types can modulate the heart rhythm. Recently, cardiac macrophages were demonstrated as important players in both electrophysiology and arrhythmogenesis. Cardiac macrophages are a heterogeneous group of immune cells including resident macrophages derived from embryonic and fetal precursors and recruited macrophages derived from circulating monocytes from the bone marrow. Recent studies suggest antiarrhythmic as well as proarrhythmic effects of cardiac macrophages. The proposed mechanisms of how cardiac macrophages affect electrophysiology vary and include both direct and indirect interactions with other cardiac cells. In this review, we provide an overview of the different subsets of macrophages in the heart and their possible interactions with cardiomyocytes under both physiologic conditions and heart disease. Furthermore, we elucidate similarities and differences between human, murine and porcine cardiac macrophages, thus providing detailed information for researchers investigating cardiac macrophages in important animal species for electrophysiologic research. Finally, we discuss the pros and cons of mice and pigs to investigate the role of cardiac macrophages in arrhythmogenesis from a translational perspective.
Collapse
Affiliation(s)
- Ruibing Xia
- Department of Medicine I, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
| | - Philipp Tomsits
- Department of Medicine I, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
| | - Simone Loy
- Department of Medicine I, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
| | - Zhihao Zhang
- Department of Medicine I, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
| | - Valerie Pauly
- Department of Medicine I, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
| | - Dominik Schüttler
- Department of Medicine I, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
| | - Sebastian Clauss
- Department of Medicine I, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
| |
Collapse
|
168
|
Young LJ, Antwi-Boasiako S, Ferrall J, Wold LE, Mohler PJ, El Refaey M. Genetic and non-genetic risk factors associated with atrial fibrillation. Life Sci 2022; 299:120529. [PMID: 35385795 PMCID: PMC9058231 DOI: 10.1016/j.lfs.2022.120529] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/08/2022] [Accepted: 03/31/2022] [Indexed: 12/15/2022]
Abstract
Atrial fibrillation (AF) is the most common arrhythmic disorder and its prevalence in the United States is projected to increase to more than twelve million cases in 2030. AF increases the risk of other forms of cardiovascular disease, including stroke. As the incidence of atrial fibrillation increases dramatically with age, it is paramount to elucidate risk factors underlying AF pathogenesis. Here, we review tissue and cellular pathways underlying AF, as well as critical components that impact AF susceptibility including genetic and environmental risk factors. Finally, we provide the latest information on potential links between SARS-CoV-2 and human AF. Improved understanding of mechanistic pathways holds promise in preventative care and early diagnostics, and also introduces novel targeted forms of therapy that might attenuate AF progression and maintenance.
Collapse
Affiliation(s)
- Lindsay J Young
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
| | - Steve Antwi-Boasiako
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
| | - Joel Ferrall
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Loren E Wold
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA; College of Nursing, The Ohio State University, Columbus, OH, USA
| | - Peter J Mohler
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA; Department of Internal Medicine, Division of Cardiovascular Medicine, The Ohio State University, Columbus, OH, USA
| | - Mona El Refaey
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA; Department of Surgery, Division of Cardiac Surgery, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
169
|
Cardiac Fibroblasts Promote Ferroptosis in Atrial Fibrillation by Secreting Exo-miR-23a-3p Targeting SLC7A11. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3961495. [PMID: 35677105 PMCID: PMC9168132 DOI: 10.1155/2022/3961495] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 04/14/2022] [Accepted: 05/05/2022] [Indexed: 12/28/2022]
Abstract
The exact mechanism of atrial fibrillation (AF) has been not well elucidated. Ferroptosis is an iron-dependent cell death due to excessive accumulation of peroxidized polyunsaturated fatty acids. However, the molecular mechanism underlying AF and ferroptosis has never been reported. Here, we established the rapid pacing model in vivo and vitro to investigate the relationship between AF and ferroptosis. In canine model of rapid atrial pacing, the content of malondialdehyde and total ions in the atrial tissue of the Pacing group was significantly increased and the exosome inhibitor GW4869 reduced ferroptosis, fibrosis, and inflammation and improved histological and electrophysiological remodeling. In rapid pacing h9c2 cells, the expression of antioxidative stress genes associated with ferroptosis presented sequential changes and proteins involved in ferroptosis such as FTH1, SLC7A11, and GPX4 were gradually depleted. Furthermore, pacing cardiac fibroblast-derived exosomes (CF-exos) exacerbated ferroptosis in h9c2 cells and pretreated pacing-CF-exos with GW4869 alleviated injury to h9c2 cells. In mechanism, our results demonstrated that pacing-CF-exos highly expressed miR-23a-3p by informatics analysis and experimental verification. Inhibitor-miR-23a-3p protected h9c2 cells from ferroptosis accompanying with upregulation of SLC7A11. In addition, SLC7A11 was shown to be the target gene of miR-23a-3p. In conclusion, our results suggest that CF-exos-miR-23a-3p may promote ferroptosis. The development of AF in a persistent direction could be prevented by intervening with exosomal miRNAs to reduce oxidative stress injury and ferroptosis.
Collapse
|
170
|
Atrial Fibrillation Global Changes after Pulmonary Vein and Posterior Wall Isolation: A Charge Density Mapping Study. J Clin Med 2022; 11:jcm11102948. [PMID: 35629074 PMCID: PMC9145946 DOI: 10.3390/jcm11102948] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/09/2022] [Accepted: 05/19/2022] [Indexed: 02/04/2023] Open
Abstract
Background: Non-contact charge density (CD) mapping allows a global visualization of left atrium (LA) activation and of activation patterns during atrial fibrillation (AF). The aim of this study was to analyze, with CD mapping, the changes in persistent AF induced by pulmonary vein isolation (PVI) and LA posterior wall isolation (LAPWI). Methods: Patients undergoing PVI + LAPWI using the Arctic Front Advance PROTM cryoballoon system were included in the study. CD maps were created during AF at baseline, after PVI and after LAPWI. Three distinct activation patterns were identified in the CD maps: localized irregular activation (LIA), localized rotational activation (LRA) and focal centrifugal activation (FCA). LA maps were divided into the following regions: anterior, septal, lateral, roof, posterior, inferior. Results: Eleven patients were included, with a total of 33 maps and 198 AF regions analyzed. Global and regional AF cycle lengths significantly increased after PVI and LAPWI. Baseline analysis demonstrated higher LIA, LRA and FCA numbers in the posterior and anterior regions. After PVI, there was no change in LIA, LRA and FCA occurrence. After PVI + LAPWI, a significant decrease in LRA was observed with no difference in LIA and FCA occurrence. In the regional analysis, there was a significant reduction in the LIA number in the inferior region, in the LRA number in the roof and posterior regions and in the FCA number in the lateral region. Conclusions: A global reduction in the LRA number was observed only after PVI + LAPWI; it was driven by a reduction in rotational activity in the roof and posterior regions.
Collapse
|
171
|
Younes R, LeBlanc CA, Hiram R. Evidence of Failed Resolution Mechanisms in Arrhythmogenic Inflammation, Fibrosis and Right Heart Disease. Biomolecules 2022; 12:biom12050720. [PMID: 35625647 PMCID: PMC9138906 DOI: 10.3390/biom12050720] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 12/14/2022] Open
Abstract
Inflammation is a complex program of active processes characterized by the well-orchestrated succession of an initiation and a resolution phase aiming to promote homeostasis. When the resolution of inflammation fails, the tissue undergoes an unresolved inflammatory status which, if it remains uncontrolled, can lead to chronic inflammatory disorders due to aggravation of structural damages, development of a fibrous area, and loss of function. Various human conditions show a typical unresolved inflammatory profile. Inflammatory diseases include cancer, neurodegenerative disease, asthma, right heart disease, atherosclerosis, myocardial infarction, or atrial fibrillation. New evidence has started to emerge on the role, including pro-resolution involvement of chemical mediators in the acute phase of inflammation. Although flourishing knowledge is available about the role of specialized pro-resolving mediators in neurodegenerative diseases, atherosclerosis, obesity, or hepatic fibrosis, little is known about their efficacy to combat inflammation-associated arrhythmogenic cardiac disorders. It has been shown that resolvins, including RvD1, RvE1, or Mar1, are bioactive mediators of resolution. Resolvins can stop neutrophil activation and infiltration, stimulate monocytes polarization into anti-inflammatory-M2-macrophages, and activate macrophage phagocytosis of inflammation-debris and neutrophils to promote efferocytosis and clearance. This review aims to discuss the paradigm of failed-resolution mechanisms (FRM) potentially promoting arrhythmogenicity in right heart disease-induced inflammatory status.
Collapse
Affiliation(s)
- Rim Younes
- Montreal Heart Institute (MHI), Montreal, QC H1T 1C8, Canada; (R.Y.); (C.-A.L.)
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Charles-Alexandre LeBlanc
- Montreal Heart Institute (MHI), Montreal, QC H1T 1C8, Canada; (R.Y.); (C.-A.L.)
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Roddy Hiram
- Montreal Heart Institute (MHI), Montreal, QC H1T 1C8, Canada; (R.Y.); (C.-A.L.)
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
- Correspondence: ; Tel.: +1-514-376-3330 (ext. 5015)
| |
Collapse
|
172
|
Small extracellular vesicles derived from patients with persistent atrial fibrillation exacerbate arrhythmogenesis via miR-30a-5p. Clin Sci (Lond) 2022; 136:621-637. [PMID: 35411927 DOI: 10.1042/cs20211141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/21/2022] [Accepted: 04/11/2022] [Indexed: 11/17/2022]
Abstract
Small extracellular vesicles (sEVs) are nanometer-sized membranous vesicles that contribute to the pathogenesis of atrial fibrillation (AF). Here, we investigated the role of sEVs derived from patients with persistent AF in the pathophysiology of AF. First, we evaluated the pathological effects of sEVs derived from the peripheral blood of patients with persistent AF (AF-sEVs). AF-sEVs treatment reduced cell viability, caused abnormal Ca2+ handling, induced reactive oxygen species (ROS) production, and led to increased CaMKII activation of non-paced and paced atrial cardiomyocytes. Next, we analyzed the miRNA profile of AF-sEVs to investigate which components of AF-sEVs promote arrhythmias, and we selected six miRNAs that correlated with CaMKII activation. qRT-PCR experiment identified that miR-30a-5p was significantly downregulated in AF-sEVs, paced cardiomyocytes, and atrial tissues of patients with persistent AF. CaMKII was predicted by bioinformatics analysis as a miR-30a-5p target gene and validated by a dual luciferase reporter; hence, we evaluated the effects of miR-30a-5p on paced cardiomyocytes and validated miR-30a-5p as a pro-arrhythmic signature of AF-sEVs. Consequently, AF-sEVs-loaded with miR-30a-5p attenuated pacing-induced Ca2+-handling abnormalities, whereas AF-sEVs-loaded with anti-miR-30a-5p reversed the change in paced cardiomyocytes. Taken together, the regulation of CaMKII by miR-30a-5p revealed that miR-30a-5p is a major mediator for AF-sEVs-mediated AF pathogenesis. Accordingly, these findings suggest that sEVs derived from patients with persistent AF exacerbate arrhythmogenesis via miR-30a-5p.
Collapse
|
173
|
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia despite substantial efforts to understand the pathophysiology of the condition and develop improved treatments. Identifying the underlying causative mechanisms of AF in individual patients is difficult and the efficacy of current therapies is suboptimal. Consequently, the incidence of AF is steadily rising and there is a pressing need for novel therapies. Research has revealed that defects in specific molecular pathways underlie AF pathogenesis, resulting in electrical conduction disorders that drive AF. The severity of this so-called electropathology correlates with the stage of AF disease progression and determines the response to AF treatment. Therefore, unravelling the molecular mechanisms underlying electropathology is expected to fuel the development of innovative personalized diagnostic tools and mechanism-based therapies. Moreover, the co-creation of AF studies with patients to implement novel diagnostic tools and therapies is a prerequisite for successful personalized AF management. Currently, various treatment modalities targeting AF-related electropathology, including lifestyle changes, pharmaceutical and nutraceutical therapy, substrate-based ablative therapy, and neuromodulation, are available to maintain sinus rhythm and might offer a novel holistic strategy to treat AF.
Collapse
Affiliation(s)
- Bianca J J M Brundel
- Department of Physiology, Amsterdam University Medical Centers, VU Universiteit, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands.
| | - Xun Ai
- Department of Physiology and Cell Biology, College of Medicine/Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | | | - Myrthe F Kuipers
- AFIPonline.org, Atrial Fibrillation Innovation Platform, Amsterdam, Netherlands
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | | |
Collapse
|
174
|
Farinha JM, Gupta D, Lip GYH. Frequent premature atrial contractions as a signalling marker of atrial cardiomyopathy, incident atrial fibrillation and stroke. Cardiovasc Res 2022; 119:429-439. [PMID: 35388889 PMCID: PMC10064848 DOI: 10.1093/cvr/cvac054] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 02/21/2022] [Accepted: 03/23/2022] [Indexed: 11/14/2022] Open
Abstract
Premature atrial contractions are a common cardiac phenomenon. Although previously considered a benign electrocardiographic finding, they have now been associated with a higher risk of incident atrial fibrillation and other adverse outcomes such as stroke and all-cause mortality. Since premature atrial contractions can be associated with these adverse clinical outcomes independently of atrial fibrillation occurrence, different explanations have being proposed. The concept of atrial cardiomyopathy, where atrial fibrillation would be an epiphenomenon outside the causal pathway between premature atrial contractions and stroke has received traction recently. This concept suggests that structural, functional and biochemical changes in the atria lead to arrhythmia occurrence and thromboembolic events. Some consensus about diagnosis and treatment of this condition have been published, but this is based on scarce evidence, highlighting the need for a clear definition of excessive premature atrial contractions and for prospective studies regarding antiarrhythmic therapies, anticoagulation or molecular targets in this group of patients.
Collapse
Affiliation(s)
- José Maria Farinha
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK
| | - Dhiraj Gupta
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
175
|
Marazzato J, Blasi F, Golino M, Verdecchia P, Angeli F, De Ponti R. Hypertension and Arrhythmias: A Clinical Overview of the Pathophysiology-Driven Management of Cardiac Arrhythmias in Hypertensive Patients. J Cardiovasc Dev Dis 2022; 9:jcdd9040110. [PMID: 35448086 PMCID: PMC9025699 DOI: 10.3390/jcdd9040110] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/26/2022] [Accepted: 04/02/2022] [Indexed: 02/06/2023] Open
Abstract
Because of demographic aging, the prevalence of arterial hypertension (HTN) and cardiac arrhythmias, namely atrial fibrillation (AF), is progressively increasing. Not only are these clinical entities strongly connected, but, acting with a synergistic effect, their association may cause a worse clinical outcome in patients already at risk of ischemic and/or haemorrhagic stroke and, consequently, disability and death. Despite the well-known association between HTN and AF, several pathogenetic mechanisms underlying the higher risk of AF in hypertensive patients are still incompletely known. Although several trials reported the overall clinical benefit of renin–angiotensin–aldosterone inhibitors in reducing incident AF in HTN, the role of this class of drugs is greatly reduced when AF diagnosis is already established, thus hinting at the urgent need for primary prevention measures to reduce AF occurrence in these patients. Through a thorough review of the available literature in the field, we investigated the basic mechanisms through which HTN is believed to promote AF, summarising the evidence supporting a pathophysiology-driven approach to prevent this arrhythmia in hypertensive patients, including those suffering from primary aldosteronism, a non-negligible and under-recognised cause of secondary HTN. Finally, in the hazy scenario of AF screening in hypertensive patients, we reviewed which patients should be screened, by which modality, and who should be offered oral anticoagulation for stroke prevention.
Collapse
Affiliation(s)
- Jacopo Marazzato
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (J.M.); (F.B.); (M.G.); (F.A.)
| | - Federico Blasi
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (J.M.); (F.B.); (M.G.); (F.A.)
| | - Michele Golino
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (J.M.); (F.B.); (M.G.); (F.A.)
| | - Paolo Verdecchia
- Fondazione Umbra Cuore e Ipertensione-ONLUS, 06100 Perugia, Italy;
- Division of Cardiology, Hospital S. Maria della Misericordia, 06100 Perugia, Italy
| | - Fabio Angeli
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (J.M.); (F.B.); (M.G.); (F.A.)
- Department of Medicine and Cardiopulmonary Rehabilitation, Maugeri Care and Research Institute, IRCCS Tradate, 21049 Tradate, Italy
| | - Roberto De Ponti
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (J.M.); (F.B.); (M.G.); (F.A.)
- Correspondence: ; Tel.: +39-0332278934
| |
Collapse
|
176
|
Gawałko M, Dobrev D. Fat chance for POAF? Pericardial adipose tissue and the arrhythmogenic substrate for postoperative atrial fibrillation. IJC HEART & VASCULATURE 2022; 39:101000. [PMID: 35402693 PMCID: PMC8984633 DOI: 10.1016/j.ijcha.2022.101000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 03/08/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Monika Gawałko
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Germany
- 1 Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Germany
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Canada
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, USA
| |
Collapse
|
177
|
Benito B, García-Elías A, Ois Á, Tajes M, Vallès E, Ble M, Yáñez Bisbe L, Giralt-Steinhauer E, Rodríguez-Campello A, Cladellas Capdevila M, Martí-Almor J, Roquer J, Cuadrado-Godia E. La concentración plasmática de microARN-1-3p se asocia con fibrilación auricular subclínica en los pacientes con ictus criptogénico. Rev Esp Cardiol 2022. [DOI: 10.1016/j.recesp.2021.11.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
178
|
Wang F, Liang Q, Ma Y, Sun M, Li T, Lin L, Sun Z, Duan J. Silica nanoparticles induce pyroptosis and cardiac hypertrophy via ROS/NLRP3/Caspase-1 pathway. Free Radic Biol Med 2022; 182:171-181. [PMID: 35219847 DOI: 10.1016/j.freeradbiomed.2022.02.027] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/21/2022] [Accepted: 02/22/2022] [Indexed: 12/11/2022]
Abstract
Growing literatures suggest that silica nanoparticles (SiNPs) exposure is correlated with adverse cardiovascular effects. Cardiac hypertrophy is one of the most common risk factors for heart failure. However, whether SiNPs involved in cardiac hypertrophy and the underlying mechanisms was remained unexploited. Our study aimed to investigate the molecular mechanisms of SiNPs on pyroptosis and cardiac hypertrophy. The in vivo results found that SiNPs induced ultrastructural change and histopathological damage, accompanied by oxidative damage occurred and increased levels of inflammatory factors (IL-18 and IL-1β) in heart tissue. In addition, SiNPs could upregulate the expressions of cardiac hypertrophy-related special marker including ANP, BNP, β-MHC, it also elevated the pyroptosis-related protein, such as NLRP3, Cleaved-Caspase-1, GSDMD, IL-18 and Cleaved-IL-1β in vivo. For in vitro study, SiNPs increased the intracellular ROS generation and activated the NLRP3/Caspase-1/GSDMD signaling pathway in cardiomyocytes. Whereas, the NADPH oxidase (NOX) inhibitor VAS2870 had effectively inhibited the ROS level and suppressed the expression of NLRP3, ASC, Pro-Caspase-1, Cleaved-Caspase-1, N-GSDMD, IL-18, Cleaved-IL-1β, ANP, BNP and β-MHC. Moreover, transfected with si-NLRP3 or adopted with Caspase-1 inhibitor VX-765 in cardiomyocytes showed an inhibitory effect on SiNPs-induced pyroptosis and cardiac hypertrophy. In summary, our results demonstrated that SiNPs could trigger pyroptosis and cardiac hypertrophy via ROS/NLRP3/Caspase-1 signaling pathway.
Collapse
Affiliation(s)
- Fenghong Wang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China; Sinopharm North Hospital, Baotou, 014040, PR China
| | - Qingqing Liang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Yuexiao Ma
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Mengqi Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Tianyu Li
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Lisen Lin
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China.
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China.
| |
Collapse
|
179
|
Fang Y, Li Q, Li X, Luo GH, Kuang SJ, Luo XS, Li QQ, Yang H, Liu Y, Deng CY, Xue YM, Wu SL, Rao F. Piezo1 Participated in Decreased L-Type Calcium Current Induced by High Hydrostatic Pressure via. CaM/Src/Pitx2 Activation in Atrial Myocytes. Front Cardiovasc Med 2022; 9:842885. [PMID: 35252406 PMCID: PMC8891577 DOI: 10.3389/fcvm.2022.842885] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 01/18/2022] [Indexed: 01/25/2023] Open
Abstract
Hypertension is a major cardiovascular risk factor for atrial fibrillation (AF) worldwide. However, the role of mechanical stress caused by hypertension on downregulating the L-type calcium current (ICa,L), which is vital for AF occurrence, remains unclear. Therefore, the aim of the present study was to investigate the role of Piezo1, a mechanically activated ion channel, in the decrease of ICa,L in response to high hydrostatic pressure (HHP, one of the principal mechanical stresses) at 40 mmHg, and to elucidate the underlying pathways. Experiments were conducted using left atrial appendages from patients with AF, spontaneously hypertensive rats (SHRs) treated with valsartan (Val) at 30 mg/kg/day and atrium-derived HL-1 cells exposed to HHP. The protein expression levels of Piezo1, Calmodulin (CaM), and Src increased, while that of the L-type calcium channel a1c subunit protein (Cav1.2) decreased in the left atrial tissue of AF patients and SHRs. SHRs were more vulnerable to AF, with decreased ICa,L and shortened action potential duration, which were ameliorated by Val treatment. Validation of these results in HL-1 cells in the context of HHP also demonstrated that Piezo1 is required for the decrease of ICa,L by regulating Ca2+ transient and activating CaM/Src pathway to increase the expression of paired like homeodomain-2 (Pitx2) in atrial myocytes. Together, these data demonstrate that HHP stimulation increases AF susceptibility through Piezo1 activation, which is required for the decrease of ICa,Lvia. the CaM/Src/Pitx2 pathway in atrial myocytes.
Collapse
Affiliation(s)
- Yuan Fang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Qian Li
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xin Li
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Guan-Hao Luo
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Su-Juan Kuang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xue-Shan Luo
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Qiao-Qiao Li
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Hui Yang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yang Liu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Chun-Yu Deng
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yu-Mei Xue
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- *Correspondence: Yu-Mei Xue
| | - Shu-Lin Wu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Shu-Lin Wu
| | - Fang Rao
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Fang Rao
| |
Collapse
|
180
|
Hohl M, Selejan SR, Wintrich J, Lehnert U, Speer T, Schneider C, Mauz M, Markwirth P, Wong DWL, Boor P, Kazakov A, Mollenhauer M, Linz B, Klinkhammer BM, Hübner U, Ukena C, Moellmann J, Lehrke M, Wagenpfeil S, Werner C, Linz D, Mahfoud F, Böhm M. Renal Denervation Prevents Atrial Arrhythmogenic Substrate Development in CKD. Circ Res 2022; 130:814-828. [PMID: 35130718 DOI: 10.1161/circresaha.121.320104] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND In patients with chronic kidney disease (CKD), atrial fibrillation (AF) is highly prevalent and represents a major risk factor for stroke and death. CKD is associated with atrial proarrhythmic remodeling and activation of the sympathetic nervous system. Whether reduction of the sympathetic nerve activity by renal denervation (RDN) inhibits AF vulnerability in CKD is unknown. METHODS Left atrial (LA) fibrosis was analyzed in samples from patients with AF and concomitant CKD (estimated GFR, <60 mL/min per 1.73 m2) using picrosirius red and compared with AF patients without CKD and patients with sinus rhythm with and without CKD. In a translational approach, male Sprague Dawley rats were fed with 0.25% adenine (AD)-containing chow for 16 weeks to induce CKD. At week 5, AD-fed rats underwent RDN or sham operation (AD). Rats on normal chow served as control. After 16 weeks, cardiac function and AF susceptibility were assessed by echocardiography, radiotelemetry, electrophysiological mapping, and burst stimulation, respectively. LA tissue was histologically analyzed for sympathetic innervation using tyrosine hydroxylase staining, and LA fibrosis was determined using picrosirius red. RESULTS Sirius red staining demonstrated significantly increased LA fibrosis in patients with AF+CKD compared with AF without CKD or sinus rhythm. In rats, AD demonstrated LA structural changes with enhanced sympathetic innervation compared with control. In AD, LA enlargement was associated with prolonged duration of induced AF episodes, impaired LA conduction latency, and increased absolute conduction inhomogeneity. RDN treatment improved LA remodeling and reduced LA diameter compared with sham-operated AD. Furthermore, RDN decreased AF susceptibility and ameliorated LA conduction latency and absolute conduction inhomogeneity, independent of blood pressure reduction and renal function. CONCLUSIONS In an experimental rat model of CKD, RDN inhibited progression of atrial structural and electrophysiological remodeling. Therefore, RDN represents a potential therapeutic tool to reduce the risk of AF in CKD, independent of changes in renal function and blood pressure.
Collapse
Affiliation(s)
- Mathias Hohl
- Department of Internal Medicine III, Saarland University Hospital and Saarland University, Homburg/Saar, Germany (M.H., S.-R.S., J.W., U.L., C.S., M. Mauz, P.M., A.K., C.U., C.W., D.L., F.M., M.B.)
| | - Simina-Ramona Selejan
- Department of Internal Medicine III, Saarland University Hospital and Saarland University, Homburg/Saar, Germany (M.H., S.-R.S., J.W., U.L., C.S., M. Mauz, P.M., A.K., C.U., C.W., D.L., F.M., M.B.)
| | - Jan Wintrich
- Department of Internal Medicine III, Saarland University Hospital and Saarland University, Homburg/Saar, Germany (M.H., S.-R.S., J.W., U.L., C.S., M. Mauz, P.M., A.K., C.U., C.W., D.L., F.M., M.B.)
| | - Ulrike Lehnert
- Department of Internal Medicine III, Saarland University Hospital and Saarland University, Homburg/Saar, Germany (M.H., S.-R.S., J.W., U.L., C.S., M. Mauz, P.M., A.K., C.U., C.W., D.L., F.M., M.B.)
| | - Thimoteus Speer
- Klinik für Innere Medizin IV, Universität des Saarlandes, Homburg/Saar, Germany (T.S.).,Translational Cardio-Renal Medicine, Saarland University, Homburg/Saar, Germany. (T.S.)
| | - Clara Schneider
- Department of Internal Medicine III, Saarland University Hospital and Saarland University, Homburg/Saar, Germany (M.H., S.-R.S., J.W., U.L., C.S., M. Mauz, P.M., A.K., C.U., C.W., D.L., F.M., M.B.)
| | - Muriel Mauz
- Department of Internal Medicine III, Saarland University Hospital and Saarland University, Homburg/Saar, Germany (M.H., S.-R.S., J.W., U.L., C.S., M. Mauz, P.M., A.K., C.U., C.W., D.L., F.M., M.B.)
| | - Philipp Markwirth
- Department of Internal Medicine III, Saarland University Hospital and Saarland University, Homburg/Saar, Germany (M.H., S.-R.S., J.W., U.L., C.S., M. Mauz, P.M., A.K., C.U., C.W., D.L., F.M., M.B.)
| | - Dickson W L Wong
- Institut für Pathologie Universitätsklinikum Aachen, Germany (D.W.L.W., P.B., B.M.K.)
| | - Peter Boor
- Institut für Pathologie Universitätsklinikum Aachen, Germany (D.W.L.W., P.B., B.M.K.)
| | - Andrey Kazakov
- Department of Internal Medicine III, Saarland University Hospital and Saarland University, Homburg/Saar, Germany (M.H., S.-R.S., J.W., U.L., C.S., M. Mauz, P.M., A.K., C.U., C.W., D.L., F.M., M.B.)
| | - Martin Mollenhauer
- Faculty of Medicine and University Hospital Cologne, Clinic III for Internal Medicine, University of Cologne, Germany (M. Mollenhauer)
| | - Benedikt Linz
- Faculty of Health and Medical Sciences, Department of Biomedical Sciences, University of Copenhagen, Denmark (B.L.)
| | | | - Ulrich Hübner
- Department of Clinical Chemistry and Laboratory Medicine, Saarland University Hospital, Homburg/Saar, Germany (U.H.)
| | - Christian Ukena
- Department of Internal Medicine III, Saarland University Hospital and Saarland University, Homburg/Saar, Germany (M.H., S.-R.S., J.W., U.L., C.S., M. Mauz, P.M., A.K., C.U., C.W., D.L., F.M., M.B.)
| | - Julia Moellmann
- Department of Internal Medicine I-Cardiology, University Hospital Aachen, Germany (J.M., M.L.)
| | - Michael Lehrke
- Department of Internal Medicine I-Cardiology, University Hospital Aachen, Germany (J.M., M.L.)
| | - Stefan Wagenpfeil
- Institut für Medizinische Biometrie, Epidemiologie und Medizinische Informatik, Saarland University, Homburg/Saar, Germany. (S.W.)
| | - Christian Werner
- Department of Internal Medicine III, Saarland University Hospital and Saarland University, Homburg/Saar, Germany (M.H., S.-R.S., J.W., U.L., C.S., M. Mauz, P.M., A.K., C.U., C.W., D.L., F.M., M.B.)
| | - Dominik Linz
- Department of Internal Medicine III, Saarland University Hospital and Saarland University, Homburg/Saar, Germany (M.H., S.-R.S., J.W., U.L., C.S., M. Mauz, P.M., A.K., C.U., C.W., D.L., F.M., M.B.).,Cardiovascular Research Institute Maastricht, University Maastricht, the Netherlands (D.L.)
| | - Felix Mahfoud
- Department of Internal Medicine III, Saarland University Hospital and Saarland University, Homburg/Saar, Germany (M.H., S.-R.S., J.W., U.L., C.S., M. Mauz, P.M., A.K., C.U., C.W., D.L., F.M., M.B.)
| | - Michael Böhm
- Department of Internal Medicine III, Saarland University Hospital and Saarland University, Homburg/Saar, Germany (M.H., S.-R.S., J.W., U.L., C.S., M. Mauz, P.M., A.K., C.U., C.W., D.L., F.M., M.B.)
| |
Collapse
|
181
|
Tu T, Li B, Li X, Zhang B, Xiao Y, Li J, Qin F, Liu N, Sun C, Liu Q, Zhou S. Dietary ω-3 fatty acids reduced atrial fibrillation vulnerability via attenuating myocardial endoplasmic reticulum stress and inflammation in a canine model of atrial fibrillation. J Cardiol 2022; 79:194-201. [PMID: 34702603 DOI: 10.1016/j.jjcc.2021.08.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 07/08/2021] [Accepted: 08/16/2021] [Indexed: 01/08/2023]
Abstract
BACKGROUND Dietary consumption of ω-3 fatty acids is correlated with a reduced incidence of cardiovascular events. Here, we investigated the effect of dietary ω-3 fatty acids on atrial fibrillation (AF) vulnerability in a canine model of AF and explored the related mechanisms. METHODS Twenty four male beagle dogs (weight, 8-10 kg) were randomly divided into four groups: (a) sham-operated group (normal chow); (b) AF+FO [AF and normal chow supplemented with fish oil (FO): 0.6 g n-3 polyunsaturated fatty acids (ω-3 PUFA) /kg/day]; (c) AF group (normal chow); (d) sham-operated FO group (chow supplemented with FO: 0.6 g ω-3 PUFA/kg/day). AF was induced by rapid atrial pacing (RAP: 400 bpm for 4 weeks). Daily oral administration of FO was initiated 1 week before surgery and continued for 4 weeks post operation. RESULTS Atrial electric remodeling was significantly attenuated and AF vulnerability were significantly reduced in AF+FO group compared to AF group. Endoplasmic reticulum (ER) stress-related protein expression levels of glucose-regulated protein78, C/EBP homologous protein, cleaved-Caspase12, and phosphorylation of protein kinase R-like ER kinase as well as inflammatory cytokines interleukin-1β, interleukin-6, tumor necrosis factor-α in left atrium (LA) were significantly downregulated in AF+FO group than in AF group (all p<0.05). In addition, Masson staining revealed lower extent of LA interstitial fibrosis in AF+FO group than in AF group (p<0.01). Myocardial apoptosis was also significantly reduced in AF+FO group than in AF group (p<0.05). CONCLUSIONS Dietary ω-3 fatty acids could significantly reduce RAP-induced AF vulnerability, possibly via attenuating myocardial ER stress, inflammation, and apoptosis in this canine model of AF.
Collapse
Affiliation(s)
- Tao Tu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Biao Li
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China; Shenzhen Traditional Chinese Medicine Hospital, 1 Fuhua Rd, Futian District, Shenzhen, Guangdong Province, China
| | - Xuping Li
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Baojian Zhang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China; Department of Cardiology, the Affiliated Chinese Medicine Hospital of Xinjiang Medical University, Urumqi City, Xinjiang, PR China
| | - Yichao Xiao
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Jiayi Li
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Fen Qin
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Na Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Chao Sun
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Qiming Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China.
| | - Shenghua Zhou
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China.
| |
Collapse
|
182
|
Greene D, Kaboudian A, Wasserstrom JA, Fenton FH, Shiferaw Y. Voltage-mediated mechanism for calcium wave synchronization and arrhythmogenesis in atrial tissue. Biophys J 2022; 121:383-395. [PMID: 34968425 PMCID: PMC8822619 DOI: 10.1016/j.bpj.2021.12.040] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/28/2021] [Accepted: 12/23/2021] [Indexed: 02/03/2023] Open
Abstract
A wide range of atrial arrythmias are caused by molecular defects in proteins that regulate calcium (Ca) cycling. In many cases, these defects promote the propagation of subcellular Ca waves in the cell, which can perturb the voltage time course and induce dangerous perturbations of the action potential (AP). However, subcellular Ca waves occur randomly in cells and, therefore, electrical coupling between cells substantially decreases their effect on the AP. In this study, we present evidence that Ca waves in atrial tissue can synchronize in-phase owing to an order-disorder phase transition. In particular, we show that, below a critical pacing rate, Ca waves are desynchronized and therefore do not induce substantial AP fluctuations in tissue. However, above this critical pacing rate, Ca waves gradually synchronize over millions of cells, which leads to a dramatic amplification of AP fluctuations. We exploit an underlying Ising symmetry of paced cardiac tissue to show that this transition exhibits universal properties common to a wide range of physical systems in nature. Finally, we show that in the heart, phase synchronization induces spatially out-of-phase AP duration alternans which drives wave break and reentry. These results suggest that cardiac tissue exhibits a phase transition that is required for subcellular Ca cycling defects to induce a life-threatening arrhythmia.
Collapse
Affiliation(s)
- D'Artagnan Greene
- Department of Physics and Astronomy, California State University, Northridge, California
| | - Abouzar Kaboudian
- School of Physics, Georgia Institute of Technology, Atlanta, Georgia
| | - John A Wasserstrom
- The Feinberg Cardiovascular and Renal Research Institute, Department of Medicine (Cardiology), Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Flavio H Fenton
- School of Physics, Georgia Institute of Technology, Atlanta, Georgia
| | - Yohannes Shiferaw
- Department of Physics and Astronomy, California State University, Northridge, California.
| |
Collapse
|
183
|
Terrar DA. Endolysosomal Calcium Release and Cardiac Physiology. Cell Calcium 2022; 104:102565. [DOI: 10.1016/j.ceca.2022.102565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 02/20/2022] [Accepted: 02/21/2022] [Indexed: 11/25/2022]
|
184
|
Linz D, Norup Hertel J, Hendriks J, Saljic A, Dobrev D, Baumert M, Jespersen T, Linz D. Sleep apnea and atrial fibrillation: challenges in clinical and translational research. Expert Rev Cardiovasc Ther 2022; 20:101-109. [PMID: 35094618 DOI: 10.1080/14779072.2022.2036606] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Sleep-disordered breathing (SDB) is present in 21-74% of all patients with atrial fibrillation (AF). Treatment of SDB by positive airway pressure may help to prevent recurrence of AF after electrical cardioversion and help to improve AF ablation success rates in non-randomized studies. AREAS COVERED In this review, the current understanding of the atrial arrhythmogenic pathophysiology of SDB is summarized, and diagnostic and therapeutic challenges in AF patients are discussed. Current international recommendations are presented, and a comprehensive literature search is undertaken. EXPERT OPINION AF patients with SDB rarely report SDB-related symptoms such as daytime sleepiness. Therefore, systematic home sleep testing evaluation should be considered for all patients eligible for rhythm control strategy. A close interdisciplinary collaboration between the electrophysiologist/cardiologist, nurses and sleep-specialists are required for the management of SDB in AF patients. An arrhythmia-orientated assessment of SDB may better quantify SDB-related AF risk in an individual patient and may help to better guide targeted and personalized SDB treatment in AF patients as a component of rhythm and symptom control strategies. Finally, randomized controlled trials are needed to confirm the relationship between SDB and AF, and the benefits of routine testing and treatment of SDB in AF patients.
Collapse
Affiliation(s)
- Dominik Linz
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Julie Norup Hertel
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jeroen Hendriks
- Centre for Heart Rhythm Disorders, South Australian Health and Medical Research Institute, University of Adelaide and Royal Adelaide Hospital, Adelaide, Australia.,Department of Cardiology, Maastricht University Medical Centre and Cardiovascular Research Institute Maastricht, The Netherlands
| | - Arnela Saljic
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany.,Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada.,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Mathias Baumert
- Centre for Heart Rhythm Disorders, South Australian Health and Medical Research Institute, University of Adelaide and Royal Adelaide Hospital, Adelaide, Australia
| | - Thomas Jespersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Dominik Linz
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Centre for Heart Rhythm Disorders, South Australian Health and Medical Research Institute, University of Adelaide and Royal Adelaide Hospital, Adelaide, Australia.,Department of Cardiology, Maastricht University Medical Centre and Cardiovascular Research Institute Maastricht, The Netherlands.,Department of Cardiology, Radboud University Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
185
|
Andelova K, Bacova BS, Sykora M, Hlivak P, Barancik M, Tribulova N. Mechanisms Underlying Antiarrhythmic Properties of Cardioprotective Agents Impacting Inflammation and Oxidative Stress. Int J Mol Sci 2022; 23:1416. [PMID: 35163340 PMCID: PMC8835881 DOI: 10.3390/ijms23031416] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/21/2022] [Accepted: 01/25/2022] [Indexed: 01/27/2023] Open
Abstract
The prevention of cardiac life-threatening ventricular fibrillation and stroke-provoking atrial fibrillation remains a serious global clinical issue, with ongoing need for novel approaches. Numerous experimental and clinical studies suggest that oxidative stress and inflammation are deleterious to cardiovascular health, and can increase heart susceptibility to arrhythmias. It is quite interesting, however, that various cardio-protective compounds with antiarrhythmic properties are potent anti-oxidative and anti-inflammatory agents. These most likely target the pro-arrhythmia primary mechanisms. This review and literature-based analysis presents a realistic view of antiarrhythmic efficacy and the molecular mechanisms of current pharmaceuticals in clinical use. These include the sodium-glucose cotransporter-2 inhibitors used in diabetes treatment, statins in dyslipidemia and naturally protective omega-3 fatty acids. This approach supports the hypothesis that prevention or attenuation of oxidative and inflammatory stress can abolish pro-arrhythmic factors and the development of an arrhythmia substrate. This could prove a powerful tool of reducing cardiac arrhythmia burden.
Collapse
Affiliation(s)
- Katarina Andelova
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, Dúbravská Cesta 9, 84104 Bratislava, Slovakia; (K.A.); (M.S.); (M.B.)
| | - Barbara Szeiffova Bacova
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, Dúbravská Cesta 9, 84104 Bratislava, Slovakia; (K.A.); (M.S.); (M.B.)
| | - Matus Sykora
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, Dúbravská Cesta 9, 84104 Bratislava, Slovakia; (K.A.); (M.S.); (M.B.)
| | - Peter Hlivak
- Department of Arrhythmias and Pacing, National Institute of Cardiovascular Diseases, Pod Krásnou Hôrkou 1, 83348 Bratislava, Slovakia;
| | - Miroslav Barancik
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, Dúbravská Cesta 9, 84104 Bratislava, Slovakia; (K.A.); (M.S.); (M.B.)
| | - Narcis Tribulova
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, Dúbravská Cesta 9, 84104 Bratislava, Slovakia; (K.A.); (M.S.); (M.B.)
| |
Collapse
|
186
|
van Wijk SW, Su W, Wijdeveld LFJM, Ramos KS, Brundel BJJM. Cytoskeletal Protein Variants Driving Atrial Fibrillation: Potential Mechanisms of Action. Cells 2022; 11:416. [PMID: 35159226 PMCID: PMC8834312 DOI: 10.3390/cells11030416] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/15/2022] [Accepted: 01/20/2022] [Indexed: 11/16/2022] Open
Abstract
The most common clinical tachyarrhythmia, atrial fibrillation (AF), is present in 1-2% of the population. Although common risk factors, including hypertension, diabetes, and obesity, frequently underlie AF onset, it has been recognized that in 15% of the AF population, AF is familial. In these families, genome and exome sequencing techniques identified variants in the non-coding genome (i.e., variant regulatory elements), genes encoding ion channels, as well as genes encoding cytoskeletal (-associated) proteins. Cytoskeletal protein variants include variants in desmin, lamin A/C, titin, myosin heavy and light chain, junctophilin, nucleoporin, nesprin, and filamin C. These cytoskeletal protein variants have a strong association with the development of cardiomyopathy. Interestingly, AF onset is often represented as the initial manifestation of cardiac disease, sometimes even preceding cardiomyopathy by several years. Although emerging research findings reveal cytoskeletal protein variants to disrupt the cardiomyocyte structure and trigger DNA damage, exploration of the pathophysiological mechanisms of genetic AF is still in its infancy. In this review, we provide an overview of cytoskeletal (-associated) gene variants that relate to genetic AF and highlight potential pathophysiological pathways that drive this arrhythmia.
Collapse
Affiliation(s)
| | | | | | | | - Bianca J. J. M. Brundel
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (S.W.v.W.); (W.S.); (L.F.J.M.W.); (K.S.R.)
| |
Collapse
|
187
|
Sirish P, Diloretto DA, Thai PN, Chiamvimonvat N. The Critical Roles of Proteostasis and Endoplasmic Reticulum Stress in Atrial Fibrillation. Front Physiol 2022; 12:793171. [PMID: 35058801 PMCID: PMC8764384 DOI: 10.3389/fphys.2021.793171] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 12/08/2021] [Indexed: 12/19/2022] Open
Abstract
Atrial fibrillation (AF) remains the most common arrhythmia seen clinically. The incidence of AF is increasing due to the aging population. AF is associated with a significant increase in morbidity and mortality, yet current treatment paradigms have proven largely inadequate. Therefore, there is an urgent need to develop new effective therapeutic strategies for AF. The endoplasmic reticulum (ER) in the heart plays critical roles in the regulation of excitation-contraction coupling and cardiac function. Perturbation in the ER homeostasis due to intrinsic and extrinsic factors, such as inflammation, oxidative stress, and ischemia, leads to ER stress that has been linked to multiple conditions including diabetes mellitus, neurodegeneration, cancer, heart disease, and cardiac arrhythmias. Recent studies have documented the critical roles of ER stress in the pathophysiological basis of AF. Using an animal model of chronic pressure overload, we demonstrate a significant increase in ER stress in atrial tissues. Moreover, we demonstrate that treatment with a small molecule inhibitor to inhibit the soluble epoxide hydrolase enzyme in the arachidonic acid metabolism significantly reduces ER stress as well as atrial electrical and structural remodeling. The current review article will attempt to provide a perspective on our recent understandings and current knowledge gaps on the critical roles of proteostasis and ER stress in AF progression.
Collapse
Affiliation(s)
- Padmini Sirish
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, Davis, CA, United States.,Department of Veterans Affairs, Northern California Health Care System, Mather, CA, United States
| | - Daphne A Diloretto
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, Davis, CA, United States
| | - Phung N Thai
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, Davis, CA, United States.,Department of Veterans Affairs, Northern California Health Care System, Mather, CA, United States
| | - Nipavan Chiamvimonvat
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, Davis, CA, United States.,Department of Veterans Affairs, Northern California Health Care System, Mather, CA, United States.,Department of Pharmacology, University of California, Davis, Davis, CA, United States
| |
Collapse
|
188
|
MicroRNA-365 regulates human cardiac action potential duration. Nat Commun 2022; 13:220. [PMID: 35017523 PMCID: PMC8752767 DOI: 10.1038/s41467-021-27856-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 12/16/2021] [Indexed: 12/21/2022] Open
Abstract
Abnormalities of ventricular action potential cause malignant cardiac arrhythmias and sudden cardiac death. Here, we aim to identify microRNAs that regulate the human cardiac action potential and ask whether their manipulation allows for therapeutic modulation of action potential abnormalities. Quantitative analysis of the microRNA targetomes in human cardiac myocytes identifies miR-365 as a primary microRNA to regulate repolarizing ion channels. Action potential recordings in patient-specific induced pluripotent stem cell-derived cardiac myocytes show that elevation of miR-365 significantly prolongs action potential duration in myocytes derived from a Short-QT syndrome patient, whereas specific inhibition of miR-365 normalizes pathologically prolonged action potential in Long-QT syndrome myocytes. Transcriptome analyses in these cells at bulk and single-cell level corroborate the key cardiac repolarizing channels as direct targets of miR-365, together with functionally synergistic regulation of additional action potential-regulating genes by this microRNA. Whole-cell patch-clamp experiments confirm miR-365-dependent regulation of repolarizing ionic current Iks. Finally, refractory period measurements in human myocardial slices substantiate the regulatory effect of miR-365 on action potential in adult human myocardial tissue. Our results delineate miR-365 to regulate human cardiac action potential duration by targeting key factors of cardiac repolarization.
Collapse
|
189
|
Conditional immortalization of human atrial myocytes for the generation of in vitro models of atrial fibrillation. Nat Biomed Eng 2022; 6:389-402. [PMID: 34992271 DOI: 10.1038/s41551-021-00827-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 10/29/2021] [Indexed: 12/12/2022]
Abstract
The lack of a scalable and robust source of well-differentiated human atrial myocytes constrains the development of in vitro models of atrial fibrillation (AF). Here we show that fully functional atrial myocytes can be generated and expanded one-quadrillion-fold via a conditional cell-immortalization method relying on lentiviral vectors and the doxycycline-controlled expression of a recombinant viral oncogene in human foetal atrial myocytes, and that the immortalized cells can be used to generate in vitro models of AF. The method generated 15 monoclonal cell lines with molecular, cellular and electrophysiological properties resembling those of primary atrial myocytes. Multicellular in vitro models of AF generated using the immortalized atrial myocytes displayed fibrillatory activity (with activation frequencies of 6-8 Hz, consistent with the clinical manifestation of AF), which could be terminated by the administration of clinically approved antiarrhythmic drugs. The conditional cell-immortalization method could be used to generate functional cell lines from other human parenchymal cells, for the development of in vitro models of human disease.
Collapse
|
190
|
Gauvrit S, Bossaer J, Lee J, Collins MM. Modeling Human Cardiac Arrhythmias: Insights from Zebrafish. J Cardiovasc Dev Dis 2022; 9:jcdd9010013. [PMID: 35050223 PMCID: PMC8779270 DOI: 10.3390/jcdd9010013] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/23/2021] [Accepted: 12/25/2021] [Indexed: 12/13/2022] Open
Abstract
Cardiac arrhythmia, or irregular heart rhythm, is associated with morbidity and mortality and is described as one of the most important future public health challenges. Therefore, developing new models of cardiac arrhythmia is critical for understanding disease mechanisms, determining genetic underpinnings, and developing new therapeutic strategies. In the last few decades, the zebrafish has emerged as an attractive model to reproduce in vivo human cardiac pathologies, including arrhythmias. Here, we highlight the contribution of zebrafish to the field and discuss the available cardiac arrhythmia models. Further, we outline techniques to assess potential heart rhythm defects in larval and adult zebrafish. As genetic tools in zebrafish continue to bloom, this model will be crucial for functional genomics studies and to develop personalized anti-arrhythmic therapies.
Collapse
|
191
|
Maudsley S, Leysen H, van Gastel J, Martin B. Systems Pharmacology: Enabling Multidimensional Therapeutics. COMPREHENSIVE PHARMACOLOGY 2022:725-769. [DOI: 10.1016/b978-0-12-820472-6.00017-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
192
|
Yu LM, Dong X, Zhao JK, Xu YL, Xu DY, Xue XD, Zhou ZJ, Huang YT, Zhao QS, Luo LY, Wang ZS, Wang HS. Activation of PKG-CREB-KLF15 by melatonin attenuates Angiotensin II-induced vulnerability to atrial fibrillation via enhancing branched-chain amino acids catabolism. Free Radic Biol Med 2022; 178:202-214. [PMID: 34864165 DOI: 10.1016/j.freeradbiomed.2021.11.043] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/27/2021] [Accepted: 11/30/2021] [Indexed: 12/20/2022]
Abstract
Mitochondrial reactive oxygen species (ROS) damage and atrial remodeling serve as the crucial substrates for the genesis of atrial fibrillation (AF). Branched-chain amino acids (BCAAs) catabolic defect plays critical roles in multiple cardiovascular diseases. However, the alteration of atrial BCAA catabolism and its role in AF remain largely unknown. This study aimed to explore the role of BCAA catabolism in the pathogenesis of AF and to further evaluate the therapeutic effect of melatonin with a focus on protein kinase G (PKG)-cAMP response element binding protein (CREB)-Krüppel-like factor 15 (KLF15) signaling. We found that angiotensin II-treated atria exhibited significantly elevated BCAA level, reduced BCAA catabolic enzyme activity, increased AF vulnerability, aggravated atrial electrical and structural remodeling, and enhanced mitochondrial ROS damage. These deleterious effects were attenuated by melatonin co-administration while exacerbated by BCAA oral supplementation. Melatonin treatment ameliorated BCAA-induced atrial damage and reversed BCAA-induced down-regulation of atrial PKGIα expression, CREB phosphorylation as well as KLF15 expression. However, inhibition of PKG partly abolished melatonin-induced beneficial actions. In summary, these data demonstrated that atrial BCAA catabolic defect contributed to the pathogenesis of AF by aggravating tissue fibrosis and mitochondrial ROS damage. Melatonin treatment ameliorated Ang II-induced atrial structural as well as electrical remodeling by activating PKG-CREB-KLF15. The present study reveals additional mechanisms contributing to AF genesis and highlights the opportunity of a novel therapy for AF by targeting BCAA catabolism. Melatonin may serve as a potential therapeutic agent for AF intervention.
Collapse
Affiliation(s)
- Li-Ming Yu
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning, 110016, PR China
| | - Xue Dong
- Outpatient Department of Liaoning Military Region, General Hospital of Northern Theater Command, 49 Beiling Road, Shenyang, Liaoning, 110032, PR China
| | - Ji-Kai Zhao
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning, 110016, PR China
| | - Yin-Li Xu
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning, 110016, PR China
| | - Deng-Yue Xu
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning, 110016, PR China
| | - Xiao-Dong Xue
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning, 110016, PR China
| | - Zi-Jun Zhou
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning, 110016, PR China
| | - Yu-Ting Huang
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning, 110016, PR China
| | - Qiu-Sheng Zhao
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning, 110016, PR China
| | - Lin-Yu Luo
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning, 110016, PR China
| | - Zhi-Shang Wang
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning, 110016, PR China
| | - Hui-Shan Wang
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning, 110016, PR China.
| |
Collapse
|
193
|
Qiu H, Huang ZY, Cao H, Zhang Z, Ma J, Li XQ, Huang S, Li X, Qiu W, Zhao Z, Ji C, Huang L, Jiang W, Yang ZQ, Xian SX, Wu H, Lu W, Ding C. Deciphering mechanism of the herbal formula WuShen in the treatment of postinfarction heart failure. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 95:153878. [PMID: 34929563 DOI: 10.1016/j.phymed.2021.153878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/15/2021] [Accepted: 12/02/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND Numerous clinical studies reported the effectiveness of herbal formula WuShen (WS) in treating cardiovascular diseases, yet relevant basic research was rarely conducted. METHODS AND RESULTS Twelve main bioactive compounds of WS decoction were identified using the ultra-performance liquid chromatography-LTQ-Orbitrap mass spectrometer. A total of 137 active compounds with 613 targets were predicted by network pharmacology; their bioinformatic annotation and human microarray data suggested that wounding healing, inflammatory response, and gap junction were potentially the major therapeutic modules. A rat model of post-myocardial infarction (MI) heart failure (HF) was used to study the effects of WS on cardiac function, adverse cardiac remodeling, and experimental arrhythmias. Rats treated with WS led to a significantly improved pump function and reduced susceptibility to both ventricular tachycardia and atrial fibrillation, and restricted adverse cardiac remodeling partly via inhibiting TGFβ1/SMADs mediated extracellular matrix deposition and Rac1/NOX2/CTGF/Connexin43 -involved gap junction remodeling. CONCLUSIONS The present study highlights that WS can be applied to the treatment of heart failure and the upstream therapy for atrial fibrillation and ventricular tachycardia through its preventive effect on adverse cardiac remodeling.
Collapse
Affiliation(s)
- Huiliang Qiu
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China; Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, P.R. China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510020, P.R. China; Division of Cardiology, University of California, San Francisco, San Francisco CA 94143, United States of America
| | - Zeng-Yan Huang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, P.R. China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, P.R. China
| | - Haiming Cao
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China; Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, P.R. China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510020, P.R. China
| | - Zezhao Zhang
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China; Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, P.R. China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510020, P.R. China
| | - Jin Ma
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China; Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, P.R. China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510020, P.R. China
| | - Xiao-Qing Li
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China; Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, P.R. China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510020, P.R. China
| | - Shen Huang
- Department of Cardiology, Affiliated Hospital of Chengdu University, Chengdu 610081, P.R. China
| | - Xiong Li
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510020, P.R. China
| | - Wencong Qiu
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510020, P.R. China
| | - Zicong Zhao
- Foshan Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Foshan 528000, P.R. China
| | - Chunlan Ji
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China; Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, P.R. China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510020, P.R. China
| | - Lihua Huang
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China; Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, P.R. China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510020, P.R. China
| | - Wei Jiang
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China; Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, P.R. China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510020, P.R. China; Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Diseases, Guangzhou 510020, P.R. China
| | - Zhong-Qi Yang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, P.R. China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, P.R. China
| | - Shao-Xiang Xian
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, P.R. China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, P.R. China
| | - Huanlin Wu
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China; Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, P.R. China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510020, P.R. China; Department of internal medicine, Beijing University of Chinese Medicine, Beijing 100000, P.R. China
| | - Weihui Lu
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China; Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, P.R. China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510020, P.R. China.
| | - Chunhua Ding
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China; Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, P.R. China; Cardiac Department, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing 100049, P.R. China.
| |
Collapse
|
194
|
Hegyi B, Ko CY, Bossuyt J, Bers DM. Two-hit mechanism of cardiac arrhythmias in diabetic hyperglycaemia: reduced repolarization reserve, neurohormonal stimulation, and heart failure exacerbate susceptibility. Cardiovasc Res 2021; 117:2781-2793. [PMID: 33483728 PMCID: PMC8683706 DOI: 10.1093/cvr/cvab006] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 01/10/2021] [Indexed: 12/11/2022] Open
Abstract
AIMS Diabetic hyperglycaemia is associated with increased arrhythmia risk. We aimed to investigate whether hyperglycaemia alone can be accountable for arrhythmias or whether it requires the presence of additional pathological factors. METHODS AND RESULTS Action potentials (APs) and arrhythmogenic spontaneous diastolic activities were measured in isolated murine ventricular, rabbit atrial, and ventricular myocytes acutely exposed to high glucose. Acute hyperglycaemia increased the short-term variability (STV) of action potential duration (APD), enhanced delayed afterdepolarizations, and the inducibility of APD alternans during tachypacing in both murine and rabbit atrial and ventricular myocytes. Hyperglycaemia also prolonged APD in mice and rabbit atrial cells but not in rabbit ventricular myocytes. However, rabbit ventricular APD was more strongly depressed by block of late Na+ current (INaL) during hyperglycaemia, consistent with elevated INaL in hyperglycaemia. All the above proarrhythmic glucose effects were Ca2+-dependent and abolished by CaMKII inhibition. Importantly, when the repolarization reserve was reduced by pharmacological inhibition of K+ channels (either Ito, IKr, IKs, or IK1) or hypokalaemia, acute hyperglycaemia further prolonged APD and further increased STV and alternans in rabbit ventricular myocytes. Likewise, when rabbit ventricular myocytes were pretreated with isoproterenol or angiotensin II, hyperglycaemia significantly prolonged APD, increased STV and promoted alternans. Moreover, acute hyperglycaemia markedly prolonged APD and further enhanced STV in failing rabbit ventricular myocytes. CONCLUSION We conclude that even though hyperglycaemia alone can enhance cellular proarrhythmic mechanisms, a second hit which reduces the repolarization reserve or stimulates G protein-coupled receptor signalling greatly exacerbates cardiac arrhythmogenesis in diabetic hyperglycaemia.
Collapse
Affiliation(s)
- Bence Hegyi
- Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, CA 95616, USA
| | - Christopher Y Ko
- Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, CA 95616, USA
| | - Julie Bossuyt
- Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, CA 95616, USA
| | - Donald M Bers
- Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, CA 95616, USA
| |
Collapse
|
195
|
Mahnkopf C, Kwon Y, Akoum N. Atrial Fibrosis, Ischaemic Stroke and Atrial Fibrillation. Arrhythm Electrophysiol Rev 2021; 10:225-229. [PMID: 35106172 PMCID: PMC8785072 DOI: 10.15420/aer.2021.51] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 11/03/2021] [Indexed: 01/02/2023] Open
Abstract
Atrial fibrosis is an important component of the arrhythmic substrate in AF. Evidence suggests that atrial fibrosis also plays a role in increasing the risk of stroke in patients with the arrhythmia. Patients with embolic stroke of undetermined source (ESUS), who are suspected to have AF but are rarely shown to have it, frequently demonstrate evidence of atrial fibrosis; measured using late-gadolinium enhancement MRI, this manifests as atrial remodelling encompassing structural, functional and electrical properties. In this review, the authors discuss the available evidence linking atrial disease, including fibrosis, with the risk of ischaemic stroke in AF, as well as in the ESUS population, in whom it has been linked to recurrent stroke and new-onset AF. They also discuss the implications of this association on future research that may elucidate the mechanism of stroke and stroke prevention strategies in the AF and ESUS populations.
Collapse
Affiliation(s)
| | - Younghoon Kwon
- Division of Cardiology, University of Washington, Seattle, WA, US
| | - Nazem Akoum
- Division of Cardiology, University of Washington, Seattle, WA, US
| |
Collapse
|
196
|
Song J, Dobrev D, Li N. Proteomics: A promising approach to discover new biomarkers for atrial fibrillation. Int J Cardiol 2021; 345:125-126. [PMID: 34710496 PMCID: PMC8876401 DOI: 10.1016/j.ijcard.2021.10.135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 11/28/2022]
Affiliation(s)
- Jia Song
- Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, Houston, TX, USA
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Centre, University Duisburg-Essen, Germany; Montréal Heart Institute and University de Montréal, Medicine and Research Center, Montréal, QC, Canada; Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Na Li
- Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, Houston, TX, USA; Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA; Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
197
|
Hiram R. Cardiac cytokine therapy? Relevance of targeting inflammatory mediators to combat cardiac arrhythmogenic remodeling. IJC HEART & VASCULATURE 2021; 37:100918. [PMID: 34849391 PMCID: PMC8607203 DOI: 10.1016/j.ijcha.2021.100918] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 12/24/2022]
Key Words
- AF, Atrial Fibrillation
- CCL2, C-C motif Chemokine Ligand 2
- CM, Cardiomyocyte
- CamKII, Calcium/calmodulin-dependent protein kinase-II
- IFN-γ, Interferon gamma
- IL, Interleukin
- LA, Left Atrium
- LVZ, Low Voltage Zone
- NLRP3, NACHT, LRR, and PYD domains-containing protein-3
- Th-cell, T helper cell
Collapse
Affiliation(s)
- Roddy Hiram
- Montreal Heart Institute (MHI), Department of Medicine, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| |
Collapse
|
198
|
Function and regulation of phosphatase 1 in healthy and diseased heart. Cell Signal 2021; 90:110203. [PMID: 34822978 DOI: 10.1016/j.cellsig.2021.110203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 11/12/2021] [Accepted: 11/18/2021] [Indexed: 12/12/2022]
Abstract
Reversible phosphorylation of ion channels and calcium-handling proteins provides precise post-translational regulation of cardiac excitation and contractility. Serine/threonine phosphatases govern dephosphorylation of the majority of cardiac proteins. Accordingly, dysfunction of this regulation contributes to the development and progression of heart failure and atrial fibrillation. On the molecular level, these changes include alterations in the expression level and phosphorylation status of Ca2+ handling and excitation-contraction coupling proteins provoked by dysregulation of phosphatases. The serine/threonine protein phosphatase PP1 is one a major player in the regulation of cardiac excitation-contraction coupling. PP1 essentially impacts on cardiac physiology and pathophysiology via interactions with the cardiac ion channels Cav1.2, NKA, NCX and KCNQ1, sarcoplasmic reticulum-bound Ca2+ handling proteins such as RyR2, SERCA and PLB as well as the contractile proteins MLC2, TnI and MyBP-C. PP1 itself but also PP1-regulatory proteins like inhibitor-1, inhibitor-2 and heat-shock protein 20 are dysregulated in cardiac disease. Therefore, they represent interesting targets to gain more insights in heart pathophysiology and to identify new treatment strategies for patients with heart failure or atrial fibrillation. We describe the genetic and holoenzymatic structure of PP1 and review its role in the heart and cardiac disease. Finally, we highlight the importance of the PP1 regulatory proteins for disease manifestation, provide an overview of genetic models to study the role of PP1 for the development of heart failure and atrial fibrillation and discuss possibilities of pharmacological interventions.
Collapse
|
199
|
Abstract
Atrial fibrillation (AF) is the most common sustained arrhythmia and associated with increased morbidity and mortality resulting from thromboembolism and heart failure. AF often presents initially as paroxysmal and may progress to a sustained form over time. Sustained forms of AF may be associated with increased symptoms and cardiovascular morbidity, and AF progression may be associated with increased risk of clinically adverse events and outcomes. The present review discusses the clinical factors of arrhythmia progression and risk stratification available to assess the probability of AF progression. Furthermore, currently available treatment options for preventing AF progression are explored and evaluated.
Collapse
Affiliation(s)
- Hisashi Ogawa
- Department of Cardiology, National Hospital Organization Kyoto Medical Center
| | - Masaharu Akao
- Department of Cardiology, National Hospital Organization Kyoto Medical Center
| |
Collapse
|
200
|
Qu Q, Sun JY, Zhang ZY, Su Y, Li SS, Li F, Wang RX. Hub microRNAs and genes in the development of atrial fibrillation identified by weighted gene co-expression network analysis. BMC Med Genomics 2021; 14:271. [PMID: 34781940 PMCID: PMC8591905 DOI: 10.1186/s12920-021-01124-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 11/08/2021] [Indexed: 01/17/2023] Open
Abstract
Co-expression network may contribute to better understanding molecular interaction patterns underlying cellular processes. To explore microRNAs (miRNAs) expression patterns correlated with AF, we performed weighted gene co-expression network analysis (WGCNA) based on the dataset GSE28954. Thereafter, we predicted target genes using experimentally verified databases (ENOCRI, miRTarBase, and Tarbase), and overlapped genes with differentially expressed genes (DEGs) from GSE79768 were identified as key genes. Integrated analysis of association between hub miRNAs and key genes was conducted to screen hub genes. In general, we identified 3 differentially expressed miRNAs (DEMs) and 320 DEGs, predominantly enriched in inflammation-related functional items. Two significant modules (red and blue) and hub miRNAs (hsa-miR-146b-5p and hsa-miR-378a-5p), which highly correlated with AF-related phenotype, were detected by WGCNA. By overlapping the DEGs and predicted target genes, 38 genes were screened out. Finally, 9 genes (i.e. ATP13A3, BMP2, CXCL1, GABPA, LIF, MAP3K8, NPY1R, S100A12, SLC16A2) located at the core region in the miRNA-gene interaction network were identified as hub genes. In conclusion, our study identified 2 hub miRNAs and 9 hub genes, which may improve the understanding of molecular mechanisms and help to reveal potential therapeutic targets against AF.
Collapse
Affiliation(s)
- Qiang Qu
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299, Qingyang Road, Wuxi, 214023, China.,Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Jin-Yu Sun
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299, Qingyang Road, Wuxi, 214023, China.,Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Zhen-Ye Zhang
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299, Qingyang Road, Wuxi, 214023, China
| | - Yue Su
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299, Qingyang Road, Wuxi, 214023, China.,Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Shan-Shan Li
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299, Qingyang Road, Wuxi, 214023, China
| | - Feng Li
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299, Qingyang Road, Wuxi, 214023, China
| | - Ru-Xing Wang
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299, Qingyang Road, Wuxi, 214023, China.
| |
Collapse
|