51
|
Ma ZG, Yuan YP, Wu HM, Zhang X, Tang QZ. Cardiac fibrosis: new insights into the pathogenesis. Int J Biol Sci 2018; 14:1645-1657. [PMID: 30416379 PMCID: PMC6216032 DOI: 10.7150/ijbs.28103] [Citation(s) in RCA: 221] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 08/02/2018] [Indexed: 12/21/2022] Open
Abstract
Cardiac fibrosis is defined as the imbalance of extracellular matrix (ECM) production and degradation, thus contributing to cardiac dysfunction in many cardiac pathophysiologic conditions. This review discusses specific markers and origin of cardiac fibroblasts (CFs), and the underlying mechanism involved in the development of cardiac fibrosis. Currently, there are no CFs-specific molecular markers. Most studies use co-labelling with panels of antibodies that can recognize CFs. Origin of fibroblasts is heterogeneous. After fibrotic stimuli, the levels of myocardial pro-fibrotic growth factors and cytokines are increased. These pro-fibrotic growth factors and cytokines bind to its receptors and then trigger the activation of signaling pathway and transcriptional factors via Smad-dependent or Smad independent-manners. These fibrosis-related transcriptional factors regulate gene expression that are involved in the fibrosis to amplify the fibrotic response. Understanding the mechanisms responsible for initiation, progression, and amplification of cardiac fibrosis are of great clinical significance to find drugs that can prevent the progression of cardiac fibrosis.
Collapse
Affiliation(s)
- Zhen-Guo Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China.,Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China.,Hubei Key Laboratory of Cardiology, Wuhan 430060, RP China
| | - Yu-Pei Yuan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China.,Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China.,Hubei Key Laboratory of Cardiology, Wuhan 430060, RP China
| | - Hai-Ming Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China.,Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China.,Hubei Key Laboratory of Cardiology, Wuhan 430060, RP China
| | - Xin Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China.,Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China.,Hubei Key Laboratory of Cardiology, Wuhan 430060, RP China
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, RP China.,Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China.,Hubei Key Laboratory of Cardiology, Wuhan 430060, RP China
| |
Collapse
|
52
|
Frangogiannis NG. Cardiac fibrosis: Cell biological mechanisms, molecular pathways and therapeutic opportunities. Mol Aspects Med 2018; 65:70-99. [PMID: 30056242 DOI: 10.1016/j.mam.2018.07.001] [Citation(s) in RCA: 586] [Impact Index Per Article: 83.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 07/23/2018] [Indexed: 12/13/2022]
Abstract
Cardiac fibrosis is a common pathophysiologic companion of most myocardial diseases, and is associated with systolic and diastolic dysfunction, arrhythmogenesis, and adverse outcome. Because the adult mammalian heart has negligible regenerative capacity, death of a large number of cardiomyocytes results in reparative fibrosis, a process that is critical for preservation of the structural integrity of the infarcted ventricle. On the other hand, pathophysiologic stimuli, such as pressure overload, volume overload, metabolic dysfunction, and aging may cause interstitial and perivascular fibrosis in the absence of infarction. Activated myofibroblasts are the main effector cells in cardiac fibrosis; their expansion following myocardial injury is primarily driven through activation of resident interstitial cell populations. Several other cell types, including cardiomyocytes, endothelial cells, pericytes, macrophages, lymphocytes and mast cells may contribute to the fibrotic process, by producing proteases that participate in matrix metabolism, by secreting fibrogenic mediators and matricellular proteins, or by exerting contact-dependent actions on fibroblast phenotype. The mechanisms of induction of fibrogenic signals are dependent on the type of primary myocardial injury. Activation of neurohumoral pathways stimulates fibroblasts both directly, and through effects on immune cell populations. Cytokines and growth factors, such as Tumor Necrosis Factor-α, Interleukin (IL)-1, IL-10, chemokines, members of the Transforming Growth Factor-β family, IL-11, and Platelet-Derived Growth Factors are secreted in the cardiac interstitium and play distinct roles in activating specific aspects of the fibrotic response. Secreted fibrogenic mediators and matricellular proteins bind to cell surface receptors in fibroblasts, such as cytokine receptors, integrins, syndecans and CD44, and transduce intracellular signaling cascades that regulate genes involved in synthesis, processing and metabolism of the extracellular matrix. Endogenous pathways involved in negative regulation of fibrosis are critical for cardiac repair and may protect the myocardium from excessive fibrogenic responses. Due to the reparative nature of many forms of cardiac fibrosis, targeting fibrotic remodeling following myocardial injury poses major challenges. Development of effective therapies will require careful dissection of the cell biological mechanisms, study of the functional consequences of fibrotic changes on the myocardium, and identification of heart failure patient subsets with overactive fibrotic responses.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, 1300 Morris Park Avenue, Forchheimer G46B, Bronx, NY, 10461, USA.
| |
Collapse
|
53
|
Chen T, Li M, Fan X, Cheng J, Wang L. Sodium Tanshinone IIA Sulfonate Prevents Angiotensin II-Induced Differentiation of Human Atrial Fibroblasts into Myofibroblasts. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:6712585. [PMID: 30140368 PMCID: PMC6081515 DOI: 10.1155/2018/6712585] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 06/25/2018] [Accepted: 07/05/2018] [Indexed: 02/06/2023]
Abstract
Differentiation of atrial fibroblasts into myofibroblasts plays a critical role in atrial fibrosis. Sodium tanshinone IIA sulfonate (DS-201), a water-soluble derivative of tanshinone IIA, has been shown to have potent antifibrotic properties. However, the protective effects of DS-201 on angiotensin II- (Ang II-) induced differentiation of atrial fibroblasts into myofibroblasts remain to be elucidated. In this study, human atrial fibroblasts were stimulated with Ang II in the presence or absence of DS-201. Then, α-smooth muscle actin (α-SMA), collagen I, and collagen III expression and reactive oxygen species (ROS) generation were measured. The expression of transforming growth factor-β1 (TGF-β1) and the downstream signaling of TGF-β1, such as phosphorylation of Smad2/3, were also determined. The results demonstrated that DS-201 significantly prevented Ang II-induced human atrial fibroblast migration and decreased Ang II-induced α-SMA, collagen I, and collagen III expression. Furthermore, increased production of ROS and expression of TGF-β1 stimulated by Ang II were also significantly inhibited by DS-201. Consistent with these results, DS-201 significantly inhibited Ang II-evoked Smad2/3 phosphorylation and periostin expression. These results and the experiments involving N-acetyl cysteine (antioxidant) and an anti-TGF-β1 antibody suggest that DS-201 prevent Ang II-induced differentiation of atrial fibroblasts to myofibroblasts, at least in part, through suppressing oxidative stress and inhibiting the activation of TGF-β1 signaling pathway. All of these data indicate the potential utility of DS-201 for the treatment of cardiac fibrosis.
Collapse
Affiliation(s)
- Tangting Chen
- Key Laboratory of Ministry of Education for Medical Electrophysiology and the Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China
| | - Miaoling Li
- Key Laboratory of Ministry of Education for Medical Electrophysiology and the Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China
| | - Xuehui Fan
- Key Laboratory of Ministry of Education for Medical Electrophysiology and the Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China
| | - Jun Cheng
- Key Laboratory of Ministry of Education for Medical Electrophysiology and the Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China
| | - Liqun Wang
- Drug Discovery Research Center, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China
| |
Collapse
|
54
|
Xu J, Wu H, Chen S, Qi B, Zhou G, Cai L, Zhao L, Wei Y, Liu S. MicroRNA-30c suppresses the pro-fibrogenic effects of cardiac fibroblasts induced by TGF-β1 and prevents atrial fibrosis by targeting TGFβRII. J Cell Mol Med 2018. [PMID: 29532993 PMCID: PMC5980214 DOI: 10.1111/jcmm.13548] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Atrial fibrosis serves as an important contributor to atrial fibrillation (AF). Recent data have suggested that microRNA-30c (miR-30c) is involved in fibrotic remodelling and cancer development, but the specific role of miR-30c in atrial fibrosis remains unclear. The purpose of this study was to investigate the role of miR-30c in atrial fibrosis and its underlying mechanisms through in vivo and in vitro experiments. Our results indicate that miR-30c is significantly down-regulated in the rat abdominal aortic constriction (AAC) model and in the cellular model of fibrosis induced by transforming growth factor-β1 (TGF-β1). Overexpression of miR-30c in cardiac fibroblasts (CFs) markedly inhibits CF proliferation, differentiation, migration and collagen production, whereas decrease in miR-30c leads to the opposite results. Moreover, we identified TGFβRII as a target of miR-30c. Finally, transferring adeno-associated virus 9 (AAV9)-miR-30c into the inferior vena cava of rats attenuated fibrosis in the left atrium following AAC. These data indicate that miR-30c attenuates atrial fibrosis via inhibition of CF proliferation, differentiation, migration and collagen production by targeting TGFβRII, suggesting that miR-30c might be a novel potential therapeutic target for preventing atrial fibrosis.
Collapse
Affiliation(s)
- Juan Xu
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haiqing Wu
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Songwen Chen
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Baozhen Qi
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Genqing Zhou
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lidong Cai
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liqun Zhao
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yong Wei
- Department of Cardiology, Shanghai Songjiang Central Hospital, Shanghai, China
| | - Shaowen Liu
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
55
|
TGF-β-Induced Endothelial-Mesenchymal Transition in Fibrotic Diseases. Int J Mol Sci 2017; 18:ijms18102157. [PMID: 29039786 PMCID: PMC5666838 DOI: 10.3390/ijms18102157] [Citation(s) in RCA: 273] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 10/06/2017] [Accepted: 10/13/2017] [Indexed: 12/22/2022] Open
Abstract
Fibrotic diseases are characterized by net accumulation of extracellular matrix proteins in affected organs leading to their dysfunction and ultimate failure. Myofibroblasts have been identified as the cells responsible for the progression of the fibrotic process, and they originate from several sources, including quiescent tissue fibroblasts, circulating CD34⁺ fibrocytes and the phenotypic conversion of various cell types into activated myofibroblasts. Several studies have demonstrated that endothelial cells can transdifferentiate into mesenchymal cells through a process termed endothelial- mesenchymal transition (EndMT) and that this can give rise to activated myofibroblasts involved in the development of fibrotic diseases. Transforming growth factor β (TGF-β) has a central role in fibrogenesis by modulating the fibroblast phenotype and function, inducing myofibroblast transdifferentiation and promoting matrix accumulation. In addition, TGF-β by inducing EndMT may further contribute to the development of fibrosis. Despite extensive investigation of the pathogenesis of fibrotic diseases, no effective treatment strategies are available. Delineation of the mechanisms responsible for initiation and progression of fibrotic diseases is crucial for the development of therapeutic strategies for the treatment of the disease. In this review, we summarize the role of the TGF-β signaling pathway and EndMT in the development of fibrotic diseases and discuss their therapeutic potential.
Collapse
|
56
|
Gene Expression Networks in the Murine Pulmonary Myocardium Provide Insight into the Pathobiology of Atrial Fibrillation. G3-GENES GENOMES GENETICS 2017; 7:2999-3017. [PMID: 28720711 PMCID: PMC5592927 DOI: 10.1534/g3.117.044651] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The pulmonary myocardium is a muscular coat surrounding the pulmonary and caval veins. Although its definitive physiological function is unknown, it may have a pathological role as the source of ectopic beats initiating atrial fibrillation. How the pulmonary myocardium gains pacemaker function is not clearly defined, although recent evidence indicates that changed transcriptional gene expression networks are at fault. The gene expression profile of this distinct cell type in situ was examined to investigate underlying molecular events that might contribute to atrial fibrillation. Via systems genetics, a whole-lung transcriptome data set from the BXD recombinant inbred mouse resource was analyzed, uncovering a pulmonary cardiomyocyte gene network of 24 transcripts, coordinately regulated by chromosome 1 and 2 loci. Promoter enrichment analysis and interrogation of publicly available ChIP-seq data suggested that transcription of this gene network may be regulated by the concerted activity of NKX2-5, serum response factor, myocyte enhancer factor 2, and also, at a post-transcriptional level, by RNA binding protein motif 20. Gene ontology terms indicate that this gene network overlaps with molecular markers of the stressed heart. Therefore, we propose that perturbed regulation of this gene network might lead to altered calcium handling, myocyte growth, and contractile force contributing to the aberrant electrophysiological properties observed in atrial fibrillation. We reveal novel molecular interactions and pathways representing possible therapeutic targets for atrial fibrillation. In addition, we highlight the utility of recombinant inbred mouse resources in detecting and characterizing gene expression networks of relatively small populations of cells that have a pathological significance.
Collapse
|
57
|
Chang SH, Yeh YH, Lee JL, Hsu YJ, Kuo CT, Chen WJ. Transforming growth factor-β-mediated CD44/STAT3 signaling contributes to the development of atrial fibrosis and fibrillation. Basic Res Cardiol 2017; 112:58. [PMID: 28871329 DOI: 10.1007/s00395-017-0647-9] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 08/29/2017] [Indexed: 11/30/2022]
Abstract
Atrial fibrillation (AF) is associated with atrial fibrosis. Inhibition of atrial fibrosis might be a plausible approach for AF prevention and therapy. This study is designed to evaluate the potential role of CD44, a membrane receptor known to regulate fibrosis, and its related signaling in the pathogenesis of atrial fibrosis and AF. Treatment of cultured rat atrial fibroblasts with transforming growth factor-β (TGF-β, a key mediator of atrial fibrosis) led to a higher expression of hyaluronan (HA), CD44, STAT3, and collagen (a principal marker of fibrosis) than that of ventricular fibroblasts. In vivo, TGF-β transgenic mice and AF patients exhibited a greater expression of HA, CD44, STAT3, and collagen in their atria than wild-type mice and sinus rhythm subjects, respectively. Treating TGF-β transgenic mice with an anti-CD44 blocking antibody resulted in a lower expression of STAT3 and collagen in their atria than those with control IgG antibody. Programmed stimulation triggered less AF episodes in TGF-β transgenic mice treated with anti-CD44 blocking antibody than in those with control IgG. Blocking CD44 signaling with anti-CD44 antibody and mutated CD44 plasmids attenuated TGF-β-induced STAT3 activation and collagen expression in cultured atrial fibroblasts. Deletion and mutational analysis of the collagen promoter along with chromatin immunoprecipitation demonstrated that STAT3 served as a vital transcription factor in collagen expression. TGF-β-mediated HA/CD44/STAT3 pathway plays a crucial role in the development of atrial fibrosis and AF. Blocking CD44-dependent signaling may be a feasible way for AF management.
Collapse
Affiliation(s)
- Shang-Hung Chang
- Cardiovascular Division, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Fu-Shin Road No. 5, Kwei-Shan, Taoyuan, 333, Taiwan
| | - Yung-Hsin Yeh
- Cardiovascular Division, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Fu-Shin Road No. 5, Kwei-Shan, Taoyuan, 333, Taiwan
| | - Jia-Lin Lee
- Department of Medical Science, Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Yu-Juei Hsu
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chi-Tai Kuo
- Cardiovascular Division, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Fu-Shin Road No. 5, Kwei-Shan, Taoyuan, 333, Taiwan
| | - Wei-Jan Chen
- Cardiovascular Division, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Fu-Shin Road No. 5, Kwei-Shan, Taoyuan, 333, Taiwan.
| |
Collapse
|
58
|
TGF-β1 affects cell-cell adhesion in the heart in an NCAM1-dependent mechanism. J Mol Cell Cardiol 2017; 112:49-57. [PMID: 28870505 DOI: 10.1016/j.yjmcc.2017.08.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 08/23/2017] [Accepted: 08/31/2017] [Indexed: 12/22/2022]
Abstract
The contractile property of the myocardium is maintained by cell-cell junctions enabling cardiomyocytes to work as a syncytium. Alterations in cell-cell junctions are observed in heart failure, a disease characterized by the activation of Transforming Growth Factor beta 1 (TGFβ1). While TGFβ1 has been implicated in diverse biologic responses, its molecular function in controlling cell-cell adhesion in the heart has never been investigated. Cardiac-specific transgenic mice expressing active TGFβ1 were generated to model the observed increase in activity in the failing heart. Activation of TGFβ1 in the heart was sufficient to drive ventricular dysfunction. To begin to understand the function of this important molecule we undertook an extensive structural analysis of the myocardium by electron microscopy and immunostaining. This approach revealed that TGFβ1 alters intercalated disc structures and cell-cell adhesion in ventricular myocytes. Mechanistically, we found that TGFβ1 induces the expression of neural adhesion molecule 1 (NCAM1) in cardiomyocytes in a p38-dependent pathway, and that selective targeting of NCAM1 was sufficient to rescue the cell adhesion defect observed when cardiomyocytes were treated with TGFβ1. Importantly, NCAM1 was upregulated in human heart samples from ischemic and non-ischemic cardiomyopathy patients and NCAM1 protein levels correlated with the degree of TGFβ1 activity in the human cardiac ventricle. Overall, we found that TGFβ1 is deleterious to the heart by regulating the adhesion properties of cardiomyocytes in an NCAM1-dependent mechanism. Our results suggest that inhibiting NCAM1 would be cardioprotective, counteract the pathological action of TGFβ1 and reduce heart failure severity.
Collapse
|
59
|
Han L, Li X, Zhang G, Xu Z, Gong D, Lu F, Liu X. Pericardial interstitial cell senescence responsible for pericardial structural remodeling in idiopathic and postsurgical constrictive pericarditis. J Thorac Cardiovasc Surg 2017; 154:966-975.e4. [PMID: 28456362 DOI: 10.1016/j.jtcvs.2017.03.115] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 03/04/2017] [Accepted: 03/20/2017] [Indexed: 01/14/2023]
Abstract
OBJECTIVE Idiopathic and postsurgical constrictive pericarditis is characterized by pericardial structural remodeling that involves fibrosis, calcification, and inflammation. This study aimed to determine whether cell senescence was responsible for pericardial structural remodeling. METHODS Pericardial interstitial cells derived from patients with idiopathic or postsurgical pericarditis (pericarditis cells) were harvested. Timing of senescence and differences in telomere length were compared between age- and sex-matched controls (nonpericarditis cells). Pericardial interstitial cells derived from normal pericardia were serially passaged until senescence (senescent cells). Apoptosis, collagen matrix, calcium deposition, chemoattractant properties, gene expression profiles, and paracrine effects of senescent cells were compared with nonsenescent cells of passage 2 (nonsenescent cells). RESULTS Pericarditis cells displayed senescent changes, including short telomere length, large flattened cell sizes, positive staining for senescence-associated β-galactosidase, and limited growth capacity. These senescent cells were resistant to apoptosis, produced more collagen matrix, deposited more calcium, and attracted more monocytes/lymphocytes than the nonsenescent cells. A cluster of genes involved in extracellular matrix deposition (connective tissue growth factor, fibronectin, collagen type I, collagen type III, and tissue inhibitors of metalloproteinase-1), calcium deposition (osteopontin, bone sialoprotein, osteonectin, and matrix Gla protein), and inflammatory cell recruitment (interleukin-6, chemoattractant protein-1, and tumor necrosis factor-α) were upregulated in senescent cells, whereas extracellular matrix-degrading enzyme (metalloproteinase-1 and metalloproteinase-3) was downregulated. Furthermore, senescent cells had the ability to promote the proliferation, differentiation, and senescence of neighboring cells. CONCLUSIONS These findings suggest that senescent cells have characteristics promoting pericardial structural remodeling, but further work is needed to establish causation.
Collapse
Affiliation(s)
- Lin Han
- Institute of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xin Li
- Institute of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Guanxin Zhang
- Institute of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Zhiyun Xu
- Institute of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Dejun Gong
- Institute of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Fanglin Lu
- Institute of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xiaohong Liu
- Institute of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China.
| |
Collapse
|
60
|
Fibroblasts in an endocardial fibroelastosis disease model mainly originate from mesenchymal derivatives of epicardium. Cell Res 2017; 27:1157-1177. [PMID: 28809397 DOI: 10.1038/cr.2017.103] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Revised: 06/18/2017] [Accepted: 06/29/2017] [Indexed: 02/06/2023] Open
Abstract
Endocardial fibroelastosis (EFE) refers to the thickening of the ventricular endocardium as a result of de novo deposition of subendocardial fibrous tissue layers during neonatal heart development. The origin of EFE fibroblasts is proposed to be postnatal endocardial cells that undergo an aberrant endothelial-to-mesenchymal transition (EndMT). Genetic lineage tracing of endocardial cells with the inducible endocardial Cre line Npr3-CreER and the endothelial cell tracing line Cdh5-CreER on an EFE-like model did not reveal any contribution of neonatal endocardial cells to fibroblasts in the EFE-like tissues. Instead, lineage tracing of embryonic epicardium by Wt1-CreER suggested that epicardium-derived mesenchymal cells (MCs) served as the major source of EFE fibroblasts. By labeling MCs using Sox9-CreER, we confirmed that MCs of the embryonic heart expand and contribute to the majority of neonatal EFE fibroblasts. During this pathological process, TGFβ signaling, the key mediator of fibroblasts activation, was highly upregulated in the EFE-like tissues. Targeting TGFβ signaling by administration of its antagonist bone morphogenetic protein 7 effectively reduced fibroblast accumulation and tissue fibrosis in the EFE-like model. Our study provides genetic evidence that excessive fibroblasts in the EFE-like tissues mainly originate from the epicardium-derived MCs through epicardial to mesenchymal transition (EpiMT). These EpiMT-derived fibroblasts within the EFE-like tissues could serve as a potential therapeutic target.
Collapse
|
61
|
Potential Biological Markers of Atrial Fibrillation: A Chance to Prevent Cryptogenic Stroke. BIOMED RESEARCH INTERNATIONAL 2017; 2017:8153024. [PMID: 28785588 PMCID: PMC5530434 DOI: 10.1155/2017/8153024] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 05/15/2017] [Accepted: 06/04/2017] [Indexed: 01/04/2023]
Abstract
Stroke affects millions of people all over the world, causing death and disability. The most frequent type of this disease is ischemic stroke, which can be caused by different factors. In approximately 25 percent of cases, no obvious cause can be found. Recent observations have shown that paroxysmal atrial fibrillation could be responsible for a significant number of cryptogenic stroke events. Short- or long-lasting ECG monitoring could help with the diagnosis of transient arrhythmias. Unfortunately, these techniques either are expensive or require good patient compliance. An alternative option is the identification of biological markers that are specific for atrial fibrillation and can be used to predict arrhythmia. In this review, we give a summary of the recent advances in the research of arrhythmia markers. Based on their structure and function, we differentiated four groups of biomarkers: markers of inflammation, markers of fibrosis, markers with hormonal activity, and other markers. In spite of intensive researches, the optimal biological marker is still not available, but there are some promising markers, like NT-proBNP/BNP.
Collapse
|
62
|
Gaborit B, Sengenes C, Ancel P, Jacquier A, Dutour A. Role of Epicardial Adipose Tissue in Health and Disease: A Matter of Fat? Compr Physiol 2017. [PMID: 28640452 DOI: 10.1002/cphy.c160034] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Epicardial adipose tissue (EAT) is a small but very biologically active ectopic fat depot that surrounds the heart. Given its rapid metabolism, thermogenic capacity, unique transcriptome, secretory profile, and simply measurability, epicardial fat has drawn increasing attention among researchers attempting to elucidate its putative role in health and cardiovascular diseases. The cellular crosstalk between epicardial adipocytes and cells of the vascular wall or myocytes is high and suggests a local role for this tissue. The balance between protective and proinflammatory/profibrotic cytokines, chemokines, and adipokines released by EAT seem to be a key element in atherogenesis and could represent a future therapeutic target. EAT amount has been found to predict clinical coronary outcomes. EAT can also modulate cardiac structure and function. Its amount has been associated with atrial fibrillation, coronary artery disease, and sleep apnea syndrome. Conversely, a beiging fat profile of EAT has been identified. In this review, we describe the current state of knowledge regarding the anatomy, physiology and pathophysiological role of EAT, and the factors more globally leading to ectopic fat development. We will also highlight the most recent findings on the origin of this ectopic tissue, and its association with cardiac diseases. © 2017 American Physiological Society. Compr Physiol 7:1051-1082, 2017.
Collapse
Affiliation(s)
- Bénédicte Gaborit
- NORT, Aix Marseille Univ, INSERM, INRA, NORT, Marseille, France.,Endocrinology Metabolic Diseases, and Nutrition Department, Pole ENDO, APHM, Aix-Marseille Univ, Marseille, France
| | - Coralie Sengenes
- STROMALab, Université de Toulouse, EFS, ENVT, Inserm U1031, ERL CNRS 5311, CHU Rangueil, Toulouse, France
| | - Patricia Ancel
- NORT, Aix Marseille Univ, INSERM, INRA, NORT, Marseille, France
| | - Alexis Jacquier
- CNRS UMR 7339, Centre de Résonance Magnétique Biologique et Médicale (CRMBM), Marseille, France.,Radiology department, CHU La Timone, Marseille, France
| | - Anne Dutour
- NORT, Aix Marseille Univ, INSERM, INRA, NORT, Marseille, France.,Endocrinology Metabolic Diseases, and Nutrition Department, Pole ENDO, APHM, Aix-Marseille Univ, Marseille, France
| |
Collapse
|
63
|
Du L, Qin M, Yi Y, Chen X, Jiang W, Zhou L, Zhang D, Xu K, Yang Y, Li C, Liu Y, Liu X, Duan SZ. Eplerenone Prevents Atrial Fibrosis via the TGF-β Signaling Pathway. Cardiology 2017; 138:55-62. [PMID: 28571007 DOI: 10.1159/000471918] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 03/22/2017] [Indexed: 01/01/2023]
Abstract
OBJECTIVES Eplerenone (EPL), an antagonist of the mineralocorticoid receptor, is beneficial for atrial fibrillation and atrial fibrosis. However, the underlying mechanism remains less well known. We aimed to investigate the effect of EPL on atrial fibrosis using a mouse with selective atrial fibrosis and to explore the underlying mechanisms. METHODS EPL-treated MHC-TGFcys33ser transgenic mice that have selective atrial fibrosis (Tx+EPL mice), as well as control mice, were used for in vivo studies including histological analyses, Western blotting, and qRT-PCR studies. TGF-β1-stimulated atrial fibroblasts were treated with EPL or vehicle for the in vitro studies including Western blotting and qRT-PCR studies. In addition, Smad7 siRNA was used to knock down Smad7. RESULTS EPL inhibited atrial fibrosis in the Tx mice. In addition, EPL suppressed the expression of fibrosis-related molecules induced by TGF-β1 in vivo and in vitro. This occurred in concert with a downregulation of Smad7 protein expression and an upregulation of p-Smad2/3 protein expression. In addition, knockdown of Smad7 by siRNA abolished the protective roles of EPL. CONCLUSIONS EPL inhibited atrial fibrosis in Tx mice. The underlying mechanism may involve increased protein expression of Smad7, which enhances the inhibitory feedback regulation of TGF-β1/Smad signaling.
Collapse
Affiliation(s)
- Lili Du
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Rani S, Sreenivasaiah PK, Kim JO, Lee MY, Kang WS, Kim YS, Ahn Y, Park WJ, Cho C, Kim DH. Tauroursodeoxycholic acid (TUDCA) attenuates pressure overload-induced cardiac remodeling by reducing endoplasmic reticulum stress. PLoS One 2017; 12:e0176071. [PMID: 28426781 PMCID: PMC5398705 DOI: 10.1371/journal.pone.0176071] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 04/05/2017] [Indexed: 12/26/2022] Open
Abstract
Pressure overload in the heart induces pathological hypertrophy and is associated with cardiac dysfunction. Apoptosis and fibrosis signaling initiated by the endoplasmic reticulum stress (ERS) is known to contribute to these maladaptive effects. The aim of this study was to investigate whether reduction of ERS by a known chemical chaperone, tauroursodeoxycholic acid (TUDCA) can attenuate pressure overload-induced cardiac remodeling in a mouse model of transverse aortic constriction (TAC). Oral administration of TUDCA at a dose of 300 mg/kg body weight (BW) in the TUDCA-TAC group reduced ERS markers (GRP78, p-PERK, and p-eIf2α), compared to the Vehicle (Veh)-TAC group. TUDCA administration, for 4 weeks after TAC significantly reduced cardiac hypertrophy as shown by the reduced heart weight (HW) to BW ratio, and expression of hypertrophic marker genes (ANF, BNP, and α-SKA). Masson's trichrome staining showed that myocardial fibrosis and collagen deposition were also significantly reduced in the TUDCA-TAC group. We also found that TUDCA significantly decreased expression of TGF-β signaling proteins and collagen isoforms. TUDCA administration also reduced cardiac apoptosis and the related proteins in the TUDCA-TAC group. Microarray analysis followed by gene ontology (GO) and pathway analysis demonstrated that extracellular matrix genes responsible for hypertrophy and fibrosis, and mitochondrial genes responsible for apoptosis and fatty acid metabolism were significantly altered in the Veh-TAC group, but the alterations were normalized in the TUDCA-TAC group, suggesting potential of TUDCA in treatment of heart diseases related to pressure-overload.
Collapse
Affiliation(s)
- Shilpa Rani
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Korea
| | | | - Jin Ock Kim
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Korea
| | - Mi Young Lee
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Korea
| | - Wan Seok Kang
- Department of Cardiology, Chonnam National University Hospital, Gwangju, Korea
| | - Yong Sook Kim
- Department of Cardiology, Chonnam National University Hospital, Gwangju, Korea
| | - Youngkeun Ahn
- Department of Cardiology, Chonnam National University Hospital, Gwangju, Korea
| | - Woo Jin Park
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Korea
| | - Chunghee Cho
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Korea
| | - Do Han Kim
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Korea
- * E-mail:
| |
Collapse
|
65
|
Plasminogen Activator Inhibitor-1 Protects Mice Against Cardiac Fibrosis by Inhibiting Urokinase-type Plasminogen Activator-mediated Plasminogen Activation. Sci Rep 2017; 7:365. [PMID: 28336948 PMCID: PMC5428408 DOI: 10.1038/s41598-017-00418-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 02/23/2017] [Indexed: 11/08/2022] Open
Abstract
Plasminogen activator inhibitor-1 (PAI-1) is known to protect mice against cardiac fibrosis. It has been speculated that PAI-1 may regulate cardiac fibrosis by inactivating urokinase-type plasminogen activator (uPA) and ultimately plasmin (Pm) generation. However, the in vivo role of PAI-1 in inactivating uPA and limiting the generation of Pm during cardiac fibrosis remains to be established. The objective of this study was to determine if the cardioprotective effect of PAI-1 is mediated through its ability to directly regulate urokinase -mediated activation of plasminogen (Pg). An Angiotensin II (AngII)-aldosterone (Ald) infusion mouse model of hypertension was utilised in this study. Four weeks after AngII-Ald infusion, PAI-1-deficient (PAI-1-/-) mice developed severe cardiac fibrosis. However, a marked reduction in cardiac fibrosis was observed in PAI-1-/-/uPA-/- double knockout mice that was associated with reduced inflammation, lower expression levels of TGF-β and proteases associated with tissue remodeling, and diminished Smad2 signaling. Moreover, total ablation of cardiac fibrosis was observed in PAI-1-/- mice that express inactive plasmin (Pm) but normal levels of zymogen Pg (PAI-1-/-/PgS743A/S743A). Our findings indicate that PAI-1 protects mice from hypertension-induced cardiac fibrosis by inhibiting the generation of active Pm.
Collapse
|
66
|
Abstract
Postoperative atrial fibrillation (PoAF), a common complication of cardiac surgery, contributes significantly to morbidity, mortality, and increasing healthcare costs. Despite advances in surgical and medical management, the overall incidence of PoAF has not changed significantly, partly because of the limited understanding of mechanisms underlying acute surgery-related factors, such as myocardial injury, inflammation, sympathetic activation, and oxidative stress, which play an important role in the initiation of PoAF, whereas a preexisting atrial substrate appears to be more important in the maintenance of this dysrhythmia. Thus, in a majority of patients, PoAF becomes a manifestation of an underlying arrhythmogenic substrate that is unmasked after acute surgical stress. As such, the ability to identify which patients have this proarrhythmic substrate and are, therefore, at high risk for developing AF postoperatively, is important for the improved selection for prophylactic interventions, closer monitoring for complications, and establishing the probability of AF in the long term. This review highlights the role of the underlying substrate in promoting PoAF, proposed mechanisms, and the potential role of serum biomarkers to identify patients at risk for PoAF.
Collapse
|
67
|
Liu LJ, Yao FJ, Lu GH, Xu CG, Xu Z, Tang K, Cheng YJ, Gao XR, Wu SH. The Role of the Rho/ROCK Pathway in Ang II and TGF-β1-Induced Atrial Remodeling. PLoS One 2016; 11:e0161625. [PMID: 27611832 PMCID: PMC5017578 DOI: 10.1371/journal.pone.0161625] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 08/09/2016] [Indexed: 02/05/2023] Open
Abstract
Objectives To study the role of the Rho/ROCK pathway in Ang II and TGF-β1-induced atrial remodeling. Methods and Results A canine atrial fibrillation (AF) model was established by rapid atrial pacing (RAP) of the left atrium. The roles of TGF-β1, the RhoA/ROCK signaling pathway and connective tissue growth factor (CTGF) in atrial remodeling were studied via both in vitro and in vivo experiments. Each of the dogs that received RAP developed persistent AF within 4 weeks. The mRNA expression levels of TGF-β1 (1.32±0.38), Collagen-I(1.33±0.91), CTGF(5.83±3.71), RhoA(1.23±0.57) and ROCK-1 (1.02±0.27) in the left atrium were significantly increased following 4 weeks of RAP. Angiotensin II (Ang II) induced the proliferation of atrial fibroblasts and up-regulated the expression of both CTGF and ROCK-1 in a dose-dependent manner. Simvastatin and Y27632 reversed Ang II-induced CFs proliferation, as well as ROCK-1(0.89±0.05 and 1.27±0.03, respectively) and CTGF (0.87±0.04 and 0.91±0.02, respectively) expression. The expression mRNA of ROCK-1(1.74±0.13) and CTGF (2.28±0.11) can upregulated by TGF-β1, and down-regulated by Simvastatin (1.22±0.03 vs 2.27±0.11), Y27632 (1.01±0.04 vs 1.64±0.03), Los (1.04±0.11 vs 1.26±0.05), respectively. Losartan and Simvastatin attenuated the effects of TGF-β1, inhibited RhoA activity as opposed to RhoA protein expression. Y27632 had no effect on either the expression or the activity of RhoA. Conclusions The increased expression of profibrotic factors (CTGF, ROCK1 and Smad2/3) played an important role in our RAP-induced AF model. Increased atrial profibrotic factors involve the activation of either the TGF-β1/RhoA/ROCK-1 or the TGF-β1/Smad2/3 signaling pathway.
Collapse
Affiliation(s)
- Li-Juan Liu
- Department of Cardiology and Department of Ultrasonography (Feng-Juan Yao), the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Feng-Juan Yao
- Department of Cardiology and Department of Ultrasonography (Feng-Juan Yao), the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Gui-Hua Lu
- Department of Cardiology and Department of Ultrasonography (Feng-Juan Yao), the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Cheng-Gui Xu
- Department of Cardiology and Department of Ultrasonography (Feng-Juan Yao), the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhe Xu
- Department of Cardiology and Department of Ultrasonography (Feng-Juan Yao), the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Kai Tang
- Department of Cardiology and Department of Ultrasonography (Feng-Juan Yao), the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yun-Jiu Cheng
- Department of Cardiology and Department of Ultrasonography (Feng-Juan Yao), the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiu-Ren Gao
- Department of Cardiology and Department of Ultrasonography (Feng-Juan Yao), the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- * E-mail: (SW); (XG)
| | - Su-Hua Wu
- Department of Cardiology and Department of Ultrasonography (Feng-Juan Yao), the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- * E-mail: (SW); (XG)
| |
Collapse
|
68
|
Polejaeva IA, Ranjan R, Davies CJ, Regouski M, Hall J, Olsen AL, Meng Q, Rutigliano HM, Dosdall DJ, Angel NA, Sachse FB, Seidel T, Thomas AJ, Stott R, Panter KE, Lee PM, Van Wettere AJ, Stevens JR, Wang Z, MacLeod RS, Marrouche NF, White KL. Increased Susceptibility to Atrial Fibrillation Secondary to Atrial Fibrosis in Transgenic Goats Expressing Transforming Growth Factor-β1. J Cardiovasc Electrophysiol 2016; 27:1220-1229. [PMID: 27447370 DOI: 10.1111/jce.13049] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 06/06/2016] [Accepted: 06/17/2016] [Indexed: 01/06/2023]
Abstract
INTRODUCTION Large animal models of progressive atrial fibrosis would provide an attractive platform to study relationship between structural and electrical remodeling in atrial fibrillation (AF). Here we established a new transgenic goat model of AF with cardiac specific overexpression of TGF-β1 and investigated the changes in the cardiac structure and function leading to AF. METHODS AND RESULTS Transgenic goats with cardiac specific overexpression of constitutively active TGF-β1 were generated by somatic cell nuclear transfer. We examined myocardial tissue, ECGs, echocardiographic data, and AF susceptibility in transgenic and wild-type control goats. Transgenic goats exhibited significant increase in fibrosis and myocyte diameters in the atria compared to controls, but not in the ventricles. P-wave duration was significantly greater in transgenic animals starting at 12 months of age, but no significant chamber enlargement was detected, suggesting conduction slowing in the atria. Furthermore, this transgenic goat model exhibited a significant increase in AF vulnerability. Six of 8 transgenic goats (75%) were susceptible to AF induction and exhibited sustained AF (>2 minutes), whereas none of 6 controls displayed sustained AF (P < 0.01). Length of induced AF episodes was also significantly greater in the transgenic group compared to controls (687 ± 212.02 seconds vs. 2.50 ± 0.88 seconds, P < 0.0001), but no persistent or permanent AF was observed. CONCLUSION A novel transgenic goat model with a substrate for AF was generated. In this model, cardiac overexpression of TGF-β1 led to an increase in fibrosis and myocyte size in the atria, and to progressive P-wave prolongation. We suggest that these factors underlie increased AF susceptibility.
Collapse
Affiliation(s)
- Irina A Polejaeva
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT
| | - Ravi Ranjan
- CARMA Center, Division of Cardiology, University of Utah, Salt Lake City, UT.,Department of Bioengineering, University of Utah, Salt Lake City, UT
| | - Christopher J Davies
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT
| | - Misha Regouski
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT
| | - Justin Hall
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT
| | - Aaron L Olsen
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT
| | - Qinggang Meng
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT
| | - Heloisa M Rutigliano
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT
| | - Derek J Dosdall
- Center for Engineering Innovation, University of Utah, Salt Lake City, UT
| | - Nathan A Angel
- CARMA Center, Division of Cardiology, University of Utah, Salt Lake City, UT
| | - Frank B Sachse
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT.,Department of Bioengineering, University of Utah, Salt Lake City, UT
| | - Thomas Seidel
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT
| | - Aaron J Thomas
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT
| | - Rusty Stott
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT
| | - Kip E Panter
- USDA ARS Poisonous Plant Research Laboratory, Logan, UT
| | - Pamela M Lee
- College of Veterinary Medicine, Washington State University, Pullman, WA
| | - Arnaud J Van Wettere
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT
| | - John R Stevens
- Department of Mathematics and Statistics, Utah State University, Logan, UT
| | - Zhongde Wang
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT
| | - Rob S MacLeod
- CARMA Center, Division of Cardiology, University of Utah, Salt Lake City, UT.,Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT
| | - Nassir F Marrouche
- CARMA Center, Division of Cardiology, University of Utah, Salt Lake City, UT
| | - Kenneth L White
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT
| |
Collapse
|
69
|
Protective role of heme oxygenase-1 in atrial remodeling. Basic Res Cardiol 2016; 111:58. [DOI: 10.1007/s00395-016-0577-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 08/22/2016] [Indexed: 12/26/2022]
|
70
|
Viral Vector-Based Targeting of miR-21 in Cardiac Nonmyocyte Cells Reduces Pathologic Remodeling of the Heart. Mol Ther 2016; 24:1939-1948. [PMID: 27545313 PMCID: PMC5154480 DOI: 10.1038/mt.2016.166] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 08/07/2016] [Indexed: 12/23/2022] Open
Abstract
Systemic inhibition of miR-21 has proven effective against myocardial fibrosis and dysfunction, while studies in cardiac myocytes suggested a protective role in this cell type. Considering potential implications for therapy, we aimed to determine the cell fraction where miR-21 exerts its pathological activity. We developed a viral vector-based strategy for gene targeting of nonmyocyte cardiac cells in vivo and compared global to cardiac myocyte-specific and nonmyocyte-specific deletion of miR-21 in chronic left ventricular pressure overload. Murine moloney virus and serotype 9 of adeno-associated virus were engineered to encode improved Cre recombinase for genetic deletion in miR-21fl/fl mice. Pericardial injection of murine moloney virus-improved Cre recombinase to neonates achieved highly selective genetic ablation of miR-21 in nonmyocyte cardiac cells, identified as cardiac fibroblasts and endothelial cells. Upon left ventricular pressure overload, cardiac function was only preserved in mice with miR-21 deficiency in nonmyocyte cardiac cells, but not in mice with global or cardiac myocyte-specific ablation. Our data demonstrate that miR-21 exerts its pathologic activity directly in cardiac nonmyocytes and encourage further development of antimiR-21 therapy toward cellular tropism.
Collapse
|
71
|
Zhao Z, Wang Y, Chen Y, Wang X, Li J, Yuan M, Liu T, Li G. Cilostazol Prevents Atrial Structural Remodeling through the MEK/ERK Pathway in a Canine Model of Atrial Tachycardia. Cardiology 2016; 135:240-248. [PMID: 27532517 DOI: 10.1159/000447769] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 06/20/2016] [Indexed: 10/11/2023]
Abstract
OBJECTIVES Atrial fibrillation (AF) is the most common sustained arrhythmia in clinical practice. Atrial structural remodeling (ASR), particularly atrial fibrosis, is an important contributor to the AF substrate. This study aimed to investigate the preventive effects of the phosphodiesterase 3 inhibitor cilostazol on ASR and its potential molecular mechanisms in a canine model of rapid atrial pacing (RAP). METHODS Thirty dogs were assigned to sham (Sham), paced/ no treatment (Paced) and paced + cilostazol 5 mg/kg/day (Paced + cilo) groups, with 10 dogs in each group. RAP at 500 beats/min was maintained for 2 weeks, while the Sham group was instrumented without pacing. Cilostazol was provided orally during pacing. Western blotting, RT-PCR and pathology were used to assess ASR. RESULTS Cilostazol attenuated atrial interstitial fibrosis and structural remodeling in canines with RAP. MEK/ERK transduction pathway gene expression was upregulated in the Paced group compared with the Sham group. Cilostazol markedly alleviated these changes in the MEK/ERK pathway. Transforming growth factor-β1 protein expression in the Paced group was significantly higher than in the Sham group (p < 0.01), and was significantly reduced by cilostazol (p < 0.01). CONCLUSIONS Our findings suggest that cilostazol is beneficial for prevention and treatment in atrial tachycardia-induced ASR in a canine model of RAP.
Collapse
Affiliation(s)
- Zhiqiang Zhao
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| | | | | | | | | | | | | | | |
Collapse
|
72
|
Lu YY, Lin YK, Kao YH, Chung CC, Yeh YH, Chen SA, Chen YJ. Collagen regulates transforming growth factor-β receptors of HL-1 cardiomyocytes through activation of stretch and integrin signaling. Mol Med Rep 2016; 14:3429-36. [PMID: 27573189 DOI: 10.3892/mmr.2016.5635] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 07/13/2016] [Indexed: 11/06/2022] Open
Abstract
The extracellular matrix (ECM) and transforming growth factor-β (TGF)-β are important in cardiac fibrosis, however, the effects of the ECM on TGF‑β signaling remain to be fully elucidated. The aims of the present study were to evaluate the role of collagen in TGF‑β signaling and examine the underlying mechanisms. In the present study, western blot analysis was used to examine TGF‑β signaling in HL‑1 cells treated with and without (control) type I collagen (10 µg/ml), which was co‑administered with either an anti‑β1 integrin antibody (10 µg/ml) or a stretch‑activated channel inhibitor (gadolinium; 50 µM). Cell proliferation and adhesion assays were used to investigate the roles of integrin, mechanical stretch and mitogen‑activated protein kinases (MAPKs) on cell proliferation and adhesion. The type I collagen (10 µg/ml)‑treated HL‑1 cells were incubated with or without anti‑β1 integrin antibody (10 µg/ml), gadolinium (50 µM) or inhibitors of p38 (SB203580; 3 µM), extracellular signal‑regulated kinase (ERK; PD98059; 50 µM) and c‑Jun N‑terminal kinase (JNK; SP600125; 50 µM). Compared with the control cells, the collagen‑treated HL‑1 cells had lower expression levels of type I and type II TGF‑β receptors (TGFβRI and TGFβRII), with an increase in phosphorylated focal adhesion kinase (FAK), p38 and ERK1/2, and a decrease in JNK. Incubation with the anti‑β1 integrin antibody reversed the collagen‑induced downregulation of the expression of TGFβRII and phosphorylated FAK. Gadolinium downregulated the expression levels of TGFβRI and small mothers against decapentaplegic (Smad)2/3, and decreased the levels of phosphorylated p38, ERK1/2 and JNK. In addition, gadolinium reversed the collagen‑induced activation of p38 and ERK1/2. In the presence of gadolinium and anti‑β1 integrin antibody, collagen regulated the expression levels of TGFβRI, TGFβRII and Smad2/3, but did not alter the phosphorylation of p38, ERK1/2 or JNK. In addition, collagen increased cell proliferation and adhesion, and this collagen‑induced cell proliferation was inhibited by the anti‑β1 integrin antibody and ERK inhibitor. Taken together, the data obtained suggested that collagen differentially regulated the expression levels of TGFβRI and TGFβRII, and modulated the phosphorylation of MAPKs through integrin‑ or stretch‑dependent and ‑independent signaling pathways.
Collapse
Affiliation(s)
- Yen-Yu Lu
- Division of Cardiology, Department of Internal Medicine, Sijhih Cathay General Hospital, New Taipei City 221, Taiwan, R.O.C
| | - Yung-Kuo Lin
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan, R.O.C
| | - Yu-Hsun Kao
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan, R.O.C
| | - Cheng-Chih Chung
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan, R.O.C
| | - Yung-Hsin Yeh
- Cardiovascular Division, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Tao‑Yuan 333, Taiwan, R.O.C
| | - Shih-Ann Chen
- School of Medicine, National Yang-Ming University, Taipei 112, Taiwan, R.O.C
| | - Yi-Jen Chen
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan, R.O.C
| |
Collapse
|
73
|
Zhang L, Zhang N, Tang X, Liu F, Luo S, Xiao H. Increased α-Actinin-2 Expression in the Atrial Myocardium of Patients with Atrial Fibrillation Related to Rheumatic Heart Disease. Cardiology 2016; 135:151-159. [PMID: 27344599 DOI: 10.1159/000446362] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 04/22/2016] [Indexed: 11/19/2022]
Abstract
OBJECTIVES Atrial fibrosis, a marker of atrial structural remodeling, plays a critical role in atrial fibrillation (AF). α- Actinin-2 is associated with structural remodeling related to stretching. The transforming growth factor-β1 (TGF-β1)/Smad pathway plays an important role in atrial fibrosis. We investigated the effects of the TGF-β1/Smad signaling pathway on α-actinin-2 in atrial fibrosis in patients with AF. METHODS Forty-one right atrial specimens obtained from patients with rheumatic heart disease (RHD) were divided into a chronic (c)AF group, i.e. RHD + cAF (n = 29), and a sinus rhythm group, i.e. RHD + sinus rhythm (n = 12). Patients with congenital heart disease (CHD) and sinus rhythm who underwent heart surgery served as controls (n = 10). Fibrosis was assessed by histological examination, and expression of α-actinin-2, TGF-β1 and Smad2/phosphorylated Smad2 (p-Smad2) was evaluated by immunohistochemistry, quantitative real-time PCR and Western blotting. In rat atrial fibroblasts treated with TGF-β1, the collagen content was measured using hydroxyproline detection, and α-actinin-2 and p-Smad2 were evaluated by semiquantitative reverse-transcription PCR and Western blotting. RESULTS The histology results revealed a significant increase in atrial fibrosis in AF patients. The collagen content, mRNA and protein expression levels of α-actinin-2 and the components of the TGF-β1/Smad signaling pathway were significantly gradually increased in the CHD + sinus rhythm, RHD + sinus rhythm and RHD + cAF groups (p < 0.05). The mRNA and protein levels of α-actinin-2 and TGF-β1 in RHD patients were positively correlated with the collagen volume fraction. A positive correlation between the expression of α-actinin-2 and TGF-β1 was also observed. In rat atrial fibroblasts treated with TGF-β1, the collagen content was greater than that in the control group (p < 0.05), and the expression levels of α- actinin-2 and p-Smad2 were also upregulated (p < 0.05). CONCLUSIONS α-Actinin-2 expression was increased in the atrial tissues of patients with AF secondary to RHD. α-Actinin-2 was upregulated via the TGF-β1/Smad pathway in atrial fibroblasts, which suggests that it may be involved in TGF-β1/Smad pathway-induced atrial fibrosis in patients with AF.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | | | | | | | | | | |
Collapse
|
74
|
Novel therapeutic strategies targeting fibroblasts and fibrosis in heart disease. Nat Rev Drug Discov 2016; 15:620-638. [PMID: 27339799 DOI: 10.1038/nrd.2016.89] [Citation(s) in RCA: 236] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Our understanding of the functions of cardiac fibroblasts has moved beyond their roles in heart structure and extracellular matrix generation and now includes their contributions to paracrine, mechanical and electrical signalling during ontogenesis and normal cardiac activity. Fibroblasts also have central roles in pathogenic remodelling during myocardial ischaemia, hypertension and heart failure. As key contributors to scar formation, they are crucial for tissue repair after interventions including surgery and ablation. Novel experimental approaches targeting cardiac fibroblasts are promising potential therapies for heart disease. Indeed, several existing drugs act, at least partially, through effects on cardiac connective tissue. This Review outlines the origins and roles of fibroblasts in cardiac development, homeostasis and disease; illustrates the involvement of fibroblasts in current and emerging clinical interventions; and identifies future targets for research and development.
Collapse
|
75
|
Eplerenone inhibits atrial fibrosis in mutant TGF-β1 transgenic mice. SCIENCE CHINA-LIFE SCIENCES 2016; 59:1042-1047. [DOI: 10.1007/s11427-016-0037-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Accepted: 05/12/2016] [Indexed: 12/14/2022]
|
76
|
Effect of irbesartan on development of atrial fibrosis and atrial fibrillation in a canine atrial tachycardia model with left ventricular dysfunction, association with p53. Heart Vessels 2016; 31:2053-2060. [PMID: 27236656 DOI: 10.1007/s00380-016-0853-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 05/20/2016] [Indexed: 12/20/2022]
Abstract
Effects of an angiotensin II receptor blocker, irbesartan (IRB), on the development of atrial fibrosis and atrial fibrillation (AF) were assessed in a canine model of atrial tachycardia remodeling (ATR) with left ventricular dysfunction, together with its possible association with involvement of p53. Atrial tachypacing (400 bpm for 4 weeks) was used to induce ATR in beagles treated with placebo (ATR-dogs, n = 6) or irbesartan (IRB-dogs, n = 5). Non-paced sham dogs served as control (Control-dogs, n = 4). ATR- and IRB-dogs developed tachycardia-induced left ventricular dysfunction. Atrial effective refractory period (AERP) shortened (83 ± 5 ms, p < 0.05), inter-atrial conduction time prolonged (72 ± 2 ms, p < 0.05), and AF duration increased (29 ± 5 s, p < 0.05 vs. baseline) after 4 weeks in ATR-dogs. ATR-dogs also had a larger area of atrial fibrous tissue (5.2 ± 0.5 %, p < 0.05 vs. Control). All these changes, except for AERP, were attenuated in IRB-dogs (92 ± 3 ms, 56 ± 3 ms, 9 ± 5 s, and 2.5 ± 0.7 %, respectively; p < 0.05 vs. ATR for each). In ATR-dogs, p53 expression in the left atrium decreased by 42 % compared with Control-dogs (p < 0.05); however, it was highly expressed in IRB-dogs (+89 % vs. ATR). Transforming growth factor (TGF)-β1 expression was enhanced in ATR-dogs (p < 0.05 vs. Control) but reduced in IRB-dogs (p < 0.05 vs. ATR). Irbesartan suppresses atrial fibrosis and AF development in a canine ATR model with left ventricular dysfunction in association with p53.
Collapse
|
77
|
Ongstad EL, Gourdie RG. Can heart function lost to disease be regenerated by therapeutic targeting of cardiac scar tissue? Semin Cell Dev Biol 2016; 58:41-54. [PMID: 27234380 DOI: 10.1016/j.semcdb.2016.05.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 05/18/2016] [Accepted: 05/23/2016] [Indexed: 01/14/2023]
Abstract
Myocardial infarction results in scar tissue that cannot actively contribute to heart mechanical function and frequently causes lethal arrhythmias. The healing response after infarction involves inflammation, biochemical signaling, changes in cellular phenotype, activity, and organization, and alterations in electrical conduction due to variations in cell and tissue geometry and alterations in protein expression, organization, and function - particularly in membrane channels. The intensive research focus on regeneration of myocardial tissues has, as of yet, only met with modest success, with no near-term prospect of improving standard-of-care for patients with heart disease. An alternative concept for novel therapeutic approach is the rejuvenation of cardiac electrical and mechanical properties through the modification of scar tissue. Several peptide therapeutics, locally applied genetic therapies, or delivery of genetically modified cells have shown promise in improving the characteristics of the fibrous scar and post-myocardial infarction prognosis in experimental models. This review highlights several factors that contribute to arrhythmogenesis in scar formation and how these might be targeted to regenerate some of the electrical and mechanical function of the post-MI scar.
Collapse
Affiliation(s)
- Emily L Ongstad
- Center for Heart and Regenerative Medicine Research, Virginia Tech Carilion Research Institute, 2 Riverside Circle, Roanoke, VA 24016, USA.
| | - Robert G Gourdie
- Center for Heart and Regenerative Medicine Research, Virginia Tech Carilion Research Institute, 2 Riverside Circle, Roanoke, VA 24016, USA; Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences, 317 Kelly Hall, Stanger Street, Blacksburg, VA 24061, USA; Department of Emergency Medicine, Carilion Clinic, 1906 Belleview Avenue, Roanoke VA 24014, USA.
| |
Collapse
|
78
|
Kunamalla A, Ng J, Parini V, Yoo S, McGee KA, Tomson TT, Gordon D, Thorp EB, Lomasney J, Zhang Q, Shah S, Browne S, Knight BP, Passman R, Goldberger JJ, Aistrup G, Arora R. Constitutive Expression of a Dominant-Negative TGF-β Type II Receptor in the Posterior Left Atrium Leads to Beneficial Remodeling of Atrial Fibrillation Substrate. Circ Res 2016; 119:69-82. [PMID: 27217399 DOI: 10.1161/circresaha.115.307878] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 05/23/2016] [Indexed: 11/16/2022]
Abstract
RATIONALE Fibrosis is an important structural contributor to formation of atrial fibrillation (AF) substrate in heart failure. Transforming growth factor-β (TGF-β) signaling is thought to be intricately involved in creation of atrial fibrosis. OBJECTIVE We hypothesized that gene-based expression of dominant-negative type II TGF-β receptor (TGF-β-RII-DN) in the posterior left atrium in a canine heart failure model will sufficiently attenuate fibrosis-induced changes in atrial conduction and restitution to decrease AF. Because AF electrograms are thought to reflect AF substrate, we further hypothesized that TGF-β-RII-DN would lead to increased fractionation and decreased organization of AF electrograms. METHODS AND RESULTS Twenty-one dogs underwent injection+electroporation in the posterior left atrium of plasmid expressing a dominant-negative TGF-β type II receptor (pUBc-TGFβ-DN-RII; n=9) or control vector (pUBc-LacZ; n=12), followed by 3 to 4 weeks of right ventricular tachypacing (240 bpm). Compared with controls, dogs treated with pUBC-TGFβ-DN-RII demonstrated an attenuated increase in conduction inhomogeneity, flattening of restitution slope and decreased duration of induced AF, with AF electrograms being more fractionated and less organized in pUBc-TGFβ-DN-RII versus pUBc-LacZ dogs. Tissue analysis revealed a significant decrease in replacement/interstitial fibrosis, p-SMAD2/3 and p-ERK1/2. CONCLUSIONS Targeted gene-based reduction of TGF-β signaling in the posterior left atrium-with resulting decrease in replacement fibrosis-led to beneficial remodeling of both conduction and restitution characteristics of the posterior left atrium, translating into a decrease in AF and increased complexity of AF electrograms. In addition to providing mechanistic insights, this data may have important diagnostic and therapeutic implications for AF.
Collapse
Affiliation(s)
- Aaron Kunamalla
- From the Feinberg Cardiovascular Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Jason Ng
- From the Feinberg Cardiovascular Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Vamsi Parini
- From the Feinberg Cardiovascular Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Shin Yoo
- From the Feinberg Cardiovascular Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Kate A McGee
- From the Feinberg Cardiovascular Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Todd T Tomson
- From the Feinberg Cardiovascular Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - David Gordon
- From the Feinberg Cardiovascular Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Edward B Thorp
- From the Feinberg Cardiovascular Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Jon Lomasney
- From the Feinberg Cardiovascular Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Qiang Zhang
- From the Feinberg Cardiovascular Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Sanjiv Shah
- From the Feinberg Cardiovascular Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Suzanne Browne
- From the Feinberg Cardiovascular Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Bradley P Knight
- From the Feinberg Cardiovascular Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Rod Passman
- From the Feinberg Cardiovascular Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Jeffrey J Goldberger
- From the Feinberg Cardiovascular Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Gary Aistrup
- From the Feinberg Cardiovascular Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Rishi Arora
- From the Feinberg Cardiovascular Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL.
| |
Collapse
|
79
|
Pellman J, Sheikh F. Atrial fibrillation: mechanisms, therapeutics, and future directions. Compr Physiol 2016; 5:649-65. [PMID: 25880508 DOI: 10.1002/cphy.c140047] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Atrial fibrillation (AF) is the most prevalent cardiac arrhythmia, affecting 1% to 2% of the general population. It is characterized by rapid and disorganized atrial activation leading to impaired atrial function, which can be diagnosed on an EKG by lack of a P-wave and irregular QRS complexes. AF is associated with increased morbidity and mortality and is a risk factor for embolic stroke and worsening heart failure. Current research on AF support and explore the hypothesis that initiation and maintenance of AF require pathophysiological remodeling of the atria, either specifically as in lone AF or secondary to other heart disease as in heart failure-associated AF. Remodeling in AF can be grouped into three categories that include: (i) electrical remodeling, which includes modulation of L-type Ca(2+) current, various K(+) currents and gap junction function; (ii) structural remodeling, which includes changes in tissues properties, size, and ultrastructure; and (iii) autonomic remodeling, including altered sympathovagal activity and hyperinnervation. Electrical, structural, and autonomic remodeling all contribute to creating an AF-prone substrate which is able to produce AF-associated electrical phenomena including a rapidly firing focus, complex multiple reentrant circuit or rotors. Although various remodeling events occur in AF, current AF therapies focus on ventricular rate and rhythm control strategies using pharmacotherapy and surgical interventions. Recent progress in the field has started to focus on the underlying substrate that drives and maintains AF (termed upstream therapies); however, much work is needed in this area. Here, we review current knowledge of AF mechanisms, therapies, and new areas of investigation.
Collapse
Affiliation(s)
- Jason Pellman
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | | |
Collapse
|
80
|
Zhu J, Zhang X, Li L, Su G. Protective effects of epigallocatechin-3 gallate on atrial electrical and structural remodeling in a rabbit rapid atrial pacing model. Cell Biochem Biophys 2015; 71:897-903. [PMID: 25312479 DOI: 10.1007/s12013-014-0280-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Epigallocatechin-3 gallate (EGCG) is the major catechin in green tea. The aim of this study is to investigate the effects of EGCG on atrial electrical and structural remodeling in a rabbit rapid atrial pacing (RAP) model. New Zealand white rabbits were subjected to RAP with or without EGCG treatment. The atrial electrophysiology was studied. ELISA, Western blots, and RT-PCR were performed to determine the level of the inflammation markers, oxidative stress, and fibrogenic agents. Atrial tissue was stained with Masson's trichrome stain for fibrosis detection. RAP rabbits showed a significantly shorter atrial effective refractory period than control rabbits. Higher AF inducibility and longer AF duration were seen in the RAP group. AERP of rabbits received high dose EGCG were prolonged compared to RAP rabbits, and AF inducibility and duration of rabbits received high dose EGCG were lower. RAP rabbits have higher inflammation markers, higher oxidative stress, and more significant fibrosis within atrium, while high dose intervention of EGCG can lower the inflammation, oxidative stress, and fibrosis induced by RAP. Results showed that EGCG have protective effects on atrial electrical and structural remodeling in a rabbit RAP model in terms of attenuating of inflammation and oxidative stress.
Collapse
Affiliation(s)
- Jifa Zhu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China,
| | | | | | | |
Collapse
|
81
|
Kim GE, Ross JL, Xie C, Su KN, Zaha VG, Wu X, Palmeri M, Ashraf M, Akar JG, Russell KS, Akar FG, Young LH. LKB1 deletion causes early changes in atrial channel expression and electrophysiology prior to atrial fibrillation. Cardiovasc Res 2015; 108:197-208. [PMID: 26378152 PMCID: PMC4571838 DOI: 10.1093/cvr/cvv212] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 07/09/2015] [Accepted: 07/14/2015] [Indexed: 12/19/2022] Open
Abstract
AIMS Liver kinase B1 (LKB1) is a protein kinase that activates the metabolic regulator AMP-activated protein kinase (AMPK) and other related kinases. Deletion of LKB1 in mice leads to cardiomyopathy and atrial fibrillation (AF). However, the specific role of the LKB1 pathway in early atrial biology remains unknown. Thus, we investigated whether LKB1 deletion altered atrial channel expression and electrophysiological function in a cardiomyocyte-specific knockout mouse model. METHODS AND RESULTS We performed a systematic comparison of αMHC-Cre LKB1(fl/fl) and littermate LKB1(fl/fl) male mice. This included analysis of gene expression, histology, and echocardiography, as well as cellular and tissue-level electrophysiology using patch-clamp recordings in vitro, optical mapping ex vivo, and ECG recordings in vivo. At postnatal day 1, atrial depolarization was prolonged, and Nav1.5 and Cx40 expression were markedly down-regulated in MHC-Cre LKB1(fl/fl) mice. Inward sodium current density was significantly decreased in MHC-Cre LKB1(fl/fl) neonatal atrial myocytes. Subsequently, additional alterations in atrial channel expression, atrial fibrosis, and spontaneous onset of AF developed by 2 weeks of age. In adult mice, abnormalities of interatrial conduction and bi-atrial electrical coupling were observed, likely promoting the perpetuation of AF. Mice with AMPK-inactivated hearts demonstrated modest overlap in channel expression with MHC-Cre LKB1(fl/fl) hearts, but retained normal structure, electrophysiological function and contractility. CONCLUSIONS Deletion of LKB1 causes early defects in atrial channel expression, action potential generation and conduction, which precede widespread atrial remodelling, fibrosis and AF. LKB1 is critical for normal atrial growth and electrophysiological function.
Collapse
Affiliation(s)
- Grace E Kim
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT 06520, USA
| | - Jenna L Ross
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Chaoqin Xie
- Cardiovascular Research Center, Mt. Sinai School of Medicine, New York, NY 10029, USA
| | - Kevin N Su
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT 06520, USA
| | - Vlad G Zaha
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Xiaohong Wu
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Monica Palmeri
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Mohammed Ashraf
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Joseph G Akar
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Kerry S Russell
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Fadi G Akar
- Cardiovascular Research Center, Mt. Sinai School of Medicine, New York, NY 10029, USA
| | - Lawrence H Young
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT 06520, USA Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| |
Collapse
|
82
|
|
83
|
Zhou T, Wang Z, Fan J, Chen S, Tan Z, Yang H, Yin Y. Angiotensin-converting enzyme-2 overexpression improves atrial remodeling and function in a canine model of atrial fibrillation. J Am Heart Assoc 2015; 4:e001530. [PMID: 25792125 PMCID: PMC4392435 DOI: 10.1161/jaha.114.001530] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Atrial fibrosis is an important factor in initiating and maintaining atrial fibrillation. The purpose of this study was to test the hypothesis that atrial angiotensin-converting enzyme-2 (ACE2) overexpression might inhibit atrial collagen accumulation and improve atrial remodeling in a canine atrial pacing model. METHODS AND RESULTS Thirty-two mongrel dogs of both genders were divided randomly into 4 groups: sham-operated, control, gene therapy with adenovirus-enhanced green fluorescent protein (Ad-EGFP), and gene therapy with Ad-ACE2. All of the dogs in the control, Ad-EGFP, and Ad-ACE2 groups were paced at 450 bpm for a period of 14 days. The dogs in the sham group were instrumented without pacing. After 2 weeks, all of the dogs underwent a thoracotomy operation and received epicardial gene painting. On post-gene transfer day 21, the animals underwent electrophysiology, histology, and molecular studies. The percentage of fibrosis in the Ad-ACE2 group was markedly lower than the percentage in the control and Ad-EGFP groups. Compared with the other groups, ACE2 expression was increased significantly in the Ad-ACE2 group. Compared with the sham and Ad-ACE2 groups, the expression levels of transforming growth factor-β1 and Smad3 were significantly higher in the Ad-EGFP and control groups; however, the expression levels of Smad7 were lower in the atrial tissue as detected by Western blot and reverse transcription polymerase chain reaction. CONCLUSIONS Our results demonstrate that the overexpression of ACE2 inhibits atrial collagen accumulation and improves left atrial remodeling and function in a canine model of atrial fibrillation. Thus, targeted gene ACE2 therapy provides a promising approach for the treatment of atrial fibrillation.
Collapse
Affiliation(s)
- Tingquan Zhou
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China (T.Z., Z.W., J.F., S.C., Z.T., H.Y., Y.Y.)
| | - Zhenglong Wang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China (T.Z., Z.W., J.F., S.C., Z.T., H.Y., Y.Y.)
| | - Jinqi Fan
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China (T.Z., Z.W., J.F., S.C., Z.T., H.Y., Y.Y.)
| | - Shaojie Chen
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China (T.Z., Z.W., J.F., S.C., Z.T., H.Y., Y.Y.)
| | - Zhen Tan
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China (T.Z., Z.W., J.F., S.C., Z.T., H.Y., Y.Y.)
| | - Hanxuan Yang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China (T.Z., Z.W., J.F., S.C., Z.T., H.Y., Y.Y.)
| | - Yuehui Yin
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China (T.Z., Z.W., J.F., S.C., Z.T., H.Y., Y.Y.)
| |
Collapse
|
84
|
Ghavami S, Cunnington RH, Gupta S, Yeganeh B, Filomeno KL, Freed DH, Chen S, Klonisch T, Halayko AJ, Ambrose E, Singal R, Dixon IMC. Autophagy is a regulator of TGF-β1-induced fibrogenesis in primary human atrial myofibroblasts. Cell Death Dis 2015; 6:e1696. [PMID: 25789971 PMCID: PMC4385916 DOI: 10.1038/cddis.2015.36] [Citation(s) in RCA: 167] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 01/09/2015] [Accepted: 01/16/2015] [Indexed: 01/07/2023]
Abstract
Transforming growth factor-β1 (TGF-β1) is an important regulator of fibrogenesis in heart disease. In many other cellular systems, TGF-β1 may also induce autophagy, but a link between its fibrogenic and autophagic effects is unknown. Thus we tested whether or not TGF-β1-induced autophagy has a regulatory function on fibrosis in human atrial myofibroblasts (hATMyofbs). Primary hATMyofbs were treated with TGF-β1 to assess for fibrogenic and autophagic responses. Using immunoblotting, immunofluorescence and transmission electron microscopic analyses, we found that TGF-β1 promoted collagen type Iα2 and fibronectin synthesis in hATMyofbs and that this was paralleled by an increase in autophagic activation in these cells. Pharmacological inhibition of autophagy by bafilomycin-A1 and 3-methyladenine decreased the fibrotic response in hATMyofb cells. ATG7 knockdown in hATMyofbs and ATG5 knockout (mouse embryonic fibroblast) fibroblasts decreased the fibrotic effect of TGF-β1 in experimental versus control cells. Furthermore, using a coronary artery ligation model of myocardial infarction in rats, we observed increases in the levels of protein markers of fibrosis, autophagy and Smad2 phosphorylation in whole scar tissue lysates. Immunohistochemistry for LC3β indicated the localization of punctate LC3β with vimentin (a mesenchymal-derived cell marker), ED-A fibronectin and phosphorylated Smad2. These results support the hypothesis that TGF-β1-induced autophagy is required for the fibrogenic response in hATMyofbs.
Collapse
Affiliation(s)
- S Ghavami
- 1] Department of Physiology, Manitoba Institute of Child Health, Winnipeg, Manitoba, Canada [2] Biology of Breathing Group, Manitoba Institute of Child Health, Winnipeg, Manitoba, Canada [3] Department of Physiology and Institute of Cardiovascular Sciences, St. Boniface Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada [4] Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Manitoba, Canada
| | - R H Cunnington
- Department of Physiology and Institute of Cardiovascular Sciences, St. Boniface Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - S Gupta
- Department of Physiology and Institute of Cardiovascular Sciences, St. Boniface Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - B Yeganeh
- 1] Department of Physiology, Manitoba Institute of Child Health, Winnipeg, Manitoba, Canada [2] Biology of Breathing Group, Manitoba Institute of Child Health, Winnipeg, Manitoba, Canada [3] Department of Physiology and Institute of Cardiovascular Sciences, St. Boniface Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - K L Filomeno
- Department of Physiology and Institute of Cardiovascular Sciences, St. Boniface Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - D H Freed
- Department of Physiology and Institute of Cardiovascular Sciences, St. Boniface Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - S Chen
- Department of Physiology and Institute of Cardiovascular Sciences, St. Boniface Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - T Klonisch
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Manitoba, Canada
| | - A J Halayko
- 1] Department of Physiology, Manitoba Institute of Child Health, Winnipeg, Manitoba, Canada [2] Biology of Breathing Group, Manitoba Institute of Child Health, Winnipeg, Manitoba, Canada [3] Department of Internal Medicine, Manitoba Institute of Child Health, Winnipeg, Manitoba, Canada
| | - E Ambrose
- Department of Physiology and Institute of Cardiovascular Sciences, St. Boniface Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - R Singal
- Cardiac Sciences Program, St. Boniface General Hospital, Winnipeg, Manitoba, Canada
| | - I M C Dixon
- Department of Physiology and Institute of Cardiovascular Sciences, St. Boniface Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
85
|
Csepe TA, Kalyanasundaram A, Hansen BJ, Zhao J, Fedorov VV. Fibrosis: a structural modulator of sinoatrial node physiology and dysfunction. Front Physiol 2015; 6:37. [PMID: 25729366 PMCID: PMC4325882 DOI: 10.3389/fphys.2015.00037] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Accepted: 01/24/2015] [Indexed: 01/01/2023] Open
Abstract
Heart rhythm is initialized and controlled by the Sinoatrial Node (SAN), the primary pacemaker of the heart. The SAN is a heterogeneous multi-compartment structure characterized by clusters of specialized cardiomyocytes enmeshed within strands of connective tissue or fibrosis. Intranodal fibrosis is emerging as an important modulator of structural and functional integrity of the SAN pacemaker complex. In adult human hearts, fatty tissue and fibrosis insulate the SAN from the hyperpolarizing effect of the surrounding atria while electrical communication between the SAN and right atrium is restricted to discrete SAN conduction pathways. The amount of fibrosis within the SAN is inversely correlated with heart rate, while age and heart size are positively correlated with fibrosis. Pathological upregulation of fibrosis within the SAN may lead to tachycardia-bradycardia arrhythmias and cardiac arrest, possibly due to SAN reentry and exit block, and is associated with atrial fibrillation, ventricular arrhythmias, heart failure and myocardial infarction. In this review, we will discuss current literature on the role of fibrosis in normal SAN structure and function, as well as the causes and consequences of SAN fibrosis upregulation in disease conditions.
Collapse
Affiliation(s)
- Thomas A Csepe
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center Columbus, OH, USA
| | - Anuradha Kalyanasundaram
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center Columbus, OH, USA
| | - Brian J Hansen
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center Columbus, OH, USA
| | - Jichao Zhao
- Auckland Bioengineering Institute, The University of Auckland Auckland, New Zealand
| | - Vadim V Fedorov
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center Columbus, OH, USA
| |
Collapse
|
86
|
Recombinant neuregulin 1 does not activate cardiomyocyte DNA synthesis in normal or infarcted adult mice. PLoS One 2014; 9:e115871. [PMID: 25545368 PMCID: PMC4278834 DOI: 10.1371/journal.pone.0115871] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 11/28/2014] [Indexed: 11/19/2022] Open
Abstract
Objectives Neuregulin 1 signaling plays an important role in cardiac trabecular development, and in sustaining functional integrity in adult hearts. Treatment with neuregulin 1 enhances adult cardiomyocyte differentiation, survival and/or function in vitro and in vivo. It has also been suggested that recombinant neuregulin 1β1 (NRG1β1) induces cardiomyocyte proliferation in normal and injured adult hearts. Here we further explore the impact of neuregulin 1 signaling on adult cardiomyocyte cell cycle activity. Methods and Results Adult mice were subjected to 9 consecutive daily injections of recombinant NRG1β1 or vehicle, and cardiomyocyte DNA synthesis was quantitated via bromodeoxyuridine (BrdU) incorporation, which was delivered using mini-osmotic pumps over the entire duration of NRG1β1 treatment. NRG1β1 treatment inhibited baseline rates of cardiomyocyte DNA synthesis in normal mice (cardiomyocyte labelling index: 0.019±0.005% vs. 0.003±0.001%, saline vs. NRG1β1, P<0.05). Acute NRG1β1 treatment did result in activation of Erk1/2 and cardiac myosin regulatory light chain (down-stream mediators of neuregulin signalling), as well as activation of DNA synthesis in non-cardiomyocytes, validating the biological activity of the recombinant protein. In other studies, mice were subjected to permanent coronary artery occlusion, and cardiomyocyte DNA synthesis was monitored via tritiated thymidine incorporation which was delivered as a single injection 7 days post-infarction. Daily NRG1β1 treatment had no impact on cardiomyocyte DNA synthesis in the infarcted myocardium (cardiomyocyte labelling index: 0.039±0.011% vs. 0.027±0.021%, saline vs. NRG1β1, P>0.05). Summary These data indicate that NRG1β1 treatment does not increase cardiomyocyte DNA synthesis (and consequently does not increase the rate of cardiomyocyte renewal) in normal or infarcted adult mouse hearts. Thus, any improvement in cardiac structure and function observed following neuregulin treatment of injured hearts likely occurs independently of overt myocardial regeneration.
Collapse
|
87
|
Lin X, Wu N, Shi Y, Wang S, Tan K, Shen Y, Dai H, Zhong J. Association between transforming growth factor β1 and atrial fibrillation in essential hypertensive patients. Clin Exp Hypertens 2014; 37:82-7. [PMID: 25496287 DOI: 10.3109/10641963.2014.913600] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Xianru Lin
- Department of Cardiology, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Jinan, China and
| | - Na Wu
- Department of Cardiology, Qingdao Municipal Hospital, Medical College, Qingdao University, Jinan, China
| | - Yue Shi
- Department of Cardiology, Qingdao Municipal Hospital, Medical College, Qingdao University, Jinan, China
| | - Shoudong Wang
- Department of Cardiology, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Jinan, China and
| | - Kai Tan
- Department of Cardiology, Qingdao Municipal Hospital, Medical College, Qingdao University, Jinan, China
| | - Yi Shen
- Department of Cardiology, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Jinan, China and
| | - Hongyan Dai
- Department of Cardiology, Qingdao Municipal Hospital, Medical College, Qingdao University, Jinan, China
| | - Jingquan Zhong
- Department of Cardiology, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Jinan, China and
| |
Collapse
|
88
|
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia, contributing to increased morbidity and reduced survival through its associations with stroke and heart failure. AF contributes to a four- to fivefold increase in the risk of stroke in the general population and is responsible for 10-15 % of all ischemic strokes. Diagnosis and treatment of AF require considerable health care resources. Current therapies to restore sinus rhythm in AF are suboptimal and are limited either by their pro-arrhythmic effects or by their procedure-related complications. These limitations have necessitated identification of newer therapeutic targets to expand the treatment options. There has been a considerable amount of research interest in investigating the mechanisms of initiation and propagation of AF. Despite extensive research focused on the pathogenesis of AF, a thorough understanding of various pathways mediating initiation and propagation of AF still remains limited. Research efforts focused on the identification of these pathways and molecular mediators have generated a great degree of interest for developing more targeted therapies. This review discusses the potential therapeutic targets and the results from experimental and clinical research investigating these targets.
Collapse
|
89
|
Ramos P, Rubies C, Torres M, Batlle M, Farre R, Brugada J, Montserrat JM, Almendros I, Mont L. Atrial fibrosis in a chronic murine model of obstructive sleep apnea: mechanisms and prevention by mesenchymal stem cells. Respir Res 2014; 15:54. [PMID: 24775918 PMCID: PMC4012097 DOI: 10.1186/1465-9921-15-54] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 03/18/2014] [Indexed: 12/31/2022] Open
Abstract
Background OSA increases atrial fibrillation (AF) risk and is associated with poor AF treatment outcomes. However, a causal association is not firmly established and the mechanisms involved are poorly understood. The aims of this work were to determine whether chronic obstructive sleep apnea (OSA) induces an atrial pro-arrhythmogenic substrate and to explore whether mesenchymal stem cells (MSC) are able to prevent it in a rat model of OSA. Methods A custom-made setup was used to mimic recurrent OSA-like airway obstructions in rats. OSA-rats (n = 16) were subjected to 15-second obstructions, 60 apneas/hour, 6 hours/day during 21 consecutive days. Sham rats (n = 14) were placed in the setup but no obstructions were applied. In a second series of rats, MSC were administered to OSA-rats and saline to Sham-rats. Myocardial collagen deposit was evaluated in Picrosirius-red stained samples. mRNA expression of genes involved in collagen turnover, inflammation and oxidative stress were quantified by real time PCR. MMP-2 protein levels were quantified by Western Blot. Results A 43% greater interstitial collagen fraction was observed in the atria, but not in the ventricles, of OSA-rats compared to Sham-rats (Sham 8.32 ± 0.46% vs OSA 11.90 ± 0.59%, P < 0.01). Angiotensin-I Converting Enzyme (ACE) and Interleukin 6 (IL-6) expression were significantly increased in both atria, while Matrix Metalloproteinase-2 (MMP-2) expression was decreased. MSC administration blunted OSA-induced atrial fibrosis (Sham + Saline 8.39 ± 0.56% vs OSA + MSC 9.57 ± 0.31%, P = 0.11), as well as changes in MMP-2 and IL-6 expression. Interleukin 1-β (IL-1β) plasma concentration correlated to atrial but not ventricular fibrosis. Notably, a 2.5-fold increase in IL-1β plasma levels was observed in the OSA group, which was prevented in rats receiving MSC. Conclusions OSA induces selective atrial fibrosis in a chronic murine model, which can be mediated in part by the systemic and local inflammation and by decreased collagen-degradation. MSCs transplantation prevents atrial fibrosis, suggesting that these stem cells could counterbalance inflammation in OSA.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Lluís Mont
- Thorax Institute, Unitat de Fibril · lació Auricular, Hospital Clínic, Universitat de Barcelona and Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain.
| |
Collapse
|
90
|
Woods CE, Olgin J. Atrial fibrillation therapy now and in the future: drugs, biologicals, and ablation. Circ Res 2014; 114:1532-46. [PMID: 24763469 PMCID: PMC4169264 DOI: 10.1161/circresaha.114.302362] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 03/03/2014] [Indexed: 01/26/2023]
Abstract
Atrial fibrillation (AF) is a complex disease with multiple inter-relating causes culminating in rapid, seemingly disorganized atrial activation. Therapy targeting AF is rapidly changing and improving. The purpose of this review is to summarize current state-of-the-art diagnostic and therapeutic modalities for treatment of AF. The review focuses on reviewing treatment as it relates to the pathophysiological basis of disease and reviews preclinical and clinical evidence for potential new diagnostic and therapeutic modalities, including imaging, biomarkers, pharmacological therapy, and ablative strategies for AF. Current ablation and drug therapy approaches to treating AF are largely based on treating the arrhythmia once the substrate occurs and is more effective in paroxysmal AF rather than persistent or permanent AF. However, there is much research aimed at prevention strategies, targeting AF substrate, so-called upstream therapy. Improved diagnostics, using imaging, genetics, and biomarkers, are needed to better identify subtypes of AF based on underlying substrate/mechanism to allow more directed therapeutic approaches. In addition, novel antiarrhythmics with more atrial specific effects may reduce limiting proarrhythmic side effects. Advances in ablation therapy are aimed at improving technology to reduce procedure time and in mechanism-targeted approaches.
Collapse
Affiliation(s)
- Christopher E Woods
- From the Division of Cardiology, University of California at San Francisco (C.E.W., J.O.); and Division of Cardiology Research, AUST Development, LLC, Mountain View, CA (C.E.W.)
| | | |
Collapse
|
91
|
Atrial fibrillation: A progressive atrial myopathy or a distinct disease? Int J Cardiol 2014; 171:126-33. [DOI: 10.1016/j.ijcard.2013.12.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 10/09/2013] [Accepted: 12/10/2013] [Indexed: 02/06/2023]
|
92
|
Kong P, Christia P, Frangogiannis NG. The pathogenesis of cardiac fibrosis. Cell Mol Life Sci 2014; 71:549-74. [PMID: 23649149 PMCID: PMC3769482 DOI: 10.1007/s00018-013-1349-6] [Citation(s) in RCA: 1215] [Impact Index Per Article: 110.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 04/19/2013] [Accepted: 04/22/2013] [Indexed: 12/16/2022]
Abstract
Cardiac fibrosis is characterized by net accumulation of extracellular matrix proteins in the cardiac interstitium, and contributes to both systolic and diastolic dysfunction in many cardiac pathophysiologic conditions. This review discusses the cellular effectors and molecular pathways implicated in the pathogenesis of cardiac fibrosis. Although activated myofibroblasts are the main effector cells in the fibrotic heart, monocytes/macrophages, lymphocytes, mast cells, vascular cells and cardiomyocytes may also contribute to the fibrotic response by secreting key fibrogenic mediators. Inflammatory cytokines and chemokines, reactive oxygen species, mast cell-derived proteases, endothelin-1, the renin/angiotensin/aldosterone system, matricellular proteins, and growth factors (such as TGF-β and PDGF) are some of the best-studied mediators implicated in cardiac fibrosis. Both experimental and clinical evidence suggests that cardiac fibrotic alterations may be reversible. Understanding the mechanisms responsible for initiation, progression, and resolution of cardiac fibrosis is crucial to design anti-fibrotic treatment strategies for patients with heart disease.
Collapse
Affiliation(s)
- Ping Kong
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, 1300 Morris Park Avenue Forchheimer G46B, Bronx, NY 10461 USA
| | - Panagiota Christia
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, 1300 Morris Park Avenue Forchheimer G46B, Bronx, NY 10461 USA
| | - Nikolaos G. Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, 1300 Morris Park Avenue Forchheimer G46B, Bronx, NY 10461 USA
| |
Collapse
|
93
|
Corradi D. Atrial fibrillation from the pathologist's perspective. Cardiovasc Pathol 2013; 23:71-84. [PMID: 24462196 DOI: 10.1016/j.carpath.2013.12.001] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 12/03/2013] [Accepted: 12/07/2013] [Indexed: 12/18/2022] Open
Abstract
Atrial fibrillation (AF), the most common sustained cardiac arrhythmia encountered in clinical practice, is associated with increased morbidity and mortality. Electrophysiologically, it is characterized by a high rate of asynchronous atrial cell depolarization causing a loss of atrial contractile function and irregular ventricular rates. For a long time, AF was considered as a pure functional disorder without any structural background. Only in recent years, have new mapping and imaging techniques identified atrial locations, which are very often involved in the initiation and maintenance of this supraventricular arrhythmia (i.e. the distal portion of the pulmonary veins and the surrounding atrial myocardium). Morphological analysis of these myocardial sites has demonstrated significant structural remodeling as well as paved the way for further knowledge of AF natural history, pathogenesis, and treatment. This architectural myocardial disarrangement is induced by the arrhythmia itself and the very frequently associated cardiovascular disorders. At the same time, the structural remodeling is also capable of sustaining AF, thereby creating a sort of pathogenetic vicious circle. This review focuses on current understanding about the structural and genetic bases of AF with reference to their classification, pathogenesis, and clinical implications.
Collapse
Affiliation(s)
- Domenico Corradi
- Department of Biomedical, Biotechnological, and Translational Sciences (S.Bi.Bi.T.), Unit of Pathology, University of Parma, Parma, Italy.
| |
Collapse
|
94
|
Zheng W, Yan C, Wang X, Luo Z, Chen F, Yang Y, Liu D, Gai X, Hou J, Huang M. TheTGFB1 functional polymorphism rs1800469 and susceptibility to atrial fibrillation in two Chinese Han populations. PLoS One 2013; 8:e83033. [PMID: 24349426 PMCID: PMC3861462 DOI: 10.1371/journal.pone.0083033] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 10/29/2013] [Indexed: 01/18/2023] Open
Abstract
Transforming growth factor-β1 (TGF-β1) is related to the degree of atrial fibrosis and plays critical roles in the induction and perpetuation of atrial fibrillation (AF). To investigate the association of the common promoter polymorphism rs1800469 in the TGF-β1 gene (TGFB1) with the risk of AF in Chinese Han population, we carried out a case-control study of two hospital-based independent populations: Southeast Chinese population (581 patients with AF and 723 controls), and Northeast Chinese population (308 AF patients and 292 controls). Two hundred and seventy-eight cases of AF were lone AF and 334 cases of AF were diagnosed as paroxysmal AF. In both populations, AF patients had larger left atrial diameters than the controls did. The rs1800469 genotypes in the TGFB1 gene were determined by polymerase chain reaction-restriction fragment length polymorphism. The genotype and allele frequencies of rs1800469 were not different between AF patients and controls of the Southeast Chinese population, Northeast Chinese population, and total Study Population. After adjustment for age, sex, hypertension and LAD, there was no association between the rs1800469 polymorphism and the risk of AF under the dominant, recessive and additive genetic models. Similar results were obtained from subanalysis of the lone and paroxymal AF subgroups. Our results do not support the role of the TGFB1 rs1800469 functional gene variant in the development of AF in the Chinese Han population.
Collapse
Affiliation(s)
- Weixing Zheng
- Department of Cardiology, Fuzhou General Hospital, Fujian Medical University, Fuzhou, China
| | - Chenghui Yan
- Department of Cardiology, Shenyang General Hospital, Shenyang, China
| | - Xiaohu Wang
- Department of Cardiology, Fujian Provincial Hospital, Fuzhou, China
| | - Zhurong Luo
- Department of Cardiology, Fuzhou General Hospital, Fujian Medical University, Fuzhou, China
| | - Fengping Chen
- Department of Cardiology, Fuzhou General Hospital, Fujian Medical University, Fuzhou, China
| | - Yuhui Yang
- Department of Cardiology, Fuzhou General Hospital, Fujian Medical University, Fuzhou, China
| | - Donglin Liu
- Department of Cardiology, Fuzhou General Hospital, Fujian Medical University, Fuzhou, China
| | - Xiaobo Gai
- Department of Cardiology, Fuzhou General Hospital, Fujian Medical University, Fuzhou, China
| | - Jianping Hou
- Department of Cardiology, Fuzhou General Hospital, Fujian Medical University, Fuzhou, China
| | - Mingfang Huang
- Department of Cardiology, Fuzhou General Hospital, Fujian Medical University, Fuzhou, China
- * E-mail:
| |
Collapse
|
95
|
Zhang X, Zhang C, Shen S, Xia YJ, Yi L, Gao Q, Wang Y. Dehydroepiandrosterone induces ovarian and uterine hyperfibrosis in female rats. Hum Reprod 2013; 28:3074-3085. [PMID: 23980058 DOI: 10.1093/humrep/det341] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
STUDY QUESTION Do dehydroepiandrosterone (DHEA)-treated rats with polycystic ovary syndrome (PCOS) demonstrate a high level of fibrosis in ovarian and uterine tissues? SUMMARY ANSWER DHEA induces ovarian and uterine hyperfibrosis in rats, probably involving a transforming growth factor-β (TGF-β)-dependent mechanism. WHAT IS KNOWN ALREADY Chronic inflammation is the typical cause of fibrosis and is involved in the pathophysiological process of PCOS. Patients with PCOS are reported to have a higher serum level of TGF-β, a well-characterized key pro-fibrotic factor. Fibrillin-3, a protein capable of interacting with TGF-β, has been reported to be partially responsible for the fetal origin of PCOS. STUDY DESIGN, SIZE, DURATION Female Sprague-Dawley rats were treated with a vehicle control or DHEA for 35 days, with subsequent analyses of changes in morphology and gene expression in ovarian and uterine tissues. Rescue groups treated with metformin or simvastatin and their corresponding controls were also analyzed. A total of 80 rats were included. PARTICIPANTS/MATERIALS, SETTING, METHODS The PCOS model was induced by daily administration of DHEA s.c. to 3-week-old female rats, and the rescue groups were injected daily with either metformin or simvastatin in addition to DHEA. Serum steroid hormone levels were measured by enzyme-linked immunosorbent assay. Samples were stained with hematoxylin and eosin for histological morphology, and Sirius Red and immunohistochemistry for revealing collagens. The expression of fibrosis-related genes was analyzed both at mRNA (real-time RT-PCR) and protein (western blot) levels. MAIN RESULTS AND THE ROLE OF CHANCE DHEA-induced rats with PCOS exhibited significantly higher levels of fibrosis (collagen IV) in both ovarian and uterine tissues. In ovarian tissue, the expression of connective tissue growth factor (CTGF) increased following DHEA treatment at both mRNA and protein levels (P < 0.05, P < 0.001 versus controls, respectively). Similar results versus controls were obtained at a protein level for TGF-β (P < 0.01) and mRNA level for fibronectin (P < 0.05) and angiotensin-II (P < 0.05). Likewise, in uterine tissue, the protein levels of both CTGF and TGF-β were higher than controls following DHEA treatment (P < 0.05). Treatment with either metformin or simvastatin attenuated the fibrosis progression induced by DHEA exposure, as evidenced by a reduction of TGF-β, plus CTGF or not, in both ovarian and uterine tissues. LIMITATIONS, REASONS FOR CAUTION The particular mechanism involved in the DHEA-induced fibrosis was not fully revealed. WIDER IMPLICATIONS OF THE FINDINGS Ovarian and uterine hyperfibrosis may occur in patients with PCOS and result in anovulation or other PCOS-related phenotypes. Anti-fibrotic therapy, for example metformin treatment, may be beneficial for patients with PCOS. STUDY FUNDING/COMPETING INTEREST(S) This study was supported by the National Natural Science Foundation of China (81170541) and the Natural Basic Research Program of China (973 program 2010CB945103). The authors declare no conflicts of interest.
Collapse
Affiliation(s)
- Xinlin Zhang
- Center for Translational Medicine & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, China
| | | | | | | | | | | | | |
Collapse
|
96
|
Treatment with bone morphogenetic protein 2 limits infarct size after myocardial infarction in mice. Shock 2013; 39:353-60. [PMID: 23376954 DOI: 10.1097/shk.0b013e318289728a] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Various strategies have been devised to reduce the clinical consequences of myocardial infarction, including acute medical care, revascularization, stem cell transplantations, and more recently, prevention of cardiomyocyte cell death. Activation of embryonic signaling pathways is a particularly interesting option to complement these strategies and to improve the functional performance and survival rate of cardiomyocytes. Here, we have concentrated on bone morphogenetic protein 2 (BMP-2), which induces ectopic formation of beating cardiomyocytes during development in the mesoderm and protects neonatal cardiomyocytes from ischemia-reperfusion injury. In a mouse model of acute myocardial infarction, an i.v. injection of BMP-2 reduced infarct size in mice when given after left anterior descending artery ligation. Mice treated with BMP-2 are characterized by a reduced rate of apoptotic cardiomyocytes both in the border zone of the infarcts and in the remote myocardium. In vitro, BMP-2 increases the frequency of spontaneously beating neonatal cardiomyocytes and the contractile performance under electrical pacing at 2 Hz, preserves cellular adenosine triphosphate stores, and decreases the rate of apoptosis despite the increased workload. In addition, BMP-2 specifically induced phosphorylation of Smad1/5/8 proteins and protected adult cardiomyocytes from long-lasting hypoxia-induced cellular damage and oxidative stress without activation of the cardiodepressant transforming growth factor-β pathway. Our data suggest that BMP-2 treatment may have considerable therapeutic potential in individuals with acute and chronic myocardial ischemia by improving the contractility of cardiomyocytes and preventing cardiomyocyte cell death.
Collapse
|
97
|
Velagapudi P, Turagam MK, Leal MA, Kocheril AG. Atrial fibrosis: a risk stratifier for atrial fibrillation. Expert Rev Cardiovasc Ther 2013; 11:155-60. [PMID: 23405837 DOI: 10.1586/erc.12.174] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Atrial fibrillation (AF), especially persistent and long-standing persistent AF, may result in electro-anatomical changes in the left atrium, resulting in remodeling and deposition of fibrous tissue. There are emerging data that atrial substrate modification may increase the risk of thromboembolic complications, including stroke. Several studies have reported that atrial fibrosis is due to complex interactions among several cellular and neurohumoral mediators. Late gadolinium enhancement MRI has been reported to allow quantitative assessment of myocardial fibrosis in patients at risk of developing a stroke. Current stroke risk stratification criteria for AF do not utilize atrial fibrosis as an independent risk factor despite its association with AF and stroke. Further research is required in developing adequate risk stratification tools for predicting the stroke risk and catheter ablation outcomes in AF.
Collapse
Affiliation(s)
- Poonam Velagapudi
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | | | | | | |
Collapse
|
98
|
King JH, Huang CLH, Fraser JA. Determinants of myocardial conduction velocity: implications for arrhythmogenesis. Front Physiol 2013; 4:154. [PMID: 23825462 PMCID: PMC3695374 DOI: 10.3389/fphys.2013.00154] [Citation(s) in RCA: 147] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 06/10/2013] [Indexed: 12/19/2022] Open
Abstract
Slowed myocardial conduction velocity (θ) is associated with an increased risk of re-entrant excitation, predisposing to cardiac arrhythmia. θ is determined by the ion channel and physical properties of cardiac myocytes and by their interconnections. Thus, θ is closely related to the maximum rate of action potential (AP) depolarization [(dV/dt)max], as determined by the fast Na+ current (INa); the axial resistance (ra) to local circuit current flow between cells; their membrane capacitances (cm); and to the geometrical relationship between successive myocytes within cardiac tissue. These determinants are altered by a wide range of pathophysiological conditions. Firstly, INa is reduced by the impaired Na+ channel function that arises clinically during heart failure, ischemia, tachycardia, and following treatment with class I antiarrhythmic drugs. Such reductions also arise as a consequence of mutations in SCN5A such as those occurring in Lenègre disease, Brugada syndrome (BrS), sick sinus syndrome, and atrial fibrillation (AF). Secondly, ra, may be increased due to gap junction decoupling following ischemia, ventricular hypertrophy, and heart failure, or as a result of mutations in CJA5 found in idiopathic AF and atrial standstill. Finally, either ra or cm could potentially be altered by fibrotic change through the resultant decoupling of myocyte–myocyte connections and coupling of myocytes with fibroblasts. Such changes are observed in myocardial infarction and cardiomyopathy or following mutations in MHC403 and SCN5A resulting in hypertrophic cardiomyopathy (HCM) or Lenègre disease, respectively. This review defines and quantifies the determinants of θ and summarizes experimental evidence that links changes in these determinants with reduced myocardial θ and arrhythmogenesis. It thereby identifies the diverse pathophysiological conditions in which abnormal θ may contribute to arrhythmia.
Collapse
Affiliation(s)
- James H King
- Physiological Laboratory, Department of Physiology, Development and Neuroscience, University of Cambridge Cambridge, UK
| | | | | |
Collapse
|
99
|
Sánchez-Quintana D, López-Mínguez JR, Pizarro G, Murillo M, Cabrera JA. Triggers and anatomical substrates in the genesis and perpetuation of atrial fibrillation. Curr Cardiol Rev 2013; 8:310-26. [PMID: 22920484 PMCID: PMC3492815 DOI: 10.2174/157340312803760721] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2012] [Revised: 05/14/2012] [Accepted: 05/31/2012] [Indexed: 12/21/2022] Open
Abstract
The definition of atrial fibrillation (AF) as a functional electrical disorder does not reflect the significant underlying structural abnormalities. Atrial and Pulmonary Vein (PV) muscle sleeve microstructural remodeling is present, and establishes a vulnerable substrate for AF maintenance. In spite of an incomplete understanding of the anatomo-functional basis for AF, current evidence demonstrates that this arrhythmia usually requires a trigger for initiation and a vulnerable electrophysiological and/or anatomical substrate for maintenance. It is still unclear whether the trigger mechanisms include focal enhanced automaticity, triggered activity and/or micro re-entry from myocardial tissue. Initiation of AF can be favored by both parasympathetic and sympathetic stimulation, which also seem to play a role in maintaining AF. Finally, evolving clinical evidence demonstrates that inflammation is associated with new-onset and recurrent AF through a mechanism that possibly involves cellular degeneration, apoptosis, and subsequent atrial fibrosis.
Collapse
Affiliation(s)
- Damián Sánchez-Quintana
- Departamento de Anatomía y Biología Celular, Facultad de Medicina, Universidad de Extremadura, Badajoz, Spain.
| | | | | | | | | |
Collapse
|
100
|
Deng W, Zong J, Bian Z, Zhou H, Yuan Y, Zhang R, Guo H, Zhang Y, Shen D, Li H, Tang Q. Indole-3-carbinol protects against pressure overload induced cardiac remodeling via activating AMPK-α. Mol Nutr Food Res 2013; 57:1680-7. [PMID: 23625645 DOI: 10.1002/mnfr.201300012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2013] [Revised: 02/18/2013] [Accepted: 02/19/2013] [Indexed: 11/06/2022]
Affiliation(s)
- Wei Deng
- Department of Cardiology; Renmin Hospital of Wuhan University; Wuhan P. R. China
- Cardiovascular Research Institute of Wuhan University; Wuhan P. R. China
| | - Jing Zong
- Department of Cardiology; Renmin Hospital of Wuhan University; Wuhan P. R. China
- Cardiovascular Research Institute of Wuhan University; Wuhan P. R. China
| | - Zhouyan Bian
- Department of Cardiology; Renmin Hospital of Wuhan University; Wuhan P. R. China
- Cardiovascular Research Institute of Wuhan University; Wuhan P. R. China
| | - Heng Zhou
- Department of Cardiology; Renmin Hospital of Wuhan University; Wuhan P. R. China
- Cardiovascular Research Institute of Wuhan University; Wuhan P. R. China
| | - Yuan Yuan
- Department of Cardiology; Renmin Hospital of Wuhan University; Wuhan P. R. China
- Cardiovascular Research Institute of Wuhan University; Wuhan P. R. China
| | - Rui Zhang
- Department of Cardiology; Renmin Hospital of Wuhan University; Wuhan P. R. China
- Cardiovascular Research Institute of Wuhan University; Wuhan P. R. China
| | - Haipeng Guo
- The Key Laboratory of Cardiovascular Remodeling and Function Research; Chinese Ministry of Education and Chinese Ministry of Health; Qilu Hospital of Shandong University; Jinan P. R. China
| | - Yan Zhang
- Department of Cardiology; Renmin Hospital of Wuhan University; Wuhan P. R. China
- Cardiovascular Research Institute of Wuhan University; Wuhan P. R. China
| | - Difei Shen
- Department of Cardiology; Renmin Hospital of Wuhan University; Wuhan P. R. China
- Cardiovascular Research Institute of Wuhan University; Wuhan P. R. China
| | - Hongliang Li
- Department of Cardiology; Renmin Hospital of Wuhan University; Wuhan P. R. China
- Cardiovascular Research Institute of Wuhan University; Wuhan P. R. China
| | - Qizhu Tang
- Department of Cardiology; Renmin Hospital of Wuhan University; Wuhan P. R. China
- Cardiovascular Research Institute of Wuhan University; Wuhan P. R. China
| |
Collapse
|