1
|
Ning ZH, Wang XH, Zhao Y, Ou Y, Yang JY, Tang HF, Hu HJ. Ferroptosis in organ fibrosis: Mechanisms and therapeutic approaches. Int Immunopharmacol 2025; 151:114341. [PMID: 40024213 DOI: 10.1016/j.intimp.2025.114341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/09/2025] [Accepted: 02/17/2025] [Indexed: 03/04/2025]
Abstract
Ferroptosis, a form of iron-dependent regulated cell death, has emerged as a critical mechanism in the pathogenesis of organ fibrosis. This review aims to provide an overview of the molecular mechanisms underlying ferroptosis and its contribution to fibrosis in various organs, including the liver, lung, heart, and kidneys. We explore how dysregulated iron metabolism, lipid peroxidation, and oxidative stress contribute to ferroptosis and subsequent tissue damage, promoting the progression of fibrosis. In addition, we highlight the complex interplay between ferroptosis and other cellular processes such as apoptosis, necrosis, and inflammation in the fibrotic microenvironment. Furthermore, this review discusses current therapeutic strategies targeting ferroptosis, including iron chelation, antioxidants, and modulators of lipid peroxidation. We also examine ongoing clinical and preclinical studies aimed at translating these findings into viable treatments for fibrotic diseases. Understanding the role of ferroptosis in organ fibrosis offers novel therapeutic opportunities, with the potential to mitigate disease progression and improve patient outcomes.
Collapse
Affiliation(s)
- Zhi-Hong Ning
- The First Affiliated Hospital, Department of Cardiovascular Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Xiu-Heng Wang
- The First Affiliated Hospital, Department of Medical-Record, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Yue Zhao
- The First Affiliated Hospital, Department of Cardiovascular Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Yun Ou
- The First Affiliated Hospital, Department of Cardiovascular Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Jia-Yan Yang
- The First Affiliated Hospital, Department of Cardiovascular Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Hui-Fang Tang
- The First Affiliated Hospital, Department of Cardiovascular Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Heng-Jing Hu
- The First Affiliated Hospital, Department of Cardiovascular Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China; Department of Cardiovascular Disease and Key Lab for Atherosclerosis of Hunan Province, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China.
| |
Collapse
|
2
|
Schotten U, Goette A, Verheule S. Translation of pathophysiological mechanisms of atrial fibrosis into new diagnostic and therapeutic approaches. Nat Rev Cardiol 2025; 22:225-240. [PMID: 39443702 DOI: 10.1038/s41569-024-01088-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/12/2024] [Indexed: 10/25/2024]
Abstract
Atrial fibrosis is one of the main manifestations of atrial cardiomyopathy, an array of electrical, mechanical and structural alterations associated with atrial fibrillation (AF), stroke and heart failure. Atrial fibrosis can be both a cause and a consequence of AF and, once present, it accelerates the progression of AF. The pathophysiological mechanisms leading to atrial fibrosis are diverse and include stretch-induced activation of fibroblasts, systemic inflammatory processes, activation of coagulation factors and fibrofatty infiltrations. Importantly, atrial fibrosis can occur in different forms, such as reactive and replacement fibrosis. The diversity of atrial fibrosis mechanisms and patterns depends on sex, age and comorbidity profile, hampering the development of therapeutic strategies. In addition, the presence and severity of comorbidities often change over time, potentially causing temporal changes in the mechanisms underlying atrial fibrosis development. This Review summarizes the latest knowledge on the molecular and cellular mechanisms of atrial fibrosis, its association with comorbidities and the sex-related differences. We describe how the various patterns of atrial fibrosis translate into electrophysiological mechanisms that promote AF, and critically appraise the clinical applicability and limitations of diagnostic tools to quantify atrial fibrosis. Finally, we provide an overview of the newest therapeutic interventions under development and discuss relevant knowledge gaps related to the association between clinical manifestations and pathological mechanisms of atrial fibrosis and to the translation of this knowledge to a clinical setting.
Collapse
Affiliation(s)
- Ulrich Schotten
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands.
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht, The Netherlands.
| | - Andreas Goette
- Department of Cardiology and Intensive Care Medicine, St. Vincenz Hospital, Paderborn, Germany
- Otto-von-Guericke University, Medical Faculty, Magdeburg, Germany
| | - Sander Verheule
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
3
|
Hou A, Shi D, Huang H, Liu Y, Zhang Y. Inflammation pathways as therapeutic targets in angiotensin II induced atrial fibrillation. Front Pharmacol 2025; 16:1515864. [PMID: 40098617 PMCID: PMC11911380 DOI: 10.3389/fphar.2025.1515864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 01/30/2025] [Indexed: 03/19/2025] Open
Abstract
Atrial fibrillation (AF), a common cardiac arrhythmia, is associated with severe complications such as stroke and heart failure. Although the precise mechanisms underlying AF remain elusive, inflammation is acknowledged as a pivotal factor in its progression. Angiotensin II (AngII) is implicated in promoting atrial remodeling and inflammation. However, the exact pathways through which AngII exacerbates AF are still not fully defined. This study explores the key molecular mechanisms involved, including dysregulation of calcium ions, altered connexin expression, and activation of signaling pathways such as TGF-β, PI3K/AKT, MAPK, NF-κB/NLRP3, and Rac1/JAK/STAT3. These pathways are instrumental in contributing to atrial fibrosis, electrical remodeling, and increased susceptibility to AF. Ang II-induced inflammation disrupts ion channel function, resulting in structural and electrical remodeling of the atria and significantly elevating the risk of AF. Anti-inflammatory treatments such as RAAS inhibitors, colchicine, and statins have demonstrated potential in reducing the incidence of AF, although clinical outcomes are inconsistent. This manuscript underscores the link between AngII-induced inflammation and the development of AF, proposing the importance of targeting inflammation in the management of AF.
Collapse
Affiliation(s)
- Ailin Hou
- Cardiovascular Department, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School of Beijing University of Chinese Medicine, Xiyuan Hospital, Beijing, China
| | - Dazhuo Shi
- Cardiovascular Department, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hongbo Huang
- Cardiovascular Department, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuxuan Liu
- Cardiovascular Department, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ying Zhang
- Cardiovascular Department, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
4
|
Liu X, Sha J, Wang L, Wang Z, Fang Z, Han X, Tan S, Chen Y, Yuan H, De The H, Zhou J, Zhu J. Rnf111 has a pivotal role in regulating development of definitive hematopoietic stem and progenitor cells through the Smad2/3-Gcsfr/NO axis in zebrafish. Haematologica 2025; 110:385-396. [PMID: 39363867 PMCID: PMC11788642 DOI: 10.3324/haematol.2024.285438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 09/23/2024] [Indexed: 10/05/2024] Open
Abstract
The ubiquitination or SUMOylation of hematopoietic-related factors plays pivotal roles in hematopoiesis. RNF111, known as a ubiquitin ligase, is a newly discovered SUMO-targeted ubiquitin ligase involved in multiple signaling pathways mediated by transforming growth factor (TGF)-β family members. However, its role in hematopoiesis remains unclear. Herein, a heritable Rnf111 mutant zebrafish line was generated by CRISPR/Cas9-mediated genome editing. Impairment of hematopoietic stem and progenitor cells (HSPC) of definitive hematopoiesis was found in Rnf111-deficient mutants. Ablation of Rnf111 resulted in decreased phosphorylation of Smad2/3 in HSPC. Definitive endoderm 2 inducer (IDE2), which specifically activates TGF-β signaling and downstream Smad2 phosphorylation, could restore definitive hematopoiesis in Rnf111-deficient embryos. Further molecular mechanism studies revealed that Gcsfr/NO signaling was an important target pathway of Smad2/3 involved in Rnf111-mediated HSPC development. In conclusion, our study demonstrated that Rnf111 contributes to the development of HSPC by maintaining Smad2/3 phosphorylation and activation of the Gcsfr/NO signaling pathway.
Collapse
Affiliation(s)
- Xiaohui Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; CNRS IRP (International research Project), Cancer, Aging and Hematology, Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025.
| | - Jinghan Sha
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; CNRS IRP (International research Project), Cancer, Aging and Hematology, Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025
| | - Luxiang Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025
| | - Zixuan Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025
| | - Zhou Fang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; CNRS IRP (International research Project), Cancer, Aging and Hematology, Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025
| | - Xiao Han
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025
| | - Shuiyi Tan
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; CNRS IRP (International research Project), Cancer, Aging and Hematology, Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025
| | - Yi Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025
| | - Hao Yuan
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; CNRS IRP (International research Project), Cancer, Aging and Hematology, Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025
| | - Hugues De The
- CNRS IRP (International research Project), Cancer, Aging and Hematology, Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Université de Paris 7/INSERM/CNRS UMR 944/7212, Equipe Labellisée Ligue Nationale Contre le Cancer, Hôpital St. Louis, Paris 75010
| | - Jun Zhou
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; CNRS IRP (International research Project), Cancer, Aging and Hematology, Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025
| | - Jun Zhu
- CNRS IRP (International research Project), Cancer, Aging and Hematology, Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Université de Paris 7/INSERM/CNRS UMR 944/7212, Equipe Labellisée Ligue Nationale Contre le Cancer, Hôpital St. Louis, Paris 75010.
| |
Collapse
|
5
|
Hu HJ, Wang XH, Zhang ZZ, Ou Y, Ning ZH, Yang JY, Huang H, Tang HF, Jiang ZS. SIRT3 sulfhydration using hydrogen sulfide inhibited angiotensin II-induced atrial fibrosis and vulnerability to atrial fibrillation via suppression of the TGF-β1/smad2/3 signalling pathway. Eur J Pharmacol 2024; 982:176900. [PMID: 39168432 DOI: 10.1016/j.ejphar.2024.176900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 07/30/2024] [Accepted: 08/14/2024] [Indexed: 08/23/2024]
Abstract
Atrial fibrosis is associated with the occurrence of atrial fibrillation (AF) and regulated by the transforming growth factor-β1 (TGF-β1)/Smad2/3 signalling pathway. Unfortunately, the mechanisms of regulation of TGF-β1/Smad2/3-induced atrial fibrosis and vulnerability to AF remain still unknown. Previous studies have shown that sirtuin3 (SIRT3) sulfhydration has strong anti-fibrotic effects. We hypothesised that SIRT3 sulfhydration inhibits angiotensin II (Ang-II)-induced atrial fibrosis via blocking the TGF-β1/Smad2/3 signalling pathway. In this study, we found that SIRT3 expression was decreased in the left atrium of patients with AF compared to that in those with sinus rhythm (SR). In vitro, SIRT3 knockdown by small interfering RNA significantly expanded Ang-II-induced atrial fibrosis and TGF-β1/Smad2/3 signalling pathway activation, whereas supplementation with Sodium Hydrosulfide (NaHS, exogenous hydrogen sulfide donor and sulfhydration agonist) and SIRT3 overexpression using adenovirus ameliorated Ang-II-induced atrial fibrosis. Moreover, we observed suppression of the TGF-β1/Smad2/3 pathway when Ang-II was combined with NaHS treatment, and the effect of this co-treatment was consistent with that of Ang-II combined with LY3200882 (Smad pathway inhibitor) on reducing atrial fibroblast proliferation and cell migration in vitro. Supplementation with dithiothreitol (DTT, a sulfhydration inhibitor) and adenovirus SIRT3 shRNA blocked the ameliorating effect of NaHS and AngII co-treatment on atrial fibrosis in vitro. Finally, continued treatment with NaHS in rats ameliorated atrial fibrosis and remodelling, and further improved AF vulnerability induced by Ang-II, which was reversed by DTT and adenovirus SIRT3 shRNA, suggesting that SIRT3 sulfhydration might be a potential therapeutic target in atrial fibrosis and AF.
Collapse
Affiliation(s)
- Heng-Jing Hu
- Department of Cardiology Laboratory, First Affiliated Hospital of University of South China, Hengyang, Hunan, 421001, PR China; Postdoctoral Research Station of Basic Medicine, University of South China, Hengyang, Hunan, 421001, PR China
| | - Xiu-Heng Wang
- Department of Medical-record, First Affiliated Hospital of University of South China, Hengyang, Hunan, 421001, PR China
| | - Zhi-Zhu Zhang
- Department of Cardiology Laboratory, First Affiliated Hospital of University of South China, Hengyang, Hunan, 421001, PR China
| | - Yun Ou
- Department of Cardiology Laboratory, First Affiliated Hospital of University of South China, Hengyang, Hunan, 421001, PR China
| | - Zhi-Hong Ning
- Department of Cardiology Laboratory, First Affiliated Hospital of University of South China, Hengyang, Hunan, 421001, PR China
| | - Jia-Yan Yang
- Department of Cardiology Laboratory, First Affiliated Hospital of University of South China, Hengyang, Hunan, 421001, PR China
| | - Hong Huang
- Department of Cardiology Laboratory, First Affiliated Hospital of University of South China, Hengyang, Hunan, 421001, PR China
| | - Hui-Fang Tang
- Department of Cardiology Laboratory, First Affiliated Hospital of University of South China, Hengyang, Hunan, 421001, PR China
| | - Zhi-Sheng Jiang
- Department of Cardiology Laboratory, First Affiliated Hospital of University of South China, Hengyang, Hunan, 421001, PR China; Postdoctoral Research Station of Basic Medicine, University of South China, Hengyang, Hunan, 421001, PR China; Institute of Cardiovascular Disease and Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan, 421001, PR China.
| |
Collapse
|
6
|
Kim D, Yang PS, Jang E, Yu HT, Kim TH, Uhm JS, Pak HN, Lee MH, Lip GYH, Sung JH, Joung B. Renal Outcomes of Rhythm Control in Patients Recently Diagnosed With Atrial Fibrillation. JACC Clin Electrophysiol 2024; 10:2431-2444. [PMID: 39243258 DOI: 10.1016/j.jacep.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/31/2024] [Accepted: 07/16/2024] [Indexed: 09/09/2024]
Abstract
BACKGROUND Atrial fibrillation (AF) is associated with impaired renal function and chronic kidney disease (CKD). OBJECTIVES This study assessed the effects of rhythm control on renal function compared with rate control among patients recently diagnosed with AF. METHODS A total of 20,886 patients with AF and available baseline estimated glomerular filtration rate (eGFR) data undergoing rhythm control (antiarrhythmic drugs or ablation) or rate control therapy, initiated within 1 year of AF diagnosis in 2005 to 2015, were identified from the Korean National Health Insurance Service database. The composite outcome of ≥30% decline in eGFR, acute kidney injury, kidney failure, or death from renal or cardiovascular causes was compared with the use of propensity overlap weighting between rhythm or rate control strategies in patients with or without significant CKD (eGFR <60 mL/min/1.73 m2). RESULTS Of the included patients (median age 62 years, 32.7% female), 2,213 (10.6%) had eGFR <60 mL/min/1.73 m2. Among patients with significant CKD, early rhythm control, compared with rate control, was associated with a lower risk of the primary composite outcome (weighted incidence rate: 2.77 vs 3.92 per 100 person-years; weighted HR: 0.70; 95% CI: 0.52-0.95). In patients without significant CKD, there was no difference in the risk of the primary composite outcome between rhythm and rate control groups (weighted incidence rate: 3.41 vs 3.21 per 100 person-years; weighted HR: 1.06; 95% CI: 0.96-1.18). No differences in safety outcomes were found between rhythm and rate control strategies in patients without or with significant CKD. CONCLUSIONS Among patients with AF and CKD, early rhythm control was associated with lower risks of adverse renal outcomes than rate control was.
Collapse
Affiliation(s)
- Daehoon Kim
- Division of Cardiology, Department of Internal Medicine, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Pil-Sung Yang
- Department of Cardiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Eunsun Jang
- Division of Cardiology, Department of Internal Medicine, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hee Tae Yu
- Division of Cardiology, Department of Internal Medicine, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Tae-Hoon Kim
- Division of Cardiology, Department of Internal Medicine, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jae-Sun Uhm
- Division of Cardiology, Department of Internal Medicine, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hui-Nam Pak
- Division of Cardiology, Department of Internal Medicine, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Moon-Hyoung Lee
- Division of Cardiology, Department of Internal Medicine, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart and Chest Hospital, Liverpool, United Kingdom; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Jung-Hoon Sung
- Department of Cardiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea.
| | - Boyoung Joung
- Division of Cardiology, Department of Internal Medicine, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
7
|
Zhu L, Liu YP, Huang YT, Zhou ZJ, Liu JF, Yu LM, Wang HS. Cellular and molecular biology of posttranslational modifications in cardiovascular disease. Biomed Pharmacother 2024; 179:117374. [PMID: 39217836 DOI: 10.1016/j.biopha.2024.117374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/25/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
Cardiovascular disease (CVD) has now become the leading cause of death worldwide, and its high morbidity and mortality rates pose a great threat to society. Although numerous studies have reported the pathophysiology of CVD, the exact pathogenesis of all types of CVD is not fully understood. Therefore, much more research is still needed to explore the pathogenesis of CVD. With the development of proteomics, many studies have successfully identified the role of posttranslational modifications in the pathogenesis of CVD, including key processes such as apoptosis, cell metabolism, and oxidative stress. In this review, we summarize the progress in the understanding of posttranslational modifications in cardiovascular diseases, including novel protein posttranslational modifications such as succinylation and nitrosylation. Furthermore, we summarize the currently identified histone deacetylase (HDAC) inhibitors used to treat CVD, providing new perspectives on CVD treatment modalities. We critically analyze the roles of posttranslational modifications in the pathogenesis of CVD-related diseases and explore future research directions related to posttranslational modifications in cardiovascular diseases.
Collapse
Affiliation(s)
- Li Zhu
- Graduate School of Dalian Medical University, Dalian 116000, Liaoning, China; State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning, China
| | - Yong-Ping Liu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Yu-Ting Huang
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning, China
| | - Zi-Jun Zhou
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning, China
| | - Jian-Feng Liu
- First School of Clinical Medicine, Shenyang Medical College, Shenyang 110034, Liaoning, China
| | - Li-Ming Yu
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning, China.
| | - Hui-Shan Wang
- Graduate School of Dalian Medical University, Dalian 116000, Liaoning, China; State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning, China.
| |
Collapse
|
8
|
Parent S, Amant JS, Remortel SV, Kahn S, Vaka R, Courtman D, Stewart DJ, Davis DR. Atrial Fibrosis and Inflammation in Postoperative Atrial Fibrillation: Comparative Effects of Amiodarone, Colchicine, or Exosomes. JACC Clin Electrophysiol 2024; 10:1037-1049. [PMID: 38639701 DOI: 10.1016/j.jacep.2024.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/01/2024] [Accepted: 02/07/2024] [Indexed: 04/20/2024]
Abstract
BACKGROUND Extracellular vesicles (EVs) isolated from human heart-derived cells have shown promise in suppressing inflammation and fibroblast proliferation. However, their precise benefits in atrial fibrillation (AF) prevention and the role of their antifibrotic/anti-inflammatory properties remain unclear. OBJECTIVES The purpose of this study was to conduct a head-to-head comparison of antiarrhythmic strategies to prevent postoperative AF using a rat model of sterile pericarditis. Specifically, we aimed to assess the efficacy of amiodarone (a classic antiarrhythmic drug), colchicine (an anti-inflammatory agent), and EVs derived from human heart-derived cells, which possess anti-inflammatory and antifibrotic properties, on AF induction, inflammation, and fibrosis progression. METHODS Heart-derived cells were cultured from human atrial appendages under serum-free xenogen-free conditions. Middle-aged Sprague Dawley rats were randomized into different groups, including sham operation, sterile pericarditis with amiodarone treatment, sterile pericarditis with colchicine treatment (2 dose levels), and sterile pericarditis with intra-atrial injection of EVs or vehicle. Invasive electrophysiological testing was performed 3 days after surgery before sacrifice. RESULTS Sterile pericarditis increased the likelihood of inducing AF. Colchicine and EVs exhibited anti-inflammatory effects, but only EV treatment significantly reduced AF probability, whereas colchicine showed a positive trend without statistical significance. EVs and high-dose colchicine reduced atrial fibrosis by 46% ± 2% and 26% ± 2%, respectively. Amiodarone prevented AF induction but had no effect on inflammation or fibrosis. CONCLUSIONS In this study, both amiodarone and EVs prevented AF, whereas treatment with colchicine was ineffective. The additional anti-inflammatory and antifibrotic effects of EVs suggest their potential as a comprehensive therapeutic approach for AF prevention, surpassing the effects of amiodarone or colchicine.
Collapse
Affiliation(s)
- Sandrine Parent
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Jennifer St Amant
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Sophie Van Remortel
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Saad Kahn
- Ottawa Hospital Research Institute, Division of Regenerative Medicine, Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Ramana Vaka
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - David Courtman
- Ottawa Hospital Research Institute, Division of Regenerative Medicine, Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Duncan John Stewart
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada; Ottawa Hospital Research Institute, Division of Regenerative Medicine, Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Darryl Raymond Davis
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada.
| |
Collapse
|
9
|
Datta M, Via LE, Dartois V, Weiner DM, Zimmerman M, Kaya F, Walker AM, Fleegle JD, Raplee ID, McNinch C, Zarodniuk M, Kamoun WS, Yue C, Kumar AS, Subudhi S, Xu L, Barry CE, Jain RK. Normalizing granuloma vasculature and matrix improves drug delivery and reduces bacterial burden in tuberculosis-infected rabbits. Proc Natl Acad Sci U S A 2024; 121:e2321336121. [PMID: 38530888 PMCID: PMC10998582 DOI: 10.1073/pnas.2321336121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/28/2024] [Indexed: 03/28/2024] Open
Abstract
Host-directed therapies (HDTs) represent an emerging approach for bacterial clearance during tuberculosis (TB) infection. While most HDTs are designed and implemented for immuno-modulation, other host targets-such as nonimmune stromal components found in pulmonary granulomas-may prove equally viable. Building on our previous work characterizing and normalizing the aberrant granuloma-associated vasculature, here we demonstrate that FDA-approved therapies (bevacizumab and losartan, respectively) can be repurposed as HDTs to normalize blood vessels and extracellular matrix (ECM), improve drug delivery, and reduce bacterial loads in TB granulomas. Granulomas feature an overabundance of ECM and compressed blood vessels, both of which are effectively reduced by losartan treatment in the rabbit model of TB. Combining both HDTs promotes secretion of proinflammatory cytokines and improves anti-TB drug delivery. Finally, alone and in combination with second-line antitubercular agents (moxifloxacin or bedaquiline), these HDTs significantly reduce bacterial burden. RNA sequencing analysis of HDT-treated lung and granuloma tissues implicates up-regulated antimicrobial peptide and proinflammatory gene expression by ciliated epithelial airway cells as a putative mechanism of the observed antitubercular benefits in the absence of chemotherapy. These findings demonstrate that bevacizumab and losartan are well-tolerated stroma-targeting HDTs, normalize the granuloma microenvironment, and improve TB outcomes, providing the rationale to clinically test this combination in TB patients.
Collapse
Affiliation(s)
- Meenal Datta
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Laura E Via
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Disease, NIH, Bethesda, MD 20892
| | - Véronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110
- Hackensack Meridian School of Medicine, Hackensack Meridian Health, Nutley, NJ 07110
| | - Danielle M Weiner
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Disease, NIH, Bethesda, MD 20892
| | - Matthew Zimmerman
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110
| | - Firat Kaya
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110
| | - April M Walker
- Tuberculosis Imaging Program, Division of Intramural Research, National Institute of Allergy and Infectious Disease, NIH, Bethesda, MD 20892
| | - Joel D Fleegle
- Tuberculosis Imaging Program, Division of Intramural Research, National Institute of Allergy and Infectious Disease, NIH, Bethesda, MD 20892
| | - Isaac D Raplee
- Bioinformatics and Computational Bioscience Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892
| | - Colton McNinch
- Bioinformatics and Computational Bioscience Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892
| | - Maksym Zarodniuk
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556
| | - Walid S Kamoun
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Changli Yue
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556
| | - Ashwin S Kumar
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Sonu Subudhi
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Lei Xu
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Clifton E Barry
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Disease, NIH, Bethesda, MD 20892
| | - Rakesh K Jain
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| |
Collapse
|
10
|
Newman JD, O'Meara E, Böhm M, Savarese G, Kelly PR, Vardeny O, Allen LA, Lancellotti P, Gottlieb SS, Samad Z, Morris AA, Desai NR, Rosano GMC, Teerlink JR, Giraldo CS, Lindenfeld J. Implications of Atrial Fibrillation for Guideline-Directed Therapy in Patients With Heart Failure: JACC State-of-the-Art Review. J Am Coll Cardiol 2024; 83:932-950. [PMID: 38418008 DOI: 10.1016/j.jacc.2023.12.033] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/29/2023] [Accepted: 12/18/2023] [Indexed: 03/01/2024]
Abstract
Atrial fibrillation (AF) and heart failure (HF) are common cardiovascular conditions that frequently coexist. Among patients with HF, more than one-half also have AF. Both are associated with significant morbidity and mortality. Moreover, the prevalence of each is increasing globally, and this trend is expected to continue owing to an aging population and increased life expectancy. Diagnosis of AF in a patient with HF is associated with greater symptom burden, more frequent hospitalizations, and a worse prognosis. Guideline-directed medical therapy (GDMT) for HF can affect the incidence of AF. Once present, AF can influence the efficacy of some components of GDMT for HF. In this review, we discuss the effect of GDMT for HF across the spectrum of ejection fraction on prevention of AF as well as the benefit of GDMT in patients with vs without AF.
Collapse
Affiliation(s)
| | - Eileen O'Meara
- Montreal Heart Institute and Université de Montréal, Montreal, Quebec, Canada
| | - Michael Böhm
- University of the Saarland, Homberg/Saar, Germany
| | - Gianluigi Savarese
- Division of Cardiology, Department of Medicine, Karolinska Institute, Stockholm, Sweden; Heart and Vascular and Neuro Theme, Karolinska University Hospital, Stockholm, Sweden
| | | | - Orly Vardeny
- University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Larry A Allen
- Division of Cardiology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | | | - Stephen S Gottlieb
- Division of Cardiovascular Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA; Baltimore Veterans Administration Medical Center, Baltimore, Maryland, USA
| | | | | | - Nihar R Desai
- Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Giuseppe M C Rosano
- Center for Clinical and Basic Research, IRCCS San Raffaele Pisana, Rome, Italy
| | | | | | - JoAnn Lindenfeld
- Vanderbilt Heart and Vascular Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
| |
Collapse
|
11
|
Parent S, Vaka R, St Amant J, Kahn S, Van Remortel S, Bi C, Courtman D, Stewart DJ, Davis DR. Inactivation of the NLRP3 inflammasome mediates exosome-based prevention of atrial fibrillation. Theranostics 2024; 14:608-621. [PMID: 38169629 PMCID: PMC10758048 DOI: 10.7150/thno.89520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 11/07/2023] [Indexed: 01/05/2024] Open
Abstract
Rationale: Extracellular vesicles (EVs) from human explant-derived cells injected directly into the atria wall muscle at the time of open chest surgery reduce atrial fibrosis, atrial inflammation, and atrial fibrillation (AF) in a rat model of sterile pericarditis. Albeit a promising solution to prevent postoperative AF, the mechanism(s) underlying this effect are unknown and it is not clear if this benefit is dependent on EV dose. Methods: To determine the dose-efficacy relationship of EVs from human explant-derived cells in a rat model of sterile pericarditis. Increasing doses of EVs (106, 107, 108 or 109) or vehicle control were injected into the atria of middle-age male Sprague-Dawley rats at the time of talc application. A sham control group was included to demonstrate background inducibility. Three days after surgery, all rats underwent invasive electrophysiological testing prior to sacrifice. Results: Pericarditis increased the likelihood of inducing AF (p<0.05 vs. sham). All doses decreased the probability of inducing AF with maximal effects seen after treatment with the highest dose (109, p<0.05 vs. vehicle). Pericarditis increased atrial fibrosis while EV treatment limited the effect of pericarditis on atrial fibrosis with maximal effects seen after treatment with 108 or 109 EVs. Increasing EV dose was associated with progressive decreases in pro-inflammatory cytokine content, inflammatory cell infiltration, and oxidative stress. EVs decreased NLRP3 (NACHT, LRR, and PYD domains-containing protein-3) inflammasome activation though a direct effect on resident atrial fibroblasts and macrophages. This suppressive effect was exclusive to EVs produced by heart-derived cells as application of EVs from bone marrow or umbilical cords did not alter NLRP3 activity. Conclusions: Intramyocardial injection of incremental doses of EVs at the time of open chest surgery led to progressive reductions in atrial fibrosis and inflammatory markers. These effects combined to render atria resistant to the pro-arrhythmic effects of pericarditis which is mechanistically related to suppression of the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Sandrine Parent
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, Ottawa, CANADA K1Y4W7
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada, K1H8M5
| | - Ramana Vaka
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, Ottawa, CANADA K1Y4W7
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada, K1H8M5
| | - Jennifer St Amant
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, Ottawa, CANADA K1Y4W7
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada, K1H8M5
| | - Saad Kahn
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, Canada, K1H8L6
| | - Sophie Van Remortel
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, Ottawa, CANADA K1Y4W7
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada, K1H8M5
| | - Christina Bi
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, Ottawa, CANADA K1Y4W7
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada, K1H8M5
| | - David Courtman
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, Canada, K1H8L6
| | - Duncan John Stewart
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, Ottawa, CANADA K1Y4W7
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada, K1H8M5
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, Canada, K1H8L6
| | - Darryl Raymond Davis
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, Ottawa, CANADA K1Y4W7
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada, K1H8M5
| |
Collapse
|
12
|
Zhang X, Tian B, Cong X, Ning Z. Corilagin inhibits angiotensin II-induced atrial fibrosis and fibrillation in mice through the PI3K-Akt pathway. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2024; 27:717-724. [PMID: 38645493 PMCID: PMC11024412 DOI: 10.22038/ijbms.2024.73281.15928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 10/17/2023] [Indexed: 04/23/2024]
Abstract
Objectives Corilagin (Cor) is reported as beiing hepatoprotective, anti-inflammatory, antibacterial, and anti-oxidant, while the effect on atrial fibrosis remains unknown. Therefore, we investigated the protective effect of Cor in angiotensin II (Ang II)-induced atrial fibrosis and atrial fibrillation (AF). Materials and Methods C57BL/6 mice (male, 8-10 weeks, n = 40) were subcutaneously infused either with saline or Ang II (2.0 mg/kg/day) and Cor (30 mg/kg) intraperitoneally injected 2 hr before Ang II infusion for 4 weeks. Mice were grouped into the control group (n=8), Cor group (n=8), Ang II group (n=8), and Ang II + Cor group (n=8). Morphological, histological, and biochemical examinations were performed. In vivo, transesophageal burst pacing was used to generate AF. Results Cor treatment markedly reduced Ang II-induced AF development in mice. Ang II + Cor therapy potentially decreased the atrial fibrotic area. It significantly decreased the increase in smooth muscle alpha-actin (α-SMA), CTGF, Collagen I, and Collagen III expressions brought on by Ang II treatment. Moreover, Ang II + Cor treatment remarkably decreased the malondialdehyde (MDA) content, whereas superoxide dismutase (SOD) and catalase (CAT) activities were potentially increased (all, P<0.001). In addition, Ang II + Cor significantly reduced Ang II-induced interleukin 1 beta (IL-1β), interleukin 6 (IL-6), and tumor necrosis factor-alpha (TNF-α) concentrations in atrial tissues. Furthermore, Cor significantly inhibited Ang II-induced p-PI3K, p-Akt, and NF-κB p-p65 protein expression in atrial tissues. Conclusion Our data speculated that Cor could have a protective effect against Ang II-induced atrial fibrosis and AF via down-regulation of the PI3K-Akt pathway.
Collapse
Affiliation(s)
- Xiaogang Zhang
- Cardiovascular Department, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Bei Tian
- Cardiovascular Department, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Xinpeng Cong
- Cardiovascular Department, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Zhongping Ning
- Cardiovascular Department, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| |
Collapse
|
13
|
Sayed A, Awad AK, Abdelfattah OM, Elsayed M, Herzallah K, Marine JE, Passman R, Bunch JT. The impact of catheter ablation in patient's heart failure and atrial fibrillation: a meta-analysis of randomized clinical trials. J Interv Card Electrophysiol 2023; 66:1487-1497. [PMID: 36572800 DOI: 10.1007/s10840-022-01451-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 12/05/2022] [Indexed: 12/28/2022]
Abstract
BACKGROUND Recent trial data suggest a benefit to catheter ablation (CA) compared to medical therapy for atrial fibrillation (AF) in patients with heart failure (HF). Nevertheless, because of mixed trial evidence, contemporary guidelines give it a class 2 recommendation. Accordingly, we sought to assess the currently available evidence for CA in HF with AF. METHODS Electronic databases were searched to identify randomized clinical trials (RCTs) comparing CA to medical therapy in patients with AF and HF. Study data was pooled using fixed and random effects, and the number needed to treat (NNT) was calculated to gauge absolute risk differences. Heterogeneity was quantified using I2. Our primary outcome was all-cause mortality. RESULTS Nine trials (CA 1075 patients; medical therapy 1083 patients) were included. Ablation reduced the relative risk of all-cause mortality by 31.5% (95% CI 13.7 to 45.6%; NNT = 23), cardiovascular mortality by 39.3% (95% CI 10.9 to 58.7%; NNT = 31), cardiovascular hospitalization by 29.1% (95% CI 9.4 to 44.6%; NNT = 9), and heart failure hospitalization by 28.5% (95% CI 6.5 to 45.4%; NNT = 22). Improvements in quality of life were observed with CA using the Minnesota Living with Heart Failure Questionnaire (mean difference - 5.26; 95% CI - 2.73 to - 7.78) and the Atrial Fibrillation Effect on Quality of Life (mean difference 5.36; 95% CI 2.72 to 8.00). CONCLUSION Compared to medical therapy, CA for AF in patients with HF reduces all-cause mortality, cardiovascular mortality, cardiovascular hospitalizations, and heart failure hospitalizations, and may improve quality of life.
Collapse
Affiliation(s)
- Ahmed Sayed
- Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Ahmed K Awad
- Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Omar M Abdelfattah
- Internal Medicine Department, Morristown Medical Center, Atlantic Health System, Morristown, NJ, USA
- Department of Cardiovascular Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - Mahmoud Elsayed
- Section of Cardiac Electrophysiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Joseph E Marine
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rod Passman
- Section of Cardiac Electrophysiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jared T Bunch
- Division of Cardiovascular Medicine, University of Utah Health Sciences Center, 30 North 1900 East, Room 4A100, Salt Lake City, UT, 84132, USA.
| |
Collapse
|
14
|
Öztürk B, Göçer K, Aksu E, Doğan K. Impaired Renal Vein Flow in Atrial Fibrillation: A Potential Risk for Renal Dysfunction. Med Sci Monit 2023; 29:e941435. [PMID: 37635347 PMCID: PMC10472836 DOI: 10.12659/msm.941435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 07/08/2023] [Indexed: 08/29/2023] Open
Abstract
BACKGROUND Atrial fibrillation (AF) is one of the most common heart rhythm disorders. Identification and early treatment of AF risk factors can improve mortality and morbidity rates. This study aimed to compare the renal venous stasis index (RVSI) and intra-renal venous flow (IRVF) patterns evaluated by intra-renal Doppler ultrasonography in patients with AF and sinus rhythm (SR). MATERIAL AND METHODS A total of 68 patients, 34 with AF (lasting >12 months AF) and 34 with SR (no previous diagnosis of AF and no AF attack in 24-h Holter monitoring) were included in the study. The RVSI was calculated, and the IRVF patterns were determined using intra-renal Doppler ultrasonography. High RVSI was defined as >0.12 RVSI. In addition, echocardiography and a 6-min walk test were performed. A model including diabetes mellitus, hypertension, creatine, Pro-BNP, left ventricular ejection fraction, presence of AF, and systolic pulmonary artery pressure was created to evaluate the effects of variables on high RVSI. RESULTS The RVSI value was significantly higher in patients with AF than in those with SR (P=0.004). The SR group exhibited a higher prevalence of the continuous flow pattern, which is one of the IRVF patterns (P=0.015). In contrast, the biphasic flow pattern was observed more frequently in patients with AF (P=0.003). The presence of AF was found to predict the high RVSI (P=0.002, OR=14.134, 95% CI 2.083-71.277). CONCLUSIONS The presence of AF may affect the IRVF and cause an increase in RVSI.
Collapse
Affiliation(s)
- Bayram Öztürk
- Department of Cardiology, Medical Park Göztepe Hospital, İstanbul, Turkey
| | - Kemal Göçer
- Department of Cardiology, Kahramanmaras Sutcu Imam University Faculty of Medicine, Kahramanmaras, Turkey
| | - Ekrem Aksu
- Department of Cardiology, Kahramanmaras Sutcu Imam University Faculty of Medicine, Kahramanmaras, Turkey
| | - Kamil Doğan
- Department of Radiology, Kahramanmaras Sutcu Imam University Faculty of Medicine, Kahramanmaras, Turkey
| |
Collapse
|
15
|
Wu CK, Teng S, Bai F, Liao XB, Zhou XM, Liu QM, Xiao YC, Zhou SH. Changes of ubiquitylated proteins in atrial fibrillation associated with heart valve disease: proteomics in human left atrial appendage tissue. Front Cardiovasc Med 2023; 10:1198486. [PMID: 37701139 PMCID: PMC10493305 DOI: 10.3389/fcvm.2023.1198486] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 06/27/2023] [Indexed: 09/14/2023] Open
Abstract
Background Correlations between posttranslational modifications and atrial fibrillation (AF) have been demonstrated in recent studies. However, it is still unclear whether and how ubiquitylated proteins relate to AF in the left atrial appendage of patients with AF and valvular heart disease. Methods Through LC-MS/MS analyses, we performed a study on tissues from eighteen subjects (9 with sinus rhythm and 9 with AF) who underwent cardiac valvular surgery. Specifically, we explored the ubiquitination profiles of left atrial appendage samples. Results In summary, after the quantification ratios for the upregulated and downregulated ubiquitination cutoff values were set at >1.5 and <1:1.5, respectively, a total of 271 sites in 162 proteins exhibiting upregulated ubiquitination and 467 sites in 156 proteins exhibiting downregulated ubiquitination were identified. The ubiquitylated proteins in the AF samples were enriched in proteins associated with ribosomes, hypertrophic cardiomyopathy (HCM), glycolysis, and endocytosis. Conclusions Our findings can be used to clarify differences in the ubiquitination levels of ribosome-related and HCM-related proteins, especially titin (TTN) and myosin heavy chain 6 (MYH6), in patients with AF, and therefore, regulating ubiquitination may be a feasible strategy for AF.
Collapse
Affiliation(s)
- Chen-Kai Wu
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Shuai Teng
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Fan Bai
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiao-Bo Liao
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xin-Min Zhou
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qi-Ming Liu
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yi-Chao Xiao
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Sheng-Hua Zhou
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
16
|
Parent S, Vaka R, Risha Y, Ngo C, Kanda P, Nattel S, Khan S, Courtman D, Stewart DJ, Davis DR. Prevention of atrial fibrillation after open-chest surgery with extracellular vesicle therapy. JCI Insight 2023; 8:e163297. [PMID: 37384420 PMCID: PMC10481795 DOI: 10.1172/jci.insight.163297] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 06/28/2023] [Indexed: 07/01/2023] Open
Abstract
Almost half of patients recovering from open-chest surgery experience atrial fibrillation (AF) that results principally from inflammation in the pericardial space surrounding the heart. Given that postoperative AF is associated with increased mortality, effective measures to prevent AF after open-chest surgery are highly desirable. In this study, we tested the concept that extracellular vesicles (EVs) isolated from human atrial explant-derived cells can prevent postoperative AF. Middle-aged female and male rats were randomized to undergo sham operation or induction of sterile pericarditis followed by trans-epicardial injection of human EVs or vehicle into the atrial tissue. Pericarditis increased the probability of inducing AF while EV treatment abrogated this effect in a sex-independent manner. EV treatment reduced infiltration of inflammatory cells and production of pro-inflammatory cytokines. Atrial fibrosis and hypertrophy seen after pericarditis were markedly attenuated by EV pretreatment, an effect attributable to suppression of fibroblast proliferation by EVs. Our study demonstrates that injection of EVs at the time of open-chest surgery shows prominent antiinflammatory effects and prevents AF due to sterile pericarditis. Translation of this finding to patients might provide an effective new strategy to prevent postoperative AF by reducing atrial inflammation and fibrosis.
Collapse
Affiliation(s)
- Sandrine Parent
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, and
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Ramana Vaka
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, and
| | - Yousef Risha
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, and
| | - Clarissa Ngo
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, and
| | - Pushpinder Kanda
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, and
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Stanley Nattel
- Research Center and Department of Medicine, Montreal Heart Institute, University of Montreal, Montreal, Quebec, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Institute of Pharmacology, West German Heart and Vascular Center, Faculty of Medicine, University of Duisburg-Essen, Essen, Germany
| | - Saad Khan
- Ottawa Hospital Research Institute, Division of Regenerative Medicine, Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - David Courtman
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Hospital Research Institute, Division of Regenerative Medicine, Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Duncan J. Stewart
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, and
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Hospital Research Institute, Division of Regenerative Medicine, Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Darryl R. Davis
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, and
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
17
|
Liu X, Zhang W, Luo J, Shi W, Zhang X, Li Z, Qin X, Liu B, Wei Y. TRIM21 deficiency protects against atrial inflammation and remodeling post myocardial infarction by attenuating oxidative stress. Redox Biol 2023; 62:102679. [PMID: 36996623 PMCID: PMC10074251 DOI: 10.1016/j.redox.2023.102679] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/05/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Atrial remodeling is a major contributor to the onset of atrial fibrillation (AF) after myocardial infarction (MI). Tripartite motif-containing protein 21 (TRIM21), an E3 ubiquitin protein ligase, is associated with pathological cardiac remodeling and dysfunction. However, the role of TRIM21 in postmyocardial infarction atrial remodeling and subsequent AF remains unclear. This study investigated the role of TRIM21 in post myocardial infarction atrial remodeling using TRIM21 knockout mice and explored the underlying mechanisms by overexpressing TRIM21 in HL-1 atrial myocytes using a lentiviral vector. The expression of TRIM21 in the left atrium of the mouse MI model was significantly elevated. TRIM21 deficiency alleviated MI-induced atrial oxidative damage, Cx43 downregulation, atrial fibrosis and enlargement, and abnormalities in electrocardiogram parameters (prolongation of the P-wave and PR interval). TRIM21 overexpression in atrial myocyte HL-1 cells further enhanced oxidative damage and Cx43 downregulation, whereas these effects were reversed by the reactive oxygen species scavenger N-acetylcysteine. The findings suggest that TRIM21 likely induces Nox2 expression mechanistically by activating the NF-κB pathway, which in turn leads to myocardial oxidative damage, inflammation, and atrial remodeling.
Collapse
Affiliation(s)
- Xiangdong Liu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Wenming Zhang
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Jiachen Luo
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Wentao Shi
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Xingxu Zhang
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Zhiqiang Li
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Xiaoming Qin
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Baoxin Liu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China.
| | - Yidong Wei
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China.
| |
Collapse
|
18
|
Liu X, Wu M, He Y, Gui C, Wen W, Jiang Z, Zhong G. Construction and integrated analysis of the ceRNA network hsa_circ_0000672/miR-516a-5p/TRAF6 and its potential function in atrial fibrillation. Sci Rep 2023; 13:7701. [PMID: 37169841 PMCID: PMC10175563 DOI: 10.1038/s41598-023-34851-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 05/09/2023] [Indexed: 05/13/2023] Open
Abstract
Atrial fibrosis is a crucial contributor to initiation and perpetuation of atrial fibrillation (AF). This study aimed to identify a circRNA-miRNA-mRNA competitive endogenous RNA (ceRNA) regulatory network related to atrial fibrosis in AF, especially to validate hsa_circ_0000672/hsa_miR-516a-5p/TRAF6 ceRNA axis in AF preliminarily. The circRNA-miRNA-mRNA ceRNA network associated with AF fibrosis was constructed using bioinformatic tools and literature reviews. Left atrium (LA) low voltage was used to represent LA fibrosis by using LA voltage matrix mapping. Ten controls with sinus rhythm (SR), and 20 patients with persistent AF including 12 patients with LA low voltage and 8 patients with LA normal voltage were enrolled in this study. The ceRNA regulatory network associated with atrial fibrosis was successfully constructed, which included up-regulated hsa_circ_0000672 and hsa_circ_0003916, down-regulated miR-516a-5p and five up-regulated hub genes (KRAS, SMAD2, TRAF6, MAPK11 and SMURF1). In addition, according to the results of Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, these hub genes were clustered in TGF-beta and MAPK signaling pathway. In the patients with persistent AF, hsa_circ_0000672 expression in peripheral blood monocytes was significantly higher than those in controls with SR by quantitative real-time polymerase chain reaction (p-value < 0.001). Furthermore, hsa_circ_0000672 expression was higher in peripheral blood monocytes of persistent AF patients with LA low voltage than those with LA normal voltage (p-value = 0.002). The dual-luciferase activity assay confirmed that hsa_circ_0000672 exerted biological functions as a sponge of miR-516a-5p to regulate expression of its target gene TRAF6. Hsa_circ_0000672 expression in peripheral blood monocytes may be associated with atrial fibrosis. The hsa_circ_0000672 may be involved in atrial fibrosis by indirectly regulating TRAF6 as a ceRNA by sponging miR-516a-5p.
Collapse
Affiliation(s)
- Xing Liu
- Department of Cardiology, Xiangtan Central Hospital, Xiangtan, China
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Mingxing Wu
- Department of Cardiology, Xiangtan Central Hospital, Xiangtan, China
| | - Yan He
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Chun Gui
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Weiming Wen
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhiyuan Jiang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.
| | - Guoqiang Zhong
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.
| |
Collapse
|
19
|
Jankauskas SS, Mone P, Avvisato R, Varzideh F, De Gennaro S, Salemme L, Macina G, Kansakar U, Cioppa A, Frullone S, Gambardella J, Di Mauro M, Tesorio T, Santulli G. miR-181c targets Parkin and SMAD7 in human cardiac fibroblasts: Validation of differential microRNA expression in patients with diabetes and heart failure with preserved ejection fraction. Mech Ageing Dev 2023; 212:111818. [PMID: 37116731 DOI: 10.1016/j.mad.2023.111818] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 04/15/2023] [Accepted: 04/19/2023] [Indexed: 04/30/2023]
Abstract
BACKGROUND Cardiac fibrosis represents a key feature in the pathophysiology of heart failure with preserved ejection fraction (HFpEF), a condition highly prevalent amongst geriatric patients, especially if diabetic. The microRNA miR-181c has been shown to be associated with the response to exercise training in HFpEF patients and has been also linked to diabetic cardiovascular complications. However, the underlying mechanisms have not been fully elucidated. OBJECTIVE To measure circulating miR-181c in elderly patients with HFpEF and DM and identify gene targets pathophysiologically relevant in HFpEF. METHODS We quantified circulating miR-181c in frail older adults with a confirmed diagnosis of HFpEF and diabetes, and, as control, we enrolled age-matched subjects without HFpEF and without diabetes. We validated in human cardiac fibroblasts the molecular mechanisms linking miR-181c to a pro-fibrotic response. RESULTS 51 frail patients were included (34 patients with diabetes and HFpEF and 17 age-matched controls. We observed that miR-181c was significantly upregulated (p<0.0001) in HFpEF patients vs controls. We confirmed in vitro that miR-181c is targeting PRKN and SMAD7. CONCLUSIONS We demonstrate that miR-181c levels are significantly increased in frail elderly adults with diabetes and HFpEF and that miR-181c targets PRKN and SMAD7 in human cardiac fibroblasts.
Collapse
Affiliation(s)
- Stanislovas S Jankauskas
- Department of Medicine, Einstein Institute for Aging Research, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Pasquale Mone
- Department of Medicine, Einstein Institute for Aging Research, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA; ASL Avellino, Avellino, 83100, Italy
| | - Roberta Avvisato
- Department of Medicine, Einstein Institute for Aging Research, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Fahimeh Varzideh
- Department of Medicine, Einstein Institute for Aging Research, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA
| | | | - Luigi Salemme
- Casa di Cura "Montevergine", Mercogliano (Avellino), 83013, Italy
| | | | - Urna Kansakar
- Department of Medicine, Einstein Institute for Aging Research, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Angelo Cioppa
- Casa di Cura "Montevergine", Mercogliano (Avellino), 83013, Italy
| | | | - Jessica Gambardella
- Department of Molecular Pharmacology, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA
| | | | - Tullio Tesorio
- Casa di Cura "Montevergine", Mercogliano (Avellino), 83013, Italy
| | - Gaetano Santulli
- Department of Medicine, Einstein Institute for Aging Research, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA; Department of Molecular Pharmacology, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA.
| |
Collapse
|
20
|
Xie DM, Zhong Q, Xu X, Li Y, Chen S, Li M, Peng C. Alpha lipoic acid-loaded electrospun fibrous patch films protect heart in acute myocardial infarction mice by inhibiting oxidative stress. Int J Pharm 2023; 632:122581. [PMID: 36608806 DOI: 10.1016/j.ijpharm.2023.122581] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/11/2022] [Accepted: 01/01/2023] [Indexed: 01/05/2023]
Abstract
Oxidative stress, characterized by excessive accumulation of reactive oxygen species (ROS), is involved in acute myocardial infarction (AMI)-related pathological processes and vascular reperfusion therapy injury. Alpha lipoic acid (LA) exhibits excellent antioxidant properties, however, its application is limited by inherent characteristics, including rapid clearance and extensive volume distribution. In this study, we hypothesized that scavenging cardiac ROS using adequately delivered LA could promote heart repair. Here, we report a new strategy for dynamic-release LA to treat AMI disease. In particular, this involves using poly(lactic-co-glycolic) (PLGA) copolymers as carriers to form a thin film (LA@PLGA) via electrospinning technology to achieve controlled release of LA, which essentially blocking local ROS production in damaged hearts. The drug-loading capacity and capsulation efficiency of this compound film could be regulated by determining the dose proportions of LA and PLGA. The incubation of LA@PLGA showed strong anti-oxidative activity and anti-apoptosis effect in hydrogen peroxide-administered primary cardiomyocytes. Patching LA@PLGA on the infarcted cardiac surfaces of AMI mice dramatically improved heart functions and reduced cardiac fibrosis throughout ventricular remodeling process. Importantly, the attenuation of detrimental pathologies was observed, including oxidative stress, senescence, DNA damage, cytokine-related processes, apoptosis, and ferroptosis. These results suggest that PLGA-carried LA can reduce ROS damage and restore heart function after myocardial damage, demonstrating a great potential for LA drugs in treating AMI disease.
Collapse
Affiliation(s)
- Dong-Mei Xie
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Cardiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Qingguo Zhong
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Cardiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Xiaochun Xu
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Cardiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Yuanlong Li
- Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou 519041, China
| | - Simin Chen
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Cardiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Cardiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Chaoquan Peng
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Cardiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| |
Collapse
|
21
|
Novel preventive effect of isorhamnetin on electrical and structural remodeling in atrial fibrillation. Clin Sci (Lond) 2022; 136:1831-1849. [DOI: 10.1042/cs20220319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 11/22/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022]
Abstract
Abstract
Isorhamnetin, a natural flavonoid, has strong antioxidant and antifibrotic effects, and a regulatory effect against Ca2+-handling. Atrial remodeling due to fibrosis and abnormal intracellular Ca2+ activities contributes to initiation and persistence of atrial fibrillation (AF). The present study investigated the effect of isorhamnetin on angiotensin II (AngII)-induced AF in mice. Wild-type male mice (C57BL/6J, 8 weeks old) were assigned to three groups: (1) control group, (2) AngII-treated group, and (3) AngII- and isorhamnetin-treated group. AngII (1000 ng/kg/min) and isorhamnetin (5 mg/kg) were administered continuously via an implantable osmotic pump for two weeks and intraperitoneally one week before initiating AngII administration, respectively. AF induction and electrophysiological studies, Ca2+ imaging with isolated atrial myocytes and HL-1 cells, and action potential duration (APD) measurements using atrial tissue and HL-1 cells were performed. AF-related molecule expression was assessed and histopathological examination was performed. Isorhamnetin decreased AF inducibility compared with the AngII group and restored AngII-induced atrial effective refractory period prolongation. Isorhamnetin eliminated abnormal diastolic intracellular Ca2+ activities induced by AngII. Isorhamnetin also abrogated AngII-induced APD prolongation and abnormal Ca2+ loading in HL-1 cells. Furthermore, isorhamnetin strongly attenuated AngII-induced left atrial enlargement and atrial fibrosis. AngII-induced elevated expression of AF-associated molecules, such as ox-CaMKII, p-RyR2, p-JNK, p-ERK, and TRPC3/6, was improved by isorhamnetin treatment. The findings of the present study suggest that isorhamnetin prevents AngII-induced AF vulnerability and arrhythmogenic atrial remodeling, highlighting its therapeutic potential as an anti-arrhythmogenic pharmaceutical or dietary supplement.
Collapse
|
22
|
Xiao Z, Xie Y, Huang F, Yang J, Liu X, Lin X, Zhu P, Zheng S. MicroRNA-205-5p plays a suppressive role in the high-fat diet-induced atrial fibrosis through regulation of the EHMT2/IGFBP3 axis. GENES & NUTRITION 2022; 17:11. [PMID: 35858845 PMCID: PMC9297569 DOI: 10.1186/s12263-022-00712-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/25/2022] [Indexed: 11/28/2022]
Abstract
Objective MicroRNAs (miRNAs) targeting has been revealed to be an appealing strategy for the treatment and management of atrial fibrillation (AF). In this research, we aimed to explore the mechanisms of miR-205-5p in reducing the high-fat diet (HFD)-induced atrial fibrosis through the EHMT2/IGFBP3 axis. Methods Expression levels of miR-205-5p, IGFBP3 and EHMT2 were determined in AF patients, cell fibrosis models and mouse atrial fibrosis models. Luciferase activity and RIP assays were performed to detect the binding between miR-205-5p and EHMT2, and ChIP assays were implemented to detect the enrichment of H3K9me2 and H3K4me3 in the promoter region of IGFBP3 in cells. The related experiments focusing on the inflammatory response, atrial fibrosis, mitochondrial damage, and metabolic abnormalities were performed to figure out the roles of miR-205-5p, IGFBP3, and EHMT2 in cell and mouse atrial fibrosis models. Results Low expression levels of miR-205-5p and IGFBP3 and a high expression of EHMT2 were found in AF patients, cell fibrosis models and mouse atrial fibrosis models. Upregulation of miR-205-5p reduced the expression of TGF-β1, α-SMA, Col III and other fibrosis-related proteins. miR-205-5p overexpression targeted EHMT2 to regulate the methylation of H3 histones to promote IGFBP3 expression, which in turn affected the fibrosis of atrial muscle cells. In HFD-induced atrial fibrosis mice, upregulated miR-205-5p or elevated IGFBP3 alleviated atrial fibrosis, mitochondrial damage, and metabolic abnormalities. Conclusion This study suggests that miR-205-5p attenuates HFD-induced atrial fibrosis via modulating the EHMT2/IGFBP3 axis. Graphical Abstract miR-205-5p alleviates high-fat diet-induced atrial fibrosis in mice via EHMT2/IGFBP3. ![]()
Collapse
|
23
|
Mining of Potential Biomarkers and Pathway in Valvular Atrial Fibrillation (VAF) via Systematic Screening of Gene Coexpression Network. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:3645402. [PMID: 36226239 PMCID: PMC9550484 DOI: 10.1155/2022/3645402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/06/2022] [Accepted: 09/19/2022] [Indexed: 11/17/2022]
Abstract
Purpose. We apply the bioinformatics method to excavate the potential genes and therapeutic targets associated with valvular atrial fibrillation (VAF). Methods. The downloaded gene expression files from the gene expression omnibus (GEO) included patients with primary severe mitral regurgitation complicated with sinus or atrial fibrillation rhythm. Subsequently, the differential gene expression in left and right atrium was analyzed by R software. Additionally, weighted correlation network analysis (WGCNA), principal component analysis (PCA), and linear model for microarray data (LIMMA) algorithm were used to determine hub genes. Then, Metascape database, DAVID database, and STRING database were used to annotate and visualize the gene ontology (GO) analysis, KEGG pathway enrichment analysis, and PPI network analysis of differentially expressed genes (DEGs). Finally, the TFs and miRNAs were predicted by using online tools, such as PASTAA and miRDB. Results. 20,484 differentially expressed genes related to atrial fibrillation were obtained through the analysis of left and right atrial tissue samples of GSE115574 gene chip, and 1,009 were with statistical significance, including 45 upregulated genes and 964 downregulated genes. And the hub genes implicated in AF of NPC2, ODC1, SNAP29, LAPTM5, ST8SIA5, and FCGR3B were screened. Finally, the main regulators of targeted candidate biomarkers and microRNAs, EIF5A2, HIF1A, ZIC2, ELF1, and STAT2, were found in this study. Conclusion. These hub genes, NPC2, ODC1, SNAP29, LAPTM5, ST8SIA5, and FCGR3B, are important for the development of VAF, and their enrichment pathways and TFs elucidate the involved molecular mechanisms and assist in the validation of drug targets.
Collapse
|
24
|
Zhou Y, Suo W, Zhang X, Lv J, Liu Z, Liu R. Roles and mechanisms of quercetin on cardiac arrhythmia: A review. Biomed Pharmacother 2022; 153:113447. [DOI: 10.1016/j.biopha.2022.113447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 07/14/2022] [Accepted: 07/18/2022] [Indexed: 11/02/2022] Open
|
25
|
Zhou M, Chen JY, Chao ML, Zhang C, Shi ZG, Zhou XC, Xie LP, Sun SX, Huang ZR, Luo SS, Ji Y. S-nitrosylation of c-Jun N-terminal kinase mediates pressure overload-induced cardiac dysfunction and fibrosis. Acta Pharmacol Sin 2022; 43:602-612. [PMID: 34011968 PMCID: PMC8888706 DOI: 10.1038/s41401-021-00674-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 03/31/2021] [Indexed: 02/04/2023]
Abstract
Cardiac fibrosis (CF) is an irreversible pathological process that occurs in almost all kinds of cardiovascular diseases. Phosphorylation-dependent activation of c-Jun N-terminal kinase (JNK) induces cardiac fibrosis. However, whether S-nitrosylation of JNK mediates cardiac fibrosis remains an open question. A biotin-switch assay confirmed that S-nitrosylation of JNK (SNO-JNK) increased significantly in the heart tissues of hypertrophic patients, transverse aortic constriction (TAC) mice, spontaneously hypertensive rats (SHRs), and neonatal rat cardiac fibroblasts (NRCFs) stimulated with angiotensin II (Ang II). Site to site substitution of alanine for cysteine in JNK was applied to determine the S-nitrosylated site. S-Nitrosylation occurred at both Cys116 and Cys163 and substitution of alanine for cysteine 116 and cysteine 163 (C116/163A) inhibited Ang II-induced myofibroblast transformation. We further confirmed that the source of S-nitrosylation was inducible nitric oxide synthase (iNOS). 1400 W, an inhibitor of iNOS, abrogated the profibrotic effects of Ang II in NRCFs. Mechanistically, SNO-JNK facilitated the nuclear translocation of JNK, increased the phosphorylation of c-Jun, and induced the transcriptional activity of AP-1 as determined by chromatin immunoprecipitation and EMSA. Finally, WT and iNOS-/- mice were subjected to TAC and iNOS knockout reduced SNO-JNK and alleviated cardiac fibrosis. Our findings demonstrate an alternative mechanism by which iNOS-induced SNO-JNK increases JNK pathway activity and accelerates cardiac fibrosis. Targeting SNO-JNK might be a novel therapeutic strategy against cardiac fibrosis.
Collapse
Affiliation(s)
- Miao Zhou
- grid.89957.3a0000 0000 9255 8984Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Nanjing Medical University, Nanjing, 201203 China
| | - Ji-yu Chen
- grid.89957.3a0000 0000 9255 8984Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Nanjing Medical University, Nanjing, 201203 China
| | - Meng-Lin Chao
- grid.89957.3a0000 0000 9255 8984Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Nanjing Medical University, Nanjing, 201203 China
| | - Chao Zhang
- grid.89957.3a0000 0000 9255 8984Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Nanjing Medical University, Nanjing, 201203 China
| | - Zhi-guang Shi
- grid.89957.3a0000 0000 9255 8984Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, 201203 China
| | - Xue-chun Zhou
- grid.89957.3a0000 0000 9255 8984Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Nanjing Medical University, Nanjing, 201203 China
| | - Li-ping Xie
- grid.89957.3a0000 0000 9255 8984Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Nanjing Medical University, Nanjing, 201203 China ,grid.89957.3a0000 0000 9255 8984Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, 201203 China
| | - Shi-xiu Sun
- grid.89957.3a0000 0000 9255 8984Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Nanjing Medical University, Nanjing, 201203 China
| | - Zheng-rong Huang
- grid.412625.6Department of Cardiology, the First Affiliated Hospital of Xiamen University, Xiamen, 361003 China
| | - Shan-shan Luo
- grid.89957.3a0000 0000 9255 8984Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Nanjing Medical University, Nanjing, 201203 China
| | - Yong Ji
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Nanjing Medical University, Nanjing, 201203, China. .,Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, 201203, China. .,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 201203, China.
| |
Collapse
|
26
|
Abstract
Transforming growth factor-β (TGFβ) isoforms are upregulated and activated in myocardial diseases and have an important role in cardiac repair and remodelling, regulating the phenotype and function of cardiomyocytes, fibroblasts, immune cells and vascular cells. Cardiac injury triggers the generation of bioactive TGFβ from latent stores, through mechanisms involving proteases, integrins and specialized extracellular matrix (ECM) proteins. Activated TGFβ signals through the SMAD intracellular effectors or through non-SMAD cascades. In the infarcted heart, the anti-inflammatory and fibroblast-activating actions of TGFβ have an important role in repair; however, excessive or prolonged TGFβ signalling accentuates adverse remodelling, contributing to cardiac dysfunction. Cardiac pressure overload also activates TGFβ cascades, which initially can have a protective role, promoting an ECM-preserving phenotype in fibroblasts and preventing the generation of injurious, pro-inflammatory ECM fragments. However, prolonged and overactive TGFβ signalling in pressure-overloaded cardiomyocytes and fibroblasts can promote cardiac fibrosis and dysfunction. In the atria, TGFβ-mediated fibrosis can contribute to the pathogenic substrate for atrial fibrillation. Overactive or dysregulated TGFβ responses have also been implicated in cardiac ageing and in the pathogenesis of diabetic, genetic and inflammatory cardiomyopathies. This Review summarizes the current evidence on the role of TGFβ signalling in myocardial diseases, focusing on cellular targets and molecular mechanisms, and discussing challenges and opportunities for therapeutic translation.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
27
|
Gordon B, González-Fernández V, Dos-Subirà L. Myocardial fibrosis in congenital heart disease. Front Pediatr 2022; 10:965204. [PMID: 36467466 PMCID: PMC9715985 DOI: 10.3389/fped.2022.965204] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 10/18/2022] [Indexed: 11/21/2022] Open
Abstract
Myocardial fibrosis resulting from the excessive deposition of collagen fibers through the myocardium is a common histopathologic finding in a wide range of cardiovascular diseases, including congenital anomalies. Interstitial fibrosis has been identified as a major cause of myocardial dysfunction since it distorts the normal architecture of the myocardium and impairs the biological function and properties of the interstitium. This review summarizes current knowledge on the mechanisms and detrimental consequences of myocardial fibrosis in heart failure and arrhythmias, discusses the usefulness of available imaging techniques and circulating biomarkers to assess this entity and reviews the current body of evidence regarding myocardial fibrosis in the different subsets of congenital heart diseases with implications in research and treatment.
Collapse
Affiliation(s)
- Blanca Gordon
- Integrated Adult Congenital Heart Disease Unit, Vall d'Hebron University Hospital-Santa Creu i Sant Pau University Hospital, Barcelona, Spain
| | - Víctor González-Fernández
- Integrated Adult Congenital Heart Disease Unit, Vall d'Hebron University Hospital-Santa Creu i Sant Pau University Hospital, Barcelona, Spain
| | - Laura Dos-Subirà
- Integrated Adult Congenital Heart Disease Unit, Vall d'Hebron University Hospital-Santa Creu i Sant Pau University Hospital, Barcelona, Spain
| |
Collapse
|
28
|
Yang Y, Bartz TM, Brown MR, Guo X, Zilhao NR, Trompet S, Weiss S, Yao J, Brody JA, Defilippi CR, Hoogeveen RC, Lin HJ, Gudnason V, Ballantyne CM, Dorr M, Jukema JW, Petersmann A, Psaty BM, Rotter JI, Boerwinkle E, Fornage M, Jun G, Yu B. Identification of Functional Genetic Determinants of Cardiac Troponin T and I in a Multiethnic Population and Causal Associations With Atrial Fibrillation. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2021; 14:e003460. [PMID: 34732054 PMCID: PMC8692416 DOI: 10.1161/circgen.121.003460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 10/26/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND Elevated cardiac troponin levels in blood are associated with increased risk of cardiovascular diseases and mortality. Cardiac troponin levels are heritable, but their genetic architecture remains elusive. METHODS We conducted a transethnic genome-wide association analysis on high-sensitivity cTnT (cardiac troponin T; hs-cTnT) and high-sensitivity cTnI (cardiac troponin I; hs-cTnI) levels in 24 617 and 14 336 participants free of coronary heart disease and heart failure from 6 population-based cohorts, followed by a series of bioinformatic analyses to decipher the genetic architecture of hs-cTnT and hs-cTnI. RESULTS We identified 4 genome-wide significant loci for hs-cTnT including a novel locus rs3737882 in PPFIA4 and 3 previously reported loci at NCOA2, TRAM1, and BCL2. One known locus at VCL was replicated for hs-cTnI. One copy of C allele for rs3737882 was associated with a 6% increase in hs-cTnT levels (minor allele frequency, 0.18; P=2.80×10-9). We observed pleiotropic loci located at BAG3 and ANO5. The proportions of variances explained by single-nucleotide polymorphisms were 10.15% and 7.74% for hs-cTnT and hs-cTnI, respectively. Single-nucleotide polymorphisms were colocalized with BCL2 expression in heart tissues and hs-cTnT and with ANO5 expression in artery, heart tissues, and whole blood and both troponins. Mendelian randomization analyses showed that genetically increased hs-cTnT and hs-cTnI levels were associated with higher odds of atrial fibrillation (odds ratio, 1.38 [95% CI, 1.25-1.54] for hs-cTnT and 1.21 [95% CI, 1.06-1.37] for hs-cTnI). CONCLUSIONS We identified a novel genetic locus associated with hs-cTnT in a multiethnic population and found that genetically regulated troponin levels were associated with atrial fibrillation.
Collapse
Affiliation(s)
- Yunju Yang
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, Houston, Texas, USA
| | - Traci M. Bartz
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, USA
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Michael R. Brown
- Department of Epidemiology, Human Genetics & Environmental Sciences and Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | | | - Stella Trompet
- Department of Cardiology, Leiden University Medical Center and Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Stefan Weiss
- Interfaculty Institute for Genetics and Functional Genomics; Department of Functional Genomics; University Medicine and University of Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Jie Yao
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Jennifer A. Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, USA
| | | | - Ron C. Hoogeveen
- Division of Atherosclerosis and Vascular Medicine, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Henry J. Lin
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Christie M. Ballantyne
- Division of Atherosclerosis and Vascular Medicine, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
- Center for Cardiovascular Disease Prevention, Methodist DeBakey Heart Center, Houston, TX, USA
| | - Marcus Dorr
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Department of Internal Medicine B - Cardiology, Pneumology, Infectious Diseases, Intensive Care Medicine, University Medicine Greifswald, Greifswald, Germany
| | - J. Wouter Jukema
- Department of Cardiology and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden and the Netherlands Heart Institute, Utrecht, the Netherlands
| | - Astrid Petersmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald and Institute of Clinical Chemistry and Laboratory Medicine, Universitätsmedizin Oldenburg, Oldenburg, Germany
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, USA
- Department of Epidemiology, and Department of Health Services, University of Washington, Seattle, Washington, USA
| | - Jerome I. Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Eric Boerwinkle
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, Houston, Texas, USA
- Department of Epidemiology, Human Genetics & Environmental Sciences and Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Myriam Fornage
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, Houston, Texas, USA
- Department of Epidemiology, Human Genetics & Environmental Sciences and Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Goo Jun
- Department of Epidemiology, Human Genetics & Environmental Sciences and Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Bing Yu
- Department of Epidemiology, Human Genetics & Environmental Sciences and Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
29
|
Park S, Lee S, Kim Y, Lee Y, Kang MW, Kim K, Kim YC, Han SS, Lee H, Lee JP, Joo KW, Lim CS, Kim YS, Kim DK. Atrial fibrillation and kidney function: a bidirectional Mendelian randomization study. Eur Heart J 2021; 42:2816-2823. [PMID: 34023889 DOI: 10.1093/eurheartj/ehab291] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 11/11/2020] [Accepted: 04/27/2021] [Indexed: 01/06/2023] Open
Abstract
AIMS The aim of this study was to investigate the causal effects between atrial fibrillation (AF) and kidney function. METHODS AND RESULTS We performed a bidirectional summary-level Mendelian randomization (MR) analysis implementing the results from a large-scale genome-wide association study for estimated glomerular filtration rate (eGFR) by the CKDGen (N = 765 348) and AF (N = 588 190) to identify genetic instruments. The inverse variance weighted method was the main MR method used. For replication, an allele score-based MR was performed by individual-level data within a UK Biobank cohort of white British ancestry individuals (N = 337 138). A genetic predisposition to AF was significantly associated with decreased eGFR [for log-eGFR, beta -0.003 (standard error, 0.0005), P < 0.001] and increased risk of chronic kidney disease [beta 0.059 (0.0126), P < 0.001]. The significance remained in MR sensitivity analyses and the causal estimates were consistent when we limited the analysis to individuals of European ancestry. Genetically predicted eGFR did not show a significant association with the risk of AF [beta -0.366 (0.275), P = 0.183]. The results were similar in allele score-based MR, as allele score for AF was significantly associated with reduced eGFR [for continuous eGFR, beta -0.079 (0.021), P < 0.001], but allele score for eGFR did not show a significant association with risk of AF [beta -0.005 (0.008), P = 0.530]. CONCLUSIONS Our study supports that AF is a causal risk factor for kidney function impairment. However, an effect of kidney function on AF was not identified in this study.
Collapse
Affiliation(s)
- Sehoon Park
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Internal Medicine, Armed Forces Capital Hospital, Gyeonggi-do 13574, Korea
| | - Soojin Lee
- Department of Internal Medicine, Uijeongbu Eulji University Medical Center, Gyeonggi-do 11759, Korea
| | - Yaerim Kim
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu 42601, Korea
| | - Yeonhee Lee
- Department of Internal Medicine, Uijeongbu Eulji University Medical Center, Gyeonggi-do 11759, Korea
| | - Min Woo Kang
- Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Korea
| | - Kwangsoo Kim
- Transdisciplinary Department of Medicine & Advanced Technology, Seoul National University Hospital, Seoul 03080, Korea
| | - Yong Chul Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Korea
| | - Seung Seok Han
- Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Korea.,Kidney Research Institute, Seoul National University, Seoul, Korea
| | - Hajeong Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Korea.,Kidney Research Institute, Seoul National University, Seoul, Korea
| | - Jung Pyo Lee
- Kidney Research Institute, Seoul National University, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Internal Medicine, SMG-SNU Boramae Medical Center, Seoul 07061, Korea
| | - Kwon Wook Joo
- Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Korea.,Kidney Research Institute, Seoul National University, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Chun Soo Lim
- Kidney Research Institute, Seoul National University, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Internal Medicine, SMG-SNU Boramae Medical Center, Seoul 07061, Korea
| | - Yon Su Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Korea.,Kidney Research Institute, Seoul National University, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Dong Ki Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Korea.,Kidney Research Institute, Seoul National University, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
30
|
Guo G, Watterson S, Zhang SD, Bjourson A, McGilligan V, Peace A, Rai TS. The role of senescence in the pathogenesis of atrial fibrillation: A target process for health improvement and drug development. Ageing Res Rev 2021; 69:101363. [PMID: 34023420 DOI: 10.1016/j.arr.2021.101363] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 01/24/2021] [Accepted: 05/12/2021] [Indexed: 12/11/2022]
Abstract
Cellular senescence is a state of growth arrest that occurs after cells encounter various stresses. Senescence contributes to tumour suppression, embryonic development, and wound healing. It impacts on the pathology of various diseases by secreting inflammatory chemokines, immune modulators and other bioactive factors. These secretory biosignatures ultimately cause inflammation, tissue fibrosis, immunosenescence and many ageing-related diseases such as atrial fibrillation (AF). Because the molecular mechanisms underpinning AF development remain unclear, current treatments are suboptimal and have serious side effects. In this review, we summarize recent results describing the role of senescence in AF. We propose that senescence factors induce AF and have a causative role. Hence, targeting senescence and its secretory phenotype may attenuate AF.
Collapse
|
31
|
Cao W, Song S, Fang G, Li Y, Wang Y, Wang QS. Cadherin-11 Deficiency Attenuates Ang-II-Induced Atrial Fibrosis and Susceptibility to Atrial Fibrillation. J Inflamm Res 2021; 14:2897-2911. [PMID: 34239314 PMCID: PMC8259948 DOI: 10.2147/jir.s306073] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 05/14/2021] [Indexed: 11/23/2022] Open
Abstract
Background Atrial fibrosis serves as a disease initiating mechanism in the development of atrial fibrillation. Angiotensin II (Ang-II), a key mediator for atrial fibrosis, aberrantly activates atrial fibroblasts (AFs) into myofibroblasts, resulting in subsequent excessive synthesis and deposition of extracellular matrix (ECM). Cadherin-11 (CDH11) is essential in the development of non-cardiac fibrotic diseases. In this study, we investigated its role in the pathogenesis and underlying mechanism of atrial fibrillation. Methods We obtained left atrial tissues from either patients with atrial fibrillation or Ang-II-induced atrial fibrosis mice. We utilized a global CDH11 knockout mouse (CDH11-/-) model to determine the effect of CDH11 on AF cell proliferation, migration, ECM synthesis/deposition. RNA-Seq of isolated AFs from CDH11-/- or normal mice was performed and differential expressed genes were analyzed. The mouse susceptibility to atrial fibrillation was examined by cardiac electrophysiology. Results We found that cadherin-11 was significantly up-regulated in fibrotic atrial tissue from patients with atrial fibrillation and Ang-II-induced mice. Both normal and CDH11-/- mice did not develop atrial fibrosis at resting state. However, after Ang-II infusion, unlike severe atrial fibrosis occurred in normal mice, CDH11-/- mice displayed a reduced atrial fibrosis. Atrial fibroblasts with CDH11 deletion from CDH11-/- mice showed reduction in Ang-II-induced cell proliferation, migration and ECM synthesis/deposition, indicating the involvement of CDH11 in atrial fibrosis. Consistently, RNA-Seq of CDH11-null AFs uncovered significant decrease in pro-fibrotic gene expression. In addition, we identified reduction of transcripts associated with Smad2/3, ERK1/2 and JNK pathways. Further, CDH11-/- mice showed a significantly attenuated Ang-II-induced susceptibility to atrial fibrillation. Conclusion Our results indicate that CDH11 potentiates Ang-II-induced activation of AFs. The pathogenesis of atrial fibrosis is through CDH11 mediated stimulation of Smad2/3, ERK1/2 and JNK pathways. Thus, CDH11 might serve as a novel therapeutic target for ameliorating the development of atrial fibrillation.
Collapse
Affiliation(s)
- Wei Cao
- Department of Cardiology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200092, People's Republic of China
| | - Shuai Song
- Department of Cardiology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200092, People's Republic of China
| | - Guojian Fang
- Department of Cardiology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200092, People's Republic of China
| | - Yingze Li
- Department of Cardiology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200092, People's Republic of China
| | - Yuepeng Wang
- Department of Cardiology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200092, People's Republic of China
| | - Qun-Shan Wang
- Department of Cardiology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200092, People's Republic of China
| |
Collapse
|
32
|
Ilov NN, Palnikova OV, Stompel DR, Nechepurenko АA. Clinical Predictors of Occurrence of Ventricular Tachyarrhythmias in Patients with Reduced Left Ventricle Ejection Fraction. Results of Single-Center Prospective Study. ACTA ACUST UNITED AC 2021; 61:32-40. [PMID: 34112073 DOI: 10.18087/cardio.2021.5.n1480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/21/2021] [Accepted: 02/26/2021] [Indexed: 11/18/2022]
Abstract
Aim To evaluate the diagnostic significance of clinical and demographic parameters for predicting a 2-year probability of ventricular tachyarrhythmias (VT) in patients with chronic heart failure and reduced left ventricular ejection fraction (CHFrLVEF).Material and methods This single-center, prospective cohort study included 175 patients with CHFrLVEF who were implanted with a cardioverter defibrillator (CD). The endpoint was a CD-detected episode of VT. Patients were followed up for 2 years with visits at 3, 12, and 24 months after CD implantation.Results The primary endpoint was observed in 43 (24.4 %) patients at an average of 20.9 months (95 % confidence interval (CI), 20-21.9). The 2-year risk of fatal ventricular arrhythmias increased with detection of unstable VT (one-factor analysis, odds ratio (OR), 4.2; 95 % CI, 1.1-16.5; р=0.041; multifactor analysis, OR, 6.3; 95 % CI, 1.5-26.3; р=0.012) and with ischemic CHFrLVEF origin (one-factor analysis, OR, 2.2; 95 % CI, 1.1-4.5; p=0.021; multifactor analysis, OR, 2.5; 95 % CI, 1.2-5.1; р=0.018). In the presence of any type of atrial fibrillation (AF) in patients with non-ischemic CHFrLVEF, the probability of VT increased threefold (one-factor analysis, OR, 2.97; 95 % CI, 1.02-8.8; р=0.047; multifactor analysis, OR, 3.5; 95 % CI, 1.1-10.9; р=0.032).Conclusion The presence of ischemic heart disease and unstable VT paroxysms can be included in the number of important clinical predictors of VT in patients with CHFrLVEF. In patients with non-ischemic CHF, the presence of AF is associated with a high risk of VT.
Collapse
Affiliation(s)
- N N Ilov
- Astrakhan State Medical University, Astrakhan; Federal Center for Cardiovascular Surgery, Astrakhan
| | - O V Palnikova
- Federal Center for Cardiovascular Surgery, Astrakhan
| | - D R Stompel
- Federal Center for Cardiovascular Surgery, Astrakhan
| | | |
Collapse
|
33
|
Zeng N, Wen YH, Pan R, Yang J, Yan YM, Zhao AZ, Zhu JN, Fang XH, Shan ZX. Dickkopf 3: a Novel Target Gene of miR-25-3p in Promoting Fibrosis-Related Gene Expression in Myocardial Fibrosis. J Cardiovasc Transl Res 2021; 14:1051-1062. [PMID: 33723747 DOI: 10.1007/s12265-021-10116-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 02/25/2021] [Indexed: 10/21/2022]
Abstract
Increasing evidence has shown that microRNAs (miRNAs) participate in cardiac fibrosis. We aimed to elucidate the effect of miRNA miR-25-3p on cardiac fibrosis. MiRNA microarray was used to profile miRNAs in the myocardium of angiotensin-II (Ang-II)-infused mice. Effect of miR-25-3p on expression of fibrosis-related genes, including Col1a1, Col3a1, and Acta2, was investigated both in vitro and in vivo. MiR-25-3p was shown increased in the myocardium of Ang-II-infused mice and patients with heart failure. MiR-25-3p enhanced fibrosis-related gene expression in mouse cardiac fibroblasts (mCFs) and in the myocardium of Ang-II-infused mice. Dickkopf 3 (Dkk3) was identified as a target gene of miR-25-3p, and Dkk3 could ameliorate Smad3 activation and fibrosis-related gene expression via enhancing Smad7 expression in mCFs. Additionally, NF-κB signal was proven to mediate upregulation of miR-25-3p in cardiac fibrosis. Our findings suggest that miR-25-3p enhances cardiac fibrosis by suppressing Dkk3 to activate Smad3 and fibrosis-related gene expression.
Collapse
Affiliation(s)
- Ni Zeng
- School of Medicine, South China University of Technology, Guangzhou, 510632, China
| | - Yi-Hong Wen
- School of Medicine, South China University of Technology, Guangzhou, 510632, China
| | - Rong Pan
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, 510632, China
| | - Jing Yang
- Department of Cardiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yu-Min Yan
- School of Pharmacy, Southern Medical University, Guangzhou, 510515, China
| | - An-Zhi Zhao
- School of Pharmacy, Southern Medical University, Guangzhou, 510515, China
| | - Jie-Ning Zhu
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Cardiovascular Institute, Guangzhou, 510080, China.,Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Xian-Hong Fang
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Cardiovascular Institute, Guangzhou, 510080, China.
| | - Zhi-Xin Shan
- School of Medicine, South China University of Technology, Guangzhou, 510632, China. .,School of Pharmacy, Southern Medical University, Guangzhou, 510515, China. .,Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.
| |
Collapse
|
34
|
Ma J, Chen Q, Ma S. Left atrial fibrosis in atrial fibrillation: Mechanisms, clinical evaluation and management. J Cell Mol Med 2021; 25:2764-2775. [PMID: 33576189 PMCID: PMC7957273 DOI: 10.1111/jcmm.16350] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 01/05/2021] [Accepted: 01/27/2021] [Indexed: 12/19/2022] Open
Abstract
Atrial fibrillation (AF), the commonest arrhythmia, shows associations with various disease conditions. Mounting evidence indicates that atrial fibrosis is an important part of the arrhythmogenic substrate, with an essential function in the generation of conduction abnormalities that underlie the transition from paroxysmal to persistent AF, which in turn contributes to AF perpetuation. Left atrial (LA) fibrosis is considered a possible major factor and predictor in AF treatment. The present review provides insights into LA fibrosis’ association with AF. The information is focused on clinical aspects and mechanisms, clinical evaluating methods that evaluate fibrosis changes and examining possible options for the prevention of atrial fibrosis.
Collapse
Affiliation(s)
- Jin Ma
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Qiuxiong Chen
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Shiyu Ma
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| |
Collapse
|
35
|
Ye T, Zhang C, Wu G, Wan W, Guo Y, Fo Y, Chen X, Liu X, Ran Q, Liang J, Shi S, Yang B. Pinocembrin Decreases Ventricular Fibrillation Susceptibility in a Rat Model of Depression. Front Pharmacol 2020; 11:547966. [PMID: 33390936 PMCID: PMC7775674 DOI: 10.3389/fphar.2020.547966] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 10/06/2020] [Indexed: 12/15/2022] Open
Abstract
Background: Depression is associated with the increased risk of mortality and morbidity and is an independent risk factor for many cardiovascular diseases. Depression may promote cardiac arrhythmias, but little is known about the mechanisms. Pinocembrin mitigated depressive-like behaviors and exhibited cardioprotective effects in several models; however, whether pinocembrin benefits ventricular arrhythmias in depression models has not been elucidated. Thus, this study was to evaluate the effects of pinocembrin on ventricular fibrillation susceptibility in rat models of depression. Methods: Male Sprague-Dawley rats were randomly assigned into control, control + pinocembrin, MDD (major depressive disorder), and MDP (MDD + pinocembrin) groups, respectively. Depressive-like behaviors, ventricular electrophysiological parameters, electrocardiogram parameters, heart rate variability, ventricular histology, serum norepinephrine, tumor necrosis factor-α, and interleukin-1β were detected. Protein levels in left ventricle were measured by Western blot assays. Results: Compared with the MDD group, pinocembrin significantly mitigated depressive-like behaviors, prolonged ventricular effective refractory period, action potential duration, QT, and corrected QT (QTc) interval, improved heart rate variability, decreased Tpeak–Tend interval, ventricular fibrillation inducibility rate, ventricular fibrosis, ventricular positive nerve densities, and protein expression of tyrosine hydroxylase and growth associated protein-43, reduced serum norepinephrine, tumor necrosis factor-α, interleukin-1β concentrations, and the expression levels of p-IκBα and p-p65, and increased the protein expression of Cx43, Cav1.2, and Kv.4.2 in the MDP group. Conclusion: Pinocembrin attenuates ventricular electrical remodeling, autonomic remodeling, and ion-channel remodeling, lowers ventricular fibrosis, and suppresses depression-induced inflammatory responses, providing new insights in pinocembrin and ventricular arrhythmias in depressed patients.
Collapse
Affiliation(s)
- Tianxin Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Cui Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Gang Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Weiguo Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yan Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yuhong Fo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Xiuhuan Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Xin Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Qian Ran
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jinjun Liang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Shaobo Shi
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Bo Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
36
|
Hanna A, Humeres C, Frangogiannis NG. The role of Smad signaling cascades in cardiac fibrosis. Cell Signal 2020; 77:109826. [PMID: 33160018 DOI: 10.1016/j.cellsig.2020.109826] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/31/2020] [Accepted: 11/02/2020] [Indexed: 12/30/2022]
Abstract
Most myocardial pathologic conditions are associated with cardiac fibrosis, the expansion of the cardiac interstitium through deposition of extracellular matrix (ECM) proteins. Although replacement fibrosis plays a reparative role after myocardial infarction, excessive, unrestrained or dysregulated myocardial ECM deposition is associated with ventricular dysfunction, dysrhythmias and adverse prognosis in patients with heart failure. The members of the Transforming Growth Factor (TGF)-β superfamily are critical regulators of cardiac repair, remodeling and fibrosis. TGF-βs are released and activated in injured tissues, bind to their receptors and transduce signals in part through activation of cascades involving a family of intracellular effectors the receptor-activated Smads (R-Smads). This review manuscript summarizes our knowledge on the role of Smad signaling cascades in cardiac fibrosis. Smad3, the best-characterized member of the family plays a critical role in activation of a myofibroblast phenotype, stimulation of ECM synthesis, integrin expression and secretion of proteases and anti-proteases. In vivo, fibroblast Smad3 signaling is critically involved in scar organization and exerts matrix-preserving actions. Although Smad2 also regulates fibroblast function in vitro, its in vivo role in rodent models of cardiac fibrosis seems more limited. Very limited information is available on the potential involvement of the Smad1/5/8 cascade in cardiac fibrosis. Dissection of the cellular actions of Smads in cardiac fibrosis, and identification of patient subsets with overactive or dysregulated myocardial Smad-dependent fibrogenic responses are critical for design of successful therapeutic strategies in patients with fibrosis-associated heart failure.
Collapse
Affiliation(s)
- Anis Hanna
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, USA
| | - Claudio Humeres
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, USA
| | - Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
37
|
Wang H, Song T, Zhao Y, Zhao J, Wang X, Fu X. Long non-coding RNA LICPAR regulates atrial fibrosis via TGF-β/Smad pathway in atrial fibrillation. Tissue Cell 2020; 67:101440. [PMID: 32971457 DOI: 10.1016/j.tice.2020.101440] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 09/04/2020] [Accepted: 09/08/2020] [Indexed: 01/11/2023]
Abstract
Long non-coding RNA predicting cardiac remodeling (lnc LIPCAR) was implicated in several human diseases, while its role in atrial fibrillation (AF) remained poorly understood. Our study aimed to discover the role of LICPAR played in AF. Samples of atrial muscle tissues from patients diagnosed with sinus rhythm (SR) and atrial fibrillation (AF) were collected, and human atrial fibroblasts were isolated and identified under immunofluorescence staining. After Angiotensin II (Ang II, as a activator of TGF-β) stimulation with LICPAR overexpression or knockdown, the viability and proliferation of atrial fibroblasts were respectively determined using cell counting kit-8 (CCK-8) assay and clone formation assay. Relative expressions of LICPAR, fibrosis- and transforming growth factor-β (TGF-β)/Smad2/3-pathway related proteins were measured using quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot as needed. LICPAR and TGF-β1 were upregulated and were positively correlated in atrial muscle tissues from AF. Atrial fibroblasts were identified as Vimentin positive. Further analysis indicated that Ang II enhanced the levels of LIPCAR, Smad2/3 phosphorylation and α-smooth muscle actin (α-SMA). Also, upregulating LIPCAR further promoted the promotive effects of Ang II on levels of LIPCAR, Collagen I, Collagen II, α-SMA and Smad2/3 phosphorylation, cell viability and proliferation of atrial fibroblasts, whereas silencing LIPCAR resulted in opposite effects. LICPAR regulates atrial fibrosis via modulating TGF-β/Smad pathway, which provided a potential therapeutic method for AF in clinical practice in the future.
Collapse
Affiliation(s)
- Haiyan Wang
- Department of Cardiovascular Medicine, Second Hospital of HeBei Medical University, No. 215, Heping Road, Xinhua District, Shijiazhuang, 050000, China
| | - Tingting Song
- Department of Cardiovascular Medicine, Second Hospital of HeBei Medical University, No. 215, Heping Road, Xinhua District, Shijiazhuang, 050000, China
| | - Ying Zhao
- Department of Cardiovascular Medicine, Second Hospital of HeBei Medical University, No. 215, Heping Road, Xinhua District, Shijiazhuang, 050000, China
| | - Jiayu Zhao
- Department of Cardiovascular Medicine, Second Hospital of HeBei Medical University, No. 215, Heping Road, Xinhua District, Shijiazhuang, 050000, China
| | - Xun Wang
- Department of Cardiovascular Medicine, Second Hospital of HeBei Medical University, No. 215, Heping Road, Xinhua District, Shijiazhuang, 050000, China
| | - Xianghua Fu
- Department of Cardiovascular Medicine, Second Hospital of HeBei Medical University, No. 215, Heping Road, Xinhua District, Shijiazhuang, 050000, China.
| |
Collapse
|
38
|
Abstract
Catheter ablation (CA) of the pulmonary veins for atrial fibrillation (AF) is growing exponentially and is the most commonly performed electrophysiologic procedure. Initial descriptions focused on CA for paroxysmal AF, and now more recently expanded in application to persistent AF and those with comorbid heart failure. Efforts to improve success have and continue to address issues such as pulmonary vein "reconnection" following ablation through different ablative energy modalities, and the use of a "hybrid" surgical/endocardial combined approach in persistent forms of AF. Technologic advances as well are concurrently seeking to improve safety, particularly regarding the incidence of atrio-esophageal fistula in this seemingly ever-growing ablation population.
Collapse
Affiliation(s)
- Aaron B. Hesselson
- Electrophysiologic Services, Division of Cardiovascular Medicine, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
39
|
Kanda P, Benavente-Babace A, Parent S, Connor M, Soucy N, Steeves A, Lu A, Cober ND, Courtman D, Variola F, Alarcon EI, Liang W, Stewart DJ, Godin M, Davis DR. Deterministic paracrine repair of injured myocardium using microfluidic-based cocooning of heart explant-derived cells. Biomaterials 2020; 247:120010. [PMID: 32259654 DOI: 10.1016/j.biomaterials.2020.120010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 03/17/2020] [Accepted: 03/26/2020] [Indexed: 02/08/2023]
Abstract
While encapsulation of cells within protective nanoporous gel cocoons increases cell retention and pro-survival integrin signaling, the influence of cocoon size and intra-capsular cell-cell interactions on therapeutic repair are unknown. Here, we employ a microfluidic platform to dissect the impact of cocoon size and intracapsular cell number on the regenerative potential of transplanted heart explant-derived cells. Deterministic increases in cocoon size boosted the proportion of multicellular aggregates within cocoons, reduced vascular clearance of transplanted cells and enhanced stimulation of endogenous repair. The latter being attributable to cell-cell stimulation of cytokine and extracellular vesicle production while also broadening of the miRNA cargo within extracellular vesicles. Thus, by tuning cocoon size and cell occupancy, the paracrine signature and retention of transplanted cells can be enhanced to promote paracrine stimulation of endogenous tissue repair.
Collapse
Affiliation(s)
- Pushpinder Kanda
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, Ottawa, K1Y4W7, Canada
| | | | - Sandrine Parent
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, Ottawa, K1Y4W7, Canada
| | - Michie Connor
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, Ottawa, K1Y4W7, Canada
| | - Nicholas Soucy
- Ottawa-Carleton Institute for Biomedical Engineering, Ottawa, K1N6N5, Canada
| | - Alexander Steeves
- Department of Mechanical Engineering, University of Ottawa, K1N6N5, Canada
| | - Aizhu Lu
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, Ottawa, K1Y4W7, Canada
| | - Nicholas David Cober
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, K1H8M5, Canada
| | - David Courtman
- Ottawa Hospital Research Institute, Division of Regenerative Medicine, Department of Medicine, University of Ottawa, Ottawa, K1H8L6, Canada
| | - Fabio Variola
- Department of Mechanical Engineering, University of Ottawa, K1N6N5, Canada
| | - Emilio I Alarcon
- University of Ottawa Heart Institute, Division of Cardiac Surgery, Department of Medicine, University of Ottawa, Ottawa, K1Y4W7, Canada; Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, K1H8M5, Canada
| | - Wenbin Liang
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, Ottawa, K1Y4W7, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, K1H8M5, Canada
| | - Duncan J Stewart
- Ottawa Hospital Research Institute, Division of Regenerative Medicine, Department of Medicine, University of Ottawa, Ottawa, K1H8L6, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, K1H8M5, Canada
| | - Michel Godin
- Department of Physics, University of Ottawa, K1N6N5, Canada; Ottawa-Carleton Institute for Biomedical Engineering, Ottawa, K1N6N5, Canada; Department of Mechanical Engineering, University of Ottawa, K1N6N5, Canada
| | - Darryl R Davis
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, Ottawa, K1Y4W7, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, K1H8M5, Canada.
| |
Collapse
|
40
|
Chen G, Huang S, Song F, Zhou Y, He X. Lnc-Ang362 is a pro-fibrotic long non-coding RNA promoting cardiac fibrosis after myocardial infarction by suppressing Smad7. Arch Biochem Biophys 2020; 685:108354. [PMID: 32240638 DOI: 10.1016/j.abb.2020.108354] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 03/26/2020] [Accepted: 03/27/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Cardiac fibrosis following myocardial infarction (MI) leads to cardiac remodeling and dysfunction. Dysregulation of Smad7 which negatively regulates the profibrotic transforming growth factor-β1 (TGF-β1)/Smad signaling promotes cardiac fibrosis. However, the molecular mechanisms underlying TGF-β1/Smad7 dysregulation remain elusive. Long non-coding RNAs (lncRNAs) are recently emerging as important regulators of cardiac diseases. Here, we report lnc-Ang362 is a novel lncRNA mediating MI-induced fibrosis through TGF-β1/Smad7 signaling pathway. METHODS AND RESULTS The MI model was established by artificial coronary artery occlusion in rats. Microarray analysis identified 215 lncRNAs (fold change > 2.0, P < 0.05) differentially expressed between MI hearts and the sham group 4 weeks after MI. Lnc-Ang362 had the highest fold upregulation and the change was validated by reverse transcription polymerase chain reaction. Also, MI caused a marked increase in TGF-β1 and collagen I/III expression, but significantly downregulated Smad7 expression. Adult rat cardiac fibroblasts (RCFs) treated with TGF-β1 showed increased lnc-Ang362 expression and decreased Smad7 expression. Moreover, overexpression and knockdown of lnc-Ang362 by small interfering RNAs reduced and increased Smad7 expression, respectively. Importantly, this result was negatively correlated with the expression of collagen I/III in RCFs. Furthermore, the luciferase reporter assays confirmed that Smad7 was a validated lnc-Ang362 target. Further silencing Smad7 attenuated the effects of lnc-Ang362 knockdown on decreasing collagen I/III expression in RCFs. CONCLUSIONS These results suggested lnc-Ang362 promoted cardiac fibrosis after MI via directly suppressing Smad7, which may decrease the inhibitory feedback regulation of TGF-β1/Smad signaling pathway. Thus, lnc-Ang362 may be a novel profibrotic lncRNA in the regulation of cardiac fibrosis post MI.
Collapse
Affiliation(s)
- Guo Chen
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.
| | - Sihui Huang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Feier Song
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.
| | - Yingling Zhou
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.
| | - Xuyu He
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.
| |
Collapse
|
41
|
Packer M. Do most patients with obesity or type 2 diabetes, and atrial fibrillation, also have undiagnosed heart failure? A critical conceptual framework for understanding mechanisms and improving diagnosis and treatment. Eur J Heart Fail 2019; 22:214-227. [PMID: 31849132 DOI: 10.1002/ejhf.1646] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 09/14/2019] [Accepted: 09/20/2019] [Indexed: 02/06/2023] Open
Abstract
Obesity and diabetes can lead to heart failure with preserved ejection fraction (HFpEF), potentially because they both cause expansion and inflammation of epicardial adipose tissue and thus lead to microvascular dysfunction and fibrosis of the underlying left ventricle. The same process also causes an atrial myopathy, which is clinically evident as atrial fibrillation (AF); thus, AF may be the first manifestation of HFpEF. Many patients with apparently isolated AF have latent HFpEF or subsequently develop HFpEF. Most patients with obesity or diabetes who have AF and exercise intolerance have increased left atrial pressures at rest or during exercise, even in the absence of diagnosed HFpEF. Among patients with AF, those who also have latent HFpEF have increased risk for systemic thromboembolism and death. The identification of HFpEF in patients with obesity or diabetes alters the risk-to-benefit relationship of commonly prescribed treatments. Bariatric surgery and statins can ameliorate AF and reduce the risk for HFpEF. Conversely, antihyperglycaemic drugs that promote adipogenesis or cause sodium retention (insulin and thiazolidinediones) may increase the risk for heart failure in patients with an underlying ventricular myopathy. Patients with obesity and diabetes who undergo catheter ablation for AF are at increased risk for AF recurrence and for post-ablation increases in pulmonary venous pressures and worsening heart failure, especially if HFpEF coexists. Therefore, AF may be the earliest indicator of HFpEF in patients with obesity or type 2 diabetes, and recognition of HFpEF alters the management of these patients.
Collapse
Affiliation(s)
- Milton Packer
- Baylor Heart and Vascular Institute, Baylor University Medical Center, Dallas, TX, USA.,Imperial College London, London, UK
| |
Collapse
|
42
|
Wang J, Han M, Han SX, Zhi C, Gao S, Li Y. Effect of c-Ski on atrial remodelling in a rapid atrial pacing canine model. J Cell Mol Med 2019; 24:1795-1803. [PMID: 31815360 PMCID: PMC6991632 DOI: 10.1111/jcmm.14876] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 09/01/2019] [Indexed: 12/21/2022] Open
Abstract
Atrial fibrosis is an important factor in the initiation and maintenance of atrial fibrillation (AF); therefore, understanding the pathogenesis of atrial fibrosis may reveal promising therapeutic targets for AF. In this study, we successfully established a rapid atrial pacing canine model and found that the inducibility and duration of AF were significantly reduced by the overexpression of c‐Ski, suggesting that this approach may have therapeutic effects. c‐Ski was found to be down‐regulated in the atrial tissues of the rapid atrial pacing canine model. We artificially up‐regulated c‐Ski expression with a c‐Ski–overexpressing adenovirus. Haematoxylin and eosin, Masson's trichrome and picrosirius red staining showed that c‐Ski overexpression alleviated atrial fibrosis. Furthermore, we found that the expression levels of collagen III and α‐SMA were higher in the groups of dogs subjected to right‐atrial pacing, and this increase was attenuated by c‐Ski overexpression. In addition, c‐Ski overexpression decreased the phosphorylation of smad2, smad3 and p38 MAPK (p38α and p38β) as well as the expression of TGF‐β1 in atrial tissues, as shown by a comparison of the right‐atrial pacing + c‐Ski‐overexpression group to the control group with right‐atrial pacing only. These results suggest that c‐Ski overexpression improves atrial remodelling in a rapid atrial pacing canine model by suppressing TGF‐β1–Smad signalling and p38 MAPK activation.
Collapse
Affiliation(s)
- Juan Wang
- Department of Cardiology, The Fifth Affiliated Hospital to Xin Jiang Medical University, Urumchi, Xin Jiang, China
| | - Min Han
- Xin Jiang Medical University, Urumchi, Xin Jiang, China
| | - Su-Xia Han
- Department of Cardiovascular Medicine, Shanghai Pudong New Area People's Hospital Affiliated to Shanghai Health University, Shanghai, China
| | - Cuiju Zhi
- Department of Cardiovascular Medicine, Shanghai Pudong New Area People's Hospital Affiliated to Shanghai Health University, Shanghai, China
| | - Suli Gao
- Department of Cardiovascular Medicine, Shanghai Pudong New Area People's Hospital Affiliated to Shanghai Health University, Shanghai, China
| | - Yao Li
- Department of Cardiovascular Medicine, Shanghai Pudong New Area People's Hospital Affiliated to Shanghai Health University, Shanghai, China
| |
Collapse
|
43
|
Tao J, Wang J, Li C, Wang W, Yu H, Liu J, Kong X, Chen Y. MiR-216a accelerates proliferation and fibrogenesis via targeting PTEN and SMAD7 in human cardiac fibroblasts. Cardiovasc Diagn Ther 2019; 9:535-544. [PMID: 32038943 DOI: 10.21037/cdt.2019.11.06] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background Heart failure (HF) is a progressive disease with relatively poor prognosis and lacks effective therapy, and the discovery of dysregulated microRNAs (miRNAs) and their role in cardiac fibroblasts have provided a new avenue for elucidating the mechanism involved in HF. Methods Two datasets of GSE53080 and GSE57338 were used to screen the miRNAs profiling and analysis the differentially expressed genes (DEGs) in HF. QRT-PCR was used to detect miR-216a between HF and healthy controls (HC). Cell counting kit-8 (CCK-8) assay and clonogenic assay were used to analyze the effect of proliferation and fibrogenesis. Then dual-luciferase activity assay and western blotting were used to confirm the key mechanism. Results In this study, the results showed that miR-216a was significantly up-regulated in HF and over-expression of miR-216a promoted proliferation and enhanced the fibrogenesis in the human cardiac fibroblasts (HCF) cells. Phosphatase and tensin homolog (PTEN) and mothers against decapentaplegic homolog 7 (SMAD7) were both validated as the direct target genes of miR-216a, which were confirmed by the dual-luciferase reporter assay. MiR-216a decreased the expression of PTEN and SMAD7 leading to the activation of Akt/mTOR and TGF-βRI/Smad2 in the HCF cells, which might act as a promoter of cardiac fibrosis. Conclusions Our study might provide a promising approach for the treatment of HF in the future.
Collapse
Affiliation(s)
- Jinsong Tao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.,Department of Cardiology, The Affiliated Hospital of Southeast University Medical College, Jiangyin 214400, China
| | - Jingyi Wang
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Chunyu Li
- Intensive Care Unit, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Weiwei Wang
- Intensive Care Unit, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Hao Yu
- Emergency Center, Kizilsu Kirghiz Autonomous Prefecture People's Hospital, Artux 845350, China
| | - Jinhui Liu
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xiangqing Kong
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yan Chen
- Emergency Center, Kizilsu Kirghiz Autonomous Prefecture People's Hospital, Artux 845350, China.,Emergency Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
44
|
Li X, Zhu F, Meng W, Zhang F, Hong J, Zhang G, Wang F. CYP2J2/EET reduces vulnerability to atrial fibrillation in chronic pressure overload mice. J Cell Mol Med 2019; 24:862-874. [PMID: 31749335 PMCID: PMC6933320 DOI: 10.1111/jcmm.14796] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 08/26/2019] [Accepted: 09/26/2019] [Indexed: 12/11/2022] Open
Abstract
Growing evidence has well established the protective effects of CYP2J2/EET on the cardiovascular system. The aim of the present study was to determine whether CYP2J2/EET has a preventive effect on atrial fibrillation (AF) and to investigate the underlying mechanisms. Wild‐type mice were injected with or without AAV9‐CYP2J2 before abdominal aortic constriction (AAC) operation. After 8 weeks, compared with wild‐type mice, AAC mice display higher AF inducibility and longer AF durations, which were remarkably attenuated with AAV9‐CYP2J2. Also, AAV9‐CYP2J2 reduced atrial fibrosis area and the deposit of collagen‐I/III in AAC mice, accompanied by the blockade of TGF‐β/Smad‐2/3 signalling pathways, as well as the recovery in Smad‐7 expression. In vitro, isolated atrial fibroblasts were administrated with TGF‐β1, EET, EEZE, GW9662, SiRNA Smad‐7 and pre‐MiR‐21, and EET was demonstrated to restrain the differentiation of atrial fibroblasts largely dependent on Smad‐7, due to the inhibition of EET on MiR‐21. In addition, increased inflammatory cytokines, as well as activated NF‐κB pathways induced by AAC surgery, were also significantly blunted by AAV9‐CYP2J2 treatment. These effects of CYP2J2/EET were partially blocked by GW9662, the antagonist of PPAR‐γ. In conclusion, this study revealed that CYP2J2/EET ameliorates atrial fibrosis through modulating atrial fibroblasts activation by disinhibition of MiR‐21 on Smad‐7, and attenuates atrial inflammatory response by repressing NF‐κB pathways, reducing the vulnerability to AF, and CYP2J2/EET exerts its role at least partially through PPAR‐γ activation. Our findings might provide a novel upstream therapeutic strategy for AF.
Collapse
Affiliation(s)
- Xuguang Li
- Department of Cardiology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Feng Zhu
- Department of Cardiology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Weidong Meng
- Department of Cardiology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Feng Zhang
- Department of Cardiology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiang Hong
- Department of Cardiology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Guobing Zhang
- Department of Cardiology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fang Wang
- Department of Cardiology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
45
|
Song S, Zhang R, Mo B, Chen L, Liu L, Yu Y, Cao W, Fang G, Wan Y, Gu Y, Wang Y, Li Y, Yu Y, Wang Q. EZH2 as a novel therapeutic target for atrial fibrosis and atrial fibrillation. J Mol Cell Cardiol 2019; 135:119-133. [PMID: 31408621 DOI: 10.1016/j.yjmcc.2019.08.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 08/04/2019] [Accepted: 08/06/2019] [Indexed: 01/20/2023]
Abstract
Angiotensin II (Ang-II)-induced fibroblast differentiation plays an important role in the development of atrial fibrosis and atrial fibrillation (AF). Here, we show that the expression of the histone methyltransferase enhancer of zeste homolog 2 (EZH2) is increased in atrial muscle and atrial fibroblasts in patients with AF, accompanied by significant atrial fibrosis and atrial fibroblast differentiation. In addition, EZH2 is induced in murine models of atrial fibrosis. Furthermore, either pharmacological GSK126 inhibition or molecular silencing of EZH2 can inhibit the differentiation of atrial fibroblasts and the ability to produce ECM induced by Ang-II. Simultaneously, inhibition of EZH2 can block the Ang-II-induced migration of atrial fibroblasts. We found that EZH2 promotes fibroblast differentiation mainly through the Smad signaling pathway and can form a transcription complex with Smad2 to bind to the promoter region of the ACTA2 gene. Finally, our in vivo experiments demonstrated that the EZH2 inhibitor GSK126 significantly inhibited Ang-II-induced atrial enlargement and fibrosis and reduced AF vulnerability. Our results demonstrate that targeting EZH2 or EZH2-regulated genes might present therapeutic potential in AF.
Collapse
Affiliation(s)
- Shuai Song
- Department of Cardiology, Xinhua Hospital Affiliated To Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Rui Zhang
- Department of Cardiology, Xinhua Hospital Affiliated To Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Binfeng Mo
- Department of Cardiology, Xinhua Hospital Affiliated To Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Long Chen
- Department of Cardiovascular Surgery, Huadong Hospital Affiliated of Fudan University, 221 Yananxi Road, Shanghai 200040, China
| | - Liang Liu
- Department of Cardiology, Shanghai Jiaotong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Yi Yu
- Department of Ultrasound, Xinhua Hospital Affiliated To Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Wei Cao
- Department of Cardiology, Xinhua Hospital Affiliated To Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Guojian Fang
- Department of Cardiology, Xinhua Hospital Affiliated To Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Yi Wan
- Department of Cardiology, Xinhua Hospital Affiliated To Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Yue Gu
- Department of Cardiology, Xinhua Hospital Affiliated To Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Yuepeng Wang
- Department of Cardiology, Xinhua Hospital Affiliated To Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Yigang Li
- Department of Cardiology, Xinhua Hospital Affiliated To Shanghai Jiaotong University School of Medicine, Shanghai 200092, China.
| | - Ying Yu
- Department of Cardiology, Xinhua Hospital Affiliated To Shanghai Jiaotong University School of Medicine, Shanghai 200092, China.
| | - Qunshan Wang
- Department of Cardiology, Xinhua Hospital Affiliated To Shanghai Jiaotong University School of Medicine, Shanghai 200092, China.
| |
Collapse
|
46
|
Effects of febuxostat on atrial remodeling in a rabbit model of atrial fibrillation induced by rapid atrial pacing. JOURNAL OF GERIATRIC CARDIOLOGY : JGC 2019; 16:540-551. [PMID: 31447893 PMCID: PMC6689522 DOI: 10.11909/j.issn.1671-5411.2019.07.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Febuxostat, a novel nonpurine selective inhibitor of xanthine oxidase (XO), may be used in the prevention and management of atrial fibrillation (AF). The purpose of this study was to evaluate the effects of febuxostat on atrial remodeling in a rabbit model of AF induced by rapid atrial pacing (RAP) and the mechanisms by which it acts. Methods Twenty-four rabbits were randomly divided into four groups: sham-operated group (Group S), RAP group (Group P), RAP with 5 mg/kg per day febuxostat group (Group LFP), and RAP with 10 mg/kg per day febuxostat group (Group HFP). All rabbits except those in Group S were subjected to RAP at 600 beats/min for four weeks. The effects of febuxostat on atrial electrical and structural remodeling, markers of inflammation and oxidative stress, and signaling pathways involved in the left atrium were examined. Results Shortened atrial effective refractory period (AERP), increased AF inducibility, decreased mRNA levels of Cav1.2 and Kv4.3, and left atrial enlargement and dysfunction were observed in Group P, and these changes were suppressed in the groups treated with febuxostat. Prominent atrial fibrosis was observed in Group P, as were increased levels of TGF-β1, Collagen I, and α-SMA and decreased levels of Smad7 and eNOS. Treatment with febuxostat attenuated these differences. Changes in inflammatory and oxidative stress markers induced by RAP were consistent with the protective effects of febuxostat. Conclusions This study is the first to find that febuxostat can inhibit atrial electrical and structural remodeling of AF by suppressing XO and inhibiting the TGF-β1/Smad signaling pathway.
Collapse
|
47
|
Li Y, Tan W, Ye F, Xue F, Gao S, Huang W, Wang Z. Identification of microRNAs and genes as biomarkers of atrial fibrillation using a bioinformatics approach. J Int Med Res 2019; 47:3580-3589. [PMID: 31218935 PMCID: PMC6726789 DOI: 10.1177/0300060519852235] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Objective We aimed to explore potential microRNAs (miRNAs) and target genes related to atrial fibrillation (AF). Methods Data for microarrays GSE70887 and GSE68475, both of which include AF and control groups, were downloaded from the Gene Expression Omnibus database. Differentially expressed miRNAs between AF and control groups were identified within each microarray, and the intersection of these two sets was obtained. These miRNAs were mapped to target genes in the miRNet database. Functional annotation and enrichment analysis of these target genes was performed in the DAVID database. The protein-protein interaction (PPI) network from the STRING database and the miRNA-target-gene network were merged into a PPI-miRNA network using Cytoscape software. Modules of this network containing miRNAs were detected and further analyzed. Results Ten differentially expressed miRNAs and 1520 target genes were identified. Three PPI-miRNA modules were constructed, which contained miR-424, miR-15a, miR-542-3p, and miR-421 as well as their target genes, CDK1, CDK6, and CCND3. Conclusion The identified miRNAs and genes may be related to the pathogenesis of AF. Thus, they may be potential biomarkers for diagnosis and targets for treatment of AF.
Collapse
Affiliation(s)
- Yingyuan Li
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University, Guangdong, China
| | - Wulin Tan
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University, Guangdong, China
| | - Fang Ye
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University, Guangdong, China
| | - Faling Xue
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University, Guangdong, China
| | - Shaowei Gao
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University, Guangdong, China
| | - Wenqi Huang
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University, Guangdong, China
| | - Zhongxing Wang
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University, Guangdong, China
| |
Collapse
|
48
|
Manfrini O, Cenko E, Ricci B, Bugiardini R. Post Cardiovascular Surgery Atrial Fibrillation. Biomarkers Determining Prognosis. Curr Med Chem 2019; 26:916-924. [DOI: 10.2174/0929867324666170727104930] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 12/14/2016] [Accepted: 12/22/2016] [Indexed: 12/30/2022]
Abstract
<p>Background: New onset of atrial fibrillation (AF) after cardiovascular surgery is associated with increased risk of complications and length of hospital stay. Identification of patients at high risk of post-operative AF (POAF) may help to act with preventive strategies having clinical and economic relevance. </P><P> Objective: The focus of this review is to summarize findings on biomarkers of myocardial fibrosis (PICP and PIIINP), profibrotic mediators (TGF-beta1), extracellular matrix remodeling (MMP-9), myocardial stretch (BNP and NTpro-BNP), inflammation (interleukins, C-reactive protein and sCD40L), and myocardial necrosis (high-sensitivity troponin T), biomarkers, that can be used in clinical practice to stratify patients at risk for POAF. </P><P> Method: We searched English-language studies on MEDLINE and PubMed. Evidence synthesis was based on cohort studies, clinical trials and meta-analysis data. International clinical practice guidelines were reviewed, as well. </P><P> Results: Factors such as cardiac remodelling, atrial pressure, surgery trauma, inflammation, oxidative stress, and sympathetic/parasympathetic activation have been implicated in the development of POAF. On the basis of multifactorial mechanism underlying the onset of POAF, several studies have investigated the predictive value of some serum biomarkers. To date, there are promising preliminary data on the clinical utility of PICP, PIINP, TGF-β1 and sCD40L, whereas data on NT-proBNP, BNP, CRP, IL- 6, and hs-cTnT are controversial. </P><P> Conclusion: Although some studies have shown promising results, there is a need for future larger studies with longer follow-up, before applying biomarkers as tools for POAF risk-stratification into clinical practice.</p>
Collapse
Affiliation(s)
- Olivia Manfrini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Edina Cenko
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Beatrice Ricci
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Raffaele Bugiardini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| |
Collapse
|
49
|
Zheng D, Zhang Y, Hu Y, Guan J, Xu L, Xiao W, Zhong Q, Ren C, Lu J, Liang J, Hou J. Long noncoding RNA Crnde attenuates cardiac fibrosis via Smad3-Crnde negative feedback in diabetic cardiomyopathy. FEBS J 2019; 286:1645-1655. [PMID: 30748104 PMCID: PMC6849551 DOI: 10.1111/febs.14780] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 01/02/2019] [Accepted: 02/10/2019] [Indexed: 01/08/2023]
Abstract
Diabetic cardiomyopathy (DCM)-ventricular dysfunction in the absence of underlying heart disease-is a common complication of diabetes and a leading cause of mortality associated with the disease. In DCM, cardiac fibrosis is the main cause of heart failure. Although it is well-established that the transforming growth factor-beta signaling pathway plays a part in inducing cardiac fibrosis in DCM, details of the molecular mechanism involved remain elusive. Therefore, it is crucial to study the gene reg;ulation of key signaling effectors in DCM-associated cardiac fibrosis. A recently emerged hotspot in the field of gene regulation is the role of long noncoding RNAs (lncRNAs). Recent evidence indicates that lncRNAs play a critical role in cardiac fibrosis; however, in DCM, the function of these regulatory RNAs have not been studied in depth. In this study, we identified a conserved cardiac-specific lncRNA named colorectal neoplasia differentially expressed (Crnde). By analyzing 376 human heart tissues, it was found that Crnde expression is negatively correlated with that of cardiac fibrosis marker genes. Moreover, Crnde expression was shown to be enriched in cardiac fibroblasts (CFs). Overexpression of Crnde attenuated cardiac fibrosis and enhanced cardiac function in mice with DCM. Further, in vitro experiments showed that Crnde negatively regulates the myofibroblast differentiation of CFs. The expression of Crnde was activated by SMAD family member 3 (Smad3), shedding light on the underlying molecular mechanism. Interestingly, Crnde also inhibited the transcriptional activation of Smad3 on target genes, thereby inhibiting the expression of myofibroblastic marker genes in CFs. Overall, our data provide valuable insights into the development of potential anti-cardiac fibrosis strategies centered on lncRNAs, for the treatment of DCM.
Collapse
Affiliation(s)
- Dezhi Zheng
- Department of Cardiovascular Surgery, The 960th Hospital of the PLA Joint Logistic Support Force, Jinan, China
| | - Yong Zhang
- Department of Cardiovascular Surgery, The 960th Hospital of the PLA Joint Logistic Support Force, Jinan, China
| | - Yonghe Hu
- Department of Pharmacy, The General Hospital of Western Theater Command, Chengdu, China
| | - Jing Guan
- Department of Radiology, The General Hospital of Western Theater Command, Chengdu, China
| | - Lianbin Xu
- Department of Cardiovascular Surgery, The 960th Hospital of the PLA Joint Logistic Support Force, Jinan, China
| | - Wenjing Xiao
- Department of Pharmacy, The General Hospital of Western Theater Command, Chengdu, China
| | - Qinyue Zhong
- Department of Pharmacy, The General Hospital of Western Theater Command, Chengdu, China
| | - Chao Ren
- Department of Cardiovascular Surgery, The 960th Hospital of the PLA Joint Logistic Support Force, Jinan, China
| | - Jinfeng Lu
- Department of Cardiovascular Surgery, The 960th Hospital of the PLA Joint Logistic Support Force, Jinan, China
| | - Jiali Liang
- Department of Cardiovascular Surgery, The 960th Hospital of the PLA Joint Logistic Support Force, Jinan, China
| | - Jun Hou
- Department of Pharmacy, The General Hospital of Western Theater Command, Chengdu, China
| |
Collapse
|
50
|
Shukla SK, Rafiq K. Proteasome biology and therapeutics in cardiac diseases. Transl Res 2019; 205:64-76. [PMID: 30342797 PMCID: PMC6372329 DOI: 10.1016/j.trsl.2018.09.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 08/30/2018] [Accepted: 09/25/2018] [Indexed: 02/07/2023]
Abstract
The ubiquitin proteasome system (UPS) is the major pathway for intracellular protein degradation in most organs, including the heart. UPS controls many fundamental biological processes such as cell cycle, cell division, immune responses, antigen presentation, apoptosis, and cell signaling. The UPS not only degrades substrates but also regulates activity of gene transcription at the post-transcription level. Emerging evidence suggests that impairment of UPS function is sufficient to cause a number of cardiac diseases, including heart failure, cardiomyopathies, hypertrophy, atrophy, ischemia-reperfusion, and atherosclerosis. Alterations in the expression of UPS components, changes in proteasomal peptidase activities and increased ubiquitinated and oxidized proteins have also been detected in diabetic cardiomyopathy (DCM). However, the pathophysiological role of the UPS in DCM has not been examined. Recently, in vitro and in vivo studies have proven highly valuable in assessing effects of various stressors on the UPS and, in some cases, suggesting a causal link between defective protein clearance and disease phenotypes in different cardiac diseases, including DCM. Translation of these findings to human disease can be greatly strengthened by corroboration of discoveries from experimental model systems using human heart tissue from well-defined patient populations. This review will summarize the general role of the UPS in different cardiac diseases, with major focus on DCM, and on recent advances in therapeutic development.
Collapse
Affiliation(s)
- Sanket Kumar Shukla
- Department of Medicine, Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Khadija Rafiq
- Department of Medicine, Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania.
| |
Collapse
|