1
|
Rohun J, Dudzik D, Raczak-Gutknecht J, Wabich E, Młodziński K, Markuszewski MJ, Daniłowicz-Szymanowicz L. Metabolomics in Atrial Fibrillation: Unlocking Novel Biomarkers and Pathways for Diagnosis, Prognosis, and Personalized Treatment. J Clin Med 2024; 14:34. [PMID: 39797116 PMCID: PMC11722095 DOI: 10.3390/jcm14010034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/16/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND/OBJECTIVES Atrial fibrillation (AF) is the most frequent arrhythmia in the adult population associated with a high rate of severe consequences leading to significant morbidity and mortality worldwide. Therefore, its prompt recognition is of high clinical importance. AF detection often remains challenging due to unspecific symptoms and a lack of reliable biomarkers for its prediction. Herein, novel bioanalytical methodologies, such as metabolomics, offer new opportunities for a better understanding of the underlying pathological mechanisms of cardiovascular diseases, including AF. The metabolome, considered a complete set of small molecules present in the organism, directly reflects the current phenotype of the studied system and is highly sensitive to any changes, including arrhythmia's onset. A growing body of evidence suggests that metabolite profiling has prognostic value in AF prediction, highlighting its potential role not only in early diagnosis but also in guiding therapeutic interventions. By identifying specific metabolites as a disease biomarker or recognising particular metabolomic pathways involved in the AF pathomechanisms, metabolomics could be of great clinical value for further clinical decision-making, risk stratification, and an individual personalised approach. The presented narrative review aims to summarise the current state of knowledge on metabolomics in AF with a special emphasis on its implications for clinical practice and personalised medicine.
Collapse
Affiliation(s)
- Justyna Rohun
- Department of Cardiology and Electrotherapy, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdańsk, Poland; (J.R.); (E.W.); (K.M.)
| | - Danuta Dudzik
- Department of Biopharmaceutics and Pharmacodynamics, Faculty of Pharmacy, Medical University of Gdansk, 80-416 Gdańsk, Poland; (D.D.); (J.R.-G.); (M.J.M.)
| | - Joanna Raczak-Gutknecht
- Department of Biopharmaceutics and Pharmacodynamics, Faculty of Pharmacy, Medical University of Gdansk, 80-416 Gdańsk, Poland; (D.D.); (J.R.-G.); (M.J.M.)
| | - Elżbieta Wabich
- Department of Cardiology and Electrotherapy, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdańsk, Poland; (J.R.); (E.W.); (K.M.)
| | - Krzysztof Młodziński
- Department of Cardiology and Electrotherapy, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdańsk, Poland; (J.R.); (E.W.); (K.M.)
| | - Michał J. Markuszewski
- Department of Biopharmaceutics and Pharmacodynamics, Faculty of Pharmacy, Medical University of Gdansk, 80-416 Gdańsk, Poland; (D.D.); (J.R.-G.); (M.J.M.)
| | - Ludmiła Daniłowicz-Szymanowicz
- Department of Cardiology and Electrotherapy, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdańsk, Poland; (J.R.); (E.W.); (K.M.)
| |
Collapse
|
2
|
Pfenniger A, Yoo S, Arora R. Oxidative stress and atrial fibrillation. J Mol Cell Cardiol 2024; 196:141-151. [PMID: 39307416 DOI: 10.1016/j.yjmcc.2024.09.011] [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: 12/11/2023] [Revised: 09/09/2024] [Accepted: 09/20/2024] [Indexed: 10/05/2024]
Abstract
Atrial fibrillation (AF) is the most common sustained arrhythmia in clinical practice. Though the pathogenesis of AF is complex and is not completely understood, many studies suggest that oxidative stress is a major mechanism in pathophysiology of AF. Through multiple mechanisms, reactive oxygen species (ROS) lead to the formation of an AF substrate that facilitates the development and maintenance of AF. In this review article, we provide an update on the different mechanisms by which oxidative stress promotes atrial remodeling. We then discuss several therapeutic strategies targeting oxidative stress for the prevention or treatment of AF. Considering the complex biology of ROS induced remodeling, and the evolution of ROS sources and compartmentalization during AF progression, there is a definite need for improvement in timing, targeting and reduction of off-target effects of therapeutic strategies targeting oxidative injury in AF.
Collapse
Affiliation(s)
- Anna Pfenniger
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
| | - Shin Yoo
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
| | - Rishi Arora
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America.
| |
Collapse
|
3
|
Ramos-Mondragón R, Lozhkin A, Vendrov AE, Runge MS, Isom LL, Madamanchi NR. NADPH Oxidases and Oxidative Stress in the Pathogenesis of Atrial Fibrillation. Antioxidants (Basel) 2023; 12:1833. [PMID: 37891912 PMCID: PMC10604902 DOI: 10.3390/antiox12101833] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/26/2023] [Accepted: 09/29/2023] [Indexed: 10/29/2023] Open
Abstract
Atrial fibrillation (AF) is the most common type of cardiac arrhythmia and its prevalence increases with age. The irregular and rapid contraction of the atria can lead to ineffective blood pumping, local blood stasis, blood clots, ischemic stroke, and heart failure. NADPH oxidases (NOX) and mitochondria are the main sources of reactive oxygen species in the heart, and dysregulated activation of NOX and mitochondrial dysfunction are associated with AF pathogenesis. NOX- and mitochondria-derived oxidative stress contribute to the onset of paroxysmal AF by inducing electrophysiological changes in atrial myocytes and structural remodeling in the atria. Because high atrial activity causes cardiac myocytes to expend extremely high energy to maintain excitation-contraction coupling during persistent AF, mitochondria, the primary energy source, undergo metabolic stress, affecting their morphology, Ca2+ handling, and ATP generation. In this review, we discuss the role of oxidative stress in activating AF-triggered activities, regulating intracellular Ca2+ handling, and functional and anatomical reentry mechanisms, all of which are associated with AF initiation, perpetuation, and progression. Changes in the extracellular matrix, inflammation, ion channel expression and function, myofibril structure, and mitochondrial function occur during the early transitional stages of AF, opening a window of opportunity to target NOX and mitochondria-derived oxidative stress using isoform-specific NOX inhibitors and mitochondrial ROS scavengers, as well as drugs that improve mitochondrial dynamics and metabolism to treat persistent AF and its transition to permanent AF.
Collapse
Affiliation(s)
- Roberto Ramos-Mondragón
- Department of Pharmacology, University of Michigan, 1150 West Medical Center Drive, 2301 Medical Science Research Building III, Ann Arbor, MI 48109, USA; (R.R.-M.); (L.L.I.)
| | - Andrey Lozhkin
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48019, USA; (A.L.); (A.E.V.); (M.S.R.)
| | - Aleksandr E. Vendrov
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48019, USA; (A.L.); (A.E.V.); (M.S.R.)
| | - Marschall S. Runge
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48019, USA; (A.L.); (A.E.V.); (M.S.R.)
| | - Lori L. Isom
- Department of Pharmacology, University of Michigan, 1150 West Medical Center Drive, 2301 Medical Science Research Building III, Ann Arbor, MI 48109, USA; (R.R.-M.); (L.L.I.)
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Nageswara R. Madamanchi
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48019, USA; (A.L.); (A.E.V.); (M.S.R.)
| |
Collapse
|
4
|
Ma S, Yan F, Hou Y. Intermedin 1-53 Ameliorates Atrial Fibrosis and Reduces Inducibility of Atrial Fibrillation via TGF-β1/pSmad3 and Nox4 Pathway in a Rat Model of Heart Failure. J Clin Med 2023; 12:jcm12041537. [PMID: 36836072 PMCID: PMC9959393 DOI: 10.3390/jcm12041537] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 02/07/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
OBJECTIVE New drugs to block the occurrence of atrial fibrillation (AF) based on atrial structural remodeling (ASR) are urgently needed. The purpose of this study was to study the role of intermedin 1-53 (IMD1-53) in ASR and AF formation in rats after myocardial infarction (MI). MATERIAL AND METHODS Heart failure was induced by MI in rats. Fourteen days after MI surgery, rats with heart failure were randomized into control (untreated MI group, n = 10) and IMD-treated (n = 10) groups. The MI group and sham group received saline injections. The rats in the IMD group received IMD1-53, 10 nmol/kg/day intraperitoneally for 4 weeks. The AF inducibility and atrial effective refractory period (AERP) were assessed with an electrophysiology test. Additionally, the left-atrial diameter was determined, and heart function and hemodynamic tests were performed. We detected the area changes of myocardial fibrosis in the left atrium using Masson staining. To detect the protein expression and mRNA expression of transforming growth factor-β1 (TGF-β1), α-SMA, collagen Ⅰ, collagen III, and NADPH oxidase (Nox4) in the myocardial fibroblasts and left atrium, we used the Western blot method and real-time quantitative polymerase chain reaction (PCR) assays. RESULTS Compared with the MI group, IMD1-53 treatment decreased the left-atrial diameter and improved cardiac function, while it also improved the left-ventricle end-diastolic pressure (LVEDP). IMD1-53 treatment attenuated AERP prolongation and reduced atrial fibrillation inducibility in the IMD group. In vivo, IMD1-53 reduced the left-atrial fibrosis content in the heart after MI surgery and inhibited the mRNA and protein expression of collagen type Ⅰ and III. IMD1-53 also inhibited the expression of TGF-β1, α-SMA, and Nox4 both in mRNA and protein. In vivo, we found that IMD1-53 inhibited the phosphorylation of Smad3. In vitro, we found that the downregulated expression of Nox4 was partly dependent on the TGF-β1/ALK5 pathway. CONCLUSIONS IMD1-53 decreased the duration and inducibility of AF and atrial fibrosis in the rats after MI operation. The possible mechanisms are related to the inhibition of TGF-β1/Smad3-related fibrosis and TGF-β1/Nox4 activity. Therefore, IMD1-53 may be a promising upstream treatment drug to prevent AF.
Collapse
Affiliation(s)
- Shenzhou Ma
- Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250100, China
- Cardiology Departments, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China
| | - Feng Yan
- Department of Emergency Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250100, China
| | - Yinglong Hou
- Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250100, China
- Cardiology Departments, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China
- Correspondence:
| |
Collapse
|
5
|
Distress-Mediated Remodeling of Cardiac Connexin-43 in a Novel Cell Model for Arrhythmogenic Heart Diseases. Int J Mol Sci 2022; 23:ijms231710174. [PMID: 36077591 PMCID: PMC9456330 DOI: 10.3390/ijms231710174] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 11/16/2022] Open
Abstract
Gap junctions and their expression pattern are essential to robust function of intercellular communication and electrical propagation in cardiomyocytes. In healthy myocytes, the main cardiac gap junction protein connexin-43 (Cx43) is located at the intercalated disc providing a clear direction of signal spreading across the cardiac tissue. Dislocation of Cx43 to lateral membranes has been detected in numerous cardiac diseases leading to slowed conduction and high propensity for the development of arrhythmias. At the cellular level, arrhythmogenic diseases are associated with elevated levels of oxidative distress and gap junction remodeling affecting especially the amount and sarcolemmal distribution of Cx43 expression. So far, a mechanistic link between sustained oxidative distress and altered Cx43 expression has not yet been identified. Here, we propose a novel cell model based on murine induced-pluripotent stem cell-derived cardiomyocytes to investigate subcellular signaling pathways linking cardiomyocyte distress with gap junction remodeling. We tested the new hypothesis that chronic distress, induced by rapid pacing, leads to increased reactive oxygen species, which promotes expression of a micro-RNA, miR-1, specific for the control of Cx43. Our data demonstrate that Cx43 expression is highly sensitive to oxidative distress, leading to reduced expression. This effect can be efficiently prevented by the glutathione peroxidase mimetic ebselen. Moreover, Cx43 expression is tightly regulated by miR-1, which is activated by tachypacing-induced oxidative distress. In light of the high arrhythmogenic potential of altered Cx43 expression, we propose miR-1 as a novel target for pharmacological interventions to prevent the maladaptive remodeling processes during chronic distress in the heart.
Collapse
|
6
|
Lai YJ, Tsai FC, Chang GJ, Chang SH, Huang CC, Chen WJ, Yeh YH. miR-181b targets semaphorin 3A to mediate TGF-β-induced endothelial-mesenchymal transition related to atrial fibrillation. J Clin Invest 2022; 132:142548. [PMID: 35775491 PMCID: PMC9246393 DOI: 10.1172/jci142548] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 05/10/2022] [Indexed: 11/17/2022] Open
Abstract
Atrial fibrosis is an essential contributor to atrial fibrillation (AF). It remains unclear whether atrial endocardial endothelial cells (AEECs) that undergo endothelial-mesenchymal transition (EndMT) are among the sources of atrial fibroblasts. We studied human atria, TGF-β-treated human AEECs, cardiac-specific TGF-β-transgenic mice, and heart failure rabbits to identify the underlying mechanism of EndMT in atrial fibrosis. Using isolated AEECs, we found that miR-181b was induced in TGF-β-treated AEECs, which decreased semaphorin 3A (Sema3A) and increased EndMT markers, and these effects could be reversed by a miR-181b antagomir. Experiments in which Sema3A was increased by a peptide or decreased by a siRNA in AEECs revealed a mechanistic link between Sema3A and LIM-kinase 1/phosphorylated cofilin (LIMK/p-cofilin) signaling and suggested that Sema3A is upstream of LIMK in regulating actin remodeling through p-cofilin. Administration of the miR-181b antagomir or recombinant Sema3A to TGF-β-transgenic mice evoked increased Sema3A, reduced EndMT markers, and significantly decreased atrial fibrosis and AF vulnerability. Our study provides a mechanistic link between the induction of EndMT by TGF-β via miR-181b/Sema3A/LIMK/p-cofilin signaling to atrial fibrosis. Blocking miR-181b and increasing Sema3A are potential strategies for AF therapeutic intervention.
Collapse
Affiliation(s)
- Ying-Ju Lai
- Cardiovascular Department, Chang Gung Memorial Hospital, Tao Yuan, Taiwan.,Department of Respiratory Therapy, Chang Gung University College of Medicine, Tao Yuan, Taiwan.,Department of Respiratory Care, Chang Gung University of Science and Technology, Chia Yi, Taiwan
| | - Feng-Chun Tsai
- Department of Thoracic and Cardiovascular Surgery, Chang Gung Memorial Hospital, Tao Yuan, Taiwan.,Department of Medicine and
| | - Gwo-Jyh Chang
- Cardiovascular Department, Chang Gung Memorial Hospital, Tao Yuan, Taiwan.,Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Tao Yuan, Taiwan
| | - Shang-Hung Chang
- Cardiovascular Department, Chang Gung Memorial Hospital, Tao Yuan, Taiwan.,Department of Medicine and
| | - Chung-Chi Huang
- Department of Respiratory Therapy, Chang Gung University College of Medicine, Tao Yuan, Taiwan.,Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Tao Yuan, Taiwan
| | - Wei-Jan Chen
- Cardiovascular Department, Chang Gung Memorial Hospital, Tao Yuan, Taiwan.,Department of Medicine and
| | - Yung-Hsin Yeh
- Cardiovascular Department, Chang Gung Memorial Hospital, Tao Yuan, Taiwan.,Department of Medicine and
| |
Collapse
|
7
|
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
|
8
|
Chen YL, Wang HT, Lin PT, Chuang JH, Yang MY. Macrophage Inflammatory Protein-1 Alpha, a Potential Biomarker for Predicting Left Atrial Remodeling in Patients With Atrial Fibrillation. Front Cardiovasc Med 2021; 8:784792. [PMID: 34957262 PMCID: PMC8695724 DOI: 10.3389/fcvm.2021.784792] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/17/2021] [Indexed: 11/13/2022] Open
Abstract
Objectives: Left atrial (LA) remodeling itself is an independent risk factor for ischemic stroke and mortality, with or without atrial fibrillation (AF). Macrophage inflammatory protein-1 alpha (MIP-1α) has been reported to be involved in the induction of autoimmune myocarditis and dilated cardiomyopathy. Little is known about whether MIP-1α can be used to predict LA remodeling, especially in patients with AF. Methods: We prospectively enrolled 78 patients who had received a cardiac implantable electronic device due to sick sinus syndrome in order to define AF accurately. AF was diagnosed clinically before enrollment, according to 12-lead electrocardiography (ECG) and 24-h Holter test in 54 (69%) patients. The serum cytokine levels and the mRNA expression levels of peripheral blood leukocytes were checked and echocardiographic study was performed on the same day within 1 week after the patients were enrolled into the study. The 12-lead ECG and 24-h Holter test were performed on the same day of the patients' enrollment, and the device interrogation was performed every 3 months after enrollment. The enrolled patients were clinically followed up for 1 year. Results: There was no difference in baseline characteristics, cytokine levels and mRNA expression between patients with and without AF. Larger LA volume was positively correlated with higher levels of MIP-1α (r = 0.461, p ≤ 0.001) and the atrial high-rate episodes (AHREs) burden (r = 0.593, p < 0.001), and negatively correlated with higher levels of transforming growth factor (TGF)-β1 (r = −0.271, p = 0.047) and TGF-β3 (r = −0.279, p = 0.041). The higher AHREs burden and MIP-1α level could predict LA volume independently. The mRNA expression of RORC was negatively associated with the MIP-1α level. Conclusions: This study showed that higher MIP-1α was significantly associated with LA remodeling and may have the potentials to predict LA remodeling in terms of a larger LA volume, and that circadian gene derangement might affect the expression of MIP-1α.
Collapse
Affiliation(s)
- Yung-Lung Chen
- Section of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan.,School of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,College of Medicine, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Hui-Ting Wang
- Emergency Department, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Pei-Ting Lin
- Section of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan.,School of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Jiin-Haur Chuang
- College of Medicine, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan.,Division of Pediatric Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Ming-Yu Yang
- College of Medicine, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan.,Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| |
Collapse
|
9
|
Chen M, Zhong J, Wang Z, Xu H, Chen H, Sun X, Lu Y, Chen L, Xie X, Zheng L. Fibroblast Growth Factor 21 Protects Against Atrial Remodeling via Reducing Oxidative Stress. Front Cardiovasc Med 2021; 8:720581. [PMID: 34708083 PMCID: PMC8542911 DOI: 10.3389/fcvm.2021.720581] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 09/06/2021] [Indexed: 11/13/2022] Open
Abstract
Aim: The structural and electrical changes in the atrium, also known as atrial remodeling, are the main characteristics of atrial fibrillation (AF). Fibroblast growth factor 21 (Fgf21) is an important endocrine factor, which has been shown to play an important role in cardiovascular diseases. However, the effects of Fgf21 on atrial remodeling have not been addressed yet. The purpose of the present study is to evaluate the effects of Fgf21 on atrial remodeling. Methods and Results: Adult mice were treated with Ang II, and randomly administrated with or without Fgf21 for 2 weeks. The susceptibility to AF was assessed by electrical stimulation and optical mapping techniques. Here, we found that Fgf21 administration attenuated the inducibility of atrial fibrillation/atrial tachycardia (AF/AT), improved epicardial conduction velocity in the mice atria. Mechanistically, Fgf21 protected against atrial fibrosis and reduced oxidative stress of the atria. Consistently, in vitro study also demonstrated that Fgf21 blocked the upregulation of collagen by Tgf-β in fibroblasts and attenuated tachypacing-induced oxidative stress including reactive oxygen species (ROS), Tgf-β, and ox-CaMKII in atrial myocytes. We further found that Fgf21 attenuated oxidative stress by inducing antioxidant genes, such as SOD2 and UCP3. Fgf21 also improved tachypacing-induced myofibril degradation, downregulation of L-type calcium channel, and upregulation of p-RyR2, which implicated protective effects of Fgf21 on structural and electrical remodeling in the atria. Moreover, Nrf2 was identified as a downstream of Fgf21 and partly mediated Fgf21-induced antioxidant gene expression in atrial myocytes. Conclusion: Fgf21 administration effectively suppressed atrial remodeling by reducing oxidative stress, which provides a novel therapeutic insight for AF.
Collapse
Affiliation(s)
- Miao Chen
- Department of Cardiology and Atrial Fibrillation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jiawei Zhong
- Department of Cardiology and Atrial Fibrillation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhen Wang
- Department of Cardiology and Atrial Fibrillation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hongfei Xu
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Heng Chen
- Department of Cardiology and Atrial Fibrillation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xingang Sun
- Department of Cardiology and Atrial Fibrillation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yunlong Lu
- Department of Cardiology and Atrial Fibrillation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lu Chen
- Department of Cardiology and Atrial Fibrillation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xudong Xie
- Department of Cardiology and Atrial Fibrillation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Liangrong Zheng
- Department of Cardiology and Atrial Fibrillation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
10
|
Chang SH, Chan YH, Chen WJ, Chang GJ, Lee JL, Yeh YH. Tachypacing-induced CREB/CD44 signaling contributes to the suppression of L-type calcium channel expression and the development of atrial remodeling. Heart Rhythm 2021; 18:1760-1771. [PMID: 34023501 DOI: 10.1016/j.hrthm.2021.05.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 05/11/2021] [Accepted: 05/14/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND Atrial fibrillation (AF), a common arrhythmia in clinics, is characterized as downregulation of L-type calcium channel (LTCC) and shortening of atrial action potential duration (APD). Our prior studies have shown the association of CD44 with AF genesis. OBJECTIVE The purpose of this study was to explore the potential role of CD44 and its related signaling in tachypacing-induced downregulation of LTCC. METHODS AND RESULTS In vitro, tachypacing in atrium-derived myocytes (HL-1 cell line) induced activation (phosphorylation) of cyclic adenosine monophosphate response element-binding protein (CREB). Furthermore, tachypacing promoted an association between CREB and CD44 in HL-1 myocytes, which was documented in atrial tissues from patients with AF. Deletion and mutational analysis of the LTCC promoter along with chromatin immunoprecipitation revealed that cyclic adenosine monophosphate response element is essential for tachypacing-inhibited LTCC transcription. Tachypacing also hindered the binding of p-CREB to the promoter of LTCC. Blockade of CREB/CD44 signaling in HL-1 cells attenuated tachypacing-triggered downregulation of LTCC and shortening of APD. Atrial myocytes isolated from CD44-/- mice exhibited higher LTCC current and longer APD than did those from wild-type mice. Ex vivo, tachypacing caused less activation of CREB in CD44-/- mice than in wild-type mice. In vivo, burst atrial pacing stimulated less inducibility of AF in CREB inhibitor-treated mice than in controls. CONCLUSION Tachypacing-induced CREB/CD44 signaling contributes to the suppression of LTCC, which provides valuable information about the pathogenesis of atrial modeling and AF.
Collapse
Affiliation(s)
- Shang-Hung Chang
- Division of Cardiology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Tao-Yuan, Taiwan
| | - Yi-Hsin Chan
- Division of Cardiology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Tao-Yuan, Taiwan
| | - Wei-Jan Chen
- Division of Cardiology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Tao-Yuan, Taiwan.
| | - Gwo-Jyh Chang
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Tao-Yuan, Taiwan
| | - Jia-Lin Lee
- Institute of Molecular and Cellular Biology and Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Yung-Hsin Yeh
- Division of Cardiology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Tao-Yuan, Taiwan
| |
Collapse
|
11
|
Berto S, Fontenot MR, Seger S, Ayhan F, Caglayan E, Kulkarni A, Douglas C, Tamminga CA, Lega BC, Konopka G. Gene-expression correlates of the oscillatory signatures supporting human episodic memory encoding. Nat Neurosci 2021; 24:554-564. [PMID: 33686299 PMCID: PMC8016736 DOI: 10.1038/s41593-021-00803-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 01/19/2021] [Indexed: 12/13/2022]
Abstract
In humans, brain oscillations support critical features of memory formation. However, understanding the molecular mechanisms underlying this activity remains a major challenge. Here, we measured memory-sensitive oscillations using intracranial electroencephalography recordings from the temporal cortex of patients performing an episodic memory task. When these patients subsequently underwent resection, we employed transcriptomics on the temporal cortex to link gene expression with brain oscillations and identified genes correlated with oscillatory signatures of memory formation across six frequency bands. A co-expression analysis isolated oscillatory signature-specific modules associated with neuropsychiatric disorders and ion channel activity, with highly correlated genes exhibiting strong connectivity within these modules. Using single-nucleus transcriptomics, we further revealed that these modules are enriched for specific classes of both excitatory and inhibitory neurons, and immunohistochemistry confirmed expression of highly correlated genes. This unprecedented dataset of patient-specific brain oscillations coupled to genomics unlocks new insights into the genetic mechanisms that support memory encoding.
Collapse
Affiliation(s)
- Stefano Berto
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, USA
| | - Miles R Fontenot
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, USA
| | - Sarah Seger
- Department of Neurosurgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Fatma Ayhan
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, USA
| | - Emre Caglayan
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, USA
| | | | - Connor Douglas
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, USA
| | - Carol A Tamminga
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX, USA
| | - Bradley C Lega
- Department of Neurosurgery, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Genevieve Konopka
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
12
|
Li CY, Zhang JR, Hu WN, Li SN. Atrial fibrosis underlying atrial fibrillation (Review). Int J Mol Med 2021; 47:9. [PMID: 33448312 PMCID: PMC7834953 DOI: 10.3892/ijmm.2020.4842] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 12/07/2020] [Indexed: 01/17/2023] Open
Abstract
Atrial fibrillation (AF) is one of the most common tachyarrhythmias observed in the clinic and is characterized by structural and electrical remodelling. Atrial fibrosis, an emblem of atrial structural remodelling, is a complex multifactorial and patient-specific process involved in the occurrence and maintenance of AF. Whilst there is already considerable knowledge regarding the association between AF and fibrosis, this process is extremely complex, involving intricate neurohumoral and cellular and molecular interactions, and it is not limited to the atrium. Current technological advances have made the non-invasive evaluation of fibrosis in the atria and ventricles possible, facilitating the selection of patient-specific ablation strategies and upstream treatment regimens. An improved understanding of the mechanisms and roles of fibrosis in the context of AF is of great clinical significance for the development of treatment strategies targeting the fibrous region. In the present review, a focus was placed on the atrial fibrosis underlying AF, outlining its role in the occurrence and perpetuation of AF, by reviewing recent evaluations and potential treatment strategies targeting areas of fibrosis, with the aim of providing a novel perspective on the management and prevention of AF.
Collapse
Affiliation(s)
- Chang Yi Li
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Jing Rui Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Wan Ning Hu
- Department of Cardiology, Laboratory of Molecular Biology, Head and Neck Surgery, Tangshan Gongren Hospital, Tangshan, Hebei 063000, P.R. China
| | - Song Nan Li
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| |
Collapse
|
13
|
Aldehyde Dehydrogenase 2 Ameliorates Chronic Alcohol Consumption-Induced Atrial Fibrillation through Detoxification of 4-HNE. Int J Mol Sci 2020; 21:ijms21186678. [PMID: 32932651 PMCID: PMC7555032 DOI: 10.3390/ijms21186678] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/08/2020] [Accepted: 09/10/2020] [Indexed: 12/22/2022] Open
Abstract
Aldehyde dehydrogenase 2 (ALDH2) is an enzyme that detoxifies reactive oxygen species (ROS)-generated aldehyde adducts such as 4-hydroxy-trans-2-nonenal (4-HNE). Previous meta-analyses have shown an increase in the risk of atrial fibrillation (AF) in patients with chronic alcohol consumption. ALDH2*2, a common dysfunctional polymorphism in the ALDH2 gene, has been linked to an increased risk of cancer and heart disease. We tested the effect of ALDH2 deficiency on alcohol-induced AF in a murine model of chronic-binge ethanol feeding, with ALDH2*2 knock-in (KI) mice generated by a CRISPR/CAS9 system. In addition, right atrial appendages were obtained from eight patients with AF undergoing open heart surgery. The results showed that burst atrial pacing induced a greater susceptibility to AF in ALDH2*2 KI mice exposed to chronic ethanol intoxication than in wild-type mice, resulting from a higher degree of 4-HNE accumulation and collagen deposition in their atria. Alda-1 attenuated transforming growth factor beta 1 (TGF-β1) expression and collagen deposition in the atria and reduced AF inducibility. Patients with AF and the ALDH2*2 allele exhibited greater oxidative stress and substrate remodeling in their atria than non-carriers. In conclusion, ALDH2 deficiency may increase the risk of chronic alcohol and tachypacing-induced AF through the accumulation of 4-HNE and increased ROS production.
Collapse
|
14
|
Tachycardia-induced CD44/NOX4 signaling is involved in the development of atrial remodeling. J Mol Cell Cardiol 2019; 135:67-78. [DOI: 10.1016/j.yjmcc.2019.08.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 07/30/2019] [Accepted: 08/12/2019] [Indexed: 12/15/2022]
|
15
|
Yang Z, Xiao Z, Guo H, Fang X, Liang J, Zhu J, Yang J, Li H, Pan R, Yuan S, Dong W, Zheng XL, Wu S, Shan Z. Novel role of the clustered miR-23b-3p and miR-27b-3p in enhanced expression of fibrosis-associated genes by targeting TGFBR3 in atrial fibroblasts. J Cell Mol Med 2019; 23:3246-3256. [PMID: 30729664 PMCID: PMC6484421 DOI: 10.1111/jcmm.14211] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 12/27/2018] [Accepted: 01/17/2019] [Indexed: 01/29/2023] Open
Abstract
Atrial fibrillation (AF) is the most common type of arrhythmia in cardiovascular diseases. Atrial fibrosis is an important pathophysiological contributor to AF. This study aimed to investigate the role of the clustered miR‐23b‐3p and miR‐27b‐3p in atrial fibrosis. Human atrial fibroblasts (HAFs) were isolated from atrial appendage tissue of patients with sinus rhythm. A cell model of atrial fibrosis was achieved in Ang‐II‐induced HAFs. Cell proliferation and migration were detected. We found that miR‐23b‐3p and miR‐27b‐3p were markedly increased in atrial appendage tissues of AF patients and in Ang‐II‐treated HAFs. Overexpression of miR‐23b‐3p and miR‐27b‐3p enhanced the expression of collagen, type I, alpha 1 (COL1A1), COL3A1 and ACTA2 in HAFs without significant effects on their proliferation and migration. Luciferase assay showed that miR‐23b‐3p and miR‐27b‐3p targeted two different sites in 3ʹ‐UTR of transforming growth factor (TGF)‐β1 receptor 3 (TGFBR3) respectively. Consistently, TGFBR3 siRNA could increase fibrosis‐related genes expression, along with the Smad1 inactivation and Smad3 activation in HAFs. Additionally, overexpression of TGFBR3 could alleviate the increase of COL1A1, COL3A1 and ACTA2 in HAFs after transfection with miR‐23b‐3p and miR‐27b‐3p respectively. Moreover, Smad3 was activated in HAFs in response to Ang‐II treatment and inactivation of Smad3 attenuated up‐regulation of miR‐23b‐3p and miR‐27b‐3p in Ang‐II‐treated HAFs. Taken together, these results suggest that the clustered miR‐23b‐3p and miR‐27b‐3p consistently promote atrial fibrosis by targeting TGFBR3 to activate Smad3 signalling in HAFs, suggesting that miR‐23b‐3p and miR‐27b‐3p are potential therapeutic targets for atrial fibrosis.
Collapse
Affiliation(s)
- Zhenzhen Yang
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Cardiovascular Institute, Guangzhou, China.,Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,School of Medicine, South China University of Technology, Guangzhou, China
| | - Zhen Xiao
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Cardiovascular Institute, Guangzhou, China.,Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Huiming Guo
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Cardiovascular Institute, Guangzhou, China
| | - Xianhong Fang
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Cardiovascular Institute, Guangzhou, China
| | - Jingnan Liang
- School of Pharmacy, Southern Medical University, Guangzhou, China
| | - Jiening Zhu
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Cardiovascular Institute, Guangzhou, China.,Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jing Yang
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Hui Li
- School of Pharmacy, Southern Medical University, Guangzhou, China
| | - Rong Pan
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Shujing Yuan
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Wenyan Dong
- Guangzhou Women and Children's Medical Center, Institute of Pediatrics, Guangzhou Medical University, Guangzhou, China
| | - Xi-Long Zheng
- Department of Biochemistry & Molecular Biology, The Libin Cardiovascular Institute of Alberta, The University of Calgary, Calgary, Alberta, Canada
| | - Shulin Wu
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Cardiovascular Institute, Guangzhou, China
| | - Zhixin Shan
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Cardiovascular Institute, Guangzhou, China.,Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
16
|
Qiu H, Ji C, Liu W, Wu Y, Lu Z, Lin Q, Xue Z, Liu X, Wu H, Jiang W, Zou C. Chronic Kidney Disease Increases Atrial Fibrillation Inducibility: Involvement of Inflammation, Atrial Fibrosis, and Connexins. Front Physiol 2018; 9:1726. [PMID: 30564139 PMCID: PMC6288485 DOI: 10.3389/fphys.2018.01726] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 11/16/2018] [Indexed: 12/13/2022] Open
Abstract
Chronic kidney disease (CKD) causes atrial structural remodeling and subsequently increases the incidence of atrial fibrillation (AF). Atrial connexins and inflammatory responses may be involved in this remodeling process. In this study, nephrectomy was used to produce the CKD rat model. Three months post-nephrectomy, cardiac structure, function and AF vulnerability were quantified using echocardiography and electrophysiology methods. The left atrial tissue was tested for quantification of fibrosis and inflammation, and for the distribution and expression of connexin (Cx) 40 and Cx43. An echocardiography showed that CKD resulted in the left atrial enlargement and left ventricular hypertrophy, but had no functional changes. CKD caused a significant increase in the AF inducible rate (91.11% in CKD group vs. 6.67% in sham group, P < 0.001) and the AF duration [107 (0–770) s in CKD vs. 0 (0–70) s in sham, P < 0.001] with prolonged P-wave duration. CKD induced severe interstitial fibrosis, activated the transforming growth factor-β1/Smad2/3 pathway with a massive extracellular matrix deposition of collagen type I and α-smooth muscle actin, and matured the NLR (nucleotide-binding domain leucine-rich repeat-containing receptor) pyrin domain-containing protein 3 (NLRP3) inflammasome with an inflammatory cascade response. CKD resulted in an increase in non-phosphorylated-Cx43, a decrease in Cx40 and phosphorylated-Cx43, and lateralized the distribution of Cx40 and Cx43 proteins with upregulations of Rac-1, connective tissue growth factor and N-cadherin. These findings implicate the transforming growth factor-β1/Smad2/3, the NLRP3 inflammasome and the connexins as potential mediators of increased AF vulnerability in CKD.
Collapse
Affiliation(s)
- Huiliang Qiu
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Chunlan Ji
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Wei Liu
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Yuchi Wu
- Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Zhaoyu Lu
- Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Qizhan Lin
- Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Zheng Xue
- Department of Cardiology, Guangzhou Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Xusheng Liu
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Huanlin Wu
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China.,Department of internal medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Wei Jiang
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Chuan Zou
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| |
Collapse
|
17
|
Liou YS, Yang FY, Chen HY, Jong GP. Antihyperglycemic drugs use and new-onset atrial fibrillation: A population-based nested case control study. PLoS One 2018; 13:e0197245. [PMID: 30161122 PMCID: PMC6116917 DOI: 10.1371/journal.pone.0197245] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 04/30/2018] [Indexed: 12/19/2022] Open
Abstract
Currently, the potential risk of atrial fibrillation associated with antihyperglycemic drug use has been a topic of considerable interest. However, it remains uncertain whether different classes of antihyperglycemic drug therapy are associated with the risk of atrial fibrillation risk. Here, we investigated the association between different classes of antihyperglycemic drugs and new-onset atrial fibrillation (NAF). A case-matched study was performed based on the National Health Insurance Program in Taiwan. Patients who had NAF were considered the NAF group and were matched in a 1:4 ratio with patients without NAF, who were assigned to the non-NAF group. Patients were matched according to sex, age, diabetes mellitus duration, index date, and Charlson Comorbidity Index score. We used multivariate logistic regression controlling for potential confounders to examine the association between different classes of antihyperglycemic drug use and the risk of NAF. Overall, we identified 2,882 cases and 11,528 matched controls for the study. After adjusting for sex, age, comorbidities, and concurrent medications, users of biguanides or thiazolidinediones were at a lower risk of developing NAF when compared with non-users (odds ratio [OR] 0.81, 95% confidence interval [CI] 0.71-0.95 and OR 0.72, 95% CI 0.63-0.83, respectively). In contrast, users of insulin were at a higher risk of developing NAF than were non-users (OR 1.19, 95% CI 1.06-1.35). Sulfonylureas, glinides, α-glucosidase inhibitors, and dipeptidyl peptidase-4 inhibitors were not associated with developing the risk of NAF. In conclusion, the use of biguanides or thiazolidinediones may be associated with a low risk of NAF, whereas insulin may be associated with a significant increase in the risk of NAF in patients with type 2 diabetes mellitus during long-term follow-up. Further prospective randomized studies should investigate which specific class of antihyperglycemic drug treatment for diabetes mellitus can prevent or postpone NAF.
Collapse
Affiliation(s)
- Yi-Sheng Liou
- Department of Family Medicine and Geriatrics, Taichung Veteran General Hospital, and School of Public Health, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Fu-Yu Yang
- Institute of Pharmacy, China Medical University, Taichung, Taiwan, ROC
| | - Hung-Yi Chen
- Institute of Pharmacy, China Medical University, Taichung, Taiwan, ROC
- Department of Pharmacy, China Medical University Beigang Hospital, Yunlin County, Taiwan, ROC
| | - Gwo-Ping Jong
- Division of Internal Cardiology, Chung Shan Medical University Hospital and Chung Shan Medical University, Taichung, Taiwan, ROC
| |
Collapse
|
18
|
The Transcription Factor ATF4 Promotes Expression of Cell Stress Genes and Cardiomyocyte Death in a Cellular Model of Atrial Fibrillation. BIOMED RESEARCH INTERNATIONAL 2018; 2018:3694362. [PMID: 30003094 PMCID: PMC5996409 DOI: 10.1155/2018/3694362] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Revised: 03/29/2018] [Accepted: 04/15/2018] [Indexed: 12/19/2022]
Abstract
Introduction Cardiomyocyte remodelling in atrial fibrillation (AF) has been associated with both oxidative stress and endoplasmic reticulum (ER) stress and is accompanied by a complex transcriptional regulation. Here, we investigated the role the oxidative stress and ER stress responsive bZIP transcription factor ATF4 plays in atrial cardiomyocyte viability and AF induced gene expression. Methods HL-1 cardiomyocytes were subjected to rapid field stimulation. Forced expression of ATF4 was achieved by adenoviral gene transfer. Using global gene expression analysis and chromatin immunoprecipitation, ATF4 dependent transcriptional regulation was studied, and tissue specimen of AF patients was analysed by immunohistochemistry. Results Oxidative stress and ER stress caused a significant reduction in cardiomyocyte viability and were associated with an induction of ATF4. Accordingly, ATF4 was also induced by rapid field stimulation mimicking AF. Forced expression of wild type ATF4 promoted cardiomyocyte death. ATF4 was demonstrated to bind to the promoters of several cell stress genes and to induce the expression of a number of ATF4 dependent stress responsive genes. Moreover, immunohistochemical analyses showed that ATF4 is expressed in the nuclei of cardiomyocytes of tissue specimen obtained from AF patients. Conclusion ATF4 is expressed in human atrial cardiomyocytes and is induced in response to different types of cell stress. High rate electrical field stimulation seems to result in ATF4 induction, and forced expression of ATF4 reduces cardiomyocyte viability.
Collapse
|
19
|
Wei W, Rao F, Liu F, Xue Y, Deng C, Wang Z, Zhu J, Yang H, Li X, Zhang M, Fu Y, Zhu W, Shan Z, Wu S. Involvement of Smad3 pathway in atrial fibrosis induced by elevated hydrostatic pressure. J Cell Physiol 2018; 233:4981-4989. [PMID: 29215718 DOI: 10.1002/jcp.26337] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 11/28/2017] [Indexed: 12/18/2022]
Affiliation(s)
- Wei Wei
- Department of Cardiology, Guangdong Cardiovascular Institute; Guangdong Academy of Medical Sciences; Guangzhou P. R. China
- Guangdong Key Laboratory of Clinical Pharmacology; Guangdong Academy of Medical Sciences; Guangzhou P. R. China
| | - Fang Rao
- Department of Cardiology, Guangdong Cardiovascular Institute; Guangdong Academy of Medical Sciences; Guangzhou P. R. China
- Guangdong Key Laboratory of Clinical Pharmacology; Guangdong Academy of Medical Sciences; Guangzhou P. R. China
- Research Center of Medical Sciences, Guangdong General Hospital; Guangdong Academy of Medical Sciences; Guangzhou P. R. China
| | - Fangzhou Liu
- Department of Cardiology, Guangdong Cardiovascular Institute; Guangdong Academy of Medical Sciences; Guangzhou P. R. China
- Guangdong Key Laboratory of Clinical Pharmacology; Guangdong Academy of Medical Sciences; Guangzhou P. R. China
| | - Yumei Xue
- Department of Cardiology, Guangdong Cardiovascular Institute; Guangdong Academy of Medical Sciences; Guangzhou P. R. China
- Guangdong Key Laboratory of Clinical Pharmacology; Guangdong Academy of Medical Sciences; Guangzhou P. R. China
| | - Chunyu Deng
- Guangdong Key Laboratory of Clinical Pharmacology; Guangdong Academy of Medical Sciences; Guangzhou P. R. China
- Research Center of Medical Sciences, Guangdong General Hospital; Guangdong Academy of Medical Sciences; Guangzhou P. R. China
| | - Zhaoyu Wang
- Guangdong Key Laboratory of Clinical Pharmacology; Guangdong Academy of Medical Sciences; Guangzhou P. R. China
- Research Center of Medical Sciences, Guangdong General Hospital; Guangdong Academy of Medical Sciences; Guangzhou P. R. China
| | - Jiening Zhu
- Guangdong Key Laboratory of Clinical Pharmacology; Guangdong Academy of Medical Sciences; Guangzhou P. R. China
- Research Center of Medical Sciences, Guangdong General Hospital; Guangdong Academy of Medical Sciences; Guangzhou P. R. China
| | - Hui Yang
- Guangdong Key Laboratory of Clinical Pharmacology; Guangdong Academy of Medical Sciences; Guangzhou P. R. China
- Research Center of Medical Sciences, Guangdong General Hospital; Guangdong Academy of Medical Sciences; Guangzhou P. R. China
| | - Xin Li
- Department of Cardiology, Guangdong Cardiovascular Institute; Guangdong Academy of Medical Sciences; Guangzhou P. R. China
- Guangdong Key Laboratory of Clinical Pharmacology; Guangdong Academy of Medical Sciences; Guangzhou P. R. China
| | - Mengzhen Zhang
- Guangdong Key Laboratory of Clinical Pharmacology; Guangdong Academy of Medical Sciences; Guangzhou P. R. China
- Research Center of Medical Sciences, Guangdong General Hospital; Guangdong Academy of Medical Sciences; Guangzhou P. R. China
| | - Yongheng Fu
- Guangdong Key Laboratory of Clinical Pharmacology; Guangdong Academy of Medical Sciences; Guangzhou P. R. China
- Research Center of Medical Sciences, Guangdong General Hospital; Guangdong Academy of Medical Sciences; Guangzhou P. R. China
| | - Wensi Zhu
- Guangdong Key Laboratory of Clinical Pharmacology; Guangdong Academy of Medical Sciences; Guangzhou P. R. China
- Research Center of Medical Sciences, Guangdong General Hospital; Guangdong Academy of Medical Sciences; Guangzhou P. R. China
| | - Zhixin Shan
- Guangdong Key Laboratory of Clinical Pharmacology; Guangdong Academy of Medical Sciences; Guangzhou P. R. China
- Research Center of Medical Sciences, Guangdong General Hospital; Guangdong Academy of Medical Sciences; Guangzhou P. R. China
| | - Shulin Wu
- Department of Cardiology, Guangdong Cardiovascular Institute; Guangdong Academy of Medical Sciences; Guangzhou P. R. China
- Guangdong Key Laboratory of Clinical Pharmacology; Guangdong Academy of Medical Sciences; Guangzhou P. R. China
| |
Collapse
|
20
|
McLaughlin D, Zhao Y, O'Neill KM, Edgar KS, Dunne PD, Kearney AM, Grieve DJ, McDermott BJ. Signalling mechanisms underlying doxorubicin and Nox2 NADPH oxidase-induced cardiomyopathy: involvement of mitofusin-2. Br J Pharmacol 2017; 174:3677-3695. [PMID: 28261787 PMCID: PMC5647180 DOI: 10.1111/bph.13773] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 02/27/2017] [Accepted: 03/01/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE The anthracycline doxorubicin (DOX), although successful as a first-line cancer treatment, induces cardiotoxicity linked with increased production of myocardial ROS, with Nox2 NADPH oxidase-derived superoxide reported to play a key role. The aim of this study was to identify novel mechanisms underlying development of cardiac remodelling/dysfunction further to DOX-stimulated Nox2 activation. EXPERIMENTAL APPROACH Nox2-/- and wild-type (WT) littermate mice were administered DOX (12 mg·kg-1 over 3 weeks) prior to study at 4 weeks. Detailed mechanisms were investigated in murine HL-1 cardiomyocytes, employing a robust model of oxidative stress, gene silencing and pharmacological tools. KEY RESULTS DOX-induced cardiac dysfunction, cardiomyocyte remodelling, superoxide production and apoptosis in WT mice were attenuated in Nox2-/- mice. Transcriptional analysis of left ventricular tissue identified 152 differentially regulated genes (using adjusted P < 0.1) in DOX-treated Nox2-/- versus WT mice, and network analysis highlighted 'Cell death and survival' as the biological function most significant to the dataset. The mitochondrial membrane protein, mitofusin-2 (Mfn2), appeared as a strong candidate, with increased expression (1.5-fold), confirmed by qPCR (1.3-fold), matching clear published evidence of promotion of cardiomyocyte cell death. In HL-1 cardiomyocytes, targeted siRNA knockdown of Nox2 decreased Mfn2 protein expression, but not vice versa. While inhibition of Nox2 activity along with DOX treatment attenuated its apoptotic and cytotoxic effects, reduced apoptosis after Mfn2 silencing reflected a sustained cytotoxic response and reduced cell viability. CONCLUSIONS AND IMPLICATIONS DOX-induced and Nox2-mediated up-regulation of Mfn2, rather than contributing to cardiomyocyte dysfunction through apoptotic pathways, appears to promote a protective mechanism. LINKED ARTICLES This article is part of a themed section on New Insights into Cardiotoxicity Caused by Chemotherapeutic Agents. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.21/issuetoc.
Collapse
Affiliation(s)
- Declan McLaughlin
- Centre for Experimental Medicine, Wellcome‐Wolfson BuildingQueen's University BelfastBelfastUK
| | - Youyou Zhao
- Centre for Experimental Medicine, Wellcome‐Wolfson BuildingQueen's University BelfastBelfastUK
| | - Karla M O'Neill
- Centre for Experimental Medicine, Wellcome‐Wolfson BuildingQueen's University BelfastBelfastUK
| | - Kevin S Edgar
- Centre for Experimental Medicine, Wellcome‐Wolfson BuildingQueen's University BelfastBelfastUK
| | - Philip D Dunne
- Centre for Cancer Research and Cell BiologyQueen's University BelfastBelfastUK
| | - Anna M Kearney
- Centre for Experimental Medicine, Wellcome‐Wolfson BuildingQueen's University BelfastBelfastUK
| | - David J Grieve
- Centre for Experimental Medicine, Wellcome‐Wolfson BuildingQueen's University BelfastBelfastUK
| | - Barbara J McDermott
- Centre for Experimental Medicine, Wellcome‐Wolfson BuildingQueen's University BelfastBelfastUK
| |
Collapse
|
21
|
Chen HY, Yang FY, Jong GP, Liou YS. Antihyperglycemic drugs use and new-onset atrial fibrillation in elderly patients. Eur J Clin Invest 2017; 47:388-393. [PMID: 28369870 DOI: 10.1111/eci.12754] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Accepted: 03/27/2017] [Indexed: 01/23/2023]
Abstract
BACKGROUND Antihyperglycemic drugs have been linked to new-onset atrial fibrillation (NAF). However, the effect of the different classes of antihyperglycemic drugs on the development of NAF in elderly patients has not been well studied. In this study, we investigated the association between different classes of antihyperglycemic drugs and NAF in elderly patients. MATERIALS AND METHODS This was a nested case-control study performed using the database of National Health Insurance programme in Taiwan. Each participant aged 65 years and older who were NAF from 2005 to 2012 were assigned to the NAF group, whereas case was sex-, age-, diabetes duration-, index date-matched, and Charlson Comorbidity Index score-matched randomly selected participant without NAF were assigned to the non-NAF group. Multivariable logistic regression model was used for the estimation of odds ratios (ORs) and 95% confidence intervals (CIs) of NAF associated with use of different classes of antihyperglycemic agents. Nonusers served as the reference group. RESULTS We identified 1958 cases and 7832 controls. The risk of NAF after adjusting for sex, age, comorbidities and concurrent medication was higher among the users of insulin than among the nonusers (OR, 1·58; 95% CI, 1·37-1·82). Patients who took dipeptidyl peptidase 4 inhibitors were at lower risk of developing NAF than the nonusers (OR, 0·65; 95% CI, 0·45-0·93). CONCLUSIONS In this population, use of dipeptidyl peptidase 4 inhibitor was associated with a low risk of NAF. Insulin use was associated with a significant increase in the risk of NAF during the long-term follow-up.
Collapse
Affiliation(s)
- Hung-Yi Chen
- Institute of Pharmacy, China Medical University, Taichung, Taiwan
- Department of Pharmacy, China Medical University Beigang Hospital, Beigang, Taiwan
| | - Fu-Yu Yang
- Institute of Pharmacy, China Medical University, Taichung, Taiwan
- Department of Pharmacy, China Medical University Beigang Hospital, Beigang, Taiwan
| | - Gwo-Ping Jong
- Division of Internal Cardiology, Chung Shan Medical University Hospital and Chung Shan Medical University, Taichung, Taiwan
- Basic Science, Central Taiwan University of Science and Technology, Taichung, Taiwan
| | - Yi-Sheng Liou
- Department of Family Medicine and Geriatrics, Taichung Veteran General Hospital, Taichung, Taiwan
- School of Public Health, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
22
|
Association evidence of CCTTT repeat polymorphism in the iNOS promoter and the risk of atrial fibrillation in Taiwanese. Sci Rep 2017; 7:42388. [PMID: 28205526 PMCID: PMC5304328 DOI: 10.1038/srep42388] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 01/09/2017] [Indexed: 11/17/2022] Open
Abstract
Inducible nitric oxide synthase (iNOS) plays an important role in the pathogenesis of atrial fibrillation (AF). The iNOS promoter has a CCTTT-repeat length polymorphism that can determine the level of gene transcription. This study enrolled 200 AF patients and 240 controls. The length of CCTTT-repeat polymorphism in the iNOS promoter region was examined by polymerase chain reactions, with the alleles with ≤11 repeats designated as S and alleles with ≥12 repeats designated as L alleles. AF patients carried significantly higher frequencies of the LL genotype than control subjects (40.0% versus 28.3%, P = 0.010). Multivariate analysis showed that the presence of LL genotype was significantly associated with AF (odds ratio: 1.87, 95% CI = 1.10–3.17, P = 0.021). In vitro, transient transfection assay in HL-1 atrial myocytes showed that the responsiveness of iNOS transcriptional activity to tachypacing was correlated with the length of the CCTTT-repeats. Right atrial tissues from patients with chronic AF were investigated with immunoconfocal microscopy. Patients with LL genotype exhibited greater oxidative stress and substrate remodeling in their atria than those with non-LL genotypes. Our results suggest that the iNOS microsatellite polymorphism may contribute to the genetic background of AF in Chinese-Taiwanese patients.
Collapse
|
23
|
Abstract
Cardiac arrhythmias can follow disruption of the normal cellular electrophysiological processes underlying excitable activity and their tissue propagation as coherent wavefronts from the primary sinoatrial node pacemaker, through the atria, conducting structures and ventricular myocardium. These physiological events are driven by interacting, voltage-dependent, processes of activation, inactivation, and recovery in the ion channels present in cardiomyocyte membranes. Generation and conduction of these events are further modulated by intracellular Ca2+ homeostasis, and metabolic and structural change. This review describes experimental studies on murine models for known clinical arrhythmic conditions in which these mechanisms were modified by genetic, physiological, or pharmacological manipulation. These exemplars yielded molecular, physiological, and structural phenotypes often directly translatable to their corresponding clinical conditions, which could be investigated at the molecular, cellular, tissue, organ, and whole animal levels. Arrhythmogenesis could be explored during normal pacing activity, regular stimulation, following imposed extra-stimuli, or during progressively incremented steady pacing frequencies. Arrhythmic substrate was identified with temporal and spatial functional heterogeneities predisposing to reentrant excitation phenomena. These could arise from abnormalities in cardiac pacing function, tissue electrical connectivity, and cellular excitation and recovery. Triggering events during or following recovery from action potential excitation could thereby lead to sustained arrhythmia. These surface membrane processes were modified by alterations in cellular Ca2+ homeostasis and energetics, as well as cellular and tissue structural change. Study of murine systems thus offers major insights into both our understanding of normal cardiac activity and its propagation, and their relationship to mechanisms generating clinical arrhythmias.
Collapse
Affiliation(s)
- Christopher L-H Huang
- Physiological Laboratory and the Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
24
|
Gasparova I, Kubatka P, Opatrilova R, Caprnda M, Filipova S, Rodrigo L, Malan L, Mozos I, Rabajdova M, Nosal V, Kobyliak N, Valentova V, Petrovic D, Adamek M, Kruzliak P. Perspectives and challenges of antioxidant therapy for atrial fibrillation. Naunyn Schmiedebergs Arch Pharmacol 2016; 390:1-14. [PMID: 27900409 DOI: 10.1007/s00210-016-1320-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 11/18/2016] [Indexed: 12/26/2022]
Abstract
Atrial fibrillation (AF) is the most common sustained arrhythmia associated with significant morbidity and mortality. The mechanisms underlying the pathogenesis of AF are poorly understood, although electrophysiological remodeling has been described as an important initiating step. There is growing evidence that oxidative stress is involved in the pathogenesis of AF. Many known triggers of oxidative stress, such as age, diabetes, smoking, and inflammation, are linked with an increased risk of arrhythmia. Numerous preclinical studies and clinical trials reported the importance of antioxidant therapy in the prevention of AF, using vitamins C and E, polyunsaturated fatty acids, statins, or nitric oxide donors. The aim of our work is to give a current overview and analysis of opportunities, challenges, and benefits of antioxidant therapy in AF.
Collapse
Affiliation(s)
- Iveta Gasparova
- Institute of Biology, Genetics and Medical Genetics, Faculty of Medicine, Comenius University and University Hospital, Bratislava, Slovak Republic, Slovakia
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovak Republic, Slovakia
| | - Radka Opatrilova
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, Brno, Czech Republic
| | - Martin Caprnda
- 2nd Department of Internal Medicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Slavomira Filipova
- Department of Cardiology, National Institute of Cardiovascular Diseases, Bratislava, Slovakia
| | - Luis Rodrigo
- Faculty of Medicine, University of Oviedo, Central University of Asturias (HUCA), Oviedo, Spain
| | - Leone Malan
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom Campus, Potchefstroom, South Africa
| | - Ioana Mozos
- Department of Functional Sciences, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Miroslava Rabajdova
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Safarik University, Kosice, Slovakia
| | - Vladimir Nosal
- Clinic of Neurology, Jessenius Faculty of Medicine, Comenius University and University Hospital in Martin, Martin, Slovak Republic
| | - Nazarii Kobyliak
- Department of Endocrinology, Bogomolets National Medical University, Kyiv, Ukraine
| | - Vanda Valentova
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovak Republic, Slovakia
| | - Daniel Petrovic
- Institute of Histology and Embryology, Faculty of Medicine, University of Ljublana, Ljublana, Slovenia
| | - Mariusz Adamek
- Department of Thoracic Surgery, Medical University of Silesia, Zabrze, Poland
| | - Peter Kruzliak
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, Brno, Czech Republic. .,2nd Department of Surgery, Faculty of Medicine, Masaryk University, Brno, Czech Republic.
| |
Collapse
|
25
|
Qiu J, Zhao J, Li J, Liang X, Yang Y, Zhang Z, Zhang X, Fu H, Korantzopoulos P, Liu T, Li G. NADPH oxidase inhibitor apocynin prevents atrial remodeling in alloxan-induced diabetic rabbits. Int J Cardiol 2016; 221:812-819. [PMID: 27434350 DOI: 10.1016/j.ijcard.2016.07.132] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 07/08/2016] [Indexed: 11/27/2022]
Abstract
OBJECTIVES Diabetes mellitus (DM) is an independent risk factor for atrial fibrillation (AF). The role of the NADPH oxidase (NOX) signaling in the setting of DM and the potential benefits of apocynin on diabetic atrial remodeling remain unknown. METHODS Sixty Japanese rabbits were randomized into 3 groups as follows: Control group (Control, n=20), alloxan-induced diabetic group (DM, n=20) and apocynin-treated diabetic group (APO, n=20). Rabbits in the APO group were orally administered apocynin (15mg/kg/day) for 8weeks. Serum malonaldehyde (MDA), superoxide dismutase (SOD) levels, and left atrial tissue NADPH oxidase (NOX) activities were measured. Isolated rabbit hearts were Langendorff perfused. Atrial refractory effective period (AERP), atrial effective refractory period dispersion (AERPD), interatrial conduction time (IACT) and vulnerability to AF were assessed. Atrial interstitial fibrosis was evaluated by Masson's trichrome staining. The protein expression of NF-κB, TGF-β, p38, P-p38, JNK, P-JNK, ERK and P-ERK was measured by Western blot analysis. RESULTS There were no significant differences regarding SBP, DBP, LVEDP and AERP in the three groups. Compared with the Control group, AF inducibility was increased in the DM group (46/450 vs. 5/450, P<0.05), and markedly reduced by apocynin (46/450 vs. 12/450, P<0.05). Apocynin also attenuated atrial structural remodeling in diabetic rabbits. Western-blot analysis indicated that apocynin reduced the DM-induced increased protein expression of TGF-β, NF-κB, P-p38, P-JNK, ERK and P-ERK. CONCLUSIONS Apocynin, a NADPH oxidase inhibitor, prevents AF and attenuates atrial remodeling in alloxan-induced diabetic rabbits.
Collapse
Affiliation(s)
- Jiuchun Qiu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, People's Republic of China
| | - Jianping Zhao
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, People's Republic of China
| | - Jian Li
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, People's Republic of China
| | - Xue Liang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, People's Republic of China
| | - Yajuan Yang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, People's Republic of China
| | - Zhiwei Zhang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, People's Republic of China
| | - Xiaowei Zhang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, People's Republic of China
| | - Huaying Fu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, People's Republic of China
| | | | - Tong Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, People's Republic of China.
| | - Guangping Li
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, People's Republic of China.
| |
Collapse
|
26
|
Abstract
Atrial fibrillation (AF) is an important cause of stroke and risk factor for heart failure and death. Current pharmacologic treatments for AF have limited efficacy, and treatments that more directly target the underlying causes of AF are needed. Oxidant stress and inflammatory activation are interrelated pathways that promote atrial electrical and structural remodeling, leading to atrial ectopy, interstitial fibrosis, and increased stroke risk. This review evaluates the impact of common stressors on atrial oxidant stress and inflammatory activation and the contribution of these pathways to atrial remodeling. Recent studies suggest that integrated efforts to target the underlying risk factors, rather than the AF per se, may have a greater impact on health and outcomes than isolated efforts focused on the electrical abnormalities.
Collapse
|
27
|
Protective role of heme oxygenase-1 in atrial remodeling. Basic Res Cardiol 2016; 111:58. [DOI: 10.1007/s00395-016-0577-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 08/22/2016] [Indexed: 12/26/2022]
|
28
|
Zhao Z, Wang Y, Chen Y, Wang X, Li J, Yuan M, Liu T, Li G. Cilostazol Prevents Atrial Structural Remodeling through the MEK/ERK Pathway in a Canine Model of Atrial Tachycardia. Cardiology 2016; 135:240-248. [PMID: 27532517 DOI: 10.1159/000447769] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 06/20/2016] [Indexed: 10/11/2023]
Abstract
OBJECTIVES Atrial fibrillation (AF) is the most common sustained arrhythmia in clinical practice. Atrial structural remodeling (ASR), particularly atrial fibrosis, is an important contributor to the AF substrate. This study aimed to investigate the preventive effects of the phosphodiesterase 3 inhibitor cilostazol on ASR and its potential molecular mechanisms in a canine model of rapid atrial pacing (RAP). METHODS Thirty dogs were assigned to sham (Sham), paced/ no treatment (Paced) and paced + cilostazol 5 mg/kg/day (Paced + cilo) groups, with 10 dogs in each group. RAP at 500 beats/min was maintained for 2 weeks, while the Sham group was instrumented without pacing. Cilostazol was provided orally during pacing. Western blotting, RT-PCR and pathology were used to assess ASR. RESULTS Cilostazol attenuated atrial interstitial fibrosis and structural remodeling in canines with RAP. MEK/ERK transduction pathway gene expression was upregulated in the Paced group compared with the Sham group. Cilostazol markedly alleviated these changes in the MEK/ERK pathway. Transforming growth factor-β1 protein expression in the Paced group was significantly higher than in the Sham group (p < 0.01), and was significantly reduced by cilostazol (p < 0.01). CONCLUSIONS Our findings suggest that cilostazol is beneficial for prevention and treatment in atrial tachycardia-induced ASR in a canine model of RAP.
Collapse
Affiliation(s)
- Zhiqiang Zhao
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Tsai FC, Lin YC, Chang SH, Chang GJ, Hsu YJ, Lin YM, Lee YS, Wang CL, Yeh YH. Differential left-to-right atria gene expression ratio in human sinus rhythm and atrial fibrillation: Implications for arrhythmogenesis and thrombogenesis. Int J Cardiol 2016; 222:104-112. [PMID: 27494721 DOI: 10.1016/j.ijcard.2016.07.103] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 07/05/2016] [Accepted: 07/07/2016] [Indexed: 12/26/2022]
Abstract
BACKGROUND Atrial fibrillation (AF) causes atrial remodeling, and the left atrium (LA) is the favored substrate for maintaining AF. It remains unclear if AF remodels both atria differently and contributes to LA arrhythmogenesis and thrombogenesis. Therefore, we wished to characterize the transcript profiles in the LA and right atrium (RA) in sinus rhythm (SR) and AF respectively. METHODS Paired LA and RA appendages acquired from patients receiving cardiac surgery were used for ion-channel- and whole-exome-based transcriptome analysis. The ultrastructure was evaluated by immunohistochemistry. RESULTS Twenty-two and twenty ion-channels and transporters were differentially expressed between the LA and RA in AF and SR, respectively. Among these, 15 genes were differentially expressed in parallel between AF and SR. AF was associated with increased LA/RA expression ratio in 9 ion channel-related genes, including genes related to calcium handling. In microarray, AF was associated with a differential LA/RA gene expression ratio in 309 genes, and was involved in atherosclerosis-related signaling. AF was associated with the upregulation of thrombogenesis-related genes in the LA appendage, including P2Y12, CD 36 and ApoE. Immunohistochemistry showed higher expressions of collagen-1, oxidative stress and TGF-β1 in the RA compared to the LA. CONCLUSIONS AF was associated with differential LA-to-RA gene expression related to specific ion channels and pathways as well as upregulation of thrombogenesis-related genes in the LA appendage. Targeting the molecular mechanisms underlying the LA-to-RA difference and AF-related remodeling in the LA appendage may help provide new therapeutic options in treating AF and preventing thromboembolism in AF.
Collapse
Affiliation(s)
- Feng-Chun Tsai
- Division of Cardiac Surgery, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, Taiwan
| | - Yen-Chen Lin
- Cardiovascular Division, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, Taiwan
| | - Shang-Hung Chang
- Cardiovascular Division, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, Taiwan
| | - Gwo-Jyh Chang
- Graduate Institute of Clinical Medical Sciences, Chang-Gung University College of Medicine, Chang-Gung University, Taiwan
| | - Yu-Juei Hsu
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yuan-Min Lin
- School of Dentistry, National Yang-Ming University, Taipei, Taiwan
| | - Yun-Shien Lee
- Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan
| | - Chun-Li Wang
- Cardiovascular Division, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, Taiwan
| | - Yung-Hsin Yeh
- Cardiovascular Division, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, Taiwan.
| |
Collapse
|
30
|
Abstract
Reactive oxygen species (ROS) and oxidative stress have long been linked to aging and diseases prominent in the elderly such as hypertension, atherosclerosis, diabetes and atrial fibrillation (AF). NADPH oxidases (Nox) are a major source of ROS in the vasculature and are key players in mediating redox signalling under physiological and pathophysiological conditions. In this review, we focus on the Nox-mediated ROS signalling pathways involved in the regulation of 'longevity genes' and recapitulate their role in age-associated vascular changes and in the development of age-related cardiovascular diseases (CVDs). This review is predicated on burgeoning knowledge that Nox-derived ROS propagate tightly regulated yet varied signalling pathways, which, at the cellular level, may lead to diminished repair, the aging process and predisposition to CVDs. In addition, we briefly describe emerging Nox therapies and their potential in improving the health of the elderly population.
Collapse
|
31
|
Kunamalla A, Ng J, Parini V, Yoo S, McGee KA, Tomson TT, Gordon D, Thorp EB, Lomasney J, Zhang Q, Shah S, Browne S, Knight BP, Passman R, Goldberger JJ, Aistrup G, Arora R. Constitutive Expression of a Dominant-Negative TGF-β Type II Receptor in the Posterior Left Atrium Leads to Beneficial Remodeling of Atrial Fibrillation Substrate. Circ Res 2016; 119:69-82. [PMID: 27217399 DOI: 10.1161/circresaha.115.307878] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 05/23/2016] [Indexed: 11/16/2022]
Abstract
RATIONALE Fibrosis is an important structural contributor to formation of atrial fibrillation (AF) substrate in heart failure. Transforming growth factor-β (TGF-β) signaling is thought to be intricately involved in creation of atrial fibrosis. OBJECTIVE We hypothesized that gene-based expression of dominant-negative type II TGF-β receptor (TGF-β-RII-DN) in the posterior left atrium in a canine heart failure model will sufficiently attenuate fibrosis-induced changes in atrial conduction and restitution to decrease AF. Because AF electrograms are thought to reflect AF substrate, we further hypothesized that TGF-β-RII-DN would lead to increased fractionation and decreased organization of AF electrograms. METHODS AND RESULTS Twenty-one dogs underwent injection+electroporation in the posterior left atrium of plasmid expressing a dominant-negative TGF-β type II receptor (pUBc-TGFβ-DN-RII; n=9) or control vector (pUBc-LacZ; n=12), followed by 3 to 4 weeks of right ventricular tachypacing (240 bpm). Compared with controls, dogs treated with pUBC-TGFβ-DN-RII demonstrated an attenuated increase in conduction inhomogeneity, flattening of restitution slope and decreased duration of induced AF, with AF electrograms being more fractionated and less organized in pUBc-TGFβ-DN-RII versus pUBc-LacZ dogs. Tissue analysis revealed a significant decrease in replacement/interstitial fibrosis, p-SMAD2/3 and p-ERK1/2. CONCLUSIONS Targeted gene-based reduction of TGF-β signaling in the posterior left atrium-with resulting decrease in replacement fibrosis-led to beneficial remodeling of both conduction and restitution characteristics of the posterior left atrium, translating into a decrease in AF and increased complexity of AF electrograms. In addition to providing mechanistic insights, this data may have important diagnostic and therapeutic implications for AF.
Collapse
Affiliation(s)
- Aaron Kunamalla
- From the Feinberg Cardiovascular Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Jason Ng
- From the Feinberg Cardiovascular Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Vamsi Parini
- From the Feinberg Cardiovascular Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Shin Yoo
- From the Feinberg Cardiovascular Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Kate A McGee
- From the Feinberg Cardiovascular Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Todd T Tomson
- From the Feinberg Cardiovascular Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - David Gordon
- From the Feinberg Cardiovascular Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Edward B Thorp
- From the Feinberg Cardiovascular Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Jon Lomasney
- From the Feinberg Cardiovascular Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Qiang Zhang
- From the Feinberg Cardiovascular Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Sanjiv Shah
- From the Feinberg Cardiovascular Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Suzanne Browne
- From the Feinberg Cardiovascular Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Bradley P Knight
- From the Feinberg Cardiovascular Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Rod Passman
- From the Feinberg Cardiovascular Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Jeffrey J Goldberger
- From the Feinberg Cardiovascular Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Gary Aistrup
- From the Feinberg Cardiovascular Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Rishi Arora
- From the Feinberg Cardiovascular Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL.
| |
Collapse
|
32
|
Liu XH, Zhang QY, Pan LL, Liu SY, Xu P, Luo XL, Zou SL, Xin H, Qu LF, Zhu YZ. NADPH oxidase 4 contributes to connective tissue growth factor expression through Smad3-dependent signaling pathway. Free Radic Biol Med 2016; 94:174-84. [PMID: 26945889 DOI: 10.1016/j.freeradbiomed.2016.02.031] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 02/21/2016] [Accepted: 02/28/2016] [Indexed: 01/28/2023]
Abstract
Transforming growth factor-β (TGF-β)/Smad signaling has been implicated in connective tissue growth factor (CTGF) expression in vascular smooth muscle cells (VSMC). Reactive oxygen species (ROS) are involved in activation of TGF-β/Smad signaling. However, detailed mechanisms underlying the process remain unclear. In present study, we demonstrated TGF-β1 strongly induced CTGF expression, Smad3 activation, NADPH oxidase 4 (Nox4) expression and increased ROS production in primary rat VSMC in vitro. NADPH oxidases inhibitor diphenylene iodonium (DPI) eliminated TGF-β1-induced CTGF expression and ROS generation. In addition, small-interfering RNA (siRNA) silencing of Smad3 or Nox4 significantly suppressed TGF-β1-mediated CTGF expression in VSMC. Furthermore, Nox4 silencing or inhibition eliminated TGF-β1-induced Smad3 activation and interaction between Nox4 and Smad3. In vivo studies further identified a positive correlation of Nox4 levels with Smad3 activation and CTGF expression in atherosclerotic arteries of patients and animal models. These data established that a novel mechanistic link of Nox4-dependent activation of Smad3 to increased TGF-β1-induced CTGF in the process of vascular remodeling, which suggested a new potential pathway for therapeutic interventions.
Collapse
Affiliation(s)
- Xin-Hua Liu
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, 826, Zhangheng Road, Pudong New District, Shanghai 201203, China
| | - Qiu-Yan Zhang
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, 826, Zhangheng Road, Pudong New District, Shanghai 201203, China
| | - Li-Long Pan
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, 826, Zhangheng Road, Pudong New District, Shanghai 201203, China
| | - Si-Yu Liu
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, 826, Zhangheng Road, Pudong New District, Shanghai 201203, China
| | - Peng Xu
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, 826, Zhangheng Road, Pudong New District, Shanghai 201203, China
| | - Xiao-Ling Luo
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, 826, Zhangheng Road, Pudong New District, Shanghai 201203, China
| | - Si-Li Zou
- Department of Vascular Surgery, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai 200003, China
| | - Hong Xin
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, 826, Zhangheng Road, Pudong New District, Shanghai 201203, China
| | - Le-Feng Qu
- Department of Vascular Surgery, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai 200003, China.
| | - Yi-Zhun Zhu
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, 826, Zhangheng Road, Pudong New District, Shanghai 201203, China; School of Pharmacy, Macau University of Science and Technology, Macau.
| |
Collapse
|
33
|
Hsiao FC, Yeh YH, Chen WJ, Chan YH, Kuo CT, Wang CL, Chang CJ, Tsai HY, Tsai FC, Hsu LA. MMP9 Rs3918242 Polymorphism Affects Tachycardia-Induced MMP9 Expression in Cultured Atrial-Derived Myocytes but Is Not a Risk Factor for Atrial Fibrillation among the Taiwanese. Int J Mol Sci 2016; 17:521. [PMID: 27070579 PMCID: PMC4848977 DOI: 10.3390/ijms17040521] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Revised: 03/23/2016] [Accepted: 03/29/2016] [Indexed: 12/19/2022] Open
Abstract
Matrix metalloproteinase (MMP) plays an important role in the pathogenesis of atrial fibrillation (AF). The MMP9 promoter has a functional polymorphism rs3918242 that can regulate the level of gene transcription. This study recruited 200 AF patients and 240 controls. The MMP9 rs3918242 was examined by polymerase chain reactions. HL-1 atrial myocytes were cultured and electrically stimulated. Right atrial appendages were obtained from six patients with AF and three controls with sinus rhythm undergoing open heart surgery. The MMP9 expression and activity were determined using immunohistochemical analysis and gelatin zymography, respectively. Rapid pacing induces MMP9 secretion from HL-1 myocytes in a time- and dose-dependent manner. The responsiveness of MMP9 transcriptional activity to tachypacing was significantly enhanced by rs3918242. The expression of MMP9 was increased in fibrillating atrial tissue than in sinus rhythm. However, the distribution of rs3918242 genotypes and allele frequencies did not significantly differ between the control and AF groups. HL-1 myocyte may secrete MMP9 in response to rapid pacing, and the secretion could be modulated by rs3918242. Although the MMP9 expression of human atrial myocyte is associated with AF, our study did not support the association of susceptibility to AF among Taiwanese subjects with the MMP9 rs3918242 polymorphism.
Collapse
Affiliation(s)
- Fu-Chih Hsiao
- Cardiovascular Division, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, No. 5, Fu-Shin Road, Kwei-Shan, Taoyuan 33305, Taiwan.
| | - Yung-Hsin Yeh
- Cardiovascular Division, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, No. 5, Fu-Shin Road, Kwei-Shan, Taoyuan 33305, Taiwan.
| | - Wei-Jan Chen
- Cardiovascular Division, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, No. 5, Fu-Shin Road, Kwei-Shan, Taoyuan 33305, Taiwan.
| | - Yi-Hsin Chan
- Cardiovascular Division, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, No. 5, Fu-Shin Road, Kwei-Shan, Taoyuan 33305, Taiwan.
| | - Chi-Tai Kuo
- Cardiovascular Division, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, No. 5, Fu-Shin Road, Kwei-Shan, Taoyuan 33305, Taiwan.
| | - Chun-Li Wang
- Cardiovascular Division, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, No. 5, Fu-Shin Road, Kwei-Shan, Taoyuan 33305, Taiwan.
| | - Chi-Jen Chang
- Cardiovascular Division, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, No. 5, Fu-Shin Road, Kwei-Shan, Taoyuan 33305, Taiwan.
| | - Hsin-Yi Tsai
- Cardiovascular Division, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, No. 5, Fu-Shin Road, Kwei-Shan, Taoyuan 33305, Taiwan.
| | - Feng-Chun Tsai
- Division of Cardiac Surgery, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 33305, Taiwan.
| | - Lung-An Hsu
- Cardiovascular Division, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, No. 5, Fu-Shin Road, Kwei-Shan, Taoyuan 33305, Taiwan.
| |
Collapse
|
34
|
Tsai FC, Chang GJ, Hsu YJ, Lin YM, Lee YS, Chen WJ, Kuo CT, Yeh YH. Proinflammatory gene expression in patients undergoing mitral valve surgery and maze ablation for atrial fibrillation. J Thorac Cardiovasc Surg 2015; 151:1673-1682.e5. [PMID: 26774166 DOI: 10.1016/j.jtcvs.2015.12.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Revised: 11/27/2015] [Accepted: 12/05/2015] [Indexed: 12/12/2022]
Abstract
OBJECTIVE It is difficult to achieve rhythm control in patients with long-standing persistent atrial fibrillation (AF). The radiofrequency maze procedure is an effective means in curing AF with a variable recurrence rate depending on patient characteristics and AF duration. In these patients, the characteristics of the atrial substrate have not been well investigated. Because the inflammatory process has been shown to be important in the pathogenesis of AF, we sought to characterize the proinflammatory gene expression in left atria obtained from patients with AF undergoing mitral valve surgery combined with the maze procedure to distinguish the changes associated with AF and its recurrence after the surgical ablation. METHODS Left atrial appendages from 35 patients receiving mitral valve surgery were used for study. Ten patients had sinus rhythm (SR) and 25 patients had persistent AF for more than 1 year and underwent the maze procedure. Among the AF patients, 13 patients remained in SR (AF-SR) and 12 patients had recurrent AF during the 1-year clinical follow-up (AF-AF). The nCounter Human Inflammation Array (NanoString Technologies, Seattle, Wash) was used for evaluating proinflammatory gene expression. Quantitative polymerase chain reaction, Western blot, and immunohistochemistry were applied for studying messenger RNA and protein expression. RESULTS Of 144 expressed proinflammatory genes, the inflammation array analysis revealed that 32 genes were differentially expressed between AF (including AF-SR and AF-AF) and SR. Thirteen genes were differentially expressed between AF-SR and AF-AF. The array and quantitative polymerase chain reaction produced parallel results in analyzing the expression of particular genes. Concordant with the gene expression difference between AF and SR patients, rapid pacing increased the expressions of SHC1, RHOA, PDGFA, and TRAF2 in HL-1 myocytes, implicating a causative effect of tachyarrhythmia on these genes. Compared with AF-SR, AF-AF expressed more intense oxidative stress, upregulations of collagen, transforming growth factor beta 1, and intranuclear nuclear factor of activated T-cells. Regression analysis showed that increased left atrial diameter was associated with the expression of RHOA and STAT1. CONCLUSIONS Differential expression profiles of proflammatory genes were presented between SR and AF and between maintained SR and recurrent AF after the maze procedure. The identified inflammatory molecules associated with AF and failed surgical ablation may provide clues for developing new potential therapeutic targets to improve AF rhythm control.
Collapse
Affiliation(s)
- Feng-Chun Tsai
- Division of Cardiac Surgery, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, Taiwan
| | - Gwo-Jyh Chang
- Graduate Institute of Clinical Medical Sciences, Chang-Gung University College of Medicine, Taoyuan, Taiwan
| | - Yu-Juei Hsu
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yuan-Min Lin
- School of Dentistry, National Yang-Ming University, Taipei, Taiwan
| | - Yun-Shien Lee
- Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan
| | - Wei-Jan Chen
- Cardiovascular Division, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, Taiwan
| | - Chi-Tai Kuo
- Cardiovascular Division, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, Taiwan
| | - Yung-Hsin Yeh
- Cardiovascular Division, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, Taiwan.
| |
Collapse
|
35
|
Irregular rhythm and atrial metabolism are key for the evolution of proarrhythmic atrial remodeling in atrial fibrillation. Basic Res Cardiol 2015; 110:41. [PMID: 26018792 DOI: 10.1007/s00395-015-0498-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 05/22/2015] [Indexed: 12/18/2022]
|
36
|
Rosuvastatin suppresses atrial tachycardia-induced cellular remodeling via Akt/Nrf2/heme oxygenase-1 pathway. J Mol Cell Cardiol 2015; 82:84-92. [DOI: 10.1016/j.yjmcc.2015.03.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 03/02/2015] [Accepted: 03/03/2015] [Indexed: 12/12/2022]
|
37
|
Oxidative stress-associated senescence in dermal papilla cells of men with androgenetic alopecia. J Invest Dermatol 2015; 135:1244-1252. [PMID: 25647436 DOI: 10.1038/jid.2015.28] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 12/12/2014] [Accepted: 12/22/2014] [Indexed: 12/19/2022]
Abstract
Dermal papilla cells (DPCs) taken from male androgenetic alopecia (AGA) patients undergo premature senescence in vitro in association with the expression of p16(INK4a), suggesting that DPCs from balding scalp are more sensitive to environmental stress than nonbalding cells. As one of the major triggers of senescence in vitro stems from the cell "culture shock" owing to oxidative stress, we have further investigated the effects of oxidative stress on balding and occipital scalp DPCs. Patient-matched DPCs from balding and occipital scalp were cultured at atmospheric (21%) or physiologically normal (2%) O2. At 21% O2, DPCs showed flattened morphology and a significant reduction in mobility, population doubling, increased levels of reactive oxygen species and senescence-associated β-Gal activity, and increased expression of p16(INK4a) and pRB. Balding DPCs secreted higher levels of the negative hair growth regulators transforming growth factor beta 1 and 2 in response to H2O2 but not cell culture-associated oxidative stress. Balding DPCs had higher levels of catalase and total glutathione but appear to be less able to handle oxidative stress compared with occipital DPCs. These in vitro findings suggest that there may be a role for oxidative stress in the pathogenesis of AGA both in relation to cell senescence and migration but also secretion of known hair follicle inhibitory factors.
Collapse
|
38
|
Identification of microRNA-mRNA dysregulations in paroxysmal atrial fibrillation. Int J Cardiol 2015; 184:190-197. [PMID: 25706326 DOI: 10.1016/j.ijcard.2015.01.075] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 12/08/2014] [Accepted: 01/26/2015] [Indexed: 01/09/2023]
Abstract
BACKGROUND The molecular mechanisms underlying the early development of atrial fibrillation (AF) remain poorly understood. Emerging evidence suggests that abnormal epigenetic modulation via microRNAs (miRNAs) might be involved in the pathogenesis of paroxysmal AF (pAF). OBJECTIVE To identify key molecular changes associated with pAF, we conducted state-of-the-art transcriptomic studies to identify the abnormal miRNA-mRNA interactions potentially driving AF development. METHODS High-quality total RNA including miRNA was isolated from atrial biopsies of age-matched and sex-matched pAF patients and control patients in sinus rhythm (SR; n=4 per group) and used for RNA-sequencing and miRNA microarray. Results were analyzed bioinformatically and validated using quantitative real-time (qRT)-PCR and 3'UTR luciferase reporter assays. RESULTS 113 genes and 49 miRNAs were differentially expressed (DE) in pAF versus SR patients. Gene ontology analysis revealed that most of the DE genes were involved in the "gonadotropin releasing hormone receptor pathway" and "p53 pathway". Of these DE genes, bioinformatic analyses identified 23 pairs of putative miRNA-mRNA interactions that were altered in pAF (involving 15 miRNAs and 17 mRNAs). Using qRT-PCR and 3'UTR luciferase reporter assays, the interaction between upregulation of miR-199a-5p and downregulation of FKBP5 was confirmed in samples from pAF patients. CONCLUSION Our combined transcriptomic analysis and miRNA microarray study of atrial samples from pAF patients revealed novel pathways and miRNA-mRNA regulations that may be relevant in the development of pAF. Future studies are required to investigate the potential involvement of the gonadotropin releasing hormone receptor and p53 pathways in AF pathogenesis.
Collapse
|
39
|
He Z, Zhang X, Chen C, Wen Z, Hoopes SL, Zeldin DC, Wang DW. Cardiomyocyte-specific expression of CYP2J2 prevents development of cardiac remodelling induced by angiotensin II. Cardiovasc Res 2015; 105:304-17. [PMID: 25618409 DOI: 10.1093/cvr/cvv018] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AIMS Cardiac remodelling is one of the key pathological changes that occur with cardiovascular disease. Previous studies have demonstrated the beneficial effects of CYP2J2 expression on cardiac injury. In the present study, we investigated the effects of cardiomyocyte-specific CYP2J2 expression and EET treatment on angiotensin II-induced cardiac remodelling and sought to determine the underlying molecular mechanisms involved in this process. METHODS AND RESULTS Eight-week-old mice with cardiomyocyte-specific CYP2J2 expression (αMHC-CYP2J2-Tr) and wild-type (WT) control mice were treated with Ang-II. Ang-II treatment of WT mice induced changes in heart morphology, cardiac hypertrophy and dysfunction, as well as collagen accumulation; however, cardiomyocyte-specific expression of CYP2J2 attenuated these effects. The cardioprotective effects observed in α-MHC-CYP2J2-Tr mice were associated with peroxisome proliferator-activated receptor (PPAR)-γ activation, reduced oxidative stress, reduced NF-κB p65 nuclear translocation, and inhibition of TGF-β1/smad pathway. The effects seen with cardiomyocyte-specific expression of CYP2J2 were partially blocked by treatment with PPAR-γ antagonist GW9662. In in vitro studies, 11,12-EET(1 μmol/L) treatment attenuated cardiomyocyte hypertrophy and remodelling-related protein (collagen I, TGF-β1, TIMP1) expression by inhibiting the oxidative stress-mediated NF-κB pathway via PPAR-γ activation. Furthermore, conditioned media from neonatal cardiomyocytes treated with 11,12-EET inhibited activation of cardiac fibroblasts and TGF-β1/smad pathway. CONCLUSION Cardiomyocyte-specific expression of CYP2J2 or treatment with EETs protects against cardiac remodelling by attenuating oxidative stress-mediated NF-κBp65 nuclear translocation via PPAR-γ activation.
Collapse
Affiliation(s)
- Zuowen He
- Department of Internal Medicine and Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Avenue, Wuhan 430030, P. R. China
| | - Xu Zhang
- Department of Physiology, Tianjin Medical University, Tianjin, P. R. China
| | - Chen Chen
- Department of Internal Medicine and Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Avenue, Wuhan 430030, P. R. China
| | - Zheng Wen
- Department of Internal Medicine and Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Avenue, Wuhan 430030, P. R. China
| | - Samantha L Hoopes
- Division of Intramural Research, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, USA
| | - Darryl C Zeldin
- Division of Intramural Research, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, USA
| | - Dao Wen Wang
- Department of Internal Medicine and Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Avenue, Wuhan 430030, P. R. China
| |
Collapse
|
40
|
Lu G, Xu S, Peng L, Huang Z, Wang Y, Gao X. Angiotensin II upregulates Kv1.5 expression through ROS-dependent transforming growth factor-beta1 and extracellular signal-regulated kinase 1/2 signalings in neonatal rat atrial myocytes. Biochem Biophys Res Commun 2014; 454:410-6. [DOI: 10.1016/j.bbrc.2014.10.088] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Accepted: 10/18/2014] [Indexed: 01/07/2023]
|
41
|
Hsu LA, Yeh YH, Kuo CT, Chen YH, Chang GJ, Tsai FC, Chen WJ. Microsatellite polymorphism in the heme oxygenase-1 gene promoter and the risk of atrial fibrillation in Taiwanese. PLoS One 2014; 9:e108773. [PMID: 25268359 PMCID: PMC4182563 DOI: 10.1371/journal.pone.0108773] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 08/25/2014] [Indexed: 11/18/2022] Open
Abstract
Background Atrial fibrillation (AF) is associated with increased oxidative stress. Emerging evidence suggests that heme oxygenase-1 (HO-1) is a potent antioxidant system against various oxidative stress-related diseases. The human HO-1 promoter has a GT-repeat length polymorphism that can determine the level of gene transcription. Objective The aim of this study is to assess the role of the GT-repeat polymorphism in the promoter region of the HO-1 gene in Chinese-Taiwanese patients with AF. Methods and Results This study enrolled 200 AF patients and 240 controls, comparable for age and gender. In each subject, the length of GT-repeat polymorphism in the HO-1 promoter region was examined by polymerase chain reactions. The frequencies of long GT-repeat alleles (≧32) were significantly higher in AF patients than in controls. Multivariate analysis showed that the presence of long allele was significantly and independently associated with AF (odds ratio: 1.91, 95% CI 1.07–3.72; P = 0.028). Right atrial tissues from patients with chronic AF were investigated with immunoconfocal microscopy. Patients homozygous for shorter GT-repeat alleles exhibited greater HO-1 expression in their atria than those homozygous for longer alleles, which was reflected by less oxidative stress, myofibril degradation, and fibrosis in the atria of patients with shorter GT-repeat. In vitro, transient transfection assay in HL-1 atrial myocytes showed that the responsiveness of HO-1 transcriptional activity to tachypacing was inversely correlated with the length of the GT-repeats. Conclusion Our results suggest that the HO-1 microsatellite polymorphism may contribute to the genetic background of AF in Chinese-Taiwanese patients.
Collapse
Affiliation(s)
- Lung-An Hsu
- Cardiovascular Division, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Tao-Yuan, Taiwan
| | - Yung-Hsin Yeh
- Cardiovascular Division, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Tao-Yuan, Taiwan
| | - Chi-Tai Kuo
- Cardiovascular Division, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Tao-Yuan, Taiwan
| | - Ying-Hwa Chen
- Division of Cardiology, Department of Internal Medicine, Taipei Veterans General Hospital, National Yang-Ming University College of Medicine, Taipei, Taiwan
- * E-mail: (YHC); (WJC)
| | - Gwo-Jyh Chang
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Tao-Yuan, Taiwan
| | - Feng-Chun Tsai
- Division of Cardiac Surgery, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Tao-Yuan, Taiwan
| | - Wei-Jan Chen
- Cardiovascular Division, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Tao-Yuan, Taiwan
- * E-mail: (YHC); (WJC)
| |
Collapse
|
42
|
Chang SH, Wu LS, Chiou MJ, Liu JR, Yu KH, Kuo CF, Wen MS, Chen WJ, Yeh YH, See LC. Association of metformin with lower atrial fibrillation risk among patients with type 2 diabetes mellitus: a population-based dynamic cohort and in vitro studies. Cardiovasc Diabetol 2014; 13:123. [PMID: 25106079 PMCID: PMC4149273 DOI: 10.1186/s12933-014-0123-x] [Citation(s) in RCA: 150] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Accepted: 08/04/2014] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Atrial fibrillation (AF), an inflammatory process involving arrhythmia, is associated with severe morbidity and mortality and commonly seen in patients with diabetes mellitus (DM). The effect of metformin, the most commonly used medication for patients with DM, on AF has not been investigated. The primary aim of this study was to examine whether metformin prevented the occurrence of AF in type 2 DM patients by analyzing a nationwide, population-based dynamic cohort. Additionally, we investigated the effect of metformin on tachycardia-induced myolysis and oxidative stress in atrial cells. METHODS The study population included 645,710 patients with type 2 diabetes and not using other anti-diabetic medication from a subset of the Taiwan National Health Insurance Research Database. Of these patients, those who used metformin were categorized as the user group, and the remaining were classified as the non-user group. The time-dependent Cox's proportional hazard model was used to examine the effect of metformin on AF and the status of metformin use was treated as a time-dependent covariate. HL-1 atrial cells were paced with or without metformin, and then troponin and heavy-chain-myosin were measured as markers of myolysis. RESULTS After 13 years of follow-up, 9,983 patients developed AF with an incidence rate of 1.5% (287 per 100,000 person-years). After adjusting for co-morbidities and medications, metformin independently protected the diabetic patients from new-onset AF with a hazard ratio of .81 (95% confidence interval 0.76-0.86, p < 0.0001). Metformin significantly decreased the extent of pacing-induced myolysis and the production of reactive oxygen species. CONCLUSION Metformin use was associated with a decreased risk of AF in patients with type 2 DM who were not using other anti-diabetic medication, probably via attenuation of atrial cell tachycardia-induced myolysis and oxidative stress.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yung-Hsin Yeh
- Chang Gung University and Department of Cardiology, Chang Gung Memorial Hospital, Kweishan 333, Taoyuan, Taiwan.
| | | |
Collapse
|
43
|
Lin YK, Chen YC, Kao YH, Tsai CF, Yeh YH, Huang JL, Cheng CC, Chen SA, Chen YJ. A monounsaturated fatty acid (oleic acid) modulates electrical activity in atrial myocytes with calcium and sodium dysregulation. Int J Cardiol 2014; 176:191-8. [PMID: 25064200 DOI: 10.1016/j.ijcard.2014.07.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 05/07/2014] [Accepted: 07/05/2014] [Indexed: 01/05/2023]
Abstract
BACKGROUND Obesity and metabolic syndrome are important risk factors for atrial fibrillation. High plasma concentrations of monounsaturated fatty acids, including oleic acid (OLA), are frequently noted in obese individuals and patients with metabolic syndrome. However, it is not clear whether monounsaturated fatty acids (MUFAs) can directly modulate the electrophysiological characteristics of atrial myocytes. METHODS Whole-cell patch clamp, indo-1 fluorescence, and Western blot analyses were used to record the action potentials (APs), ionic currents, and protein expressions of HL-1 myocytes incubated with and without (control) OLA (0.5mM) for 24h. RESULTS Compared to control myocytes (n=14), OLA-treated myocytes (n=16) had shorter APD90 (65 ± 6 vs. 85 ± 6 ms, p<0.05) and APD50 (24 ± 6 vs. 38 ± 4 ms, p<0.05) with a higher incidence of delayed afterdepolarizations (35.7% vs. 7%, p<0.05), which were suppressed by 4,4'-diisothiocyanatostilbene-2,2'-disulfonic acid (DIDS, a blocker of the calcium-activated chloride current). In addition, OLA-treated myocytes (n=19) exhibited larger calcium transients (0.54 ± 0.06 vs. 0.38 ± 0.05 R410/485, p<0.05), and sarcoplasmic reticular calcium contents (0.91 ± 0.05 vs. 0.64 ± 0.08 R410/485, p<0.05) than control myocytes (n=15). OLA-treated myocytes had larger late sodium currents, smaller sodium-calcium exchanger currents, and smaller sodium-potassium pump currents. Moreover OLA-treated myocytes had higher expressions of sarcoplasmic reticular Ca(2+)-ATPase and calmodulin kinase II, but lower expression of the sodium-potassium ATPase protein than control myocytes. CONCLUSIONS MUFAs can regulate atrial electrophysiological characteristics with calcium and sodium dysregulation, which may contribute to atrial arrhythmogenesis.
Collapse
Affiliation(s)
- Yung-Kuo Lin
- Division of Cardiovascular Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yao-Chang Chen
- Department of Biomedical Engineering, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Hsun Kao
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Chin-Feng Tsai
- Division of Cardiology, Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan; School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Yung-Hsin Yeh
- The First Cardiovascular Division, Chang-Gung Memorial Hospital, Chang-Gung University, Taoyuan, Taiwan
| | - Jin-Long Huang
- Cardiovascular Center, Taichung Veterans General Hospital, Taichung, Taiwan; Faculty of Medicine and Institute of Clinical Medicine, and Cardiovascular Research Institute, National Yang-Ming University, Taipei, Taiwan
| | | | - Shih-Ann Chen
- National Yang-Ming University, School of Medicine, Division of Cardiology and Cardiovascular Research Center, Veterans General Hospital-Taipei, Taipei, Taiwan
| | - Yi-Jen Chen
- Division of Cardiovascular Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
44
|
Andrade J, Khairy P, Dobrev D, Nattel S. The clinical profile and pathophysiology of atrial fibrillation: relationships among clinical features, epidemiology, and mechanisms. Circ Res 2014; 114:1453-68. [PMID: 24763464 DOI: 10.1161/circresaha.114.303211] [Citation(s) in RCA: 874] [Impact Index Per Article: 79.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Atrial fibrillation (AF) is the most common arrhythmia (estimated lifetime risk, 22%-26%). The aim of this article is to review the clinical epidemiological features of AF and to relate them to underlying mechanisms. Long-established risk factors for AF include aging, male sex, hypertension, valve disease, left ventricular dysfunction, obesity, and alcohol consumption. Emerging risk factors include prehypertension, increased pulse pressure, obstructive sleep apnea, high-level physical training, diastolic dysfunction, predisposing gene variants, hypertrophic cardiomyopathy, and congenital heart disease. Potential risk factors are coronary artery disease, kidney disease, systemic inflammation, pericardial fat, and tobacco use. AF has substantial population health consequences, including impaired quality of life, increased hospitalization rates, stroke occurrence, and increased medical costs. The pathophysiology of AF centers around 4 general types of disturbances that promote ectopic firing and reentrant mechanisms, and include the following: (1) ion channel dysfunction, (2) Ca(2+)-handling abnormalities, (3) structural remodeling, and (4) autonomic neural dysregulation. Aging, hypertension, valve disease, heart failure, myocardial infarction, obesity, smoking, diabetes mellitus, thyroid dysfunction, and endurance exercise training all cause structural remodeling. Heart failure and prior atrial infarction also cause Ca(2+)-handling abnormalities that lead to focal ectopic firing via delayed afterdepolarizations/triggered activity. Neural dysregulation is central to atrial arrhythmogenesis associated with endurance exercise training and occlusive coronary artery disease. Monogenic causes of AF typically promote the arrhythmia via ion channel dysfunction, but the mechanisms of the more common polygenic risk factors are still poorly understood and under intense investigation. Better recognition of the clinical epidemiology of AF, as well as an improved appreciation of the underlying mechanisms, is needed to develop improved methods for AF prevention and management.
Collapse
Affiliation(s)
- Jason Andrade
- From Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montreal, Quebec, Canada (J.A., P.K., S.N.); Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada (J.A.); and Faculty of Medicine, Institute of Pharmacology, University Duisburg-Essen, Essen, Germany (D.D.)
| | | | | | | |
Collapse
|
45
|
Mira YELA, Muhuyati, Lu WH, He PY, Liu ZQ, Yang YC. TGF-β1 signal pathway in the regulation of inflammation in patients with atrial fibrillation. ASIAN PAC J TROP MED 2014; 6:999-1003. [PMID: 24144036 DOI: 10.1016/s1995-7645(13)60180-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 10/15/2013] [Accepted: 11/15/2013] [Indexed: 10/26/2022] Open
Abstract
OBJECTIVE To observe the expression changes of inflammatory markers TGF-β1, Smad3 and IL-6 in patients with atrial fibrillation (AF), and to explore the significance of TGF-β1 signaling pathway in the structural remodeling of AF. METHODS The expression of TGF-β1, Smad3 and IL-6 in 50 cases with AF and 30 normal cases were detected by RT-PCR and ELISA. RESULTS The TGF-β1, Smad3 and IL-6 mRNA and protein expression levels in patients with AF were significantly higher than that in the control group (P<0.05), but there was no significantly different between the paroxysmal AF group and the persistent AF group (P>0.05). The TGF-β1mRNA expression in the ⩾ 50 years subgroup was significantly higher than that in the <50 years subgroups, and it was higher in the NYHA III subgroup than in the I/II grade subgroup. It was also higher in the left ventricular ejection fraction (LVEF) <50% subgroup than in LVEF ⩾ 50% group, and it was significantly higher in the AF time ⩾ 36 months subgroup than that in <36 months subgroup (P<0.05). The Smad3 and IL-6 expressions in the in the LVEF <50% subgroup were both high that than that in LVEF ⩾ 50% group, and higher in the AF time ⩾ 36 months subgroup than that in <36 months subgroup (P<0.05). There were a positive correlation between TGF-β1, Smad3 and IL-6 (r=0.687, r=0.547). There were also a positive correlation between Smad3 and IL-6 mRNA (r=0.823). CONCLUSIONS AF is associated with inflammation, and the inflammation is also involved in the fibrillation and sustain of AF. The TGF-β1 signal pathway may be involved in the process of atrial structural remodeling in patients with AF, and iss related with the occurrence and maintenance of AF.
Collapse
Affiliation(s)
- Ye Erbo Lati Ali Mira
- Comprehensive Cardiology Department, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830011, China
| | | | | | | | | | | |
Collapse
|
46
|
Woods CE, Olgin J. Atrial fibrillation therapy now and in the future: drugs, biologicals, and ablation. Circ Res 2014; 114:1532-46. [PMID: 24763469 PMCID: PMC4169264 DOI: 10.1161/circresaha.114.302362] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 03/03/2014] [Indexed: 01/26/2023]
Abstract
Atrial fibrillation (AF) is a complex disease with multiple inter-relating causes culminating in rapid, seemingly disorganized atrial activation. Therapy targeting AF is rapidly changing and improving. The purpose of this review is to summarize current state-of-the-art diagnostic and therapeutic modalities for treatment of AF. The review focuses on reviewing treatment as it relates to the pathophysiological basis of disease and reviews preclinical and clinical evidence for potential new diagnostic and therapeutic modalities, including imaging, biomarkers, pharmacological therapy, and ablative strategies for AF. Current ablation and drug therapy approaches to treating AF are largely based on treating the arrhythmia once the substrate occurs and is more effective in paroxysmal AF rather than persistent or permanent AF. However, there is much research aimed at prevention strategies, targeting AF substrate, so-called upstream therapy. Improved diagnostics, using imaging, genetics, and biomarkers, are needed to better identify subtypes of AF based on underlying substrate/mechanism to allow more directed therapeutic approaches. In addition, novel antiarrhythmics with more atrial specific effects may reduce limiting proarrhythmic side effects. Advances in ablation therapy are aimed at improving technology to reduce procedure time and in mechanism-targeted approaches.
Collapse
Affiliation(s)
- Christopher E Woods
- From the Division of Cardiology, University of California at San Francisco (C.E.W., J.O.); and Division of Cardiology Research, AUST Development, LLC, Mountain View, CA (C.E.W.)
| | | |
Collapse
|
47
|
Abed HS, Wittert GA. Obesity and atrial fibrillation. Obes Rev 2013; 14:929-38. [PMID: 23879190 DOI: 10.1111/obr.12056] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Revised: 05/03/2013] [Accepted: 05/24/2013] [Indexed: 12/31/2022]
Abstract
Atrial fibrillation (AF) is an increasing public health problem, often described as the epidemic of the new millennium. The rising health economic impact of AF, its association with poor quality of life and independent probability of increased mortality, has recently been highlighted. Although population ageing is regarded as an important contributor to this epidemic, obesity and its associated cardiometabolic comorbidities may represent the principal driving factor behind the current and projected AF epidemic. Obesity-related risk factors, such as hypertension, vascular disease, obstructive sleep apnea and pericardial fat, are thought to result in atrial electro-structural dysfunction. In addition, insulin resistance, its associated abnormalities in nutrient utilization and intermediary metabolic by-products are associated with structural and functional abnormalities, ultimately promoting AF. Recent elucidation of molecular pathways, including those responsible for atrial fibrosis, have provided mechanistic insights and the potential for targeted pharmacotherapy. In this article, we review the evidence for an obesity-related atrial electromechanical dysfunction, the mechanisms behind this and its impact on AF therapeutic outcomes. In light of the recently described mechanisms, we illustrate proposed management approaches and avenues for further investigations.
Collapse
Affiliation(s)
- H S Abed
- Department of Medicine, University of Adelaide, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | | |
Collapse
|
48
|
Yeh YH, Kuo CT, Chang GJ, Qi XY, Nattel S, Chen WJ. Nicotinamide adenine dinucleotide phosphate oxidase 4 mediates the differential responsiveness of atrial versus ventricular fibroblasts to transforming growth factor-β. Circ Arrhythm Electrophysiol 2013; 6:790-8. [PMID: 23884197 DOI: 10.1161/circep.113.000338] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Atrial fibrosis, a common feature of atrial fibrillation, is thought to originate from the differential response of atrium versus ventricle to pathological insult. However, detailed mechanisms underlying the regional differences remain unclear. The aim of this study was to investigate the related factor(s) in mediating atrial vulnerability to fibrotic processes. METHODS AND RESULTS We first compared the response of cultured atrial versus ventricular fibroblasts with transforming growth factor-β (TGF-β), a key mediator of myocardial fibrosis. Atrial fibroblasts showed a stronger response to TGF-β1 in producing extracellular matrix protein (collagen and fibronectin) than ventricular fibroblasts. Furthermore, TGF-β1 activated its downstream signaling (Smads) and induced pronounced oxidative stress, including up-regulation of nicotinamide adenine dinucleotide phosphate oxidase in atrial fibroblasts, and to a lesser extent in ventricular fibroblasts. Nicotinamide adenine dinucleotide phosphate oxidase inhibitors and small-interfering RNA for Nox4 eliminated TGF-β-induced difference between atrial and ventricular fibroblasts, suggesting the crucial role of Nox4 in mediating the atrial-ventricular discrepancy. Small-interfering RNA for Smad3 also suppressed the differential responsiveness of atrial versus ventricular fibroblasts to TGF-β1, including Nox4 activation, implicating a crosstalk between nicotinamide adenine dinucleotide phosphate oxidases and Smad. In vivo, the increased TGF-β1 responsiveness and Nox4 expression were documented in the atria of transgenic mice with cardiac overexpression of TGF-β1. CONCLUSIONS Atrial fibroblasts show greater fibrotic and oxidative responses to TGF-β1 than ventricular fibroblasts. Nox4-derived reactive oxygen species production mediates the susceptibility of atrial fibroblasts to TGF-β1 via activating TGF-β1/Smad signaling cascade, which provides a novel insight into the pathogenesis of atrial fibrosis.
Collapse
Affiliation(s)
- Yung-Hsin Yeh
- First Cardiovascular Division, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Kuei-Shan, Taoyuan, Taiwan
| | | | | | | | | | | |
Collapse
|
49
|
Oxidative stress in atrial fibrillation: an emerging role of NADPH oxidase. J Mol Cell Cardiol 2013; 62:72-9. [PMID: 23643589 DOI: 10.1016/j.yjmcc.2013.04.019] [Citation(s) in RCA: 166] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Revised: 03/22/2013] [Accepted: 04/18/2013] [Indexed: 02/07/2023]
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia. Patients with AF have up to seven-fold higher risk of suffering from ischemic stroke. Better understanding of etiologies of AF and its thromboembolic complications are required for improved patient care, as current anti-arrhythmic therapies have limited efficacy and off target effects. Accumulating evidence has implicated a potential role of oxidative stress in the pathogenesis of AF. Excessive production of reactive oxygen species (ROS) is likely involved in the structural and electrical remodeling of the heart, contributing to fibrosis and thrombosis. In particular, NADPH oxidase (NOX) has emerged as a potential enzymatic source for ROS production in AF based on growing evidence from clinical and animal studies. Indeed, NOX can be activated by known upstream triggers of AF such as angiotensin II and atrial stretch. In addition, treatments such as statins, antioxidants, ACEI or AT1RB have been shown to prevent post-operative AF; among which ACEI/AT1RB and statins can attenuate NOX activity. On the other hand, detailed molecular mechanisms by which specific NOX isoform(s) are involved in the pathogenesis of AF and the extent to which activation of NOX plays a causal role in AF development remains to be determined. The current review discusses causes and consequences of oxidative stress in AF with a special focus on the emerging role of NOX pathways.
Collapse
|
50
|
|