1
|
Jiang XX, Zhang R, Wang HS. Neferine mitigates angiotensin II-induced atrial fibrillation and fibrosis via upregulation of Nrf2/HO-1 and inhibition of TGF-β/p-Smad2/3 pathways. Aging (Albany NY) 2024; 16:8630-8644. [PMID: 38775722 PMCID: PMC11164477 DOI: 10.18632/aging.205829] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 04/10/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND Atrial fibrillation (AF) is often associated with atrial fibrosis and oxidative stress. Neferine, a bisbenzylisoquinoline alkaloid, has been reported to exert an antiarrhythmic effect. However, its impact on Angiotensin II (Ang II) infusion-induced AF and the underlying mechanism remains unclear. This study aimed to investigate whether neferine alleviates Ang II-induced AF and explore the underlying mechanisms. METHODS Mice subjected to Ang II infusion to induce AF were concurrently treated with neferine or saline. AF incidence, myocardial cell size, fibrosis, and oxidative stress were then examined. RESULTS Neferine treatment inhibited Ang II-induced AF, atrial size augmentation, and atrial fibrosis. Additionally, we observed that Ang II increased reactive oxygen species (ROS) generation, induced mitochondrial membrane potential depolarization, and reduced glutathione (GSH) and superoxide dismutase (SOD) levels, which were reversed to some extent by neferine. Mechanistically, neferine activated the Nrf2/HO-1 signaling pathway and inhibited TGF-β/p-Smad2/3 in Ang II-infused atria. Zinc Protoporphyrin (ZnPP), an HO-1 inhibitor, reduced the anti-oxidative effect of neferine to some extent and subsequently abolished the beneficial effect of neferine on Ang II-induced AF. CONCLUSIONS These findings provide hitherto undocumented evidence that the protective role of neferine in Ang II-induced AF is dependent on HO-1.
Collapse
Affiliation(s)
- Xiao-Xiao Jiang
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Ri Zhang
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Hui-Shan Wang
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Command, Shenyang 110016, Liaoning, China
| |
Collapse
|
2
|
Yafasov M, Olsen FJ, Shabib A, Skaarup KG, Lassen MCH, Johansen ND, Jensen MT, Jensen GB, Schnohr P, Møgelvang R, Biering-Sørensen T. Even mild mitral regurgitation is associated with incident atrial fibrillation in the general population. Eur Heart J Cardiovasc Imaging 2024; 25:579-586. [PMID: 38078897 DOI: 10.1093/ehjci/jead337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 10/19/2023] [Accepted: 12/05/2023] [Indexed: 02/04/2024] Open
Abstract
AIMS Mitral regurgitation (MR) can be difficult to quantify. We sought to investigate whether the MR jet area to left atrial (LA) area ratio (MR/LA ratio) method for quantifying MRs can be used to predict incident atrial fibrillation (AF) in the general population. METHODS AND RESULTS The study included 4466 participants from the 5th Copenhagen City Heart Study, a prospective general population study, who underwent transthoracic echocardiography. MR jet area was measured and indexed to LA area. The endpoint was incident AF. MR was quantified in 4042 participants (mean age: 57 years, 43% men). Of these, 198 (4.9%) developed AF during a median follow-up period of 5.3 years (interquartile range: 4.4-6.1 years). MR was present in 1938 participants (48%) including 1593 (39%) trace/mild MRs (MR/LA ratio ≤ 20% and ≤4 cm2). In unadjusted analysis, MR/LA ratio was associated with incident AF [HR: 1.06 (1.00-1.13), P = 0.042 per 5% increase] but not after adjusting for CHARGE-AF score. However, the association was modified by age (P for interaction = 0.034), such that MR/LA ratio was associated with AF only in participants ≤ 73 years. In these participants, MR/LA ratio 'was' independently associated with AF after adjusting for CHARGE-AF score [HR: 1.14 (1.06-1.24), P = 0.001, per 5% increase]. This finding persisted when restricting the analysis to participants without moderate or severe MR and normal LA size [HR: 1.35 (1.09-1.68), P = 0.005, per 5% increase]. CONCLUSION MR, including even trace regurgitations quantified by MR/LA ratio, is independently associated with incident AF in individuals ≤ 73 years of age.
Collapse
Affiliation(s)
- Marat Yafasov
- Department of Cardiology, Copenhagen University Hospital-Herlev and Gentofte, Niels Andersens Vej 65, entrance 8, 3rd floor on the right, p. 835, 2900 Hellerup, Denmark
- The Copenhagen City Heart Study, Copenhagen University Hospital-Herlev Hospital, Borgmester Ib Juuls Vej 73, opgang 7, 4. etage, M1, 2730 Herlev, Copenhagen, Denmark
| | - Flemming Javier Olsen
- Department of Cardiology, Copenhagen University Hospital-Herlev and Gentofte, Niels Andersens Vej 65, entrance 8, 3rd floor on the right, p. 835, 2900 Hellerup, Denmark
- The Copenhagen City Heart Study, Copenhagen University Hospital-Herlev Hospital, Borgmester Ib Juuls Vej 73, opgang 7, 4. etage, M1, 2730 Herlev, Copenhagen, Denmark
| | - Ali Shabib
- Department of Cardiology, Copenhagen University Hospital-Herlev and Gentofte, Niels Andersens Vej 65, entrance 8, 3rd floor on the right, p. 835, 2900 Hellerup, Denmark
- The Copenhagen City Heart Study, Copenhagen University Hospital-Herlev Hospital, Borgmester Ib Juuls Vej 73, opgang 7, 4. etage, M1, 2730 Herlev, Copenhagen, Denmark
| | - Kristoffer Grundtvig Skaarup
- Department of Cardiology, Copenhagen University Hospital-Herlev and Gentofte, Niels Andersens Vej 65, entrance 8, 3rd floor on the right, p. 835, 2900 Hellerup, Denmark
- The Copenhagen City Heart Study, Copenhagen University Hospital-Herlev Hospital, Borgmester Ib Juuls Vej 73, opgang 7, 4. etage, M1, 2730 Herlev, Copenhagen, Denmark
| | - Mats Christian Højbjerg Lassen
- Department of Cardiology, Copenhagen University Hospital-Herlev and Gentofte, Niels Andersens Vej 65, entrance 8, 3rd floor on the right, p. 835, 2900 Hellerup, Denmark
- The Copenhagen City Heart Study, Copenhagen University Hospital-Herlev Hospital, Borgmester Ib Juuls Vej 73, opgang 7, 4. etage, M1, 2730 Herlev, Copenhagen, Denmark
| | - Niklas Dyrby Johansen
- Department of Cardiology, Copenhagen University Hospital-Herlev and Gentofte, Niels Andersens Vej 65, entrance 8, 3rd floor on the right, p. 835, 2900 Hellerup, Denmark
- The Copenhagen City Heart Study, Copenhagen University Hospital-Herlev Hospital, Borgmester Ib Juuls Vej 73, opgang 7, 4. etage, M1, 2730 Herlev, Copenhagen, Denmark
- Center for Translational Cardiology and Pragmatic Randomized Trials, Dept. of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Magnus T Jensen
- The Copenhagen City Heart Study, Copenhagen University Hospital-Herlev Hospital, Borgmester Ib Juuls Vej 73, opgang 7, 4. etage, M1, 2730 Herlev, Copenhagen, Denmark
- Steno Diabetes Center Copenhagen, Borgmester Ib Juuls Vej 83, 2730 Herlev, Denmark
| | - Gorm Boje Jensen
- The Copenhagen City Heart Study, Copenhagen University Hospital-Herlev Hospital, Borgmester Ib Juuls Vej 73, opgang 7, 4. etage, M1, 2730 Herlev, Copenhagen, Denmark
| | - Peter Schnohr
- The Copenhagen City Heart Study, Copenhagen University Hospital-Herlev Hospital, Borgmester Ib Juuls Vej 73, opgang 7, 4. etage, M1, 2730 Herlev, Copenhagen, Denmark
| | - Rasmus Møgelvang
- The Copenhagen City Heart Study, Copenhagen University Hospital-Herlev Hospital, Borgmester Ib Juuls Vej 73, opgang 7, 4. etage, M1, 2730 Herlev, Copenhagen, Denmark
- Department of Cardiology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Tor Biering-Sørensen
- Department of Cardiology, Copenhagen University Hospital-Herlev and Gentofte, Niels Andersens Vej 65, entrance 8, 3rd floor on the right, p. 835, 2900 Hellerup, Denmark
- The Copenhagen City Heart Study, Copenhagen University Hospital-Herlev Hospital, Borgmester Ib Juuls Vej 73, opgang 7, 4. etage, M1, 2730 Herlev, Copenhagen, Denmark
- Center for Translational Cardiology and Pragmatic Randomized Trials, Dept. of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
3
|
Bae H, Kim T, Lim I. Carbon monoxide activation of delayed rectifier potassium currents of human cardiac fibroblasts through diverse pathways. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2022; 26:25-36. [PMID: 34965993 PMCID: PMC8723981 DOI: 10.4196/kjpp.2022.26.1.25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/28/2021] [Accepted: 09/28/2021] [Indexed: 06/14/2023]
Abstract
To identify the effect and mechanism of carbon monoxide (CO) on delayed rectifier K+ currents (IK) of human cardiac fibroblasts (HCFs), we used the wholecell mode patch-clamp technique. Application of CO delivered by carbon monoxidereleasing molecule-3 (CORM3) increased the amplitude of outward K+ currents, and diphenyl phosphine oxide-1 (a specific IK blocker) inhibited the currents. CORM3- induced augmentation was blocked by pretreatment with nitric oxide synthase blockers (L-NG-monomethyl arginine citrate and L-NG-nitro arginine methyl ester). Pretreatment with KT5823 (a protein kinas G blocker), 1H-[1,-2,-4] oxadiazolo-[4,-3-a] quinoxalin-1-on (ODQ, a soluble guanylate cyclase blocker), KT5720 (a protein kinase A blocker), and SQ22536 (an adenylate cyclase blocker) blocked the CORM3 stimulating effect on IK. In addition, pretreatment with SB239063 (a p38 mitogen-activated protein kinase [MAPK] blocker) and PD98059 (a p44/42 MAPK blocker) also blocked the CORM3's effect on the currents. When testing the involvement of S-nitrosylation, pretreatment of N-ethylmaleimide (a thiol-alkylating reagent) blocked CO-induced IK activation and DL-dithiothreitol (a reducing agent) reversed this effect. Pretreatment with 5,10,15,20-tetrakis(1-methylpyridinium-4-yl)-21H,23H porphyrin manganese (III) pentachloride and manganese (III) tetrakis (4-benzoic acid) porphyrin chloride (superoxide dismutase mimetics), diphenyleneiodonium chloride (an NADPH oxidase blocker), or allopurinol (a xanthine oxidase blocker) also inhibited CO-induced IK activation. These results suggest that CO enhances IK in HCFs through the nitric oxide, phosphorylation by protein kinase G, protein kinase A, and MAPK, S-nitrosylation and reduction/oxidation (redox) signaling pathways.
Collapse
Affiliation(s)
- Hyemi Bae
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| | - Taeho Kim
- Department of Internal Medicine, College of Medicine, Chung-Ang University Hospital, Seoul 06973, Korea
| | - Inja Lim
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| |
Collapse
|
4
|
Saljic A, Friederike Fenner M, Winters J, Flethøj M, Eggert Eggertsen C, Carstensen H, Dalgas Nissen S, Melis Hesselkilde E, van Hunnik A, Schotten U, Sørensen U, Jespersen T, Verheule S, Buhl R. Increased fibroblast accumulation in the equine heart following persistent atrial fibrillation. IJC HEART & VASCULATURE 2021; 35:100842. [PMID: 34355058 PMCID: PMC8322305 DOI: 10.1016/j.ijcha.2021.100842] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 06/30/2021] [Accepted: 07/06/2021] [Indexed: 01/06/2023]
Abstract
BACKGROUND Fibroblasts maintain the extracellular matrix homeostasis and may couple to cardiomyocytes through gap junctions and thereby increase the susceptibility to slow conduction and cardiac arrhythmias, such as atrial fibrillation (AF). In this study, we used an equine model of persistent AF to characterize structural changes and the role of fibroblasts in the development of an arrhythmogenic substrate for AF. MATERIAL AND METHODS Eleven horses were subjected to atrial tachypacing until self-sustained AF developed and were kept in AF for six weeks. Horses in sinus rhythm (SR) served as control. In terminal open-chest experiments conduction velocity (CV) was measured. Tissue was harvested and stained from selected sites. Automated image analysis was performed to assess fibrosis, fibroblasts, capillaries and various cardiomyocyte characteristics. RESULTS Horses in SR showed a rate-dependent slowing of CV, while in horses with persistent AF this rate-dependency was completely abolished (CV•basic cycle length relation p = 0.0295). Overall and interstitial amounts of fibrosis were unchanged, but an increased fibroblast count was found in left atrial appendage, Bachmann's bundle, intraatrial septum and pulmonary veins (p < 0.05 for all) in horses with persistent AF. The percentage of α-SMA expressing fibroblasts remained the same between the groups. CONCLUSION Persistent AF resulted in fibroblast accumulation in several regions, particularly in the left atrial appendage. The increased number of fibroblasts could be a mediator of altered electrophysiology during AF. Targeting the fibroblast proliferation and differentiation could potentially serve as a novel therapeutic target slowing down the structural remodeling associated with AF.
Collapse
Affiliation(s)
- Arnela Saljic
- Laboratory of Cardiac Physiology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen, Denmark
| | - Merle Friederike Fenner
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Agrovej 8, DK-2630 Taastrup, Denmark
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Grønnegårdsvej 7, 1870 Frederiksberg, Denmark
| | - Joris Winters
- Department of Physiology, Maastricht University, Maastricht, Netherlands
| | - Mette Flethøj
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Agrovej 8, DK-2630 Taastrup, Denmark
| | - Caroline Eggert Eggertsen
- Laboratory of Cardiac Physiology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen, Denmark
| | - Helena Carstensen
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Agrovej 8, DK-2630 Taastrup, Denmark
| | - Sarah Dalgas Nissen
- Laboratory of Cardiac Physiology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen, Denmark
| | - Eva Melis Hesselkilde
- Laboratory of Cardiac Physiology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen, Denmark
| | - Arne van Hunnik
- Department of Physiology, Maastricht University, Maastricht, Netherlands
| | - Ulrich Schotten
- Department of Physiology, Maastricht University, Maastricht, Netherlands
| | | | - Thomas Jespersen
- Laboratory of Cardiac Physiology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen, Denmark
| | - Sander Verheule
- Department of Physiology, Maastricht University, Maastricht, Netherlands
| | - Rikke Buhl
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Agrovej 8, DK-2630 Taastrup, Denmark
| |
Collapse
|
5
|
Li Y, Zhang YX, Ning DS, Chen J, Li SX, Mo ZW, Peng YM, He SH, Chen YT, Zheng CJ, Gao JJ, Yuan HX, Ou JS, Ou ZJ. Simvastatin inhibits POVPC-mediated induction of endothelial-to-mesenchymal cell transition. J Lipid Res 2021; 62:100066. [PMID: 33711324 PMCID: PMC8063863 DOI: 10.1016/j.jlr.2021.100066] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 02/22/2021] [Accepted: 03/05/2021] [Indexed: 11/16/2022] Open
Abstract
Endothelial-to-mesenchymal transition (EndMT), the process by which an endothelial cell (EC) undergoes a series of molecular events that result in a mesenchymal cell phenotype, plays an important role in atherosclerosis. 1-Palmitoyl-2-(5-oxovaleroyl)-sn-glycero-3-phosphocholine (POVPC), derived from the oxidation of 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphatidylcholine, is a proinflammatory lipid found in atherosclerotic lesions. Whether POVPC promotes EndMT and how simvastatin influences POVPC-mediated EndMT remains unclear. Here, we treated human umbilical vein ECs with POVPC, simvastatin, or both, and determined their effect on EC viability, morphology, tube formation, proliferation, and generation of NO and superoxide anion (O2•-). Expression of specific endothelial and mesenchymal markers was detected by immunofluorescence and immunoblotting. POVPC did not affect EC viability but altered cellular morphology from cobblestone-like ECs to a spindle-like mesenchymal cell morphology. POVPC increased O2- generation and expression of alpha-smooth muscle actin, vimentin, Snail-1, Twist-1, transforming growth factor-beta (TGF-β), TGF-β receptor II, p-Smad2/3, and Smad2/3. POVPC also decreased NO production and expression of CD31 and endothelial NO synthase. Simvastatin inhibited POVPC-mediated effects on cellular morphology, production of O2•- and NO, and expression of specific endothelial and mesenchymal markers. These data demonstrate that POVPC induces EndMT by increasing oxidative stress, which stimulates TGF-β/Smad signaling, leading to Snail-1 and Twist-1 activation. Simvastatin inhibited POVPC-induced EndMT by decreasing oxidative stress, suppressing TGF-β/Smad signaling, and inactivating Snail-1 and Twist-1. Our findings reveal a novel mechanism of atherosclerosis that can be inhibited by simvastatin.
Collapse
Affiliation(s)
- Yan Li
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China
| | - Yi-Xin Zhang
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China
| | - Da-Sheng Ning
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China
| | - Jing Chen
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; Division of Hypertension and Vascular Diseases, Department of Cardiology, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Shang-Xuan Li
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China
| | - Zhi-Wei Mo
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China
| | - Yue-Ming Peng
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China
| | - Shi-Hui He
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China
| | - Ya-Ting Chen
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China
| | - Chun-Juan Zheng
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China
| | - Jian-Jun Gao
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China
| | - Hao-Xiang Yuan
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China
| | - Jing-Song Ou
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People's Republic of China.
| | - Zhi-Jun Ou
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, People's Republic of China; Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, People's Republic of China; Division of Hypertension and Vascular Diseases, Department of Cardiology, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China.
| |
Collapse
|
6
|
Wang P, Tian X, Tang J, Duan X, Wang J, Cao H, Qiu X, Wang W, Mai M, Yang Q, Liao R, Yan F. Artemisinin protects endothelial function and vasodilation from oxidative damage via activation of PI3K/Akt/eNOS pathway. Exp Gerontol 2021; 147:111270. [PMID: 33556535 DOI: 10.1016/j.exger.2021.111270] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/29/2021] [Accepted: 01/31/2021] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Previous studies showed that artemisinin (ART) may be useful in the protection against the early development of atherosclerosis, but the effects of ART on vasodilation and eNOS remained unclear. OBJECTIVES AND METHODS In the current study, we investigated the protective effect of ART on endothelial cell injury induced by oxidative stress and its underlying mechanism via MTT assay, Flow Cytometry Assay, Vasodilation study, Western blotting and vivo assay. RESULTS We found that pretreatment of human umbilical vein endothelial cells (HUVECs) with ART significantly suppressed H2O2-induced cell death by decreasing the extent of oxidation and MDA activity, activating SOD, increasing NO production and inhibiting caspase 3/7 activity. Meanwhile, we also found that ART was able to activate PI3K/Akt/eNOS pathway. PI3K inhibitor LY294002 or Akt kinase specific inhibitor Akt inhibitor VIII blocked the protective effect of ART. To explore the effect of ART in the damage of vasodilation induced by H2O2 in mice, we treated the aortic ring from C57BL/6 mice with H2O2 with or without ART, the results demonstrated that ART ameliorated endothelium-dependent vasodilation damage induced by H2O2. CONCLUSION Taken together, these data suggest that ART is able to protect endothelial function and vasodilation from oxidative damage, at least in part through activation of PI3K/Akt/eNOS pathway. Our findings indicate that artemisinin maybe as a potential therapeutic agent for patients with atherosclerosis.
Collapse
Affiliation(s)
- Peng Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaoying Tian
- School of Medical Science, Jinan University, Guangzhou, China
| | - Juxian Tang
- Department of Hematology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510630, China
| | - Xiao Duan
- Department of Rehabilitation, the First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Jinying Wang
- Department of Rehabilitation, the First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Huan Cao
- School of Medical Science, Jinan University, Guangzhou, China
| | - Xiaoyuan Qiu
- School of Medical Science, Jinan University, Guangzhou, China
| | - Wenxuan Wang
- School of Medical Science, Jinan University, Guangzhou, China
| | - Mengfei Mai
- School of Medical Science, Jinan University, Guangzhou, China
| | - Qiaohong Yang
- School of Medical Science, Jinan University, Guangzhou, China.
| | - Rifang Liao
- Department of pharmacy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Fengxia Yan
- School of Medical Science, Jinan University, Guangzhou, China.
| |
Collapse
|
7
|
Callegari S, Macchi E, Monaco R, Magnani L, Tafuni A, Croci S, Nicastro M, Garrapa V, Banchini A, Becchi G, Corradini E, Goldoni M, Rocchio F, Sala R, Benussi S, Ferrara D, Alfieri O, Corradi D. Clinicopathological Bird's-Eye View of Left Atrial Myocardial Fibrosis in 121 Patients With Persistent Atrial Fibrillation: Developing Architecture and Main Cellular Players. Circ Arrhythm Electrophysiol 2020; 13:e007588. [PMID: 32538131 DOI: 10.1161/circep.119.007588] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Scientific research on atrial fibrosis in atrial fibrillation (AF) has mainly focused on quantitative or molecular features. The purpose of this study was to perform a clinicoarchitectural/structural investigation of fibrosis to provide one key to understanding the electrophysiological/clinical aspects of AF. METHODS We characterized the fibrosis (amount, architecture, cellular components, and ultrastructure) in left atrial biopsies from 121 patients with persistent/long-lasting persistent AF (group 1; 59 males; 60±11 years; 91 mitral disease-related AF, 30 nonmitral disease-related AF) and from 39 patients in sinus rhythm with mitral valve regurgitation (group 2; 32 males; 59±12 years). Ten autopsy hearts served as controls. RESULTS Qualitatively, the fibrosis exhibited the same characteristics in all cases and displayed particular architectural scenarios (which we arbitrarily subdivided into 4 stages) ranging from isolated foci to confluent sclerotic areas. The percentage of fibrosis was larger and at a more advanced stage in group 1 versus group 2 and, within group 1, in patients with rheumatic disease versus nonrheumatic cases. In patients with AF with mitral disease and no rheumatic disease, the percentage of fibrosis and the fibrosis stages correlated with both left atrial volume index and AF duration. The fibrotic areas mainly consisted of type I collagen with only a minor cellular component (especially fibroblasts/myofibroblasts; average value range 69-150 cells/mm2, depending on the areas in AF biopsies). A few fibrocytes-circulating and bone marrow-derived mesenchymal cells-were also detectable. The fibrosis-entrapped cardiomyocytes showed sarcolemmal damage and connexin 43 redistribution/internalization. CONCLUSIONS Atrial fibrosis is an evolving and inhomogeneous histological/architectural change that progresses through different stages ranging from isolated foci to confluent sclerotic zones which-seemingly-constrain impulse conduction across restricted regions of electrotonically coupled cardiomyocytes. The fibrotic areas mainly consist of type I collagen extracellular matrix and, only to a lesser extent, mesenchymal cells.
Collapse
Affiliation(s)
- Sergio Callegari
- Center of Excellence for Toxicological Research (CERT) (S.C.), University of Parma, Italy
| | - Emilio Macchi
- Department of Chemistry, Life Sciences and Environmental Sustainability (E.M., L.M., V.G.), University of Parma, Italy
| | - Rodolfo Monaco
- Pathology Unit (R.M., A.T., G.B., E.C., D.C.), Department of Medicine and Surgery, University of Parma, Italy
| | - Luca Magnani
- Department of Chemistry, Life Sciences and Environmental Sustainability (E.M., L.M., V.G.), University of Parma, Italy
| | - Alessandro Tafuni
- Pathology Unit (R.M., A.T., G.B., E.C., D.C.), Department of Medicine and Surgery, University of Parma, Italy
| | - Stefania Croci
- Clinical Immunology, Allergy & Advanced Biotechnologies Unit, Azienda Unità, Sanitaria Locale-IRCCS, Reggio Emilia, Italy (S.C., M.N.)
| | - Maria Nicastro
- Clinical Immunology, Allergy & Advanced Biotechnologies Unit, Azienda Unità, Sanitaria Locale-IRCCS, Reggio Emilia, Italy (S.C., M.N.)
| | - Valentina Garrapa
- Department of Chemistry, Life Sciences and Environmental Sustainability (E.M., L.M., V.G.), University of Parma, Italy
| | - Antonio Banchini
- Forensic Medicine Unit (A.B.), Department of Medicine and Surgery, University of Parma, Italy
| | - Gabriella Becchi
- Pathology Unit (R.M., A.T., G.B., E.C., D.C.), Department of Medicine and Surgery, University of Parma, Italy
| | - Emilia Corradini
- Pathology Unit (R.M., A.T., G.B., E.C., D.C.), Department of Medicine and Surgery, University of Parma, Italy
| | - Matteo Goldoni
- Laboratory of Industrial Toxicology (M.G.), Department of Medicine and Surgery, University of Parma, Italy
| | - Francesca Rocchio
- International Centre for T1D, Paediatric Clinical Research Center Fondazione "Romeo ed Enrica Invernizzi", Department of Biomedical & Clinical Science, Hospital "L. Sacco", University of Milan, Italy (F.R.)
| | - Roberto Sala
- General Pathology Unit (R.S.), Department of Medicine and Surgery, University of Parma, Italy
| | | | - David Ferrara
- Cardiothoracic Surgery Unit, Department of Cardiology, San Raffaele University Hospital, Milan, Italy (D.F., O.A.)
| | - Ottavio Alfieri
- Cardiothoracic Surgery Unit, Department of Cardiology, San Raffaele University Hospital, Milan, Italy (D.F., O.A.)
| | - Domenico Corradi
- Pathology Unit (R.M., A.T., G.B., E.C., D.C.), Department of Medicine and Surgery, University of Parma, Italy
| |
Collapse
|
8
|
Al-Owais MM, Hettiarachchi NT, Boyle JP, Scragg JL, Elies J, Dallas ML, Lippiat JD, Steele DS, Peers C. Multiple mechanisms mediating carbon monoxide inhibition of the voltage-gated K + channel Kv1.5. Cell Death Dis 2017; 8:e3163. [PMID: 29095440 PMCID: PMC5775415 DOI: 10.1038/cddis.2017.568] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 09/19/2017] [Accepted: 09/21/2017] [Indexed: 12/13/2022]
Abstract
The voltage-gated K+ channel has key roles in the vasculature and in atrial excitability and contributes to apoptosis in various tissues. In this study, we have explored its regulation by carbon monoxide (CO), a product of the cytoprotective heme oxygenase enzymes, and a recognized toxin. CO inhibited recombinant Kv1.5 expressed in HEK293 cells in a concentration-dependent manner that involved multiple signalling pathways. CO inhibition was partially reversed by superoxide dismutase mimetics and by suppression of mitochondrial reactive oxygen species. CO also elevated intracellular nitric oxide (NO) levels. Prevention of NO formation also partially reversed CO inhibition of Kv1.5, as did inhibition of soluble guanylyl cyclase. CO also elevated intracellular peroxynitrite levels, and a peroxynitrite scavenger markedly attenuated the ability of CO to inhibit Kv1.5. CO caused nitrosylation of Kv1.5, an effect that was also observed in C331A and C346A mutant forms of the channel, which had previously been suggested as nitrosylation sites within Kv1.5. Augmentation of Kv1.5 via exposure to hydrogen peroxide was fully reversed by CO. Native Kv1.5 recorded in HL-1 murine atrial cells was also inhibited by CO. Action potentials recorded in HL-1 cells were increased in amplitude and duration by CO, an effect mimicked and occluded by pharmacological inhibition of Kv1.5. Our data indicate that Kv1.5 is a target for modulation by CO via multiple mechanisms. This regulation has important implications for diverse cellular functions, including excitability, contractility and apoptosis.
Collapse
Affiliation(s)
- Moza M Al-Owais
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Nishani T Hettiarachchi
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - John P Boyle
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Jason L Scragg
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Jacobo Elies
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Mark L Dallas
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Jon D Lippiat
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Derek S Steele
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Chris Peers
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
9
|
Zhao G, Zhou J, Gao J, Liu Y, Gu S, Zhang X, Su P. Genome-wide DNA methylation analysis in permanent atrial fibrillation. Mol Med Rep 2017; 16:5505-5514. [DOI: 10.3892/mmr.2017.7221] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 04/25/2017] [Indexed: 11/06/2022] Open
|
10
|
Ou ZJ, Chen J, Dai WP, Liu X, Yang YK, Li Y, Lin ZB, Wang TT, Wu YY, Su DH, Cheng TP, Wang ZP, Tao J, Ou JS. 25-Hydroxycholesterol impairs endothelial function and vasodilation by uncoupling and inhibiting endothelial nitric oxide synthase. Am J Physiol Endocrinol Metab 2016; 311:E781-E790. [PMID: 27600825 DOI: 10.1152/ajpendo.00218.2016] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Accepted: 09/02/2016] [Indexed: 12/24/2022]
Abstract
Endothelial dysfunction is a key early step in atherosclerosis. 25-Hydroxycholesterol (25-OHC) is found in atherosclerotic lesions. However, whether 25-OHC promotes atherosclerosis is unclear. Here, we hypothesized that 25-OHC, a proinflammatory lipid, can impair endothelial function, which may play an important role in atherosclerosis. Bovine aortic endothelial cells were incubated with 25-OHC. Endothelial cell proliferation, migration, and tube formation were measured. Nitric oxide (NO) production and superoxide anion generation were determined. The expression and phosphorylation of endothelial NO synthase (eNOS) and Akt as well as the association of eNOS and heat shock protein (HSP)90 were detected by immunoblot analysis and immunoprecipitation. Endothelial cell apoptosis was monitored by TUNEL staining and caspase-3 activity, and expression of Bcl-2, Bax, cleaved caspase-9, and cleaved caspase-3 were detected by immunoblot analysis. Finally, aortic rings from Sprague-Dawley rats were isolated and treated with 25-OHC, and endothelium-dependent vasodilation was evaluated. 25-OHC significantly inhibited endothelial cell proliferation, migration, and tube formation. 25-OHC markedly decreased NO production and increased superoxide anion generation. 25-OHC reduced the phosphorylation of Akt and eNOS and the association of eNOS and HSP90. 25-OHC also enhanced endothelial cell apoptosis by decreasing Bcl-2 expression and increasing cleaved caspase-9 and cleaved caspase-3 expressions as well as caspase-3 activity. 25-OHC impaired endothelium-dependent vasodilation. These data demonstrated that 25-OHC could impair endothelial function by uncoupling and inhibiting eNOS activity as well as by inducing endothelial cell apoptosis. Our findings indicate that 25-OHC may play an important role in regulating atherosclerosis.
Collapse
Affiliation(s)
- Zhi-Jun Ou
- Division of Hypertension and Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; The Key Laboratory of Assisted Circulation, Ministry of Health, Guangzhou, China; Guangdong Province Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; National and Guangdong Province Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; and
| | - Jing Chen
- Division of Hypertension and Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; The Key Laboratory of Assisted Circulation, Ministry of Health, Guangzhou, China; Guangdong Province Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; National and Guangdong Province Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; and
| | - Wei-Ping Dai
- Division of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; The Key Laboratory of Assisted Circulation, Ministry of Health, Guangzhou, China; Guangdong Province Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; National and Guangdong Province Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; and
| | - Xiang Liu
- Division of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; The Key Laboratory of Assisted Circulation, Ministry of Health, Guangzhou, China; Guangdong Province Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; National and Guangdong Province Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; and
| | - Yin-Ke Yang
- Division of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; The Key Laboratory of Assisted Circulation, Ministry of Health, Guangzhou, China; Guangdong Province Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; National and Guangdong Province Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; and
| | - Yan Li
- Division of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; The Key Laboratory of Assisted Circulation, Ministry of Health, Guangzhou, China; Guangdong Province Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; National and Guangdong Province Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; and
| | - Ze-Bang Lin
- Division of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; The Key Laboratory of Assisted Circulation, Ministry of Health, Guangzhou, China; Guangdong Province Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; National and Guangdong Province Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; and
| | - Tian-Tian Wang
- Division of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; The Key Laboratory of Assisted Circulation, Ministry of Health, Guangzhou, China; Guangdong Province Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; National and Guangdong Province Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; and
| | - Ying-Ying Wu
- Division of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; The Key Laboratory of Assisted Circulation, Ministry of Health, Guangzhou, China; Guangdong Province Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; National and Guangdong Province Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; and
| | - Dan-Hong Su
- Division of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; The Key Laboratory of Assisted Circulation, Ministry of Health, Guangzhou, China; Guangdong Province Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; National and Guangdong Province Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; and
| | - Tian-Pu Cheng
- Division of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; The Key Laboratory of Assisted Circulation, Ministry of Health, Guangzhou, China; Guangdong Province Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; National and Guangdong Province Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; and
| | - Zhi-Ping Wang
- Division of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; The Key Laboratory of Assisted Circulation, Ministry of Health, Guangzhou, China; Guangdong Province Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; National and Guangdong Province Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; and
| | - Jun Tao
- Division of Hypertension and Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; The Key Laboratory of Assisted Circulation, Ministry of Health, Guangzhou, China; Guangdong Province Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; National and Guangdong Province Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; and
| | - Jing-Song Ou
- Division of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; The Key Laboratory of Assisted Circulation, Ministry of Health, Guangzhou, China; Guangdong Province Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; National and Guangdong Province Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; and Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, China
| |
Collapse
|
11
|
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]
|
12
|
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
|
13
|
Opacic D, van Bragt KA, Nasrallah HM, Schotten U, Verheule S. Atrial metabolism and tissue perfusion as determinants of electrical and structural remodelling in atrial fibrillation. Cardiovasc Res 2016; 109:527-41. [DOI: 10.1093/cvr/cvw007] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 01/12/2016] [Indexed: 12/14/2022] Open
|
14
|
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]
|
15
|
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
|
16
|
Beinart R, Nazarian S. Role of magnetic resonance imaging in atrial fibrillation ablation. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2014; 16:316. [PMID: 24748019 DOI: 10.1007/s11936-014-0316-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OPINION STATEMENT Ablation therapy is widely used for treatment of drug-resistant atrial fibrillation (AF). Ablation success for AF, however, is relatively low, often requiring repeated procedures for long-term suppression of the arrhythmia. Utilization of imaging techniques that visualize cardiac anatomy, function, and tissue characteristics may improve ablation results. Compared to other imaging modalities, cardiac magnetic resonance (CMR) has several advantages, including the lack of ionizing radiation and unsurpassed soft tissue resolution. Chamber morphology images can be registered onto electroanatomic maps acquired during the procedure, thus improving procedural safety and efficacy. In addition, the ability of CMR to characterize myocardial tissues may optimize patient selection for ablation and thromboembolic risk stratification. Post-procedure CMR can be used to detect potential complications, and with improved resolution, it has the potential to assess the integrity of ablation lesions. In this paper we will review the role of CMR in the pre-ablation diagnostic workup of AF patients as well as during and after catheter ablation.
Collapse
Affiliation(s)
- Roy Beinart
- Department of Medicine/Cardiology, Johns Hopkins University, 702 Rutland Avenue, Traylor 903, Baltimore, MD, 21287, USA,
| | | |
Collapse
|
17
|
Corradi D, Callegari S, Manotti L, Ferrara D, Goldoni M, Alinovi R, Pinelli S, Mozzoni P, Andreoli R, Asimaki A, Pozzoli A, Becchi G, Mutti A, Benussi S, Saffitz JE, Alfieri O. Persistent lone atrial fibrillation: clinicopathologic study of 19 cases. Heart Rhythm 2014; 11:1250-8. [PMID: 24560692 DOI: 10.1016/j.hrthm.2014.02.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Indexed: 11/26/2022]
Abstract
BACKGROUND The extent to which atrial myocardium is remodeled in patients with persistent lone atrial fibrillation (LAF) is largely unknown. OBJECTIVE The purpose of this study was to perform a clinicopathologic investigation in patients with persistent LAF. METHODS We characterized structural and molecular remodeling in atrial biopsies from 19 patients (17 males, mean age 49 years) with persistent (>7 days; n = 8) or long-lasting persistent (>1 year; n = 11) LAF who underwent surgical ablation. Atrial tissue from 15 autopsy samples without clinicopathologic evidence of heart disease served as controls. RESULTS Morphometric analysis showed cardiomyocyte hypertrophy and greater amounts of myolytic damage and interstitial fibrosis in persistent LAF patients compared to controls (P <.0001). Atrial tissue levels of heme oxygenase-1 and 3-nitrotyrosine were increased in persistent LAF patients (P <.001), consistent with oxidative stress. Levels of superoxide dismutase-2, interleukin-8, interleukin-10, tumor necrosis factor-α, and thiobarbituric acid reactive substance were greater in controls than in persistent LAF patients. Immunoreactive signal for connexin43 was reduced more frequently in persistent LAF patients than controls. There was no correlation between features of structural or molecular remodeling and clinical parameters, including persistent LAF duration. CONCLUSION In persistent LAF patients, the atria are modified by structural remodeling and molecular changes of oxidative stress. Tissue changes in persistent LAF appear to occur early after its onset and are qualitatively no different than those observed in patients with atrial fibrillation related to conventional risk factors. These findings suggest that different types of atrial fibrillation are associated with the same spectrum of tissue lesions. Early intervention to restore sinus rhythm in persistent LAF patients may prevent irreversible tissue change, especially interstitial fibrosis.
Collapse
Affiliation(s)
- Domenico Corradi
- Department of Biomedical, Biotechnological, and Translational Sciences (S.Bi.Bi.T.), Unit of Pathology, University of Parma, Parma, Italy.
| | | | - Laura Manotti
- Department of Biomedical, Biotechnological, and Translational Sciences (S.Bi.Bi.T.), Unit of Pathology, University of Parma, Parma, Italy
| | - David Ferrara
- Cardiothoracic Surgery Unit, Department of Cardiology, San Raffaele University Hospital, Milan, Italy
| | - Matteo Goldoni
- Department of Clinical and Experimental Medicine, Laboratory of Industrial Toxicology, University of Parma, Parma, Italy
| | - Rossella Alinovi
- Department of Clinical and Experimental Medicine, Laboratory of Industrial Toxicology, University of Parma, Parma, Italy
| | - Silvana Pinelli
- Department of Clinical and Experimental Medicine, Laboratory of Industrial Toxicology, University of Parma, Parma, Italy
| | - Paola Mozzoni
- Department of Clinical and Experimental Medicine, Laboratory of Industrial Toxicology, University of Parma, Parma, Italy; Italian Workers' Compensation Authority (INAIL) Research Center at the University of Parma, Parma, Italy
| | - Roberta Andreoli
- Department of Clinical and Experimental Medicine, Laboratory of Industrial Toxicology, University of Parma, Parma, Italy; Italian Workers' Compensation Authority (INAIL) Research Center at the University of Parma, Parma, Italy
| | - Angeliki Asimaki
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Alberto Pozzoli
- Cardiothoracic Surgery Unit, Department of Cardiology, San Raffaele University Hospital, Milan, Italy
| | - Gabriella Becchi
- Department of Biomedical, Biotechnological, and Translational Sciences (S.Bi.Bi.T.), Unit of Pathology, University of Parma, Parma, Italy
| | - Antonio Mutti
- Department of Clinical and Experimental Medicine, Laboratory of Industrial Toxicology, University of Parma, Parma, Italy
| | - Stefano Benussi
- Cardiothoracic Surgery Unit, Department of Cardiology, San Raffaele University Hospital, Milan, Italy
| | - Jeffrey E Saffitz
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Ottavio Alfieri
- Cardiothoracic Surgery Unit, Department of Cardiology, San Raffaele University Hospital, Milan, Italy
| |
Collapse
|
18
|
Zhou J, Gao J, Liu Y, Gu S, Zhang X, An X, Yan J, Xin Y, Su P. Human atrium transcript analysis of permanent atrial fibrillation. Int Heart J 2014; 55:71-7. [PMID: 24463922 DOI: 10.1536/ihj.13-196] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Atrial fibrillation (AF) is the most common sustained cardiac arrhythmia and is associated with increased risks of stroke and heart failure. However, the exact mechanisms of left atrium remodeling and AF-related biological behaviors are not completely understood.The transcripts of left atrium in permanent atrium fibrillation patients (n = 7) were compared with those of healthy heart donors (n = 4) in sinus rhythm using Agilent 4x44K microarrays. Differently expressed genes were analyzed based on Gene Ontology and KEGG and Biocarta pathway analysis databases.We identified 567 down- and 420 up-regulated genes in atrial fibrillation. The majority of the down-regulated genes participated in metabolic processes, particularly that for fatty acids. The most remarkable up-regulating effects were immune and platelet activation. In addition, atrial remodeling including structural, contractile, electrophysiological, neurohormone, and oxidant stress was also observed, suggesting various pathophysiology changes in fibrillating atrium. Nine AF closely related genes were validated by real-time RT-PCR.Some AF specific genes were determined which may be a complement to the mechanism of left atrium remodeling. Metabolic changes and inflammation could promote or aggravate atrial fibrillation.
Collapse
Affiliation(s)
- Jian Zhou
- Department of Cardiac Surgery, Beijing Chaoyang Hospital, Capital Medical University, Ministry of Education
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Corradi D. Atrial fibrillation from the pathologist's perspective. Cardiovasc Pathol 2013; 23:71-84. [PMID: 24462196 DOI: 10.1016/j.carpath.2013.12.001] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 12/03/2013] [Accepted: 12/07/2013] [Indexed: 12/18/2022] Open
Abstract
Atrial fibrillation (AF), the most common sustained cardiac arrhythmia encountered in clinical practice, is associated with increased morbidity and mortality. Electrophysiologically, it is characterized by a high rate of asynchronous atrial cell depolarization causing a loss of atrial contractile function and irregular ventricular rates. For a long time, AF was considered as a pure functional disorder without any structural background. Only in recent years, have new mapping and imaging techniques identified atrial locations, which are very often involved in the initiation and maintenance of this supraventricular arrhythmia (i.e. the distal portion of the pulmonary veins and the surrounding atrial myocardium). Morphological analysis of these myocardial sites has demonstrated significant structural remodeling as well as paved the way for further knowledge of AF natural history, pathogenesis, and treatment. This architectural myocardial disarrangement is induced by the arrhythmia itself and the very frequently associated cardiovascular disorders. At the same time, the structural remodeling is also capable of sustaining AF, thereby creating a sort of pathogenetic vicious circle. This review focuses on current understanding about the structural and genetic bases of AF with reference to their classification, pathogenesis, and clinical implications.
Collapse
Affiliation(s)
- Domenico Corradi
- Department of Biomedical, Biotechnological, and Translational Sciences (S.Bi.Bi.T.), Unit of Pathology, University of Parma, Parma, Italy.
| |
Collapse
|
20
|
Fibrosis and electrophysiological characteristics of the atrial appendage in patients with atrial fibrillation and structural heart disease. J Interv Card Electrophysiol 2013; 38:85-93. [PMID: 24026967 DOI: 10.1007/s10840-013-9820-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2013] [Accepted: 06/27/2013] [Indexed: 10/26/2022]
Abstract
PURPOSE This study was conducted to investigate the degree of fibrosis in atrial appendages of patients with and without atrial fibrillation (AF) undergoing cardiac surgery. In addition, we hypothesized that areas of atrial fibrosis can be identified by electrogram fractionation and low voltage for potential ablation therapy. METHODS Interstitial fibrosis from right (RAA) and/or left atrial appendages (LAA) was studied in patients with sinus rhythm (SR, n = 8), paroxysmal (n = 21), and persistent AF (n = 20) undergoing coronary artery bypass and/or aortic or mitral valve surgery. Atrial fibrosis quantification was performed with Masson trichrome staining. Intraoperative bipolar epicardial electrophysiological measurements were performed to correlate fibrosis to electrogram fractionation, voltage, and AF cycle length. RESULTS The average degree of fibrosis was 11.2 ± 7.2 % in the LAA and 22.8 ± 7.6 % in the RAA (p < 0.001). Fibrosis was not significantly higher in paroxysmal AF patients compared to SR subjects (18.2 ± 8.7 versus 20.7 ± 5.3 %). Persistent AF patients had a higher degree of LAA and RAA fibrosis compared to paroxysmal AF patients (LAA 14.6 ± 8.7 versus 8.6 ± 4.7 %, p = 0.02, and RAA 28.2 ± 7.9 versus 18.2 ± 8.7 %, respectively, p = 0.04). The left atrial end diastolic volume index was higher in persistent AF patients compared to SR controls (38.3 ± 16.4 and 28 ± 11 ml/m(2), respectively, p = 0.04). No correlation between atrial fibrosis and electrogram fractionation or voltage was found. CONCLUSION Patients with structural heart disease undergoing cardiac surgery have more fibrosis in the RAA than in the LAA. Furthermore, RAA fibrosis is increased in persistent AF but not paroxysmal AF patients compared to control subjects. Electrogram fractionation and low voltage did not provide accurate identification of the fibrotic substrate.
Collapse
|
21
|
Ci HB, Ou ZJ, Chang FJ, Liu DH, He GW, Xu Z, Yuan HY, Wang ZP, Zhang X, Ou JS. Endothelial microparticles increase in mitral valve disease and impair mitral valve endothelial function. Am J Physiol Endocrinol Metab 2013; 304:E695-E702. [PMID: 23384770 DOI: 10.1152/ajpendo.00016.2013] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mitral valve endothelial cells are important for maintaining lifelong mitral valve integrity and function. Plasma endothelial microparticles (EMPs) increased in various pathological conditions related to activation of endothelial cells. However, whether EMPs will increase in mitral valve disease and their relationship remains unclear. Here, 81 patients with mitral valve disease and 45 healthy subjects were analyzed for the generation of EMPs by flow cytometry. Human mitral valve endothelial cells (HMVECs) were treated with EMPs. The phosphorylation of Akt and endothelial nitric oxide synthase (eNOS), the association of eNOS and heat shock protein 90 (HSP90), and the generation of nitric oxide (NO) and superoxide anion (O(2)(∙-)) were measured. EMPs were increased significantly in patients with mitral valve disease compared with those in healthy subjects. EMPs were negatively correlated with mitral valve area in patients with isolated mitral stenosis. EMPs were significantly higher in the group with severe mitral regurgitation than those in the group with mild and moderate mitral regurgitation. Furthermore, EMPs were decreased dramatically in both Akt and eNOS phosphorylation and the association of HSP90 with eNOS in HMVECs. EMPs decreased NO production but increased O(2)(∙-) generation in HMVECs. Our data demonstrated that EMPs were significantly increased in patients with mitral valve disease. The increase of EMPs can in turn impair HMVEC function by inhibiting the Akt/eNOS-HSP90 signaling pathway. These findings suggest that EMPs may be a therapeutic target for mitral valve disease.
Collapse
Affiliation(s)
- Hong-Bo Ci
- Division of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
|
23
|
CORRADI DOMENICO, CALLEGARI SERGIO, MAESTRI ROBERTA, FERRARA DAVID, MANGIERI DOMENICA, ALINOVI ROSSELLA, MOZZONI PAOLA, PINELLI SILVANA, GOLDONI MATTEO, PRIVITERA YLENIAADELAIDE, BARTOLI VERONICA, ASTORRI ETTORE, MACCHI EMILIO, VAGLIO AUGUSTO, BENUSSI STEFANO, ALFIERI OTTAVIO. Differential Structural Remodeling of the Left-Atrial Posterior Wall in Patients Affected by Mitral Regurgitation with or Without Persistent Atrial Fibrillation: A Morphological and Molecular Study. J Cardiovasc Electrophysiol 2011; 23:271-9. [DOI: 10.1111/j.1540-8167.2011.02187.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
24
|
Chen MC, Chang JP, Wang YH, Liu WH, Ho WC, Chang HW. Autophagy as a mechanism for myolysis of cardiomyocytes in mitral regurgitation. Eur J Clin Invest 2011; 41:299-307. [PMID: 21070221 DOI: 10.1111/j.1365-2362.2010.02410.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Myolysis of atrial cardiomyocytes occurs in patients with severe mitral and tricuspid regurgitation. This morphological remodelling may involve autophagy. METHODS This study comprised 20 patients (10 with long-standing persistent atrial fibrillation and 10 with sinus rhythm) with severe mitral and tricuspid regurgitation. Atrial appendageal tissues were obtained during surgery. The appearance of autophagosomes (LC3B) in myocytes can reflect autophagy induction. Complement 9 is used as a reliable marker of oncosis. RESULTS In the fibrillating right atria, 68·4 ± 18·9% of total myocytes showed moderate-to-severe myolysis, while 64·2 ± 15·8% of total myocytes comprised these cells in right atrial myocardium with sinus rhythm. Immunohistochemical study revealed LC3B-positive myocytes in 8·0% of myocytes without myolysis, 11·9% of myocytes with mild myolysis and 49·4% of myocytes with moderate-to-severe myolysis in right atrial myocardium with sinus rhythm (P < 0·0001). Similarly, in the fibrillating right atria, LC3B-positive myocytes were observed in 5·9% of myocytes without myolysis, 12·2% of myocytes with mild myolysis and 50·7% of myocytes with moderate-to-severe myolysis (P < 0·0001). Moreover, in the fibrillating left atria, LC3B-positive myocytes were observed in 4·9% of myocytes without myolysis, 12·6% of myocytes with mild myolysis and 52·0% of myocytes with moderate-to-severe myolysis (P < 0·0001). None of the atrial myocytes displayed intracellular deposition of complement 9. CONCLUSIONS Induction of autophagy, but not oncosis, occurs in most cases of atrial cardiomyocytes with severe mitral and tricuspid regurgitation, even those without atrial fibrillation, and is closely associated with the development of myolysis in this disease.
Collapse
Affiliation(s)
- Mien-Cheng Chen
- Division of Cardiology and Department of Internal Medicine, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, Kaohsiung, Taiwan.
| | | | | | | | | | | |
Collapse
|
25
|
CORRADI DOMENICO, MAESTRI ROBERTA, MACCHI EMILIO, CALLEGARI SERGIO. Clinical Reviews: The Atria: From Morphology to Function. J Cardiovasc Electrophysiol 2010; 22:223-35. [DOI: 10.1111/j.1540-8167.2010.01887.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
26
|
Lin YK, Lin FZ, Chen YC, Cheng CC, Lin CI, Chen YJ, Chen SA. Oxidative stress on pulmonary vein and left atrium arrhythmogenesis. Circ J 2010; 74:1547-56. [PMID: 20562495 DOI: 10.1253/circj.cj-09-0999] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Oxidative stress and pulmonary veins (PVs) play critical roles in the pathophysiology of atrial fibrillation. The purpose of the present study was to investigate whether oxidative stress and antioxidant agents can change the electrophysiological characteristics of the left atrium (LA) and PVs. METHODS AND RESULTS Conventional microelectrodes were used to record the action potentials (APs) in isolated rabbit PV and LA specimens before and after H(2)O(2) administration with or without ascorbic acid or N-mercaptopropionyl-glycine (N-MPG, a free radical .OH scavenger). H(2)O(2) (0.02 and 0.2 mmol/L) decreased the PV spontaneous rates from 2.0+/-0.1 Hz to 1.6+/-0.1 Hz, and 1.7+/-0.1 Hz (n=10, P<0.05), but H(2)O(2) (2 mmol/L) increased PV spontaneous rates from 2.0+/-0.1 Hz to 2.8+/-0.2 Hz. H(2)O(2) easily induced PV burst firing and early afterdepolarizations, but not in the LA. H(2)O(2) shortened the AP duration and increased the contractile force to a greater extent in the LA than in PVs. In addition, the H(2)O(2)-induced PV burst firing and increasing spontaneous rates were suppressed or attenuated by pretreatment with ascorbic acid (1 mmol/L) or N-MPG (10 mmol/L). CONCLUSIONS H(2)O(2) significantly changed the electrophysiological characteristics of PV and LA through activation of free radicals and may facilitate the occurrence of atrial fibrillation.
Collapse
Affiliation(s)
- Yung-Kuo Lin
- Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
27
|
Chang JP, Chen MC, Liu WH, Yang CH, Chen CJ, Chen YL, Pan KL, Tsai TH, Chang HW. Atrial myocardial nox2 containing NADPH oxidase activity contribution to oxidative stress in mitral regurgitation: potential mechanism for atrial remodeling. Cardiovasc Pathol 2010; 20:99-106. [PMID: 20080418 DOI: 10.1016/j.carpath.2009.12.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2009] [Revised: 12/21/2009] [Accepted: 12/23/2009] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Oxidative stress is linked with several cardiovascular diseases. However, the NADPH oxidase activity in severe mitral regurgitation patients with and without atrial fibrillation has not yet been explored. METHODS This study involved 16 adult patients (eight patients with persistent atrial fibrillation and eight with sinus rhythm) with severe mitral and moderate-to-severe tricuspid regurgitation and five control patients without mitral and tricuspid disease. Atrial tissues of the right and left atrial appendages were obtained during surgery. Superoxide anion production was measured by lucigenin-enhanced chemiluminescence, and the expression of nox2 containing NADPH oxidase mRNA was measured by quantitative real-time RT-PCR. Additionally, immunohistochemical study was performed. RESULTS NADPH-stimulated superoxide release was significantly higher than basal superoxide production from right [5671.9±3498.7 vs. 232.7±70.0 relative light units per second per milligram of protein (RLU s(-1) mg protein(-1)), P=.008) and left atrial homogenates (6475.1±1890.8 vs. 229.0±79.6 RLU s(-1) mg protein(-1), P=.008) in atrial fibrillation patients. The NADPH-stimulated superoxide release from right atrial homogenates was also significantly higher than basal superoxide production in sinus patients (6809.1±1327.1 vs. 244.2±65.5 RLU s(-1) mg protein(-1), P=.008). Additionally, there was a borderline significant correlation between NADPH-stimulated superoxide production from left atrial homogenates and left atrial sizes (r=0.683, P=.062) in atrial fibrillation patients. Membrane-bound nox2 containing NADPH oxidase mRNA expression was increased and was similar in both the atrial fibrillation patients and sinus patients. The NADPH-stimulated superoxide production in right atrial homogenates in control atrial samples was 1863.7±137.2 RLU s(-1) mg protein(-1). Immunohistochemical study demonstrated increased expression of nox2 in myocytes with moderate-to-severe myolysis and hypertrophy. CONCLUSIONS Results of this study demonstrate that membrane-bound nox2 containing NADPH oxidase activity and expression in the atrial myocardium is increased in patients with severe mitral regurgitation, possibly contributing to atrial remodeling in this clinical setting.
Collapse
Affiliation(s)
- Jen-Ping Chang
- Department of Internal Medicine, Chang Gung University College of Medicine, Kaohsiung Hsien 83301, Taiwan, Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Chen MC, Chang JP, Liu WH, Yang CH, Chen CJ, Fang CY, Hsieh YK, Wang YH, Chang HW. Increased serum oxidative stress in patients with severe mitral regurgitation: A new finding and potential mechanism for atrial enlargement. Clin Biochem 2009; 42:943-8. [DOI: 10.1016/j.clinbiochem.2009.04.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2009] [Revised: 04/11/2009] [Accepted: 04/23/2009] [Indexed: 01/22/2023]
|
29
|
Rodrigo R, Vinay J, Castillo R, Cereceda M, Asenjo R, Zamorano J, Araya J, Castillo-Koch R, Espinoza J, Larraín E. Use of vitamins C and E as a prophylactic therapy to prevent postoperative atrial fibrillation. Int J Cardiol 2009; 138:221-8. [PMID: 19446899 DOI: 10.1016/j.ijcard.2009.04.043] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2008] [Revised: 04/07/2009] [Accepted: 04/23/2009] [Indexed: 02/07/2023]
Abstract
Oxidative stress has been strongly involved in the underlying mechanism of atrial fibrillation, particularly in the arrhythmia occurring in patients undergoing cardiac surgery with extracorporeal circulation (postoperative atrial fibrillation). The ischemia/reperfusion injury thus occurring in the myocardial tissue contributes to the development of tissue remodeling, thought to be responsible for the functional heart impairment. Consequently, structural changes due to the cardiac tissue biomolecules attack by reactive oxygen and/or nitrogen species could account for functional changes in ion channels, transporters, membrane conductance, cytosolic transduction signals, and other events, all associated with the occurrence of arrhythmic consequences. The lack of success and significant side effects of anti-arrhythmic drugs have given rise to attempts aimed to develop alternative novel pharmacologic treatments. On this line, the biological properties of the antioxidant vitamins C and E suggest that they could decrease the vulnerability of the heart to the oxidative damage. Nevertheless, very few studies to assess their anti-arrhythmic effects have been reported in humans. The clinical and experimental evidence supporting the view that the pharmacological use of antioxidant vitamins could contribute to prevent postoperative atrial fibrillation is presented.
Collapse
Affiliation(s)
- Ramón Rodrigo
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, University of Chile, Independencia 1027, Casilla 70058, Santiago 7, Chile.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Corradi D, Callegari S, Maestri R, Benussi S, Alfieri O. Structural remodeling in atrial fibrillation. ACTA ACUST UNITED AC 2008; 5:782-96. [DOI: 10.1038/ncpcardio1370] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2008] [Accepted: 08/08/2008] [Indexed: 02/07/2023]
|