1
|
Jiang Z, Lu H, Gao B, Huang J, Ding Y. Transcriptomic Analysis of Cardiac Tissues in a Rodent Model of Coronary Microembolization. J Inflamm Res 2024; 17:6645-6659. [PMID: 39345897 PMCID: PMC11437660 DOI: 10.2147/jir.s469297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 09/13/2024] [Indexed: 10/01/2024] Open
Abstract
Purpose Coronary microembolization (CME) can result in cardiac dysfunction, severe arrhythmias, and a reduced coronary flow reserve. Impairment of mitochondrial energy metabolism has been implicated in the progression and pathogenesis of CME; however, its role remains largely undetermined. This study aimed to explore alterations in mitochondria-related genes in CME. Methods A rat model of CME was successfully established by injecting plastic microspheres into the left ventricle. The cardiac tissues of the two groups were sequenced and mitochondrial functions were assessed. Results Using RNA-Seq, together with GO and KEGG enrichment analyses, we identified 3822 differentially expressed genes (DEGs) in CME rats compared to control rats, and 101 DEGs were mitochondria-related genes. Notably, 36 DEGs were up-regulated and 65 DEGs were down-regulated (CME vs control). In particular, the oxidative phosphorylation (OXPHOS) and mitochondrial electron transport were obviously down-regulated in the CME group. Functional analysis revealed that CME mice exhibited marked reductions in ATP and mitochondrial membrane potential (MMP), by contrast, the production of reactive oxygen species (ROS) was much higher in CME mice than in controls. Protein-protein interaction (PPI) and quantitative PCR (qPCR) validation suggested that eight hub genes including Cmpk2, Isg15, Acsl1, Etfb, Ndufa8, Adhfe1, Gabarapl1 and Acot13 were down-regulated in CME, whereas Aldh18a1 and Hspa5 were up-regulated. Conclusion Our findings suggest that dysfunctions in mitochondrial activity and metabolism are important mechanisms for CME, and mitochondria-related DEGs may be potential therapeutic targets for CME.
Collapse
Affiliation(s)
- Zhaochang Jiang
- Department of Pathology, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang, 310009, People's Republic of China
| | - Haohao Lu
- Zhejiang Center of Laboratory Animals, Hangzhou Medical College, Hangzhou, Zhejiang, 310063, People's Republic of China
| | - Beibei Gao
- Department of Cardiology, Hangzhou First People's Hospital, Hangzhou, Zhejiang, 310006, People's Republic of China
| | - Jinyu Huang
- Department of Cardiology, Hangzhou First People's Hospital, Hangzhou, Zhejiang, 310006, People's Republic of China
| | - Yu Ding
- Department of Clinical Laboratory, Hangzhou First People's Hospital, Hangzhou, Zhejiang, 310006, People's Republic of China
| |
Collapse
|
2
|
Mekhael M, Marrouche N, Hajjar AHE, Donnellan E. The relationship between atrial fibrillation and coronary artery disease: Understanding common denominators. Trends Cardiovasc Med 2024; 34:91-98. [PMID: 36182022 DOI: 10.1016/j.tcm.2022.09.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/13/2022] [Accepted: 09/21/2022] [Indexed: 01/04/2023]
Abstract
Atrial fibrillation (AF) and coronary artery disease (CAD) are highly prevalent cardiovascular conditions. The coexistence of both diseases is common as they share similar risk factors and common pathophysiological characteristics. Systemic inflammatory conditions are associated with an increased incidence of both AF and CAD. The presence of both entities increases the incidence of complications and adverse outcomes. Furthermore, their coexistence poses challenges for the management of patients, particularly with respect to anticoagulation and rhythm management. In this review, we aim to better understand the relationship between AF and CAD by detailing basic molecular pathophysiology, assessing therapeutic guidelines, and describing interactions between the two conditions.
Collapse
Affiliation(s)
- Mario Mekhael
- Tulane University School of Medicine, New Orleans, LA, USA
| | | | | | - Eoin Donnellan
- Tulane University School of Medicine, New Orleans, LA, USA.
| |
Collapse
|
3
|
Song J, Wu J, Robichaux DJ, Li T, Wang S, Arredondo Sancristobal MJ, Dong B, Dobrev D, Karch J, Thomas SS, Li N. A High-Protein Diet Promotes Atrial Arrhythmogenesis via Absent-in-Melanoma 2 Inflammasome. Cells 2024; 13:108. [PMID: 38247800 PMCID: PMC10814244 DOI: 10.3390/cells13020108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 12/29/2023] [Accepted: 01/03/2024] [Indexed: 01/23/2024] Open
Abstract
High-protein diets (HPDs) offer health benefits, such as weight management and improved metabolic profiles. The effects of HPD on cardiac arrhythmogenesis remain unclear. Atrial fibrillation (AF), the most common arrhythmia, is associated with inflammasome activation. The role of the Absent-in-Melanoma 2 (AIM2) inflammasome in AF pathogenesis remains unexplored. In this study, we discovered that HPD increased susceptibility to AF. To demonstrate the involvement of AIM2 signaling in the pathogenesis of HPD-induced AF, wildtype (WT) and Aim2-/- mice were fed normal-chow (NC) and HPD, respectively. Four weeks later, inflammasome activity was upregulated in the atria of WT-HPD mice, but not in the Aim2-/--HPD mice. The increased AF vulnerability in WT-HPD mice was associated with abnormal sarcoplasmic reticulum (SR) Ca2+-release events in atrial myocytes. HPD increased the cytoplasmic double-strand (ds) DNA level, causing AIM2 activation. Genetic inhibition of AIM2 in Aim2-/- mice reduced susceptibility to AF, cytoplasmic dsDNA level, mitochondrial ROS production, and abnormal SR Ca2+-release in atrial myocytes. These data suggest that HPD creates a substrate conducive to AF development by activating the AIM2-inflammasome, which is associated with mitochondrial oxidative stress along with proarrhythmic SR Ca2+-release. Our data imply that targeting the AIM2 inflammasome might constitute a novel anti-AF strategy in certain patient subpopulations.
Collapse
Affiliation(s)
- Jia Song
- Department of Medicine, Section of Cardiovascular Research, Baylor College of Medicine, Houston, TX 77030, USA (M.J.A.S.)
| | - Jiao Wu
- Department of Medicine, Section of Nephrology, Houston, TX 77030, USA
| | - Dexter J. Robichaux
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA (D.D.)
| | - Tingting Li
- Department of Medicine, Section of Cardiovascular Research, Baylor College of Medicine, Houston, TX 77030, USA (M.J.A.S.)
| | - Shuyue Wang
- Department of Medicine, Section of Gastroenterology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Bingning Dong
- Department of Medicine, Section of Gastroenterology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Dobromir Dobrev
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA (D.D.)
- Institute of Pharmacology, University Duisburg-Essen, 45147 Essen, Germany
- Department of Medicine, Montreal Heart Institute, Université de Montréal, Montréal, QC H1T 1C8, Canada
| | - Jason Karch
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA (D.D.)
| | - Sandhya S. Thomas
- Department of Medicine, Section of Nephrology, Houston, TX 77030, USA
- Michael E. Debakey VA Medical Center, Houston, TX 77030, USA
| | - Na Li
- Department of Medicine, Section of Cardiovascular Research, Baylor College of Medicine, Houston, TX 77030, USA (M.J.A.S.)
| |
Collapse
|
4
|
Zacher J, Filipovic K, Predel G, Schmidt T. Exercise and Atrial Fibrillation: The Dose Makes the Poison? A Narrative Review. Int J Sports Med 2024; 45:17-22. [PMID: 37802082 PMCID: PMC10776210 DOI: 10.1055/a-2152-7628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 08/03/2023] [Indexed: 10/08/2023]
Abstract
"All things are poison, and nothing is without poison; the dosage alone makes it so a thing is not a poison" (Paracelsus, ~ 1538 AD). This well-known quote seems to aptly summarize the current understanding of the interaction between exercise and atrial fibrillation (AF). A host of data strongly suggests that regular exercise has a protective effect against developing AF. A small but well-conducted group of trials also demonstrates beneficial effects of exercise in the treatment of AF. Recently, however, potentially detrimental effects of large volumes of high-intensity exercise on the probability of developing AF have moved into the sports-cardiological focus. This effect is well documented for elite athletes; data regarding the general population is less clear. This review presents the current data regarding the protective, therapeutic and potentially risk-enhancing effects of exercise regarding AF. The authors demonstrate that the benefits are clear and strongly outweigh the potential disadvantages.
Collapse
Affiliation(s)
- Jonas Zacher
- Institute of Cardiology and Sports Medicine, Preventative and
Rehabilitative Sports and Performance Medicine, German Sport University Cologne,
Köln, Germany
| | - Karlo Filipovic
- Department of Electrophysiology, University of Cologne, Heart Center,
Uniklinik Köln, Köln, Germany
| | - Georg Predel
- Institute of Cardiology and Sports Medicine, Preventative and
Rehabilitative Sports and Performance Medicine, German Sport University Cologne,
Köln, Germany
| | - Thomas Schmidt
- Institute of Cardiology and Sports Medicine, Preventative and
Rehabilitative Sports and Performance Medicine, German Sport University Cologne,
Köln, Germany
- Schüchtermann-Klinik Bad Rothenfelde, Bad Rothenfelde,
Germany
| |
Collapse
|
5
|
Suga N, Ikeda Y, Yoshikawa S, Taniguchi K, Sawamura H, Matsuda S. Non-Coding RNAs and Gut Microbiota in the Pathogenesis of Cardiac Arrhythmias: The Latest Update. Genes (Basel) 2023; 14:1736. [PMID: 37761875 PMCID: PMC10530369 DOI: 10.3390/genes14091736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/24/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023] Open
Abstract
Non-coding RNAs (ncRNAs) are indispensable for adjusting gene expression and genetic programming throughout development and for health as well as cardiovascular diseases. Cardiac arrhythmia is a frequent cardiovascular disease that has a complex pathology. Recent studies have shown that ncRNAs are also associated with cardiac arrhythmias. Many non-coding RNAs and/or genomes have been reported as genetic background for cardiac arrhythmias. In general, arrhythmias may be affected by several functional and structural changes in the myocardium of the heart. Therefore, ncRNAs might be indispensable regulators of gene expression in cardiomyocytes, which could play a dynamic role in regulating the stability of cardiac conduction and/or in the remodeling process. Although it remains almost unclear how ncRNAs regulate the expression of molecules for controlling cardiac conduction and/or the remodeling process, the gut microbiota and immune system within the intricate networks might be involved in the regulatory mechanisms. This study would discuss them and provide a research basis for ncRNA modulation, which might support the development of emerging innovative therapies against cardiac arrhythmias.
Collapse
Affiliation(s)
| | | | | | | | | | - Satoru Matsuda
- Department of Food Science and Nutrition, Nara Women’s University, Kita-Uoya Nishimachi, Nara 630-8506, Japan; (N.S.); (Y.I.); (S.Y.); (K.T.); (H.S.)
| |
Collapse
|
6
|
Leventopoulos G, Koros R, Travlos C, Perperis A, Chronopoulos P, Tsoni E, Koufou EE, Papageorgiou A, Apostolos A, Kaouris P, Davlouros P, Tsigkas G. Mechanisms of Atrial Fibrillation: How Our Knowledge Affects Clinical Practice. Life (Basel) 2023; 13:1260. [PMID: 37374043 PMCID: PMC10303005 DOI: 10.3390/life13061260] [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: 04/15/2023] [Revised: 05/17/2023] [Accepted: 05/19/2023] [Indexed: 06/29/2023] Open
Abstract
Atrial fibrillation (AF) is a very common arrhythmia that mainly affects older individuals. The mechanism of atrial fibrillation is complex and is related to the pathogenesis of trigger activation and the perpetuation of arrhythmia. The pulmonary veins in the left atrium arei confirm that onfirm the most common triggers due to their distinct anatomical and electrophysiological properties. As a result, their electrical isolation by ablation is the cornerstone of invasive AF treatment. Multiple factors and comorbidities affect the atrial tissue and lead to myocardial stretch. Several neurohormonal and structural changes occur, leading to inflammation and oxidative stress and, consequently, a fibrotic substrate created by myofibroblasts, which encourages AF perpetuation. Several mechanisms are implemented into daily clinical practice in both interventions in and the medical treatment of atrial fibrillation.
Collapse
Affiliation(s)
- Georgios Leventopoulos
- Cardiology Department, University Hospital of Patras, 26504 Patras, Greece; (R.K.); (C.T.); (A.P.); (P.C.); (E.T.); (E.-E.K.); (A.P.); (A.A.); (P.K.); (P.D.); (G.T.)
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Desantis V, Potenza MA, Sgarra L, Nacci C, Scaringella A, Cicco S, Solimando AG, Vacca A, Montagnani M. microRNAs as Biomarkers of Endothelial Dysfunction and Therapeutic Target in the Pathogenesis of Atrial Fibrillation. Int J Mol Sci 2023; 24:5307. [PMID: 36982382 PMCID: PMC10049145 DOI: 10.3390/ijms24065307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/07/2023] [Accepted: 03/08/2023] [Indexed: 03/12/2023] Open
Abstract
The pathophysiology of atrial fibrillation (AF) may involve atrial fibrosis/remodeling and dysfunctional endothelial activities. Despite the currently available treatment approaches, the progression of AF, its recurrence rate, and the high mortality risk of related complications underlay the need for more advanced prognostic and therapeutic strategies. There is increasing attention on the molecular mechanisms controlling AF onset and progression points to the complex cell to cell interplay that triggers fibroblasts, immune cells and myofibroblasts, enhancing atrial fibrosis. In this scenario, endothelial cell dysfunction (ED) might play an unexpected but significant role. microRNAs (miRNAs) regulate gene expression at the post-transcriptional level. In the cardiovascular compartment, both free circulating and exosomal miRNAs entail the control of plaque formation, lipid metabolism, inflammation and angiogenesis, cardiomyocyte growth and contractility, and even the maintenance of cardiac rhythm. Abnormal miRNAs levels may indicate the activation state of circulating cells, and thus represent a specific read-out of cardiac tissue changes. Although several unresolved questions still limit their clinical use, the ease of accessibility in biofluids and their prognostic and diagnostic properties make them novel and attractive biomarker candidates in AF. This article summarizes the most recent features of AF associated with miRNAs and relates them to potentially underlying mechanisms.
Collapse
Affiliation(s)
- Vanessa Desantis
- Department of Precision and Regenerative Medicine and Ionian Area, Pharmacology Section, University of Bari Aldo Moro Medical School, 70124 Bari, Italy
| | - Maria Assunta Potenza
- Department of Precision and Regenerative Medicine and Ionian Area, Pharmacology Section, University of Bari Aldo Moro Medical School, 70124 Bari, Italy
| | - Luca Sgarra
- General Hospital “F. Miulli” Acquaviva delle Fonti, 70021 Bari, Italy
| | - Carmela Nacci
- Department of Precision and Regenerative Medicine and Ionian Area, Pharmacology Section, University of Bari Aldo Moro Medical School, 70124 Bari, Italy
| | - Antonietta Scaringella
- Department of Precision and Regenerative Medicine and Ionian Area, Pharmacology Section, University of Bari Aldo Moro Medical School, 70124 Bari, Italy
| | - Sebastiano Cicco
- Department of Precision and Regenerative Medicine and Ionian Area, Unit of Internal Medicine and Clinical Oncology, University of Bari Aldo Moro Medical School, 70124 Bari, Italy
| | - Antonio Giovanni Solimando
- Department of Precision and Regenerative Medicine and Ionian Area, Unit of Internal Medicine and Clinical Oncology, University of Bari Aldo Moro Medical School, 70124 Bari, Italy
| | - Angelo Vacca
- Department of Precision and Regenerative Medicine and Ionian Area, Unit of Internal Medicine and Clinical Oncology, University of Bari Aldo Moro Medical School, 70124 Bari, Italy
| | - Monica Montagnani
- Department of Precision and Regenerative Medicine and Ionian Area, Pharmacology Section, University of Bari Aldo Moro Medical School, 70124 Bari, Italy
| |
Collapse
|
8
|
Dong Y, Zhai Z, Wang J, Xia Z, Xia Z, Zhu B, Dong Q, Li Q, Li J. Angiotensin receptor-neprilysin inhibitor delays progression from paroxysmal to persistent atrial fibrillation. Sci Rep 2023; 13:3140. [PMID: 36823222 PMCID: PMC9950488 DOI: 10.1038/s41598-023-30349-w] [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: 12/04/2022] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Progression from paroxysmal to persistent atrial fibrillation (AF) is linked to adverse clinical outcomes. The present study sought to clarify whether angiotensin receptor-neprilysin inhibitor (ARNI) can delay AF progression. A retrospective cohort study was conducted on consecutive patients with paroxysmal AF admitted at the Second Affiliated Hospital of Nanchang University between January 2017 and January 2022. The risk of AF progression from paroxysmal to persistent was compared between paroxysmal patients treated with ARNI and those who received an angiotensin receptor blocker (ARB). Seven-day Holter monitoring was performed to identify persistent AF. Propensity-score matched analysis was performed to compare the two groups. Cox-regression was used to estimate the hazard ratio (HR) for AF progression events. A total of 1083 patients were screened, and 113 patients in the ARB group and 57 patients in the ARNI group were eligible for analysis. Before propensity-score matching, the ARNI therapy was associated with a lower risk of AF progression than the ARB therapy (HR 0.34; 95% confidence interval [CI] 0.14-0.81; P = 0.015) after a median follow-up of 705 (interquartile range [IQR] 512 to 895) days. Among 170 patients, 47 ARNI-treated patients were successfully matched to 47 ARB-treated patients. After a median follow-up of 724 (541-929) days, compared to ARB, ARNI significantly reduced the risk of AF progression (HR 0.32; 95% CI 0.12-0.88; P = 0.016). ARNI may be superior to ARB in reducing the risk of progression from paroxysmal to persistent AF.
Collapse
Affiliation(s)
- Youzheng Dong
- grid.412455.30000 0004 1756 5980Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, No.1 of Minde Road, Nanchang, 330006 China
| | - Zhenyu Zhai
- grid.412455.30000 0004 1756 5980Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, No.1 of Minde Road, Nanchang, 330006 China
| | - Jihong Wang
- grid.412455.30000 0004 1756 5980Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, No.1 of Minde Road, Nanchang, 330006 China
| | - Zhen Xia
- grid.412455.30000 0004 1756 5980Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, No.1 of Minde Road, Nanchang, 330006 China
| | - Zirong Xia
- grid.412455.30000 0004 1756 5980Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, No.1 of Minde Road, Nanchang, 330006 China
| | - Bo Zhu
- grid.412455.30000 0004 1756 5980Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, No.1 of Minde Road, Nanchang, 330006 China
| | - Quanbing Dong
- grid.412455.30000 0004 1756 5980Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, No.1 of Minde Road, Nanchang, 330006 China
| | - Qing Li
- grid.412455.30000 0004 1756 5980Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, No.1 of Minde Road, Nanchang, 330006 China
| | - Juxiang Li
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, No.1 of Minde Road, Nanchang, 330006, China.
| |
Collapse
|
9
|
Feng R, Wan J, He Y, Gong H, Xu Z, Feng J. Angiotensin-receptor blocker losartan alleviates atrial fibrillation in rats by downregulating frizzled 8 and inhibiting the activation of WNT-5A pathway. Clin Exp Pharmacol Physiol 2023; 50:19-27. [PMID: 36047789 DOI: 10.1111/1440-1681.13715] [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: 05/20/2022] [Revised: 08/19/2022] [Accepted: 08/30/2022] [Indexed: 12/13/2022]
Abstract
Atrial fibrillation (AF) is a common arrhythmia. Angiotensin-receptor blocker (ARB) is related to AF treatment. This study explored the mechanism of ARB in AF. AF rat models were established by Ach-CaCl2 mixed solution injection. Rats were treated with ARB by gavage and injected with pcDNA3.1-based frizzled homolog 8 (FZD8) overexpression plasmids (oe-FZD8) through the tail vein. The 12-lead electrocardiogram was recorded by biological signal acquisition and processing system and AF duration was recorded, and atrial effective refractory period (AERP) was monitored by electrophysiology. Atrial fibrosis degree, FZD8 messenger RNA and protein levels, collagen I, collagen III, transforming growth factor β1 (TGF-β1), fibronectin, α smooth muscle actin (α-SMA), WBT-5B, and p-JNK1/2 levels, interleukin 1 β (IL-1β) and interleukin 6 (IL-6) levels were detected by Masson staining, reverse transcription quantitative polymerase chain reaction, western blot assay, immunohistochemistry, and enzyme-linked immunosorbent assay. ACh-CaCl2-induced AF rats showed a large area of fused necrosis, abnormal collagen fibre proliferation, high atrial fibrosis degree, and increased atrial fibrosis area in atrial interstitium, elevated collagen I, collagen III, TGF-β1, fibronectin, α-SMA, IL-1β, and IL-6 levels, whereas these trends were averted by ARB treatment. FZD8 was highly expressed in AF rat myocardium. ARB repressed FZD8 expression, prolonged AERP and reduced AF incidence. FZD8 overexpression annulled the effects of ARB on improving AF rat myocardial fibrosis. ARB inactivated the WNT-5A pathway by suppressing FZD8. ARB inactivated the WNT-5A pathway by silencing FZD8, therefore, alleviating AF rat atrial fibrosis.
Collapse
Affiliation(s)
- Ronghua Feng
- Department of Cardiovascular Medicine, The Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang City, Jiangxi Province, China
| | - Jinjie Wan
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, China
| | - Yongsheng He
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, China
| | - Hui Gong
- Department of Cardiovascular Medicine, The Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang City, Jiangxi Province, China
| | - Zeqin Xu
- Department of Cardiovascular Medicine, The Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang City, Jiangxi Province, China
| | - Jiugeng Feng
- Department of Postgraduate, Medical College of Nanchang University, Nanchang City, Jiangxi Province, China
| |
Collapse
|
10
|
Hao H, Dai C, Han X, Li Y. A novel therapeutic strategy for alleviating atrial remodeling by targeting exosomal miRNAs in atrial fibrillation. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119365. [PMID: 36167158 DOI: 10.1016/j.bbamcr.2022.119365] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 08/29/2022] [Accepted: 09/18/2022] [Indexed: 06/16/2023]
Abstract
Atrial fibrillation (AF) is one of the most frequent cardiac arrhythmias, and atrial remodeling is related to the progression of AF. Although several therapeutic approaches have been presented in recent years, the continuously increasing mortality rate suggests that more advanced strategies for treatment are urgently needed. Exosomes regulate pathological processes through intercellular communication mediated by microribonucleic acid (miRNA) in various cardiovascular diseases (CVDs). Exosomal miRNAs associated with signaling pathways have added more complexity to an already complex direct cell-to-cell interaction. Exosome delivery of miRNAs is involved in cardiac regeneration and cardiac protection. Recent studies have found that exosomes play a critical role in the diagnosis and treatment of cardiac fibrosis. By improving exosome stability and modifying surface epitopes, specific pharmaceutical agents can be supplied to improve tropism and targeting to cells and tissues in vivo. Exosomes harboring miRNAs may have clinical utility in cell-free therapeutic approaches and may serve as prognostic and diagnostic biomarkers for AF. Currently, limitations challenge pharmaceutic design, therapeutic utility and in vivo targeted delivery to patients. The aim of this article is to review the developmental features of AF associated with exosomal miRNAs and relate them to underlying mechanisms.
Collapse
Affiliation(s)
- Hongting Hao
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Chenguang Dai
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Xuejie Han
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Yue Li
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China; NHC Key Laboratory of Cell Translation, Harbin Medical University, Heilongjiang 150001, China; Key Laboratory of Hepatosplenic Surgery, Harbin Medical University, Ministry of Education, Harbin 150001, China; Key Laboratory of Cardiac Diseases and Heart Failure, Harbin Medical University, Harbin 150001, China; Heilongjiang Key Laboratory for Metabolic Disorder & Cancer Related Cardiovascular Diseases, Harbin 150081, China; Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Harbin, China.
| |
Collapse
|
11
|
Lu Y, Zhao N, Du Y. Comprehensive bioinformatics analysis reveals common potential mechanisms, progression markers, and immune cells of coronary virus disease 2019 and atrial fibrillation. Front Cardiovasc Med 2022; 9:1027026. [PMID: 36352845 PMCID: PMC9637541 DOI: 10.3389/fcvm.2022.1027026] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 10/07/2022] [Indexed: 12/01/2023] Open
Abstract
OBJECTIVES Atrial fibrillation (AF) is the most common arrhythmia in coronary virus disease 2019 (COVID-19) patients, especially in severe patients. A history of AF can exacerbate COVID-19 symptoms. COVID-19 Patients with new-onset AF have prolonged hospital stays and increased death risk. However, the mechanisms and targets of the interaction between COVID-19 and AF have not been elucidated. MATERIALS AND METHODS We used a series of bioinformatics analyses to understand biological pathways, protein-protein interaction (PPI) networks, gene regulatory networks (GRNs), and protein-chemical interactions between COVID-19 and AF and constructed an AF-related gene signature to assess COVID-19 severity and prognosis. RESULTS We found folate and one-carbon metabolism, calcium regulation, and TFG-β signaling pathway as potential mechanisms linking COVID-19 and AF, which may be involved in alterations in neutrophil metabolism, inflammation, and endothelial cell function. We identified hug genes and found that NF-κb, hsa-miR-1-3p, hsa-miR-124-3p, valproic acid, and quercetin may be key regulatory molecules. We constructed a 3-gene signature consisting of ARG1, GIMAP7, and RFX2 models for the assessment of COVID-19 severity and prognosis, and found that they are associated with neutrophils, T cells, and hematopoietic stem cells, respectively. CONCLUSION Our study reveals a dysregulation of metabolism, inflammation, and immunity between COVID-19 and AF, and identified several therapeutic targets and progression markers. We hope that the results will reveal important insights into the complex interactions between COVID-19 and AF that will drive novel drug development and help in severity assessment.
Collapse
Affiliation(s)
- Yang Lu
- Department of Cardiology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
- Research Center of Ion Channelopathy, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
- Tongji Medical College, Union Hospital, Institute of Cardiology, Huazhong University of Science and Technology, Wuhan, China
- Key Lab for Biological Targeted Therapy of Education Ministry and Hubei Province, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ning Zhao
- Department of Cardiology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
- Research Center of Ion Channelopathy, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
- Tongji Medical College, Union Hospital, Institute of Cardiology, Huazhong University of Science and Technology, Wuhan, China
- Key Lab for Biological Targeted Therapy of Education Ministry and Hubei Province, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yimei Du
- Department of Cardiology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
- Research Center of Ion Channelopathy, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
- Tongji Medical College, Union Hospital, Institute of Cardiology, Huazhong University of Science and Technology, Wuhan, China
- Key Lab for Biological Targeted Therapy of Education Ministry and Hubei Province, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
12
|
Adeleke I, Kersh JS, Sweitzer B. Perioperative management and optimization of atrial fibrillation for noncardiac surgery. Int Anesthesiol Clin 2022; 60:16-23. [PMID: 34897218 DOI: 10.1097/aia.0000000000000352] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Ibukun Adeleke
- Department of Anesthesiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Jay S Kersh
- Department of Anesthesiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | | |
Collapse
|
13
|
Li H, Yang M, Lou D. Troxerutin regulates HIF-1α by activating JAK2/STAT3 signaling to inhibit oxidative stress, inflammation, and apoptosis of cardiomyocytes induced by H 2 O 2. Drug Dev Res 2021; 83:552-563. [PMID: 34622462 DOI: 10.1002/ddr.21885] [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: 08/03/2021] [Revised: 08/24/2021] [Accepted: 08/26/2021] [Indexed: 11/06/2022]
Abstract
Heart failure (HF) is greatly threatening human health and affecting morbidity and mortality worldwide. Troxerutin can alleviate myocardial injury induced by ischemia and hypoxia. The present study aimed to investigate the protective effect of troxerutin on H2 O2 -induced cardiomyocytes and the underlying molecular mechanism. Primary mouse cardiomyocytes morphology induced by H2 O2 in a different duration time was observed by a microscope. After indicated treatment, the viability and apoptosis of cardiomyocytes were detected by CCK-8 assay and flow cytometry analysis. The expression of inflammatory factors and oxidative stress biomarkers was detected by Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and assay kits. Hypoxia inducible factor-1a (HIF-1α) expression was determined by western blot analysis, RT-qPCR analysis and immunofluorescence staining. The apoptosis-related protein expression and the phosphorylation level of janus-activated kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) were detected by the western blot analysis. As a result, after the H2 O2 treatment in a different duration time, the primary mouse cardiomyocytes gradually stopped beating and the morphology of cardiomyocytes treated with H2 O2 was changed significantly from fusiform shape to round shape. The viability of cardiomyocytes was decreased after H2 O2 induction. The HIF-1α expression was increased after the H2 O2 treatment within 30 min while decreased over 30 min. In addition, troxerutin improved viability and suppressed apoptosis, inflammation and oxidative stress of H2 O2 -induced cardiomyocytes, which was reversed by KC7F2 (a HIF-1α inhibitor) or CHZ868 (a JAK inhibitor). To sum up, troxerutin could regulate HIF-1α by activating JAK2/STAT3 signaling to inhibit oxidative stress, inflammation, and apoptosis of cardiomyocytes induced by H2 O2 .
Collapse
Affiliation(s)
- Hui Li
- Department of Cardiology, Shanxi Chinese Medical Hospital, Taiyuan, Shanxi, China
| | - Min Yang
- Department of Medical Oncology, The Second Affiliate Hospital of Zhe Jiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Danfei Lou
- Department of Geriatrics, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
14
|
Xintarakou A, Tzeis S, Psarras S, Asvestas D, Vardas P. Atrial fibrosis as a dominant factor for the development of atrial fibrillation: facts and gaps. Europace 2021; 22:342-351. [PMID: 31998939 DOI: 10.1093/europace/euaa009] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 01/03/2020] [Indexed: 01/08/2023] Open
Abstract
Atrial fibrillation (AF), the most commonly diagnosed arrhythmia, affects a notable percentage of the population and constitutes a major risk factor for thromboembolic events and other heart-related conditions. Fibrosis plays an important role in the onset and perpetuation of AF through structural and electrical remodelling processes. Multiple molecular pathways are involved in atrial substrate modification and the subsequent maintenance of AF. In this review, we aim to recapitulate underlying molecular pathways leading to atrial fibrosis and to indicate existing gaps in the complex interplay of atrial fibrosis and AF.
Collapse
Affiliation(s)
| | - Stylianos Tzeis
- Cardiology Department, Mitera General Hospital, Hygeia Group, Athens, Greece
| | - Stelios Psarras
- Center of Basic Research, Biomedical Research Foundation Academy of Athens, Greece
| | - Dimitrios Asvestas
- Cardiology Department, Mitera General Hospital, Hygeia Group, Athens, Greece
| | - Panos Vardas
- Heart Sector, Hygeia Hospitals Group, 5, Erithrou Stavrou, Marousi, Athens 15123, Greece
| |
Collapse
|
15
|
Hu J, Zhang JJ, Li L, Wang SL, Yang HT, Fan XW, Zhang LM, Hu GL, Fu HX, Song WF, Yan LJ, Liu JJ, Wu JT, Kong B. PU.1 inhibition attenuates atrial fibrosis and atrial fibrillation vulnerability induced by angiotensin-II by reducing TGF-β1/Smads pathway activation. J Cell Mol Med 2021; 25:6746-6759. [PMID: 34132026 PMCID: PMC8278085 DOI: 10.1111/jcmm.16678] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 05/02/2021] [Accepted: 05/08/2021] [Indexed: 12/13/2022] Open
Abstract
Fibrosis serves a critical role in driving atrial remodelling‐mediated atrial fibrillation (AF). Abnormal levels of the transcription factor PU.1, a key regulator of fibrosis, are associated with cardiac injury and dysfunction following acute viral myocarditis. However, the role of PU.1 in atrial fibrosis and vulnerability to AF remain unclear. Here, an in vivo atrial fibrosis model was developed by the continuous infusion of C57 mice with subcutaneous Ang‐II, while the in vitro model comprised atrial fibroblasts that were isolated and cultured. The expression of PU.1 was significantly up‐regulated in the Ang‐II‐induced group compared with the sham/control group in vivo and in vitro. Moreover, protein expression along the TGF‐β1/Smads pathway and the proliferation and differentiation of atrial fibroblasts induced by Ang‐II were significantly higher in the Ang‐II‐induced group than in the sham/control group. These effects were attenuated by exposure to DB1976, a PU.1 inhibitor, both in vivo and in vitro. Importantly, in vitro treatment with small interfering RNA against Smad3 (key protein of TGF‐β1/Smads signalling pathway) diminished these Ang‐II‐mediated effects, and the si‐Smad3‐mediated effects were, in turn, antagonized by the addition of a PU.1‐overexpression adenoviral vector. Finally, PU.1 inhibition reduced the atrial fibrosis induced by Ang‐II and attenuated vulnerability to AF, at least in part through the TGF‐β1/Smads pathway. Overall, the study implicates PU.1 as a potential therapeutic target to inhibit Ang‐II‐induced atrial fibrosis and vulnerability to AF.
Collapse
Affiliation(s)
- Juan Hu
- Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, China
| | - Jing-Jing Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei, China
| | - Li Li
- Department of Cardiology, Qitai Farm Hospital, Xinjiang, China
| | - Shan-Ling Wang
- Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, China
| | - Hai-Tao Yang
- Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, China
| | - Xian-Wei Fan
- Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, China
| | - Lei-Ming Zhang
- Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, China
| | - Guang-Ling Hu
- Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, China
| | - Hai-Xia Fu
- Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, China
| | - Wei-Feng Song
- Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, China
| | - Li-Jie Yan
- Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, China
| | - Jing-Jing Liu
- Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, China
| | - Jin-Tao Wu
- Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, China
| | - Bin Kong
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei, China
| |
Collapse
|
16
|
Martins RC, Pintalhão M, Leite-Moreira A, Castro-Chaves P. Relaxin and the Cardiovascular System: from Basic Science to Clinical Practice. Curr Mol Med 2021; 20:167-184. [PMID: 31642776 DOI: 10.2174/1566524019666191023121607] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 08/07/2019] [Accepted: 10/07/2019] [Indexed: 12/16/2022]
Abstract
The peptide hormone relaxin was originally linked to reproductive physiology, where it is believed to mediate systemic and renal hemodynamic adjustments to pregnancy. Recently, its broad range of effects in the cardiovascular system has been the focus of intensive research regarding its implications under pathological conditions and potential therapeutic potential. An understanding of the multitude of cardioprotective actions prompted the study of serelaxin, recombinant human relaxin-2, for the treatment of acute heart failure. Despite early promising results from phase II studies, recently revealed RELAX-AHF-2 outcomes were rather disappointing and the treatment for acute heart failure remains an unmet medical need. This article reviews the physiologic actions of relaxin on the cardiovascular system and its relevance in the pathophysiology of cardiovascular disease. We summarize the most updated clinical data and discuss future directions of serelaxin for the treatment of acute heart failure. This should encourage additional work to determine how can relaxin's beneficial effects be exploited for the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Rafael Clara Martins
- Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal.,Cardiovascular Research Centre, Porto, Portugal.,Internal Medicine Department, São João Hospital Centre, Porto, Portugal
| | - Mariana Pintalhão
- Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal.,Cardiovascular Research Centre, Porto, Portugal.,Internal Medicine Department, São João Hospital Centre, Porto, Portugal
| | - Adelino Leite-Moreira
- Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal.,Cardiovascular Research Centre, Porto, Portugal.,Cardiothoracic Surgery Department, São João Hospital Centre, Porto, Portugal
| | - Paulo Castro-Chaves
- Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal.,Cardiovascular Research Centre, Porto, Portugal.,Internal Medicine Department, São João Hospital Centre, Porto, Portugal
| |
Collapse
|
17
|
Cui W, Wu X, Shi Y, Guo W, Luo J, Liu H, Zheng L, Du Y, Wang P, Wang Q, Feng D, Ge S, Qu Y. 20-HETE synthesis inhibition attenuates traumatic brain injury-induced mitochondrial dysfunction and neuronal apoptosis via the SIRT1/PGC-1α pathway: A translational study. Cell Prolif 2020; 54:e12964. [PMID: 33314534 PMCID: PMC7848954 DOI: 10.1111/cpr.12964] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/02/2020] [Accepted: 11/17/2020] [Indexed: 12/13/2022] Open
Abstract
Objectives 20‐hydroxyeicosatetraenoic acid (20‐HETE) is a metabolite of arachidonic acid catalysed by cytochrome P450 enzymes and plays an important role in cell death and proliferation. We hypothesized that 20‐HETE synthesis inhibition may have protective effects in traumatic brain injury (TBI) and investigated possible underlying molecular mechanisms. Materials and methods Neurologic deficits, and lesion volume, reactive oxygen species (ROS) levels and cell death as assessed using immunofluorescence staining, transmission electron microscopy and Western blotting were used to determine post‐TBI effects of HET0016, an inhibitor of 20‐HETE synthesis, and their underlying mechanisms. Results The level of 20‐HETE was found to be increased significantly after TBI in mice. 20‐HETE synthesis inhibition reduced neuronal apoptosis, ROS production and damage to mitochondrial structures after TBI. Mechanistically, HET0016 decreased the Drp1 level and increased the expression of Mfn1 and Mfn2 after TBI, indicating a reversal of the abnormal post‐TBI mitochondrial dynamics. HET0016 also promoted the restoration of SIRT1 and PGC‐1α in vivo, and a SIRT1 activator (SRT1720) reversed the downregulation of SIRT1 and PGC‐1α and the abnormal mitochondrial dynamics induced by 20‐HETE in vitro. Furthermore, plasma 20‐HETE levels were found to be higher in TBI patients with unfavourable neurological outcomes and were correlated with the GOS score. Conclusions The inhibition of 20‐HETE synthesis represents a novel strategy to mitigate TBI‐induced mitochondrial dysfunction and neuronal apoptosis by regulating the SIRT1/PGC‐1α pathway.
Collapse
Affiliation(s)
- Wenxing Cui
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xun Wu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yingwu Shi
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Wei Guo
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jianing Luo
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Haixiao Liu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Longlong Zheng
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yong Du
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Ping Wang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Qiang Wang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Dayun Feng
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Shunnan Ge
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yan Qu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
18
|
Li Q, Rempel JD, Ball TB, Aukema H, Minuk GY. Plasma Oxylipins Levels in Nonalcoholic Fatty Liver Disease. Dig Dis Sci 2020; 65:3605-3613. [PMID: 31997053 DOI: 10.1007/s10620-020-06095-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 01/18/2020] [Indexed: 01/22/2023]
Abstract
BACKGROUND Activation of innate immunity by gut-derived immunogens such as lipopolysaccharides (LPS) may play an important role in the pathogenesis of nonalcoholic fatty liver disease (NAFLD). Whether NAFLD-associated lipid disturbances and polyunsaturated fatty acid (PUFA) metabolism in particular contribute to heightened innate immunity, remains to be determined. OBJECTIVE To determine if oxylipins, metabolic products of PUFA metabolism, enhance innate immune reactivity alone and/or following exposure to LPS. METHODS Plasma and peripheral blood mononuclear cells (PBMC) were collected from 35 NAFLD patients and 8 healthy controls. Oxylipin levels were documented by HPLC-MS/MS, cytokines (IL-1, IL-6, IL-10, and TNF-α) by ELISA, and chemokine receptors (CCR1 and CCR2) by flow cytometry. RESULTS Mean plasma levels of four pro-inflammatory oxylipins (Tetranor 12-HETE, 20-HETE, 8-HETrE, and 7-HDoHE) were significantly elevated in NAFLD patients compared to healthy controls. However, the levels did not correlate with the severity of liver injury as reflected by serum aminotransferases, ck18M30, and Fib-4 determinations. In vitro, 20-HETE (0.01-100 nM), the plasma oxylipin with the most significantly elevated plasma levels, did not alter NAFLD or control PBMC cytokine release or enhance the increases in cytokine release following 24 h of LPS exposure. Similarly, 20-HETE alone did not alter PBMC CCR1 or CCR2 expression or LPS-induced downregulation of these receptors. CONCLUSIONS Pro-inflammatory oxylipin levels are increased in NAFLD, but these metabolites do not appear to drive short-term direct or LPS-induced increases in PBMC cytokine release or chemotaxis.
Collapse
Affiliation(s)
- Qian Li
- Morberg Family Chair in Hepatology, Section of Hepatology, Department of Medicine, John Buhler Research Centre, University of Manitoba, 715 McDermot Ave., Winnipeg, MB, R3E 3P4, Canada
| | - Julia D Rempel
- Morberg Family Chair in Hepatology, Section of Hepatology, Department of Medicine, John Buhler Research Centre, University of Manitoba, 715 McDermot Ave., Winnipeg, MB, R3E 3P4, Canada
| | - Terry B Ball
- Medical Microbiology and Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - Harold Aukema
- Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Gerald Y Minuk
- Morberg Family Chair in Hepatology, Section of Hepatology, Department of Medicine, John Buhler Research Centre, University of Manitoba, 715 McDermot Ave., Winnipeg, MB, R3E 3P4, Canada.
| |
Collapse
|
19
|
Pedro B, Fontes-Sousa AP, Gelzer AR. Canine atrial fibrillation: Pathophysiology, epidemiology and classification. Vet J 2020; 265:105548. [PMID: 33129553 DOI: 10.1016/j.tvjl.2020.105548] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 09/20/2020] [Accepted: 09/21/2020] [Indexed: 12/14/2022]
Abstract
Atrial fibrillation (AF) is the most common non-physiological arrhythmia in dogs and humans. Its high prevalence in both species and the impact it has on survival time and quality of life of affected patients, makes it a very relevant topic of medical research. Significant developments in understanding the mechanisms underlying this arrhythmia in humans has occurred over the last decades and some of this knowledge is being applied to veterinary medicine, despite the many differences between species. This article reviews the current understanding of the pathophysiology of AF. The epidemiology and classification of AF in dogs will also be discussed.
Collapse
Affiliation(s)
- Brigite Pedro
- Willows Veterinary Centre and Referral Service, Highlands Road, Solihull, West Midlands B90 4NH, UK.
| | - Ana Patrícia Fontes-Sousa
- Laboratório de Farmacologia e Neurobiologia, Centro de Investigação Farmacológica e Inovação Medicamentosa (MedInUP), Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto (ICBAS-UP), Porto, Portugal
| | - Anna R Gelzer
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
20
|
Gunasekaran S, Kim D. Is Otsu thresholding the answer to reproducible quantification of left atrial scar from late gadolinium-enhancement MRI? J Cardiovasc Electrophysiol 2020; 31:2833-2835. [PMID: 32931626 DOI: 10.1111/jce.14742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 11/30/2022]
Affiliation(s)
- Suvai Gunasekaran
- Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Daniel Kim
- Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.,Department of Biomedical Engineering, Northwestern University, Evanston, Illinois, USA
| |
Collapse
|
21
|
Ma S, Ma J, Tu Q, Zheng C, Chen Q, Lv W. Isoproterenol Increases Left Atrial Fibrosis and Susceptibility to Atrial Fibrillation by Inducing Atrial Ischemic Infarction in Rats. Front Pharmacol 2020; 11:493. [PMID: 32351393 PMCID: PMC7174760 DOI: 10.3389/fphar.2020.00493] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 03/30/2020] [Indexed: 11/24/2022] Open
Abstract
Left atrial (LA) fibrosis is a major arrhythmogenic substrate for atrial fibrillation (AF). The purpose of this study was to assess whether isoproterenol (ISO) induces LA fibrosis and increases susceptibility to AF, exploring the underlying mechanisms. Male Sprague-Dawley rats were subcutaneously injected ISO once per day for 2 days. Five weeks after injection, the ISO group had higher susceptibility AF and prolonged AF duration compared with the control group. ISO decreased LA conduction velocity (CV) and increased LA conduction heterogeneity. ISO increased fibrosise areas and the protein levels of collagen types I and III in the left atrium. Antifibrosis drug pirfenidone decreased AF occurrence and reduced LA fibrosis in ISO treated rats. ISO injection induced atrial ischemia infarction by increasing heart rate and decreasing diastolic and systolic blood pressures. These findings demonstrated that ISO increases susceptibility to AF by increasing LA fibrosis and LA conduction abnormalities 5 weeks after injection. ISO injection induces atrial ischemic injury is the main cause of fibrosis. Rats with ISO-induced LA fibrosis may be used in further AF research.
Collapse
Affiliation(s)
- Shiyu Ma
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Jin Ma
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Qingqiang Tu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Chaoyang Zheng
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Qiuxiong Chen
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Weihui Lv
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| |
Collapse
|
22
|
Gunasekaran S, Lee DC, Knight BP, Collins JD, Fan L, Trivedi A, Ragin AB, Carr JC, Passman RS, Kim D. Left ventricular extracellular volume expansion does not predict recurrence of atrial fibrillation following catheter ablation. Pacing Clin Electrophysiol 2020; 43:159-166. [PMID: 31797387 PMCID: PMC7024017 DOI: 10.1111/pace.13853] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 10/17/2019] [Accepted: 10/28/2019] [Indexed: 01/25/2023]
Abstract
INTRODUCTION A recent study reported that diffuse left ventricular (LV) fibrosis is a predictor of atrial fibrillation (AF) recurrence following catheter ablation, by measuring postcontrast cardiac T1 (an error prone metric as per the 2017 Society for Cardiovascular Magnetic Resonance consensus statement) using an inversion-recovery pulse sequence (an error prone method in arrhythmia) in AF ablation candidates. The purpose of this study was to verify the prior study, by measuring extracellular volume (ECV) fraction (an accurate metric) using a saturation-recovery pulse sequence (accurate method in arrhythmia). METHODS AND RESULTS This study examined 100 AF patients (mean age = 62 ± 11 years, 69 males and 31 females, 67 paroxysmal [pAF] and 33 persistent [peAF]) who underwent a preablation cardiovascular magnetic resonance (CMR) exam. LV ECV and left atrial (LA) and LV functional parameters were quantified using standard analysis methods. During an average follow-up period of 457 ± 261 days with 4 ± 3 rhythm checks per patient, 72 patients maintained sinus rhythm. Between those who maintained sinus rhythm (n = 72) and those who reverted to AF (n = 28), the only clinical characteristic that was significantly different was age (60 ± 12 years vs 66 ± 9 years); for CMR metrics, neither mean LV ECV (25.1 ± 3.3% vs 24.7 ± 3.7%), native LV T1 (1093.8 ± 73.5 ms vs 1070.2 ± 115.9 ms), left ventricular ejection fraction (54.1 ± 11.2% vs 55.7 ± 7.1%), nor LA end diastolic volume/body surface area (42.4 ± 14.8 mL/m2 vs 43.4 ± 19.6 mL/m2 ) were significantly different (P ≥ .23). According to Cox regression tests, none of the clinical and imaging variables predict AF recurrence. CONCLUSION Neither LV ECV nor other CMR metrics predict recurrence of AF following catheter ablation.
Collapse
Affiliation(s)
- Suvai Gunasekaran
- Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, United States
| | - Daniel C. Lee
- Division of Cardiology, Department of Internal Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Bradley P. Knight
- Division of Cardiology, Department of Internal Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Jeremy D. Collins
- Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Department of Radiology, Mayo Clinic, Rochester, MN, United States
| | - Lexiaozi Fan
- Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, United States
| | - Amar Trivedi
- Division of Cardiology, Department of Internal Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Ann B. Ragin
- Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - James C. Carr
- Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Rod S. Passman
- Division of Cardiology, Department of Internal Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Daniel Kim
- Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, United States
| |
Collapse
|
23
|
Abstract
Circular RNAs (circRNAs) are a type of single-stranded RNA molecules that normally do not encode proteins. circRNAs are involved in many physiological processes as well as the pathogenesis of diseases. Cardiac fibrosis is increasingly recognized as a pathological force in advanced heart diseases. A growing number of studies have reported that the occurrence and development of cardiac fibrosis is closely associated with the regulation of circRNAs. This review summarizes the current understanding of circRNA biogenesis and function and will highlight the recent updates regarding the involvement of circRNAs in cardiac fibrosis, and their potential as emerging biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Fatemeh Yousefi
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box 14115-154, Tehran, Iran
| | - Bahram M Soltani
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box 14115-154, Tehran, Iran.
| |
Collapse
|
24
|
Inhibition of UCHL1 by LDN-57444 attenuates Ang II-Induced atrial fibrillation in mice. Hypertens Res 2019; 43:168-177. [PMID: 31700166 PMCID: PMC8075865 DOI: 10.1038/s41440-019-0354-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 09/23/2019] [Accepted: 10/01/2019] [Indexed: 12/22/2022]
Abstract
Atrial fibrillation (AF) is the most common human arrhythmia in clinical practice and may be promoted by atrial inflammation and fibrosis. Ubiquitination is an important posttranslational modification process that is reversed by deubiquitinating enzymes (DUBs). DUBs play critical roles in modulating the degradation, activity, trafficking, and recycling of substrates. However, less research has focused on the role of DUBs in AF. Here, we investigated the effect of ubiquitin C-terminal hydrolase 1 (UCHL1), an important DUB, on the development of AF induced by angiotensin II (Ang II). Male wild-type mice were treated with the UCHL1 inhibitor LDN57444 (LDN) at a dose of 40 μg/kg and infused with Ang II (2000 ng/kg/min) for 3 weeks. Our results showed that Ang II-infused wild-type (WT) mice had higher systolic blood pressure and an increased incidence and duration of AF. Conversely, this effect was attenuated in LDN-treated mice. Moreover, the administration of LDN significantly reduced Ang II-induced left atrial dilation, fibrosis, inflammatory cell infiltration, and reactive oxygen species (ROS) production. Mechanistically, LDN treatment inhibited the activation of multiple signaling pathways (the AKT, ERK1/2, HIF-1α, and TGF-β/smad2/3 pathways) and the expression of CX43 protein in atrial tissues compared with that in vehicle-treated control mice. Overall, our study identified UCHL1 as a novel regulator that contributes to Ang II-induced AF and suggests that the administration of LDN may represent a potential therapeutic approach for treating hypertensive AF.
Collapse
|
25
|
Moreno-Ruiz LA, Madrid-Miller A, Martínez-Flores JE, González-Hermosillo JA, Arenas-Fonseca J, Zamorano-Velázquez N, Mendoza-Pérez B. Left atrial longitudinal strain by speckle tracking as independent predictor of recurrence after electrical cardioversion in persistent and long standing persistent non-valvular atrial fibrillation. Int J Cardiovasc Imaging 2019; 35:1587-1596. [PMID: 30993507 PMCID: PMC6700045 DOI: 10.1007/s10554-019-01597-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 04/03/2019] [Indexed: 11/01/2022]
Abstract
Atrial fibrillation (AF) is the most common arrhythmia in humans. After successful cardioversion, there is a recurrence of 60% due to atrial remodeling, and it has been shown that the global peak atrial longitudinal strain (GPALS) is decreased in these subjects. The aim of this study was to evaluate the predictive value of GPALS for AF recurrence. A prospective cohort of patients with persistent (PnVAF) and long standing persistent non-valvular AF (LSPnVAF) which underwent electrical cardioversion was evaluated with standard echocardiographic variables and GPALS quantification. The primary endpoint was AF recurrence at 6 months. We included PnVAF (n = 50, aged 68.4 ± 10.2 years, female 46%, lasted AF 6 months) and LSPnVAF (n = 81, aged 66.5 ± 13.1 years, female 36%, lasted AF 18 months). At 6 months there were a 68% of recurrence of AF in PnVAF and 53% in LSPnVAF group. GPALS was lower in recurrence 7.8 ± 2.0% versus 21.2 ± 8.9% (p < 0.001) for PnVAF and 7.3 ± 2.7% versus 20.7 ± 7.6% (p < 0.001) in LSPnVAF. GPALS ≤ 10.75% discriminates recurrence at 6 months with a sensitivity of 85%, specificity 99%, PPV 85%, NPV 90%, LR + 8.5 and LR- 0.17. The independent predictors of recurrence in PnVAF were GPALS ≤ 10.75% HR 8.89 [(2.2-35.7), p < 0.01] meanwhile in LSPnVAF were age HR 1.039 [(1.007-1.071), p = 0.01], and GPALS ≤ 10.75% HR 28.1 [(7.2-109.1), p < 0.001]. In subjects with PnVAF and LSPnVAF with successful electrical cardioversion, GPALS ≤ 10.75% predicts arrhythmia recurrence at 6-month follow-up.
Collapse
Affiliation(s)
- Luis Antonio Moreno-Ruiz
- Division of Cardiology, UMAE Hospital de Cardiología, Centro Médico Nacional Siglo XXI, IMSS, 330 Cuauhtémoc Av, Doctores, Cuauhtémoc, 06720, Mexico City, Mexico.
| | - Alejandra Madrid-Miller
- Direction of Education and Research, UMAE Hospital de Cardiología, Centro Médico Nacional Siglo XXI, IMSS, 330 Cuauhtémoc Av, Doctores, Cuauhtémoc, 06720, Mexico City, Mexico
| | - Jerónimo Enrique Martínez-Flores
- Department of Electrophysiology, UMAE Hospital de Cardiología, Centro Médico Nacional Siglo XXI, IMSS, 330 Cuauhtémoc Av, Doctores, Cuauhtémoc, 06720, Mexico City, Mexico
| | - Jesús Antonio González-Hermosillo
- Medical Subadrees of Innovation and Development Project, Instituto Nacional de Cardiología "Ignacio Chávez", 1 Juan Badiano, Sección XVI, Tlalpan, 14080, Mexico City, Mexico
| | - Jorge Arenas-Fonseca
- Department of Echocardiography, UMAE Hospital de Cardiología, Centro Médico Nacional Siglo XXI, IMSS, 330 Cuauhtémoc Av, Doctores, Cuauhtémoc, 06720, Mexico City, Mexico
| | - Noé Zamorano-Velázquez
- Department of Echocardiography, UMAE Hospital de Cardiología, Centro Médico Nacional Siglo XXI, IMSS, 330 Cuauhtémoc Av, Doctores, Cuauhtémoc, 06720, Mexico City, Mexico
| | - Beatriz Mendoza-Pérez
- Division of Cardiology, UMAE Hospital de Cardiología, Centro Médico Nacional Siglo XXI, IMSS, 330 Cuauhtémoc Av, Doctores, Cuauhtémoc, 06720, Mexico City, Mexico
| |
Collapse
|
26
|
Tsai YT, Lin FY, Lin CS, Loh SH, Li CY, Lin CY, Lin YW, Tsai CS. B-type natriuretic peptide enhances fibrotic effects via matrix metalloproteinase-2 expression in the mouse atrium in vivo and in human atrial myofibroblasts in vitro. Transl Res 2019; 208:30-46. [PMID: 30857762 DOI: 10.1016/j.trsl.2019.02.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 01/04/2019] [Accepted: 02/15/2019] [Indexed: 12/13/2022]
Abstract
B-type natriuretic peptide (BNP) was approved by the US Food and Drug Administration in 2001 for the treatment of heart failure. However, the effects of BNP in clinical applications are controversial and uncertain. Recently, study indicated that high BNP levels are associated with an increased risk of developing atrial fibrillation. In this study, we investigated the direct effects of BNP on TNF-α-induced atrial fibrosis mice, as well as its effects on human atrial myofibroblasts. We found that injecting TNF-α-induced mice with recombinant human BNP enhanced atrial fibrosis via matrix metalloproteinase-2 (MMP-2) expression and collagen accumulation. Furthermore, we found that BNP stimulated MMP-2 expression in human atrial myofibroblasts. Treatment of human atrial myofibroblasts with cycloheximide had no effect on this outcome; however, treatment of cells with MG132 enhanced BNP-induced MMP-2 expression, indicating that protein stability and inhibition of proteasome-mediated protein degradation pathways are potentially involved. Inhibition of SIRT1 significantly decreased BNP-induced MMP-2 expression. Additionally, confocal and coimmunoprecipitation data indicated that BNP-regulated MMP-2 expression are likely to be mediated through direct interaction with SIRT1, which is thought to deacetylate MMP-2 and to increase its protein stability in human atrial myofibroblasts. Finally, we confirmed that SIRT1 is expressed and cytoplasmically redistributed as well as colocalized with MMP-2 in mouse fibrotic atrial tissue. We suggest a possible fibrosis-promoting role of BNP in the atrium, although the antifibrotic properties of BNP in the ventricle have been reported in previous studies, and that the coordination between MMP-2 and SIRT1 in BNP-induced atrial myofibroblasts participates in atrial fibrosis.
Collapse
Affiliation(s)
- Yi-Ting Tsai
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan; Division of Cardiovascular Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Feng-Yen Lin
- Division of Cardiology and Cardiovascular Research Center, TaipeiMedical University Hospital, Taipei, Taiwan; Departments of Internal Medicine, College of Medicine, School of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chin-Sheng Lin
- Division of Cardiology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Hurng Loh
- Department of Pharmacology & Graduate Institute of Pharmacology, National Defense Medical Center, Taipei, Taiwan
| | - Chi-Yuan Li
- Department of Anesthesiology, China Medical University and Hospital, Taichung, Taiwan
| | - Cheng-Yen Lin
- Departments of Internal Medicine, College of Medicine, School of Medicine, Taipei Medical University, Taipei, Taiwan; Healthcare Information and Management Department, Ming Chuan University, Taipei, Taiwan
| | - Yi-Wen Lin
- Division of Cardiovascular Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan; Institute of Oral Biology, National Yang-Ming University, Taipei, Taiwan.
| | - Chien-Sung Tsai
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan; Division of Cardiovascular Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan; Department of Pharmacology & Graduate Institute of Pharmacology, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
27
|
Ferkh A, Brown P, O'Keefe E, Zada M, Duggins A, Thiagalingam A, Altman M, Boyd A, Byth K, Kizana E, Denniss AR, Thomas L. Clinical and echocardiographic characteristics of cardioembolic stroke. Eur J Neurol 2019; 26:1310-1317. [PMID: 31062440 DOI: 10.1111/ene.13981] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 04/30/2019] [Indexed: 12/01/2022]
Abstract
BACKGROUND AND PURPOSE Ischaemic stroke frequently has a cardioembolic (CE) source. Clinical and echocardiographic parameters associated with CE stroke were evaluated. METHODS In all, 93 consecutive ischaemic stroke patients who underwent a transthoracic echocardiogram were retrospectively analysed; strokes were classified by TOAST (Trial of Org 10172 in Acute Stroke Treatment) criteria. Echocardiographic parameters related to CE stroke, including left atrial volumes and function, were compared to 73 healthy controls. RESULTS Of 93 patients (mean age 66.1 years, 56% male), nine (10%) had large artery atherosclerosis, 38 (41%) CE stroke, two (2%) small vessel disease, two (2%) other and 42 (45%) undetermined aetiology. Left atrial (LA) maximum volumes (LAVImax ) and minimum volumes (LAVImin ) were larger in the CE group than the non-CE group (45 vs. 32 ml/m2 , 32 vs. 13 ml/m2 , respectively, P < 0.001), whilst LA function indices including LA emptying fraction and LA function index (LAFI) were lower in the CE group (34% vs. 55%, and 0.12 vs. 0.35, respectively, P < 0.001). Adjusting for clinical characteristics, LAFI ≤0.3 was an independent predictor of CE stroke (adjusted odds ratio 5.3, P = 0.001). Additionally, LAVImax and LAVImin were larger (61 vs. 44 and 32 vs. 24 ml/m2 respectively, P < 0.01) and LAFI significantly lower (0.34 vs. 0.52, P < 0.001) in the undetermined aetiology group versus healthy controls. CONCLUSIONS Left atrial enlargement with reduced LA function was associated with CE stroke and LAFI was the best independent predictor. LA parameters were also altered in the undetermined aetiology group, suggesting an underlying LA myopathy in this subset.
Collapse
Affiliation(s)
- A Ferkh
- University of Sydney, Camperdown, NSW, Australia.,Cardiology Department, Westmead Hospital, Sydney, NSW, Australia
| | - P Brown
- Cardiology Department, Westmead Hospital, Sydney, NSW, Australia
| | - E O'Keefe
- Cardiology Department, Westmead Hospital, Sydney, NSW, Australia
| | - M Zada
- Cardiology Department, Westmead Hospital, Sydney, NSW, Australia
| | - A Duggins
- Neurology Department, Westmead Hospital, Sydney, NSW, Australia
| | - A Thiagalingam
- University of Sydney, Camperdown, NSW, Australia.,Cardiology Department, Westmead Hospital, Sydney, NSW, Australia
| | - M Altman
- Cardiology Department, Westmead Hospital, Sydney, NSW, Australia
| | - A Boyd
- University of Sydney, Camperdown, NSW, Australia
| | - K Byth
- University of Sydney, Camperdown, NSW, Australia
| | - E Kizana
- University of Sydney, Camperdown, NSW, Australia.,Cardiology Department, Westmead Hospital, Sydney, NSW, Australia.,Westmead Institute of Medical Research, Sydney, NSW, Australia
| | - A R Denniss
- University of Sydney, Camperdown, NSW, Australia.,Cardiology Department, Westmead Hospital, Sydney, NSW, Australia
| | - L Thomas
- University of Sydney, Camperdown, NSW, Australia.,Cardiology Department, Westmead Hospital, Sydney, NSW, Australia.,South Western Clinical School, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
28
|
Serum relaxin level predicts recurrence of atrial fibrillation after radiofrequency catheter ablation. Heart Vessels 2019; 34:1543-1551. [PMID: 30937522 DOI: 10.1007/s00380-019-01386-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 03/15/2019] [Indexed: 01/10/2023]
Abstract
Relaxin, an emerging biomarker in heart failure, is involved in fibrosis and inflammation. The value of relaxin in predicting recurrence of atrial fibrillation (AF) after radiofrequency catheter ablation (RFCA) is unknown and the subject of this study. We prospectively enrolled 248 consecutive patients with AF (paroxysmal in 127 and persistent in 121) who underwent RFCA at our center after measurement of circulating levels of relaxin by ELISA. Kaplan-Meier analysis with log-rank test and multivariate analysis were used to assess the association between pre-RFCA relaxin levels and post-RFCA AF recurrence at 18 months follow-up. At mean 16.3 ± 3.8 months post-RFCA, 195 (78.6%) patients maintained sinus rhythm, and their pre-RFCA relaxin level was lower than that in patients with AF recurrence (P < 0.001). From lowest to highest pre-RFCA relaxin level tertiles (T1; 82.10-< 234.36; T2; 234.36-< 342.26; and T3; 342.26-740.63 ng/L), AF recurrence rate increased significantly (8.5%, 20.5% and 34.9%, respectively; Kaplan-Meier analysis with log-rank test, χ2 = 18.44, P < 0.001). Using a cutoff of 285.4 ng/L, pre-RFCA relaxin level predicted AF recurrence during follow-up with sensitivity of 77.4% and specificity of 55.9% (area under the receiver operating characteristic curve = 0.71). On multivariate Cox proportional hazard model, relaxin level by tertile (T2, hazard ratio 2.678; 95% confidence interval 1.110-6.460; P = 0.028, and T3, hazard ratio 4.745; 95% confidence interval 2.075-10.854; P < 0.001, respectively compared with the T1) was the independent factor predicting recurrence. Elevated pre-RFCA relaxin level is associated with post-RFCA AF recurrence. A simple measurement of relaxin level therefore might help identify patients at high risk of AF recurrence after RFCA.Clinical Trial Registration chictr.org.cn identifier: ChiCTR-OOC-15006130.
Collapse
|
29
|
Li G, Yang Q, Yang Y, Yang G, Wan J, Ma Z, Du L, Sun Y, Ζhang G. Laminar shear stress alters endothelial KCa2.3 expression in H9c2 cells partially via regulating the PI3K/Akt/p300 axis. Int J Mol Med 2019; 43:1289-1298. [PMID: 30664154 PMCID: PMC6365081 DOI: 10.3892/ijmm.2019.4063] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 01/10/2019] [Indexed: 12/11/2022] Open
Abstract
In cardiac tissues, myoblast atrial myocytes continue to be exposed to mechanical forces including shear stress. However, little is known about the effects of shear stress on atrial myocytes, particularly on ion channel function, in association with disease. The present study demonstrated that the Ca2+-activated K+ channel (KCa)2.3 serves a vital role in regulating arterial tone. As increased intracellular Ca2+ levels and activation of histone acetyltransferase p300 (p300) are early responses to laminar shear stress (LSS) that result in the transcriptional activation of genes, the role of p300 and the phosphoinositide3-kinase (PI3K)/protein kinase B (Akt) pathway, an intracellular pathway that promotes the growth and proliferation rather than the differentiation of adult cells, in the LSS-dependent regulation of KCa2.3 in cardiac myoblasts was examined. In cultured H9c2 cells, exposure to LSS (15 dyn/cm2) for 12 h markedly increased KCa2.3 mRNA expression. Inhibiting PI3K attenuated the LSS-induced increases in the expression and channel activity of KCa2.3, and decreased the phosphorylation levels of p300. The upregulation of these channels was abolished by the inhibition of Akt through decreasing p300 phosphorylation. ChIP assays indicated that p300 was recruited to the promoter region of the KCa2.3 gene. Therefore, the PI3K/Akt/p300 axis serves a crucial role in the LSS-dependent induction of KCa2.3 expression, by regulating cardiac myoblast function and adaptation to hemodynamic changes. The key novel insights gained from the present study are: i) KCa2.3 was upregulated in patients with atrial fibrillation (AF) and in patients with AF combined with mitral value disease; ii) LSS induced a profound upregulation of KCa2.3 mRNA and protein expression in H9c2 cells; iii) PI3K activation was associated with LSS-induced upregulation of the KCa2.3 channel; iv) PI3K activation was mediated by PI3K/Akt-dependent Akt activation; and v) LSS induction of KCa2.3 involved the binding of p300 to transcription factors in the promoter region of the KCa2.3 gene.
Collapse
Affiliation(s)
- Guojian Li
- Department of Vascular Surgery, The Second People's Hospital of Yunnan Province, Kunming Medical University, Kunming, Yunnan 650200, P.R. China
| | - Qionghui Yang
- Department of Pharmacy, The Third People's Hospital of Yunnan Province, Kunming, Yunnan 650200, P.R. China
| | - Yong Yang
- Department of Vascular Surgery, The Second People's Hospital of Yunnan Province, Kunming Medical University, Kunming, Yunnan 650200, P.R. China
| | - Guokai Yang
- Department of Vascular Surgery, The Second People's Hospital of Yunnan Province, Kunming Medical University, Kunming, Yunnan 650200, P.R. China
| | - Jia Wan
- Department of Vascular Surgery, The Second People's Hospital of Yunnan Province, Kunming Medical University, Kunming, Yunnan 650200, P.R. China
| | - Zhenhuan Ma
- Department of Vascular Surgery, The Second People's Hospital of Yunnan Province, Kunming Medical University, Kunming, Yunnan 650200, P.R. China
| | - Lingjuan Du
- Department of Vascular Surgery, The Second People's Hospital of Yunnan Province, Kunming Medical University, Kunming, Yunnan 650200, P.R. China
| | - Yi Sun
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650021, P.R. China
| | - Guimin Ζhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650021, P.R. China
| |
Collapse
|
30
|
Inflammatory and Imaging-based Predictors of Atrial Fibrillation Recurrence after Pulmonary Vein Isolation Using Electroanatomical Mapping – the INFLAMAP Study. JOURNAL OF INTERDISCIPLINARY MEDICINE 2018. [DOI: 10.2478/jim-2018-0007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Abstract
Atrial fibrillation (AF) is the most frequent form of supraventricular arrhythmia in medical practice. It is characterized by chaotic electrical activity in the atria, which often leads to irregular and fast ventricular contractions. Pulmonary veins (PV) play an essential part in the genesis of AF. There are a series of risk factors that trigger the development and recurrence of AF after PV isolation. Despite advanced medical technology, the success rate of AF ablation is not satisfactory. The purpose of this study is to assess the preprocedural imaging and serum biomarkers linked to an increased recurrence of AF after PV isolation. The primary endpoint is represented by AF recurrence after PV isolation. In addition, the rate of cardiovascular death and the rate of major adverse cardiovascular events will be assessed in relation to the enlargement of the left atrium and the volume of epicardial adipose tissue surrounding the heart.
Collapse
|
31
|
Tryptase Promotes the Profibrotic Phenotype Transfer of Atrial Fibroblasts by PAR2 and PPARγ Pathway. Arch Med Res 2018; 49:568-575. [DOI: 10.1016/j.arcmed.2018.12.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 11/23/2018] [Accepted: 12/05/2018] [Indexed: 12/12/2022]
|
32
|
Chen T, Li M, Fan X, Cheng J, Wang L. Sodium Tanshinone IIA Sulfonate Prevents Angiotensin II-Induced Differentiation of Human Atrial Fibroblasts into Myofibroblasts. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:6712585. [PMID: 30140368 PMCID: PMC6081515 DOI: 10.1155/2018/6712585] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 06/25/2018] [Accepted: 07/05/2018] [Indexed: 02/06/2023]
Abstract
Differentiation of atrial fibroblasts into myofibroblasts plays a critical role in atrial fibrosis. Sodium tanshinone IIA sulfonate (DS-201), a water-soluble derivative of tanshinone IIA, has been shown to have potent antifibrotic properties. However, the protective effects of DS-201 on angiotensin II- (Ang II-) induced differentiation of atrial fibroblasts into myofibroblasts remain to be elucidated. In this study, human atrial fibroblasts were stimulated with Ang II in the presence or absence of DS-201. Then, α-smooth muscle actin (α-SMA), collagen I, and collagen III expression and reactive oxygen species (ROS) generation were measured. The expression of transforming growth factor-β1 (TGF-β1) and the downstream signaling of TGF-β1, such as phosphorylation of Smad2/3, were also determined. The results demonstrated that DS-201 significantly prevented Ang II-induced human atrial fibroblast migration and decreased Ang II-induced α-SMA, collagen I, and collagen III expression. Furthermore, increased production of ROS and expression of TGF-β1 stimulated by Ang II were also significantly inhibited by DS-201. Consistent with these results, DS-201 significantly inhibited Ang II-evoked Smad2/3 phosphorylation and periostin expression. These results and the experiments involving N-acetyl cysteine (antioxidant) and an anti-TGF-β1 antibody suggest that DS-201 prevent Ang II-induced differentiation of atrial fibroblasts to myofibroblasts, at least in part, through suppressing oxidative stress and inhibiting the activation of TGF-β1 signaling pathway. All of these data indicate the potential utility of DS-201 for the treatment of cardiac fibrosis.
Collapse
Affiliation(s)
- Tangting Chen
- Key Laboratory of Ministry of Education for Medical Electrophysiology and the Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China
| | - Miaoling Li
- Key Laboratory of Ministry of Education for Medical Electrophysiology and the Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China
| | - Xuehui Fan
- Key Laboratory of Ministry of Education for Medical Electrophysiology and the Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China
| | - Jun Cheng
- Key Laboratory of Ministry of Education for Medical Electrophysiology and the Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China
| | - Liqun Wang
- Drug Discovery Research Center, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China
| |
Collapse
|
33
|
Abstract
20-HETE, the ω-hydroxylation product of arachidonic acid catalyzed by enzymes of the cytochrome P450 (CYP) 4A and 4F gene families, is a bioactive lipid mediator with potent effects on the vasculature including stimulation of smooth muscle cell contractility, migration and proliferation as well as activation of endothelial cell dysfunction and inflammation. Clinical studies have shown elevated levels of plasma and urinary 20-HETE in human diseases and conditions such as hypertension, obesity and metabolic syndrome, myocardial infarction, stroke, and chronic kidney diseases. Studies of polymorphic associations also suggest an important role for 20-HETE in hypertension, stroke and myocardial infarction. Animal models of increased 20-HETE production are hypertensive and are more susceptible to cardiovascular injury. The current review summarizes recent findings that focus on the role of 20-HETE in the regulation of vascular and cardiac function and its contribution to the pathology of vascular and cardiac diseases.
Collapse
Affiliation(s)
- Petra Rocic
- Department of Pharmacology, New York Medical College School of Medicine, Valhalla, NY, United States
| | | |
Collapse
|
34
|
Sarfraz M, Afzal A, Raza SM, Bashir S, Madni A, Khan MW, Ma X, Xiang G. Liposomal co-delivered oleanolic acid attenuates doxorubicin-induced multi-organ toxicity in hepatocellular carcinoma. Oncotarget 2018; 8:47136-47153. [PMID: 28525367 PMCID: PMC5564550 DOI: 10.18632/oncotarget.17559] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 04/18/2017] [Indexed: 01/05/2023] Open
Abstract
Doxorubicin in combination with other cytotoxic drugs has clinical advantages. However, doxorubicin-induced cardiotoxicity negatively impacts clinical utility and outcomes. Cardiotoxicity can result from increased oxidative stress or from a local cytochrome P450 mediated increase in 20-hydroxy-5, 8, 11, 14-eicosatetraenoic acid (20-HETE). Oleanolic acid (OA) is a natural pentacyclic triterpenoid with free radical scavenging, cardioprotective, and P450-mediated cyclooxygenase-upregulating properties. We investigated co-delivery of liposomal OA and doxorubicin in a HepG2 model of hepatocellular carcinoma (HCC). OA attenuated the cardiotoxicity induced by doxorubicin without compromising its anticancer activity. Apoptosis assays revealed that co-delivery of DOX and OA produced a synergistic anticancer effect. However, the drugs had antagonistic effects on cardiomyocytes. Female BALB/c nude mice treated with OA- and DOX-loaded liposomes (ODLs) exhibited reduced tumor growth, stable body weight, and stable organ indices. Reduced 20-HETE production suggested ODLs had limited cardiotoxicity. No changes in biochemical or histopathological markers were observed in mice treated with ODLs. Tailored co-delivery of OA and DOX may thus be an effective therapeutic strategy for treating HCC.
Collapse
Affiliation(s)
- Muhammad Sarfraz
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.,Department of Pharmacy, University of Sargodha, Sargodha, 40100, Punjab, Pakistan
| | - Attia Afzal
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.,Institute of Pharmacy, Lahore College for Women University, Lahore, 54610, Punjab, Pakistan
| | - Shahid Masood Raza
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Sajid Bashir
- Department of Pharmacy, University of Sargodha, Sargodha, 40100, Punjab, Pakistan
| | - Asadullah Madni
- Faculty of Pharmacy and Alternative Medicine, The Islamia University of Bahawalpur, Bahawalpur, 63100, Punjab, Pakistan
| | - Muhammad Waseem Khan
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Xiang Ma
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Guangya Xiang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| |
Collapse
|
35
|
Xu J, Wu H, Chen S, Qi B, Zhou G, Cai L, Zhao L, Wei Y, Liu S. MicroRNA-30c suppresses the pro-fibrogenic effects of cardiac fibroblasts induced by TGF-β1 and prevents atrial fibrosis by targeting TGFβRII. J Cell Mol Med 2018. [PMID: 29532993 PMCID: PMC5980214 DOI: 10.1111/jcmm.13548] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Atrial fibrosis serves as an important contributor to atrial fibrillation (AF). Recent data have suggested that microRNA-30c (miR-30c) is involved in fibrotic remodelling and cancer development, but the specific role of miR-30c in atrial fibrosis remains unclear. The purpose of this study was to investigate the role of miR-30c in atrial fibrosis and its underlying mechanisms through in vivo and in vitro experiments. Our results indicate that miR-30c is significantly down-regulated in the rat abdominal aortic constriction (AAC) model and in the cellular model of fibrosis induced by transforming growth factor-β1 (TGF-β1). Overexpression of miR-30c in cardiac fibroblasts (CFs) markedly inhibits CF proliferation, differentiation, migration and collagen production, whereas decrease in miR-30c leads to the opposite results. Moreover, we identified TGFβRII as a target of miR-30c. Finally, transferring adeno-associated virus 9 (AAV9)-miR-30c into the inferior vena cava of rats attenuated fibrosis in the left atrium following AAC. These data indicate that miR-30c attenuates atrial fibrosis via inhibition of CF proliferation, differentiation, migration and collagen production by targeting TGFβRII, suggesting that miR-30c might be a novel potential therapeutic target for preventing atrial fibrosis.
Collapse
Affiliation(s)
- Juan Xu
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haiqing Wu
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Songwen Chen
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Baozhen Qi
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Genqing Zhou
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lidong Cai
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liqun Zhao
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yong Wei
- Department of Cardiology, Shanghai Songjiang Central Hospital, Shanghai, China
| | - Shaowen Liu
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
36
|
Ma J, Ma S, Yin C, Wu H. Matrine reduces susceptibility to postinfarct atrial fibrillation in rats due to antifibrotic properties. J Cardiovasc Electrophysiol 2018; 29:616-627. [PMID: 29377366 DOI: 10.1111/jce.13448] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 12/18/2017] [Accepted: 01/08/2018] [Indexed: 12/19/2022]
Abstract
This study aimed to investigate whether matrine could prevent atrial fibrillation (AF) after myocardial infarction by reducing left atrial fibrosis, and to determine the underlying mechanisms in isolated cardiac fibroblasts (CFs). Five weeks after MI, matrine-treated rats had lower rates of AF inducibility and shorter AF duration than MI rats. Matrine improved the left atrial conduction velocity and homogeneity. Matrine decreased the fibrosis positive areas and the protein levels of type I collagen and type III collagen in the left atrium. Matrine inhibited CFs differentiation to myofibroblasts and the expression of transforming growth factor-beta 1 and matrix metalloproteinase 9. In vitro, matrine inhibited the CFs proliferation, migration, differentiation, and secretion ability. These in vitro and in vivo data demonstrated that matrine has the potential to reduce susceptibility to AF after MI due, at least in part, to reduced atrial fibrosis via inhibiting CFs proliferation, migration, differentiation, and secretion ability.
Collapse
Affiliation(s)
- Jin Ma
- Heart Center, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, PR, China
| | - Shiyu Ma
- Department of Critical-Care Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, PR, China
| | - Chunxia Yin
- Heart Center, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, PR, China
| | - Huanlin Wu
- Heart Center, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, PR, China
| |
Collapse
|
37
|
Panwar P, Xue L, Søe K, Srivastava K, Law S, Delaisse JM, Brömme D. An Ectosteric Inhibitor of Cathepsin K Inhibits Bone Resorption in Ovariectomized Mice. J Bone Miner Res 2017; 32:2415-2430. [PMID: 28745432 DOI: 10.1002/jbmr.3227] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 07/19/2017] [Accepted: 07/22/2017] [Indexed: 12/13/2022]
Abstract
The potent cathepsin K (CatK) inhibitor, Tanshinone IIA sulfonic sodium (T06), was tested for its in vitro and in vivo antiresorptive activities. T06 binds in an ectosteric site of CatK remote from its active site and selectively inhibits collagen degradation with an IC50 value of 2.7 ± 0.2 μM (CatK:T06 molar ratio of 1:5). However, it does not suppress fluorogenic peptide cleavage and gelatinolysis at a 2500-fold molar excess. Contrary to active site-directed CatK inhibitors, such as odanacatib, T06 suppresses bone resorption in both human and mouse osteoclasts equally well (IC50 value for human and mouse osteoclasts: 237 ± 60 nM and 245 ± 55 nM, respectively) and its antiresorptive activity is fully reversible in both cell types. Moreover, T06 affects neither the metabolic activity of osteoclasts nor osteoclastogenesis. In in vivo studies, 40 mg T06/kg/d given to 12-week-old ovariectomized (OVX) mice for 3 months reduced plasma CTx-1 by 20% and increased osteoblast numbers and plasma P1NP by ∼28% when compared with the OVX control. μCT analysis of T06-treated OVX mice showed a 35% increase in bone mineral density and other femoral trabecular bone parameters when compared with OVX animals. T06 did not alter the number of osteoclasts, had no estrogenic effect on the uterus, did not change plasma estradiol levels, and did not inhibit fibroblast-mediated TGF-ß1 processing or degradation and cognitive functions in OVX mice. This study indicates that the ectosteric inhibitor, T06, is a selective antiresorptive CatK inhibitor that may overcome the shortcomings of side effect-prone active site-directed drugs, which all failed in clinical trials. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Preety Panwar
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, Canada.,Clinical Cell Biology, Vejle Hospital/Lillebaelt Hospital, Institute of Regional Health Research, University of Southern Denmark, Vejle, Denmark.,Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Liming Xue
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, Canada
| | - Kent Søe
- Clinical Cell Biology, Vejle Hospital/Lillebaelt Hospital, Institute of Regional Health Research, University of Southern Denmark, Vejle, Denmark
| | - Kamini Srivastava
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, Canada
| | - Simon Law
- Centre for Blood Research, University of British Columbia, Vancouver, Canada.,Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Jean-Marie Delaisse
- Clinical Cell Biology, Vejle Hospital/Lillebaelt Hospital, Institute of Regional Health Research, University of Southern Denmark, Vejle, Denmark
| | - Dieter Brömme
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, Canada.,Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| |
Collapse
|
38
|
Oltean-Péter B, Korodi S, Benedek I, Lázár E, Kéri J, Pakucs A, Kovács I, Bordi L, Mitre A, Benedek I, Benedek T, Benedek I. Imaging-derived Biomarkers Associated with Atrial FIBROsis, Structural Remodeling and the Risk of Cardioembolic Events in Patients with Atrial Fibrillation – the FIBROS Study. JOURNAL OF INTERDISCIPLINARY MEDICINE 2017. [DOI: 10.1515/jim-2017-0095] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Abstract
Recent studies demonstrated that despite restoration of the sinus rhythm, patients with a positive history of atrial fibrillation (AF) are still at risk of thromboembolic events. The primary objective of this study is to identify new imaging-derived biomarkers provided by modern imaging technologies, such as cardiac computed tomography angiography, delayed enhancement magnetic resonance imaging, or speckle tracking echocardiography, as well as hematological biomarkers, associated with the risk of intracavitary thrombosis in patients with AF, in order to identify the imaging-derived characteristics associated with an increased risk of cardioembolic events. Imaging data collected will be post-processed using advanced techniques of computational modeling, in order to fully characterize the degree of structural remodeling and the amount of atrial fibrosis. The primary endpoint of the study is represented by the rate of thromboembolic events. The rate of cardiovascular death, the rate of major adverse cardiovascular events, and the rate of AF recurrence will also be determined in relation to the degree of structural remodeling and atrial fibrosis.
Collapse
Affiliation(s)
- Balázs Oltean-Péter
- University of Medicine and Pharmacy , Tîrgu Mureș , Romania
- Clinic of Hematology and Bone Marrow Transplantation Unit , Tîrgu Mureș , Romania
| | - Szilamér Korodi
- University of Medicine and Pharmacy , Tîrgu Mureș , Romania
- Center of Advanced Research in Multimodality Cardiac Imaging, Cardio Med Medical Center , Tîrgu Mureș , Romania
| | - István Benedek
- University of Medicine and Pharmacy , Tîrgu Mureș , Romania
- Clinic of Hematology and Bone Marrow Transplantation Unit , Tîrgu Mureș , Romania
| | - Erzsébet Lázár
- University of Medicine and Pharmacy , Tîrgu Mureș , Romania
- Clinic of Hematology and Bone Marrow Transplantation Unit , Tîrgu Mureș , Romania
| | - Johanna Kéri
- University of Medicine and Pharmacy , Tîrgu Mureș , Romania
- Clinic of Hematology and Bone Marrow Transplantation Unit , Tîrgu Mureș , Romania
| | - Annamária Pakucs
- University of Medicine and Pharmacy , Tîrgu Mureș , Romania
- Clinic of Hematology and Bone Marrow Transplantation Unit , Tîrgu Mureș , Romania
| | - István Kovács
- University of Medicine and Pharmacy , Tîrgu Mureș , Romania
- Center of Advanced Research in Multimodality Cardiac Imaging, Cardio Med Medical Center , Tîrgu Mureș , Romania
| | - Lehel Bordi
- University of Medicine and Pharmacy , Tîrgu Mureș , Romania
- Center of Advanced Research in Multimodality Cardiac Imaging, Cardio Med Medical Center , Tîrgu Mureș , Romania
| | - Adriana Mitre
- University of Medicine and Pharmacy , Tîrgu Mureș , Romania
| | - Imre Benedek
- University of Medicine and Pharmacy , Tîrgu Mureș , Romania
- Center of Advanced Research in Multimodality Cardiac Imaging, Cardio Med Medical Center , Tîrgu Mureș , Romania
| | - Theodora Benedek
- University of Medicine and Pharmacy , Tîrgu Mureș , Romania
- Center of Advanced Research in Multimodality Cardiac Imaging, Cardio Med Medical Center , Tîrgu Mureș , Romania
| | - István Benedek
- University of Medicine and Pharmacy , Tîrgu Mureș , Romania
- Clinic of Hematology and Bone Marrow Transplantation Unit , Tîrgu Mureș , Romania
| |
Collapse
|
39
|
Degiovanni A, Boggio E, Prenna E, Sartori C, De Vecchi F, Marino PN. Association between left atrial phasic conduit function and early atrial fibrillation recurrence in patients undergoing electrical cardioversion. Clin Res Cardiol 2017; 107:329-337. [PMID: 29181725 PMCID: PMC5869942 DOI: 10.1007/s00392-017-1188-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 11/23/2017] [Indexed: 01/08/2023]
Abstract
Background Diastolic dysfunction promotes atrial fibrillation (AF) inducing left atrial (LA) remodeling, with chamber dilation and fibrosis. Predominance of LA phasic conduit (LAC) function should reflect not only chamber alterations but also underlying left ventricular (LV) filling impairment. Thus, LAC was tested as possible predictor of early AF relapse after electrical cardioversion (EC). Methods 96 consecutive patients, who underwent EC for persistent non-valvular AF, were prospectively enrolled. Immediately after successful EC (3 h ± 15 min), an echocardiographic apical four-chamber view was acquired with transmitral velocities, annular tissue Doppler and simultaneous LV and LA three-dimensional full-volume datasets. Then, from LA–LV volumetric curves we computed LAC as: [(LV maximum − LV minimum) − (LA maximum − LA minimum) volume], expressed as % LV stroke volume. LA pump, immediately post-EC, was assumed and verified as being negligible. Sinus rhythm persistence at 1 month was checked with ECG-Holter monitoring. Results At 1 month 62 patients were in sinus rhythm and 34 in AF. AF patients presented pre-EC higher E/é values (p = 0.012), no major LA volume differences (p = NS), but a stiffer LV cavity (p = 0.012) for a comparable LV capacitance (p = 0.461). Conduit contributed more (p < 0.001) to LV stroke volume in AF subpopulation. Multiple regression revealed LAC as the most significant AF predictor (p = 0.013), even after correction for biometric characteristics and pharmacotherapy (p = 0.008). Conclusion Our data suggest that LAC larger contribution to LV filling soon after EC reflects LA–LV stiffening, which skews atrioventricular interaction leading to AF perpetuation and makes conduit dominance a powerful predictor of early AF recurrence. Electronic supplementary material The online version of this article (10.1007/s00392-017-1188-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anna Degiovanni
- Department of Translational Medicine, Clinical Cardiology, Università del Piemonte Orientale, Azienda Ospedaliera Universitaria "Maggiore della Carità", Corso Mazzini 18, 28100, Novara, Italy
| | - Enrico Boggio
- Department of Translational Medicine, Clinical Cardiology, Università del Piemonte Orientale, Azienda Ospedaliera Universitaria "Maggiore della Carità", Corso Mazzini 18, 28100, Novara, Italy
| | - Eleonora Prenna
- Department of Translational Medicine, Clinical Cardiology, Università del Piemonte Orientale, Azienda Ospedaliera Universitaria "Maggiore della Carità", Corso Mazzini 18, 28100, Novara, Italy
| | - Chiara Sartori
- Department of Translational Medicine, Clinical Cardiology, Università del Piemonte Orientale, Azienda Ospedaliera Universitaria "Maggiore della Carità", Corso Mazzini 18, 28100, Novara, Italy
| | - Federica De Vecchi
- Department of Translational Medicine, Clinical Cardiology, Università del Piemonte Orientale, Azienda Ospedaliera Universitaria "Maggiore della Carità", Corso Mazzini 18, 28100, Novara, Italy
| | - Paolo N Marino
- Department of Translational Medicine, Clinical Cardiology, Università del Piemonte Orientale, Azienda Ospedaliera Universitaria "Maggiore della Carità", Corso Mazzini 18, 28100, Novara, Italy.
| | | |
Collapse
|
40
|
Zhang J, Feng C, Song C, Ai B, Bai X, Liu Y, Li X, Zhao J, Shi S, Chen X, Su X, Li C. Identification and analysis of a key long non-coding RNAs (lncRNAs)-associated module reveal functional lncRNAs in cardiac hypertrophy. J Cell Mol Med 2017; 22:892-903. [PMID: 29154475 PMCID: PMC5783834 DOI: 10.1111/jcmm.13376] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 08/14/2017] [Indexed: 01/28/2023] Open
Abstract
Cardiac hypertrophy (CH) is a common disease that originates from long-term heart pressure overload and finally leads to heart failure. Recently, long non-coding RNAs (lncRNAs) have attracted attention because they have broad and crucial functions in regulating complex biological processes. Some studies had found that lncRNAs play vital roles in complex cardiovascular diseases. However, the function and mechanism of lncRNAs in CH have not been elucidated. In our study, to investigate the potential roles of lncRNAs in CH, the Cardiac Hypertrophy-associated LncRNAs-Protein coding genes Network (CHLPN) was constructed by integrating gene microarray re-annotation and subpathway enrichment analyses. After performing random walking with restart in CHLPN, we predicted 21 significant risk lncRNAs, of which 7 (Kis2, 1700110K17Rik, Gm17501, E330017L17Rik, C630043F03Rik, Gm9866 and Ube4bos1) formed a close module with their co-expressed protein-coding genes (PCGs). We found that the module might play crucial roles in the development of CH. In particular, 44 PCGs that were co-expressed with six lncRNAs were enriched in CH-related biological processes and pathways. We also found that some lncRNAs participated in the competitive endogenous RNA cross-talk that might be involved in CH. These results indicate that the functional lncRNAs are related to post-transcriptional regulation and could shed light on a new molecular diagnostic target of CH.
Collapse
Affiliation(s)
- Jian Zhang
- School of Medical Informatics, Harbin Medical University, Daqing, China
| | - Chenchen Feng
- School of Medical Informatics, Harbin Medical University, Daqing, China
| | - Chao Song
- Department of Pharmacology, Harbin Medical University, Daqing, China
| | - Bo Ai
- School of Medical Informatics, Harbin Medical University, Daqing, China
| | - Xuefeng Bai
- School of Medical Informatics, Harbin Medical University, Daqing, China
| | - Yuejuan Liu
- School of Medical Informatics, Harbin Medical University, Daqing, China
| | - Xuecang Li
- School of Medical Informatics, Harbin Medical University, Daqing, China
| | - Jianmei Zhao
- School of Medical Informatics, Harbin Medical University, Daqing, China
| | - Shengshu Shi
- College of Computer Science and Technology, Heilongjiang University, Harbin Medical University, Harbin, China
| | - Xin Chen
- College Food and Biological Engineering, Jimei University, Xiamen, China
| | - Xiaojie Su
- College of Medical Laboratory Science and Technology, Harbin Medical University, Daqing, China
| | - Chunquan Li
- School of Medical Informatics, Harbin Medical University, Daqing, China
| |
Collapse
|
41
|
Zhao S, Li M, Ju W, Gu L, Zhang F, Chen H, Gu K, Yang B, Chen M. Serum level of transforming growth factor beta 1 is associated with left atrial voltage in patients with chronic atrial fibrillation. Indian Pacing Electrophysiol J 2017; 18:95-99. [PMID: 29155027 PMCID: PMC5986266 DOI: 10.1016/j.ipej.2017.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 11/12/2017] [Accepted: 11/14/2017] [Indexed: 11/26/2022] Open
Abstract
Background Atrial tissue fibrosis can cause electrical or structural remodeling in patients with atrial fibrillation. Transforming growth factor beta 1(TGF-β1) signaling acts as a central role in fibroblast activation. In this report, we aimed to investigate the relationship between serum level of TGF-β1 and mean left atrial voltage in patients with chronic atrial fibrillation (CAF). Methods A total of 16 consecutive adult patients with CAF who underwent catheter ablation were enrolled. Blood samples for measurement of TGF-β1 were collected from periphery veins and coronary sinus before pulmonary vein isolation. The measurement was performed with a commercially available ELISA kit. Cardiac indices were measured using echocardiography. The left atrial electroanatomic mapping was performed after pulmonary vein isolation. Results Serum level of TGF-β1 in peripheral blood was higher than that in coronary sinus (p < 0.001). TGF-β1 serum level in coronary sinus negatively correlated with mean left atrial voltage (r = -0.650, p = 0.012), While periphery TGF-β1 level tended to be negatively correlated with mean left atrial voltage(r = -0.492, p = 0.053). Patients who treated with angiotensin II receptor antagonists had lower coronary sinus TGF-β1 serum level than those who did not treated with angiotensin II receptor antagonists (p = 0.046). Conclusion Level of TGF-β1 in peripheral serum is higher than that in coronary sinus, and serum level of TGF-β1 in coronary sinus is negatively associated with mean left atrial voltage in patients with CAF, angiotensin II receptor antagonists could affect TGF-β1 serum level.
Collapse
Affiliation(s)
- Shilu Zhao
- Cardiology Division, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Mingfang Li
- Cardiology Division, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Weizhu Ju
- Cardiology Division, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Lingyun Gu
- Cardiology Division, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Fengxiang Zhang
- Cardiology Division, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Hongwu Chen
- Cardiology Division, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Kai Gu
- Cardiology Division, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Bing Yang
- Cardiology Division, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Minglong Chen
- Cardiology Division, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China.
| |
Collapse
|
42
|
Ghavami S, Yeganeh B, Zeki AA, Shojaei S, Kenyon NJ, Ott S, Samali A, Patterson J, Alizadeh J, Moghadam AR, Dixon IMC, Unruh H, Knight DA, Post M, Klonisch T, Halayko AJ. Autophagy and the unfolded protein response promote profibrotic effects of TGF-β 1 in human lung fibroblasts. Am J Physiol Lung Cell Mol Physiol 2017; 314:L493-L504. [PMID: 29074489 DOI: 10.1152/ajplung.00372.2017] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a lethal fibrotic lung disease in adults with limited treatment options. Autophagy and the unfolded protein response (UPR), fundamental processes induced by cell stress, are dysregulated in lung fibroblasts and epithelial cells from humans with IPF. Human primary cultured lung parenchymal and airway fibroblasts from non-IPF and IPF donors were stimulated with transforming growth factor-β1 (TGF-β1) with or without inhibitors of autophagy or UPR (IRE1 inhibitor). Using immunoblotting, we monitored temporal changes in abundance of protein markers of autophagy (LC3βII and Atg5-12), UPR (BIP, IRE1α, and cleaved XBP1), and fibrosis (collagen 1α2 and fibronectin). Using fluorescent immunohistochemistry, we profiled autophagy (LC3βII) and UPR (BIP and XBP1) markers in human non-IPF and IPF lung tissue. TGF-β1-induced collagen 1α2 and fibronectin protein production was significantly higher in IPF lung fibroblasts compared with lung and airway fibroblasts from non-IPF donors. TGF-β1 induced the accumulation of LC3βII in parallel with collagen 1α2 and fibronectin, but autophagy marker content was significantly lower in lung fibroblasts from IPF subjects. TGF-β1-induced collagen and fibronectin biosynthesis was significantly reduced by inhibiting autophagy flux in fibroblasts from the lungs of non-IPF and IPF donors. Conversely, only in lung fibroblasts from IPF donors did TGF-β1 induce UPR markers. Treatment with an IRE1 inhibitor decreased TGF-β1-induced collagen 1α2 and fibronectin biosynthesis in IPF lung fibroblasts but not those from non-IPF donors. The IRE1 arm of the UPR response is uniquely induced by TGF-β1 in lung fibroblasts from human IPF donors and is required for excessive biosynthesis of collagen and fibronectin in these cells.
Collapse
Affiliation(s)
- Saeid Ghavami
- Department of Human Anatomy and Cell Science, University of Manitoba, Manitoba, Canada.,Biology of Breathing Group, Children's Hospital Research Institute of Manitoba, Manitoba, Canada
| | - Behzad Yeganeh
- Biology of Breathing Group, Children's Hospital Research Institute of Manitoba, Manitoba, Canada.,Department of Physiology and Pathophysiology, University of Manitoba, Manitoba, Canada.,Department of Physiology and Experimental Medicine, University of Toronto , Toronto , Canada.,Hospital for Sick Children Research Institute , Toronto , Canada
| | - Amir A Zeki
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of California , Davis, California
| | - Shahla Shojaei
- Department of Human Anatomy and Cell Science, University of Manitoba, Manitoba, Canada
| | - Nicholas J Kenyon
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of California , Davis, California
| | - Sean Ott
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of California , Davis, California
| | - Afshin Samali
- Apoptosis Research Centre, National University of Ireland , Galway , Ireland
| | | | - Javad Alizadeh
- Department of Human Anatomy and Cell Science, University of Manitoba, Manitoba, Canada.,Biology of Breathing Group, Children's Hospital Research Institute of Manitoba, Manitoba, Canada
| | - Adel Rezaei Moghadam
- Department of Human Anatomy and Cell Science, University of Manitoba, Manitoba, Canada.,Biology of Breathing Group, Children's Hospital Research Institute of Manitoba, Manitoba, Canada
| | - Ian M C Dixon
- Department of Physiology and Pathophysiology, University of Manitoba, Manitoba, Canada.,St. Boniface Research Centre , Winnipeg , Canada
| | - Helmut Unruh
- Department of Internal Medicine, University of Manitoba, Manitoba, Canada
| | - Darryl A Knight
- School of Biomedical Science and Pharmacy, University of Newcastle , Newcastle , Australia
| | - Martin Post
- Department of Physiology and Experimental Medicine, University of Toronto , Toronto , Canada.,Hospital for Sick Children Research Institute , Toronto , Canada
| | - Thomas Klonisch
- Department of Human Anatomy and Cell Science, University of Manitoba, Manitoba, Canada
| | - Andrew J Halayko
- Biology of Breathing Group, Children's Hospital Research Institute of Manitoba, Manitoba, Canada.,Department of Physiology and Pathophysiology, University of Manitoba, Manitoba, Canada.,St. Boniface Research Centre , Winnipeg , Canada
| |
Collapse
|
43
|
Dogan A, Parmaksız M, Elçin AE, Elçin YM. Extracellular Matrix and Regenerative Therapies from the Cardiac Perspective. Stem Cell Rev Rep 2017; 12:202-13. [PMID: 26668014 DOI: 10.1007/s12015-015-9641-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cardiovascular diseases are the leading cause of death and a major cause of financial burden. Regenerative therapies for heart diseases bring the promise of alternative treatment modalities for myocardial infarction, ischemic heart disease, and congestive heart failure. Although, clinical trials attest to the safety of stem cell injection therapies, researchers need to overcome the underlying mechanisms that are limiting the success of future regenerative options. This article aims to review the basic scientific concepts in the field of mechanobiology and the effects of extracellular functions on stem cell fate.
Collapse
Affiliation(s)
- Arin Dogan
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science, and Ankara University Stem Cell Institute, Degol Caddesi, Tandogan, 06100, Ankara, Turkey
| | - Mahmut Parmaksız
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science, and Ankara University Stem Cell Institute, Degol Caddesi, Tandogan, 06100, Ankara, Turkey
| | - A Eser Elçin
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science, and Ankara University Stem Cell Institute, Degol Caddesi, Tandogan, 06100, Ankara, Turkey
| | - Y Murat Elçin
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science, and Ankara University Stem Cell Institute, Degol Caddesi, Tandogan, 06100, Ankara, Turkey.
| |
Collapse
|
44
|
Kim N, Jung Y, Nam M, Sun Kang M, Lee MK, Cho Y, Choi EK, Hwang GS, Soo Kim H. Angiotensin II affects inflammation mechanisms via AMPK-related signalling pathways in HL-1 atrial myocytes. Sci Rep 2017; 7:10328. [PMID: 28871102 PMCID: PMC5583339 DOI: 10.1038/s41598-017-09675-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 07/27/2017] [Indexed: 11/30/2022] Open
Abstract
Inflammation is a common cause of cardiac arrhythmia. Angiotensin ІІ (Ang ІІ) is a major contributing factor in the pathogenesis of cardiac inflammation; however, its underlying molecular mechanism remains unclear. Here, we explored the effect of Ang ІІ on inflammatory mechanisms and oxidative stress using HL-1 atrial myocytes. We showed that Ang ІІ activated c-Jun N-terminal kinase (JNK) phosphorylation and other inflammatory markers, such as transforming growth factor-β1 (TGF-β1) and tumor necrosis factor-α (TNF-α). Ang ІІ decreased oxygen consumption rate, which resulted in reactive oxygen species (ROS) generation and inhibition of ROS blocked Ang II-mediated JNK phosphorylation and TGF-β1 induction. Ang ІІ induced the expression of its specific receptor, AT1R. Ang II-induced intracellular calcium production associated with Ang ІІ-mediated signalling pathways. In addition, the generated ROS and calcium stimulated AMPK phosphorylation. Inhibiting AMPK blocked Ang II-mediated JNK and TGF-β signalling pathways. Ang ІІ concentration, along with TGF-β1 and tumor necrosis factor-α levels, was slightly increased in plasma of patients with atrial fibrillation. Taken together, these results suggest that Ang ІІ induces inflammation mechanisms through an AMPK-related signalling pathway. Our results provide new molecular targets for the development of therapeutics for inflammation-related conditions, such as atrial fibrillation.
Collapse
Affiliation(s)
- Nami Kim
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, 120-140, Republic of Korea
| | - Youngae Jung
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, 120-140, Republic of Korea
| | - Miso Nam
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, 120-140, Republic of Korea
| | - Mi Sun Kang
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, 120-140, Republic of Korea
| | - Min Kyung Lee
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, 120-140, Republic of Korea
| | - Youngjin Cho
- Division of Cardiology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Eue-Keun Choi
- Division of Cardiology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Geum-Sook Hwang
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, 120-140, Republic of Korea. .,Chemistry & Nanoscience, Ewha Womans University, Seoul, Republic of Korea.
| | - Hyeon Soo Kim
- Department of Anatomy, Korea University College of Medicine, Seoul, 02841, Republic of Korea.
| |
Collapse
|
45
|
Guo F, Yi X, Li M, Fu J, Li S. Snail1 is positively correlated with atrial fibrosis in patients with atrial fibrillation and rheumatic heart disease. Exp Ther Med 2017; 14:4231-4237. [PMID: 29104639 PMCID: PMC5658748 DOI: 10.3892/etm.2017.5084] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 06/05/2017] [Indexed: 02/07/2023] Open
Abstract
The present study investigated the association between Snail1 and atrial fibrosis in patients with atrial fibrillation (AF) and rheumatic heart disease (RHD) and to determine the possible mechanism underlying this interrelation. A total of 19 patients were included in the current study and were divided into two groups: A sinus rhythm (SR) group (n=9) and an AF group (n=10). All patients underwent heart valve replacement surgery, during which ~200 mg right atrium tissue was obtained. Hematoxylin and eosin and Masson's trichrome-stained sections were used to evaluate the morphological changes of cardiomyocytes and the level of fibrosis. Immunohistochemistry was applied to observe the location and expression of Snail1. Reverse transcription-quantitative polymerase chain reaction was used to measure Snail1 mRNA levels. Western blotting was used to determine changes in the expression of Snail1, as well as in the expression of proteins involved in the Wnt pathway, including Wnt1, Wnt 3a, Wnt8a, Wnt5a and Wnt11. Compared with the SR group, expanded cardiomyocytes and higher collagen deposition was detected in the atrial tissue of the AF group. The expression of Snail1 mRNA and protein was significantly higher in the AF group than in the SR group (P<0.05). Additionally, the expression of Wnt1, 3a and 8a in the canonical Wnt signaling pathway, and Wnt5a and 11 in the noncanonical Wnt signaling pathway were significantly increased in the AF group. Furthermore, the phosphorylation level of glycogen synthase kinase 3β (GSK3β) and the levels of β-catenin and GSK3β were significantly increased in the AF group compared with the SR group (P<0.05). Snail1 may be involved in the development and maintenance of atrial fibrosis in patients with atrial fibrillation and rheumatic heart disease and may be developed as a novel biomarker to evaluate myocardial fibrosis in the future. Additionally, the current study suggests that the Wnt signaling pathway may participate in the process of increased Snail1 expression and atrial fibrosis in patients with AF and RHD.
Collapse
Affiliation(s)
- Furong Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China.,Cardiovascular Research Institute, Wuhan, Hubei 430060, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xin Yi
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China.,Cardiovascular Research Institute, Wuhan, Hubei 430060, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Mingjiang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China.,Cardiovascular Research Institute, Wuhan, Hubei 430060, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jinrong Fu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China.,Cardiovascular Research Institute, Wuhan, Hubei 430060, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Sha Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China.,Cardiovascular Research Institute, Wuhan, Hubei 430060, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
46
|
Colombero C, Papademetrio D, Sacca P, Mormandi E, Alvarez E, Nowicki S. Role of 20-Hydroxyeicosatetraenoic Acid (20-HETE) in Androgen-Mediated Cell Viability in Prostate Cancer Cells. HORMONES & CANCER 2017; 8:243-256. [PMID: 28639228 PMCID: PMC10355871 DOI: 10.1007/s12672-017-0299-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Accepted: 06/05/2017] [Indexed: 01/18/2023]
Abstract
20-Hydroxyeicosatetraenoic acid (20-HETE) is generated intracellularly through the ω-hydroxylation of arachidonic acid by the cytochrome P450 (in humans, CYP4A11 and CYP4F2). 20-HETE induces mitogenic responses in different cancer cells. The aim of this study was to analyze how 20-HETE impacts cell survival, proliferation, and apoptosis in prostate cancer cells. Incubation of the human androgen-sensitive cells (LNCaP) with 1-10 μM HET0016 (a selective inhibitor of 20-HETE synthesis) reduced cell viability by 49*-64%* (*p < 0.05 vs. control). This was explained by a reduction in cell proliferation (vehicle, 46 ± 3%; 1 μM, 23 ± 3%*; 10 μM, 28 ± 3%*) and by an increase in apoptosis (vehicle, 2.1 ± 0%; 1 μM, 16 ± 4%*; 10 μM, 31 ± 3%*). Furthermore, the increase in LNCaP cell viability induced by dihydrotestosterone (DHT, 0.1 nM) was abrogated by 30*-42%* by 1-10 μM HET0016. Incubation with 20-HETE (5-1000 nM) increased LNCaP cell viability up to 50%*, together with a 70%* reduction in apoptosis. PC-3 (androgen-insensitive) cell viability was not affected by either HET0016 or 20-HETE. In LNCaP cells, HET0016 (10 μM) diminished the expression of androgen receptors (AR): messenger RNA (mRNA) (40%*) and protein (50%*). DHT (10 nM) augmented CYP4F2 protein expression (1.9-fold*) and 20-HETE levels (50%*). Oppositely, enzalutamide (AR antagonist) reduced CYP4F2 mRNA and protein expressions by 30 and 25%, respectively. Thus, intracellular availability of 20-HETE is necessary to sustain LNCaP cell viability. 20-HETE may act as a signaling molecule in the pathways involved in LNCaP cell viability upon stimulation of the AR. This effect may be partially attributed to its role on securing normal AR expression levels that in turn contribute to maintain intracellular levels of 20-HETE.
Collapse
Affiliation(s)
- Cecilia Colombero
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Fundación de Endocrinología Infantil, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Gallo 1360, C1425EFD, Buenos Aires, Argentina
| | - Daniela Papademetrio
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquimica - Instituto de Estudios de la Inmunidad Humoral Prof Ricardo Margni. Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Junin 954, C1113AAD, Buenos Aires, Argentina
| | - Paula Sacca
- Instituto de Biología y Medicina Experimental (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Eduardo Mormandi
- Laboratorio de Endocrinología, División Endocrinología, Hospital Carlos G. Durand, Av. Díaz Vélez 5044, C1405DCS, Buenos Aires, Argentina
| | - Elida Alvarez
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquimica - Instituto de Estudios de la Inmunidad Humoral Prof Ricardo Margni. Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Junin 954, C1113AAD, Buenos Aires, Argentina
| | - Susana Nowicki
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Fundación de Endocrinología Infantil, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Gallo 1360, C1425EFD, Buenos Aires, Argentina.
| |
Collapse
|
47
|
Maheshwari A, Norby FL, Soliman EZ, Koene RJ, Rooney MR, O'Neal WT, Alonso A, Chen LY. Abnormal P-Wave Axis and Ischemic Stroke: The ARIC Study (Atherosclerosis Risk In Communities). Stroke 2017; 48:2060-2065. [PMID: 28626057 PMCID: PMC5534350 DOI: 10.1161/strokeaha.117.017226] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 05/12/2017] [Accepted: 05/22/2017] [Indexed: 01/12/2023]
Abstract
BACKGROUND AND PURPOSE Abnormal P-wave axis (aPWA) has been linked to incident atrial fibrillation and mortality; however, the relationship between aPWA and stroke has not been reported. We hypothesized that aPWA is associated with ischemic stroke independent of atrial fibrillation and other stroke risk factors and tested our hypothesis in the ARIC study (Atherosclerosis Risk In Communities), a community-based prospective cohort study. METHODS We included 15 102 participants (aged 54.2±5.7 years; 55.2% women; 26.5% blacks) who attended the baseline examination (1987-1989) and without prevalent stroke. We defined aPWA as any value outside 0 to 75° using 12-lead ECGs obtained during study visits. Each case of incident ischemic stroke was classified in accordance with criteria from the National Survey of Stroke by a computer algorithm and adjudicated by physician review. Multivariable Cox regression was used to estimate hazard ratios and 95% confidence intervals for the association of aPWA with stroke. RESULTS During a mean follow-up of 20.2 years, there were 657 incident ischemic stroke cases. aPWA was independently associated with a 1.50-fold (95% confidence interval, 1.22-1.85) increased risk of ischemic stroke in the multivariable model that included atrial fibrillation. When subtyped, aPWA was associated with a 2.04-fold (95% confidence interval, 1.42-2.95) increased risk of cardioembolic stroke and a 1.32-fold (95% confidence interval, 1.03-1.71) increased risk of thrombotic stroke. CONCLUSIONS aPWA is independently associated with ischemic stroke. This association seems to be stronger for cardioembolic strokes. Collectively, our findings suggest that alterations in atrial electric activation may predispose to cardiac thromboembolism independent of atrial fibrillation.
Collapse
Affiliation(s)
- Ankit Maheshwari
- Cardiovascular Division, Department of Medicine (A.M., R.K., L.Y.C.) and Division of Epidemiology and Community Health, School of Public Health (F.L.N., M.R.R.), University of Minnesota, Minneapolis; Epidemiological Cardiology Research Center (EPICARE), Wake Forest School of Medicine, Winston-Salem, NC (E.Z.S.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (W.T.O.); and Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA (A.A.).
| | - Faye L Norby
- Cardiovascular Division, Department of Medicine (A.M., R.K., L.Y.C.) and Division of Epidemiology and Community Health, School of Public Health (F.L.N., M.R.R.), University of Minnesota, Minneapolis; Epidemiological Cardiology Research Center (EPICARE), Wake Forest School of Medicine, Winston-Salem, NC (E.Z.S.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (W.T.O.); and Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA (A.A.)
| | - Elsayed Z Soliman
- Cardiovascular Division, Department of Medicine (A.M., R.K., L.Y.C.) and Division of Epidemiology and Community Health, School of Public Health (F.L.N., M.R.R.), University of Minnesota, Minneapolis; Epidemiological Cardiology Research Center (EPICARE), Wake Forest School of Medicine, Winston-Salem, NC (E.Z.S.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (W.T.O.); and Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA (A.A.)
| | - Ryan J Koene
- Cardiovascular Division, Department of Medicine (A.M., R.K., L.Y.C.) and Division of Epidemiology and Community Health, School of Public Health (F.L.N., M.R.R.), University of Minnesota, Minneapolis; Epidemiological Cardiology Research Center (EPICARE), Wake Forest School of Medicine, Winston-Salem, NC (E.Z.S.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (W.T.O.); and Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA (A.A.)
| | - Mary R Rooney
- Cardiovascular Division, Department of Medicine (A.M., R.K., L.Y.C.) and Division of Epidemiology and Community Health, School of Public Health (F.L.N., M.R.R.), University of Minnesota, Minneapolis; Epidemiological Cardiology Research Center (EPICARE), Wake Forest School of Medicine, Winston-Salem, NC (E.Z.S.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (W.T.O.); and Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA (A.A.)
| | - Wesley T O'Neal
- Cardiovascular Division, Department of Medicine (A.M., R.K., L.Y.C.) and Division of Epidemiology and Community Health, School of Public Health (F.L.N., M.R.R.), University of Minnesota, Minneapolis; Epidemiological Cardiology Research Center (EPICARE), Wake Forest School of Medicine, Winston-Salem, NC (E.Z.S.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (W.T.O.); and Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA (A.A.)
| | - Alvaro Alonso
- Cardiovascular Division, Department of Medicine (A.M., R.K., L.Y.C.) and Division of Epidemiology and Community Health, School of Public Health (F.L.N., M.R.R.), University of Minnesota, Minneapolis; Epidemiological Cardiology Research Center (EPICARE), Wake Forest School of Medicine, Winston-Salem, NC (E.Z.S.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (W.T.O.); and Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA (A.A.)
| | - Lin Y Chen
- Cardiovascular Division, Department of Medicine (A.M., R.K., L.Y.C.) and Division of Epidemiology and Community Health, School of Public Health (F.L.N., M.R.R.), University of Minnesota, Minneapolis; Epidemiological Cardiology Research Center (EPICARE), Wake Forest School of Medicine, Winston-Salem, NC (E.Z.S.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (W.T.O.); and Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA (A.A.)
| |
Collapse
|
48
|
The crucial role of activin A/ALK4 pathway in the pathogenesis of Ang-II-induced atrial fibrosis and vulnerability to atrial fibrillation. Basic Res Cardiol 2017. [PMID: 28639003 DOI: 10.1007/s00395-017-0634-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Atrial fibrosis, the hallmark of structural remodeling associated with atrial fibrillation (AF), is characterized by abnormal proliferation of atrial fibroblasts and excessive deposition of extracellular matrix. Transforming growth factor-β1 (TGF-β1)/activin receptor-like kinase 5 (ALK5)/Smad2/3/4 pathway has been reported to be involved in the process. Recent studies have implicated both activin A and its specific downstream component activin receptor-like kinase 4 (ALK4) in stimulating fibrosis in non-cardiac organs. We recently reported that ALK4 haplodeficiency attenuated the pressure overload- and myocardial infarction-induced ventricular fibrosis. However, the role of activin A/ALK4 in the pathogenesis of atrial fibrosis and vulnerability to AF remains unknown. Our study provided experimental and clinical evidence for the involvement of activin A and ALK4 in the pathophysiology of atrial fibrosis and AF. Patients with AF had higher activin A and ALK4 expression in atriums as compared to individuals devoid of AF. After angiotensin-II (Ang-II) stimulation which mimicked atrial fibrosis progression, ALK4-deficient mice showed lower expression of ALK4 in atriums, reduced activation of atrial fibroblasts, blunted atrial enlargement and atrial fibrosis, and further reduced AF vulnerability upon right atrial electrophysiological studies as compared to wild-type littermates. Moreover, we found that apart from the well-known TGF-β1/ALK5 pathway, the activation of activin A/ALK4/smad2/3 pathway played an important role in the pathogenesis of Ang-II-mediated atrial fibrosis and inducibility of AF, suggesting that targeting ALK4 might be a potential therapy for atrial fibrosis and AF.
Collapse
|
49
|
Role of the MAPKs/TGF-β1/TRAF6 signaling pathway in postoperative atrial fibrillation. PLoS One 2017; 12:e0173759. [PMID: 28323847 PMCID: PMC5360308 DOI: 10.1371/journal.pone.0173759] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Accepted: 02/27/2017] [Indexed: 12/25/2022] Open
Abstract
Objectives To explore the relationship between the MAPKs/TGF-β1/TRAF6 signaling pathway and atrial fibrosis in patients with rheumatic heart disease (RHD) and its role in atrial fibrillation (AF) after cardiac surgery on the basis of our previous animal study of the MAPKs/TGF-β1/TRAF6 signaling pathway in atrial fibrosis. Methods A total of 57 patients with RHD without a history of AF consented to left atrial biopsy. Histopathology quantified the percentage of fibrosis, and real-time PCR and western blot assessed the mRNA and protein expression of TGF-β1, TRAF6, and connective tissue growth factor (CTGF), respectively. Western blot was also used to measure the protein expression of phosphorylated MAPKs and TGF-β-activated kinase 1 (TAK1). Serum angiotensin II (Ang II) levels were assayed using enzyme-linked immunosorbent assay (ELISA). Results Eighteen patients developed AF, whereas 39 remained in sinus rhythm (SR). The severity of atrial fibrosis was significantly higher in patients who developed AF versus those who remained in SR; the mRNA and protein expression of TGF-β1, TRAF6 and CTGF were significantly higher in patients with AF. The protein expression of phosphorylated MAPKs and TAK1 was significantly increased in patients who developed AF compared with the patients who remained in SR. Serum Ang II levels were significantly higher in patients who developed AF versus those who remained in SR. Conclusion The MAPKs/TGF-β1/TRAF6 signaling pathway is involved in atrial fibrosis in patients with RHD, which results in the occurrence of AF after cardiac surgery.
Collapse
|
50
|
Lee SL, Daimon M, Nakao T, Singer DE, Shinozaki T, Kawata T, Kimura K, Hirokawa M, Kato TS, Mizuno Y, Watanabe M, Yatomi Y, Yamazaki T, Komuro I. Factors influencing left atrial volume in a population with preserved ejection fraction: Left ventricular diastolic dysfunction or clinical factors? J Cardiol 2016; 68:275-81. [DOI: 10.1016/j.jjcc.2016.02.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 01/16/2016] [Accepted: 02/02/2016] [Indexed: 11/29/2022]
|