1
|
Basiouny SM, Zaki HF, Elshazly SM, Mohamed AF. Berberine ameliorates seizure activity and cardiac dysfunction in pentylenetetrazol-kindling seizures in rats: Modulation of sigma1 receptor, Akt/eNOS signaling, and ferroptosis. Neuropharmacology 2025; 267:110295. [PMID: 39800083 DOI: 10.1016/j.neuropharm.2025.110295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 01/03/2025] [Accepted: 01/03/2025] [Indexed: 01/15/2025]
Abstract
Seizures can lead to cardiac dysfunction. Multiple pathways contribute to this phenomenon, of which the chaperone sigma-1 receptor (S1R) signaling represents a promising nexus between the abnormalities seen in both epilepsy and ensuing cardiac complications. The study explored the potential of Berberine (BER), a promising S1R agonist, in treating epilepsy and associated cardiac abnormalities in a pentylenetetrazol (PTZ) kindling rat model of epilepsy. Male Wistar albino rats received PTZ (35 mg/kg) every other day alone, with BER, with phenytoin (PHT), with both BER and PHT and with both BER and an S1R blocker (NE-100) over 27 days. BER decreased seizure severity and improved hemodynamic parameters. Histopathological abnormalities were more pronounced in the PTZ, and blocker group than in other groups, in heart tissue. In cardiac tissue, BER enhanced the AKT/eNOS signaling pathway and mitigated ferroptosis by boosting the cystine/glutamate transporter/Glutathione/Glutathione Peroxidase 4 (XCT/GSH/GPX4) system and ferritin heavy chain-1 (FTH-1) expression, while reducing iron and Transferrin receptor protein 1 (TFR1) levels. Such effects were largely negated by NE-100 pretreatment. In conclusion, BER shows protective effects on cardiac dysfunction induced by the PTZ kindling model by acting as an S1R agonist and influencing the AKT/eNOS signaling pathway and ferroptosis.
Collapse
Affiliation(s)
- Shrouk M Basiouny
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Sinai University, Egypt.
| | - Hala F Zaki
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Shimaa M Elshazly
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Zagazig University, Egypt
| | - Ahmed F Mohamed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt; Pharmacology and Toxicology Department, Faculty of Pharmacy, King Salman International University (KSIU), South Sinai, 46612, Egypt
| |
Collapse
|
2
|
Zheng E, Warchoł I, Mejza M, Możdżan M, Strzemińska M, Bajer A, Madura P, Żak J, Plewka M. Exploring Anti-Inflammatory Treatment as Upstream Therapy in the Management of Atrial Fibrillation. J Clin Med 2025; 14:882. [PMID: 39941553 PMCID: PMC11818443 DOI: 10.3390/jcm14030882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/05/2025] [Accepted: 01/14/2025] [Indexed: 02/16/2025] Open
Abstract
Inflammation has been widely recognized as one of the major pathophysiological drivers of the development of atrial fibrillation (AF), which works in tandem with other risk factors of AF including obesity, diabetes, hypertension, and heart failure (HF). Our current understanding of the role of inflammation in the natural history of AF remains elusive; however, several key players, including the NLRP3 (NLR family pyrin domain containing 3) inflammasome, have been acknowledged to be heavily influential on chronic inflammation in the atrial myocardium, which leads to fibrosis and eventual degradation of its electrical function. Nevertheless, our current methods of pharmacological modalities with reported immunomodulatory properties, including well-established classes of drugs e.g., drugs targeting the renin-angiotensin-aldosterone system (RAAS), statins, and vitamin D, have proven effective in reducing the overall risk of developing AF, the onset of postoperative atrial fibrillation (POAF), and reducing overall mortality among patients with AF. This might bring hope for further progress in developing new treatment modalities targeting cellular checkpoints of the NLRP3 inflammasome pathway, or revisiting other well-known anti-inflammatory drugs e.g., colchicine, vitamin C, nonsteroidal anti-inflammatory drugs (NSAIDs), glucocorticosteroids, and antimalarial drugs. In our review, we aim to find relevant upstream anti-inflammatory treatment methods for the management of AF and present the most current real-world evidence of their clinical utility.
Collapse
|
3
|
Qi F, Yang L, Chang G, Wang X, Tao G, Xiao H. Comprehensive mendelian randomization reveals atrial fibrillation-breast cancer relationship and explores common druggable targets. Front Pharmacol 2024; 15:1435545. [PMID: 39170695 PMCID: PMC11335625 DOI: 10.3389/fphar.2024.1435545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 07/26/2024] [Indexed: 08/23/2024] Open
Abstract
Background Atrial fibrillation (AF) and breast cancer pose significant risks to human health. The reasons behind the concurrent occurrence of AF and breast cancer remain unclear, leading to complex treatment approaches. Mendelian Randomization (MR) analyses aim to offer genetic evidence supporting the causation of AF and breast cancer and to investigate common druggable genes associated with both conditions. Methods We used two-samples of MR to sequentially explore the causal relationship between atrial fibrillation and breast cancer, and between atrial fibrillation and breast cancer therapeutic drugs, and verified the stability of the results through colocalization analysis. We utilized the Connectivity map database to infer the direction of drug effects on disease. Finally, we explored druggable genes that play a role in AF and breast cancer and performed a Phenome-wide MR analysis to analyze the potential side effects of drug targets. Results We found 15 breast cancer therapeutic drugs that significantly support a causal association between AF and breast cancer through expression in blood and/or atrial appendage tissue. Among these, activation of ANXA5 by Docetaxel, inhibition of EIF5A by Fulvestrant, and inhibition of GNA12 by Tamoxifen increased the risk of AF, while inhibition of ANXA5 by Gemcitabine and Vinorebine and inhibition of PCGF6 by Paclitaxel reduced the risk of AF. Inhibition of MSH6 and SF3B1 by Cyclophosphamide, as well as inhibition of SMAD4 and PSMD2 and activation of ASAH1 and MLST8 by Doxorubicin can have bidirectional effects on AF occurrence. XBP1 can be used as a common druggable gene for AF and breast cancer, and there are no potential side effects of treatment against this target. Conclusion This study did not find a direct disease causality between AF and breast cancer but identified 40 target genes for 15 breast cancer therapeutic drugs associated with AF, clarified the direction of action of 8 breast cancer therapeutic drugs on AF, and finally identified one common druggable target for AF and breast cancer.
Collapse
Affiliation(s)
- Fenglin Qi
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lunzhe Yang
- Department of Neurosurgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Guanglei Chang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiangbin Wang
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Guanghong Tao
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hua Xiao
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
4
|
Lai YJ, Chang SH, Tung YC, Chang GJ, Almeida CD, Chen WJ, Yeh YH, Tsai FC. Naringin activates semaphorin 3A to ameliorate TGF-β-induced endothelial-to-mesenchymal transition related to atrial fibrillation. J Cell Physiol 2024; 239:e31248. [PMID: 38501506 DOI: 10.1002/jcp.31248] [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: 07/25/2023] [Revised: 02/25/2024] [Accepted: 02/29/2024] [Indexed: 03/20/2024]
Abstract
The loss of semaphorin 3A (Sema3A), which is related to endothelial-to-mesenchymal transition (EndMT) in atrial fibrosis, is implicated in the pathogenesis of atrial fibrillation (AF). To explore the mechanisms by which EndMT affects atrial fibrosis and assess the potential of a Sema3A activator (naringin) to prevent atrial fibrosis by targeting transforming growth factor-beta (TGF-β)-induced EndMT, we used human atria, isolated human atrial endocardial endothelial cells (AEECs), and used transgenic mice expressing TGF-β specifically in cardiac tissues (TGF-β transgenic mice). We evaluated an EndMT marker (Twist), a proliferation marker (proliferating cell nuclear antigen; PCNA), and an endothelial cell (EC) marker (CD31) through triple immunohistochemistry and confirmed that both EndMT and EC proliferation contribute to atrial endocardial fibrosis during AF in TGF-β transgenic mice and AF patient tissue sections. Additionally, we investigated the impact of naringin on EndMT and EC proliferation in AEECs and atrial fibroblasts. Naringin exhibited an antiproliferative effect, to which AEECs were more responsive. Subsequently, we downregulated Sema3A in AEECs using small interfering RNA to clarify a correlation between the reduction in Sema3A and the elevation of EndMT markers. Naringin treatment induced the expression of Sema3A and a concurrent decrease in EndMT markers. Furthermore, naringin administration ameliorated AF and endocardial fibrosis in TGF-β transgenic mice by stimulating Sema3A expression, inhibiting EndMT markers, reducing atrial fibrosis, and lowering AF vulnerability. This suggests therapeutic potential for naringin in AF treatment.
Collapse
Affiliation(s)
- Ying-Ju Lai
- Cardiovascular Department, Chang-Gung Memorial Hospital, Tao-Yuan, Taiwan
- Department of Respiratory Therapy, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
- Department of Respiratory Care, Chang-Gung University of Science and Technology, Chia-Yi, Puzi, Taiwan
| | - Shang-Hung Chang
- Cardiovascular Department, Chang-Gung Memorial Hospital, Tao-Yuan, Taiwan
- Department of Medicine, College of Medicine, Chang Gung University Tao-Yuan, Tao-Yuan, Taiwan
| | - Ying-Chang Tung
- Cardiovascular Department, Chang-Gung Memorial Hospital, Tao-Yuan, Taiwan
- Department of Medicine, College of Medicine, Chang Gung University Tao-Yuan, Tao-Yuan, Taiwan
| | - Gwo-Jyh Chang
- Cardiovascular Department, Chang-Gung Memorial Hospital, Tao-Yuan, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang-Gung University Tao-Yuan, Tao-Yuan, Taiwan
| | - Celina De Almeida
- Department of Respiratory Therapy, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang-Gung University Tao-Yuan, Tao-Yuan, Taiwan
| | - Wei-Jan Chen
- Cardiovascular Department, Chang-Gung Memorial Hospital, Tao-Yuan, Taiwan
- Department of Medicine, College of Medicine, Chang Gung University Tao-Yuan, Tao-Yuan, Taiwan
| | - Yung-Hsin Yeh
- Cardiovascular Department, Chang-Gung Memorial Hospital, Tao-Yuan, Taiwan
- Department of Medicine, College of Medicine, Chang Gung University Tao-Yuan, Tao-Yuan, Taiwan
| | - Feng-Chun Tsai
- Department of Surgery, Division of Cardiovascular Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Surgery, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
5
|
Mascarenhas L, Downey M, Schwartz G, Adabag S. Antiarrhythmic effects of metformin. Heart Rhythm O2 2024; 5:310-320. [PMID: 38840768 PMCID: PMC11148504 DOI: 10.1016/j.hroo.2024.04.003] [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] [Indexed: 06/07/2024] Open
Abstract
Atrial fibrillation/flutter (AF) is a major public health problem and is associated with stroke, heart failure, dementia, and death. It is estimated that 20%-30% of Americans will develop AF at some point in their life. Current medications to prevent AF have limited efficacy and significant adverse effects. Newer and safer therapies to prevent AF are needed. Ventricular arrhythmias are less prevalent than AF but may have significant consequences including sudden cardiac death. Metformin is the most prescribed, first-line medication for treatment of diabetes mellitus (DM). It decreases hepatic glucose production but also reduces inflammation and oxidative stress. Experimental studies have shown that metformin improves metabolic, electrical, and histologic risk factors associated with AF and ventricular arrhythmias. Furthermore, in large clinical observational studies, metformin has been associated with a reduced risk of AF in people with DM. These data suggest that metformin may have antiarrhythmic properties and may be a candidate to be repurposed as a medication to prevent cardiac arrhythmias. In this article, we review the clinical observational and experimental evidence for the association between metformin and cardiac arrhythmias. We also discuss the potential antiarrhythmic mechanisms underlying this association. Repurposing a well-tolerated, safe, and inexpensive medication to prevent cardiac arrhythmias has significant positive public health implications.
Collapse
Affiliation(s)
- Lorraine Mascarenhas
- Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Michael Downey
- Department of Cardiology, Hennepin County Medical Center, Minneapolis, Minnesota
| | - Gregory Schwartz
- Cardiology Section, Rocky Mountain Regional VA Medical Center and University of Colorado School of Medicine, Aurora, Colorado
| | - Selcuk Adabag
- Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota
- Department of Cardiology, Minneapolis Veterans Affairs Medical Center and University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
6
|
Wang YF, An ZY, Li JW, Dong ZK, Jin WL. MG53/TRIM72: multi-organ repair protein and beyond. Front Physiol 2024; 15:1377025. [PMID: 38681139 PMCID: PMC11046001 DOI: 10.3389/fphys.2024.1377025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/01/2024] [Indexed: 05/01/2024] Open
Abstract
MG53, a member of the tripartite motif protein family, possesses multiple functionalities due to its classic membrane repair function, anti-inflammatory ability, and E3 ubiquitin ligase properties. Initially recognized for its crucial role in membrane repair, the therapeutic potential of MG53 has been extensively explored in various diseases including muscle injury, myocardial damage, acute lung injury, and acute kidney injury. However, further research has revealed that the E3 ubiquitin ligase characteristics of MG53 also contribute to the pathogenesis of certain conditions such as diabetic cardiomyopathy, insulin resistance, and metabolic syndrome. Moreover, recent studies have highlighted the anti-tumor effects of MG53 in different types of cancer, such as small cell lung cancer, liver cancer, and colorectal cancer; these effects are closely associated with their E3 ubiquitin ligase activities. In summary, MG53 is a multifunctional protein that participates in important physiological and pathological processes of multiple organs and is a promising therapeutic target for various human diseases. MG53 plays a multi-organ protective role due to its membrane repair function and its exertion of anti-tumor effects due to its E3 ubiquitin ligase properties. In addition, the controversial aspect of MG53's E3 ubiquitin ligase properties potentially causing insulin resistance and metabolic syndrome necessitates further cross-validation for clarity.
Collapse
Affiliation(s)
- Yong-Fei Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, Lanzhou, China
| | - Zi-Yi An
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, Lanzhou, China
| | - Jian-Wen Li
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, Lanzhou, China
| | - Zi-Kai Dong
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, Lanzhou, China
| | - Wei-Lin Jin
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
7
|
Kostin S, Richter M, Ganceva N, Sasko B, Giannakopoulos T, Ritter O, Szalay Z, Pagonas N. Atrial fibrillation in human patients is associated with increased collagen type V and TGFbeta1. INTERNATIONAL JOURNAL OF CARDIOLOGY. HEART & VASCULATURE 2024; 50:101327. [PMID: 38419608 PMCID: PMC10899732 DOI: 10.1016/j.ijcha.2023.101327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 03/02/2024]
Abstract
Background and aim Atrial fibrosis is an important factor in initiating and maintaining atrial fibrillation (AF). Collagen V belongs to fibrillar collagens. There are, however no data on collagen V in AF. The aim of this work was to study the quantity of collagen V and its relationship with the number of fibroblasts and TGF- b 1 expression in patients in sinus rhythm (SR) and in patients with atrial fibrillation (AF). Methods We used quantitative immuhistochemistry to study collagen V in right and left atrial biopsies obtained from 35 patients in SR, 35 patients with paroxysmal AF (pAF) and 27 patients with chronic, long-standing persistent AF (cAF). In addition, we have quantified the number of vimentin-positive fibroblasts and expression levels of TGF-β1. Results Compared to patients in SR, collagen V was increased 1.8- and 3.1-fold in patients with pAF and cAF, respectively. In comparison with SR patients, the number of vimentin-positive cells increased significantly 1.46- and 1.8-fold in pAF and cAF patients, respectively.Compared to SR patients, expression levels of TGF-ß1, expressed as fluorescence units per tissue area, was significantly increased by 77 % and 300 % in patients with pAF and cAF, respectively. Similar to intensity measurements, the number of TGFß1-positive cells per 1 mm2 atrial tissue increased significantly from 35.5 ± 5.5 cells in SR patients to 61.9 ± 12.4 cells in pAF and 131.5 ± 23.5 cells in cAF. In both types of measurements, there was a statistically significant difference between pAF and cAF groups. Conclusions This is the first study to show that AF is associated with increased expression levels of collagen V and TGF-ß1indicating its role in the pathogenesis of atrial fibrosis. In addition, increases in collagen V correlate with increased number of fibroblasts and TGF-β1 and are more pronounced in cAF patients than those in pAF patients.
Collapse
Affiliation(s)
- Sawa Kostin
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
| | - Manfred Richter
- Department of Cardiac Surgery, Kerckhoff-Clinic, Bad Nauheim, Germany
| | - Natalia Ganceva
- Department of Anesthesiology and Intensive Care, Kerckoff-Clinic, Bad Nauheim, Germany
| | - Benjamin Sasko
- Medical Department II, Marien Hospital Herne, Ruhr-University of Bochum, Germany
| | | | - Oliver Ritter
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
- Department of Cardiology, University Hospital Brandenburg, Brandenburg an der Havel, Germany
| | - Zoltan Szalay
- Department of Cardiac Surgery, Kerckhoff-Clinic, Bad Nauheim, Germany
| | - Nikolaos Pagonas
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
- Department of Internal Medicine, University Hospital Ruppin-Brandenburg, Neuruppin, Germany
| |
Collapse
|
8
|
Liu J, Liu X, Luo Y, Huang F, Xie Y, Zheng S, Jia B, Xiao Z. Sphingolipids: drivers of cardiac fibrosis and atrial fibrillation. J Mol Med (Berl) 2024; 102:149-165. [PMID: 38015241 PMCID: PMC10858135 DOI: 10.1007/s00109-023-02391-8] [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: 06/29/2023] [Revised: 10/13/2023] [Accepted: 10/23/2023] [Indexed: 11/29/2023]
Abstract
Sphingolipids (SLs) are vital constituents of the plasma membrane of animal cells and concurrently regulate numerous cellular processes. An escalating number of research have evinced that SLs assume a crucial part in the progression of tissue fibrosis, a condition for which no efficacious cure exists as of now. Cardiac fibrosis, and in particular, atrial fibrosis, is a key factor in the emergence of atrial fibrillation (AF). AF has become one of the most widespread cardiac arrhythmias globally, with its incidence continuing to mount, thereby propelling it to the status of a major public health concern. This review expounds on the structure and biosynthesis pathways of several pivotal SLs, the pathophysiological mechanisms of AF, and the function of SLs in cardiac fibrosis. Delving into the influence of sphingolipid levels in the alleviation of cardiac fibrosis offers innovative therapeutic strategies to address cardiac fibrosis and AF.
Collapse
Affiliation(s)
- Junjie Liu
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ximao Liu
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yucheng Luo
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fangze Huang
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yu Xie
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shaoyi Zheng
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Bo Jia
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China.
| | - Zezhou Xiao
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
9
|
Liu SM, Zhao Q, Li WJ, Zhao JQ. Advances in the Study of MG53 in Cardiovascular Disease. Int J Gen Med 2023; 16:6073-6082. [PMID: 38152078 PMCID: PMC10752033 DOI: 10.2147/ijgm.s435030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 11/29/2023] [Indexed: 12/29/2023] Open
Abstract
Cardiovascular diseases represent a global health crisis, and understanding the intricate molecular mechanisms underlying cardiac pathology is crucial for developing effective diagnostic and therapeutic strategies. Mitsugumin-53 (MG53) plays a pivotal role in cell membrane repair, has emerged as a multifaceted player in cardiovascular health. MG53, also known as TRIM72, is primarily expressed in cardiac and skeletal muscle and actively participates in membrane repair processes essential for maintaining cardiomyocyte viability. It promotes k-ion currents, ensuring action potential integrity, and actively engages in repairing myocardial and mitochondrial membranes, preserving cardiac function in the face of oxidative stress. This study discusses the dual impact of MG53 on cardiac health, highlighting its cardioprotective role during ischemia/reperfusion injury, its modulation of cardiac arrhythmias, and its influence on cardiomyopathy. MG53's regulation of metabolic pathways, such as lipid metabolism, underlines its role in diabetic cardiomyopathy, while its potential to mitigate the effects of various cardiac disorders, including those induced by antipsychotic medications and alcohol consumption, warrants further exploration. Furthermore, we examine MG53's diagnostic potential as a biomarker for cardiac injury. Research has shown that MG53 levels correlate with cardiomyocyte damage and may predict major adverse cardiovascular events, highlighting its value as a biomarker. Additionally, exogenous recombinant human MG53 (rhMG53) emerges as a promising therapeutic option, demonstrating its ability to reduce infarct size, inhibit apoptosis, and attenuate fibrotic responses. In summary, MG53's diagnostic and therapeutic potential in cardiovascular diseases presents an exciting avenue for improved patient care and outcomes.
Collapse
Affiliation(s)
- Shan-Mei Liu
- Bayannur Hospital Department of Cardiology, Bayannur City, Inner Mongolia, 015000, People’s Republic of China
| | - Qin Zhao
- Bayannur Hospital Department of Cardiology, Bayannur City, Inner Mongolia, 015000, People’s Republic of China
| | - Wen-Jun Li
- Tangshan Central Hospital, Tangshan, Hebei, 063008, People’s Republic of China
| | - Jian-Quan Zhao
- Bayannur Hospital Department of Cardiology, Bayannur City, Inner Mongolia, 015000, People’s Republic of China
| |
Collapse
|
10
|
Winters J, Isaacs A, Zeemering S, Kawczynski M, Maesen B, Maessen J, Bidar E, Boukens B, Hermans B, van Hunnik A, Casadei B, Fabritz L, Chua W, Sommerfeld L, Guasch E, Mont L, Batlle M, Hatem S, Kirchhof P, Wakili R, Sinner M, Stoll M, Goette A, Verheule S, Schotten U. Heart Failure, Female Sex, and Atrial Fibrillation Are the Main Drivers of Human Atrial Cardiomyopathy: Results From the CATCH ME Consortium. J Am Heart Assoc 2023; 12:e031220. [PMID: 37982389 PMCID: PMC10727294 DOI: 10.1161/jaha.123.031220] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 09/22/2023] [Indexed: 11/21/2023]
Abstract
BACKGROUND Atrial cardiomyopathy (atCM) is an emerging prognostic factor in cardiovascular disease. Fibrotic remodeling, cardiomyocyte hypertrophy, and capillary density are hallmarks of atCM. The contribution of etiological factors and atrial fibrillation (AF) to the development of differential atCM phenotypes has not been quantified. This study aimed to evaluate the association between histological features of atCM and the clinical phenotype. METHODS AND RESULTS We examined left atrial (LA, n=95) and right atrial (RA, n=76) appendages from a European cohort of patients undergoing cardiac surgery. Quantification of histological atCM features was performed following wheat germ agglutinin/CD31/vimentin staining. The contributions of AF, heart failure, sex, and age to histological characteristics were determined with multiple linear regression models. Persistent AF was associated with increased endomysial fibrosis (LA: +1.13±0.47 μm, P=0.038; RA: +0.94±0.38 μm, P=0.041), whereas total extracellular matrix content was not. Men had larger cardiomyocytes (LA: +1.92±0.72 μm, P<0.001), while women had more endomysial fibrosis (LA: +0.99±0.56 μm, P=0.003). Patients with heart failure showed more endomysial fibrosis (LA: +1.85±0.48 μm, P<0.001) and extracellular matrix content (LA: +3.07±1.29%, P=0.016), and a higher capillary density (LA: +0.13±0.06, P=0.007) and size (LA: +0.46±0.22 μm, P=0.044). Fuzzy k-means clustering of histological features identified 2 subtypes of atCM: 1 characterized by enhanced endomysial fibrosis (LA: +3.17 μm, P<0.001; RA: +2.86 μm, P<0.001), extracellular matrix content (LA: +3.53%, P<0.001; RA: +6.40%, P<0.001) and fibroblast density (LA: +4.38%, P<0.001), and 1 characterized by cardiomyocyte hypertrophy (LA: +1.16 μm, P=0.008; RA: +2.58 μm, P<0.001). Patients with fibrotic atCM were more frequently female (LA: odds ratio [OR], 1.33, P=0.002; RA: OR, 1.54, P=0.004), with persistent AF (LA: OR, 1.22, P=0.036) or heart failure (LA: OR, 1.62, P<0.001). Hypertrophic features were more common in men (LA: OR=1.33, P=0.002; RA: OR, 1.54, P=0.004). CONCLUSIONS Fibrotic atCM is associated with female sex, persistent AF, and heart failure, while hypertrophic features are more common in men.
Collapse
Affiliation(s)
- Joris Winters
- Department of Physiology, Cardiovascular Research Institute MaastrichtUniversity MaastrichtMaastrichtThe Netherlands
| | - Aaron Isaacs
- Department of Physiology, Cardiovascular Research Institute MaastrichtUniversity MaastrichtMaastrichtThe Netherlands
- Maastricht Centre for Systems BiologyUniversity MaastrichtMaastrichtThe Netherlands
| | - Stef Zeemering
- Department of Physiology, Cardiovascular Research Institute MaastrichtUniversity MaastrichtMaastrichtThe Netherlands
| | - Michal Kawczynski
- Department of Physiology, Cardiovascular Research Institute MaastrichtUniversity MaastrichtMaastrichtThe Netherlands
- Department of Cardiothoracic SurgeryMaastricht University Medical Centre+MaastrichtThe Netherlands
| | - Bart Maesen
- Department of Cardiothoracic SurgeryMaastricht University Medical Centre+MaastrichtThe Netherlands
| | - Jos Maessen
- Department of Cardiothoracic SurgeryMaastricht University Medical Centre+MaastrichtThe Netherlands
| | - Elham Bidar
- Department of Cardiothoracic SurgeryMaastricht University Medical Centre+MaastrichtThe Netherlands
| | - Bas Boukens
- Department of Physiology, Cardiovascular Research Institute MaastrichtUniversity MaastrichtMaastrichtThe Netherlands
| | - Ben Hermans
- Department of Physiology, Cardiovascular Research Institute MaastrichtUniversity MaastrichtMaastrichtThe Netherlands
| | - Arne van Hunnik
- Department of Physiology, Cardiovascular Research Institute MaastrichtUniversity MaastrichtMaastrichtThe Netherlands
| | - Barbara Casadei
- Division of Cardiovascular Medicine, BHF Centre of Research ExcellenceUniversity of OxfordOxfordUnited Kingdom
| | - Larissa Fabritz
- Institute of Cardiovascular SciencesBirminghamUnited Kingdom
- University Center of Cardiovascular ScienceUKE HamburgHamburgGermany
- University Heart and Vascular Center, University Hospital Hamburg EppendorfHamburgGermany
- DZHK, Standort Hamburg/Kiel/LübeckLübeckGermany
| | - Winnie Chua
- Institute of Cardiovascular SciencesBirminghamUnited Kingdom
| | - Laura Sommerfeld
- Institute of Cardiovascular SciencesBirminghamUnited Kingdom
- University Center of Cardiovascular ScienceUKE HamburgHamburgGermany
- University Heart and Vascular Center, University Hospital Hamburg EppendorfHamburgGermany
- DZHK, Standort Hamburg/Kiel/LübeckLübeckGermany
| | - Eduard Guasch
- Institute of Biomedical Research August Pi Sunyer (IDIBAPS)BarcelonaSpain
| | - Luis Mont
- Clinic Barcelona, Universitat de BarcelonaBarcelonaSpain
| | - Montserrat Batlle
- Institute of Biomedical Research August Pi Sunyer (IDIBAPS)BarcelonaSpain
- Centro de Investigación Biomédica en Red—Cardiovascular (CIBERCV)MadridSpain
| | | | - Paulus Kirchhof
- Institute of Cardiovascular SciencesBirminghamUnited Kingdom
- University Heart and Vascular Center, University Hospital Hamburg EppendorfHamburgGermany
- DZHK, Standort Hamburg/Kiel/LübeckLübeckGermany
| | - Reza Wakili
- Department of Medicine and CardiologyGoethe UniversityFrankfurtGermany
| | - Mortiz Sinner
- University Heart and Vascular Center, University Hospital Hamburg EppendorfHamburgGermany
- DZHK, Standort Hamburg/Kiel/LübeckLübeckGermany
- Department of CardiologyUniversity Hospital of MunichMunichGermany
| | - Monica Stoll
- Maastricht Centre for Systems BiologyUniversity MaastrichtMaastrichtThe Netherlands
- Department of Biochemistry, Genetic Epidemiology and Statistical GeneticsUniversity MaastrichtMaastrichtThe Netherlands
- Department of Genetic Epidemiology, Institute of Human GeneticsUniversity of MünsterMünsterGermany
| | - Andreas Goette
- Department of Cardiology and Intensive Care MedicineSt. Vincenz Hospital PaderbornPaderbornGermany
| | - Sander Verheule
- Department of Physiology, Cardiovascular Research Institute MaastrichtUniversity MaastrichtMaastrichtThe Netherlands
| | - Ulrich Schotten
- Department of Physiology, Cardiovascular Research Institute MaastrichtUniversity MaastrichtMaastrichtThe Netherlands
- Department of CardiologyMaastricht University Medical Centre+MaastrichtThe Netherlands
| |
Collapse
|
11
|
Zhang Q, Luo T, Yuan D, Liu J, Fu Y, Yuan J. Qilongtian ameliorate bleomycin-induced pulmonary fibrosis in mice via inhibiting IL-17 signal pathway. Sci Rep 2023; 13:6002. [PMID: 37045911 PMCID: PMC10092933 DOI: 10.1038/s41598-023-31439-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 03/11/2023] [Indexed: 04/14/2023] Open
Abstract
Pulmonary fibrosis (PF) is a special type of pulmonary parenchymal disease, with chronic, progressive, fibrosis, and high mortality. There is a lack of safe, effective, and affordable treatment methods. Qilongtian (QLT) is a traditional Chinese prescription that is composed of Panax notoginseng, Earthworm, and Rhodiola, and shows the remarkable clinical curative effect of PF. However, the mechanism of QLT remains to be clarified. Therefore, we studied the effectivity of QLT in treating Bleomycin (BLM) induced PF mice. 36 C57BL/6 J mice were randomized into the control group, the model group, the low-, medium- and high-dose QLT group, and Pirfenidone group. After establishing a model of pulmonary fibrosis in mice, the control and model groups were infused with a normal saline solution, and the delivery group was infused with QLT. Pulmonary function in the mice from each group was detected. Pulmonary tissue morphologies and collagen deposition were stained by HE and Masson. The content of hydroxyproline (HYP) was detected by alkaline hydrolysis and the mRNA and protein expression of related genes in pulmonary tissues were detected by using q-PCR, ELISA, and Western blot. Our studies have shown that QLT significantly reduced the inflammatory injury, hydroxy-proline content, and collagen deposition of pulmonary tissue in BLM-induced PF mice and down-regulated the cytokine related to inflammation and fibrosis and PF expression on the mRNA and protein level in PF mice. To identify the mechanism of QLT, the Transcriptome was measured and the IL-17 signal pathway was screened out for further research. Further studies indicated that QLT reduced the mRNAs and protein levels of interleukin 17 (IL-17), c-c motif chemokine ligand 12 (CCL12), c-x-c motif chemokine ligand 5 (CXCL5), fos-like antigen 1 (FOSL1), matrix metalloproteinase-9 (MMP9), and amphiregulin (AREG), which are inflammation and fibrosis-related genes in the IL-17 signal pathway. The results indicated that the potential mechanism for QLT in the prevention of PF progression was by inhibiting inflammation resulting in the IL-17 signal pathway. Our study provides the novel scientific basis of QLT and represents new therapeutics for PF in clinical.
Collapse
Affiliation(s)
- Qiang Zhang
- School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Pudong District, Shanghai, 201203, China.
- Yunnan Provincial Key Laboratory of Molecular Biology for Sinomedicine, Yunnan University of Chinese Medicine, Kunming, 650500, China.
| | - Ting Luo
- Yunnan Provincial Key Laboratory of Molecular Biology for Sinomedicine, Yunnan University of Chinese Medicine, Kunming, 650500, China
- Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Dezheng Yuan
- Yunnan University of Chinese Medicine, Kunming, 650500, China
- The third Affiliated Hospital of Yunnan University of Chinese Medicine: Kunming Municipal Hospital of Traditional Chinese Medicine, Kunming, 650500, China
| | - Jing Liu
- Yunnan University of Chinese Medicine, Kunming, 650500, China
- The third Affiliated Hospital of Yunnan University of Chinese Medicine: Kunming Municipal Hospital of Traditional Chinese Medicine, Kunming, 650500, China
| | - Yi Fu
- Yunnan University of Chinese Medicine, Kunming, 650500, China
- The third Affiliated Hospital of Yunnan University of Chinese Medicine: Kunming Municipal Hospital of Traditional Chinese Medicine, Kunming, 650500, China
| | - Jiali Yuan
- School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Pudong District, Shanghai, 201203, China
- Yunnan Provincial Key Laboratory of Molecular Biology for Sinomedicine, Yunnan University of Chinese Medicine, Kunming, 650500, China
- Yunnan University of Chinese Medicine, Kunming, 650500, China
| |
Collapse
|
12
|
Zhu M, Yan T, Zhu S, Weng F, Zhu K, Wang C, Guo C. Identification and verification of FN1, P4HA1 and CREBBP as potential biomarkers in human atrial fibrillation. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2023; 20:6947-6965. [PMID: 37161136 DOI: 10.3934/mbe.2023300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
BACKGROUND Atrial fibrillation (AF) is a common arrhythmia that can lead to cardiac complications. The mechanisms involved in AF remain elusive. We aimed to explore the potential biomarkers and mechanisms underpinning AF. METHODS An independent dataset, GSE2240, was obtained from the Gene Expression Omnibus database. The R package, "limma", was used to screen for differentially expressed genes (DEGs) in individuals with AF and normal sinus rhythm (SR). Weighted gene co-expression network analysis (WGCNA) was applied to cluster DEGs into different modules based on functional disparities. Enrichment analyses were performed using the Database for Annotation, Visualization and Integrated Discovery. A protein-protein interaction network was constructed, and hub genes were identified using cytoHubba. Quantitative reverse-transcription PCR was used to validate mRNA expression in individuals with AF and SR. RESULTS We identified 2, 589 DEGs clustered into 10 modules using WGCNA. Gene Ontology analysis showed specific clustered genes significantly enriched in pathways associated with the extracellular matrix and collagen organization. Kyoto Encyclopedia of Genes and Genomes pathway analysis revealed that the target genes were mainly enriched for proteoglycans in cancer, extracellular matrix-receptor interaction, focal adhesion, and the PI3K-Akt signaling pathway. Three hub genes, FN1, P4HA1 and CREBBP, were identified, which were highly correlated with AF endogenesis. mRNA expression of hub genes in patients with AF were higher than in individuals with normal SR, consistent with the results of bioinformatics analysis. CONCLUSIONS FN1, P4HA1, and CREBBP may play critical roles in AF. Using bioinformatics, we found that expression of these genes was significantly elevated in patients with AF than in individuals with normal SR. Furthermore, these genes were elevated at core positions in the mRNA interaction network. These genes should be further explored as novel biomarkers and target candidates for AF therapy.
Collapse
Affiliation(s)
- Miao Zhu
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Tao Yan
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Shijie Zhu
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Fan Weng
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Kai Zhu
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Chunsheng Wang
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Changfa Guo
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| |
Collapse
|
13
|
Mechanism and prevention of atrial remodeling and their related genes in cardiovascular disorders. Curr Probl Cardiol 2022; 48:101414. [PMID: 36155200 DOI: 10.1016/j.cpcardiol.2022.101414] [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: 09/09/2022] [Accepted: 09/20/2022] [Indexed: 11/23/2022]
Abstract
Atrial fibrillation (AF) is associated with profound structural and functional changes in the atrium. Inflammation mediated atrial fibrosis is one of the key mechanisms in the pathogenesis of AF. The collagen deposition in extracellular matrix (ECM) is mainly mediated by transforming growth factor β1 (TGF-β1) which promotes AF via controlling smads mediated-collagen gene transcription and regulating the balance of metalloproteinases (MMPs)/ tissue inhibitor of metalloproteinases (TIMPs). Although many processes can alter atrial properties and promote AF, animal models and clinical studies have provided insights into two major forms of atrial remodeling: Atrial tachycardia remodeling (ATR), which occurs with rapid atrial tachyarrhythmia's such as AF and atrial flutter, and atrial structural remodeling (ASR), which is associated with CHF and other fibrosis-promoting conditions. The mechanism of atrial remodeling such as atrial enlargement, ultra structural changes of atrial muscle tissue and myocardial interstitial fibrosis in AF is still unclear. At present, many studies focus on calcium overload, renin angiotensin aldosterone system and transforming growth factor β1, that effect on atrial structural remodeling. Recent experimental studies and clinical investigations have provided structural remodeling is important contributor to the AF. This paper reviews the current understanding of the progresses about mechanism of atrial structural remodeling, and highlights the potential therapeutic approaches aimed at attenuating structural remodeling to prevent AF. Now some recent advancements of this area are reviewed in this paper.
Collapse
|
14
|
miR-29b ameliorates atrial fibrosis in rats with atrial fibrillation by targeting TGFβRΙ and inhibiting the activation of Smad-2/3 pathway. J Bioenerg Biomembr 2022; 54:81-91. [PMID: 35322290 DOI: 10.1007/s10863-022-09934-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 02/23/2022] [Indexed: 10/18/2022]
Abstract
OBJECTIVE Atrial fibrillation (AF) is a major cause of stroke with lifetime risks. microRNAs (miRNAs) are associated with AF attenuation, yet the mechanism remains unknown. This study investigated the functional mechanism of miR-29b in atrial fibrosis in AF. METHODS The AF rat model was established by a 7-day intravenous injection of Ach-CaCl2 mixture. AF rats were injected with adeno-associated virus (AAv)-miR-29b and TGFβRΙ overexpression plasmid. AF duration was recorded by electrocardiogram. Atrial fibrosis was observed by Masson staining. Expressions of COL1A1, COL3A1, TGFβRΙ, TGFβΙ, miR-29b and Smad-2/3 pathway-related proteins in atrial tissues were detected by RT-qPCR and Western blot. Binding sites of miR-29b and TGFβRΙ were predicted and their target relationship was verified by dual-luciferase reporter assay. RESULTS miR-29b was poorly expressed and expressions of COL1A1, COL3A1, TGFβRΙ, and TGFβ1 were increased in atrial tissues of AF rats. miR-29b overexpression alleviated atrial fibrosis, reduced expressions of COL1A1, COL3A1, and TGFβ1, and shortened AF duration in AF rats. TGFβRΙ was highly expressed in atrial tissues of AF rats. miR-29b targeted TGFβRΙ. TGFβRΙ overexpression overcame the improving effect of miR-29b overexpression on AF. miR-29b overexpression decreased ratios of p-Smad-2/3 and Smad-2/3 and inhibited the Smad-2/3 pathway. CONCLUSION miR-29b might mitigate atrial fibrosis in AF rats by targeting TGFβRΙ and inhibiting the Smad-2/3 pathway.
Collapse
|
15
|
Abstract
Transforming growth factor-β (TGFβ) isoforms are upregulated and activated in myocardial diseases and have an important role in cardiac repair and remodelling, regulating the phenotype and function of cardiomyocytes, fibroblasts, immune cells and vascular cells. Cardiac injury triggers the generation of bioactive TGFβ from latent stores, through mechanisms involving proteases, integrins and specialized extracellular matrix (ECM) proteins. Activated TGFβ signals through the SMAD intracellular effectors or through non-SMAD cascades. In the infarcted heart, the anti-inflammatory and fibroblast-activating actions of TGFβ have an important role in repair; however, excessive or prolonged TGFβ signalling accentuates adverse remodelling, contributing to cardiac dysfunction. Cardiac pressure overload also activates TGFβ cascades, which initially can have a protective role, promoting an ECM-preserving phenotype in fibroblasts and preventing the generation of injurious, pro-inflammatory ECM fragments. However, prolonged and overactive TGFβ signalling in pressure-overloaded cardiomyocytes and fibroblasts can promote cardiac fibrosis and dysfunction. In the atria, TGFβ-mediated fibrosis can contribute to the pathogenic substrate for atrial fibrillation. Overactive or dysregulated TGFβ responses have also been implicated in cardiac ageing and in the pathogenesis of diabetic, genetic and inflammatory cardiomyopathies. This Review summarizes the current evidence on the role of TGFβ signalling in myocardial diseases, focusing on cellular targets and molecular mechanisms, and discussing challenges and opportunities for therapeutic translation.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
16
|
Chen YL, Wang HT, Lin PT, Chuang JH, Yang MY. Macrophage Inflammatory Protein-1 Alpha, a Potential Biomarker for Predicting Left Atrial Remodeling in Patients With Atrial Fibrillation. Front Cardiovasc Med 2021; 8:784792. [PMID: 34957262 PMCID: PMC8695724 DOI: 10.3389/fcvm.2021.784792] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/17/2021] [Indexed: 11/13/2022] Open
Abstract
Objectives: Left atrial (LA) remodeling itself is an independent risk factor for ischemic stroke and mortality, with or without atrial fibrillation (AF). Macrophage inflammatory protein-1 alpha (MIP-1α) has been reported to be involved in the induction of autoimmune myocarditis and dilated cardiomyopathy. Little is known about whether MIP-1α can be used to predict LA remodeling, especially in patients with AF. Methods: We prospectively enrolled 78 patients who had received a cardiac implantable electronic device due to sick sinus syndrome in order to define AF accurately. AF was diagnosed clinically before enrollment, according to 12-lead electrocardiography (ECG) and 24-h Holter test in 54 (69%) patients. The serum cytokine levels and the mRNA expression levels of peripheral blood leukocytes were checked and echocardiographic study was performed on the same day within 1 week after the patients were enrolled into the study. The 12-lead ECG and 24-h Holter test were performed on the same day of the patients' enrollment, and the device interrogation was performed every 3 months after enrollment. The enrolled patients were clinically followed up for 1 year. Results: There was no difference in baseline characteristics, cytokine levels and mRNA expression between patients with and without AF. Larger LA volume was positively correlated with higher levels of MIP-1α (r = 0.461, p ≤ 0.001) and the atrial high-rate episodes (AHREs) burden (r = 0.593, p < 0.001), and negatively correlated with higher levels of transforming growth factor (TGF)-β1 (r = −0.271, p = 0.047) and TGF-β3 (r = −0.279, p = 0.041). The higher AHREs burden and MIP-1α level could predict LA volume independently. The mRNA expression of RORC was negatively associated with the MIP-1α level. Conclusions: This study showed that higher MIP-1α was significantly associated with LA remodeling and may have the potentials to predict LA remodeling in terms of a larger LA volume, and that circadian gene derangement might affect the expression of MIP-1α.
Collapse
Affiliation(s)
- Yung-Lung Chen
- Section of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan.,School of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,College of Medicine, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Hui-Ting Wang
- Emergency Department, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Pei-Ting Lin
- Section of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan.,School of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Jiin-Haur Chuang
- College of Medicine, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan.,Division of Pediatric Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Ming-Yu Yang
- College of Medicine, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan.,Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| |
Collapse
|
17
|
van den Berg NWE, Kawasaki M, Fabrizi B, Nariswari FA, Verduijn AC, Neefs J, Wesselink R, Al‐Shama RFM, van der Wal AC, de Boer OJ, Aten J, Driessen AHG, Jongejan A, de Groot JR. Epicardial and endothelial cell activation concurs with extracellular matrix remodeling in atrial fibrillation. Clin Transl Med 2021; 11:e558. [PMID: 34841686 PMCID: PMC8567047 DOI: 10.1002/ctm2.558] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 08/11/2021] [Accepted: 08/16/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Improved understanding of the interconnectedness of structural remodeling processes in atrial fibrillation (AF) in patients could identify targets for future therapies. METHODS We present transcriptome sequencing of atrial tissues of patients without AF, with paroxysmal AF, and persistent AF (total n = 64). RNA expression levels were validated in the same and an independent cohort with qPCR. Biological processes were assessed with histological and immunohistochemical analyses. RESULTS In AF patients, epicardial cell gene expression decreased, contrasting with an upregulation of epithelial-to-mesenchymal transition (EMT) and mesenchymal cell gene expression. Immunohistochemistry demonstrated thickening of the epicardium and an increased proportion of (myo)fibroblast-like cells in the myocardium, supporting enhanced EMT in AF. We furthermore report an upregulation of endothelial cell proliferation, angiogenesis, and endothelial signaling. EMT and endothelial cell proliferation concurred with increased interstitial (myo)fibroblast-like cells and extracellular matrix gene expression including enhanced tenascin-C, thrombospondins, biglycan, and versican. Morphological analyses discovered increased and redistributed glycosaminoglycans and collagens in the atria of AF patients. Signaling pathways, including cell-matrix interactions, PI3K-AKT, and Notch signaling that could regulate mesenchymal cell activation, were upregulated. CONCLUSION Our results suggest that EMT and endothelial cell proliferation work in concert and characterize the (myo)fibroblast recruitment and ECM remodeling of AF. These processes could guide future research toward the discovery of targets for AF therapy.
Collapse
Affiliation(s)
- Nicoline W. E. van den Berg
- Department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular SciencesAmsterdam UMC, University of Amsterdam, Heart CenterAmsterdamThe Netherlands
| | - Makiri Kawasaki
- Department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular SciencesAmsterdam UMC, University of Amsterdam, Heart CenterAmsterdamThe Netherlands
| | - Benedetta Fabrizi
- Department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular SciencesAmsterdam UMC, University of Amsterdam, Heart CenterAmsterdamThe Netherlands
| | - Fransisca A. Nariswari
- Department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular SciencesAmsterdam UMC, University of Amsterdam, Heart CenterAmsterdamThe Netherlands
| | - Arianne C. Verduijn
- Department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular SciencesAmsterdam UMC, University of Amsterdam, Heart CenterAmsterdamThe Netherlands
| | - Jolien Neefs
- Department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular SciencesAmsterdam UMC, University of Amsterdam, Heart CenterAmsterdamThe Netherlands
| | - Robin Wesselink
- Department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular SciencesAmsterdam UMC, University of Amsterdam, Heart CenterAmsterdamThe Netherlands
| | - Rushd F. M. Al‐Shama
- Department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular SciencesAmsterdam UMC, University of Amsterdam, Heart CenterAmsterdamThe Netherlands
| | - Allard C. van der Wal
- Department of Clinical PathologyAmsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
| | - Onno J. de Boer
- Department of Clinical PathologyAmsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
| | - Jan Aten
- Department of Clinical PathologyAmsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
| | - Antoine H. G. Driessen
- Department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular SciencesAmsterdam UMC, University of Amsterdam, Heart CenterAmsterdamThe Netherlands
| | - Aldo Jongejan
- Department of Epidemiology & Data ScienceAmsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
| | - Joris R. de Groot
- Department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular SciencesAmsterdam UMC, University of Amsterdam, Heart CenterAmsterdamThe Netherlands
| |
Collapse
|
18
|
Xu D, Murakoshi N, Tajiri K, Duo F, Okabe Y, Murakata Y, Yuan Z, Li S, Aonuma K, Song Z, Shimoda Y, Mori H, Sato A, Nogami A, Aonuma K, Ieda M. Xanthine oxidase inhibitor febuxostat reduces atrial fibrillation susceptibility by inhibition of oxidized CaMKII in Dahl salt-sensitive rats. Clin Sci (Lond) 2021; 135:2409-2422. [PMID: 34386810 DOI: 10.1042/cs20210405] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 08/06/2021] [Accepted: 08/13/2021] [Indexed: 11/17/2022]
Abstract
Oxidative stress could be a possible mechanism and a therapeutic target of atrial fibrillation (AF). However, the effects of the xanthine oxidase (XO) inhibition for AF remain to be fully elucidated. We investigated the effects of a novel XO inhibitor febuxostat on AF compared with allopurinol in hypertension rat model. Five-week-old Dahl salt-sensitive rats were fed either low-salt (LS) (0.3% NaCl) or high-salt (HS) (8% NaCl) diet. After 4 weeks of diet, HS diet rats were divided into three groups: orally administered to vehicle (HS-C), febuxostat (5 mg/kg/day) (HS-F), or allopurinol (50 mg/kg/day) (HS-A). After 4 weeks of treatment, systolic blood pressure (SBP) was significantly higher in HS-C than LS, and it was slightly but significantly decreased by treatment with each XO inhibitor. AF duration was significantly prolonged in HS-C compared with LS, and significantly suppressed in both HS-F and HS-A (LS; 5.8 ± 3.5 s, HS-C; 33.9 ± 23.7 s, HS-F; 15.0 ± 14.1 s, HS-A; 20.1 ± 11.9 s: P<0.05). Ca2+ spark frequency was obviously increased in HS-C rats and reduced in the XO inhibitor-treated rats, especially in HS-F group. Western blotting revealed that the atrial expression levels of Met281/282-oxidized Ca2+/Calmodulin-dependent kinase II (CaMKII) and Ser2814-phosphorylated ryanodine receptor 2 were significantly increased in HS-C, and those were suppressed in HS-F and HS-A. Decreased expression of gap junction protein connexin 40 in HS-C was partially restored by treatment with each XO inhibitor. In conclusion, XO inhibitor febuxostat, as well as allopurinol, could reduce hypertension-related increase in AF perpetuation by restoring Ca2+ handling and gap junction.
Collapse
Affiliation(s)
- DongZhu Xu
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Nobuyuki Murakoshi
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kazuko Tajiri
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Feng Duo
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yuta Okabe
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yoshiko Murakata
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Zixun Yuan
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Siqi Li
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kazuhiro Aonuma
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Zonghu Song
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yuzuno Shimoda
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Haruka Mori
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Akira Sato
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Akihiko Nogami
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kazutaka Aonuma
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Masaki Ieda
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
19
|
Jiang J, Gu X, Wang H, Ding S. Resveratrol improves cardiac function and left ventricular fibrosis after myocardial infarction in rats by inhibiting NLRP3 inflammasome activity and the TGF- β1/SMAD2 signaling pathway. PeerJ 2021; 9:e11501. [PMID: 34123595 PMCID: PMC8166236 DOI: 10.7717/peerj.11501] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 05/02/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Several studies have shown that resveratrol (RES), a naturally occurring polyphenol found in many plants, is beneficial for preventing cardiovascular diseases. However, the mechanism underlying the RES-mediated protection against myocardial infarction has not yet been revealed entirely. In this study, we investigated the protective effects of RES on cardiac function in a rat model of acute myocardial infarction (AMI) and the related underlying mechanisms. METHODS Male Sprague-Dawley rats were randomly divided into four groups: Sham (sham operation), Sham-RES, AMI (AMI induction), and AMI-RES. The rat AMI model was established by the permanent ligation of left anterior descending coronary artery method. The rats in the RES-treated groups were gavaged with RES (50 mg/kg/day) daily for 45 days after the Sham operation or AMI induction; rats in the Sham and AMI groups were gavaged with deionized water. Cardiac function was evaluated by echocardiography. Atrial interstitial fibrosis was assessed by hematoxylin-eosin or Masson's trichrome staining. Real-time PCR and western blotting analyses were performed to examine the levels of signaling pathway components. RESULTS RES supplementation decreased the inflammatory cytokine levels, improved the cardiac function, and ameliorated atrial interstitial fibrosis in the rats with AMI. Furthermore, RES supplementation inhibited NLRP3 inflammasome activity, decreased the TGF-β1 production, and downregulated the p-SMAD2/SMAD2 expression in the heart. CONCLUSION RES shows notable cardioprotective effects in a rat model of AMI; the possible mechanisms underlying these effects may involve the improvement of cardiac function and atrial interstitial fibrosis via the RES-mediated suppression of NLRP3 inflammasome activity and inhibition of the TGF-β1/SMAD2 signaling pathway in the heart.
Collapse
Affiliation(s)
- Jinjin Jiang
- Jiangsu Vocational College of Medicine, Yancheng, Jiangsu, China
| | - Xiuping Gu
- Department of Cardiology, General Hospital of TISCO, Taiyuan, Shanxi, China
| | - Huifeng Wang
- Department of Cardiology, General Hospital of TISCO, Taiyuan, Shanxi, China
| | - Shibin Ding
- Jiangsu Vocational College of Medicine, Yancheng, Jiangsu, China
| |
Collapse
|
20
|
Oh Y, Yang S, Liu X, Jana S, Izaddoustdar F, Gao X, Debi R, Kim DK, Kim KH, Yang P, Kassiri Z, Lakin R, Backx PH. Transcriptomic Bioinformatic Analyses of Atria Uncover Involvement of Pathways Related to Strain and Post-translational Modification of Collagen in Increased Atrial Fibrillation Vulnerability in Intensely Exercised Mice. Front Physiol 2020; 11:605671. [PMID: 33424629 PMCID: PMC7793719 DOI: 10.3389/fphys.2020.605671] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 11/26/2020] [Indexed: 02/06/2023] Open
Abstract
Atrial Fibrillation (AF) is the most common supraventricular tachyarrhythmia that is typically associated with cardiovascular disease (CVD) and poor cardiovascular health. Paradoxically, endurance athletes are also at risk for AF. While it is well-established that persistent AF is associated with atrial fibrosis, hypertrophy and inflammation, intensely exercised mice showed similar adverse atrial changes and increased AF vulnerability, which required tumor necrosis factor (TNF) signaling, even though ventricular structure and function improved. To identify some of the molecular factors underlying the chamber-specific and TNF-dependent atrial changes induced by exercise, we performed transcriptome analyses of hearts from wild-type and TNF-knockout mice following exercise for 2 days, 2 or 6 weeks of exercise. Consistent with the central role of atrial stretch arising from elevated venous pressure in AF promotion, all 3 time points were associated with differential regulation of genes in atria linked to mechanosensing (focal adhesion kinase, integrins and cell-cell communications), extracellular matrix (ECM) and TNF pathways, with TNF appearing to play a permissive, rather than causal, role in gene changes. Importantly, mechanosensing/ECM genes were only enriched, along with tubulin- and hypertrophy-related genes after 2 days of exercise while being downregulated at 2 and 6 weeks, suggesting that early reactive strain-dependent remodeling with exercise yields to compensatory adjustments. Moreover, at the later time points, there was also downregulation of both collagen genes and genes involved in collagen turnover, a pattern mirroring aging-related fibrosis. By comparison, twofold fewer genes were differentially regulated in ventricles vs. atria, independently of TNF. Our findings reveal that exercise promotes TNF-dependent atrial transcriptome remodeling of ECM/mechanosensing pathways, consistent with increased preload and atrial stretch seen with exercise. We propose that similar preload-dependent mechanisms are responsible for atrial changes and AF in both CVD patients and athletes.
Collapse
Affiliation(s)
- Yena Oh
- Department of Biology, York University, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.,University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Sibao Yang
- Department of Biology, York University, Toronto, ON, Canada.,Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xueyan Liu
- Department of Biology, York University, Toronto, ON, Canada.,Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Sayantan Jana
- Department of Physiology, Cardiovascular Research Center, University of Alberta, Edmonton, AB, Canada
| | | | - Xiaodong Gao
- Department of Biology, York University, Toronto, ON, Canada
| | - Ryan Debi
- Department of Biology, York University, Toronto, ON, Canada
| | - Dae-Kyum Kim
- Donnelly Centre, University of Toronto, Toronto, ON, Canada.,Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Kyoung-Han Kim
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.,University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Ping Yang
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zamaneh Kassiri
- Department of Physiology, Cardiovascular Research Center, University of Alberta, Edmonton, AB, Canada
| | - Robert Lakin
- Department of Biology, York University, Toronto, ON, Canada
| | - Peter H Backx
- Department of Biology, York University, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
21
|
Babapoor-Farrokhran S, Tarighati Rasekhi R, Gill D, Alzubi J, Mainigi SK. How transforming growth factor contributes to atrial fibrillation? Life Sci 2020; 266:118823. [PMID: 33309721 DOI: 10.1016/j.lfs.2020.118823] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/20/2020] [Accepted: 11/21/2020] [Indexed: 12/12/2022]
Abstract
Atrial fibrillation (AF) is the most common clinically significant arrhythmia. There are four fundamental pathophysiological mechanisms of AF including: electrical remodeling, structural remodeling, autonomic nervous system changes, and Ca2+ handling abnormalities. The transforming growth factor-β (TGF-β) superfamily are cytokines that have the ability to regulate numerous cell functions including proliferation, differentiation, apoptosis, epithelial-mesenchymal transition, and production of extracellular matrix. During the last decade numerous studies have demonstrated that TGF-β affects the architecture of the heart. TGF-β1 has been shown to be involved in the development and propagation of atrial fibrillation (AF). Investigators have studied TGF-β signaling in AF with the aim of discovering potential therapeutic agents. In this review we discuss the role of TGF-β in atrial fibrillation and specifically its role in atrial structural and electrical remodeling.
Collapse
Affiliation(s)
| | - Roozbeh Tarighati Rasekhi
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Deanna Gill
- Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jafar Alzubi
- Department of Medicine, Division of Cardiology, Einstein Medical Center, Philadelphia, PA 19141, USA
| | - Sumeet K Mainigi
- Department of Medicine, Division of Cardiology, Einstein Medical Center, Philadelphia, PA 19141, USA; Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
22
|
The Predictive Value of Growth Differentiation Factor-15 in Recurrence of Atrial Fibrillation after Catheter Ablation. Mediators Inflamm 2020; 2020:8360936. [PMID: 32904560 PMCID: PMC7456492 DOI: 10.1155/2020/8360936] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 07/13/2020] [Accepted: 07/15/2020] [Indexed: 11/25/2022] Open
Abstract
The mechanisms underlying the recurrence of atrial fibrillation (AF) after radiofrequency catheter ablation (RFCA) are not well concerned. The study sought to explore the association between growth differentiation factor-15 (GDF-15) and the incidence of recurrent events among AF patients after the ablation procedure. We prospectively included 150 consecutive AF patients who underwent RFCA. Clinical information about the patients was collected. Blood samples on the second morning of hospital admission and three months after RFCA were collected, and enzyme-linked immunosorbent assay (ELISA) was used to measure the concentration of GDF-15. All participants were followed up at specific times (1st/3rd/6th/12th/18th/24th months) after RFCA to record recurrences events. During a median follow-up of 14.0 months, AF recurrence occurred in 37(24.7%) patients. Baseline serum GDF-15 level in the persistent AF group was significantly higher than the paroxysmal AF group [1140(854~1701)ng/L vs. 1062(651~1374)ng/L, P = 0.039]. Baseline serum GDF-15 level in the recurrence group was significantly higher than the nonrecurrence group [1287(889~1768) ng/L vs. 1062(694~1373)ng/L, P = 0.022]. Serum GDF-15 level at three months after RFCA was significantly lower than the baseline [870 (579~1270) ng/L vs. 1155 (735~1632)ng/L, P < 0.001]. The baseline GDF-15 correlated significantly with LAP (r = 0.296, P < 0.001) and LAAV(r = −0.235, P = 0.003). Kaplan-Meier analysis showed a significantly lower event-free survival time in the high baseline GDF-15 (≥1287.3 ng/L) group than the low baseline GDF-15 (<1287.3 ng/L) group (17.1 months vs. 20.4 months, Log Rank P = 0.017). In the multivariate Cox regression, baseline GDF-15(HR 1.053, 95% CI 1.007-1.100, P = 0.022) and LAD (HR 1.124, 95% CI 1.011-1.250, P = 0.030) were independent predictors of AF recurrence after RFCA. Our study indicated increased preprocedural GDF-15 is associated with left atrial remodeling and acts as a predictor of AF recurrence after ablation.
Collapse
|
23
|
Abstract
While AF most often occurs in the setting of atrial disease, current assessment and treatment of patients with AF does not focus on the extent of the atrial myopathy that serves as the substrate for this arrhythmia. Atrial myopathy, in particular atrial fibrosis, may initiate a vicious cycle in which atrial myopathy leads to AF, which in turn leads to a worsening myopathy. Various techniques, including ECG, plasma biomarkers, electroanatomical voltage mapping, echocardiography, and cardiac MRI, can help to identify and quantify aspects of the atrial myopathy. Current therapies, such as catheter ablation, do not directly address the underlying atrial myopathy. There is emerging research showing that by targeting this myopathy we can help decrease the occurrence and burden of AF.
Collapse
Affiliation(s)
- Harold Rivner
- Cardiovascular Division, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, US
| | - Raul D Mitrani
- Cardiovascular Division, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, US
| | - Jeffrey J Goldberger
- Cardiovascular Division, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, US
| |
Collapse
|
24
|
The Atrium and Embolic Stroke. JACC Clin Electrophysiol 2020; 6:251-261. [DOI: 10.1016/j.jacep.2019.12.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 11/25/2019] [Accepted: 12/20/2019] [Indexed: 12/30/2022]
|
25
|
De Pascale MR, Della Mura N, Vacca M, Napoli C. Useful applications of growth factors for cardiovascular regenerative medicine. Growth Factors 2020; 38:35-63. [PMID: 33028111 DOI: 10.1080/08977194.2020.1825410] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Novel advances for cardiovascular diseases (CVDs) include regenerative approaches for fibrosis, hypertrophy, and neoangiogenesis. Studies indicate that growth factor (GF) signaling could promote heart repair since most of the evidence is derived from preclinical models. Observational studies have evaluated GF serum/plasma levels as feasible biomarkers for risk stratification of CVDs. Noteworthy, two clinical interventional published studies showed that the administration of growth factors (GFs) induced beneficial effect on left ventricular ejection fraction (LVEF), myocardial perfusion, end-systolic volume index (ESVI). To date, large scale ongoing studies are in Phase I-II and mostly focussed on intramyocardial (IM), intracoronary (IC) or intravenous (IV) administration of vascular endothelial growth factor (VEGF) and fibroblast growth factor-23 (FGF-23) which result in the most investigated GFs in the last 10 years. Future data of ongoing randomized controlled studies will be crucial in understanding whether GF-based protocols could be in a concrete way effective in the clinical setting.
Collapse
Affiliation(s)
| | | | - Michele Vacca
- Division of Immunohematology and Transfusion Medicine, Cardarelli Hospital, Naples, Italy
| | - Claudio Napoli
- IRCCS Foundation SDN, Naples, Italy
- Clinical Department of Internal Medicine and Specialistics, Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
26
|
Zhang PP, Sun J, Li W. Genome-wide profiling reveals atrial fibrillation-related circular RNAs in atrial appendages. Gene 2019; 728:144286. [PMID: 31838248 DOI: 10.1016/j.gene.2019.144286] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 11/28/2019] [Accepted: 12/10/2019] [Indexed: 12/11/2022]
Abstract
Atrial fibrillation (AF) is an abnormal heart rhythm characterized by rapid and irregular beating of the atria. The non-coding RNAs (ncRNAs) have attracted much attention of AF researchers, as they play a critical role in the transcriptional and post-transcriptional regulation, which could greatly benefit the interpretation of the pathogenesis of AF. However, circRNAs, as a special member of the ncRNAs, and their role in the pathogenesis of AF is less understood. In the present study, we detected a total of 14,215 circRNAs in AF patients and healthy controls. Differential expression analysis of these circRNAs revealed 20 upregulated and 3 downregulated circRNAs, which were differentially expressed in both left and right atrial appendages. The association analysis of the AF-related circRNAs and their parental genes revealed that hsa_circ_0003965 had significantly negative correlation with its parental gene TMEM245 (PCC = -0.51), suggesting that the dysregulation of hsa_circ_0003965 was not regulated by the transcription of its parental gene, but could be associated with glucagon signaling pathway. The competing endogenous RNA (ceRNA) network analysis revealed two upregulated genes, IFNG and GDF7, and one downregulated gene, BMP7, all of which were involved in TGF-beta signaling pathway, which further suggested that these circRNAs, namely hsa_circ_0000075 and hsa_circ_0082096, participated in the AF pathogenesis via TGF-beta signaling pathway. Consistently, TGF-beta signaling pathway was a well-recognized player for its association with atrial fibrosis in AF. In summary, we aimed to discover and provide key circRNAs involved in AF for AF-related researchers, which had the potential to greatly improve our understanding of the underlying mechanism behind circRNAs and AF.
Collapse
Affiliation(s)
- Peng-Pai Zhang
- Department of Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Jian Sun
- Department of Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Wei Li
- Department of Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
27
|
Wang J, Han M, Han SX, Zhi C, Gao S, Li Y. Effect of c-Ski on atrial remodelling in a rapid atrial pacing canine model. J Cell Mol Med 2019; 24:1795-1803. [PMID: 31815360 PMCID: PMC6991632 DOI: 10.1111/jcmm.14876] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 09/01/2019] [Indexed: 12/21/2022] Open
Abstract
Atrial fibrosis is an important factor in the initiation and maintenance of atrial fibrillation (AF); therefore, understanding the pathogenesis of atrial fibrosis may reveal promising therapeutic targets for AF. In this study, we successfully established a rapid atrial pacing canine model and found that the inducibility and duration of AF were significantly reduced by the overexpression of c‐Ski, suggesting that this approach may have therapeutic effects. c‐Ski was found to be down‐regulated in the atrial tissues of the rapid atrial pacing canine model. We artificially up‐regulated c‐Ski expression with a c‐Ski–overexpressing adenovirus. Haematoxylin and eosin, Masson's trichrome and picrosirius red staining showed that c‐Ski overexpression alleviated atrial fibrosis. Furthermore, we found that the expression levels of collagen III and α‐SMA were higher in the groups of dogs subjected to right‐atrial pacing, and this increase was attenuated by c‐Ski overexpression. In addition, c‐Ski overexpression decreased the phosphorylation of smad2, smad3 and p38 MAPK (p38α and p38β) as well as the expression of TGF‐β1 in atrial tissues, as shown by a comparison of the right‐atrial pacing + c‐Ski‐overexpression group to the control group with right‐atrial pacing only. These results suggest that c‐Ski overexpression improves atrial remodelling in a rapid atrial pacing canine model by suppressing TGF‐β1–Smad signalling and p38 MAPK activation.
Collapse
Affiliation(s)
- Juan Wang
- Department of Cardiology, The Fifth Affiliated Hospital to Xin Jiang Medical University, Urumchi, Xin Jiang, China
| | - Min Han
- Xin Jiang Medical University, Urumchi, Xin Jiang, China
| | - Su-Xia Han
- Department of Cardiovascular Medicine, Shanghai Pudong New Area People's Hospital Affiliated to Shanghai Health University, Shanghai, China
| | - Cuiju Zhi
- Department of Cardiovascular Medicine, Shanghai Pudong New Area People's Hospital Affiliated to Shanghai Health University, Shanghai, China
| | - Suli Gao
- Department of Cardiovascular Medicine, Shanghai Pudong New Area People's Hospital Affiliated to Shanghai Health University, Shanghai, China
| | - Yao Li
- Department of Cardiovascular Medicine, Shanghai Pudong New Area People's Hospital Affiliated to Shanghai Health University, Shanghai, China
| |
Collapse
|
28
|
Piché J, Van Vliet PP, Pucéat M, Andelfinger G. The expanding phenotypes of cohesinopathies: one ring to rule them all! Cell Cycle 2019; 18:2828-2848. [PMID: 31516082 PMCID: PMC6791706 DOI: 10.1080/15384101.2019.1658476] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 08/13/2019] [Accepted: 08/17/2019] [Indexed: 12/13/2022] Open
Abstract
Preservation and development of life depend on the adequate segregation of sister chromatids during mitosis and meiosis. This process is ensured by the cohesin multi-subunit complex. Mutations in this complex have been associated with an increasing number of diseases, termed cohesinopathies. The best characterized cohesinopathy is Cornelia de Lange syndrome (CdLS), in which intellectual and growth retardations are the main phenotypic manifestations. Despite some overlap, the clinical manifestations of cohesinopathies vary considerably. Novel roles of the cohesin complex have emerged during the past decades, suggesting that important cell cycle regulators exert important biological effects through non-cohesion-related functions and broadening the potential pathomechanisms involved in cohesinopathies. This review focuses on non-cohesion-related functions of the cohesin complex, gene dosage effect, epigenetic regulation and TGF-β in cohesinopathy context, especially in comparison to Chronic Atrial and Intestinal Dysrhythmia (CAID) syndrome, a very distinct cohesinopathy caused by a homozygous Shugoshin-1 (SGO1) mutation (K23E) and characterized by pacemaker failure in both heart (sick sinus syndrome followed by atrial flutter) and gut (chronic intestinal pseudo-obstruction) with no intellectual or growth delay. We discuss the possible impact of SGO1 alterations in human pathologies and the potential impact of the SGO1 K23E mutation in the sinus node and gut development and functions. We suggest that the human phenotypes observed in CdLS, CAID syndrome and other cohesinopathies can inform future studies into the less well-known non-cohesion-related functions of cohesin complex genes. Abbreviations: AD: Alzheimer Disease; AFF4: AF4/FMR2 Family Member 4; ANKRD11: Ankyrin Repeat Domain 11; APC: Anaphase Promoter Complex; ASD: Atrial Septal Defect; ATRX: ATRX Chromatin Remodeler; ATRX: Alpha Thalassemia X-linked intellectual disability syndrome; BIRC5: Baculoviral IAP Repeat Containing 5; BMP: Bone Morphogenetic Protein; BRD4: Bromodomain Containing 4; BUB1: BUB1 Mitotic Checkpoint Serine/Threonine Kinase; CAID: Chronic Atrial and Intestinal Dysrhythmia; CDK1: Cyclin Dependent Kinase 1; CdLS: Cornelia de Lange Syndrome; CHD: Congenital Heart Disease; CHOPS: Cognitive impairment, coarse facies, Heart defects, Obesity, Pulmonary involvement, Short stature, and skeletal dysplasia; CIPO: Chronic Intestinal Pseudo-Obstruction; c-kit: KIT Proto-Oncogene Receptor Tyrosine Kinase; CoATs: Cohesin Acetyltransferases; CTCF: CCCTC-Binding Factor; DDX11: DEAD/H-Box Helicase 11; ERG: Transcriptional Regulator ERG; ESCO2: Establishment of Sister Chromatid Cohesion N-Acetyltransferase 2; GJC1: Gap Junction Protein Gamma 1; H2A: Histone H2A; H3K4: Histone H3 Lysine 4; H3K9: Histone H3 Lysine 9; HCN4: Hyperpolarization Activated Cyclic Nucleotide Gated Potassium and Sodium Channel 4;p HDAC8: Histone deacetylases 8; HP1: Heterochromatin Protein 1; ICC: Interstitial Cells of Cajal; ICC-MP: Myenteric Plexus Interstitial cells of Cajal; ICC-DMP: Deep Muscular Plexus Interstitial cells of Cajal; If: Pacemaker Funny Current; IP3: Inositol trisphosphate; JNK: C-Jun N-Terminal Kinase; LDS: Loeys-Dietz Syndrome; LOAD: Late-Onset Alzheimer Disease; MAPK: Mitogen-Activated Protein Kinase; MAU: MAU Sister Chromatid Cohesion Factor; MFS: Marfan Syndrome; NIPBL: NIPBL, Cohesin Loading Factor; OCT4: Octamer-Binding Protein 4; P38: P38 MAP Kinase; PDA: Patent Ductus Arteriosus; PDS5: PDS5 Cohesin Associated Factor; P-H3: Phospho Histone H3; PLK1: Polo Like Kinase 1; POPDC1: Popeye Domain Containing 1; POPDC2: Popeye Domain Containing 2; PP2A: Protein Phosphatase 2; RAD21: RAD21 Cohesin Complex Component; RBS: Roberts Syndrome; REC8: REC8 Meiotic Recombination Protein; RNAP2: RNA polymerase II; SAN: Sinoatrial node; SCN5A: Sodium Voltage-Gated Channel Alpha Subunit 5; SEC: Super Elongation Complex; SGO1: Shogoshin-1; SMAD: SMAD Family Member; SMC1A: Structural Maintenance of Chromosomes 1A; SMC3: Structural Maintenance of Chromosomes 3; SNV: Single Nucleotide Variant; SOX2: SRY-Box 2; SOX17: SRY-Box 17; SSS: Sick Sinus Syndrome; STAG2: Cohesin Subunit SA-2; TADs: Topology Associated Domains; TBX: T-box transcription factors; TGF-β: Transforming Growth Factor β; TGFBR: Transforming Growth Factor β receptor; TOF: Tetralogy of Fallot; TREK1: TREK-1 K(+) Channel Subunit; VSD: Ventricular Septal Defect; WABS: Warsaw Breakage Syndrome; WAPL: WAPL Cohesin Release Factor.
Collapse
Affiliation(s)
- Jessica Piché
- Cardiovascular Genetics, Department of Pediatrics, CHU Sainte-Justine, Montréal, QC, Canada
| | - Patrick Piet Van Vliet
- Cardiovascular Genetics, Department of Pediatrics, CHU Sainte-Justine, Montréal, QC, Canada
- LIA (International Associated Laboratory), CHU Sainte-Justine, Montréal, QC, Canada
- LIA (International Associated Laboratory), INSERM, Marseille, U1251-13885, France
| | - Michel Pucéat
- LIA (International Associated Laboratory), CHU Sainte-Justine, Montréal, QC, Canada
- LIA (International Associated Laboratory), INSERM, Marseille, U1251-13885, France
- INSERM U-1251, MMG,Aix-Marseille University, Marseille, 13885, France
| | - Gregor Andelfinger
- Cardiovascular Genetics, Department of Pediatrics, CHU Sainte-Justine, Montréal, QC, Canada
| |
Collapse
|
29
|
Yu JZ, Ying Y, Liu Y, Sun CB, Dai C, Zhao S, Tian SZ, Peng J, Han NP, Yuan JL, Yan JY, Yang ZS. Antifibrotic action of Yifei Sanjie formula enhanced autophagy via PI3K-AKT-mTOR signaling pathway in mouse model of pulmonary fibrosis. Biomed Pharmacother 2019; 118:109293. [DOI: 10.1016/j.biopha.2019.109293] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/26/2019] [Accepted: 07/31/2019] [Indexed: 12/25/2022] Open
|
30
|
Sun W, Wu Y, Gao M, Tian Y, Qi P, Shen Y, Huang L, Shi L, Wang Y, Liu X. C-reactive protein promotes inflammation through TLR4/NF-κB/TGF-β pathway in HL-1 cells. Biosci Rep 2019; 39:BSR20190888. [PMID: 31391207 PMCID: PMC6712437 DOI: 10.1042/bsr20190888] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 07/25/2019] [Accepted: 08/02/2019] [Indexed: 01/08/2023] Open
Abstract
Atrial fibrillation (AF) is the most common type of heart arrhythmia. Currently, the pathogenesis of AF is not fully understood yet. A growing body of evidence highlighted the strong association between inflammation and the pathogenesis of AF. C-reactive protein (CRP) is an inflammation marker with increased expression in AF. Therefore, the aim of this study was to determine if CRP promotes inflammation, which may sequentially mediate the onset of AF and the concurrent atrial fibrosis, through TLR4/NF-κB/TGF-β pathway. HL-1 cells were treated with either 25 or 50 μg/ml recombinant human CRP. TGF-β1 and NF-κB inhibitors were given either solely or together to the 50 μg/ml CRP-treated cells. Cell proliferation, apoptosis, the expression of apoptotic factors and TLR4, IL-6, TGF-β1, Smad2, and the phosphorylation of Smad2 were determined. Data showed that CRP induced dose-dependent inhibition on cell proliferation and promoted cell apoptosis, which was induced through both intrinsic and extrinsic pathways. Such effects were reversed by inhibiting TGF-β1 and/or NF-κB. Inhibition of TGF-β1 and/or NF-κB also reduced the expression of TLR4 and IL-6. Inhibition of NF-κB alone weakened the expression of TGF-β1 and phosphorylation of Smad2. Our study demonstrated that CRP is not only a marker, but also an important mediator in the induction of inflammation and likely the pathogenesis of AF. We for the first time reported CRP-induced activation and cross-talk between TLR4 and NF-κB/TGF-β1 signaling pathway in a cardiomyocyte model. Reducing CRP and targeting TLR4/NF-κB/TGF-β1 pathway may provide new insights in the therapeutic interventions to inflammation-induced AF.
Collapse
Affiliation(s)
- Weiping Sun
- Department of Cardiology, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100037, China
- Department of Cardiology, Beijing Luhe Hospital, Capital Medical University, Beijing 100037, China
| | - Yongquan Wu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Mingyang Gao
- Department of Cardiology, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100037, China
| | - Ying Tian
- Department of Cardiology, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100037, China
| | - Peng Qi
- Department of Cardiology, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100037, China
| | - Yujing Shen
- Department of Cardiology, Fuwai Hospital, Chinese academy of Medicine sciences, Beijing 100037, China
| | - Lihong Huang
- Department of Cardiology, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100037, China
| | - Liang Shi
- Department of Cardiology, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100037, China
| | - Yanjiang Wang
- Department of Cardiology, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100037, China
| | - Xingpeng Liu
- Department of Cardiology, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100037, China
| |
Collapse
|
31
|
Ferreira RR, Abreu RDS, Vilar-Pereira G, Degrave W, Meuser-Batista M, Ferreira NVC, da Cruz Moreira O, da Silva Gomes NL, Mello de Souza E, Ramos IP, Bailly S, Feige JJ, Lannes-Vieira J, de Araújo-Jorge TC, Waghabi MC. TGF-β inhibitor therapy decreases fibrosis and stimulates cardiac improvement in a pre-clinical study of chronic Chagas' heart disease. PLoS Negl Trop Dis 2019; 13:e0007602. [PMID: 31365537 PMCID: PMC6690554 DOI: 10.1371/journal.pntd.0007602] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 08/12/2019] [Accepted: 07/03/2019] [Indexed: 01/11/2023] Open
Abstract
TGF-β involvement in Chagas disease cardiomyopathy has been clearly demonstrated. The TGF-β signaling pathway is activated in the cardiac tissue of chronic phase patients and is associated with an increase in extracellular matrix protein expression. The aim of this study was to investigate the effect of GW788388, a selective inhibitor of TβR1/ALK5, on cardiac function in an experimental model of chronic Chagas' heart disease. To this end, C57BL/6 mice were infected with Trypanosoma cruzi (102 parasites from the Colombian strain) and treated orally with 3mg/kg GW788388 starting at 120 days post-infection (dpi), when 100% of the infected mice show cardiac damage, and following three distinct treatment schedules: i) single dose; ii) one dose per week; or iii) three doses per week during 30 days. The treatment with GW788388 improved several cardiac parameters: reduced the prolonged PR and QTc intervals, increased heart rate, and reversed sinus arrhythmia, and atrial and atrioventricular conduction disorders. At 180 dpi, 30 days after treatment interruption, the GW3x-treated group remained in a better cardiac functional condition. Further, GW788388 treatment reversed the loss of connexin-43 enriched intercellular plaques and reduced fibrosis of the cardiac tissue. Inhibition of the TGF-β signaling pathway reduced TGF-β/pSmad2/3, increased MMP-9 and Sca-1, reduced TIMP-1/TIMP-2/TIMP-4, and partially restored GATA-6 and Tbox-5 transcription, supporting cardiac recovery. Moreover, GW788388 administration did not modify cardiac parasite load during the infection but reduced the migration of CD3+ cells to the heart tissue. Altogether, our data suggested that the single dose schedule was not as effective as the others and treatment three times per week during 30 days seems to be the most effective strategy. The therapeutic effects of GW788388 are promising and suggest a new possibility to treat cardiac fibrosis in the chronic phase of Chagas' heart disease by TGF-β inhibitors.
Collapse
Affiliation(s)
- Roberto Rodrigues Ferreira
- Laboratório de Genômica Funcional e Bioinformática—Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro RJ, Brasil
| | - Rayane da Silva Abreu
- Laboratório de Genômica Funcional e Bioinformática—Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro RJ, Brasil
| | - Glaucia Vilar-Pereira
- Laboratório de Biologia das Interações—Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro RJ, Brasil
| | - Wim Degrave
- Laboratório de Genômica Funcional e Bioinformática—Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro RJ, Brasil
| | - Marcelo Meuser-Batista
- Laboratório de Inovações em Terapias, Ensino e Bioprodutos—Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro RJ, Brasil
- Departamento de Anatomia Patológica e Citopatologia, Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brasil
| | - Nilma Valéria Caldeira Ferreira
- Departamento de Anatomia Patológica e Citopatologia, Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brasil
| | - Otacílio da Cruz Moreira
- Laboratório de Biologia Molecular e Doenças Endêmicas, Instituto Oswaldo Cruz (FIOCRUZ/RJ), Rio de Janeiro, Brazil
| | - Natália Lins da Silva Gomes
- Laboratório de Biologia Molecular e Doenças Endêmicas, Instituto Oswaldo Cruz (FIOCRUZ/RJ), Rio de Janeiro, Brazil
| | - Elen Mello de Souza
- Laboratório de Virologia Molecular—Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro RJ, Brasil
| | - Isalira P. Ramos
- UFRJ, Centro Nacional de Biologia Estrutural e Bioimagem, Rio de Janeiro, RJ, Brazil
| | - Sabine Bailly
- Université Grenoble-Alpes, Inserm, CEA, Biology of Cancer and Infection Laboratory, Grenoble, France
| | - Jean-Jacques Feige
- Université Grenoble-Alpes, Inserm, CEA, Biology of Cancer and Infection Laboratory, Grenoble, France
| | - Joseli Lannes-Vieira
- Laboratório de Biologia das Interações—Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro RJ, Brasil
| | - Tania C. de Araújo-Jorge
- Laboratório de Inovações em Terapias, Ensino e Bioprodutos—Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro RJ, Brasil
| | - Mariana Caldas Waghabi
- Laboratório de Genômica Funcional e Bioinformática—Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro RJ, Brasil
- * E-mail:
| |
Collapse
|
32
|
Effects of febuxostat on atrial remodeling in a rabbit model of atrial fibrillation induced by rapid atrial pacing. JOURNAL OF GERIATRIC CARDIOLOGY : JGC 2019; 16:540-551. [PMID: 31447893 PMCID: PMC6689522 DOI: 10.11909/j.issn.1671-5411.2019.07.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Febuxostat, a novel nonpurine selective inhibitor of xanthine oxidase (XO), may be used in the prevention and management of atrial fibrillation (AF). The purpose of this study was to evaluate the effects of febuxostat on atrial remodeling in a rabbit model of AF induced by rapid atrial pacing (RAP) and the mechanisms by which it acts. Methods Twenty-four rabbits were randomly divided into four groups: sham-operated group (Group S), RAP group (Group P), RAP with 5 mg/kg per day febuxostat group (Group LFP), and RAP with 10 mg/kg per day febuxostat group (Group HFP). All rabbits except those in Group S were subjected to RAP at 600 beats/min for four weeks. The effects of febuxostat on atrial electrical and structural remodeling, markers of inflammation and oxidative stress, and signaling pathways involved in the left atrium were examined. Results Shortened atrial effective refractory period (AERP), increased AF inducibility, decreased mRNA levels of Cav1.2 and Kv4.3, and left atrial enlargement and dysfunction were observed in Group P, and these changes were suppressed in the groups treated with febuxostat. Prominent atrial fibrosis was observed in Group P, as were increased levels of TGF-β1, Collagen I, and α-SMA and decreased levels of Smad7 and eNOS. Treatment with febuxostat attenuated these differences. Changes in inflammatory and oxidative stress markers induced by RAP were consistent with the protective effects of febuxostat. Conclusions This study is the first to find that febuxostat can inhibit atrial electrical and structural remodeling of AF by suppressing XO and inhibiting the TGF-β1/Smad signaling pathway.
Collapse
|
33
|
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
|
34
|
Wu YX, Han X, Chen C, Zou LX, Dong ZC, Zhang YL, Li HH. Time Series Gene Expression Profiling and Temporal Regulatory Pathway Analysis of Angiotensin II Induced Atrial Fibrillation in Mice. Front Physiol 2019; 10:597. [PMID: 31191333 PMCID: PMC6548816 DOI: 10.3389/fphys.2019.00597] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 04/26/2019] [Indexed: 11/24/2022] Open
Abstract
Background/Aim: Angiotensin II (Ang II) and hypertension play critical roles in the pathogenesis of the atrial remodeling that contributes to atrial fibrillation (AF). However, the gene expression profiles and signaling pathways in atria during the development of AF induced by Ang II remain unknown. Methods: Wild-type male mice (C57BL/6 background, 10 weeks old) were administered an infusion of Ang II (2000 ng/kg/min) using an osmotic pump for 1, 2, and 3 weeks. Blood pressure (BP) was measured by the tail-cuff method. AF was induced and recorded. Atrial enlargement and remodeling were examined by echocardiography and Masson’s trichrome staining. Time-series microarray analyses were conducted to examine gene expression profiles and pathways. Results: Ang II infusion resulted in marked elevation of systolic BP, increased AF incidence and duration, atrial enlargement, fibrosis, and atrial infiltration of myofibroblasts and F4/80-positive macrophages in a time-dependent manner. Microarray results showed that 1,719 genes were differentially expressed in the atrium at weeks 1, 2, and 3 after Ang II infusion. Gene ontology showed that these genes participate mainly in immune system processes, and regulation of cell migration, cell adhesion, complement activation, and the inflammatory response. Significant pathways included lysosomal and phagosomal pathways, which are involved in antigen processing and presentation, as well as chemokine signaling, and extracellular matrix–receptor interaction, which are known to play important roles in Ang II-induced AF. Moreover, these differentially expressed genes were classified into 50 profiles by hierarchical cluster analysis. Of these, eight profiles were significant and contained a total of 1,157 genes. Gene co-expression network analysis identified that Pik3cg (also known as phosphoinositide-3-kinase regulatory subunit 3) was localized in the core of the gene network, and was the most highly expressed among the Pik3 isoforms at different time points. Conclusion: The present findings revealed that many genes are involved in Ang II-induced AF, and highlighted that Pik3cg may play a central role in this disease.
Collapse
Affiliation(s)
- Yu-Xuan Wu
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China.,Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian, China
| | - Xiao Han
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian, China
| | - Chen Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian, China
| | - Lei-Xin Zou
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian, China
| | - Zhi-Chao Dong
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yun-Long Zhang
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China.,Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian, China
| | - Hui-Hua Li
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China.,Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian, China
| |
Collapse
|
35
|
Krisciunas GP, Vakharia A, Lazarus C, Taborda SG, Martino R, Hutcheson K, McCulloch T, Langmore SE. Application of Manual Therapy for Dysphagia in Head and Neck Cancer Patients: A Preliminary National Survey of Treatment Trends and Adverse Events. Glob Adv Health Med 2019; 8:2164956119844151. [PMID: 31041144 PMCID: PMC6482656 DOI: 10.1177/2164956119844151] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/24/2019] [Accepted: 03/11/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Radiation-associated dysphagia is a common and debilitating consequence of treatment for head and neck cancer (HNC). Since commonly employed dysphagia therapy programs for HNC patients still lack authoritative efficacy, some speech-language pathologists (SLPs) have started employing manual therapy (MT) techniques in an attempt to prevent or rehabilitate dysphagia in this patient population. However, exceptionally little is known about the use of MT in this patient population. OBJECTIVES The purpose of this study was to describe practice patterns as well as the rate, type, and severity of adverse events associated with SLP provision of MT to HNC patients. METHODS An Internet-based questionnaire geared toward SLPs who practice MT was developed and sent to SLPs practicing in the United States, 3 times, through 3 national listservs (American Speech Language Hearing Association [ASHA] Special Interest Division 13, ASHA Special Interest Division 3, and University of Iowa Voiceserv), over the course of 4 weeks. RESULTS Of the 255 respondents, 116 (45.5%) performed MT on HNC patients. Of these 116 SLPs, 27.6% provided proactive MT during radiation, 62.1% provided 1 to 2 sessions per week, and 94.8% prescribed a MT home program. The rate, type, and severity of reported adverse events were similar between HNC and non-HNC patients. CONCLUSION This preliminary survey demonstrated that SLPs provide MT to HNC patients during and after cancer treatment, and that reported adverse events paralleled those experienced by noncancer patients. However, these results should be taken with caution, and a well-designed prospective study is needed to formally establish the safety and the preliminary efficacy of this novel clinical intervention.
Collapse
Affiliation(s)
- Gintas P Krisciunas
- Department of Otolaryngology, Boston University Medical Center, Boston, Massachusetts
| | - Aneri Vakharia
- Department of Otolaryngology, Boston University Medical Center, Boston, Massachusetts
- Aneri Vakharia, Boston University Medical Center, BCD Building, 800 Harrison Avenue, Boston, MA 02218, USA.
| | - Cathy Lazarus
- Department of Otolaryngology, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | - Rosemary Martino
- Department of Otolaryngology, University of Toronto, Toronto, Ontario, Canada
| | - Katherine Hutcheson
- Department of Head & Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Timothy McCulloch
- Department of Otolaryngology, University of Wisconsin, Madison, Wisconsin
| | - Susan E Langmore
- Department of Otolaryngology, Boston University Medical Center, Boston, Massachusetts
| |
Collapse
|
36
|
Lakin R, Polidovitch N, Yang S, Guzman C, Gao X, Wauchop M, Burns J, Izaddoustdar F, Backx PH. Inhibition of soluble TNFα prevents adverse atrial remodeling and atrial arrhythmia susceptibility induced in mice by endurance exercise. J Mol Cell Cardiol 2019; 129:165-173. [DOI: 10.1016/j.yjmcc.2019.01.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 12/17/2018] [Accepted: 01/15/2019] [Indexed: 10/27/2022]
|
37
|
Umbarkar P, Singh AP, Gupte M, Verma VK, Galindo CL, Guo Y, Zhang Q, McNamara JW, Force T, Lal H. Cardiomyocyte SMAD4-Dependent TGF-β Signaling is Essential to Maintain Adult Heart Homeostasis. ACTA ACUST UNITED AC 2019; 4:41-53. [PMID: 30847418 PMCID: PMC6390466 DOI: 10.1016/j.jacbts.2018.10.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 08/10/2018] [Accepted: 08/10/2018] [Indexed: 12/25/2022]
Abstract
SMAD4 is the central intracellular mediator of TGF-β pathway. CM-specific loss of SMAD4 causes cardiac dysfunction independent of fibrotic remodeling. Deletion CM-SMAD4 affects CM survival. CM-SMAD4 loss leads to down-regulation of several ion channels’ genes, resulting in cardiac conduction abnormalities. CM-SMAD4 deletion alters sarcomere shortening kinetics, in parallel with reduction in cardiac myosin-binding protein C levels. These results demonstrate a fundamental role for CM-SMAD4–dependent TGF-β signaling in adult heart homeostasis.
The role of the transforming growth factor (TGF)-β pathway in myocardial fibrosis is well recognized. However, the precise role of this signaling axis in cardiomyocyte (CM) biology is not defined. In TGF-β signaling, SMAD4 acts as the central intracellular mediator. To investigate the role of TGF-β signaling in CM biology, the authors deleted SMAD4 in adult mouse CMs. We demonstrate that CM-SMAD4–dependent TGF-β signaling is critical for maintaining cardiac function, sarcomere kinetics, ion-channel gene expression, and cardiomyocyte survival. Thus, our findings raise a significant concern regarding the therapeutic approaches that rely on systemic inhibition of the TGF-β pathway for the management of myocardial fibrosis.
Collapse
Key Words
- CM, cardiomyocyte
- CSA, cross-sectional area
- CTL, control
- DCM, dilated cardiomyopathy
- KO, knockout
- LV, left ventricle/ventricular
- MAPK, mitogen-activated protein kinase
- MCM, MerCreMer
- PI3K, phosphoinositide-3 kinase
- RNA-Seq, RNA sequencing
- SMAD4
- TAK1, transforming growth factor beta–activated kinase 1
- TAM, tamoxifen
- TGF, transforming growth factor
- TGF-β
- cMyBP-C, cardiac myosin-binding protein C
- cardiomyocyte
- cardiomyopathy
- fibrosis
- heart failure
Collapse
Affiliation(s)
- Prachi Umbarkar
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Anand P Singh
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Manisha Gupte
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Vipin K Verma
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Cristi L Galindo
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Yuanjun Guo
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Pharmacology, Vanderbilt University, Nashville, Tennessee
| | - Qinkun Zhang
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - James W McNamara
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Thomas Force
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Hind Lal
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
38
|
Piché J, Gosset N, Legault LM, Pacis A, Oneglia A, Caron M, Chetaille P, Barreiro L, Liu D, Qi X, Nattel S, Leclerc S, Breton-Larrivée M, McGraw S, Andelfinger G. Molecular Signature of CAID Syndrome: Noncanonical Roles of SGO1 in Regulation of TGF-β Signaling and Epigenomics. Cell Mol Gastroenterol Hepatol 2018; 7:411-431. [PMID: 30739867 PMCID: PMC6369230 DOI: 10.1016/j.jcmgh.2018.10.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 10/17/2018] [Accepted: 10/17/2018] [Indexed: 01/14/2023]
Abstract
BACKGROUND & AIMS A generalized human pacemaking syndrome, chronic atrial and intestinal dysrhythmia (CAID) (OMIM 616201), is caused by a homozygous SGO1 mutation (K23E), leading to chronic intestinal pseudo-obstruction and arrhythmias. Because CAID patients do not show phenotypes consistent with perturbation of known roles of SGO1, we hypothesized that noncanonical roles of SGO1 drive the clinical manifestations observed. METHODS To identify a molecular signature for CAID syndrome, we achieved unbiased screens in cell lines and gut tissues from CAID patients vs wild-type controls. We performed RNA sequencing along with stable isotope labeling with amino acids in cell culture. In addition, we determined the genome-wide DNA methylation and chromatin accessibility signatures using reduced representative bisulfite sequencing and assay for transposase-accessible chromatin with high-throughput sequencing. Functional studies included patch-clamp, quantitation of transforming growth factor-β (TGF-β) signaling, and immunohistochemistry in CAID patient gut biopsy specimens. RESULTS Proteome and transcriptome studies converge on cell-cycle regulation, cardiac conduction, and smooth muscle regulation as drivers of CAID syndrome. Specifically, the inward rectifier current, an important regulator of cellular function, was disrupted. Immunohistochemistry confirmed overexpression of Budding Uninhibited By Benzimidazoles 1 (BUB1) in patients, implicating the TGF-β pathway in CAID pathogenesis. Canonical TGF-β signaling was up-regulated and uncoupled from noncanonical signaling in CAID patients. Reduced representative bisulfite sequencing and assay for transposase-accessible chromatin with high-throughput sequencing experiments showed significant changes of chromatin states in CAID, pointing to epigenetic regulation as a possible pathologic mechanism. CONCLUSIONS Our findings point to impaired inward rectifier potassium current, dysregulation of canonical TGF-β signaling, and epigenetic regulation as potential drivers of intestinal and cardiac manifestations of CAID syndrome. Transcript profiling and genomics data are as follows: repository URL: https://www.ncbi.nlm.nih.gov/geo; SuperSeries GSE110612 was composed of the following subseries: GSE110309, GSE110576, and GSE110601.
Collapse
Affiliation(s)
- Jessica Piché
- Cardiovascular Genetics, Department of Pediatrics, Centre Hospitalier Universitaire Sainte Justine Research Center, Université de Montréal, Montréal, Québec, Canada
| | - Natacha Gosset
- Cardiovascular Genetics, Department of Pediatrics, Centre Hospitalier Universitaire Sainte Justine Research Center, Université de Montréal, Montréal, Québec, Canada
| | - Lisa-Marie Legault
- Department of Biochemistry and Molecular Medicine, Centre Hospitalier Universitaire Sainte Justine Research Center, Université de Montréal, Montréal, Québec, Canada
| | - Alain Pacis
- Department of Genetics, Centre Hospitalier Universitaire Sainte Justine Research Center, Université de Montréal, Montréal, Québec, Canada,Department of Biochemistry, Université de Montréal, Montréal, Québec, Canada
| | - Andrea Oneglia
- Cardiovascular Genetics, Department of Pediatrics, Centre Hospitalier Universitaire Sainte Justine Research Center, Université de Montréal, Montréal, Québec, Canada
| | - Maxime Caron
- Centre Hospitalier Universitaire Sainte Justine Research Center, Université de Montréal, Montréal, Québec, Canada
| | - Philippe Chetaille
- Service of Pediatric Cardiology, Department of Pediatrics, Centre Mère Enfants Soleil, Centre Hospitalier de l’Université de Québec, Québec City, Québec, Canada
| | - Luis Barreiro
- Department of Genetics, Centre Hospitalier Universitaire Sainte Justine Research Center, Université de Montréal, Montréal, Québec, Canada,Department of Biochemistry, Université de Montréal, Montréal, Québec, Canada,Department of Pediatrics, Université de Montréal, Québec, Canada
| | - Donghai Liu
- Research Center, Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | - Xioyan Qi
- Research Center, Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | - Stanley Nattel
- Research Center, Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | - Séverine Leclerc
- Cardiovascular Genetics, Department of Pediatrics, Centre Hospitalier Universitaire Sainte Justine Research Center, Université de Montréal, Montréal, Québec, Canada
| | - Mélanie Breton-Larrivée
- Department of Biochemistry and Molecular Medicine, Centre Hospitalier Universitaire Sainte Justine Research Center, Université de Montréal, Montréal, Québec, Canada
| | | | - Serge McGraw
- Department of Biochemistry and Molecular Medicine, Centre Hospitalier Universitaire Sainte Justine Research Center, Université de Montréal, Montréal, Québec, Canada,Departement of Obstetrics and Gynecology, Centre Hospitalier Universitaire Sainte Justine Research Center, Université de Montréal, Montréal, Québec, Canada
| | - Gregor Andelfinger
- Cardiovascular Genetics, Department of Pediatrics, Centre Hospitalier Universitaire Sainte Justine Research Center, Université de Montréal, Montréal, Québec, Canada,Correspondence Address correspondence to: Gregor Andelfinger, MD, FRCPC, Service of Cardiology, Department of Pediatrics, Cardiovascular Genetics Research Laboratory, Centre Hospitalier Sainte Justine Research Center, Université de Montréal 3175, Chemin Côte Sainte Catherine, Montréal, Québec, H3T 1C5 Canada. fax: (514) 345-4896.
| |
Collapse
|
39
|
Wang J, Jia CH, Wang F, Xie ML. Stevioside inhibits transforming growth factor-β1-induced cardiac fibroblast differentiation and collagen synthesis by modulation of Smad signaling pathway. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.09.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
40
|
Zhan G, Wenhua G, Jie H, Xiang Q, Lingzhi C, Xue X, Xing-Xing C, Qianli Z, Weijian H, Hao Z. Potential roles of circulating matrix metalloproteinase-28 (MMP-28) in patients with atrial fibrillation. Life Sci 2018; 204:15-19. [DOI: 10.1016/j.lfs.2018.04.053] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Revised: 04/14/2018] [Accepted: 04/27/2018] [Indexed: 10/17/2022]
|
41
|
da Silva AMG, de Araújo JNG, de Oliveira KM, Novaes AEM, Lopes MB, de Sousa JCV, Filho AADA, Luchessi AD, de Rezende AA, Hirata MH, Silbiger VN. Circulating miRNAs in acute new-onset atrial fibrillation and their target mRNA network. J Cardiovasc Electrophysiol 2018; 29:1159-1166. [PMID: 29676832 DOI: 10.1111/jce.13612] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 04/14/2018] [Accepted: 04/17/2018] [Indexed: 02/05/2023]
Abstract
BACKGROUND MicroRNAs (miRNAs) are involved in the pathogenesis of atrial fibrillation (AF), acting on development and progression. Our pilot study investigated the expression of six miRNAs and their miRNA-mRNA interactions in patients with acute new-onset AF, well-controlled AF, and normal sinus rhythm (controls). METHODS AND RESULTS Plasma of acute new-onset AF patients (n = 5) was collected in the emergency room when patients presented with irregular and fast-atrial fibrillation rhythm. Samples from well-controlled AF (n = 16) and control (n = 15) patients were collected during medical appointments following an ECG. Expression of miR-21, miR-133a, miR-133b, miR-150, miR-328, and miR-499 was analyzed by real-time PCR. Ingenuity Pathway Analysis and the TargetScan database identified the top 30 mRNA targets of these miRNA, seeking the miRNA-mRNA interactions in cardiovascular process. Increased expression of miR-133b (1.4-fold), miR-328 (2.0-fold), and miR-499 (2.3-fold) was observed in patients with acute new-onset AF, compared with well-controlled AF and control patients. Decreased expression of miR-21 was seen in patients with well-controlled AF compared to those with acute new-onset AF and controls (0.6-fold). The miRNA-mRNA interaction demonstrated that SMAD7 and FASLG genes were the targets of miR-21, miR-133b, and miR-499 and were directly related to AF, being involved in apoptosis and fibrosis. CONCLUSION The miRNAs had different expression profiles dependent on the AF condition, with higher expression in the acute new-onset AF than well-controlled AF. Clinically, this may contribute to an effective assessment for patients, leading to early detection of AF and monitoring to reduce the risk of other serious cardiovascular events.
Collapse
Affiliation(s)
| | | | - Katiene Macêdo de Oliveira
- Department of Clinical Analysis and Toxicology, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Ana Eloísa Melo Novaes
- Department of Integrated Medicine, Hospital Onofre Lopes, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Mariana Borges Lopes
- Department of Clinical Analysis and Toxicology, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Júlio César Vieira de Sousa
- Department of Integrated Medicine, Hospital Onofre Lopes, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | | | - André Ducati Luchessi
- Department of Clinical Analysis and Toxicology, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Adriana Augusto de Rezende
- Department of Clinical Analysis and Toxicology, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Mário Hiroyuki Hirata
- Department of Clinical Analysis and Toxicology, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Vivian Nogueira Silbiger
- Department of Clinical Analysis and Toxicology, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| |
Collapse
|
42
|
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
|
43
|
The role of pro-fibrotic biomarkers in paroxysmal and persistent atrial fibrillation. Cytokine 2018; 103:63-68. [DOI: 10.1016/j.cyto.2017.12.026] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 12/14/2017] [Accepted: 12/26/2017] [Indexed: 01/05/2023]
|
44
|
Jiang H, Wang W, Wang C, Xie X, Hou Y. Association of pre-ablation level of potential blood markers with atrial fibrillation recurrence after catheter ablation: a meta-analysis. Europace 2017; 19:392-400. [PMID: 27386883 DOI: 10.1093/europace/euw088] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 03/09/2016] [Indexed: 11/13/2022] Open
Abstract
Aims The meta-analysis was aimed to search for candidate blood markers whose pre-ablation level was associated with atrial fibrillation (AF) recurrence after radiofrequency catheter ablation (RFCA). Methods and results A systematic literature search of PubMed, EMBASE, Springer Link, Web of Science, Wiley-Cochrane library, and supplemented with Google scholar search engine was performed. Thirty-six studies covering 11 blood markers were qualified for this meta-analysis. Compared with the nonrecurrence group, the recurrence group had increased pre-ablation level of atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), N-terminal pro-brain natriuretic peptide (NT-pro-BNP), interleukin-6 (IL-6), C-reactive protein, low density lipoprotein (LDL), and tissue inhibitor of metal loproteinase-2 (TIMP-2) [standardized mean difference (95% confidence interval): 0.37 (0.13-0.61), 0.77 (0.40-1.14), 1.25 (0.64-1.87), 0.37 (0.21-0.52), 0.35 (0.10-0.60), 0.24 (0.07-0.42), 0.17 (0.00-0.34), respectively], while no statistical difference of pre-ablation level of white blood cell, total cholesterol, triglyceride, and transforming growth factor-β1 was found. Subgroup analysis demonstrated that ANP was associated with AF recurrence in participants who had no concomitant structural heart diseases (SHD); however, not in participants who had SHD, C-reactive protein was associated with AF recurrence in Asian studies, whereas not in European studies. Conclusion Increased pre-ablation level of ANP, BNP, NT-pro-BNP, IL-6, C-reactive protein, LDL, and TIMP-2 was associated with greater risk of AF recurrence after RFCA.
Collapse
Affiliation(s)
- Hui Jiang
- Department of Clinical Medicine, School of Medicine, Shandong University, Jinan, Shandong Province, China.,Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, No. 16766 Jingshi Road, Jinan, Shandong Province 250014, China
| | - Weizong Wang
- Department of Clinical Medicine, School of Medicine, Shandong University, Jinan, Shandong Province, China.,Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, No. 16766 Jingshi Road, Jinan, Shandong Province 250014, China
| | - Cong Wang
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, No. 16766 Jingshi Road, Jinan, Shandong Province 250014, China
| | - Xinxing Xie
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, No. 16766 Jingshi Road, Jinan, Shandong Province 250014, China
| | - Yinglong Hou
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, No. 16766 Jingshi Road, Jinan, Shandong Province 250014, China
| |
Collapse
|
45
|
Jeon CS, Shim MS, Park SJ, Jeong DS, Park KM, On YK, Kim JS, Park PW. The Absence of Atrial Contraction as a Predictor of Permanent Pacemaker Implantation after Maze Procedure with Cryoablation. THE KOREAN JOURNAL OF THORACIC AND CARDIOVASCULAR SURGERY 2017; 50:163-170. [PMID: 28593151 PMCID: PMC5460962 DOI: 10.5090/kjtcs.2017.50.3.163] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 12/23/2016] [Accepted: 12/27/2016] [Indexed: 11/16/2022]
Abstract
BACKGROUND The absence of atrial contraction (AC) after the maze procedure has been reported to cause subsequent annular dilatation and to increase the risk of embolic stroke. We hypothesized that the lack of AC could increase the risk of permanent pacemaker (PPM) implantation in patients undergoing the maze procedure. METHODS In 376 consecutive patients who had undergone a cryo-maze procedure and combined valve operation, recovery of AC was assessed at baseline and at immediate (≤2 weeks), early (≤1 year, 4.6±3.8 months), and late (>1 year, 3.5±1.1 years) postoperative stages. RESULTS With a median follow-up of 53 months, 10 patients underwent PPM implantation. Seven PPM implants were for sinus node dysfunction (pauses of 9.6±2.4 seconds), one was for marked sinus bradycardia, and two were for advanced/complete atrioventricular block. The median (interquartile range) time to PPM implantation was 13.8 (0.5-68.2) months. Our time-varying covariate Cox models showed that the absence of AC was a risk factor for PPM implantation (hazard ratio, 11.92; 95% confidence interval, 2.52 to 56.45; p=0.002). CONCLUSION The absence of AC may be associated with a subsequent risk of PPM implantation.
Collapse
Affiliation(s)
- Chang-Seok Jeon
- Department of Thoracic and Cardiovascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine
| | - Man-Shik Shim
- Department of Thoracic and Cardiovascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine
| | - Seung-Jung Park
- Division of Cardiology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine
| | - Dong Seop Jeong
- Department of Thoracic and Cardiovascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine
| | - Kyoung-Min Park
- Division of Cardiology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine
| | - Young Keun On
- Division of Cardiology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine
| | - June Soo Kim
- Division of Cardiology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine
| | - Pyo Won Park
- Department of Thoracic and Cardiovascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine
| |
Collapse
|
46
|
Qiao G, Xia D, Cheng Z, Zhang G. Role of Sprouty1 (Spry1) in the pathogenesis of atrial fibrosis. Pathol Res Pract 2017; 214:308-313. [PMID: 29096943 DOI: 10.1016/j.prp.2017.04.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 03/28/2017] [Accepted: 04/20/2017] [Indexed: 11/19/2022]
Abstract
Atrial fibrosis is the hallmark of atrial fibrillation (AF) dependent structure remodeling. Besides, sprouty 1 (Spry1) plays a key role in the process of fibrosis. In this study, we investigated whether Spry1 could regulate TGF-β1 in atrial fibrosis. Ten dogs or patients were assigned to control (n=4) and AF group (n=6). The left atrium of dogs or right atrial appendage of patients was taken. After that, cardiac fibroblasts were treated with or without angiotensin II (Ang II). Furthermore, cardiac fibroblasts were transfected with lentivirus of Spry1 over-expression vector, Spry1 shRNA or negative control (NC). And the protein expression of Spry1 and TGF-β1 was analyzed by western blot and immunohistochemistry. The results showed that TGF-β1 was highly expressed while Spry1 was lowly expressed in the models of human and canine with AF. Besides, the protein expression of TGF-β1 was up-regulated and Spry1 was down-regulated in Ang II stimulated cardiac fibroblasts. Furthermore, when Spry1 was knockdown in Ang II-induced cardiac fibroblasts, the cell proliferation and the TGF-β1 protein expression increased significantly, while Spry1 over-expression showed inverse results. Our results demonstrated that Spry1 may target TGF-β1 in regulating fibrosis. These findings may provide possible therapeutic targets in atrial fibrosis.
Collapse
Affiliation(s)
- Gang Qiao
- Department of Cardiovascular Surgery, Henan Provincial Hospital, Zhengzhou University 450003, Zhengzhou, China
| | - Dongsheng Xia
- Department of Cardiovascular Surgery, Henan Provincial Hospital, Zhengzhou University 450003, Zhengzhou, China
| | - Zhaoyun Cheng
- Department of Cardiovascular Surgery, Henan Provincial Hospital, Zhengzhou University 450003, Zhengzhou, China
| | - Guobao Zhang
- Department of Cardiovascular Surgery, Henan Provincial Hospital, Zhengzhou University 450003, Zhengzhou, China.
| |
Collapse
|
47
|
Impact of Compliance on Dysphagia Rehabilitation in Head and Neck Cancer Patients: Results from a Multi-center Clinical Trial. Dysphagia 2016; 32:327-336. [PMID: 27848021 DOI: 10.1007/s00455-016-9760-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 11/08/2016] [Indexed: 10/20/2022]
Abstract
A 5-year, 16-site, randomized controlled trial enrolled 170 HNC survivors into active (estim + swallow exercise) or control (sham estim + swallowing exercise) arms. Primary analyses showed that estim did not enhance swallowing exercises. This secondary analysis determined if/how patient compliance impacted outcomes. A home program, performed 2 times/day, 6 days/week, for 12 weeks included stretches and 60 swallows paired with real or sham estim. Regular clinic visits ensured proper exercise execution, and detailed therapy checklists tracked patient compliance which was defined by mean number of sessions performed per week (0-12 times) over the 12-week intervention period. "Compliant" was defined as performing 10-12 sessions/week. Outcomes were changes in PAS, HNCI, PSS, OPSE, and hyoid excursion. ANCOVA analyses determined if outcomes differed between real/sham and compliant/noncompliant groups after 12 weeks of therapy. Of the 170 patients enrolled, 153 patients had compliance data. The mean number of sessions performed was 8.57/week (median = 10.25). Fifty-four percent of patients (n = 83) were considered "compliant." After 12 weeks of therapy, compliant patients in the sham estim group realized significantly better PAS scores than compliant patients in the active estim group (p = 0.0074). When pooling all patients together, there were no significant differences in outcomes between compliant and non-compliant patients. The addition of estim to swallowing exercises resulted in worse swallowing outcomes than exercises alone, which was more pronounced in compliant patients. Since neither compliant nor non-compliant patients benefitted from swallowing exercises, the proper dose and/or efficacy of swallowing exercises must also be questioned in this patient population.
Collapse
|
48
|
Abstract
Postoperative atrial fibrillation (PoAF), a common complication of cardiac surgery, contributes significantly to morbidity, mortality, and increasing healthcare costs. Despite advances in surgical and medical management, the overall incidence of PoAF has not changed significantly, partly because of the limited understanding of mechanisms underlying acute surgery-related factors, such as myocardial injury, inflammation, sympathetic activation, and oxidative stress, which play an important role in the initiation of PoAF, whereas a preexisting atrial substrate appears to be more important in the maintenance of this dysrhythmia. Thus, in a majority of patients, PoAF becomes a manifestation of an underlying arrhythmogenic substrate that is unmasked after acute surgical stress. As such, the ability to identify which patients have this proarrhythmic substrate and are, therefore, at high risk for developing AF postoperatively, is important for the improved selection for prophylactic interventions, closer monitoring for complications, and establishing the probability of AF in the long term. This review highlights the role of the underlying substrate in promoting PoAF, proposed mechanisms, and the potential role of serum biomarkers to identify patients at risk for PoAF.
Collapse
|
49
|
Jia W, Qi X, Li Q. Association Between Rs3807989 Polymorphism in Caveolin-1 (CAV1) Gene and Atrial Fibrillation: A Meta-Analysis. Med Sci Monit 2016; 22:3961-3966. [PMID: 27775682 PMCID: PMC5091217 DOI: 10.12659/msm.896826] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Atrial fibrillation (AF) is the most common sustained arrhythmia affected by multiple cardiovascular risk factors. It is reported that caveolin-1 gene (CAV1) rs3807989 polymorphism might be associated with AF risk. The goal of this meta-analysis was to confirm the association between CAV1 rs3807989 polymorphism and susceptibility to AF. Material/Methods We carried out a comprehensive literature search through the electronic databases PubMed, MEDLIN, and Web of Science. We performed a meta-analysis of all selected studies based on CAV1 rs3807989 polymorphism genotypes, including 3758 cases and 6126 controls. Results After meta-analysis with fixed- or random-effects models, we found significant associations in all 5 comparisons: allelic model (G/A; OR=1.228, 95%CI: 1.061–1.420; P=0.006), homozygote model (GG/AA; OR=1.439, 95%CI: 1.094–1.894; P=0.009), heterozygote model (GG/GA; OR=1.257, 95%CI: 1.064–1.486; P=0.007), dominant model (GG/AA+GA; OR=1.287, 95%CI: 1.076–1.540; P=0.006), and recessive model (AA/GA+GG; OR=0.738, 95%CI: 0.629–0.867; P<0.001). Sensitivity analysis results revealed the overall results were robust. Conclusions The results revealed a significant association between CAV1 gene rs3807989 polymorphism and susceptibility to AF, suggesting that the presence of allelic G might be one of the genetic factors conferring susceptibility to AF. To confirm this association, further well-designed studies are necessary.
Collapse
Affiliation(s)
- Wenjun Jia
- Department of Cardiology, Tianjin Union Medical Center, Tianjin, China (mainland)
| | - Xin Qi
- Department of Cardiology, Tianjin Union Medical Center, Tianjin, China (mainland)
| | - Qi Li
- Department of Cardiology, Tianjin Union Medical Center, Tianjin, China (mainland)
| |
Collapse
|
50
|
Kornej J, Ueberham L, Schmidl J, Husser D, Adams V, Hindricks G, Bollmann A. Addition of TGF-β1 to existing clinical risk scores does not improve prediction for arrhythmia recurrences after catheter ablation of atrial fibrillation. Int J Cardiol 2016; 221:52-4. [PMID: 27400297 DOI: 10.1016/j.ijcard.2016.06.290] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 06/28/2016] [Indexed: 10/21/2022]
Affiliation(s)
- Jelena Kornej
- University of Leipzig, Heart Center, Department of Electrophysiology, Leipzig, Germany.
| | - Laura Ueberham
- University of Leipzig, Heart Center, Department of Electrophysiology, Leipzig, Germany
| | - Josephin Schmidl
- University of Leipzig, Heart Center, Department of Electrophysiology, Leipzig, Germany
| | - Daniela Husser
- University of Leipzig, Heart Center, Department of Electrophysiology, Leipzig, Germany
| | - Volker Adams
- University of Leipzig, Heart Center, Department of Cardiology, Leipzig, Germany
| | - Gerhard Hindricks
- University of Leipzig, Heart Center, Department of Electrophysiology, Leipzig, Germany
| | - Andreas Bollmann
- University of Leipzig, Heart Center, Department of Electrophysiology, Leipzig, Germany
| |
Collapse
|