1
|
Abbas M, Gaye A. Emerging roles of noncoding RNAs in cardiovascular pathophysiology. Am J Physiol Heart Circ Physiol 2025; 328:H603-H621. [PMID: 39918596 DOI: 10.1152/ajpheart.00681.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/17/2024] [Accepted: 01/30/2025] [Indexed: 02/27/2025]
Abstract
This review comprehensively examines the diverse roles of noncoding RNAs (ncRNAs) in the pathogenesis and treatment of cardiovascular disease (CVD), focusing on microRNA (miRNA), long noncoding RNA (lncRNA), piwi-interacting RNA (piRNA), small-interfering RNA (siRNA), circular RNA (circRNA), and vesicle-associated RNAs. These ncRNAs are integral regulators of key cellular processes, including gene expression, inflammation, and fibrosis, and they hold great potential as both diagnostic biomarkers and therapeutic targets. The review highlights novel insights into how these RNA species, particularly miRNAs, lncRNAs, and piRNAs, contribute to various CVDs such as hypertension, atherosclerosis, and myocardial infarction. In addition, it explores the emerging role of extracellular vesicles (EVs) in intercellular communication and their therapeutic potential in cardiovascular health. The review underscores the need for continued research into ncRNAs and RNA-based therapies, with a focus on advancing delivery systems and expanding personalized medicine approaches to improve cardiovascular outcomes.
Collapse
Affiliation(s)
- Malak Abbas
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States
| | - Amadou Gaye
- Department of Integrative Genomics and Epidemiology, School of Graduate Studies, Meharry Medical School, Nashville, Tennessee, United States
| |
Collapse
|
2
|
Bai S, Wang X, Hou Y, Cui Y, Song Q, Du J, Zhang Y, Xu J. lncRNA-056298 Regulates GAP43 and Promotes Cardiac Intrinsic Autonomic Nerve Remodelling in a Canine Model of Atrial Fibrillation Induction after Ganglionated Plexus Ablation. Curr Med Chem 2025; 32:136-159. [PMID: 38299396 DOI: 10.2174/0109298673289298240129103537] [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: 10/25/2023] [Revised: 12/20/2023] [Accepted: 01/11/2024] [Indexed: 02/02/2024]
Abstract
BACKGROUND Cardiac intrinsic autonomic nerve remodelling has been reported to play an important role in the recurrence of atrial fibrillation after radiofrequency ablation, which significantly affects the long-term efficacy of this procedure. lncRNAs have been shown to interact in the pathological processes underlying heart diseases. However, the roles and mechanisms of lncRNAs in cardiac intrinsic autonomic nerve remodelling during atrial fibrillation reduction after ganglionated plexus ablation remain unknown. OBJECTIVE The aim of this study was to investigate the mechanism by which lncRNA- 056298 modulates GAP43 to affect cardiac intrinsic autonomic nerve remodelling and facilitate the induction of atrial fibrillation after ganglionated plexus ablation. METHODS A canine model of right atrial ganglionated plexus ablation was established. The atrial electrophysiological characteristics and neural markers were detected before and after 6 months of ganglionated plexus ablation. High-throughput sequencing was used to screen differentially expressed lncRNAs in target atrial tissues, and lncRNA- 056298 was selected to further explore its effects and mechanisms on cardiac intrinsic autonomic nerve remodelling. RESULTS The induction rate of atrial fibrillation increased in dogs after ganglionated plexus ablation. Overexpression of lncRNA-056298 by lentivirus can shorten the atrial effective refractory period and increase the induction of atrial fibrillation. lncRNA- 056298 promoted cardiac intrinsic autonomic nerve remodelling via endogenous competition with cfa-miR-185 to induce transcription of its target gene GAP43, thereby affecting the induction of atrial fibrillation. CONCLUSION lncRNA-056298 regulates GAP43 by sponging miR-185, which affects cardiac intrinsic autonomic nerve remodelling and mediates atrial fibrillation induction after ganglionated plexus ablation.
Collapse
Affiliation(s)
- Shuting Bai
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Ximin Wang
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Yinglong Hou
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Yansong Cui
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Qiyuan Song
- Department of Cardiology, Shandong First Medical University, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Juanjuan Du
- Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Yujiao Zhang
- Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Jingwen Xu
- Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| |
Collapse
|
3
|
Li S, Xu Z, Zhang S, Sun H, Qin X, Zhu L, Jiang T, Zhou J, Yan F, Deng Q. Non-coding RNAs in acute ischemic stroke: from brain to periphery. Neural Regen Res 2025; 20:116-129. [PMID: 38767481 PMCID: PMC11246127 DOI: 10.4103/nrr.nrr-d-23-01292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 11/09/2023] [Accepted: 12/18/2023] [Indexed: 05/22/2024] Open
Abstract
Acute ischemic stroke is a clinical emergency and a condition with high morbidity, mortality, and disability. Accurate predictive, diagnostic, and prognostic biomarkers and effective therapeutic targets for acute ischemic stroke remain undetermined. With innovations in high-throughput gene sequencing analysis, many aberrantly expressed non-coding RNAs (ncRNAs) in the brain and peripheral blood after acute ischemic stroke have been found in clinical samples and experimental models. Differentially expressed ncRNAs in the post-stroke brain were demonstrated to play vital roles in pathological processes, leading to neuroprotection or deterioration, thus ncRNAs can serve as therapeutic targets in acute ischemic stroke. Moreover, distinctly expressed ncRNAs in the peripheral blood can be used as biomarkers for acute ischemic stroke prediction, diagnosis, and prognosis. In particular, ncRNAs in peripheral immune cells were recently shown to be involved in the peripheral and brain immune response after acute ischemic stroke. In this review, we consolidate the latest progress of research into the roles of ncRNAs (microRNAs, long ncRNAs, and circular RNAs) in the pathological processes of acute ischemic stroke-induced brain damage, as well as the potential of these ncRNAs to act as biomarkers for acute ischemic stroke prediction, diagnosis, and prognosis. Findings from this review will provide novel ideas for the clinical application of ncRNAs in acute ischemic stroke.
Collapse
Affiliation(s)
- Shuo Li
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Zhaohan Xu
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Shiyao Zhang
- Department of Neurology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| | - Huiling Sun
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xiaodan Qin
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Lin Zhu
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Teng Jiang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Junshan Zhou
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Fuling Yan
- Department of Neurology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| | - Qiwen Deng
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
4
|
Xi S, Wang H, Chen J, Gan T, Zhao L. LncRNA GAS5 Attenuates Cardiac Electrical Remodeling Induced by Rapid Pacing via the miR-27a-3p/HOXa10 Pathway. Int J Mol Sci 2023; 24:12093. [PMID: 37569470 PMCID: PMC10419054 DOI: 10.3390/ijms241512093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/13/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
Previous studies indicated long non-coding RNAs (lncRNAs) participated in the pathogenesis of atrial fibrillation (AF). However, little is known about the role of lncRNAs in AF-induced electrical remodeling. This study aimed to investigate the regulatory effect of lncRNA GAS5 (GAS5) on the electrical remodeling of neonatal rat cardiomyocytes (NRCMs) induced by rapid pacing (RP). RNA microarray analysis yielded reduced GAS5 level in NRCMs after RP. RT-qPCR, western blot, and immunofluorescence yielded downregulated levels of Nav1.5, Kv4.2, and Cav1.2 after RP, and whole-cell patch-clamp yielded decreased sodium, potassium, and calcium current. Overexpression of GAS5 attenuated electrical remodeling. Bioinformatics tool prediction analysis and dual luciferase reporter assay confirmed a direct negative regulatory effect for miR-27a-3p on lncRNA-GAS5 and HOXa10. Further analysis demonstrated that either miR-27a-3p overexpression or the knockdown of HOXa10 further downregulated Nav1.5, Kv4.2, and Cav1.2 expression. GAS5 overexpression antagonized such effects in Nav1.5 and Kv4.2 but not in Cav1.2. These results indicate that, in RP-treated NRCMs, GAS5 could restore Nav1.5 and Kv4.2 expression via the miR-27a-3p/HOXa10 pathway. However, the mechanism of GAS5 restoring Cav1.2 level remains unclear. Our study suggested that GAS5 regulated cardiac ion channels via the GAS5/miR-27a-3p/HOXa10 pathway and might be a potential therapeutic target for AF.
Collapse
Affiliation(s)
| | | | | | | | - Liang Zhao
- Department of Cardiology, Shanghai Chest Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai 200003, China; (S.X.); (H.W.); (J.C.); (T.G.)
| |
Collapse
|
5
|
Ding J, Cao Y, Qi C, Zong Z. Dysregulated microRNAs participate in the crosstalk between colorectal cancer and atrial fibrillation. Hum Cell 2023:10.1007/s13577-023-00899-2. [PMID: 36964414 DOI: 10.1007/s13577-023-00899-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 03/16/2023] [Indexed: 03/26/2023]
Abstract
Colorectal cancer and atrial fibrillation share several common risk factors, and the incidence of the two diseases also exhibits a certain correlation. The above facts suggest a potential interaction mechanism between them, which has obtained increasing attention in the scientific community but remains to be further explored. Participating in diverse physiological and pathological processes, miRNAs exert important roles in both occurrence and growth of colorectal cancer and atrial fibrillation. To fill the gap in the understanding of the potential linkage between two diseases, the present study collected dysregulated miRNAs of colorectal cancer and atrial fibrillation from previous studies and then selected the miRNAs with the same change trends in both diseases. Finally, we reviewed the potential crosstalk of two diseases focusing on the roles of 6 dysregulated miRNAs, including 3 co-downregulated miRNAs (hsa-mir-126, hsa-mir-133a and hsa-mir-150) and 3 co-upregulated miRNAs (hsa-mir-106a, hsa-mir-155 and hsa-mir-21). The molecular mechanisms mediated by these miRNAs in colorectal cancer and atrial fibrillation were reviewed, and the possible crosstalk between the two diseases was discussed from the perspective of miRNAs. This study also provides potential common targets for preventive and curative measures against both colorectal cancer and atrial fibrillation.
Collapse
Affiliation(s)
- Jiatong Ding
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, 1 MinDe Road, Nanchang, 330006, China
- The Second Clinical Medicine School, Nanchang University, Nanchang, 330006, China
| | - Yuke Cao
- School of Ophthalmology and Optometry, Nanchang University, Nanchang, 330006, China
| | - Chaofan Qi
- The First Clinical Medicine School, Nanchang University, Nanchang, 330006, China
| | - Zhen Zong
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, 1 MinDe Road, Nanchang, 330006, China.
| |
Collapse
|
6
|
Long non-coding RNA and circular RNA: new perspectives for molecular pathophysiology of atrial fibrillation. Mol Biol Rep 2023; 50:2835-2845. [PMID: 36596997 DOI: 10.1007/s11033-022-08216-0] [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: 07/12/2022] [Accepted: 12/15/2022] [Indexed: 01/05/2023]
Abstract
Many studies have demonstrated the association of atrial fibrillation (AF) with endogenous genetic regulatory mechanisms. These interactions could advance the understanding of the AF pathophysiological process, supporting the search for early biomarkers to improve diagnosis and disease monitoring. Among the endogenous genetic regulatory mechanisms, long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) have gained special attention, and studies have demonstrated their involvement in AF development and other AF-related diseases such as coronary artery disease and cardiomyopathy. This review describes the main experimental results reported by studies that analyzed the expression of lncRNAs and circRNAs in AF associated with miRNA or mRNA. The search was conducted in PubMed public database using the terms "lncRNA and atrial fibrillation" or "long ncRNA and atrial fibrillation" or "long non-coding RNA and atrial fibrillation" or "circular RNA and atrial fibrillation" or "circRNA and atrial fibrillation". There was no overlapping of lncRNA or circRNA among the studies, attributed to the different sample types, methods, species, and patient classification evaluated in these studies. Although the regulatory mechanisms in which these molecules are involved are not yet well understood, the studies analyzed show their importance in the pathophysiological process of AF, supporting the idea that lncRNAs and circRNAs are involved in miRNA or mRNA regulation in the molecular mechanism of this disease.
Collapse
|
7
|
Emami Meybodi SM, Soleimani N, Yari A, Javadifar A, Tollabi M, Karimi B, Emami Meybodi M, Seyedhossaini S, Brouki Milan P, Dehghani Firoozabadi A. Circulatory long noncoding RNAs (circulatory-LNC-RNAs) as novel biomarkers and therapeutic targets in cardiovascular diseases: Implications for cardiovascular diseases complications. Int J Biol Macromol 2023; 225:1049-1071. [PMID: 36414082 DOI: 10.1016/j.ijbiomac.2022.11.167] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/16/2022] [Indexed: 11/21/2022]
Abstract
Cardiovascular diseases (CVDs) are a group of disorders with major global health consequences. The prevalence of CVDs continues to grow due to population-aging and lifestyle modifications. Non-coding RNAs (ncRNAs) as key regulators of cell signaling pathways have gained attention in the occurrence and development of CVDs. Exosomal-lncRNAs (exos-lncRNAs) are emerging biomarkers due to their high sensitivity and specificity, stability, accuracy and accessibility in the biological fluids. Recently, circulatory and exos-based-lncRNAs are emerging and novel bio-tools in various pathogenic conditions. It is worth mentioning that dysregulation of these molecules has been found in different types of CVDs. In this regard, we aimed to discuss the knowledge gaps and suggest research priorities regarding circulatory and exos-lncRNAs as novel bio-tools and therapeutic targets for CVDs.
Collapse
Affiliation(s)
- Seyed Mahdi Emami Meybodi
- Yazd Cardiovascular Research Center, Non-communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| | - Nafiseh Soleimani
- Yazd Cardiovascular Research Center, Non-communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| | - Abolfazl Yari
- Cellular and Molecular Research Center, Birjand University of Medical Mciences, Birjand, Iran.
| | - Amin Javadifar
- Immunology Research Center, Inflammation and Inflammatory Disease Division, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mohammad Tollabi
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran; Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Bahareh Karimi
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| | - Mahmoud Emami Meybodi
- Yazd Cardiovascular Research Center, Non-communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| | - Seyedmostafa Seyedhossaini
- Yazd Cardiovascular Research Center, Non-communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| | - Peiman Brouki Milan
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran; Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Ali Dehghani Firoozabadi
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran; Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Wang M, An G, Wang B, Chen Y, Liu G, Wang X, Liu S, Zhang D, Sun D, Zhang Y, Shen T, Li X. Integrated analysis of the lncRNA-miRNA-mRNA network based on competing endogenous RNA in atrial fibrillation. Front Cardiovasc Med 2023; 10:1099124. [PMID: 37180786 PMCID: PMC10174322 DOI: 10.3389/fcvm.2023.1099124] [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: 11/15/2022] [Accepted: 04/07/2023] [Indexed: 05/16/2023] Open
Abstract
Objective Long non-coding RNAs (lncRNAs) play pivotal roles in the transcriptional regulation of atrial fibrillation (AF) by acting as competing endogenous RNAs (ceRNAs). In the present study, the expression levels of lncRNAs of sinus rhythm (SR) patients and AF patients were investigated with transcriptomics technology, and the lncRNA-miRNA-mRNA network based on the ceRNA theory in AF was elaborated. Methods Left atrial appendage (LAA) tissues were obtained from patients with valvular heart disease during cardiac surgery, and they were divided into SR and AF groups. The expression characterizations of differentially expressed (DE) lncRNAs in the two groups were revealed by high-throughput sequencing methods. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed, and the lncRNA-miRNA-mRNA-mediated ceRNA network was constructed. Results A total of differentially expressed 82 lncRNAs, 18 miRNAs, and 495 mRNAs in human atrial appendage tissues were targeted. Compared to SR patients, the following changes were found in AF patients: 32 upregulated and 50 downregulated lncRNAs; 7 upregulated and 11 downregulated miRNAs; and 408 upregulated and 87 downregulated mRNAs. A lncRNA-miRNA-mRNA network was constructed, which included 44 lncRNAs, 18 miRNAs, and 347 mRNAs. qRT-PCR was performed to verify these findings. GO and KEGG analyses suggested that inflammatory response, chemokine signaling pathway, and other biological processes play important roles in the pathogenesis of AF. Network analysis based on the ceRNA theory identified that lncRNA XR_001750763.2 and Toll-like receptor 2 (TLR2) compete for binding to miR-302b-3p. In AF patients, lncRNA XR_001750763.2 and TLR2 were upregulated, and miR-302b-3p was downregulated. Conclusion We identified a lncRNA XR_001750763.2/miR-302b-3p/TLR2 network based on the ceRNA theory in AF. The present study shed light on the physiological functions of lncRNAs and provided information for exploring potential treatments for AF.
Collapse
Affiliation(s)
- Manman Wang
- Jining Key Laboratory for Diagnosis and Treatment of Cardiovascular Diseases, Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, China
- Correspondence: Manman Wang Xiangting Li
| | - Guoying An
- Shandong Provincial Key Laboratory of Cardiac Disease Diagnosis and Treatment, Department of Cardiac Surgery, Affiliated Hospital of Jining Medical University, Jining, China
| | - Benxuan Wang
- Department of Neurology, Jinnan Hospital, Tianjin, China
| | - Yuanyuan Chen
- Jining Key Laboratory for Diagnosis and Treatment of Cardiovascular Diseases, Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Genli Liu
- Jining Key Laboratory for Diagnosis and Treatment of Cardiovascular Diseases, Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Xin Wang
- Jining Key Laboratory for Diagnosis and Treatment of Cardiovascular Diseases, Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Shuai Liu
- Jining Key Laboratory for Diagnosis and Treatment of Cardiovascular Diseases, Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Daozou Zhang
- Jining Key Laboratory for Diagnosis and Treatment of Cardiovascular Diseases, Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Dandan Sun
- Jining Key Laboratory for Diagnosis and Treatment of Cardiovascular Diseases, Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Yanyan Zhang
- Admission and Patient Service Center, Affiliated Hospital of Jining Medical University, Jining, China
| | - Tong Shen
- Jining Key Laboratory for Diagnosis and Treatment of Cardiovascular Diseases, Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Xiangting Li
- Jining Key Laboratory for Diagnosis and Treatment of Cardiovascular Diseases, Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, China
- Correspondence: Manman Wang Xiangting Li
| |
Collapse
|
9
|
Kang JY, Mun D, Kim H, Yun N, Joung B. Serum exosomal long non-coding RNAs as a diagnostic biomarker for atrial fibrillation. Heart Rhythm 2022; 19:1450-1458. [PMID: 35660473 DOI: 10.1016/j.hrthm.2022.05.033] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 05/01/2022] [Accepted: 05/25/2022] [Indexed: 01/22/2023]
Abstract
BACKGROUND Exosomal long non-coding RNAs (lncRNAs) are known as ideal diagnostic biomarkers of various diseases. However, there are no reports on the use of serum exosomal lncRNAs as diagnostic biomarkers for atrial fibrillation (AF). OBJECTIVE The purpose of this study was to explore serum exosomal lncRNAs as a useful tool for diagnosing AF. METHODS First, serum exosomes from patients with persistent AF and controls were isolated using a polymer-based exosome precipitation kit. Next, we conducted a multi-phase process including screening and two independent validation phases. In the screening phase, serum exosomal lncRNA expression profiles were examined using RNA-sequencing analysis. In two validation phases, we evaluated the expression levels of candidate exosomal lncRNAs using qRT-PCR. Finally, we performed different statistical and functional analyses. RESULTS After the screening phase, we identified 26 differentially expressed lncRNAs (i.e., 15 up-regulated and 11 down-regulated lncRNAs with a |fold change| ≥ 2 and p < 0.05) in serum exosomes from patients with persistent AF compared with the controls. We then screened out six exosomal lncRNAs as biomarker candidates following parameters: read length ≥ 200 nucleotides; exon number ≥ 2; and coding potential score < 0.1. In two validation phases, exosomal lncRNAs LOC105377989 and LOC107986997 were consistently up-regulated in serum of patients with persistent AF, compared with the controls (p < 0.0001). Moreover, both exosomal lncRNAs exhibited significant diagnostic validity for AF. Notably, exosomal lncRNA LOC107986997 was involved in AF-related pathophysiological mechanisms. CONCLUSION Serum-derived exosomal lncRNA LOC107986997 could serve as a potential biomarker for AF diagnosis.
Collapse
Affiliation(s)
- Ji-Young Kang
- Division of Cardiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Dasom Mun
- Division of Cardiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Hyoeun Kim
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Nuri Yun
- Institute of Life Science & Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Boyoung Joung
- Division of Cardiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
| |
Collapse
|
10
|
Li MP, Hao ZC, Yan MQ, Xia CL, Wang ZH, Feng YQ. Possible causes of atherosclerosis: lncRNA COLCA1 induces oxidative stress in human coronary artery endothelial cells and impairs wound healing. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:286. [PMID: 35434044 PMCID: PMC9011302 DOI: 10.21037/atm-22-507] [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: 01/07/2022] [Accepted: 03/04/2022] [Indexed: 11/06/2022]
Abstract
Background Atherosclerosis is the most common cause of cardiovascular disease, accompanied by high mortality and poor prognosis. Low-density lipoprotein (LDL) and its oxidized form oxidized low-density lipoprotein (oxLDL) play an important role in atherosclerosis. This article will explore the role of the lncRNA COLCA1 (colorectal cancer associated 1)/hsa-miR-371a-5p/SPP1 (secreted phosphoprotein 1) pathway in oxLDL in causing human coronary artery endothelial cells (HCAECs) inflammation and related biological function changes. Methods OxLDL was used to stimulate HCAECs. The inflammatory response and biological function changes of HCAECs were analyzed, total RNA-seq was performed on HCAECs before and after stimulation, and RT-Qpcr (real-time quantitative PCR) was used to verify the differential genes. Interference of the expression of COLCA1 in HCAECs was performed by siRNA interference technology to verify the role of COLCA1 in the biological function changes of HCAECs after oxLDL stimulation, and further prove that COLCA1 affects SPP1 through hsa-miR-371a-5p. Results OxLDL can affect the oxidative stress response of HCAECs, which in turn affects the apoptosis and wound healing ability of HCAECs. COLCA1 and SPP1 were highly expressed after oxLDL stimulation, while hsa-miR-371a-5p was the opposite. After COLCA1 interference, the oxidative stress level of HCAECs stimulated by oxLDL decreased, the apoptosis level also significantly decreased, and the wound healing ability was enhanced. After simultaneous COLCA1 interference and recovery of the expression of hsa-miR-371a-5p, these improved functions disappeared. The dual-luciferase assay confirmed that hsa-miR-371a-5p and COLCA1, hsa-miR-371a-5p and SPP1 has binding targets. Conclusions OxLDL can up-regulate the expression of COLCA1 in HCAECs, which in turn affects the intracellular COLCA1/hsa-miR-371a-5p/SPP1 pathway to regulate the level of oxidative stress in cells. This in turn affects the level of apoptosis and wound healing ability, which causes cells to produce a continuous inflammatory response.
Collapse
Affiliation(s)
- Ming-Peng Li
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Department of Cardiovascular, Chenzhou No. 1 People's Hospital, The First Affiliated Hospital of Xiangnan University, Chenzhou, China
| | - Zi-Chen Hao
- Department of Cardiovascular, Chenzhou No. 1 People's Hospital, The First Affiliated Hospital of Xiangnan University, Chenzhou, China
| | - Meng-Qi Yan
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Chun-Li Xia
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhong-Hua Wang
- Department of Cardiovascular, Chenzhou No. 1 People's Hospital, The First Affiliated Hospital of Xiangnan University, Chenzhou, China
| | - Ying-Qing Feng
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
11
|
Zhao B, Wang W, Liu Y, Guan S, Wang M, Song F, Shangguan W, Miao S, Zhang X, Liu H, Liu E, Liang X. Establishment of a lncRNA-miRNA-mRNA network in a rat model of atrial fibrosis by whole transcriptome sequencing. J Interv Card Electrophysiol 2022; 63:723-736. [DOI: 10.1007/s10840-022-01120-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 01/05/2022] [Indexed: 10/19/2022]
|
12
|
Li D, Nie J, Han Y, Ni L. Epigenetic Mechanism and Therapeutic Implications of Atrial Fibrillation. Front Cardiovasc Med 2022; 8:763824. [PMID: 35127848 PMCID: PMC8815458 DOI: 10.3389/fcvm.2021.763824] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 12/08/2021] [Indexed: 12/28/2022] Open
Abstract
Atrial fibrillation (AF) is the most common arrhythmia attacking 1. 5–2.0% of general population worldwide. It has a significant impact on morbidity and mortality globally and its prevalence increases exponentially with age. Therapies like catheter ablation or conventional antiarrhythmic drugs have not provided effective solution to the recurrence for AF over the past decades. Over 100 genetic loci have been discovered to be associated with AF by Genome-wide association studies (GWAS) but none has led to a therapy. Recently potential involvement of epigenetics (DNA methylation, histone modification, and non-coding RNAs) in the initiation and maintenance of AF has partly emerged as proof-of-concept in the mechanism and management of AF. Here we reviewed the epigenetic features involved in AF pathophysiology and provided an update of their implications in AF therapy.
Collapse
|
13
|
Zhang L, Wang X, Huang C. A narrative review of non-coding RNAs in atrial fibrillation: potential therapeutic targets and molecular mechanisms. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1486. [PMID: 34734038 PMCID: PMC8506732 DOI: 10.21037/atm-21-4483] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/16/2021] [Indexed: 11/11/2022]
Abstract
Objective This review summarizes the advances in the study of ncRNAs and atrial remodeling mechanisms to explore potential therapeutic targets and strategies for AF. Background Atrial fibrillation (AF) is one of the most common arrhythmias, and its morbidity and mortality rates are gradually increasing. Non-coding ribonucleic acid RNAs (ncRNAs) are transcribed from the genome and do not have the ability to be translated into proteins. A growing body of evidence has shown ncRNAs are extensively involved in the pathophysiological processes underlying AF. However, the precise molecular mechanisms of these associations have not been fully elucidated. Atrial remodeling plays a key role in the occurrence and development of AF, and includes electrical remodeling, structural remodeling, and autonomic nerve remodeling. Research has shown that ncRNA expression is altered in the plasma and tissues of AF patients that mediate cardiac excitation and arrhythmia, and is closely related to atrial remodeling. Methods Literatures about ncRNAs and atrial fibrillation were extensively reviewed to discuss and analyze. Conclusions The biology of ncRNAs represents a relatively new field of research and is still in an emerging stage. Recent studies have laid a foundation for understanding the molecular mechanisms of AF, future studies aimed at identifying how ncRNAs act on atrial fibrillation to provide potentially promising therapeutic targets for the treatment of atrial fibrillation.
Collapse
Affiliation(s)
- Lan Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
14
|
Yan B, Liu T, Yao C, Liu X, Du Q, Pan L. LncRNA XIST shuttled by adipose tissue-derived mesenchymal stem cell-derived extracellular vesicles suppresses myocardial pyroptosis in atrial fibrillation by disrupting miR-214-3p-mediated Arl2 inhibition. J Transl Med 2021; 101:1427-1438. [PMID: 34389797 DOI: 10.1038/s41374-021-00635-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 06/17/2021] [Accepted: 06/18/2021] [Indexed: 01/17/2023] Open
Abstract
The mechanisms underlying atrial fibrillation (AF), a type of heart arrhythmia, have not been fully identified. Long noncoding RNAs (lncRNAs) have been implicated in the progression of AF. The current study aimed to ascertain the means by which X-inactive specific transcript (XIST), a lncRNA, contributes to the pathogenesis of AF in an animal model or in atrial myocytes. Extracellular vesicles (EVs) derived from mouse adipose tissue-derived mesenchymal stem cells (AMSCs) were isolated, transfected with XIST, and either injected into AF mouse models or incubated with atrial myocytes. The in vitro and in vivo effects of EV-derived XIST on myocardial pyroptosis were determined by Western blot analysis of pyroptosis-related protein and an ELISA for inflammatory factors. Bioinformatics analysis revealed a relationship between XIST, microRNA (miR)-214-3p, and Arl2, which was subsequently verified by a dual luciferase assay and RNA immunoprecipitation. Functional experiments were performed to elucidate whether changes in miR-214-3p or Arl2 regulated the effect of XIST on myocardial pyroptosis. Overexpressed XIST from AMSC-EVs were found to decrease myocardial pyroptosis while alleviating inflammation, which was demonstrated by reduced expression of nucleotide-binding and oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3), apoptosis-associated speck-like protein containing a CARD (ASC), cleared-caspase-1/caspase-1 and gasdermin D (GSDMD), as well as the amount of interleukin (IL)-1β and IL-18 in both the cardiomyocytes and AF mouse tissues. Mechanistically, XIST is a competing endogenous RNA (ceRNA) of miR-214-3p, triggering upregulation of its target gene Arl2. Silencing of Arl2 or overexpression miR-214-3p reversed the effects of XIST on inflammation and pyroptosis. Taken together, the key findings of our study suggest that XIST may blunt myocardial pyroptosis by absorbing miR-214-3p to promote Arl2 expression, providing encouraging insight into XIST-based targeted therapy for AF.
Collapse
Affiliation(s)
- Boyu Yan
- Department of Cardiology, Pingxiang People's Hospital, Pingxiang, People's Republic of China
| | - Ting Liu
- Department of Pharmacy, Pingxiang People's Hospital, Pingxiang, People's Republic of China
| | - Chang Yao
- Department of Cardiology, Pingxiang People's Hospital, Pingxiang, People's Republic of China
| | - Xinglong Liu
- Department of Cardiology, Pingxiang People's Hospital, Pingxiang, People's Republic of China
| | - Qian Du
- Department of Cardiology, Pingxiang People's Hospital, Pingxiang, People's Republic of China
| | - Lihua Pan
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, People's Republic of China.
| |
Collapse
|
15
|
Rey F, Messa L, Pandini C, Barzaghini B, Micheletto G, Raimondi MT, Bertoli S, Cereda C, Zuccotti GV, Cancello R, Carelli S. Transcriptional characterization of subcutaneous adipose tissue in obesity affected women highlights metabolic dysfunction and implications for lncRNAs. Genomics 2021; 113:3919-3934. [PMID: 34555498 DOI: 10.1016/j.ygeno.2021.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 09/03/2021] [Accepted: 09/17/2021] [Indexed: 10/20/2022]
Abstract
Obesity is a complex disease with multifactorial causes, and its prevalence is becoming a serious health crisis. For this reason, there is a crucial need to identify novel targets and players. With this aim in mind, we analyzed via RNA-sequencing the subcutaneous adipose tissue of normal weight and obesity-affected women, highlighting the differential expression in the two tissues. We specifically focused on long non-coding RNAs, as 6 of these emerged as dysregulated in the diseased-tissue (COL4A2-AS2, RPS21-AS, PELATON, ITGB2-AS1, ACER2-AS and CTEPHA1). For each of them, we performed both a thorough in silico dissection and in vitro validation, to predict their function during adipogenesis. We report the lncRNAs expression during adipose derived stem cells differentiation to adipocytes as model of adipogenesis and their potential modulation by adipogenesis-related transcription factors (C/EBPs and PPARγ). Moreover, inhibiting CTEPHA1 expression we investigated its impact on adipogenesis-related transcription factors, showing its significative dysregulation of C/EBPα expression. Lastly, we dissected the subcellular localization, pathway involvement and disease-correlation for coding differentially expressed genes. Together, these findings highlight a transcriptional deregulation at the basis of obesity, impacted by both coding and long non-coding RNAs.
Collapse
Affiliation(s)
- Federica Rey
- Department of Biomedical and Clinical Sciences "L. Sacco", University of Milan, Via Grassi 74, 20157 Milan, Italy; Pediatric Clinical Research Centre Fondazione "Romeo ed Enrica Invernizzi", University of Milano, Milano, Italy
| | - Letizia Messa
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milano, Italy
| | - Cecilia Pandini
- Genomic and post-Genomic Centre, IRCCS Mondino Foundation, 27100 Pavia, Italy
| | - Bianca Barzaghini
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milano, Italy
| | - Giancarlo Micheletto
- Department of Pathophysiology and Transplantation, INCO, Department of General Surgery, Istituto Clinico Sant'Ambrogio, University of Milan, Milan, Italy
| | - Manuela Teresa Raimondi
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milano, Italy
| | - Simona Bertoli
- Obesity Unit, Laboratory of Nutrition and Obesity Research, Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy; International Center for the Assessment of Nutritional Status (ICANS), Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
| | - Cristina Cereda
- Genomic and post-Genomic Centre, IRCCS Mondino Foundation, 27100 Pavia, Italy
| | - Gian Vincenzo Zuccotti
- Department of Biomedical and Clinical Sciences "L. Sacco", University of Milan, Via Grassi 74, 20157 Milan, Italy; Pediatric Clinical Research Centre Fondazione "Romeo ed Enrica Invernizzi", University of Milano, Milano, Italy; Department of Pediatrics, Children's Hospital "V. Buzzi", Milan, Italy
| | - Raffaella Cancello
- Obesity Unit, Laboratory of Nutrition and Obesity Research, Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Stephana Carelli
- Department of Biomedical and Clinical Sciences "L. Sacco", University of Milan, Via Grassi 74, 20157 Milan, Italy; Pediatric Clinical Research Centre Fondazione "Romeo ed Enrica Invernizzi", University of Milano, Milano, Italy.
| |
Collapse
|
16
|
Gao J, Chen X, Wei P, Wang Y, Li P, Shao K. Regulation of pyroptosis in cardiovascular pathologies: Role of noncoding RNAs. MOLECULAR THERAPY-NUCLEIC ACIDS 2021; 25:220-236. [PMID: 34458007 PMCID: PMC8368762 DOI: 10.1016/j.omtn.2021.05.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Cardiovascular disease (CVD) is one of the most important diseases endangering human life. The pathogenesis of CVDs is complex. Pyroptosis, which differs from traditional apoptosis and necrosis, is characterized by cell swelling until membrane rupture, resulting in the release of cell contents and activation of a strong inflammatory response. Recent studies have revealed that inflammation and pyroptosis play important roles in the progression of CVDs. Noncoding RNAs (ncRNAs) are considered promising biomarkers and potential therapeutic targets for the diagnosis and treatment of various diseases, including CVDs. Growing evidence has revealed that ncRNAs can mediate the transcriptional or posttranscriptional regulation of pyroptosis-related genes by participating in the pyroptosis regulatory network. The role and molecular mechanism of pyroptosis-regulating ncRNAs in cardiovascular pathologies are attracting increasing attention. Here, we summarize research progress on pyroptosis and the role of ncRNAs, particularly microRNAs (miRNAs), long ncRNAs (lncRNAs), and circular RNAs (circRNAs), in the regulation of pyroptosis in CVD pathologies. Identifying these disease-related ncRNAs is important for understanding the pathogenesis of CVDs and providing new targets and ideas for their prevention and treatment.
Collapse
Affiliation(s)
- Jinning Gao
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Xiatian Chen
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Pengcheng Wei
- College of Medicine, Qingdao University, Qingdao 266073, China
| | - Yin Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Kai Shao
- Department of Central Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, Shandong 266035, China
| |
Collapse
|
17
|
Huang S, Deng Y, Xu J, Liu J, Liu L, Fan C. The Role of Exosomes and Their Cargos in the Mechanism, Diagnosis, and Treatment of Atrial Fibrillation. Front Cardiovasc Med 2021; 8:712828. [PMID: 34395566 PMCID: PMC8355361 DOI: 10.3389/fcvm.2021.712828] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/07/2021] [Indexed: 12/28/2022] Open
Abstract
Atrial fibrillation (AF) is the most common persistent arrhythmia, but the mechanism of AF has not been fully elucidated, and existing approaches to diagnosis and treatment face limitations. Recently, exosomes have attracted considerable interest in AF research due to their high stability, specificity and cell-targeting ability. The aim of this review is to summarize recent literature, analyze the advantages and limitations of exosomes, and to provide new ideas for their use in understanding the mechanism and improving the diagnosis and treatment of AF.
Collapse
Affiliation(s)
- Shengyuan Huang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yating Deng
- Xiangya Medical College of Central South University, Changsha, China
| | - Jiaqi Xu
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Jiachen Liu
- Xiangya Medical College of Central South University, Changsha, China
| | - Liming Liu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chengming Fan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
18
|
Rey F, Urrata V, Gilardini L, Bertoli S, Calcaterra V, Zuccotti GV, Cancello R, Carelli S. Role of long non-coding RNAs in adipogenesis: State of the art and implications in obesity and obesity-associated diseases. Obes Rev 2021; 22:e13203. [PMID: 33443301 PMCID: PMC8244036 DOI: 10.1111/obr.13203] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/11/2020] [Accepted: 12/13/2020] [Indexed: 12/14/2022]
Abstract
Obesity is an evolutionary, chronic, and relapsing disease that consists of a pathological accumulation of adipose tissue able to increase morbidity for high blood pressure, type 2 diabetes, metabolic syndrome, and obstructive sleep apnea in adults, children, and adolescents. Despite intense research over the last 20 years, obesity remains today a disease with a complex and multifactorial etiology. Recently, long non-coding RNAs (lncRNAs) are emerging as interesting new regulators as different lncRNAs have been found to play a role in early and late phases of adipogenesis and to be implicated in obesity-associated complications onset. In this review, we discuss the most recent advances on the role of lncRNAs in adipocyte biology and in obesity-associated complications. Indeed, more and more researchers are focusing on investigating the underlying roles that these molecular modulators could play. Even if a significant number of evidence is correlation-based, with lncRNAs being differentially expressed in a specific disease, recent works are now focused on deeply analyzing how lncRNAs can effectively modulate the disease pathogenesis onset and progression. LncRNAs possibly represent new molecular markers useful in the future for both the early diagnosis and a prompt clinical management of patients with obesity.
Collapse
Affiliation(s)
- Federica Rey
- Department of Biomedical and Clinical Sciences "L. Sacco", University of Milan, Milan, Italy.,Pediatric Clinical Research Center Fondazione "Romeo ed Enrica Invernizzi", University of Milan, Milan, Italy
| | - Valentina Urrata
- Department of Biomedical and Clinical Sciences "L. Sacco", University of Milan, Milan, Italy.,Pediatric Clinical Research Center Fondazione "Romeo ed Enrica Invernizzi", University of Milan, Milan, Italy
| | - Luisa Gilardini
- Obesity Unit-Laboratory of Nutrition and Obesity Research, Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Simona Bertoli
- Obesity Unit-Laboratory of Nutrition and Obesity Research, Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy.,International Center for the Assessment of Nutritional Status (ICANS), Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
| | - Valeria Calcaterra
- Pediatrics and Adolescentology Unit, Department of Internal Medicine, University of Pavia, Pavia, Italy.,Department of Pediatrics, Children's Hospital "V. Buzzi", Milan, Italy
| | - Gian Vincenzo Zuccotti
- Department of Biomedical and Clinical Sciences "L. Sacco", University of Milan, Milan, Italy.,Pediatric Clinical Research Center Fondazione "Romeo ed Enrica Invernizzi", University of Milan, Milan, Italy.,Department of Pediatrics, Children's Hospital "V. Buzzi", Milan, Italy
| | - Raffaella Cancello
- Obesity Unit-Laboratory of Nutrition and Obesity Research, Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Stephana Carelli
- Department of Biomedical and Clinical Sciences "L. Sacco", University of Milan, Milan, Italy.,Pediatric Clinical Research Center Fondazione "Romeo ed Enrica Invernizzi", University of Milan, Milan, Italy
| |
Collapse
|
19
|
Identification of Pivotal MicroRNAs and Target Genes Associated with Persistent Atrial Fibrillation Based on Bioinformatics Analysis. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:6680211. [PMID: 33747117 PMCID: PMC7960048 DOI: 10.1155/2021/6680211] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/06/2021] [Accepted: 02/22/2021] [Indexed: 12/13/2022]
Abstract
Atrial fibrillation (AF) is one of the most common supraventricular arrhythmias worldwide. However, the specific molecular mechanism underlying AF remains unclear. Our study is aimed at identifying pivotal microRNAs (miRNAs) and targeting genes associated with persistent AF (pAF) using bioinformatics analysis. Three gene expression array datasets (GSE31821, GSE41177, and GSE79768) and an miRNA expression array dataset (GSE68475) associated with pAF were downloaded. Differentially expressed genes (DEGs) were identified using the LIMMA package, and differentially expressed miRNAs (DEMs) were screened from GSE68475. Target genes for DEMs were predicted using the miRTarBase database, and intersections between these target genes and DEGs were selected for further analysis, including the generation of protein–protein interaction (PPI) network, miRNA–transcription factor–target regulatory network, and drug–gene network. A total of 264 DEGs and 40 DEMs were identified between the pAF and control groups. Functional and pathway enrichment analyses of up- and downregulated DEGs were performed. The common genes (CGs) were primarily enriched in the phosphoinositide 3-kinase- (PI3K-) protein kinase B (Akt) signaling pathway, negative regulation of cell division, and response to hypoxia. The PPI network, miRNA–transcription factor–target regulatory network, and drug–gene network were constructed using Cytoscape. The present study revealed several novel miRNAs and genes involved in pAF. We speculated that miR-4298, miR-3125, miR-4306, and miR-671-5p could represent significant miRNAs that act on the target gene superoxide dismutase 2 (SOD2) during the development of pAF and may serve as essential biomarkers for pAF diagnosis and treatment. Moreover, MYC might function in pAF pathogenesis through the PI3K–Akt signaling pathway.
Collapse
|
20
|
Noubiap JJ, Sanders P, Nattel S, Lau DH. Biomarkers in Atrial Fibrillation: Pathogenesis and Clinical Implications. Card Electrophysiol Clin 2021; 13:221-233. [PMID: 33516400 DOI: 10.1016/j.ccep.2020.10.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Biomarkers derived from the key components of the pathophysiology of atrial fibrillation (AF) and its complications have the potential to play an important role in earlier characterization of AF phenotype and in risk prediction of adverse clinical events, which may translate into improved management strategies. C-reactive protein, natriuretic peptides, cardiac troponins, growth differentiation factor-15, and fibroblast growth factor-23 have been shown to be the most promising biomarkers in AF. Some biomarkers have already been included in clinical risk scores to predict postoperative AF, thromboembolism, major bleeding, and death. Considerably more work is needed to bring these novel biomarkers into routine clinical management of patients with AF.
Collapse
Affiliation(s)
| | - Prashanthan Sanders
- Centre for Heart Rhythm Disorders, University of Adelaide, Adelaide, Australia; Department of Cardiology, Royal Adelaide Hospital, Adelaide, Australia
| | - Stanley Nattel
- Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Canada
| | - Dennis H Lau
- Centre for Heart Rhythm Disorders, University of Adelaide, Adelaide, Australia; Department of Cardiology, Royal Adelaide Hospital, Adelaide, Australia.
| |
Collapse
|
21
|
Babapoor-Farrokhran S, Gill D, Alzubi J, Mainigi SK. Atrial fibrillation: the role of hypoxia-inducible factor-1-regulated cytokines. Mol Cell Biochem 2021; 476:2283-2293. [PMID: 33575876 DOI: 10.1007/s11010-021-04082-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 01/25/2021] [Indexed: 11/25/2022]
Abstract
Atrial fibrillation (AF) is a common arrhythmia that has major morbidity and mortality. Hypoxia plays an important role in AF initiation and maintenance. Hypoxia-inducible factor (HIF), the master regulator of oxygen homeostasis in cells, plays a fundamental role in the regulation of multiple chemokines and cytokines that are involved in different physiological and pathophysiological pathways. HIF is also involved in the pathophysiology of AF induction and propagation mostly through structural remodeling such as fibrosis; however, some of the cytokines discussed have even been implicated in electrical remodeling of the atria. In this article, we highlight the association between HIF and some of its related cytokines with AF. Additionally, we provide an overview of the potential diagnostic benefits of using the mentioned cytokines as AF biomarkers. Research discussed in this review suggests that the expression of these cytokines may correlate with patients who are at an increased risk of developing AF. Furthermore, cytokines that are elevated in patients with AF can assist clinicians in the diagnosis of suspect paroxysmal AF patients. Interestingly, some of the cytokines have been elevated specifically when AF is associated with a hypercoagulable state, suggesting that they could be helpful in the clinician's and patient's decision to begin anticoagulation. Finally, more recent research has demonstrated the promise of targeting these cytokines for the treatment of AF. While still in its early stages, tools such as neutralizing antibodies have proved to be efficacious in targeting the HIF pathway and treating or preventing AF.
Collapse
Affiliation(s)
- Savalan Babapoor-Farrokhran
- Division of Cardiology, Department of Medicine, Einstein Medical Center, 5501 Old York Road, Philadelphia, PA, 19141, USA.
| | - Deanna Gill
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Jafar Alzubi
- Division of Cardiology, Department of Medicine, Einstein Medical Center, 5501 Old York Road, Philadelphia, PA, 19141, USA
| | - Sumeet K Mainigi
- Division of Cardiology, Department of Medicine, Einstein Medical Center, 5501 Old York Road, Philadelphia, PA, 19141, USA
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| |
Collapse
|
22
|
Epigenetics in atrial fibrillation: A reappraisal. Heart Rhythm 2021; 18:824-832. [PMID: 33440248 DOI: 10.1016/j.hrthm.2021.01.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 12/23/2020] [Accepted: 01/01/2021] [Indexed: 11/21/2022]
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia and an important cause of morbidity and mortality globally. Atrial remodeling includes changes in ion channel expression and function, structural alterations, and neural remodeling, which create an arrhythmogenic milieu resulting in AF initiation and maintenance. Current therapeutic strategies for AF involving ablation and antiarrhythmic drugs are associated with relatively high recurrence and proarrhythmic side effects, respectively. Over the last 2 decades, in an effort to overcome these issues, research has sought to identify the genetic basis for AF thereby gaining insight into the regulatory mechanisms governing its pathophysiology. Despite identification of multiple gene loci associated with AF, thus far none has led to a therapy, indicating additional contributors to pathology. Recently, in the context of expanding knowledge of the epigenome (DNA methylation, histone modifications, and noncoding RNAs), its potential involvement in the onset and progression of AF pathophysiology has started to emerge. Probing the role of various epigenetic mechanisms that contribute to AF may improve our knowledge of this complex disease, identify potential therapeutic targets, and facilitate targeted therapies. Here, we provide a comprehensive review of growing epigenetic features involved in AF pathogenesis and summarize the emerging epigenomic targets for therapy that have been explored in preclinical models of AF.
Collapse
|
23
|
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
|
24
|
Long Noncoding RNA HOTAIR Functions as a Competitive Endogenous RNA to Regulate Connexin43 Remodeling in Atrial Fibrillation by Sponging MicroRNA-613. Cardiovasc Ther 2020; 2020:5925342. [PMID: 33294032 PMCID: PMC7688347 DOI: 10.1155/2020/5925342] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/22/2020] [Accepted: 10/24/2020] [Indexed: 12/16/2022] Open
Abstract
Several studies have indicated that long noncoding RNAs (lncRNAs)-HOX transcript antisense RNA (HOTAIR) is involved in some cardiovascular diseases by regulating gene expression as a competitive endogenous RNA (ceRNA). GJA1 encoding Cx43 is one potential target gene of microRNA-613 (miR-613). Meanwhile, there is a potential target regulatory relationship between HOTAIR and miR-613. The present study is aimed at investigating whether HOTAIR functions as a ceRNA to regulate the Cx43 expression in atrial fibrillation (AF) by sponging miR-613. The expressions of HOTAIR, miR-613, and Cx43 were detected in the right atrial appendages of 45 patients with heart valve disease, including 23 patients with chronic AF. The HOTAIR overexpressed and underexpressed HL-1 cell model were constructed to confirm the effect of HOTAIR on Cx43. Then, the Cx43 expression was detected to testify the interplay between HOTAIR and miR-613 after cotransfecting HOTAIR and miR-613. Furthermore, luciferase assays were performed to verify that HOTAIR could regulate Cx43 remolding as a ceRNA by sponging miR-613. The expression of HOTAIR and Cx43 was significantly downregulated in chronic AF group. HOTAIR regulated positively the Cx43 expression in HL-1 cells. The upregulated effect of HOTAIR on the Cx43 expression could be remarkably attenuated by miR-613. Moreover, the inhibitory effect of miR-613 on the Cx43 expression could be obviously mitigated by HOTAIR. At last, luciferase assays confirmed HOTAIR functioned as a ceRNA in the Cx43 expression by sponging miR-613. Our study suggests that HOTAIR, functioning as a ceRNA by sponging miR-613, is an important contributor to Cx43 remolding in AF.
Collapse
|
25
|
Bektik E, Cowan DB, Wang DZ. Long Non-Coding RNAs in Atrial Fibrillation: Pluripotent Stem Cell-Derived Cardiomyocytes as a Model System. Int J Mol Sci 2020; 21:ijms21155424. [PMID: 32751460 PMCID: PMC7432754 DOI: 10.3390/ijms21155424] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 12/19/2022] Open
Abstract
Atrial fibrillation (AF) is a type of sustained arrhythmia in humans often characterized by devastating alterations to the cardiac conduction system as well as the structure of the atria. AF can lead to decreased cardiac function, heart failure, and other complications. Long non-coding RNAs (lncRNAs) have been shown to play important roles in the cardiovascular system, including AF; however, a large group of lncRNAs is not conserved between mouse and human. Furthermore, AF has complex networks showing variations in mechanisms in different species, making it challenging to utilize conventional animal models to investigate the functional roles and potential therapeutic benefits of lncRNAs for AF. Fortunately, pluripotent stem cell (PSC)-derived cardiomyocytes (CMs) offer a reliable platform to study lncRNA functions in AF because of certain electrophysiological and molecular similarities with native human CMs. In this review, we first summarize the broad aspects of lncRNAs in various heart disease settings, then focus on their potential roles in AF development and pathophysiology. We also discuss current uses of PSCs in AF research and describe how these studies could be developed into novel therapeutics for AF and other cardiovascular diseases.
Collapse
Affiliation(s)
- Emre Bektik
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood, Boston, MA 02115, USA; (E.B.); (D.B.C.)
| | - Douglas B. Cowan
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood, Boston, MA 02115, USA; (E.B.); (D.B.C.)
| | - Da-Zhi Wang
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood, Boston, MA 02115, USA; (E.B.); (D.B.C.)
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
- Correspondence:
| |
Collapse
|