1
|
Bolha L, Hočevar A, Jurčić V. Current state of epigenetics in giant cell arteritis: Focus on microRNA dysregulation. Autoimmun Rev 2025; 24:103739. [PMID: 39732382 DOI: 10.1016/j.autrev.2024.103739] [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: 09/18/2024] [Revised: 12/23/2024] [Accepted: 12/25/2024] [Indexed: 12/30/2024]
Abstract
Giant cell arteritis (GCA) is a primary systemic vasculitis affecting the elderly, characterized by a granulomatous vessel wall inflammation of large- and medium-sized arteries. The immunopathology of GCA is complex, involving both the innate and adaptive arms of the immune system, where a maladaptive inflammatory-driven vascular repair process ultimately results in vessel wall thickening, intramural vascular smooth muscle cell proliferation, neovascularization and vessel lumen occlusion, which can lead to serious ischemic complications such as visual loss and ischemic stroke. Over the past decade, microRNA (miRNA) dysregulation has been highlighted as an important contributing factor underlying the pathogenesis of GCA. Since current understanding of miRNA involvement in GCA remains largely based on extrapolation of previously determined miRNA functions in vitro or in loss- or gain-of-function studies, an overall insight into the role of miRNA alteration in GCA pathophysiology remains limited. In this narrative review, we summarize the current knowledge on aberrantly expressed miRNAs in GCA and thoroughly discuss the impact of their altered regulatory role in the context of GCA setting. Furthermore, we address challenges and future perspectives in utilization of miRNA-based diagnostic and prognostic biomarkers of GCA in clinical settings.
Collapse
Affiliation(s)
- Luka Bolha
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.
| | - Alojzija Hočevar
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia; Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Vesna Jurčić
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.
| |
Collapse
|
2
|
Li J, Yao H, Zhao F, An J, Wang Q, Mu J, Liu Z, Zou MH, Xie Z. Pycard deficiency inhibits microRNA maturation and prevents neointima formation by promoting chaperone-mediated autophagic degradation of AGO2/argonaute 2 in adipose tissue. Autophagy 2024; 20:629-644. [PMID: 37963060 PMCID: PMC10936599 DOI: 10.1080/15548627.2023.2277610] [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: 08/29/2022] [Revised: 10/10/2023] [Accepted: 10/26/2023] [Indexed: 11/16/2023] Open
Abstract
PYCARD (PYD and CARD domain containing), a pivotal adaptor protein in inflammasome assembly and activation, contributes to innate immunity, and plays an essential role in the pathogenesis of atherosclerosis and restenosis. However, its roles in microRNA biogenesis remain unknown. Therefore, this study aimed to investigate the roles of PYCARD in miRNA biogenesis and neointima formation using pycard knockout (pycard-/-) mice. Deficiency of Pycard reduced circulating miRNA profile and inhibited Mir17 seed family maturation. The systemic pycard knockout also selectively reduced the expression of AGO2 (argonaute RISC catalytic subunit 2), an important enzyme in regulating miRNA biogenesis, by promoting chaperone-mediated autophagy (CMA)-mediated degradation of AGO2, specifically in adipose tissue. Mechanistically, pycard knockout increased PRMT8 (protein arginine N-methyltransferase 8) expression in adipose tissue, which enhanced AGO2 methylation, and subsequently promoted its binding to HSPA8 (heat shock protein family A (Hsp70) member 8) that targeted AGO2 for lysosome degradation through chaperone-mediated autophagy. Finally, the reduction of AGO2 and Mir17 family expression prevented vascular injury-induced neointima formation in Pycard-deficient conditions. Overexpression of AGO2 or administration of mimic of Mir106b (a major member of the Mir17 family) prevented Pycard deficiency-mediated inhibition of neointima formation in response to vascular injury. These data demonstrate that PYCARD inhibits CMA-mediated degradation of AGO2, which promotes microRNA maturation, thereby playing a critical role in regulating neointima formation in response to vascular injury independently of inflammasome activity and suggest that modulating PYCARD expression and function may represent a powerful therapeutic strategy for neointima formation.Abbreviations: 6-AN: 6-aminonicotinamide; ACTB: actin, beta; aDMA: asymmetric dimethylarginine; AGO2: argonaute RISC catalytic subunit 2; CAL: carotid artery ligation; CALCOCO2: calcium binding and coiled-coil domain 2; CMA: chaperone-mediated autophagy; CTSB: cathepsin B; CTSD: cathepsin D; DGCR8: DGCR8 microprocessor complex subunit; DOCK2: dedicator of cyto-kinesis 2; EpiAdi: epididymal adipose tissue; HSPA8: heat shock protein family A (Hsp70) member 8; IHC: immunohistochemical; ISR: in-stent restenosis; KO: knockout; LAMP2: lysosomal-associated membrane protein 2; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; miRNA: microRNA; NLRP3: NLR family pyrin domain containing 3; N/L: ammonium chloride combined with leupeptin; PRMT: protein arginine methyltransferase; PVAT: peri-vascular adipose tissues; PYCARD: PYD and CARD domain containing; sDMA: symmetric dimethylarginine; ULK1: unc-51 like kinase 1; VSMCs: vascular smooth muscle cells; WT: wild-type.
Collapse
Affiliation(s)
- Jian Li
- Center of Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia
| | - Hongmin Yao
- Center of Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia
| | - Fujie Zhao
- Center of Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia
| | - Junqing An
- Center of Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia
| | - Qilong Wang
- Center of Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia
| | - Jing Mu
- Center of Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia
| | - Zhixue Liu
- Center of Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia
| | - Ming-Hui Zou
- Center of Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia
| | - Zhonglin Xie
- Center of Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia
| |
Collapse
|
3
|
ZENUNAJ G. The role of circulating miRNAs as predictive biomarkers for the neointimal hyperplasia after femoro-popliteal endovascular procedures for symptomatic peripheral arterial disease. ITALIAN JOURNAL OF VASCULAR AND ENDOVASCULAR SURGERY 2024; 30. [DOI: 10.23736/s1824-4777.23.01634-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2025]
|
4
|
Wu KL, Chen CL, Thi Nguyen MH, Tsai JC, Wang SC, Chiang WF, Hsiao PJ, Chan JS, Hou JJ, Ma N. MicroRNA regulators of vascular pathophysiology in chronic kidney disease. Clin Chim Acta 2023; 551:117610. [PMID: 37863246 DOI: 10.1016/j.cca.2023.117610] [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: 08/19/2023] [Revised: 10/14/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023]
Abstract
Coronary artery disease (CAD) is a severe comorbidity in chronic kidney disease (CKD) due to heavy calcification in the medial layer and inflamed plaques. Chronic inflammation, endothelial dysfunction and vascular calcification are major contributors that lead to artherosclerosis in CKD. The lack of specific symptoms and signs of CAD and decreased accuracy of noninvasive diagnostic tools result in delayed diagnosis leading to increased mortality. MicroRNAs (miRNAs) are post-transcriptional regulators present in various biofluids throughout the body. In the circulation, miRNAs have been reported to be encapsulated in extracellular vesicles and serve as stable messengers for crosstalk among cells. miRNAs are involved in pathophysiologic mechanisms including CAD and can potentially be extended from basic research to clinical translational practice.
Collapse
Affiliation(s)
- Kun-Lin Wu
- Department of Biomedical Sciences and Engineering, Institute of Systems Biology and Bioinformatics, National Central University, Taoyuan, Taiwan; Division of Nephrology, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan; Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chien-Lung Chen
- Division of Nephrology, Department of Medicine, Landseed International Hospital, Taoyuan, Taiwan
| | - Mai-Huong Thi Nguyen
- Department of Biomedical Sciences and Engineering, Institute of Systems Biology and Bioinformatics, National Central University, Taoyuan, Taiwan
| | - Jen-Chieh Tsai
- Department of Biomedical Sciences and Engineering, Institute of Systems Biology and Bioinformatics, National Central University, Taoyuan, Taiwan; Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan; Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan
| | - Sun-Chong Wang
- Department of Biomedical Sciences and Engineering, Institute of Systems Biology and Bioinformatics, National Central University, Taoyuan, Taiwan
| | - Wen-Fang Chiang
- Division of Nephrology, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan; Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Po-Jen Hsiao
- Division of Nephrology, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan; Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Jenq-Shyong Chan
- Division of Nephrology, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan; Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ju Jung Hou
- Kaohsiung Medical University Hospital, Department of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Nianhan Ma
- Department of Biomedical Sciences and Engineering, Institute of Systems Biology and Bioinformatics, National Central University, Taoyuan, Taiwan.
| |
Collapse
|
5
|
Efovi D, Xiao Q. Noncoding RNAs in Vascular Cell Biology and Restenosis. BIOLOGY 2022; 12:24. [PMID: 36671717 PMCID: PMC9855655 DOI: 10.3390/biology12010024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/17/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022]
Abstract
In-stent restenosis (ISR), characterised by ≥50% re-narrowing of the target vessel, is a common complication following stent implantation and remains a significant challenge to the long-term success of angioplasty procedures. Considering the global burden of cardiovascular diseases, improving angioplasty patient outcomes remains a key priority. Noncoding RNAs (ncRNAs) including microRNA (miRNA), long noncoding RNA (lncRNA) and circular RNA (circRNA) have been extensively implicated in vascular cell biology and ISR through multiple, both distinct and overlapping, mechanisms. Vascular smooth muscle cells, endothelial cells and macrophages constitute the main cell types involved in the multifactorial pathophysiology of ISR. The identification of critical regulators exemplified by ncRNAs in all these cell types and processes makes them an exciting therapeutic target in the field of restenosis. In this review, we will comprehensively explore the potential functions and underlying molecular mechanisms of ncRNAs in vascular cell biology in the context of restenosis, with an in-depth focus on vascular cell dysfunction during restenosis development and progression. We will also discuss the diagnostic biomarker and therapeutic target potential of ncRNAs in ISR. Finally, we will discuss the current shortcomings, challenges, and perspectives toward the clinical application of ncRNAs.
Collapse
Affiliation(s)
- Denis Efovi
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Qingzhong Xiao
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
- Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences, Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| |
Collapse
|
6
|
Wu R, Zhou S, Liu M, An H, Wang Z, Liu T. Clinical Significance of miR-21-5p in Predicting Occurrence and Progression of Uremic Vascular Calcification in Patients with End-Stage Renal Disease. Yonsei Med J 2022; 63:252-258. [PMID: 35184427 PMCID: PMC8860934 DOI: 10.3349/ymj.2022.63.3.252] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 10/29/2021] [Accepted: 11/12/2021] [Indexed: 11/27/2022] Open
Abstract
PURPOSE Vascular calcification (VC) is a common complication of end-stage renal disease (ESRD). This study aimed to examine changes in the expression of miR-21-5p in ESRD patients with VC and to explore its clinical value in predicting the occurrence and progression of uremic VC. MATERIALS AND METHODS 120 ESRD patients were divided into patients without VC group (n=38) and patients with VC group (n=82). All patients were followed up for 2 years to evaluate VC progression. qRT-PCR was used to detect serum miR-21-5p levels. Receiver operating characteristic curves were constructed to assess diagnostic value. Kaplan-Meier and log-rank methods were utilized to calculate associations between VC progression and risk factors. RESULTS Serum miR-21-5p levels were significantly higher in ESRD patients with VC than in those without VC and increased progressively with increasing disease severity. Serum miR-21-5p levels were able to distinguish patients with VC from those without VC, with an area under the curve value of 0.883, a sensitivity of 81.7%, and a specificity of 84.2%. After 2 years of follow-up, miR-21-5p expression had increased in patients with worse VC severity, compared with those with stable VC severity. Patients with high miR-21-5p levels were more likely to develop more severe VC, indicating an association between miR-21-5p and VC progression (log-rank p=0.002). Multivariable Cox regression analysis suggested that serum miR-21-5p is an independent predictive factor of VC progression in ESRD patients (hazard ratio=2.064, 95% confidence interval=1.225-3.478, p=0.006). CONCLUSION miR-21-5p is overexpressed in the serum of ESRD patients with VC. Our results suggest that overexpression of miR-21-5p is closely associated with VC progression.
Collapse
Affiliation(s)
- Rong Wu
- Department of Nephropathy, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Sen Zhou
- Department of Nephropathy, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Minglong Liu
- Department of Nephropathy, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Haiqian An
- Department of Nephropathy, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Zhe Wang
- Department of Nephropathy, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Tianxi Liu
- Department of Nephropathy, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.
| |
Collapse
|
7
|
Differential Expression of miRNA-223 in Coronary In-Stent Restenosis. J Clin Med 2022; 11:jcm11030849. [PMID: 35160300 PMCID: PMC8836934 DOI: 10.3390/jcm11030849] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/03/2022] [Accepted: 01/27/2022] [Indexed: 12/11/2022] Open
Abstract
Objective: In-stent restenosis (ISR) is an unfavorable complication that occurs in patients after coronary stenting. Despite the progress with advent of modern DES and new antiplatelet agents, restenosis still hampers PCI short- and long-term results. The aim of this study was to investigate whether circulating miRNA-223, which is associated with HDL particles and involved in cholesterol efflux pathway, have diagnostic capability for determining ISR. Methods: This case–control study comprised 21 ISR and 26 NISR patients. The level of miRNA-223 expression was evaluated by TaqMan Real-Time PCR, quantified by the comparative method (fold change) and normalized to U6 expression. Results: Patients in ISR and NISR groups were not different in terms of demographic, clinical, and biochemical parameters, except that the percentage of patients who had DES was significantly greater in the NISR group (88.9%) in comparison with the ISR group (50%). The serum expression of miRNA-223 in ISR patients was 3.277 ± 0.9 times greater than that in NISR group (p = 0.016). In addition, the results of binary logistic regression demonstrated that the high level of serum miRNA-223 was strongly and positively associated with the ISR risk (OR: 17.818, 95% CI: 1.115–284.623, p = 0.042) after adjustment for age, sex, HDL-C, LDL-C, FBS, and statin consumption. Conclusion: Elevated serum level of miRNA-223 might be helpful in predicting the occurrence of ISR. Further confirmation in future large-scale studies is warranted.
Collapse
|
8
|
Poursaleh A, Beigee FS, Esfandiari G, Najafi M. Adhesion of monocytes and endothelial cells isolated from the human aorta suppresses by miRNA-PEI particles. BMC Cardiovasc Disord 2021; 21:395. [PMID: 34399692 PMCID: PMC8369609 DOI: 10.1186/s12872-021-02203-2] [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: 06/14/2021] [Accepted: 08/07/2021] [Indexed: 12/31/2022] Open
Abstract
Background Knowledge of stenosis in coronary arteries requires an understanding of the cellular and molecular processes that occur throughout the leukocyte rolling process. In this study, the roles of miR-125a-5p and miR-495-3p were investigated on the adhesion of endothelial cells (ECs) isolated from the human aorta. Methods Human primary endothelial cells were obtained from the aorta of people who had died of brain death. Whole blood was used to isolate the monocytes. The miR-125 and miR-495 were predicted and transfected into ECs using Poly Ethylene Imine (PEI). The expression levels of adhesion molecules and monocyte recruitment were identified by the RT-qPCR technique and Leukocyte-Endothelial Adhesion Assay kit, respectively. Results The ICAM-1, ICAM-2 and VCAM-1 expression levels decreased significantly in the miR-495/PEI-transfected ECs (P < 0.05) while in the miR-125/PEI-transfected ECs only the ICAM-2 and ITGB-2 expression levels decreased significantly (P < 0.05) as compared to the miR-synthetic/PEI-transfected ECs. Furthermore, the monocyte adhesion was decreased in the miR-125 and miR-mix/PEI-transfected ECs as compared to the miR-synthetic/PEI-transfected ECs (P = 0.01 and P = 0.04, respectively). Conclusion According to the findings, the efficient relations between miR-125 and adhesion molecules may be responsible for the inhibition of monocyte rolling.
Collapse
Affiliation(s)
- Adeleh Poursaleh
- Biochemistry Department, Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Farnaz Sadegh Beigee
- Masih Daneshvari Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Golnaz Esfandiari
- Biochemistry Department, Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Najafi
- Biochemistry Department, Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Roles of MicroRNAs in Peripheral Artery In-Stent Restenosis after Endovascular Treatment. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9935671. [PMID: 34368362 PMCID: PMC8337102 DOI: 10.1155/2021/9935671] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 07/13/2021] [Indexed: 12/16/2022]
Abstract
Endovascular repair including percutaneous transluminal angioplasty (PTA) and stent implantation has become the standard approach for the treatment of peripheral arterial disease; however, restenosis is still the main limited complication for the long-term success of the endovascular repair. Endothelial denudation and regeneration, inflammatory response, and neointimal hyperplasia are major pathological processes occurring during in-stent restenosis (ISR). MicroRNAs exhibit great potential in regulating several vascular biological events in different cell types and have been identified as novel therapeutic targets as well as biomarkers for ISR prevention. This review summarized recent experimental and clinical studies on the role of miRNAs in ISR modification, with the aim of unraveling the underlying mechanism and potential therapeutic strategy of ISR.
Collapse
|
10
|
Maheronnaghsh M, Niktab I, Enayati S, Amoli MM, Hosseini SK, Tavakkoly-Bazzaz J. Differentially expressed miR-152, a potential biomarker for in-stent restenosis (ISR) in peripheral blood mononuclear cells (PBMCs) of coronary artery disease (CAD) patients. Nutr Metab Cardiovasc Dis 2021; 31:1137-1147. [PMID: 33712363 DOI: 10.1016/j.numecd.2020.09.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 09/24/2020] [Accepted: 09/25/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIMS In-stent restenosis (ISR) remains the most daunting challenge of current treatments of coronary artery disease (CAD). MicroRNAs (miRNAs) are prominent regulators of key pathological processes leading to restenosis and used as diagnostic tools in different studies. miR-152 and miR-148a are implicated to contribute in the putative intracellular mechanisms of ISR. The aim of present study is to investigate the potential early-stage diagnostic role of miR-152 and miR-148a expression levels for ISR in peripheral blood mononuclear cells (PBMCs) of patients who underwent stent implantation. METHODS AND RESULTS The miRNAs that are supposed to be involved in the ISR were nominated by bioinformatics approach mainly using miRWalk3. Then by quantitative real-time PCR, we determined the relative expression of miR-152 and miR-148a of PBMCs from ISR patients with their age/sex-matched controls. RESULTS The presence of ISR significantly coincided with a decrease in the relative expression of miR-152. The area under the curve (AUC) for miR-152 receiver operating characteristic (ROC) curve was 0.717 (95% CI; 0.60-0.83) with a sensitivity of 70% and a specificity of 67%, suggesting that the miRNA expression level might be employed to identify patients at risk of ISR. CONCLUSIONS To the best of our knowledge, this is the first work to show that the miR-152 expression level can possibly be applied to predict CAD patients at risk of ISR. The results suggest that the expression levels of miR-152 in PBMCs may serve as a biomarker for ISR. Our finding suggests the importance of miRNA levels in PBMCs as a novel biological tool to detect diseases in their early clinical stages.
Collapse
Affiliation(s)
- M Maheronnaghsh
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - I Niktab
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - S Enayati
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - M M Amoli
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - S K Hosseini
- Department of Cardiovascular Disorders, Division of Interventional Cardiology, Tehran University of Medical Sciences, Tehran, Iran.
| | - J Tavakkoly-Bazzaz
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
11
|
Yang X, Yang Y, Guo J, Meng Y, Li M, Yang P, Liu X, Aung LHH, Yu T, Li Y. Targeting the epigenome in in-stent restenosis: from mechanisms to therapy. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 23:1136-1160. [PMID: 33664994 PMCID: PMC7896131 DOI: 10.1016/j.omtn.2021.01.024] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Coronary artery disease (CAD) is one of the most common causes of death worldwide. The introduction of percutaneous revascularization has revolutionized the therapy of patients with CAD. Despite the advent of drug-eluting stents, restenosis remains the main challenge in treating patients with CAD. In-stent restenosis (ISR) indicates the reduction in lumen diameter after percutaneous coronary intervention, in which the vessel's lumen re-narrowing is attributed to the aberrant proliferation and migration of vascular smooth muscle cells (VSMCs) and dysregulation of endothelial cells (ECs). Increasing evidence has demonstrated that epigenetics is involved in the occurrence and progression of ISR. In this review, we provide the latest and comprehensive analysis of three separate but related epigenetic mechanisms regulating ISR, namely, DNA methylation, histone modification, and non-coding RNAs. Initially, we discuss the mechanism of restenosis. Furthermore, we discuss the biological mechanism underlying the diverse epigenetic modifications modulating gene expression and functions of VSMCs, as well as ECs in ISR. Finally, we discuss potential therapeutic targets of the small molecule inhibitors of cardiovascular epigenetic factors. A more detailed understanding of epigenetic regulation is essential for elucidating this complex biological process, which will assist in developing and improving ISR therapy.
Collapse
Affiliation(s)
- Xi Yang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Road No. 59 Haier, Qingdao 266100, Shandong, People’s Republic of China
| | - Yanyan Yang
- Department of Immunology, School of Basic Medicine, Qingdao University, No. 308 Ningxia Road, Qingdao 266071, People’s Republic of China
| | - Junjie Guo
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Road No. 59 Haier, Qingdao 266100, Shandong, People’s Republic of China
| | - Yuanyuan Meng
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266000, People’s Republic of China
| | - Min Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao 266021, People’s Republic of China
| | - Panyu Yang
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266000, People’s Republic of China
| | - Xin Liu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Road No. 59 Haier, Qingdao 266100, Shandong, People’s Republic of China
| | - Lynn Htet Htet Aung
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao 266021, People’s Republic of China
| | - Tao Yu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266000, People’s Republic of China
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao 266021, People’s Republic of China
| | - Yonghong Li
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Road No. 59 Haier, Qingdao 266100, Shandong, People’s Republic of China
| |
Collapse
|
12
|
Ma H, Dong A. Dysregulation of lncRNA SNHG1/miR-145 axis affects the biological function of human carotid artery smooth muscle cells as a mechanism of carotid artery restenosis. Exp Ther Med 2021; 21:423. [PMID: 33777187 PMCID: PMC7967805 DOI: 10.3892/etm.2021.9867] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 02/02/2021] [Indexed: 12/14/2022] Open
Abstract
Carotid angioplasty and stenting have developed into reliable options for patients with carotid stenosis. However, postoperative restenosis remains a serious and unresolved problem. Restenosis is partly caused by the proliferation of vascular smooth muscle cells. As certain long non-coding RNAs (lncRNAs) affect cell proliferation and migration, the present study aimed to investigate them as novel biomarkers for restenosis development and to further reveal the potential underlying mechanisms. The expression of lncRNA small nucleolar RNA host gene 1 (SNHG1) and microRNA145 (miR-145) in human carotid artery smooth muscle cells (hHCtASMCs) was analyzed using reverse transcription-quantitative PCR. In addition, a luciferase reporter assay was performed to investigate the interaction between SNHG1 and miR-145. The effects of the SNHG1/miR-145 axis on the proliferation and migration of hHCtASMCs were evaluated by Cell Counting Kit-8 and Transwell assays. Serum SNHG1 and miR-145 expression levels were increased and decreased, respectively, in patients with restenosis (all P<0.001). High SNHG1 and low miR-145 were identified as risk factors for restenosis onset (all P<0.01). Furthermore, decreasing SNHG1 expression levels in hHCtASMCs inhibited cell proliferation and migration. The luciferase reporter assay and expression results demonstrated that miR-145 may be a target of SNHG1 and mediated the effects of SNHG1 on hHCtASMC proliferation and migration. The results obtained suggested that abnormal expression of SNHG1 and miR-145 may be risk factors for restenosis. The present study revealed that the SNHG1/miR-145 axis regulates hHCtASMC proliferation and migration, indicating its potential for restenosis prevention and treatment.
Collapse
Affiliation(s)
- Huanhuan Ma
- Department of Neurology, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Aiqin Dong
- Department of Neurology, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| |
Collapse
|
13
|
Bolha L, Pižem J, Frank-Bertoncelj M, Hočevar A, Tomšič M, Jurčić V. Identification of microRNAs and their target gene networks implicated in arterial wall remodelling in giant cell arteritis. Rheumatology (Oxford) 2021; 59:3540-3552. [PMID: 32594153 DOI: 10.1093/rheumatology/keaa204] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 03/31/2020] [Indexed: 01/24/2023] Open
Abstract
OBJECTIVES To identify dysregulated microRNAs (miRNAs) and their gene targets in temporal arteries from GCA patients, and determine their association with GCA pathogenesis and related arterial wall remodelling. METHODS We included 93 formalin-fixed, paraffin-embedded temporal artery biopsies (TABs) from treatment-naïve patients: 54 positive and 17 negative TABs from clinically proven GCA patients, and 22 negative TABs from non-GCA patients. miRNA expression analysis was performed with miRCURY LNA miRNome Human PCR Panels and quantitative real-time PCR. miRNA target gene prediction and pathway enrichment analysis was performed using the miRDB and Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) databases, respectively. RESULTS Dysregulation of 356 miRNAs was determined in TAB-positive GCA arteries, among which 78 were significantly under-expressed and 22 significantly overexpressed above 2-fold, when compared with non-GCA controls. Specifically, TAB-positive GCA arteries were characterized by a significant overexpression of 'pro-synthetic' (miR-21-3p/-21-5p/-146a-5p/-146b-5p/-424-5p) and under-expression of 'pro-contractile' (miR-23b-3p/-125a-5p/-143-3p/-143-5p/-145-3p/-145-5p/-195-5p/-365a-3p) vascular smooth muscle cell phenotype-associated regulatory miRNAs. These miRNAs targeted gene pathways involved in the arterial remodelling and regulation of the immune system, and their expression correlated with the extent of intimal hyperplasia in TABs from GCA patients. Notably, the expression of miR-21-3p/-21-5p/-146a-5p/-146b-5p/-365a-3p differentiated between TAB-negative GCA arteries and non-GCA temporal arteries, revealing these miRNAs as potential biomarkers of GCA. CONCLUSION Identification of dysregulated miRNAs involved in the regulation of the vascular smooth muscle cell phenotype and intimal hyperplasia in GCA arterial lesions, and detection of their expression profiles, enables a novel insight into the complexity of GCA pathogenesis and implies their potential utilization as diagnostic and prognostic biomarkers of GCA.
Collapse
Affiliation(s)
- Luka Bolha
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Jože Pižem
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Mojca Frank-Bertoncelj
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, Schlieren, Switzerland
| | - Alojzija Hočevar
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana.,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Matija Tomšič
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana.,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Vesna Jurčić
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
14
|
Ma H, Dong A. Long non-coding RNA cyclin-dependent kinase inhibitor 2B antisense ribonucleic acid 1 is associated with in-stent restenosis and promotes human carotid artery smooth muscle cell proliferation and migration by sponging miR-143-3p. Exp Ther Med 2021; 21:234. [PMID: 33603842 PMCID: PMC7851615 DOI: 10.3892/etm.2021.9665] [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: 08/21/2020] [Accepted: 12/22/2020] [Indexed: 02/07/2023] Open
Abstract
Carotid angioplasty and stenting (CAS) is an efficient therapeutic approach for carotid stenosis. However, in-stent restenosis (ISR) frequently occurs and seriously affects the therapeutic efficacy of CAS. Certain non-coding (nc)RNAs serve potential roles in ISR development and progression. Thus, the goals of the present study were to investigate novel biomarkers for ISR development and to further uncover the mechanisms underlying the progression of ISR. The expression of long ncRNA cyclin-dependent kinase inhibitor (CDKN)2B-antisense 1 (AS1) and microRNA (miR)-143-3p in patients with ISR and human carotid artery smooth muscle cells (hHCtASMCs) was analyzed using reverse transcription-quantitative PCR. A luciferase reporter assay was performed to examine the interaction between CDKN2B-AS1 and miR-143-3p. The effects of the CDKN2B/miR-143-3p axis on hHCtASMC proliferation and migration were assessed using Cell Counting Kit-8 and Transwell assays. The results indicated that serum CDKN2B-AS1 was increased and miR-143-3p was decreased in patients with ISR as compared with that in patients with no ISR (all P<0.001). CDKN2B-AS1 and miR-143-3p were identified as risk factors for ISR onset (all P<0.05) and knockdown of CDKN2B-AS1 in hHCtASMCs led to inhibited cell proliferation and migration. Furthermore, the luciferase reporter assay and expression analysis indicated that miR-143-3p is a target of CDKN2B-AS1 and may mediate the effects of CDKN2B-AS1 on hHCtASMC proliferation and migration. In conclusion, dysregulation of CDKN2B-AS1 and miR-143-3p may represent risk factors for the occurrence of ISR. The in vitro results suggested that the CDKN2B-AS1/miR-143-3p axis may regulate the proliferation and migration of hHCtASMCs, indicating its potential to be developed as a target for preventative measures and therapies for ISR.
Collapse
Affiliation(s)
- Huanhuan Ma
- Department of Neurology, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Aiqin Dong
- Department of Neurology, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| |
Collapse
|
15
|
Kibel A, Lukinac AM, Dambic V, Juric I, Selthofer-Relatic K. Oxidative Stress in Ischemic Heart Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6627144. [PMID: 33456670 PMCID: PMC7785350 DOI: 10.1155/2020/6627144] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 11/27/2020] [Accepted: 12/07/2020] [Indexed: 02/06/2023]
Abstract
One of the novel interesting topics in the study of cardiovascular disease is the role of the oxidation system, since inflammation and oxidative stress are known to lead to cardiovascular diseases, their progression and complications. During decades of research, many complex interactions between agents of oxidative stress, oxidation, and antioxidant systems have been elucidated, and numerous important pathophysiological links to na number of disorders and diseases have been established. This review article will present the most relevant knowledge linking oxidative stress to vascular dysfunction and disease. The review will focus on the role of oxidative stress in endotheleial dysfunction, atherosclerosis, and other pathogenetic processes and mechanisms that contribute to the development of ischemic heart disease.
Collapse
Affiliation(s)
- Aleksandar Kibel
- Department for Heart and Vascular Diseases, Osijek University Hospital, Osijek, Croatia
- Department of Physiology and Immunology, Faculty of Medicine, University J.J. Strossmayer in Osijek, Osijek, Croatia
| | - Ana Marija Lukinac
- Department of Rheumatology and Clinical Immunology, Osijek University Hospital, Osijek, Croatia
- Faculty of Medicine, University J.J. Strossmayer in Osijek, Osijek, Croatia
| | - Vedran Dambic
- Faculty of Medicine, University J.J. Strossmayer in Osijek, Osijek, Croatia
- Department for Emergency Medical Services of the Osijek-Baranja county, Osijek, Croatia
| | - Iva Juric
- Department for Heart and Vascular Diseases, Osijek University Hospital, Osijek, Croatia
- Department of Internal Medicine, Faculty of Medicine, University J.J. Strossmayer in Osijek, Osijek, Croatia
| | - Kristina Selthofer-Relatic
- Department for Heart and Vascular Diseases, Osijek University Hospital, Osijek, Croatia
- Department of Internal Medicine, Faculty of Medicine, University J.J. Strossmayer in Osijek, Osijek, Croatia
| |
Collapse
|
16
|
Circulating miRNA-23b and miRNA-143 Are Potential Biomarkers for In-Stent Restenosis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2509039. [PMID: 33015157 PMCID: PMC7519453 DOI: 10.1155/2020/2509039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/22/2020] [Accepted: 09/03/2020] [Indexed: 11/27/2022]
Abstract
In-stent restenosis (ISR) is one of the main complications in patients undergoing percutaneous coronary angioplasty, and microRNAs participate in the contractile-to-synthetic phenotypic switch of vascular smooth muscle cells, a hallmark of restenosis development. MicroRNAs (miRNAs) can be released into circulation from injured tissues, enticing a potential role as noninvasive biomarkers. We aimed to evaluate circulating levels of miRNA-23b, miRNA-143, and miRNA-145 as diagnostic markers of ISR. 142 patients with coronary artery disease undergoing successful angioplasty and a follow-up angiography were included. Subjects were classified according to the degree of obstruction at the angioplasty site into cases (≥50%) or controls (<50%). Total RNA was isolated from plasma to quantify circulating miRNAs levels, and the ROC curves were constructed. Among circulating miRNAs assessed, miRNA-23b and miRNA-143 were significantly lower in cases (miRNA-23b: 18.4x10−5 and miRNA-143: 13.7x10−5) than controls (miRNA-23b: 5.2x10−5, p < 0.0001; miRNA-143: 4.0x10−5, p < 0.0001). Plasma levels of miRNA-145 showed no significant differences. The analysis of the ROC curves showed an area under the curve for miRNA-23b of 0.71 (95% CI: 0.62-0.80, p < 0.0001) and 0.69 for miRNA-143 (95% CI: 0.60-0.78; p < 0.0001). Our data suggest that plasma levels of miRNA-23b and miRNA-143 could be useful as noninvasive biomarkers of ISR.
Collapse
|
17
|
Guan X, Yang X, Wang C, Bi R. In silico analysis of the molecular regulatory networks in peripheral arterial occlusive disease. Medicine (Baltimore) 2020; 99:e20404. [PMID: 32481342 PMCID: PMC7250035 DOI: 10.1097/md.0000000000020404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Peripheral arterial occlusive disease (PAOD) is a global public health concern that decreases the quality of life of the patients and can lead to disabilities and death. The aim of this study was to identify the genes and pathways associated with PAOD pathogenesis, and the potential therapeutic targets. METHODS Differentially expressed genes (DEGs) and miRNAs related to PAOD were extracted from the GSE57691 dataset and through text mining. Additionally, bioinformatics analysis was applied to explore gene ontology, pathways and protein-protein interaction of those DEGs. The potential miRNAs targeting the DEGs and the transcription factors (TFs) regulating miRNAs were predicted by multiple different databases. RESULTS A total of 59 DEGs were identified, which were significantly enriched in the inflammatory response, immune response, chemokine-mediated signaling pathway and JAK-STAT signaling pathway. Thirteen genes including IL6, CXCL12, IL1B, and STAT3 were hub genes in protein-protein interaction network. In addition, 513 miRNA-target gene pairs were identified, of which CXCL12 and PTPN11 were the potential targets of miRNA-143, and IL1B of miRNA-21. STAT3 was differentially expressed and regulated 27 potential target miRNAs including miRNA-143 and miRNA-21 in TF-miRNA regulatory network. CONCLUSION In summary, inflammation, immune response and STAT3-mediated miRNA-target genes axis play an important role in PAOD development and progression.
Collapse
Affiliation(s)
| | - Xiaoyan Yang
- Geriatric Department, First People's Hospital of Jingmen City, Jingmen, Hubei Province
| | - Chunming Wang
- Department of Intervention, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | | |
Collapse
|
18
|
Siasos G, Bletsa E, Stampouloglou PK, Oikonomou E, Tsigkou V, Paschou SA, Vlasis K, Marinos G, Vavuranakis M, Stefanadis C, Tousoulis D. MicroRNAs in cardiovascular disease. Hellenic J Cardiol 2020; 61:165-173. [PMID: 32305497 DOI: 10.1016/j.hjc.2020.03.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 03/08/2020] [Accepted: 03/19/2020] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular disease (CVD) remains the predominant cause of human morbidity and mortality in developed countries. Currently, microRNAs have been investigated in many diseases as well-promising biomarkers for diagnosis, prognosis, and disease monitoring. Plenty studies have been designed so as to elucidate the properties of microRNAs in the classification and risk stratification of patients with CVD and also to evaluate their potentials in individualized management and guide treatment decisions. Therefore, in this review article, we aimed to present the most recent data concerning the role of microRNAs as potential novel biomarkers for cardiovascular disease.
Collapse
Affiliation(s)
- Gerasimos Siasos
- Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece; Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Evanthia Bletsa
- Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Panagiota K Stampouloglou
- Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Evangelos Oikonomou
- Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Vasiliki Tsigkou
- Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Stavroula A Paschou
- Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Konstantinos Vlasis
- Department of Anatomy, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Georgios Marinos
- Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Manolis Vavuranakis
- Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Christodoulos Stefanadis
- Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Dimitris Tousoulis
- Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| |
Collapse
|
19
|
Maleknia M, Ansari N, Haybar H, Maniati M, Saki N. Inflammatory Growth Factors and In-Stent Restenosis: Effect of Cytokines and Growth Factors. ACTA ACUST UNITED AC 2020. [DOI: 10.1007/s42399-020-00240-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
20
|
Abstract
Cardiovascular diseases are one of the most common causes of death in both developing and developed countries worldwide. Even though there have been improvements in primary prevention, the prevalence of cardiovascular diseases continues to increase in recent years. Hence, it is crucial to both investigate the molecular pathophysiology of cardiovascular diseases in-depth and find novel biomarkers regarding the early and proper prevention and diagnosis of these diseases. MicroRNAs, or miRNAs, are endogenous, conserved, single-stranded non-coding RNAs of 21-25 nucleotides in length. miRNAs have important roles in various cellular events such as embryogenesis, proliferation, vasculogenesis, apoptosis, cell growth, differentiation, and tumorigenesis. They also have potential roles in the cardiovascular system, including angiogenesis, cardiac cell contractility, control of lipid metabolism, plaque formation, the arrangement of cardiac rhythm, and cardiac cell growth. Circulating miRNAs are promising novel biomarkers for purposes of the diagnosis and prognosis of cardiovascular diseases. Cell or tissue specificity, stability in serum or plasma, resistance to degradative factors such as freeze-thaw cycles or enzymes in the blood, and fast-release kinetics, provide the potential for miRNAs to be surrogate markers for the early and accurate diagnosis of disease and for predicting middle- or long-term prognosis. Moreover, it may be a logical approach to combine miRNAs with traditional biomarkers to improve risk stratification and long-term prognosis. In addition to their efficacy in both diagnosis and prognosis, miRNA-based therapeutics may be beneficial for treating cardiovascular diseases using novel platforms and computational tools and in combination with traditional methods of analysis. microRNAs are promising, novel therapeutic agents, which can affect multiple genes using different signaling pathways. miRNAs therapeutic modulation techniques have been used in the settings of atherosclerosis, acute myocardial infarction, restenosis, vascular remodeling, arrhythmias, hypertrophy and fibrosis, angiogenesis and cardiogenesis, aortic aneurysm, pulmonary hypertension, and ischemic injury. This review presents detailed information about miRNAs regarding structure and biogenesis, stages of synthesis and functions, expression profiles in serum/plasma of living organisms, diagnostic and prognostic potential as novel biomarkers, and therapeutic applications in various diseases.
Collapse
Affiliation(s)
| | - Mehmet Demir
- Department of Cardiology, University of Health Sciences, Bursa Yüksek İhtisas Research and Training Hospital, Bursa, Turkey
| |
Collapse
|
21
|
Varela N, Lanas F, Salazar LA, Zambrano T. The Current State of MicroRNAs as Restenosis Biomarkers. Front Genet 2020; 10:1247. [PMID: 31998354 PMCID: PMC6967329 DOI: 10.3389/fgene.2019.01247] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 11/13/2019] [Indexed: 12/21/2022] Open
Abstract
In-stent restenosis corresponds to the diameter reduction of coronary vessels following percutaneous coronary intervention (PCI), an invasive procedure in which a stent is deployed into the coronary arteries, producing profuse neointimal hyperplasia. The reasons for this process to occur still lack a clear answer, which is partly why it remains as a clinically significant problem. As a consequence, there is a vigorous need to identify useful non-invasive biomarkers to differentiate and follow-up subjects at risk of developing restenosis, and due to their extraordinary stability in several bodily fluids, microRNA research has received extensive attention to accomplish this task. This review depicts the current understanding, diagnostic potential and clinical challenges of microRNA molecules as possible blood-based restenosis biomarkers.
Collapse
Affiliation(s)
- Nelson Varela
- Laboratory of Chemical Carcinogenesis and Pharmacogenetics, Department of Basic-Clinical Oncology, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Fernando Lanas
- Department of Internal Medicine, Faculty of Medicine, Universidad de La Frontera, Temuco, Chile.,Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Temuco, Chile
| | - Luis A Salazar
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Temuco, Chile
| | - Tomás Zambrano
- Department of Medical Technology, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| |
Collapse
|
22
|
Khachigian LM. Transcription Factors Targeted by miRNAs Regulating Smooth Muscle Cell Growth and Intimal Thickening after Vascular Injury. Int J Mol Sci 2019; 20:ijms20215445. [PMID: 31683712 PMCID: PMC6861964 DOI: 10.3390/ijms20215445] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/20/2019] [Accepted: 10/15/2019] [Indexed: 12/21/2022] Open
Abstract
Neointima formation after percutaneous coronary intervention (PCI) is a manifestation of “phenotype switching” by vascular smooth muscle cells (SMC), a process that involves de-differentiation from a contractile quiescent phenotype to one that is richly synthetic. In response to injury, SMCs migrate, proliferate, down-regulate SMC-specific differentiation genes, and later, can revert to the contractile phenotype. The vascular response to injury is regulated by microRNAs (or miRNAs), small non-coding RNAs that control gene expression. Interactions between miRNAs and transcription factors impact gene regulatory networks. This article briefly reviews the roles of a range of miRNAs in molecular and cellular processes that control intimal thickening, focusing mainly on transcription factors, some of which are encoded by immediate-early genes. Examples include Egr-1, junB, KLF4, KLF5, Elk-1, Ets-1, HMGB1, Smad1, Smad3, FoxO4, SRF, Rb, Sp1 and c-Myb. Such mechanistic information could inform the development of strategies that block SMC growth, neointima formation, and potentially overcome limitations of lasting efficacy following PCI.
Collapse
Affiliation(s)
- Levon M Khachigian
- Vascular Biology and Translational Research, School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney NSW 2052, Australia.
| |
Collapse
|
23
|
Jiang F, Zhang X, Lu YM, Li YG, Zhou X, Wang YS. Elevated level of miR-17 along with decreased levels of TIMP-1 and IL-6 in plasma associated with the risk of in-stent restenosis. Biosci Trends 2019; 13:423-429. [PMID: 31666440 DOI: 10.5582/bst.2019.01274] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
In-stent restenosis is highly related to the deposition of inflammatory extracellular matrix and the migration of endothelial and vascular smooth muscle cells. The miR-17/TIMP-1/interleukin pathway regulates vascular matrix remodeling and plays an important role in the inflammatory reaction. This study identified miR-17 and its related biomarkers in serum that potentially indicated susceptibility to in-stent restenosis (ISR) after coronary artery stenting. Subjects were 42 patients with single de novo coronary artery lesions who underwent regular coronary angiography one year after percutaneous coronary intervention. The clinical baseline information was recorded. Serum levels of biomarkers (including miR-17, TIMP-1, IL-6, IL-8, IL-2R, TNF-alpha, IL-10, and IL-1beta) were measured with real-time PCR or ELISA. Intergroup comparisons were used to compare patients with or without ISR. Compared to levels in the non-restenosis group, the serum miR-17 level was significantly higher (3.13 ± 0.22 vs. 1.06 ± 0.04, p < 0.01) and the serum TIMP-1 and IL-6 levels were significantly lower in the ISR group (TIMP-1: 0.33 ± 0.04 vs. 1.00 ± 0.05, p < 0.01; IL-6: 1.64 ± 0.18 vs. 3.52 ± 0.11, p < 0.01). Moreover, the levels of TIMP-1 and IL-6 decreased as the level of miR-17 increased. Spearman's correlation analysis indicated that the miR-17 level was inversely correlated with TIMP-1 and IL-6 levels. Findings suggest that an elevated level of miR-17 and decreased levels of TIMP-1 and IL-6 may be associated with the risk of ISR, which is in accordance with vascular matrix remodeling and an inflammatory reaction during the pathologic process of ISR. This study highlighted the potential for miR-17, TIMP-1, and IL-6 to serve as biomarkers for ISR.
Collapse
Affiliation(s)
- Feng Jiang
- Clinical Research Unit, Xinhua Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Scientific Research Center, Xinhua Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Chongming Branch, Shanghai, China
| | - Xi Zhang
- Clinical Research Unit, Xinhua Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying-Min Lu
- Cardiology, Xinhua Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Chongming Branch, Shanghai, China
| | - Yi-Gang Li
- Cardiology, Xinhua Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Zhou
- Clinical Research Unit, Xinhua Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yao-Sheng Wang
- Clinical Research Unit, Xinhua Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Cardiology, Xinhua Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
24
|
Yang Q, Yang K, Li Z. MiR‐22 restrains proliferation of rheumatoid arthritis by targeting IL6R and may be concerned with the suppression of NF‐κB pathway. Kaohsiung J Med Sci 2019; 36:20-26. [PMID: 31483954 DOI: 10.1002/kjm2.12124] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 08/06/2019] [Indexed: 12/21/2022] Open
Affiliation(s)
- Qing‐Yi Yang
- Department of Joint OrthopaedicAffiliated Hospital of Shandong University of Traditional Chinese Medicine Jinan China
| | - Kai‐Peng Yang
- Department of bone, People's Hospital of Zouping City Binzhou China
| | - Zhi‐Zhou Li
- Department of OrthopeadicsChian‐Japan Union Hospital of Jilin University Changchun China
| |
Collapse
|
25
|
Melak T, Baynes HW. Circulating microRNAs as possible biomarkers for coronary artery disease: a narrative review. EJIFCC 2019; 30:179-194. [PMID: 31263392 PMCID: PMC6599194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Coronary artery disease is one of the most common cardiovascular diseases in the world. Involvement of microRNAs on the pathogenesis of this disease was reported either in beneficial or detrimental way. Different studies have also speculated that circulating microRNAs can be applied as promising biomarkers for the diagnosis of coronary artery disease. Particularly, microRNA-133a seems to fulfill the criteria of ideal biomarkers due to its role in the diagnosis, severity assessment and in prognosis. The panel of circulating microRNAs has also improved the predictive power of coronary artery disease compared to single microRNAs. In this review, the role of circulating microRNAs for early detection, severity assessment and prognosis of coronary artery disease were reviewed.
Collapse
Affiliation(s)
- Tadele Melak
- Department of Clinical Chemistry, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Northwest Ethiopia, Ethiopia
| | | |
Collapse
|
26
|
O'Sullivan JF, Neylon A, Fahy EF, Yang P, McGorrian C, Blake GJ. MiR-93-5p is a novel predictor of coronary in-stent restenosis. HEART ASIA 2019; 11:e011134. [PMID: 31031831 PMCID: PMC6454319 DOI: 10.1136/heartasia-2018-011134] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 01/28/2019] [Accepted: 02/03/2019] [Indexed: 01/31/2023]
Abstract
Aims MicroRNAs (miRNAs), small non-coding RNAs, have been implicated as regulators of multiple phases of atherothrombosis, and some reports have suggested altered levels in coronary artery in-stent restenosis (ISR). We recently demonstrated that miR-93-5 p was able to discriminate between patients with stable coronary artery disease (CAD) and those with no CAD, after adjusting for traditional risk factors (RFs). Thus, we wanted to determine if circulating miRNAs could predict coronary ISR. Objective To determine if circulating miRNAs have diagnostic capability for determining ISR in a cohort of matched patients with and without ISR. Approach and results To determine if miRNA plasma levels are elevated in coronary ISR, we conducted a study comprising 78 patients (39 with no ISR and 39 with ISR) and measured plasma miRNAs in each. We then determined the predictive ability of differential miRNAs, adjusting for Framingham Heart Study (FHS) RFs, and stent length and diameter, to discriminate between ISR and no ISR. After correction for multiple testing, two miRNAs—miR425-5p and miR-93-5 p—were differential between patients with ISR and patients without ISR. Only miR-93-5 p remained a strong independent predictor of ISR after correction for FHS RFs (OR 6.30, p=0.008) and FHS RFs plus stent length and diameter (OR 4.80, p=0.02) and improved discriminatory power for ISR over FHS RFs alone in receiver operator characteristic curve analysis. Conclusion This novel finding that miR-93-5 p independently predicts ISR extends our recent observation that miR-93-5 p predicted CAD after adjustment for traditional CAD RFs. These data suggest further potential diagnostic utility.
Collapse
Affiliation(s)
- John F O'Sullivan
- Department of Cardiology, Mater Misericordiae University Hospital, Dublin, Ireland.,The University of Sydney - Department of Cardiology, Royal Prince Alfred Hospital; Heart Research Institute, Charles Perkins Centre, Johns Hopkins Drive, The University of Sydney, Sydney, New South Wales, Australia
| | - Antoinette Neylon
- Department of Cardiology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Eoin F Fahy
- Department of Cardiology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Pengyi Yang
- The University of Sydney - Computational Trans-Regulatory Biology Group, School of Mathematics and Statistics, The University of Sydney, Sydney, New South Wales, Australia
| | - Catherine McGorrian
- Department of Cardiology, Mater Misericordiae University Hospital, Dublin, Ireland.,School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | - Gavin J Blake
- Department of Cardiology, Mater Misericordiae University Hospital, Dublin, Ireland.,School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
27
|
Diagnostic Value of Circulating microRNAs for In-Stent Restenosis in Patients with Lower Extremity Arterial Occlusive Disease. Sci Rep 2019; 9:1402. [PMID: 30718561 PMCID: PMC6361988 DOI: 10.1038/s41598-018-36295-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 11/13/2018] [Indexed: 12/15/2022] Open
Abstract
In-stent restenosis (ISR) is still a major cause of failure of endovascular stenting treatment in patients with lower extremity arterial occlusive disease (LEAOD). Sensitive and reliable biomarkers for early diagnosis to predict ISR should be considered. This study was conducted to explore the diagnostic value of microRNA in predicting ISR in patients with LEAOD after endovascular stenting treatment. From March 2014 to July 2016, 208 patients (170 males and 38 females) with LEAOD undergoing interventional treatment were enrolled in this research. Patients were divided into the restenosis and non-restenosis groups according to routine postoperative angiography. Circulating microRNAs expression were detected in 208 participants, including 78 ISR patients, 68 non-ISR patients and 62 healthy volunteers. We selected 6 microRNAs from microarray screening as candidates for further testing via qRT-PCR. A receiver operating characteristic (ROC) curve was generated to assess the diagnostic value of circulating microRNAs in predicting ISR for LEAOD patients. The results showed that circulating microRNA-320a and microRNA-572 in patients with ISR (n = 78) had significantly higher expression levels than it from non-ISR and healthy volunteers. By receiver operating characteristic curve analysis, the sensitivity was 82.1% and the specificity was 63.8% for microRNA-320a; the sensitivity was 69.2% and the specificity was 68.9% for microRNA-572, and the AUC was 0.766 and 0.690 for detection of ISR, respectively. Furthermore, 78 patients with ISR had significantly higher circulating expression levels of microRNA-3937 and microRNA-642a-3p and lower circulating expression levels of microRNA-4669 and microRNA-3138 compared with 68 non-ISR patients and 62 healthy volunteers, but they have no significant difference. We found that differential circulating microRNA expression in patients after stenting with ISR, and the data indicate that circulating microRNA-320a and microRNA-572 have promising value in diagnosing ISR in patients with LEAOD.
Collapse
|
28
|
Fragoso JM, Ramírez-Bello J, Martínez-Ríos MA, Peña-Duque MA, Posadas-Sánchez R, Delgadillo-Rodríguez H, Jiménez-Morales M, Posadas-Romero C, Vargas-Alarcón G. miR-196a2 (rs11614913) polymorphism is associated with coronary artery disease, but not with in-stent coronary restenosis. Inflamm Res 2018; 68:215-221. [PMID: 30560371 DOI: 10.1007/s00011-018-1206-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 12/02/2018] [Accepted: 12/10/2018] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE The aim of the study was to evaluate the association of miRNA-146a G/C (rs2910164), and miRNA-196a2 C/T (rs11614913) polymorphisms with the presence of coronary artery disease (CAD) and/or restenosis in patients with coronary stent. MATERIALS AND METHODS The polymorphisms were determined in 218 patients with CAD who underwent coronary artery stenting (66 with restenosis and 152 without restenosis) and 611 healthy controls using 5' exonuclease TaqMan assays. RESULTS The distribution of both polymorphisms was similar in patients with and without restenosis. However, when the whole group of patients (with and without restenosis) was compared to healthy controls, under co-dominant, dominant and additive genetic models, the T allele of the miRNA-196a2 C/T (rs11614913) polymorphism was associated with increased risk of CAD (OR = 2.18, Pco-dom = 0.006, OR = 1.86, Pdom = 0.002, and OR = 1.52, Padd = 0.002, respectively). All models were adjusted for age, type 2 diabetes mellitus, dyslipidemia, hypertension and smoking habit. The "GT" haplotype was associated with increased risk of developing CAD (OR = 1.36, P = 0.046). CONCLUSIONS Our data suggests that the T allele of the miRNA-196a2 C/T (rs11614913) polymorphism is associated with the risk of developing CAD, but no association with restenosis was observed.
Collapse
Affiliation(s)
- José Manuel Fragoso
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano 1, Sección XVI, Del. Tlalpan, 14080, Mexico City, Mexico
| | - Julian Ramírez-Bello
- Endocrine and Metabolic Diseases Research Unit, Hospital Juárez de Mexico, Mexico City, Mexico
| | | | - Marco Antonio Peña-Duque
- Interventional Cardiology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | | | | | - Mayra Jiménez-Morales
- Endocrine and Metabolic Diseases Research Unit, Hospital Juárez de Mexico, Mexico City, Mexico
| | - Carlos Posadas-Romero
- Department of Endocrinology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Gilberto Vargas-Alarcón
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano 1, Sección XVI, Del. Tlalpan, 14080, Mexico City, Mexico.
| |
Collapse
|
29
|
Johnson JL. Elucidating the contributory role of microRNA to cardiovascular diseases (a review). Vascul Pharmacol 2018; 114:31-48. [PMID: 30389614 PMCID: PMC6445803 DOI: 10.1016/j.vph.2018.10.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 07/13/2018] [Accepted: 10/28/2018] [Indexed: 02/07/2023]
Abstract
Cardiovascular diseases encompassing atherosclerosis, aortic aneurysms, restenosis, and pulmonary arterial hypertension, remain the leading cause of morbidity and mortality worldwide. In response to a range of stimuli, the dynamic interplay between biochemical and biomechanical mechanisms affect the behaviour and function of multiple cell types, driving the development and progression of cardiovascular diseases. Accumulating evidence has highlighted microRNAs (miRs) as significant regulators and micro-managers of key cellular and molecular pathophysiological processes involved in predominant cardiovascular diseases, including cell mitosis, motility and viability, lipid metabolism, generation of inflammatory mediators, and dysregulated proteolysis. Human pathological and clinical studies have aimed to identify select microRNA which may serve as biomarkers of disease and their progression, which are discussed within this review. In addition, I provide comprehensive coverage of in vivo investigations elucidating the modulation of distinct microRNA on the pathophysiology of atherosclerosis, abdominal aortic aneurysms, restenosis, and pulmonary arterial hypertension. Collectively, clinical and animal studies have begun to unravel the complex and often diverse effects microRNAs and their targets impart during the development of cardiovascular diseases and revealed promising therapeutic strategies through which modulation of microRNA function may be applied clinically.
Collapse
Affiliation(s)
- Jason L Johnson
- Laboratory of Cardiovascular Pathology, Bristol Medical School, University of Bristol, UK.
| |
Collapse
|
30
|
Indolfi C, Iaconetti C, Gareri C, Polimeni A, De Rosa S. Non-coding RNAs in vascular remodeling and restenosis. Vascul Pharmacol 2018; 114:49-63. [PMID: 30368024 DOI: 10.1016/j.vph.2018.10.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Revised: 10/08/2018] [Accepted: 10/18/2018] [Indexed: 02/06/2023]
Abstract
Vascular smooth muscle cells (VSMCs) and endothelial cells (ECs) are crucial in vascular remodeling. They exert pivotal roles in the development and progression of atherosclerosis, vascular response to injury, and restenosis after transcatheter angioplasty. As a witness of their importance in the cardiovascular system, a large body of evidence has accumulated about the role played by micro RNAs (miRNA) in modulating both VSMCs and ECs. More recently, a growing number of long noncoding RNA (lncRNAs) came beneath the spotlights in this research field. Several mechanisms have been revealed by which lncRNAs are able to exert a relevant biological impact on vascular remodeling. The aim of this review is to provide an integrated summary of ncRNAs that exert a relevant biological function in VSMCs and ECs of the vascular wall, with emphasis on the available clinical evidence of the potential usefulness of these molecules as circulating biomarkers of in-stent restenosis.
Collapse
Affiliation(s)
- Ciro Indolfi
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Italy; URT CNR of IFC, University Magna Graecia, Italy.
| | - Claudio Iaconetti
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Italy
| | - Clarice Gareri
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Italy
| | - Alberto Polimeni
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Italy
| | - Salvatore De Rosa
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Italy
| |
Collapse
|
31
|
Endothelial cell activation is attenuated by everolimus via transcriptional and post-transcriptional regulatory mechanisms after drug-eluting coronary stenting. PLoS One 2018; 13:e0197890. [PMID: 29889836 PMCID: PMC5995375 DOI: 10.1371/journal.pone.0197890] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 05/10/2018] [Indexed: 01/26/2023] Open
Abstract
We previously found higher level of endothelial cell (EC) activation in patients who suffered from in-stent restenosis after bare-metal stenting compared to subjects who underwent drug-eluting stenting (DES) showing no complications. Here we investigated the potential transcriptional and post-transcriptional regulatory mechanisms by which everolimus attenuated EC activation after DES. We studied the effect of everolimus on E-selectin (SELE) and VCAM1 mRNA levels when human coronary artery (HCAECs) and human umbilical vein ECs were challenged with recombinant TNF-α (100 ng/mL) for 1–24 hours in the presence or absence of everolimus using 0.5 μM concentration locally maintained by DES. EC activation was evaluated via the levels of IL-1β and IL-6 mRNAs with miR-155 expression by RT-qPCR as well as the nuclear translocation of nuclear factor kappa beta (NF-κB) detected by fluorescence microscopy. To investigate the transcriptional regulation of E-selectin and VCAM-1, TNF-α-induced enhancer RNA (eRNA) expression at p65-bound enhancers in the neighboring genomic regions of SELE and VCAM1 genes, including SELE_-11Kb and VCAM1_-10Kb, were measured in HCAECs. Mature and precursor levels of E-selectin and VCAM-1 repressor miR-181b were quantified to analyze the post-transcriptional regulation of these genes in HCAECs. Circulating miR-181b was analyzed in plasma samples of stented subjects by stem-loop RT-qPCR. TNF-α highly elevated E-selectin and VCAM-1 expression at transcriptional level in ECs. Levels of mature, pre- and pri-miR-181b were repressed in ECs by TNF-α, while everolimus acted as a negative regulator of EC activation via inhibited translocation of NF-κB p65 subunit into cell nuclei, lowered eRNA expression at SELE and VCAM1 genes-associated enhancers and modulated expression of their post-transcriptional repressor miR-181b. Significant negative correlation was observed between plasma miR-181b and soluble E-selectin and VCAM-1 in patients. In conclusion, everolimus attenuates EC activation via reduced NF-κB p65 translocation causing decreased E-selectin and VCAM-1 expression at transcriptional and post-transcriptional level after DES.
Collapse
|
32
|
Stojkovic S, Jurisic M, Kopp CW, Koppensteiner R, Huber K, Wojta J, Gremmel T. Circulating microRNAs identify patients at increased risk of in-stent restenosis after peripheral angioplasty with stent implantation. Atherosclerosis 2018; 269:197-203. [PMID: 29366993 DOI: 10.1016/j.atherosclerosis.2018.01.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Revised: 12/24/2017] [Accepted: 01/12/2018] [Indexed: 11/18/2022]
Abstract
BACKGROUND AND AIMS Target lesion restenosis is the most frequent complication after angioplasty and stenting for peripheral artery disease (PAD). MicroRNAs (miRs) regulate crucial pathophysiological processes leading to in-stent restenosis and thrombosis. The aim of this study was to investigate the predictive value of 11 miRs for the composite endpoint of target lesion restenosis and atherothrombotic events (primary endpoint), and target vessel revascularization (TVR, secondary endpoint) in 62 consecutive PAD patients after infrainguinal angioplasty with stent implantation. METHODS Circulating miRs were assessed using quantitative real-time polymerase chain reactions. RESULTS Within the 2 years of follow-up, the primary endpoint occurred in 26 patients (41.9%), and 21 patients (33.9%) underwent TVR. miR-92a and miR-195 were identified as independent predictors of the primary endpoint after adjustment for age, sex and clinical risk factors with respective HR per 1 increase of standard deviation (1-SD) of 0.55 (95% CI: 0.34-0.88, p = 0.013) and HR per 1-SD of 0.40 (95% CI: 0.23-0.68, p = 0.001). MiR-195 independently predicted TVR with HR per 1-SD of 0.40 (95% CI: 0.22-0.75, p = 0.005). Adding miR-195 to clinical risk factors improved Harrell's C-index to 0.75 (95% CI: 0.66-0.85, p = 0.03) and was superior to a model with miR-92a (C-index: 0.70, 95% CI: 0.60-0.80, p for comparison =0 .012). Assessment of both miR-92a and miR-195 had no incremental value when compared to miR-195 alone (C-index: 0.79, 95% CI: 0.69-0.88, p = 0.313). CONCLUSIONS Circulating miR-195 predicts adverse ischemic events and TVR after infrainguinal angioplasty with stent implantation. MiR-195 could improve risk stratification after peripheral endovascular revascularizations.
Collapse
Affiliation(s)
- Stefan Stojkovic
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Magdalena Jurisic
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Christoph W Kopp
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Renate Koppensteiner
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Kurt Huber
- 3rd Medical Department for Cardiology and Emergency Medicine, Wilhelminen Hospital, Vienna, Austria; Sigmund Freud Private University, Medical School, Vienna, Austria
| | - Johann Wojta
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria; Core Facilities, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Cluster for Cardiovascular Research, Vienna, Austria
| | - Thomas Gremmel
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
33
|
Reddy S, Hu DQ, Zhao M, Blay E, Sandeep N, Ong SG, Jung G, Kooiker KB, Coronado M, Fajardo G, Bernstein D. miR-21 is associated with fibrosis and right ventricular failure. JCI Insight 2017; 2:91625. [PMID: 28469078 DOI: 10.1172/jci.insight.91625] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 03/23/2017] [Indexed: 12/14/2022] Open
Abstract
Combined pulmonary insufficiency (PI) and stenosis (PS) is a common long-term sequela after repair of many forms of congenital heart disease, causing progressive right ventricular (RV) dilation and failure. Little is known of the mechanisms underlying this combination of preload and afterload stressors. We developed a murine model of PI and PS (PI+PS) to identify clinically relevant pathways and biomarkers of disease progression. Diastolic dysfunction was induced (restrictive RV filling, elevated RV end-diastolic pressures) at 1 month after generation of PI+PS and progressed to systolic dysfunction (decreased RV shortening) by 3 months. RV fibrosis progressed from 1 month (4.4% ± 0.4%) to 3 months (9.2% ± 1%), along with TGF-β signaling and tissue expression of profibrotic miR-21. Although plasma miR-21 was upregulated with diastolic dysfunction, it was downregulated with the onset of systolic dysfunction), correlating with RV fibrosis. Plasma miR-21 in children with PI+PS followed a similar pattern. A model of combined RV volume and pressure overload recapitulates the evolution of RV failure unique to patients with prior RV outflow tract surgery. This progression was characterized by enhanced TGF-β and miR-21 signaling. miR-21 may serve as a plasma biomarker of RV failure, with decreased expression heralding the need for valve replacement.
Collapse
Affiliation(s)
- Sushma Reddy
- Department of Pediatrics, Division of Cardiology, Stanford University, Stanford, California, USA
| | - Dong-Qing Hu
- Department of Pediatrics, Division of Cardiology, Stanford University, Stanford, California, USA
| | - Mingming Zhao
- Department of Pediatrics, Division of Cardiology, Stanford University, Stanford, California, USA
| | - Eddie Blay
- Department of Surgery, Temple University, Philadelphia, Pennsylvania, USA
| | - Nefthi Sandeep
- Department of Pediatrics, Division of Cardiology, Stanford University, Stanford, California, USA
| | - Sang-Ging Ong
- Cardiovascular Institute, Stanford University, Stanford, California, USA
| | - Gwanghyun Jung
- Department of Pediatrics, Division of Cardiology, Stanford University, Stanford, California, USA
| | - Kristina B Kooiker
- Department of Pediatrics, Division of Cardiology, Stanford University, Stanford, California, USA
| | - Michael Coronado
- Department of Pediatrics, Division of Cardiology, Stanford University, Stanford, California, USA
| | - Giovanni Fajardo
- Department of Pediatrics, Division of Cardiology, Stanford University, Stanford, California, USA
| | - Daniel Bernstein
- Department of Pediatrics, Division of Cardiology, Stanford University, Stanford, California, USA
| |
Collapse
|
34
|
Circulating mircoRNA-21 as a predictor for vascular restenosis after interventional therapy in patients with lower extremity arterial occlusive disease. Biosci Rep 2017; 37:BSR20160502. [PMID: 28250135 PMCID: PMC5484015 DOI: 10.1042/bsr20160502] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 02/25/2017] [Accepted: 02/28/2017] [Indexed: 12/27/2022] Open
Abstract
The present study was designed to investigate the role of circulating miRNA-21
(miR-21) in vascular restenosis of lower extremity arterial occlusive disease
(LEAOD) patients after interventional therapy. A total of 412 LEAOD patients were enrolled
randomly in the present study. According to computed tomography angiography (CTA) and
ankle-brachial index (ABI), patients were assigned into the restenosis group and the
non-restenosis group. miR-21 expression was detected with quantitative
real-time PCR (qRT-PCR) before and after patients underwent interventional therapy. A follow-up
period of 6 months was achieved. A receiver operating characteristic (ROC) curve was drawn and
the area under the curve (AUC) was calculated to assess the predictive value of
miR-21 in vascular restenosis. Patients were older in the restenosis group
than in the non-restenosis group. The percentages of patients with diabetes and hypertension
were higher in the restenosis group than in the non-restenosis group, and the Fontaine stage
exhibited a significant difference between the two groups. miR-21 expression
was higher in the restenosis group than in the non-restenosis group. miR-21
expression level was related to age, diabetes and hypertension in the restenosis group. Using
miR-21 to predict vascular restenosis yielded an AUC of 0.938 (95%
confidence interval (CI): 0.898–0.977), with Youden index of 0.817, sensitivity of
83.5% and specificity of 98.2%. Logistic regression analysis revealed that
diabetes and miR-21 expression were the major risk factors for vascular
restenosis of LEAOD. miR-21 can be used as a predictive indicator for vascular
restenosis of LEAOD after interventional therapy.
Collapse
|
35
|
Schulte C, Karakas M, Zeller T. microRNAs in cardiovascular disease - clinical application. Clin Chem Lab Med 2017; 55:687-704. [PMID: 27914211 DOI: 10.1515/cclm-2016-0576] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 11/02/2016] [Indexed: 12/11/2022]
Abstract
microRNAs (miRNAs) are well-known, powerful regulators of gene expression, and their potential to serve as circulating biomarkers is widely accepted. In cardiovascular disease (CVD), numerous studies have suggested miRNAs as strong circulating biomarkers with high diagnostic as well as prognostic power. In coronary artery disease (CAD) and heart failure (HF), miRNAs have been suggested as reliable biomarkers matching up to established protein-based such as cardiac troponins (cT) or natriuretic peptides. Also, in other CVD entities, miRNAs were identified as surprisingly specific biomarkers - with great potential for clinical applicability, especially in those entities that lack specific protein-based biomarkers such as atrial fibrillation (AF) and acute pulmonary embolism (APE). In this regard, miRNA signatures, comprising a set of miRNAs, yield high sensitivity and specificity. Attempts to utilize miRNAs as therapeutic agents have led to promising results. In this article, we review the clinical applicability of circulating miRNAs in CVD. We are giving an overview of miRNAs as biomarkers in numerous CVD entities to depict the variety of their potential clinical deployment. We illustrate the function of miRNAs by means of single miRNA examples in CVD.
Collapse
Affiliation(s)
- Christian Schulte
- Department of General and Interventional Cardiology, University Heart Center Hamburg Eppendorf, Hamburg
| | - Mahir Karakas
- Department of General and Interventional Cardiology, University Heart Center Hamburg Eppendorf, Hamburg
| | - Tanja Zeller
- Department of General and Interventional Cardiology, University Heart Center Hamburg Eppendorf, Hamburg
| |
Collapse
|
36
|
Yu ZH, Wang HT, Tu C. Diagnostic value of microRNA-143 in predicting in-stent restenosis for patients with lower extremity arterial occlusive disease. Eur J Med Res 2017; 22:2. [PMID: 28057086 PMCID: PMC5217340 DOI: 10.1186/s40001-016-0240-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 11/24/2016] [Indexed: 01/26/2023] Open
Abstract
Purpose This study was conducted to explore the diagnostic value of microRNA-143 (miRNA-143) in predicting in-stent restenosis (ISR) of lower extremity arterial occlusive disease (LEAOD). Methods From February 2012 to March 2015, 165 patients (112 males and 53 females) with LEAOD undergoing interventional treatment were enrolled in this study. Serum miRNA-143 expression was detected using quantitative real-time polymerase chain reaction (qRT-PCR). Patients were assigned into the restenosis and non-restenosis groups according to routine surveillance postoperative angiography. A logistic regression analysis was conducted to analyze the risk factors for ISR in LEAOD patients. A receiver operating characteristic (ROC) curve was drawn to evaluate the diagnostic value of miRNA-143 in predicting ISR for LEAOD patients. Results There were 74 and 91 patients in the restenosis and non-restenosis groups, respectively. Before the treatment, there were significant differences in history of diabetes, smoking status, blood sugar level (BSL) at admission, low-density lipoprotein cholesterol (LDL-C) level, and stent diameter between the restenosis and non-restenosis groups (all P < 0.05). Serum miRNA-143 expression was lower in the restenosis group than in the non-restenosis group (P < 0.05). Serum miRNA-143 expression in the restenosis group was correlated with smoking status, history of diabetes, BSL, and LDL-C (all P < 0.05). Logistic regression analysis demonstrated that miRNA-143, LDL-C, and smoking status were correlated with the postoperative ISR (all P < 0.05). ROC curve analysis revealed that the area under the curve (AUC) of miRNA-143 in predicting ISR for LEAOD patients was 0.866. Conclusion Our results indicate that miRNA-143 can be a promising tool for predicting the ISR in LEAOD patients.
Collapse
Affiliation(s)
- Zhi-Hai Yu
- Department of Interventional, The Affiliated Hospital of School of Medicine of Ningbo University, No. 247 Renmin Road, Jiangbei District, Ningbo, 315020, Zhejiang Province, People's Republic of China.
| | - Hai-Tao Wang
- Department of Interventional, The Affiliated Hospital of School of Medicine of Ningbo University, No. 247 Renmin Road, Jiangbei District, Ningbo, 315020, Zhejiang Province, People's Republic of China
| | - Can Tu
- Department of Interventional, The Affiliated Hospital of School of Medicine of Ningbo University, No. 247 Renmin Road, Jiangbei District, Ningbo, 315020, Zhejiang Province, People's Republic of China
| |
Collapse
|
37
|
Pujol-López M, Ortega-Paz L, Garabito M, Brugaletta S, Sabaté M, Paula Dantas A. miRNA Update: A Review Focus on Clinical Implications of miRNA in Vascular Remodeling. AIMS MEDICAL SCIENCE 2017. [DOI: 10.3934/medsci.2017.1.99] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
38
|
Abstract
PURPOSE OF REVIEW Noncoding RNAs (ncRNAs), such as microRNAs (miRNAs) and long noncoding RNAs (lncRNAs) are becoming fundamentally important in the pathophysiology relating to injury-induced vascular remodelling. We highlight recent studies that demonstrate the involvement of ncRNAs in vein graft disease, in in-stent restenosis and in pulmonary arterial hypertension, with a particular focus on endothelial cell and vascular smooth muscle cell function. We also briefly discuss the emerging role of exosomal-derived ncRNAs and how this mechanism impacts on vascular function. RECENT FINDINGS ncRNAs have been described as novel regulators in the pathophysiology of vascular injury, inflammation, and vessel wall remodelling. In particular, several studies have demonstrated that manipulation of miRNAs can reduce the burden of pathological vascular remodelling. Such studies have also shown that exosomal miRNA-mediated, cell-to-cell communication between endothelial cells and vascular smooth muscle cells is critical in the disease process. In addition to miRNAs, lncRNAs are emerging as regulators of vascular function in health and disease. Although lncRNAs are complex in both their sheer numbers and mechanisms of action, identifying their contribution to vascular disease is essential. SUMMARY Given the important roles of ncRNAs in vascular injury and remodelling together will their capacity for cell-to-cell communication, manipulating ncRNA might provide novel therapeutic interventions.
Collapse
Affiliation(s)
- Lin Deng
- aBHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow bCentre for Cardiovascular Science, Queen's Medical Research Institute, BHF/University of Edinburgh, Edinburgh, UK
| | | | | |
Collapse
|
39
|
Gareri C, De Rosa S, Indolfi C. MicroRNAs for Restenosis and Thrombosis After Vascular Injury. Circ Res 2016; 118:1170-84. [PMID: 27034278 DOI: 10.1161/circresaha.115.308237] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 03/01/2016] [Indexed: 12/21/2022]
Abstract
Percutaneous revascularization revolutionized the therapy of patients with coronary artery disease. Despite continuous technical advances that substantially improved patients' outcome after percutaneous revascularization, some issues are still open. In particular, restenosis still represents a challenge, even though it was dramatically reduced with the advent of drug-eluting stents. At the same time, drug-eluting stent thrombosis emerged as a major concern because of incomplete or delayed re-endothelialization after vascular injury. The discovery of microRNAs revealed a previously unknown layer of regulation for several biological processes, increasing our knowledge on the biological mechanisms underlying restenosis and stent thrombosis, revealing novel promising targets for more efficient and selective therapies. The present review summarizes recent experimental and clinical evidence on the role of microRNAs after arterial injury, focusing on practical aspects of their potential therapeutic application for selective inhibition of smooth muscle cell proliferation, enhancement of endothelial regeneration, and inhibition of platelet activation after coronary interventions. Application of circulating microRNAs as potential biomarkers is also discussed.
Collapse
Affiliation(s)
- Clarice Gareri
- From the Department of Medicine, Duke University, Durham, NC (C.G.); Division of Cardiology, Department of Medical and Surgical Science, "Magna Graecia" University, Catanzaro, Italy (S.D.R., C.I.); and URT-CNR, Department of Medicine, URT of Consiglio Nazionale delle Ricerche, Catanzaro, Italy (C.I.)
| | - Salvatore De Rosa
- From the Department of Medicine, Duke University, Durham, NC (C.G.); Division of Cardiology, Department of Medical and Surgical Science, "Magna Graecia" University, Catanzaro, Italy (S.D.R., C.I.); and URT-CNR, Department of Medicine, URT of Consiglio Nazionale delle Ricerche, Catanzaro, Italy (C.I.)
| | - Ciro Indolfi
- From the Department of Medicine, Duke University, Durham, NC (C.G.); Division of Cardiology, Department of Medical and Surgical Science, "Magna Graecia" University, Catanzaro, Italy (S.D.R., C.I.); and URT-CNR, Department of Medicine, URT of Consiglio Nazionale delle Ricerche, Catanzaro, Italy (C.I.).
| |
Collapse
|
40
|
Ballantyne MD, Pinel K, Dakin R, Vesey AT, Diver L, Mackenzie R, Garcia R, Welsh P, Sattar N, Hamilton G, Joshi N, Dweck MR, Miano JM, McBride MW, Newby DE, McDonald RA, Baker AH. Smooth Muscle Enriched Long Noncoding RNA (SMILR) Regulates Cell Proliferation. Circulation 2016; 133:2050-65. [PMID: 27052414 PMCID: PMC4872641 DOI: 10.1161/circulationaha.115.021019] [Citation(s) in RCA: 172] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 03/28/2016] [Indexed: 01/07/2023]
Abstract
BACKGROUND Phenotypic switching of vascular smooth muscle cells from a contractile to a synthetic state is implicated in diverse vascular pathologies, including atherogenesis, plaque stabilization, and neointimal hyperplasia. However, very little is known about the role of long noncoding RNA (lncRNA) during this process. Here, we investigated a role for lncRNAs in vascular smooth muscle cell biology and pathology. METHODS AND RESULTS Using RNA sequencing, we identified >300 lncRNAs whose expression was altered in human saphenous vein vascular smooth muscle cells following stimulation with interleukin-1α and platelet-derived growth factor. We focused on a novel lncRNA (Ensembl: RP11-94A24.1), which we termed smooth muscle-induced lncRNA enhances replication (SMILR). Following stimulation, SMILR expression was increased in both the nucleus and cytoplasm, and was detected in conditioned media. Furthermore, knockdown of SMILR markedly reduced cell proliferation. Mechanistically, we noted that expression of genes proximal to SMILR was also altered by interleukin-1α/platelet-derived growth factor treatment, and HAS2 expression was reduced by SMILR knockdown. In human samples, we observed increased expression of SMILR in unstable atherosclerotic plaques and detected increased levels in plasma from patients with high plasma C-reactive protein. CONCLUSIONS These results identify SMILR as a driver of vascular smooth muscle cell proliferation and suggest that modulation of SMILR may be a novel therapeutic strategy to reduce vascular pathologies.
Collapse
Affiliation(s)
- Margaret D Ballantyne
- From BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (M.D.B., R.D., L.D., R.M., R.G., P.W., N.S., M.W.N., R.A.M., A.H.B.); British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, Edinburgh, United Kingdom (M.D.B., K.P., A.T.V., N.J., M.R.D., D.E.N., R.A.M., A.H.B.); Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, The University of Glasgow, United Kingdom (G.H.); and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Karine Pinel
- From BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (M.D.B., R.D., L.D., R.M., R.G., P.W., N.S., M.W.N., R.A.M., A.H.B.); British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, Edinburgh, United Kingdom (M.D.B., K.P., A.T.V., N.J., M.R.D., D.E.N., R.A.M., A.H.B.); Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, The University of Glasgow, United Kingdom (G.H.); and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Rachel Dakin
- From BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (M.D.B., R.D., L.D., R.M., R.G., P.W., N.S., M.W.N., R.A.M., A.H.B.); British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, Edinburgh, United Kingdom (M.D.B., K.P., A.T.V., N.J., M.R.D., D.E.N., R.A.M., A.H.B.); Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, The University of Glasgow, United Kingdom (G.H.); and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Alex T Vesey
- From BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (M.D.B., R.D., L.D., R.M., R.G., P.W., N.S., M.W.N., R.A.M., A.H.B.); British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, Edinburgh, United Kingdom (M.D.B., K.P., A.T.V., N.J., M.R.D., D.E.N., R.A.M., A.H.B.); Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, The University of Glasgow, United Kingdom (G.H.); and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Louise Diver
- From BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (M.D.B., R.D., L.D., R.M., R.G., P.W., N.S., M.W.N., R.A.M., A.H.B.); British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, Edinburgh, United Kingdom (M.D.B., K.P., A.T.V., N.J., M.R.D., D.E.N., R.A.M., A.H.B.); Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, The University of Glasgow, United Kingdom (G.H.); and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Ruth Mackenzie
- From BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (M.D.B., R.D., L.D., R.M., R.G., P.W., N.S., M.W.N., R.A.M., A.H.B.); British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, Edinburgh, United Kingdom (M.D.B., K.P., A.T.V., N.J., M.R.D., D.E.N., R.A.M., A.H.B.); Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, The University of Glasgow, United Kingdom (G.H.); and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Raquel Garcia
- From BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (M.D.B., R.D., L.D., R.M., R.G., P.W., N.S., M.W.N., R.A.M., A.H.B.); British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, Edinburgh, United Kingdom (M.D.B., K.P., A.T.V., N.J., M.R.D., D.E.N., R.A.M., A.H.B.); Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, The University of Glasgow, United Kingdom (G.H.); and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Paul Welsh
- From BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (M.D.B., R.D., L.D., R.M., R.G., P.W., N.S., M.W.N., R.A.M., A.H.B.); British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, Edinburgh, United Kingdom (M.D.B., K.P., A.T.V., N.J., M.R.D., D.E.N., R.A.M., A.H.B.); Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, The University of Glasgow, United Kingdom (G.H.); and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Naveed Sattar
- From BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (M.D.B., R.D., L.D., R.M., R.G., P.W., N.S., M.W.N., R.A.M., A.H.B.); British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, Edinburgh, United Kingdom (M.D.B., K.P., A.T.V., N.J., M.R.D., D.E.N., R.A.M., A.H.B.); Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, The University of Glasgow, United Kingdom (G.H.); and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Graham Hamilton
- From BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (M.D.B., R.D., L.D., R.M., R.G., P.W., N.S., M.W.N., R.A.M., A.H.B.); British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, Edinburgh, United Kingdom (M.D.B., K.P., A.T.V., N.J., M.R.D., D.E.N., R.A.M., A.H.B.); Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, The University of Glasgow, United Kingdom (G.H.); and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Nikhil Joshi
- From BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (M.D.B., R.D., L.D., R.M., R.G., P.W., N.S., M.W.N., R.A.M., A.H.B.); British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, Edinburgh, United Kingdom (M.D.B., K.P., A.T.V., N.J., M.R.D., D.E.N., R.A.M., A.H.B.); Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, The University of Glasgow, United Kingdom (G.H.); and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Marc R Dweck
- From BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (M.D.B., R.D., L.D., R.M., R.G., P.W., N.S., M.W.N., R.A.M., A.H.B.); British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, Edinburgh, United Kingdom (M.D.B., K.P., A.T.V., N.J., M.R.D., D.E.N., R.A.M., A.H.B.); Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, The University of Glasgow, United Kingdom (G.H.); and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Joseph M Miano
- From BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (M.D.B., R.D., L.D., R.M., R.G., P.W., N.S., M.W.N., R.A.M., A.H.B.); British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, Edinburgh, United Kingdom (M.D.B., K.P., A.T.V., N.J., M.R.D., D.E.N., R.A.M., A.H.B.); Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, The University of Glasgow, United Kingdom (G.H.); and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Martin W McBride
- From BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (M.D.B., R.D., L.D., R.M., R.G., P.W., N.S., M.W.N., R.A.M., A.H.B.); British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, Edinburgh, United Kingdom (M.D.B., K.P., A.T.V., N.J., M.R.D., D.E.N., R.A.M., A.H.B.); Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, The University of Glasgow, United Kingdom (G.H.); and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - David E Newby
- From BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (M.D.B., R.D., L.D., R.M., R.G., P.W., N.S., M.W.N., R.A.M., A.H.B.); British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, Edinburgh, United Kingdom (M.D.B., K.P., A.T.V., N.J., M.R.D., D.E.N., R.A.M., A.H.B.); Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, The University of Glasgow, United Kingdom (G.H.); and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Robert A McDonald
- From BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (M.D.B., R.D., L.D., R.M., R.G., P.W., N.S., M.W.N., R.A.M., A.H.B.); British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, Edinburgh, United Kingdom (M.D.B., K.P., A.T.V., N.J., M.R.D., D.E.N., R.A.M., A.H.B.); Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, The University of Glasgow, United Kingdom (G.H.); and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.)
| | - Andrew H Baker
- From BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (M.D.B., R.D., L.D., R.M., R.G., P.W., N.S., M.W.N., R.A.M., A.H.B.); British Heart Foundation/University of Edinburgh Centre for Cardiovascular Science, Edinburgh, United Kingdom (M.D.B., K.P., A.T.V., N.J., M.R.D., D.E.N., R.A.M., A.H.B.); Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, The University of Glasgow, United Kingdom (G.H.); and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.M.M.).
| |
Collapse
|