1
|
Enteshari-Moghadam A, Fouladi N, Pordel S, Jeddi F, Asghariazar V, Eterafi M, Safarzadeh E. Evaluation of the miRNA-126 and VCAM-1 in scleroderma patients and its association with clinical characteristics. Am J Med Sci 2024:S0002-9629(24)01474-5. [PMID: 39326739 DOI: 10.1016/j.amjms.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 08/30/2024] [Accepted: 09/19/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND Systemic sclerosis (SSc) has the highest level of mortality and disability among all rheumatological diseases. Being heterogenous leads to no predictable method for clinical courses. The aim of this study was to evaluate the levels of miRNA-126 and soluble VCAM-1 protein markers in patients with SSc, and to examine the assossiation of their levels with the severity of clinical and paraclinical parameters in patients with SSc. METHOD In current study tweny six patients with SSc along with twenty-three SSc-free controls were recruited. Enzyme-linked immunosorbent assay (ELISA) was performed to measure the VCAM-1 protein. MiRNA-126 amounts in serum were detected by quantitative real-time polymerase chain reaction (PCR). RESULT SSc patients' average age was 45.42 years and control group 49.85. The mean±SD for circulating miR-126 levels were significantly lower in SSc patients compared with healthy donors (p = 0.02), 0.48 ± 0.72 vs 1.11 ± 0.61 respectively. A significant difference was also observed in the serum level of miRNA-126 in SSc patients who suffer from pulmonary artery hypertension (P = 0.03) and pulmonary fibrosis (P = 0.04). In contrast, analysis of the serum VCAM-1 levels in the study groups uncovered a significant increase in SSc patients (5.92 ± 3.52 µg/ml) compared to control group (2.62 ± 1.2 µg/ml) (P value < 0.001). CONCLUSION Significant change in circulating levels of miR-126 and VCAM-1 in the SSc patients supporting its role in the pathogenesis of the disease. It could also proposed potential role as a predictor of pulmonary complications for miRNA-126.
Collapse
Affiliation(s)
- Afsaneh Enteshari-Moghadam
- Department of Internal Medicine, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Nasrin Fouladi
- School of Medicine and Allied Medical Sciences, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Shohreh Pordel
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Farhad Jeddi
- Department of Genetics and Pathology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Vahid Asghariazar
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Majid Eterafi
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Elham Safarzadeh
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran; Department of Microbiology, Parasitology and Immunology, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
2
|
Gareev I, Beylerli O, Zhao B. MiRNAs as potential therapeutic targets and biomarkers for non-traumatic intracerebral hemorrhage. Biomark Res 2024; 12:17. [PMID: 38308370 PMCID: PMC10835919 DOI: 10.1186/s40364-024-00568-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 01/20/2024] [Indexed: 02/04/2024] Open
Abstract
Non-traumatic intracerebral hemorrhage (ICH) is the most common type of hemorrhagic stroke, most often occurring between the ages of 45 and 60. Hypertension is most often the cause of ICH. Less often, atherosclerosis, blood diseases, inflammatory changes in cerebral vessels, intoxication, vitamin deficiencies, and other reasons cause hemorrhages. Cerebral hemorrhage can occur by diapedesis or as a result of a ruptured vessel. This very dangerous disease is difficult to treat, requires surgery and can lead to disability or death. MicroRNAs (miRNAs) are a class of non-coding RNAs (about 18-22 nucleotides) that are involved in a variety of biological processes including cell differentiation, proliferation, apoptosis, etc., through gene repression. A growing number of studies have demonstrated miRNAs deregulation in various cardiovascular diseases, including ICH. In addition, given that computed tomography (CT) and/or magnetic resonance imaging (MRI) are either not available or do not show clear signs of possible vessel rupture, accurate and reliable analysis of circulating miRNAs in biological fluids can help in early diagnosis for prevention of ICH and prognosis patient outcome after hemorrhage. In this review, we highlight the up-to-date findings on the deregulated miRNAs in ICH, and the potential use of miRNAs in clinical settings, such as therapeutic targets and non-invasive diagnostic/prognostic biomarker tools.
Collapse
Affiliation(s)
- Ilgiz Gareev
- Bashkir State Medical University, Ufa, 450008, Russia
| | - Ozal Beylerli
- Bashkir State Medical University, Ufa, 450008, Russia
| | - Boxian Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, Harbin, 150001, China.
- Harbin Medical University No, 157, Baojian Road, Nangang District, Harbin, 150001, China.
| |
Collapse
|
3
|
Dahiya N, Kaur M, Singh V. Potential roles of circulatory microRNAs in the onset and progression of renal and cardiac diseases: a focussed review for clinicians. Acta Cardiol 2023; 78:863-877. [PMID: 37318070 DOI: 10.1080/00015385.2023.2221150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 05/14/2023] [Accepted: 05/30/2023] [Indexed: 06/16/2023]
Abstract
The signalling mechanisms involving the kidney and heart are a niche of networks causing pathological conditions inducing inflammation, reactive oxidative species, cell apoptosis, and organ dysfunction during the onset of clinical complications. The clinical manifestation of the kidney and heart depends on various biochemical processes that influence organ dysfunction coexistence through circulatory networks, which hold utmost importance. The cells of both organs also influence remote communication, and evidence states that it may be explicitly by circulatory small noncoding RNAs, i.e. microRNAs (miRNAs). Recent developments target miRNAs as marker panels for disease diagnosis and prognosis. Circulatory miRNAs expressed in renal and cardiac disease can reveal relevant information about the niche of networks and gene transcription and regulated networks. In this review, we discuss the pertinent roles of identified circulatory miRNAs regulating signal transduction pathways critical in the onset of renal and cardiac disease, which can hold promising future targets for clinical diagnostic and prognostic purposes.
Collapse
Affiliation(s)
- Neha Dahiya
- Centre for Life Sciences, Chitkara School of Health Sciences, Chitkara University, Punjab, India
| | - Manpreet Kaur
- Centre for Life Sciences, Chitkara School of Health Sciences, Chitkara University, Punjab, India
| | - Varsha Singh
- Centre for Life Sciences, Chitkara School of Health Sciences, Chitkara University, Punjab, India
| |
Collapse
|
4
|
Li Y, Xue JY, Chen S, Wang C, Sun P, Fu S, Li Y, Zhao P, Tian J, Du GQ. LncRNA PVT1 is a novel mediator promoting the angiogenesis response associated with collateral artery formation. Int J Biochem Cell Biol 2022; 151:106294. [PMID: 36041701 DOI: 10.1016/j.biocel.2022.106294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/21/2022] [Accepted: 08/25/2022] [Indexed: 10/15/2022]
Abstract
AIMS Angiogenesis plays a key role in coronary collateral circulation (CCC), the compensatory formation of new blood vessels during chronic total coronary occlusion. This study aimed to determine whether plasmacytoma variant translocation 1 (PVT1), a long non-coding (lnc) RNA involved in tumor angiogenesis, plays a role in regulating angiogenesis during chronic coronary ischemia. MAIN METHODS Patients with coronary artery disease, and ≥90% stenosis, were examined and divided into "Good" and "Poor" CCC groups based on Rentrop Cohen classification. RNA samples were obtained from all patients, as well as from oxygen and glucose-deprived (OGD) HUVECs. PVT1, miR-15b-5p and AKT3 levels were measured with RT-qPCR or Western blot, while HUVEC migration and angiogenesis were detected by, respectively, wound-healing and tube formation assays. Luciferase reporter assay confirmed direct PVT1-miR-15b-5p binding. KEY FINDINGS Increased PVT1 was found in "Good CCC" patient plasma, along with being highly expressed among OGD HUVECs; PVT1 knockdown reduced HUVEC migration, tube formation, and pro-angiogenic factor expression. Conversely, OGD HUVECs had downregulated miR-15b-5p, and miR-15b-5p overexpression significantly depressed their angiogenic capabilities. These PVT1 knockdown- or miR-15b-5p overexpression-associated reductions in angiogenic effects were reversed by AKT3 overexpression. In vivo, neovascularization and functioning in both ischemic mice hind-limbs and infarcted myocardium injected with ADV-sh-PVT1 were reduced, which were ameliorated by concurrent antagomiR-15b-5p injections. SIGNIFICANCE Circulating PVT1 may serve as a useful biomarker to distinguish between good versus poor CCC, as it is involved in orchestrating angiogenesis via the miR-15b-5p-AKT3 axis; it thus has potential as a target for treating ischemic disease.
Collapse
Affiliation(s)
- You Li
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China
| | - Jing-Yi Xue
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Department of Cardiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Shuang Chen
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China
| | - Chao Wang
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China
| | - Ping Sun
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China
| | - Shuai Fu
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China
| | - Yitong Li
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Peng Zhao
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China
| | - Jiawei Tian
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Guo-Qing Du
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China; Department of Ultrasound, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.
| |
Collapse
|
5
|
Fan YY, Liu CH, Wu AL, Chen HC, Hsueh YJ, Chen KJ, Lai CC, Huang CY, Wu WC. MicroRNA-126 inhibits pathological retinal neovascularization via suppressing vascular endothelial growth factor expression in a rat model of retinopathy of prematurity. Eur J Pharmacol 2021; 900:174035. [PMID: 33727052 DOI: 10.1016/j.ejphar.2021.174035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 03/06/2021] [Accepted: 03/11/2021] [Indexed: 01/11/2023]
Abstract
Vascular endothelial growth factor (VEGF) is the principal growth factor responsible for the retinal neovascularization in the pathogenesis of retinopathy of prematurity (ROP). Current therapies for ROP include laser ablation and intravitreal anti-VEGF injection. However, these treatments either destroy the peripheral retina or associate with problems of persistent peripheral avascular retina or later recurrence of ROP. In the present study we investigated a new therapeutic approach by exploring the potential role of a specific microRNA, miR-126, in regulating VEGFA expression and retinal neovascularization in a rat oxygen-induced retinopathy (OIR) model. We demonstrated that miR-126 mimic and plasmid effectively suppresses VEGFA mRNA expression in both human and rat retinal pigment epithelium cell lines, quantified with qRT-PCR. Animal experiments on rat OIR model revealed that intravitreal injection of miR-126 plasmid efficiently downregulated VEGFA expression in the intraocular fluid and retinal tissues measured by ELISA, and significantly suppressed retinal neovascularization, which was confirmed by calculating sizes of neovascularization areas on fluorescence microscopic images of flat mounted retina stained with Alexa Fluor 594-conjugated isolectin B4 to visualize blood vessels. Together, these results showed that intravitreal injection of miR-126 plasmid could inhibit retinal neovascularization by down-regulating VEGFA expression, suggesting a potential therapeutic effect for ROP.
Collapse
Affiliation(s)
- Yuan-Yao Fan
- Department of Ophthalmology, Chang Gung Memorial Hospital, Taoyuan, Taiwan; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chi-Hsien Liu
- Graduate Institute of Biochemical and Biomedical Engineering, Chang Gung University, Taoyuan, Taiwan
| | - An-Lun Wu
- Department of Ophthalmology, Mackay Memorial Hospital, Hsinchu, Taiwan
| | - Hung-Chi Chen
- Department of Ophthalmology, Chang Gung Memorial Hospital, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan; Limbal Stem Cell Laboratory, Department of Ophthalmology, Chang Gung Memorial Hospital, Taoyuan, Taiwan; Center for Tissue Engineering, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yi-Jen Hsueh
- Limbal Stem Cell Laboratory, Department of Ophthalmology, Chang Gung Memorial Hospital, Taoyuan, Taiwan; Center for Tissue Engineering, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Kuan-Jen Chen
- Department of Ophthalmology, Chang Gung Memorial Hospital, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chi-Chun Lai
- Department of Ophthalmology, Chang Gung Memorial Hospital, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chung-Ying Huang
- Department of Ophthalmology, Chang Gung Memorial Hospital, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Wei-Chi Wu
- Department of Ophthalmology, Chang Gung Memorial Hospital, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
6
|
Zhao C, Zong Z, Zhu Q, Wang Y, Li X, Zhang C, Ma C, Xue Y. The lncRNA MALAT1 participates in regulating coronary slow flow endothelial dysfunction through the miR-181b-5p-MEF2A-ET-1 axis. Vascul Pharmacol 2021; 138:106841. [PMID: 33545365 DOI: 10.1016/j.vph.2021.106841] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/30/2020] [Accepted: 01/29/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND Coronary slow flow (CSF) refers to coronary arteries with no obvious stenosis but have slow coronary flow without effective treatment. The main cause of CSF is endothelial dysfunction. The long non-coding RNA (lncRNA) MALAT1 is involved in regulating endothelial dysfunction, but its role in CSF endothelial dysfunction is still unclear. METHODS We included 41 CSF patients and 37 controls in the study, who all underwent coronary angiography, echocardiography, and brachial artery flow-mediated dilatation (FMD) examination. Human umbilical vein endothelial cells (HUVECs) stimulated by oxygen-glucose deprivation were used as CSF-induced HUVECs. Plasma endothelin-1 (ET-1) concentrations were determined by enzyme-linked immunosorbent assay (ELISA). The expression levels of MALAT1, miR-181b-5p, myocyte enhancer factor 2A (MEF2A), and ET-1 were measured by qRT-PCR or western blotting. Cell proliferation was determined by 5-ethynyl-2'-deoxyuridine (EdU) and Cell Counting Kit-8 (CCK-8) assays. Apoptosis was examined by flow cytometry. The relationship between miR-181b-5p and MALAT1 or MEF2A was verified by dual-luciferase reporter assay. MEF2A binding directly to the ET-1 promoter region was verified via chromatin immunoprecipitation (ChIP) assay. RESULTS MALAT1 and ET-1 were increased, and miR-181b-5p was decreased in the peripheral blood of the CSF patients, and could be used as predictors of CSF. In the CSF-induced HUVECs, MALAT1 was highly expressed, and MALAT1 knockdown improved endothelial function. In contrast, miR-181b-5p was downregulated in the CSF-induced HUVECs, and miR-181b-5p overexpression improved endothelial function. While MEF2A was highly enriched in CSF-induced HUVECs, MEF2A knockdown reduced ET-1 and increased the endothelial function of CSF-induced HUVECs as a transcriptional regulator of ET-1. MALAT1 modulated MEF2A expression positively by sponging miR-181b-5p. CONCLUSIONS Endothelial function is reduced in CSF. MALAT1 participates in regulating CSF endothelial dysfunction through the miR-181b-5p-MEF2A-ET-1 axis, and could provide a new target for CSF treatment.
Collapse
Affiliation(s)
- Cuiting Zhao
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, China
| | - Zhihong Zong
- Teaching Center for Basic Medical Experiment, China Medical University, Shenyang, China
| | - Qing Zhu
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, China
| | - Yonghuai Wang
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, China
| | - Xinxin Li
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, China
| | - Chenghong Zhang
- Teaching Center for Basic Medical Experiment, China Medical University, Shenyang, China
| | - Chunyan Ma
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, China.
| | - Yixue Xue
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, China; Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, China; Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, China
| |
Collapse
|
7
|
Na Nakorn P, Pannengpetch S, Isarankura-Na-Ayudhya P, Thippakorn C, Lawung R, Sathirapongsasuti N, Kitiyakara C, Sritara P, Vathesatogkit P, Isarankura-Na-Ayudhya C. Roles of kininogen-1, basement membrane specific heparan sulfate proteoglycan core protein, and roundabout homolog 4 as potential urinary protein biomarkers in diabetic nephropathy. EXCLI JOURNAL 2020; 19:872-891. [PMID: 32665774 PMCID: PMC7355151 DOI: 10.17179/excli2020-1396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 06/15/2020] [Indexed: 12/24/2022]
Abstract
Diabetic nephropathy, a major complication of diabetes mellitus (DM), is increasing worldwide and the large majority of patients have type 2 DM. Microalbuminuria has been used as a diagnostic marker of diabetic nephropathy. But owing to its insufficient sensitivity and specificity, other biomarkers are being sought. In addition, the pathophysiology of diabetic nephropathy is not fully understood and declines in renal function occur even without microalbuminuria. In this study, we investigated urinary proteins from three study groups (controls, and type 2 diabetic subjects with or without microalbuminuria). Non-targeted label-free Nano-LC QTOF analysis was conducted to discover underlying mechanisms and protein networks, and targeted label-free Nano-LC QTOF with SWATH was performed to qualify discovered protein candidates. Twenty-eight proteins were identified as candidates and functionally analyzed via String DB, gene ontology and pathway analysis. Four predictive mechanisms were analyzed: i) response to stimulus, ii) platelet activation, signaling and aggregation, iii) ECM-receptor interaction, and iv) angiogenesis. These mechanisms can provoke kidney dysfunction in type 2 diabetic patients via endothelial cell damage and glomerulus structural alteration. Based on these analyses, three proteins (kininogen-1, basement membrane-specific heparan sulfate proteoglycan core protein, and roundabout homolog 4) were proposed for further study as potential biomarkers. Our findings provide insights that may improve methods for both prevention and diagnosis of diabetic nephropathy.
Collapse
Affiliation(s)
- Piyada Na Nakorn
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Supitcha Pannengpetch
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Nakornpathom, Thailand
| | | | - Chadinee Thippakorn
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Nakornpathom, Thailand
| | - Ratana Lawung
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Nuankanya Sathirapongsasuti
- Section for Translational Medicine, Research Center, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Chagriya Kitiyakara
- Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Piyamitr Sritara
- Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Prin Vathesatogkit
- Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | | |
Collapse
|
8
|
Intravenous Administration of Allogenic Cell-Derived Microvesicles of Healthy Origins Defend Against Atherosclerotic Cardiovascular Disease Development by a Direct Action on Endothelial Progenitor Cells. Cells 2020; 9:cells9020423. [PMID: 32059493 PMCID: PMC7072151 DOI: 10.3390/cells9020423] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 02/03/2020] [Accepted: 02/05/2020] [Indexed: 02/06/2023] Open
Abstract
Atherosclerosis and cardiovascular disease development is the outcome of intermediate processes where endothelial dysfunction and vascular inflammation are main protagonists. Cell-derived microvesicles (MVs), endothelial progenitor cells (EPCs), and circulating microRNAs (miRNAs) are known as biomarkers and potential regulators for atherosclerotic vascular disease, but their role in the complexity of the inflammatory process and in the mechanism of vascular restoration is far from clear. We aimed to evaluate the biological activity and functional role of MVs, in particular of the EPCs-derived MVs (MVEs), of healthy origins in reducing atherosclerotic vascular disease development. The experiments were performed on hamsters divided into the following groups: simultaneously hypertensive–hyperlipidemic (HH group) by combining two feeding conditions for 4 months; HH with retro-orbital sinus injection containing 1 × 105 MVs or MVEs from control hamsters, one dose per month for 4 months of HH diet, to prevent atherosclerosis (HH-MVs or HH-MVEs group); and controls (C group), age-matched normal healthy animals. We found that circulating MV and MVE transplantation of healthy origins significantly reduces atherosclerosis development via (1) the mitigation of dyslipidemia, hypertension, and circulating EPC/cytokine/chemokine levels and (2) the structural and functional remodeling of arterial and left ventricular walls. We also demonstrated that (1) circulating MVs contain miRNAs; this was demonstrated by validating MVs and MVEs as transporters of Ago2-miRNA, Stau1-miRNA, and Stau2-miRNA complexes and (2) MV and MVE administration significantly protect against atherosclerotic cardiovascular disease via transfer of miR-223, miR-21, miR-126, and miR-146a to circulating late EPCs. It should be mentioned that the favorable effects of MVEs were greater than those of MVs. Our findings suggest that allogenic MV and MVE administration of healthy origins could counteract HH diet-induced detrimental effects by biologically active miR-10a, miR-21, miR-126, and miR-146a transfer to circulating EPCs, mediating their vascular repair function in atherosclerosis processes.
Collapse
|
9
|
Liu CH, Huang S, Britton WR, Chen J. MicroRNAs in Vascular Eye Diseases. Int J Mol Sci 2020; 21:ijms21020649. [PMID: 31963809 PMCID: PMC7014392 DOI: 10.3390/ijms21020649] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 01/16/2020] [Indexed: 12/12/2022] Open
Abstract
Since the discovery of the first microRNA (miRNA) decades ago, studies of miRNA biology have expanded in many biomedical research fields, including eye research. The critical roles of miRNAs in normal development and diseases have made miRNAs useful biomarkers or molecular targets for potential therapeutics. In the eye, ocular neovascularization (NV) is a leading cause of blindness in multiple vascular eye diseases. Current anti-angiogenic therapies, such as anti-vascular endothelial growth factor (VEGF) treatment, have their limitations, indicating the need for investigating new targets. Recent studies established the roles of various miRNAs in the regulation of pathological ocular NV, suggesting miRNAs as both biomarkers and therapeutic targets in vascular eye diseases. This review summarizes the biogenesis of miRNAs, and their functions in the normal development and diseases of the eye, with a focus on clinical and experimental retinopathies in both human and animal models. Discovery of novel targets involving miRNAs in vascular eye diseases will provide insights for developing new treatments to counter ocular NV.
Collapse
Affiliation(s)
| | | | | | - Jing Chen
- Correspondence: ; Tel.: +1-617-919-2525
| |
Collapse
|
10
|
SUR2B/Kir6.1 channel openers correct endothelial dysfunction in chronic heart failure via the miR-1-3p/ET-1 pathway. Biomed Pharmacother 2018; 110:431-439. [PMID: 30530045 DOI: 10.1016/j.biopha.2018.11.135] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/26/2018] [Accepted: 11/27/2018] [Indexed: 01/05/2023] Open
Abstract
The SUR2B/Kir6.1 channel openers iptakalim and natakalim reverse cardiac remodeling and ameliorate endothelial dysfunction by re-establishing the balance between the nitric oxide and endothelin systems. In this study, we investigated the microRNAs (miRs) involved in the molecular mechanisms of SUR2B/Kir6.1 channel opening in chronic heart failure. Both iptakalim and natakalim significantly upregulated the expression of miR-1-3p, suggesting that this miR is closely associated with the therapeutic effects against chronic heart failure. Bioinformatic analysis showed that many of the 183 target genes of miR-1-3p are involved in cardiovascular diseases, suggesting that miR-1-3p plays a vital role in such diseases and vascular remodeling. Target gene prediction showed that miR-1-3p combines with the 3' untranslated region (UTR) of endothelin-1 (ET-1) mRNA. Iptakalim and natakalim upregulated miR-1-3p expression and downregulated ET-1 mRNA expression in vitro. The dual luciferase assay confirmed that there is a complementary binding sequence between miR-1-3p and the 3' UTR 158-165 sequence of ET-1 mRNA. To verify the effect of miR-1-3p on ET-1, lentiviral vectors overexpressing or inhibiting miR-1-3p were constructed for the transduction of rat primary cardiac microvascular endothelial cells. The results showed that natakalim enhanced the miR-1-3p level. miR-1-3p overexpression downregulated the expression of ET-1, whereas miR-1-3p inhibition had the opposite effect. Therefore, we verified that SUR2B/Kir6.1 channel openers could correct endothelial imbalance and ameliorate chronic heart failure through the miR-1-3p/ET-1 pathway in endothelial cells. Our study provides comprehensive insights into the molecular mechanisms behind the SUR2B/Kir6.1 channel's activity against chronic heart failure.
Collapse
|
11
|
Affiliation(s)
- Mark W Majesky
- From the Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, and Departments of Pediatrics and Pathology, University of Washington.
| |
Collapse
|
12
|
Thanikachalam PV, Ramamurthy S, Wong ZW, Koo BJ, Wong JY, Abdullah MF, Chin YH, Chia CH, Tan JY, Neo WT, Tan BS, Khan WF, Kesharwani P. Current attempts to implement microRNA-based diagnostics and therapy in cardiovascular and metabolic disease: a promising future. Drug Discov Today 2018; 23:460-480. [DOI: 10.1016/j.drudis.2017.10.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 10/09/2017] [Accepted: 10/23/2017] [Indexed: 12/12/2022]
|
13
|
Zhang Y, Cai S, Jia Y, Qi C, Sun J, Zhang H, Wang F, Cao Y, Li X. Decoding Noncoding RNAs: Role of MicroRNAs and Long Noncoding RNAs in Ocular Neovascularization. Am J Cancer Res 2017; 7:3155-3167. [PMID: 28839470 PMCID: PMC5566112 DOI: 10.7150/thno.19646] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 06/15/2017] [Indexed: 12/11/2022] Open
Abstract
Ocular neovascularization is a pathological sequel of multiple eye diseases. Based on the anatomical site into which the abnormal neovessels grow, ocular neovascularization can be categorized into corneal neovascularization, choroidal neovascularization, and retinal neovascularization. Each category is intractable, and may lead to blindness if not appropriately treated. However, the current therapeutic modalities, including laser photocoagulation, vitrectomy surgery, and anti-VEGF drugs, raise concerns due to limited efficacy, damage on retinal parenchyma and vasculature, and the patients' unresponsiveness to the treatments. Therefore, the in-depth study on pathogenesis of and the search for novel therapeutic targets to the ocular neovascularization are needed. During the last 10 years or so, a large number of literatures have emerged indicating a critical role of noncoding RNAs, particularly microRNAs (miRNAs) and long noncoding RNAs (lncRNAs), in the pathogenesis and regulation of the ocular neovascularization. This review summarizes the current understanding of the biosynthesis and functions of the miRNAs and lncRNAs, the regulation of the miRNAs and lncRNAs in neovascular eye diseases, as well as the roles of these noncoding RNAs in the disease models of ocular neovascularization, in the hope that it could provide clues for the pathogenesis of and molecular targets to the ocular neovascularization.
Collapse
|
14
|
Liu J, Wang Y, Cui J, Sun M, Pu Z, Wang C, Du W, Liu X, Wu J, Hou J, Zhang S, Yu B. miR199a-3p regulates P53 by targeting CABLES1 in mouse cardiac c-kit + cells to promote proliferation and inhibit apoptosis through a negative feedback loop. Stem Cell Res Ther 2017; 8:127. [PMID: 28583208 PMCID: PMC5460483 DOI: 10.1186/s13287-017-0515-4] [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: 07/27/2016] [Revised: 02/09/2017] [Accepted: 02/21/2017] [Indexed: 01/08/2023] Open
Abstract
Background MicroRNAs (miRNAs) have emerged as crucial factors that regulate proliferation and apoptosis of cardiac c-kit+ cells. Although much is known about their role in maintaining cardiac c-kit+ cell pluripotency, the mechanisms by which they affect cell fate decisions that are an essential part of the repair of heart failure remain poorly understood. Methods Cardiac c-kit+ cells were obtained from Balb/c mice and cultured in vitro. Lentiviral vectors of miR199a-3p, its corresponding anti-miRNA, or short hairpin RNA against Cables1 were transfected into cells. The proliferation of cardiac c-kit+ cells was evaluated using EdU and flow cytometry. Furthermore, we examined cell apoptosis by flow cytometry under treatment with 200nM angiotensin II for 48 h. The levels of miR199a-3p and Cables1 mRNA were measured by quantitative real-time polymerase chain reaction (qRT-PCR). Western blot was performed to examine the expression of Cables1 and P53 proteins. Results We demonstrated a significantly decreased expression of miR199a-3p in heart failure samples compared with healthy donors. Meanwhile, we identified miR199a-3p as a proliferation- and apoptosis-associated regulator impacted through Cdk5 and Abl enzyme substrate 1 (CABLES1) targeting, and also attributed their repression to P53 protein expression. We further demonstrated that P53 induced miR199a-3p expression and, in turn, miR199-3p decreased P53 activity. Conclusion Collectively, our findings uncover one new mechanism by which P53 induced miR199a-3p expression and, in turn, miR199-3p decreased P53 activity. Therefore, miR199a-3p and P53 are coupled through CABLES1 and comprise a novel negative feedback loop that likely contributes to cardiac c-kit+ cell proliferation and apoptosis.
Collapse
Affiliation(s)
- Jingjin Liu
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China.,Key Laboratories of the Education Ministry for Myocardial Ischemia Mechanisms and Treatment, Harbin, Heilongjiang Province, China
| | - Yongshun Wang
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China
| | - Jinjin Cui
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China
| | - Meng Sun
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China.,Key Laboratories of the Education Ministry for Myocardial Ischemia Mechanisms and Treatment, Harbin, Heilongjiang Province, China
| | - Zhongyue Pu
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China.,Key Laboratories of the Education Ministry for Myocardial Ischemia Mechanisms and Treatment, Harbin, Heilongjiang Province, China
| | - Chao Wang
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China.,Key Laboratories of the Education Ministry for Myocardial Ischemia Mechanisms and Treatment, Harbin, Heilongjiang Province, China
| | - Wenjuan Du
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China.,Key Laboratories of the Education Ministry for Myocardial Ischemia Mechanisms and Treatment, Harbin, Heilongjiang Province, China
| | - Xinxin Liu
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China.,Key Laboratories of the Education Ministry for Myocardial Ischemia Mechanisms and Treatment, Harbin, Heilongjiang Province, China
| | - Jian Wu
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China
| | - Jingbo Hou
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China
| | - Shuo Zhang
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China
| | - Bo Yu
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China. .,Key Laboratories of the Education Ministry for Myocardial Ischemia Mechanisms and Treatment, Harbin, Heilongjiang Province, China.
| |
Collapse
|
15
|
Juni RP, Abreu RC, da Costa Martins PA. Regulation of microvascularization in heart failure - an endothelial cell, non-coding RNAs and exosome liaison. Noncoding RNA Res 2017; 2:45-55. [PMID: 30159420 PMCID: PMC6096416 DOI: 10.1016/j.ncrna.2017.01.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Accepted: 01/26/2017] [Indexed: 12/22/2022] Open
Abstract
Heart failure is a complex syndrome involving various pathophysiological processes. An increasing body of evidence shows that the myocardial microvasculature is essential for the homeostasis state and that a decompensated heart is associated with microvascular dysfunction as a result of impaired endothelial angiogenic capacity. The intercellular communication between endothelial cells and cardiomyocytes through various signaling molecules, such as vascular endothelial growth factor, nitric oxide, and non-coding RNAs is an important determinant of cardiac microvascular function. Non-coding RNAs are transported from endothelial cells to cardiomyocytes, and vice versa, regulating microvascular properties and angiogenic processes in the heart. Small-exocytosed vesicles, called exosomes, which are secreted by both cell types, can mediate this intercellular communication. The purpose of this review is to highlight the contribution of the microvasculature to proper heart function maintenance by focusing on the interaction between cardiac endothelial cells and myocytes with a specific emphasis on non-coding RNAs (ncRNAs) in this form of cell-to-cell communication. Finally, the potential of ncRNAs as targets for angiogenesis therapy will also be discussed.
Collapse
Affiliation(s)
- Rio P. Juni
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Ricardo C. Abreu
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Paula A. da Costa Martins
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
16
|
Exercise Training and Epigenetic Regulation: Multilevel Modification and Regulation of Gene Expression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1000:281-322. [PMID: 29098627 DOI: 10.1007/978-981-10-4304-8_16] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Exercise training elicits acute and adaptive long term changes in human physiology that mediate the improvement of performance and health state. The responses are integrative and orchestrated by several mechanisms, as gene expression. Gene expression is essential to construct the adaptation of the biological system to exercise training, since there are molecular processes mediating oxidative and non-oxidative metabolism, angiogenesis, cardiac and skeletal myofiber hypertrophy, and other processes that leads to a greater physiological status. Epigenetic is the field that studies about gene expression changes heritable by meiosis and mitosis, by changes in chromatin and DNA conformation, but not in DNA sequence, that studies the regulation on gene expression that is independent of genotype. The field approaches mechanisms of DNA and chromatin conformational changes that inhibit or increase gene expression and determine tissue specific pattern. The three major studied epigenetic mechanisms are DNA methylation, Histone modification, and regulation of noncoding RNA-associated genes. This review elucidates these mechanisms, focusing on the relationship between them and their relationship with exercise training, physical performance and the enhancement of health status. On this chapter, we clarified the relationship of epigenetic modulations and their intimal relationship with acute and chronic effect of exercise training, concentrating our effort on skeletal muscle, heart and vascular responses, that are the most responsive systems against to exercise training and play crucial role on physical performance and improvement of health state.
Collapse
|
17
|
MicroRNA-126 suppresses inflammation in endothelial cells under hyperglycemic condition by targeting HMGB1. Vascul Pharmacol 2016; 88:48-55. [PMID: 27993686 DOI: 10.1016/j.vph.2016.12.002] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Revised: 12/05/2016] [Accepted: 12/11/2016] [Indexed: 12/13/2022]
Abstract
MicroRNA-126(miR-126) targets involved in inflammation need to be identified. In this study, we aim to investigate whether high-mobility group box 1(HMGB1), an inflammation-related gene, is the target of miR-126 in diabetic vascular endothelium. The diabetic apoE-/- mice model, a classical diabetic atherosclerosis model, was established. The aorta of diabetic apoE-/- mice showed decrease of miR-126 and elevation of HMGB1 and inflammation. Next, we employed several in vitro experiments to address the role of miRNA-126 on the regulation of HMGB1 in endothelial cells under hyperglycemic and inflammatory conditions. Manipulation of miRNA levels in human umbilical vein endothelial cells (HUVECs) was achieved by transfecting cells with miR-126 mimic and antagomir. Overexpression of miR-126 could decrease the expression of downstream components of HMGB1 including TNF-α, ROS, and NADPH oxidase activity in HUVECs under hyperglycemic condition. Nevertheless, such phenomenon was completely reversed by miR-126 antagomir. The expression of HMGB1 protein rather than HMGB1 mRNA was down-regulated after transfection with miR-126 mimic, which indicated the modulation of HMGB1 mediated by miR-126 was at the posttranslational level. Luciferase reporter assay confirmed the 3'-UTR of HMGB1 gene was a direct target of miR-126. Western blot analysis also indicated that overexpression of miR-126 contributed to the elevation of p-eNOS, eNOS and p-AKT expressions, respectively. In summary, our findings suggest that miR-126 may suppress inflammation and ROS production in endothelial cells treated by high glucose through modulating the expression of HMGB1. Our study provides a novel pathogenic link between dysregulated miRNA expression and inflammation in diabetic vascular endothelium.
Collapse
|
18
|
Maffei A, Di Mauro V, Catalucci D, Lembo G. MiR-153/Kv7.4: a novel molecular axis in the regulation of hypertension. Cardiovasc Res 2016; 112:530-531. [DOI: 10.1093/cvr/cvw208] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
|
19
|
Licholai S, Blaż M, Kapelak B, Sanak M. Unbiased Profile of MicroRNA Expression in Ascending Aortic Aneurysm Tissue Appoints Molecular Pathways Contributing to the Pathology. Ann Thorac Surg 2016; 102:1245-52. [PMID: 27234576 DOI: 10.1016/j.athoracsur.2016.03.061] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Revised: 01/19/2016] [Accepted: 03/16/2016] [Indexed: 11/28/2022]
Abstract
BACKGROUND Complex etiopathogenesis of ascending aortic aneurysm suggests contribution of epigenetic mechanisms in its development. Several studies appointed microRNAs (miRs) as essential epigenetic factors in various human diseases; however, little is known about their role in ascending aortic aneurysm. Therefore, the aim of this study was to perform unbiased molecular screening of miRs expression in aneurysmal tissue and establish their functions on a transcriptional level. METHODS Samples of ascending aortic tissue were obtained from 15 patients, and total RNA was isolated separately from aneurysmal and unaffected aortic tissue obtained from the same patient. Expression of the complete panel of human miRs was assessed by quantitative real-time polymerase chain reaction. Using bioinformatic tools, 13 genes were selected that were putatively regulated by overexpressed miRs. Expression level of transcripts were evaluated by quantitative real-time polymerase chain reaction and correlated with their targeting miRs. RESULTS Overexpression of 10 miRs distinguished aneurysmal tissue from the unchanged one. These miRs were involved in cell senescence (miR-191-5p), maintenance of vascular integrity (miR-126-3p and miR-374-5p), nitric oxide-dependent vascular relaxation (miR-21-5p), smooth muscle differentiation, and contractility (miR-145- 3p, miR-29c-3p, miR-133a-3p, miR-186-5p, miR-143-3p, and miR-24-3p), and correlated with abundance of its miR targets. CONCLUSIONS Altered expression of particular miRs selectively in the affected tissue indicate their role as factors that trigger pathways of aneurysmal transformation. Limited reparative properties due to overexpression of miR-191 may play a crucial role for aneurysm enlargement, whereas nitric oxide-dependent relaxation of vascular smooth muscle mediated by miR-21 offers an attractive explanation of the aneurysm's initiation, and is confirmed in experimental conditions.
Collapse
Affiliation(s)
- Sabina Licholai
- Division of Molecular Biology and Clinical Genetics, Department of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Michal Blaż
- Students' Research Group, Division of Molecular Biology and Clinical Genetics, Department of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Boguslaw Kapelak
- Department of Cardiovascular Surgery and Transplantology, Jagiellonian University, John Paul II Hospital in Krakow, Krakow, Poland
| | - Marek Sanak
- Division of Molecular Biology and Clinical Genetics, Department of Medicine, Jagiellonian University Medical College, Krakow, Poland.
| |
Collapse
|
20
|
Schumann J. Does plasma membrane lipid composition impact the miRNA-mediated regulation of vascular inflammation? Med Hypotheses 2016; 88:57-9. [PMID: 26880639 DOI: 10.1016/j.mehy.2016.01.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 01/08/2016] [Accepted: 01/19/2016] [Indexed: 11/27/2022]
Abstract
BACKGROUND Both PUFA and miRNAs are believed to be of importance in vascular diseases. On the one hand diverse nutrition societies recommend PUFA consumption to dampen inflammatory processes. On the other hand scientists intensify efforts to use miRNAs for diagnostics or therapy in context of vascular disorders. PRESENTATION OF THE HYPOTHESIS There might be is a causal link between the plasma membrane lipid composition and the miRNA expression of monocytes and endothelial cells. PUFA enrichment of cells may affect the type and the amount of particular miRNAs produced. In this way dietary fatty acids are supposed to impact the miRNA-mediated regulation of vascular inflammatory processes. PROPOSED EXPERIMENTAL SETTING TO TEST THE HYPOTHESIS PUFA-supplemented monocytes and endothelial cells are analyzed with respect to membrane fatty acid patterns, typical markers of vascular inflammation and miRNA expression. Experiments are performed both for undifferentiated/unstimulated as well as for differentiated/stimulated cells. Verification of identified miRNA targets is performed by means of mimics/antagomirs. IMPLICATIONS OF THE HYPOTHESIS Innovative mechanism of action, which could point the way to a new understanding of the PUFA-mediated modulation of cellular signal transduction. If confirmed experimentally, it might stimulate vascular inflammation research and immunologic lipid science, hence, acting as source of inspiration for future therapeutic interventions in vascular diseases.
Collapse
Affiliation(s)
- Julia Schumann
- Clinic for Anesthesiology and Surgical Intensive Care, University Hospital Halle (Saale), Franzosenweg 1a, 06112 Halle (Saale), Germany.
| |
Collapse
|
21
|
Bretschneider M, Busch B, Mueller D, Nolze A, Schreier B, Gekle M, Grossmann C. Activated mineralocorticoid receptor regulates micro-RNA-29b in vascular smooth muscle cells. FASEB J 2016; 30:1610-22. [PMID: 26728178 DOI: 10.1096/fj.15-271254] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 12/11/2015] [Indexed: 12/17/2022]
Abstract
Inappropriately activated mineralocorticoid receptor (MR) is a risk factor for vascular remodeling with unclear molecular mechanism. Recent findings suggest that post-transcriptional regulation by micro-RNAs (miRs) may be involved. Our aim was to search for MR-dependent miRs in vascular smooth muscle cells (VSMCs) and to explore the underlying molecular mechanism and the pathologic relevance. We detected that aldosteroneviathe MR reduces miR-29bin vivoin murine aorta and in human primary and cultured VSMCs (ED50= 0.07 nM) but not in endothelial cells [quantitative PCR (qPCR), luciferase assays]. This effect was mediated by an increased decay of miR-29b in the cytoplasm with unchanged miR-29 family member or primary-miR levels. Decreased miR-29b led to an increase in extracellular matrix measured by ELISA and qPCR and enhanced VSMC migration in single cell-tracking experiments. Additionally, cell proliferation and the apoptosis/necrosis ratio (caspase/lactate dehydrogenase assay) was modulated by miR-29b. Enhanced VSMC migration by aldosterone required miR-29b regulation. Control experiments were performed with scrambled RNA and empty plasmids, by comparing aldosterone-stimulated with vehicle-incubated cells. Overall, our findings provide novel insights into the molecular mechanism of aldosterone-mediated vascular pathogenesis by identifying miR-29b as a pathophysiologic relevant target of activated MR in VSMCs and by highlighting the importance of miR processing for miR regulation.-Bretschneider, M., Busch, B., Mueller, D., Nolze, A., Schreier, B., Gekle, M., Grossmann, C. Activated mineralocorticoid receptor regulates micro-RNA-29b in vascular smooth muscle cells.
Collapse
Affiliation(s)
- Maria Bretschneider
- *Julius Bernstein Institute of Physiology and Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Bianca Busch
- *Julius Bernstein Institute of Physiology and Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Daniel Mueller
- *Julius Bernstein Institute of Physiology and Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Alexander Nolze
- *Julius Bernstein Institute of Physiology and Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Barbara Schreier
- *Julius Bernstein Institute of Physiology and Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Michael Gekle
- *Julius Bernstein Institute of Physiology and Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Claudia Grossmann
- *Julius Bernstein Institute of Physiology and Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| |
Collapse
|
22
|
Role of MicroRNAs in Renin-Angiotensin-Aldosterone System-Mediated Cardiovascular Inflammation and Remodeling. Int J Inflam 2015; 2015:101527. [PMID: 26064773 PMCID: PMC4438140 DOI: 10.1155/2015/101527] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 04/18/2015] [Indexed: 12/27/2022] Open
Abstract
MicroRNAs are endogenous regulators of gene expression either by inhibiting translation or protein degradation. Recent studies indicate that microRNAs play a role in cardiovascular disease and renin-angiotensin-aldosterone system- (RAAS-) mediated cardiovascular inflammation, either as mediators or being targeted by RAAS pharmacological inhibitors. The exact role(s) of microRNAs in RAAS-mediated cardiovascular inflammation and remodeling is/are still in early stage of investigation. However, few microRNAs have been shown to play a role in RAAS signaling, particularly miR-155, miR-146a/b, miR-132/122, and miR-483-3p. Identification of specific microRNAs and their targets and elucidating microRNA-regulated mechanisms associated RAS-mediated cardiovascular inflammation and remodeling might lead to the development of novel pharmacological strategies to target RAAS-mediated vascular pathologies. This paper reviews microRNAs role in inflammatory factors mediating cardiovascular inflammation and RAAS genes and the effect of RAAS pharmacological inhibition on microRNAs and the resolution of RAAS-mediated cardiovascular inflammation and remodeling. Also, this paper discusses the advances on microRNAs-based therapeutic approaches that may be important in targeting RAAS signaling.
Collapse
|
23
|
|
24
|
Neves VJD, Fernandes T, Roque FR, Soci UPR, Melo SFS, Oliveira EMD. Exercise training in hypertension: Role of microRNAs. World J Cardiol 2014; 6:713-727. [PMID: 25228951 PMCID: PMC4163701 DOI: 10.4330/wjc.v6.i8.713] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2013] [Revised: 03/25/2014] [Accepted: 05/29/2014] [Indexed: 02/06/2023] Open
Abstract
Hypertension is a complex disease that constitutes an important public health problem and demands many studies in order to understand the molecular mechanisms involving his pathophysiology. Therefore, an increasing number of studies have been conducted and new therapies are continually being discovered. In this context, exercise training has emerged as an important non-pharmacological therapy to treat hypertensive patients, minimizing the side effects of pharmacological therapies and frequently contributing to allow pharmacotherapy to be suspended. Several mechanisms have been associated with the pathogenesis of hypertension, such as hyperactivity of the sympathetic nervous system and renin-angiotensin aldosterone system, impaired endothelial nitric oxide production, increased oxygen-reactive species, vascular thickening and stiffening, cardiac hypertrophy, impaired angiogenesis, and sometimes genetic predisposition. With the advent of microRNAs (miRNAs), new insights have been added to the perspectives for the treatment of this disease, and exercise training has been shown to be able to modulate the miRNAs associated with it. Elucidation of the relationship between exercise training and miRNAs in the pathogenesis of hypertension is fundamental in order to understand how exercise modulates the cardiovascular system at genetic level. This can be promising even for the development of new drugs. This article is a review of how exercise training acts on hypertension by means of specific miRNAs in the heart, vascular system, and skeletal muscle.
Collapse
|
25
|
Batkai S, Thum T. Analytical approaches in microRNA therapeutics. J Chromatogr B Analyt Technol Biomed Life Sci 2014; 964:146-52. [DOI: 10.1016/j.jchromb.2014.03.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 03/17/2014] [Accepted: 03/19/2014] [Indexed: 02/06/2023]
|
26
|
Mikaelian I, Cameron M, Dalmas DA, Enerson BE, Gonzalez RJ, Guionaud S, Hoffmann PK, King NMP, Lawton MP, Scicchitano MS, Smith HW, Thomas RA, Weaver JL, Zabka TS. Nonclinical Safety Biomarkers of Drug-induced Vascular Injury. Toxicol Pathol 2014; 42:635-57. [DOI: 10.1177/0192623314525686] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Better biomarkers are needed to identify, characterize, and/or monitor drug-induced vascular injury (DIVI) in nonclinical species and patients. The Predictive Safety Testing Consortium (PSTC), a precompetitive collaboration of pharmaceutical companies and the U.S. Food and Drug Administration (FDA), formed the Vascular Injury Working Group (VIWG) to develop and qualify translatable biomarkers of DIVI. The VIWG focused its research on acute DIVI because early detection for clinical and nonclinical safety monitoring is desirable. The VIWG developed a strategy based on the premise that biomarkers of DIVI in rat would be translatable to humans due to the morphologic similarity of vascular injury between species regardless of mechanism. The histomorphologic lexicon for DIVI in rat defines degenerative and adaptive findings of the vascular endothelium and smooth muscles, and characterizes inflammatory components. We describe the mechanisms of these changes and their associations with candidate biomarkers for which advanced analytical method validation was completed. Further development is recommended for circulating microRNAs, endothelial microparticles, and imaging techniques. Recommendations for sample collection and processing, analytical methods, and confirmation of target localization using immunohistochemistry and in situ hybridization are described. The methods described are anticipated to aid in the identification and qualification of translational biomarkers for DIVI.
Collapse
Affiliation(s)
- Igor Mikaelian
- Hoffmann-La Roche Inc, Nutley, New Jersey, USA
- Abbvie, Worcester, Massachusetts, USA
| | | | | | | | - Raymond J. Gonzalez
- Merck Research Laboratories, Merck and Co, Inc, West Point, Pennsylvania, USA
| | - Silvia Guionaud
- Shire, Hampshire International Business Park, Basingstoke, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
Abstract
Lymphatic vasculature is increasingly recognized as an important factor both in the regulation of normal tissue homeostasis and immune response and in many diseases, such as inflammation, cancer, obesity, and hypertension. In the last few years, in addition to the central role of vascular endothelial growth factor (VEGF)-C/VEGF receptor-3 signaling in lymphangiogenesis, significant new insights were obtained about Notch, transforming growth factor β/bone morphogenetic protein, Ras, mitogen-activated protein kinase, phosphatidylinositol 3 kinase, and Ca2+/calcineurin signaling pathways in the control of growth and remodeling of lymphatic vessels. An emerging picture of lymphangiogenic signaling is complex and in many ways distinct from the regulation of angiogenesis. This complexity provides new challenges, but also new opportunities for selective therapeutic targeting of lymphatic vasculature.
Collapse
|
28
|
Agrawal S, Chaqour B. MicroRNA signature and function in retinal neovascularization. World J Biol Chem 2014; 5:1-11. [PMID: 24600510 PMCID: PMC3942538 DOI: 10.4331/wjbc.v5.i1.1] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 12/11/2013] [Accepted: 01/06/2014] [Indexed: 02/05/2023] Open
Abstract
Ischemic retinopathies are clinically well-defined chronic microvascular complications characterized by gradually progressive alterations in the retinal microvasculature and a compensatory aberrant neovascularization of the eye. The subsequent metabolic deficiencies result in structural and functional alterations in the retina which is highly susceptible to injurious stimuli such as diabetes, trauma, hyperoxia, inflammation, aging and dysplipidemia. Emerging evidence indicates that an effective therapy may require targeting multiple components of the angiogenic pathway. Conceptually, mircoRNA (miRNA)-based therapy provides the rationale basis for an effective antiangiogenic treatment. miRNAs are an evolutionarily conserved family of short RNAs, each regulating the expression of multiple protein-coding genes. The activity of specific miRNAs is important for vascular cell signaling and blood vessel formation and function. Recently, important progress has been made in mapping the miRNA-gene target network and miRNA-mediated gene expression control. Here we highlight the latest findings on angiogenic and antiangiogenic miRNAs and their targets as well as potential implications in ocular neovascular diseases. Emphasis is placed on how specific vascular-enriched miRNAs regulate cell responses to various cues by targeting several factors, receptors and/or signaling molecules in order to maintain either vascular function or dysfunction. Further improvement of our knowledge in not only miRNA specificity, turnover, and transport but also how miRNA sequences and functions can be altered will enhance the therapeutic utility of such molecules.
Collapse
|
29
|
Ye P, Liu J, He F, Xu W, Yao K. Hypoxia-induced deregulation of miR-126 and its regulative effect on VEGF and MMP-9 expression. Int J Med Sci 2014; 11:17-23. [PMID: 24396282 PMCID: PMC3880987 DOI: 10.7150/ijms.7329] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Accepted: 11/25/2013] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE miR-126, the miRNA considered to be specially expressed in endothelial cells and hematopoietic progenitor cells, is strongly associated with angiogenesis. The purpose is to evaluate the role of miR-126 in hypoxia-induced angiogenesis and the possible mechanisms. METHODS The expression of miR-126 was detected in hypoxia-treated RF/6A cells and diabetic retinas using real-time PCR. The miR-126 was up- or down-regulated by transfecting miR-126-mimics or inhibitors into RF/6A cells. Cell cycle analysis was performed using flow cytometry. The protein levels of vascular endothelial growth factor (VEGF) and matrix metalloproteinase-9 (MMP-9) were assessed by immunoblotting. RESULTS A significantly decreased expression of miR-126 was found in hypoxia-treated RF/6A cells in a time-dependent manner compared with normoxic condition. The expression of miR-126 was also reduced in the retina tissue of streptozotocin-induced diabetic rats. The expression of VEGF and MMP-9 proteins was increased in hypoxia-induced RF/6A cells. In the functional analysis, miR-126-mimic significantly reduced the percentage of RF/6A cells in S phases compared with the negative control under hypoxic conditions. Furthermore, the VEGF and MMP-9 protein levels were sharply decreased in hypoxia-induced RF/6A cells pretreated with miR-126-mimics and increased in the cells pretreated with miR-126-inhibitors. CONCLUSIONS miR-126 is down-regulated under hypoxic condition both in vitro and in vivo and may halt the hypoxia-induce neovascularization by suspending the cell cycle progression and inhibiting the expression of VEGF and MMP-9.
Collapse
Affiliation(s)
- Panpan Ye
- 1. Eye Center, Second Affiliated Hospital, School of Medicine, Zhejiang University; ; 2. Zhejiang Provincial Key Lab of Ophthalmology, China
| | - Jian Liu
- 1. Eye Center, Second Affiliated Hospital, School of Medicine, Zhejiang University; ; 2. Zhejiang Provincial Key Lab of Ophthalmology, China
| | - Fengying He
- 1. Eye Center, Second Affiliated Hospital, School of Medicine, Zhejiang University; ; 2. Zhejiang Provincial Key Lab of Ophthalmology, China
| | - Wen Xu
- 1. Eye Center, Second Affiliated Hospital, School of Medicine, Zhejiang University; ; 2. Zhejiang Provincial Key Lab of Ophthalmology, China
| | - Ke Yao
- 1. Eye Center, Second Affiliated Hospital, School of Medicine, Zhejiang University; ; 2. Zhejiang Provincial Key Lab of Ophthalmology, China
| |
Collapse
|
30
|
Doridot L, Miralles F, Barbaux S, Vaiman D. Trophoblasts, invasion, and microRNA. Front Genet 2013; 4:248. [PMID: 24312123 PMCID: PMC3836020 DOI: 10.3389/fgene.2013.00248] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 10/30/2013] [Indexed: 12/15/2022] Open
Abstract
MicroRNAs (miRNAs) have recently become essential actors in various fields of physiology and medicine, especially as easily accessible circulating biomarkers, or as modulators of cell differentiation. To this respect, terminal differentiation of trophoblasts (the characteristic cells of the placenta in Therian mammals) into syncytiotrophoblast, villous trophoblast, or extravillous trophoblast constitutes a good example of such a choice, where miRNAs have recently been shown to play an important role. The aim of this review is to provide a snapshot of what is known today in placentation mechanisms that are mediated by miRNA, under the angles of materno–fetal immune dialog regulation, trophoblast differentiation, and angiogenesis at the materno–fetal interface. Also, two aspects of regulation of these issues will be highlighted: the part played by oxygen concentration and the specific function of imprinted genes in the developing placenta.
Collapse
Affiliation(s)
- Ludivine Doridot
- Institut Cochin, INSERM U1016-CNRS UMR8104, Université Paris Descartes Paris, France
| | | | | | | |
Collapse
|
31
|
Role of miR-181 family in regulating vascular inflammation and immunity. Trends Cardiovasc Med 2013; 24:105-12. [PMID: 24183793 DOI: 10.1016/j.tcm.2013.09.002] [Citation(s) in RCA: 130] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 08/30/2013] [Accepted: 08/30/2013] [Indexed: 01/08/2023]
Abstract
The microRNA family, miR-181, plays diverse roles in regulating key aspects of cellular growth, development, and activation. Accumulating evidence supports a central role for the miR-181 family in vascular inflammation by controlling critical signaling pathways, such as downstream NF-κB signaling, and targets relevant to endothelial cell activation and immune cell homeostasis. This review examines the current knowledge of the miR-181 family's role in key cell types that critically control cardiovascular inflammation under pathological and physiological stimuli.
Collapse
|
32
|
Lorenzen JM, Batkai S, Thum T. Regulation of cardiac and renal ischemia-reperfusion injury by microRNAs. Free Radic Biol Med 2013; 64:78-84. [PMID: 23845966 DOI: 10.1016/j.freeradbiomed.2013.06.044] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 06/25/2013] [Accepted: 06/26/2013] [Indexed: 01/12/2023]
Abstract
Tissue damage caused by ischemia-reperfusion (I/R) injury represents a serious event, which often leads to deterioration or even loss of organ function. I/R injury is associated with transient tissue oxygen deprivation due to vessel occlusion and a subsequent reperfusion period following restoration of blood flow. Initial tissue damage inflicted by ischemia is aggravated in the reperfusion period through mechanisms such as burst of reactive oxygen and nitrogen species and inflammatory reactions. I/R injury occurs during surgical interventions, organ transplantation, diseases such as myocardial infarction, circulatory shock, and toxic insults. Recently, microRNAs have come into focus as powerful regulators of gene expression and potential diagnostic tools during I/R injury. These small noncoding ribonucleotides (~22 nucleotides in length) posttranscriptionally target mRNAs, culminating in suppression of protein synthesis or increase in mRNA degradation, thus fundamentally influencing organ function. This review highlights the latest developments regarding the role of microRNAs in cardiac and renal I/R injury.
Collapse
Affiliation(s)
- Johan M Lorenzen
- Institute of Molecular and Translational Therapeutic Strategies, Germany.
| | | | | |
Collapse
|
33
|
Wagner J, Riwanto M, Besler C, Knau A, Fichtlscherer S, Röxe T, Zeiher AM, Landmesser U, Dimmeler S. Characterization of Levels and Cellular Transfer of Circulating Lipoprotein-Bound MicroRNAs. Arterioscler Thromb Vasc Biol 2013; 33:1392-400. [DOI: 10.1161/atvbaha.112.300741] [Citation(s) in RCA: 252] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Jasmin Wagner
- From the Institute of Cardiovascular Regeneration (J.W., A.K., T.R., S.D.) and Division of Cardiology, Department of Medicine III (S.F., A.M.Z.), Centre for Molecular Medicine, Goethe-University Frankfurt, Frankfurt, Germany; and Cardiovascular Research, Institute of Physiology, University of Zurich, Zurich, Switzerland (M.R., C.B., U.L.)
| | - Meliana Riwanto
- From the Institute of Cardiovascular Regeneration (J.W., A.K., T.R., S.D.) and Division of Cardiology, Department of Medicine III (S.F., A.M.Z.), Centre for Molecular Medicine, Goethe-University Frankfurt, Frankfurt, Germany; and Cardiovascular Research, Institute of Physiology, University of Zurich, Zurich, Switzerland (M.R., C.B., U.L.)
| | - Christian Besler
- From the Institute of Cardiovascular Regeneration (J.W., A.K., T.R., S.D.) and Division of Cardiology, Department of Medicine III (S.F., A.M.Z.), Centre for Molecular Medicine, Goethe-University Frankfurt, Frankfurt, Germany; and Cardiovascular Research, Institute of Physiology, University of Zurich, Zurich, Switzerland (M.R., C.B., U.L.)
| | - Andrea Knau
- From the Institute of Cardiovascular Regeneration (J.W., A.K., T.R., S.D.) and Division of Cardiology, Department of Medicine III (S.F., A.M.Z.), Centre for Molecular Medicine, Goethe-University Frankfurt, Frankfurt, Germany; and Cardiovascular Research, Institute of Physiology, University of Zurich, Zurich, Switzerland (M.R., C.B., U.L.)
| | - Stephan Fichtlscherer
- From the Institute of Cardiovascular Regeneration (J.W., A.K., T.R., S.D.) and Division of Cardiology, Department of Medicine III (S.F., A.M.Z.), Centre for Molecular Medicine, Goethe-University Frankfurt, Frankfurt, Germany; and Cardiovascular Research, Institute of Physiology, University of Zurich, Zurich, Switzerland (M.R., C.B., U.L.)
| | - Tino Röxe
- From the Institute of Cardiovascular Regeneration (J.W., A.K., T.R., S.D.) and Division of Cardiology, Department of Medicine III (S.F., A.M.Z.), Centre for Molecular Medicine, Goethe-University Frankfurt, Frankfurt, Germany; and Cardiovascular Research, Institute of Physiology, University of Zurich, Zurich, Switzerland (M.R., C.B., U.L.)
| | - Andreas M. Zeiher
- From the Institute of Cardiovascular Regeneration (J.W., A.K., T.R., S.D.) and Division of Cardiology, Department of Medicine III (S.F., A.M.Z.), Centre for Molecular Medicine, Goethe-University Frankfurt, Frankfurt, Germany; and Cardiovascular Research, Institute of Physiology, University of Zurich, Zurich, Switzerland (M.R., C.B., U.L.)
| | - Ulf Landmesser
- From the Institute of Cardiovascular Regeneration (J.W., A.K., T.R., S.D.) and Division of Cardiology, Department of Medicine III (S.F., A.M.Z.), Centre for Molecular Medicine, Goethe-University Frankfurt, Frankfurt, Germany; and Cardiovascular Research, Institute of Physiology, University of Zurich, Zurich, Switzerland (M.R., C.B., U.L.)
| | - Stefanie Dimmeler
- From the Institute of Cardiovascular Regeneration (J.W., A.K., T.R., S.D.) and Division of Cardiology, Department of Medicine III (S.F., A.M.Z.), Centre for Molecular Medicine, Goethe-University Frankfurt, Frankfurt, Germany; and Cardiovascular Research, Institute of Physiology, University of Zurich, Zurich, Switzerland (M.R., C.B., U.L.)
| |
Collapse
|
34
|
Alexandru N, Georgescu A. Circulating microparticles and microRNAs as players in atherosclerosis. World J Hematol 2013; 2:16-19. [DOI: 10.5315/wjh.v2.i2.16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 03/21/2013] [Accepted: 04/28/2013] [Indexed: 02/05/2023] Open
Abstract
Microparticles (MPs) are extracellular membrane vesicles released from normal, apoptotic and pathological cells following a process of detachment from cells of origin. MPs are typically defined by their size, exposure of phosphatidylserine, the expression of surface antigens, proteins and genetic material, originating from their donor cells, and as important vehicles of intercellular communication across numerous biological processes. MPs contain the major source of systemic RNA including microRNA (miRNA) of which aberrant expression appears to be associated with stage and progression of atherosclerosis. The involvement and influence of miRNA during the onset and progression of atherosclerotic disease have generated a lot of interest in assessing the feasibility of therapeutic regulation of miRNAs to manipulate them with a special focus on cardiovascular disease. We speculate on the future developments of MPs which contain miRNA as new therapeutic targets for proliferative vascular diseases such as atherosclerosis.
Collapse
|
35
|
Matejuk A, Collet G, Nadim M, Grillon C, Kieda C. MicroRNAs and tumor vasculature normalization: impact on anti-tumor immune response. Arch Immunol Ther Exp (Warsz) 2013; 61:285-99. [PMID: 23575964 DOI: 10.1007/s00005-013-0231-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Accepted: 01/15/2013] [Indexed: 12/21/2022]
Abstract
Inefficient immune response is a major glitch during tumor growth and progression. Chaotic and leaky blood vessels created in the process of angiogenesis allow tumor cells to escape and extricate anti-cancer immunity. Proangiogenic characteristics of hypoxic tumor microenvironment maintained by low oxygen tension attract endothelial progenitor cells, drive expansion of cancer stem cells, and deviantly differentiate monocyte descendants. Such cellular milieu further boosts immune tolerance and eventually appoint immunity for cancer advantage. Blood vessel normalization strategies that equilibrate oxygen levels within tumor and fix abnormal vasculature bring exciting promises to future anticancer therapies especially when combined with conventional chemotherapy. Recently, a new group of microRNAs (miRs) engaged in angiogenesis, called angiomiRs and hypoxamiRs, emerged as new therapeutic targets in cancer. Some of those miRs were found to efficiently regulate cancer immunity and their dysregulation efficiently programs aberrant angiogenesis and cancer metastasis. The present review highlights new findings in the field of miRs proficiency to normalize aberrant angiogenesis and to restore anti-tumor immune responses.
Collapse
Affiliation(s)
- Agata Matejuk
- Centre de Biophysique Moléculaire, CNRS UPR 4301, rue Charles Sadron, 45071 Orléans, France.
| | | | | | | | | |
Collapse
|
36
|
Baumgarten A, Bang C, Tschirner A, Engelmann A, Adams V, von Haehling S, Doehner W, Pregla R, Anker MS, Blecharz K, Meyer R, Hetzer R, Anker SD, Thum T, Springer J. TWIST1 regulates the activity of ubiquitin proteasome system via the miR-199/214 cluster in human end-stage dilated cardiomyopathy. Int J Cardiol 2013; 168:1447-52. [PMID: 23360823 DOI: 10.1016/j.ijcard.2012.12.094] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 12/18/2012] [Accepted: 12/24/2012] [Indexed: 12/28/2022]
Abstract
BACKGROUND The transcription factor TWIST1 has been described to regulate the microRNA (miR)-199/214 cluster. Genetic disruption of TWIST1 resulted in a cachectic phenotype and early death of the knock-out mice. This might be connected to the activity of the ubiquitin-proteasome-system (UPS), as miR-199a has been suggested to regulate the ubiquitin E2 ligases Ube2i and Ube2g1. METHODS Cardiac tissue from explanted hearts of 42 patients with dilated cardiomyopathy and 20 healthy donor hearts were analysed for protein expression of TWIST1 and its inhibitors Id-1, MuRF-1 and MAFbx, the expression of miR-199a, -199b and -214, as well as the activity of the UPS by using specific fluorogenic substrates. RESULTS TWIST1 was repressed in patients with dilated cardiomyopathy by 43% (p=0.003), while Id1 expression was unchanged. This was paralleled by a reduced expression of miR-199a by 38 ± 9% (p=0.053), miR-199b by 36 ± 13% (p=0.019) and miR-214 by 41 ± 11% (p=0.0158) compared to donor hearts. An increased peptidylglutamyl-peptide-hydrolysing activity (p<0.0001) was observed in the UPS, while the chymotrypsin-like and trypsin-like activities were unchanged. The protein levels of the rate limiting ubiquitin E3-ligases MuRF-1 and MAFbx were up-regulated (p=0.005 and p=0.0156, respectively). Mechanistically silencing of TWIST1 using siRNA in primary rat cardiomyocytes led to a down-regulation of the miR-199/214 cluster and to a subsequent up-regulation of Ube2i. CONCLUSION The TWIST1/miR-199/214 axis is down-regulated in dilated cardiomyopathy, which is likely to play a role in the increased activity of the UPS. This may contribute to the loss of cardiac mass during dilatation of the heart.
Collapse
Affiliation(s)
- Anna Baumgarten
- Center for Cardiovascular Research, Charité Medical School, Berlin, Germany; Applied Cachexia Research, Department of Cardiology, Charité Medical School, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Anand S. A brief primer on microRNAs and their roles in angiogenesis. Vasc Cell 2013; 5:2. [PMID: 23324117 PMCID: PMC3554556 DOI: 10.1186/2045-824x-5-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 01/11/2013] [Indexed: 01/08/2023] Open
Abstract
Development of the vasculature is a complex, dynamic process orchestrated by a balance of pro and anti-angiogenic signaling pathways. The same signaling pathways are mis-regulated and exploited during pathological angiogenesis in cancer, inflammation and cardiovascular diseases and contribute to disease progression. In the last decade, small non-coding RNA molecules termed microRNAs (miRs) have emerged as key regulators of several cellular processes including angiogenesis. It is becoming clear that miRs function in complex networks and regulate gene expression both at the mRNA and protein levels thereby altering cellular signaling responses to specific stimuli. In the vasculature, miRs can function either in a pro-angiogenic manner and potentiate angiogenesis or act as anti-angiogenic miRs by enhancing cell death and decreasing endothelial proliferation. This review aims to provide an update on how microRNAs regulate gene expression and illustrate miR function in the vasculature with a discussion of potential applications of miRs as anti-angiogenic therapeutics.
Collapse
Affiliation(s)
- Sudarshan Anand
- Moores UCSD Cancer Center, 3855 Health Sciences Drive #0803, La Jolla, CA, 92093, USA.
| |
Collapse
|
38
|
Abbott NJ, Friedman A. Overview and introduction: the blood-brain barrier in health and disease. Epilepsia 2013; 53 Suppl 6:1-6. [PMID: 23134489 DOI: 10.1111/j.1528-1167.2012.03696.x] [Citation(s) in RCA: 221] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This article introduces the special issue on "Blood-Brain Barrier and Epilepsy." We review briefly current understanding of the structure and function of the blood-brain barrier (BBB), including its development and normal physiology, and ways in which it can be affected in pathology. The BBB formed by the endothelium of cerebral blood vessels is one of three main barrier sites protecting the central nervous system (CNS). The barrier is not a rigid structure, but a dynamic interface with a range of interrelated functions, resulting from extremely effective tight junctions, transendothelial transport systems, enzymes, and regulation of leukocyte permeation, which thereby generates the physical, transport, enzymatic, and immune regulatory functions of the BBB. The brain endothelial cells are important components of a "modular" structure, the neurovascular unit (NVU), with several associated cell types and extracellular matrix components. Modern methods have helped in identifying a range of proteins involved in barrier structure and function, and recent studies have revealed important stages, cell types, and signaling pathways important in BBB development. There is a growing list of CNS pathologies showing BBB dysfunction, with strong evidence that this can play a major role in certain disease etiologies. The articles that follow in this issue summarize in more detail reports and discussions of the recent international meeting on "BBB in Neurological Dysfunctions," which took place recently at Ben-Gurion University of the Negev Desert Campus (Beer-Sheva, Israel), focusing on the link between experimental and clinical studies, and the ways in which these lead to improved drug treatments.
Collapse
Affiliation(s)
- N Joan Abbott
- Institute of Pharmaceutical Science, Blood-Brain Barrier Group, King's College London, London, United Kingdom
| | | |
Collapse
|
39
|
Cervelli T, Borghini A, Galli A, Andreassi MG. DNA damage and repair in atherosclerosis: current insights and future perspectives. Int J Mol Sci 2012; 13:16929-44. [PMID: 23443128 PMCID: PMC3546731 DOI: 10.3390/ijms131216929] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 11/20/2012] [Accepted: 12/05/2012] [Indexed: 11/16/2022] Open
Abstract
Atherosclerosis is the leading cause of morbidity and mortality among Western populations. Over the past two decades, considerable evidence has supported a crucial role for DNA damage in the development and progression of atherosclerosis. These findings support the concept that the prolonged exposure to risk factors (e.g., dyslipidemia, smoking and diabetes mellitus) leading to reactive oxygen species are major stimuli for DNA damage within the plaque. Genomic instability at the cellular level can directly affect vascular function, leading to cell cycle arrest, apoptosis and premature vascular senescence. The purpose of this paper is to review current knowledge on the role of DNA damage and DNA repair systems in atherosclerosis, as well as to discuss the cellular response to DNA damage in order to shed light on possible strategies for prevention and treatment.
Collapse
Affiliation(s)
- Tiziana Cervelli
- Institute of Clinical Physiology, CNR (The National Research Council), via Moruzzi 1, 56124 Pisa, Italy.
| | | | | | | |
Collapse
|
40
|
Li C, Pei F, Zhu X, Duan DD, Zeng C. Circulating microRNAs as novel and sensitive biomarkers of acute myocardial Infarction. Clin Biochem 2012; 45:727-32. [PMID: 22713968 DOI: 10.1016/j.clinbiochem.2012.04.013] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2011] [Revised: 04/01/2012] [Accepted: 04/09/2012] [Indexed: 12/12/2022]
Abstract
Coronary artery disease and acute myocardial infarction (AMI) are the leading causes of death for both men and women. Serum cardiac-specific troponin level is now used for the "early" diagnosis of AMI. However, due to the "delayed" release of troponin, an earlier, more sensitive and specific biomarker is urgently demanded to further reduce AMI mortality. Recent studies have found that circulating microRNAs (miRNAs) are closely linked to myocardial injury. Due to the cell-specific physiological functions and the stability of miRNAs in plasma, serum, and urine, they are emerging as sensitive biomarkers of AMI. This review summarizes the latest insights into the identification and potential application of plasma and serum miRNAs as novel biomarkers for diagnosis and prognosis of AMI.
Collapse
Affiliation(s)
- Chuanwei Li
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, PR China
| | | | | | | | | |
Collapse
|
41
|
Thomas RA, Scicchitano MS, Mirabile RC, Chau NT, Frazier KS, Thomas HC. MicroRNA changes in rat mesentery and serum associated with drug-induced vascular injury. Toxicol Appl Pharmacol 2012; 262:310-20. [DOI: 10.1016/j.taap.2012.05.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Revised: 05/11/2012] [Accepted: 05/13/2012] [Indexed: 12/16/2022]
|
42
|
Abstract
MicroRNAs (miRNAs) are a class of highly conserved, noncoding short RNA molecules that regulate gene expression on the post-transcriptional level. MiRNAs are involved in a variety of processes such as proliferation, differentiation, and apoptosis. Deregulated expression of miRNAs has been linked to the development of diseases including cardiovascular disorders. Recently, the miR-23/27/24 cluster has been shown to be involved in angiogenesis and endothelial apoptosis in cardiac ischemia and retinal vascular development. In the present review, we summarize and discuss the role and importance of the miRNA-23/27/24 cluster during cardiovascular angiogenesis. Moreover, we illustrate a novel therapeutic application of the miRNA-23/27/24 cluster in vascular disorders and ischemic heart disease.
Collapse
Affiliation(s)
- Claudia Bang
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | | | | |
Collapse
|
43
|
Abstract
Hypertension is a complex, multifactorial disease, and its development is determined by a combination of genetic susceptibility and environmental factors. Several mechanisms have been implicated in the pathogenesis of hypertension: increased activity of the sympathetic nervous system, overactivation of the renin-angiotensin aldosterone system (RAAS), dysfunction of vascular endothelium, impaired platelet function, thrombogenesis, vascular smooth muscle and cardiac hypertrophy, and altered angiogenesis. MicroRNAs are short, noncoding nucleotides regulating target messenger RNAs at the post-transcriptional level. MicroRNAs are involved in virtually all biologic processes, including cellular proliferation, apoptosis, and differentiation. Thus, microRNA deregulation often results in impaired cellular function and disease development, so microRNAs have potential therapeutic relevance. Many aspects of the development of essential hypertension at the molecular level are still unknown. The elucidation of these processes regulated by microRNAs and the identification of novel microRNA targets in the pathogenesis of hypertension is a highly valuable and exciting strategy that may eventually led to the development of novel treatment approaches for hypertension. This article reviews the potential role of microRNAs in the mechanisms associated with the development and consequences of hypertension and discusses advances in microRNA-based approaches that may be important in treating hypertension.
Collapse
|
44
|
Collet G, Skrzypek K, Grillon C, Matejuk A, El Hafni-Rahbi B, Lamerant-Fayel N, Kieda C. Hypoxia control to normalize pathologic angiogenesis: potential role for endothelial precursor cells and miRNAs regulation. Vascul Pharmacol 2012; 56:252-61. [PMID: 22446152 DOI: 10.1016/j.vph.2012.03.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2012] [Revised: 02/18/2012] [Accepted: 03/02/2012] [Indexed: 01/12/2023]
Abstract
Tumor microenvironment is a complex and highly dynamic milieu that provides very important clues on tumor development and progression mechanisms. Tumor-associated endothelial cells play a key role in stroma organization. They achieve tumor angiogenesis, a formation of tumor-associated (angiogenic) vessels mainly through sprouting from locally preexisting vessels and/or recruitment of bone marrow-derived endothelial progenitor cells. This process participates to supply nutritional support and oxygen to the growing tumor. Endothelial cells constitute the interface between circulating blood cells, tumor cells and the extracellular matrix, thereby controlling leukocyte recruitment, tumor cell behavior and metastasis formation. Hypoxia, a critical parameter of the tumor microenvironment, controls endothelial/tumor cell interactions and is the key to tumor angiogenesis development. Under hypoxic stress, tumor cells produce factors that promote angiogenesis, vasculogenesis, tumor cell motility, metastasis and cancer stem cell selection. Targeting tumor vessels is a therapeutic strategy that has lately been fast evolving from antiangiogenesis to vessel normalization as discussed in this review. We shall focus on the pivotal role of endothelial cells within the tumor microenvironment, the specific features and the part played by circulating endothelial precursors cells. Attention is stressed on their recruitment to the tumor site and their role in tumor angiogenesis where they are submitted to miRNAs-mediated de/regulation. Here the compensation of the tumor deregulated angiogenic miRNAs - angiomiRs - is emphasized as a potential therapeutic approach. The strategy is to over express anti-angiomiRs in the tumor angiogenesis site upon selective delivery by precursor endothelial cells as miRs carriers.
Collapse
Affiliation(s)
- Guillaume Collet
- Centre de Biophysique Moléculaire, CNRS UPR 4301, rue Charles Sadron, 45071 Orleans, France
| | | | | | | | | | | | | |
Collapse
|
45
|
Aroor AR, Mandavia C, Ren J, Sowers JR, Pulakat L. Mitochondria and Oxidative Stress in the Cardiorenal Metabolic Syndrome. Cardiorenal Med 2012; 2:87-109. [PMID: 22619657 DOI: 10.1159/000335675] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Accepted: 12/08/2011] [Indexed: 12/16/2022] Open
Abstract
Mitochondria play a fundamental role in the maintenance of normal structure, function, and survival of tissues. There is considerable evidence for mitochondrial dysfunction in association with metabolic diseases including insulin resistance, obesity, diabetes, and the cardiorenal metabolic syndrome. The phenomenon of reactive oxygen species (ROS)-induced ROS release through interactions between cytosolic and mitochondrial oxidative stress contributes to a vicious cycle of enhanced oxidative stress and mitochondrial dysfunction. Activation of the cytosolic and mitochondrial NADPH oxidase system, impairment of the mitochondrial electron transport, activation of p66shc pathway-targeting mitochondria, endoplasmic reticular stress, and activation of the mammalian target of the rapamycin-S6 kinase pathway underlie dysregulation of mitochondrial dynamics and promote mitochondrial oxidative stress. These processes are further modulated by acetyltransferases including sirtuin 1 and sirtuin 3, the former regulating nuclear acetylation and the latter regulating mitochondrial acetylation. The regulation of mitochondrial functions by microRNAs forms an additional layer of molecular control of mitochondrial oxidative stress. Alcohol further exacerbates mitochondrial oxidative stress induced by overnutrition and promotes the development of metabolic diseases.
Collapse
Affiliation(s)
- Annayya R Aroor
- Department of Internal Medicine, University of Missouri School of Medicine, Columbia, Mo., USA
| | | | | | | | | |
Collapse
|
46
|
Diehl P, Fricke A, Sander L, Stamm J, Bassler N, Htun N, Ziemann M, Helbing T, El-Osta A, Jowett JBM, Peter K. Microparticles: major transport vehicles for distinct microRNAs in circulation. Cardiovasc Res 2012; 93:633-44. [PMID: 22258631 PMCID: PMC3291092 DOI: 10.1093/cvr/cvs007] [Citation(s) in RCA: 383] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Aims Circulating microRNAs (miRNAs) have attracted major interest as biomarkers for cardiovascular diseases. Since RNases are abundant in circulating blood, there needs to be a mechanism protecting miRNAs from degradation. We hypothesized that microparticles (MP) represent protective transport vehicles for miRNAs and that these are specifically packaged by their maternal cells. Methods and results Conventional plasma preparations, such as the ones used for biomarker detection, are shown to contain substantial numbers of platelet-, leucocyte-, and endothelial cell-derived MP. To analyse the widest spectrum of miRNAs, Next Generation Sequencing was used to assess miRNA profiles of MP and their corresponding stimulated and non-stimulated cells of origin. THP-1 (monocytic origin) and human umbilical vein endothelial cell (HUVEC) MP were used for representing circulating MP at a high purity. miRNA profiles of MP differed significantly from those of stimulated and non-stimulated maternal THP-1 cells and HUVECs, respectively. Quantitative reverse transcription–polymerase chain reaction of miRNAs which have been associated with cardiovascular diseases also demonstrated significant differences in miRNA profiles between platelets and their MP. Notably, the main fraction of miRNA in plasma was localized in MP. Furthermore, miRNA profiles of MP differed significantly between patients with stable and unstable coronary artery disease. Conclusion Circulating MP represent transport vehicles for large numbers of specific miRNAs, which have been associated with cardiovascular diseases. miRNA profiles of MP are significantly different from their maternal cells, indicating an active mechanism of selective ‘packaging’ from cells into MP. These findings describe an interesting mechanism for transferring gene-regulatory function from MP-releasing cells to target cells via MP circulating in blood.
Collapse
Affiliation(s)
- Philipp Diehl
- Atherothrombosis and Vascular Biology, BakerIDI Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Lorenzen JM, Martino F, Thum T. Epigenetic modifications in cardiovascular disease. Basic Res Cardiol 2012; 107:245. [PMID: 22234702 PMCID: PMC3329881 DOI: 10.1007/s00395-012-0245-9] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Revised: 12/12/2011] [Accepted: 12/31/2011] [Indexed: 01/29/2023]
Abstract
Epigenetics represents a phenomenon of altered heritable phenotypic expression of genetic information occurring without changes in DNA sequence. Epigenetic modifications control embryonic development, differentiation and stem cell (re)programming. These modifications can be affected by exogenous stimuli (e.g., diabetic milieu, smoking) and oftentimes culminate in disease initiation. DNA methylation has been studied extensively and represents a well-understood epigenetic mechanism. During this process cytosine residues preceding a guanosine in the DNA sequence are methylated. CpG-islands are short-interspersed DNA sequences with clusters of CG sequences. The abnormal methylation of CpG islands in the promoter region of genes leads to a silencing of genetic information and finally to alteration of biological function. Emerging data suggest that these epigenetic modifications also impact on the development of cardiovascular disease. Histone modifications lead to the modulation of the expression of genetic information through modification of DNA accessibility. In addition, RNA-based mechanisms (e.g., microRNAs and long non-coding RNAs) influence the development of disease. We here outline the recent work pertaining to epigenetic changes in a cardiovascular disease setting.
Collapse
Affiliation(s)
- Johan M Lorenzen
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany.
| | | | | |
Collapse
|