101
|
Zhao X, Wang Y, Meng C, Fang N. FMRP regulates endothelial cell proliferation and angiogenesis via the miR-181a-CaM-CaMKII pathway. Cell Biol Int 2018; 42:1432-1444. [PMID: 30080293 DOI: 10.1002/cbin.11039] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 07/29/2018] [Indexed: 12/25/2022]
Abstract
RNA binding proteins (RBPs) and microRNAs have emerged as crucial post-transcriptional regulators of gene expression. Although the role of Fragile X mental retardation protein (FMRP) has been well studied in the brain, the function of FMRP in endothelial cells remains unknown. In our study, we showed that FMRP controlled human umbilical vein endothelial cells (HUVECs) proliferation and angiogenesis via the miR-181a-mediated calmodulin (CaM)/CaMKII pathway. The knockdown of FMRP induced miR-181a expression and contributed to endothelial cell proliferation and angiogenesis. Furthermore, we identified CaM as a downstream target of miR-181a in endothelial cells. Additionally, tumor necrosis factor-ɑ (TNF-ɑ) treatment specifically decreased the activity of the CaM/CaMKII pathway through the dephosphorylation of FMRP and upregulation of miR-181a. Finally, the overexpression of constitutively phosphorylated FMRP rescued the TNF-ɑ-impaired endothelial cell proliferation and angiogenesis by activating the CaM/CaMKII pathway and downregulating miR-181a, which suggested there was a pivotal role of FMRP in vascular integrity in response to inflammatory stimuli. Thus, our study supports a novel function and mechanism involving FMRP and the miR-181a-CaM-CaMKII pathway may be a therapeutic target for protecting against inflammation-induced vascular diseases.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Geriatrics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, No. 160 Pujian Road, Pudong New Area, Shanghai 200127, China
| | - Yang Wang
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, No.138 Yixueyuan Road, Shanghai 200032, China
| | - Chao Meng
- Department of Geriatrics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, No. 160 Pujian Road, Pudong New Area, Shanghai 200127, China
| | - Ningyuan Fang
- Department of Geriatrics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, No. 160 Pujian Road, Pudong New Area, Shanghai 200127, China
| |
Collapse
|
102
|
Kokkonen-Simon KM, Saberi A, Nakamura T, Ranek MJ, Zhu G, Bedja D, Kuhn M, Halushka MK, Lee DI, Kass DA. Marked disparity of microRNA modulation by cGMP-selective PDE5 versus PDE9 inhibitors in heart disease. JCI Insight 2018; 3:121739. [PMID: 30089721 DOI: 10.1172/jci.insight.121739] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 06/26/2018] [Indexed: 12/13/2022] Open
Abstract
MicroRNAs (miRs) posttranscriptionally regulate mRNA and its translation into protein, and are considered master controllers of genes modulating normal physiology and disease. There is growing interest in how miRs change with drug treatment, and leveraging this for precision guided therapy. Here we contrast 2 closely related therapies, inhibitors of phosphodiesterase type 5 or type 9 (PDE5-I, PDE9-I), given to mice subjected to sustained cardiac pressure overload (PO). Both inhibitors augment cyclic guanosine monophosphate (cGMP) to activate protein kinase G, with PDE5-I regulating nitric oxide (NO) and PDE9-I natriuretic peptide-dependent signaling. While both produced strong phenotypic improvement of PO pathobiology, they surprisingly showed binary differences in miR profiles; PDE5-I broadly reduces more than 120 miRs, including nearly half those increased by PO, whereas PDE9-I has minimal impact on any miR (P < 0.0001). The disparity evolves after pre-miR processing and is organ specific. Lastly, even enhancing NO-coupled cGMP by different methods leads to altered miR regulation. Thus, seemingly similar therapeutic interventions can be barcoded by profound differences in miR signatures, and reversing disease-associated miR changes is not required for therapy success.
Collapse
Affiliation(s)
- Kristen M Kokkonen-Simon
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, Maryland, USA.,Cellular and Molecular Medicine Graduate Program, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Amir Saberi
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, Maryland, USA
| | - Taishi Nakamura
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, Maryland, USA
| | - Mark J Ranek
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, Maryland, USA
| | - Guangshuo Zhu
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, Maryland, USA
| | - Djahida Bedja
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, Maryland, USA
| | - Michaela Kuhn
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Marc K Halushka
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Dong Ik Lee
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, Maryland, USA
| | - David A Kass
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, Maryland, USA.,Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
103
|
Mollazadeh H, Carbone F, Montecucco F, Pirro M, Sahebkar A. Oxidative burden in familial hypercholesterolemia. J Cell Physiol 2018; 233:5716-5725. [PMID: 29323716 DOI: 10.1002/jcp.26466] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 01/05/2018] [Indexed: 12/19/2022]
Abstract
Familial hypercholesterolemia (FH) is a genetic disorder characterized by high serum levels of low-density lipoprotein cholesterol (LDL-c). FH is characterized by accelerated development of atherosclerosis and represents the most frequent hereditary cause of premature coronary heart disease. Mutations of the LDL receptor gene are the genetic signature of FH, resulting in abnormal levels of circulating LDLs. Moreover, FH promotes the generation of reactive oxygen species (ROS) which is another key mechanism involved in atherosclerosis development and progression. The aim of this narrative review is to update the current knowledge on the pathophysiological mechanisms linking FH to ROS generation and their detrimental impact on atherosclerotic pathophysiology. With this purpose, we reviewed experimental and clinical data on the association between FH and OS and the functional role of OS as a promoter of inflammation and atherosclerosis. In this regard, oxidant species such as oxidized LDL, malondialdehyde, ROS, and isoprostanes emerged as leading mediators of the oxidative injury in FH. In conclusion, targeting oxidative stress may be a promising therapeutic strategy to reduce atherogenesis in patients with FH.
Collapse
Affiliation(s)
- Hamid Mollazadeh
- Department of Physiology and Pharmacology, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- Ospedale Policlinico San Martino, Genoa, Italy
- Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Matteo Pirro
- Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases, Department of Medicine, University of Perugia, Perugia, Italy
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
104
|
MicroRNAs distribution in different phenotypes of Aortic Stenosis. Sci Rep 2018; 8:9953. [PMID: 29967333 PMCID: PMC6028376 DOI: 10.1038/s41598-018-28246-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 05/22/2018] [Indexed: 01/01/2023] Open
Abstract
Aortic valve stenosis (AVS) represents a cluster of different phenotypes, considering gradient and flow pattern. Circulating micro RNAs may reflect specific pathophysiological processes and could be useful biomarkers to identify disease. We assessed 80 patients (81, 76.7–84 years; 46, 57.5%females) with severe AVS. We performed bio-humoral evaluation (including circulating miRNA-1, 21, 29, 133) and 2D-echocardiography. Patients were classified according to ACC/AHA groups (D1-D3) and flow-gradient classification, considering normal/low flow, (NF/LF) and normal/high gradient, (NG/HG). Patients with reduced ejection fractionwere characterized by higher levels of miRNA1 (p = 0.003) and miRNA 133 (p = 0.03). LF condition was associated with higher levels of miRNA1 (p = 0.02) and miRNA21 (p = 0.02). Levels of miRNA21 were increased in patients with reduced Global longitudinal strain (p = 0.03). LF-HG and LF-LG showed higher levels of miRNA1 expression (p = 0.005). At one-year follow-up miRNA21 and miRNA29 levels resulted significant independent predictors of reverse remodeling and systolic function increase, respectively. Different phenotypes of AVS may express differential levels and types of miRNAs, which may retain a pathophysiological role in pro-hypertrophic and pro-fibrotic processes.
Collapse
|
105
|
TGF-β signaling alters H4K20me3 status via miR-29 and contributes to cellular senescence and cardiac aging. Nat Commun 2018; 9:2560. [PMID: 29967491 PMCID: PMC6028646 DOI: 10.1038/s41467-018-04994-z] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 05/22/2018] [Indexed: 12/21/2022] Open
Abstract
Cellular senescence is a well-orchestrated programmed process involved in age-related pathologies, tumor suppression and embryonic development. TGF-β/Smad is one of the predominant pathways that regulate damage-induced and developmentally programmed senescence. Here we show that canonical TGF-β signaling promotes senescence via miR-29-induced loss of H4K20me3. Mechanistically, oxidative stress triggers TGF-β signaling. Activated TGF-β signaling gives rise to acute accumulation of miR-29a and miR-29c, both of which directly suppress their novel target, Suv4-20h, thus reducing H4K20me3 abundance in a Smad-dependent manner, which compromises DNA damage repair and genome maintenance. Loss of H4K20me3 mediated by the senescent TGF-β/miR-29 pathway contributes to cardiac aging in vivo. Disruption of TGF-β signaling restores H4K20me3 and improves cardiac function in aged mice. Our study highlights the sequential mechanisms underlying the regulation of senescence, from senescence-inducing triggers to activation of responsive signaling followed by specific epigenetic alterations, shedding light on potential therapeutic interventions in cardiac aging.
Collapse
|
106
|
Li J, Huang J, Yang X, Yang Y, Quan K, Xie N, Wu Y, Ma C, Wang K. Gold nanoparticle-based 2′-O-methyl modified DNA probes for breast cancerous theranostics. Talanta 2018; 183:11-17. [DOI: 10.1016/j.talanta.2018.02.036] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 02/07/2018] [Accepted: 02/08/2018] [Indexed: 12/15/2022]
|
107
|
Steffensen LB, Feddersen S, Preil SR, Rasmussen LM. No detectable differential microRNA expression between non-atherosclerotic arteries of type 2 diabetic patients (treated or untreated with metformin) and non-diabetic patients. Cardiovasc Diabetol 2018; 17:72. [PMID: 29773082 PMCID: PMC5958402 DOI: 10.1186/s12933-018-0715-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 05/10/2018] [Indexed: 12/15/2022] Open
Abstract
Background Type 2 diabetes mellitus (T2DM) is an independent risk factor of cardiovascular disease (CVD), however, the underlying mechanisms are largely unknown. Using non-atherosclerotic internal thoracic arteries (ITAs) obtained from coronary artery bypass grafting, we previously identified a distinct elevation in the level of proteins comprising the arterial basement membrane in T2DM patients not treated with metformin. Altered transcription of genes encoding these proteins has not been observed, indicating alternative mechanisms of dysregulation. Methods In this study we screened for differential expression of arterial microRNAs (miRNAs) in T2DM patients to test the hypothesis that the arterial protein signature of diabetic patients is associated with dysregulation at the miRNA level, and further to lay the foundation for novel hypotheses addressing the increased CVD risk of T2DM patients. MiRNA isolated from fresh frozen ITAs [from 18 T2DM- (10 of which were subject to metformin treatment) and 30 non-diabetes mellitus (non-DM) patients] were analyzed by microarray, and miRNAs isolated from formalin-fixated paraffin-embedded (FFPE) ITAs were analyzed by quantitative PCR (qPCR) in an independent study group [26 T2DM- (15 of which were subject to metformin treatment) and 26 non-DM patients] to determine expression levels of miRNAs in a pre-defined panel of 12 miRNAs. Results Unexpectedly, no miRNAs were found to be affected by T2DM status in either of the two study groups. Conclusions Our data suggest that alternatives to microRNA dysregulation underlie T2DM-associated protein changes in non-atherosclerotic arteries. Electronic supplementary material The online version of this article (10.1186/s12933-018-0715-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lasse Bach Steffensen
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark. .,Centre for Individualized Medicine in Arterial Diseases (CIMA), Odense University Hospital, Sdr. Boulevard 29, 5000, Odense C, Denmark. .,Department of Clinical Research, University of Southern Denmark, Odense, Denmark. .,Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.
| | - Søren Feddersen
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Simone Rørdam Preil
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark.,Centre for Individualized Medicine in Arterial Diseases (CIMA), Odense University Hospital, Sdr. Boulevard 29, 5000, Odense C, Denmark
| | - Lars Melholt Rasmussen
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark.,Centre for Individualized Medicine in Arterial Diseases (CIMA), Odense University Hospital, Sdr. Boulevard 29, 5000, Odense C, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
108
|
Liu B, Lee BW, Nakanishi K, Villasante A, Williamson R, Metz J, Kim J, Kanai M, Bi L, Brown K, Di Paolo G, Homma S, Sims PA, Topkara VK, Vunjak-Novakovic G. Cardiac recovery via extended cell-free delivery of extracellular vesicles secreted by cardiomyocytes derived from induced pluripotent stem cells. Nat Biomed Eng 2018; 2:293-303. [PMID: 30271672 PMCID: PMC6159913 DOI: 10.1038/s41551-018-0229-7] [Citation(s) in RCA: 257] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 03/20/2018] [Indexed: 12/16/2022]
Abstract
The ability of extracellular vesicles (EVs) to regulate a broad range of cellular processes has recently been exploited for the treatment of diseases. For example, EVs secreted by stem cells injected into infarcted hearts can induce recovery through the delivery of stem-cell-specific miRNAs. However, the retention of the EVs and the therapeutic effects are short-lived. Here, we show that an engineered hydrogel patch capable of slowly releasing EVs secreted from cardiomyocytes derived from induced pluripotent stem (iPS) cells reduced arrhythmic burden, promoted ejection-fraction recovery, decreased cardiomyocyte apoptosis 24 hours after infarction, and reduced infarct size and cell hypertrophy 4 weeks post-infarction when implanted onto infarcted rat hearts. We also show that the EVs are enriched with cardiac-specific miRNAs known to modulate cardiomyocyte-specific processes. The extended delivery of EVs secreted from iPS-cell-derived cardiomyocytes into the heart may help understand heart recovery and treat heart injury.
Collapse
Affiliation(s)
- Bohao Liu
- College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Medicine, Columbia University, New York, NY, USA
| | - Benjamin W Lee
- College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Koki Nakanishi
- Department of Medicine, Columbia University, New York, NY, USA
| | - Aranzazu Villasante
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Rebecca Williamson
- College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Jordan Metz
- College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Jinho Kim
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Mariko Kanai
- Department of Medicine, Columbia University, New York, NY, USA
| | - Lynn Bi
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Kristy Brown
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Gilbert Di Paolo
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Shunichi Homma
- Department of Medicine, Columbia University, New York, NY, USA
| | - Peter A Sims
- Department of Systems Biology, Columbia University, New York, NY, USA
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Veli K Topkara
- Department of Medicine, Columbia University, New York, NY, USA
| | - Gordana Vunjak-Novakovic
- Department of Medicine, Columbia University, New York, NY, USA.
- Department of Biomedical Engineering, Columbia University, New York, NY, USA.
| |
Collapse
|
109
|
Effects of microRNA-292-5p on myocardial ischemia-reperfusion injury through the peroxisome proliferator-activated receptor-α/-γ signaling pathway. Gene Ther 2018; 25:234-248. [PMID: 29670247 DOI: 10.1038/s41434-018-0014-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 02/10/2018] [Accepted: 02/26/2018] [Indexed: 12/30/2022]
Abstract
Ischemia-reperfusion injury (IRI) is a major cause of cardiac damage following various pathological processes, such as free radical damage and cell apoptosis. This study aims to investigate whether microRNA-292-5p (miR-292-5p) protects against myocardial ischemia-reperfusion injury (IRI) via the peroxisome proliferator-activated receptor (PPAR)-α/-γ signaling pathway in myocardial IRI mice models. Mouse models of myocardial IRI were established. Adult male C57BL/6 mice were divided into different groups. The hemodynamic indexes, levels of related inflammatory factors and serum myocardial enzymes, and malondialdehyde (MDA) content and the activity of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) were detected. The 2,3,5-triphenyltetrazolium chloride (TTC) staining was applied to determine infarct size. TUNEL staining was used to detect cardiomyocyte apoptosis. RT-qPCR and western blotting were performed to measure the related gene expressions. Compared with the model group and the T0070907 + miR-292-5p inhibitor, the miR-292-5p inhibitor group exhibited decreased incidence and duration time of ventricular tachycardia and ventricular fibrillation, serum myocardial enzymes, TNF-α, IL-6, IL-1β, MDA, cardiomyocyte apoptosis, expressions of Bax and p53 in addition to increased SOD and GSH-Px activity, and increased expressions of Bcl-2, PPARα, PPARγ, PLIN5, AQP7, and PCK1. The T0070907 group exhibited opposite results compared to the miR-292-5p inhibitor group. The results indicate that miR-292-5p downregulation protects against myocardial IRI through activation of the PPAR-α/PPAR-γ signaling pathway.
Collapse
|
110
|
Xu J, Wu H, Chen S, Qi B, Zhou G, Cai L, Zhao L, Wei Y, Liu S. MicroRNA-30c suppresses the pro-fibrogenic effects of cardiac fibroblasts induced by TGF-β1 and prevents atrial fibrosis by targeting TGFβRII. J Cell Mol Med 2018. [PMID: 29532993 PMCID: PMC5980214 DOI: 10.1111/jcmm.13548] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Atrial fibrosis serves as an important contributor to atrial fibrillation (AF). Recent data have suggested that microRNA-30c (miR-30c) is involved in fibrotic remodelling and cancer development, but the specific role of miR-30c in atrial fibrosis remains unclear. The purpose of this study was to investigate the role of miR-30c in atrial fibrosis and its underlying mechanisms through in vivo and in vitro experiments. Our results indicate that miR-30c is significantly down-regulated in the rat abdominal aortic constriction (AAC) model and in the cellular model of fibrosis induced by transforming growth factor-β1 (TGF-β1). Overexpression of miR-30c in cardiac fibroblasts (CFs) markedly inhibits CF proliferation, differentiation, migration and collagen production, whereas decrease in miR-30c leads to the opposite results. Moreover, we identified TGFβRII as a target of miR-30c. Finally, transferring adeno-associated virus 9 (AAV9)-miR-30c into the inferior vena cava of rats attenuated fibrosis in the left atrium following AAC. These data indicate that miR-30c attenuates atrial fibrosis via inhibition of CF proliferation, differentiation, migration and collagen production by targeting TGFβRII, suggesting that miR-30c might be a novel potential therapeutic target for preventing atrial fibrosis.
Collapse
Affiliation(s)
- Juan Xu
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haiqing Wu
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Songwen Chen
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Baozhen Qi
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Genqing Zhou
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lidong Cai
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liqun Zhao
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yong Wei
- Department of Cardiology, Shanghai Songjiang Central Hospital, Shanghai, China
| | - Shaowen Liu
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
111
|
Zhang X, Yin J, Zhang X. A Semi-Supervised Learning Algorithm for Predicting Four Types MiRNA-Disease Associations by Mutual Information in a Heterogeneous Network. Genes (Basel) 2018; 9:genes9030139. [PMID: 29498680 PMCID: PMC5867860 DOI: 10.3390/genes9030139] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 02/20/2018] [Accepted: 02/22/2018] [Indexed: 01/05/2023] Open
Abstract
Increasing evidence suggests that dysregulation of microRNAs (miRNAs) may lead to a variety of diseases. Therefore, identifying disease-related miRNAs is a crucial problem. Currently, many computational approaches have been proposed to predict binary miRNA-disease associations. In this study, in order to predict underlying miRNA-disease association types, a semi-supervised model called the network-based label propagation algorithm is proposed to infer multiple types of miRNA-disease associations (NLPMMDA) by mutual information derived from the heterogeneous network. The NLPMMDA method integrates disease semantic similarity, miRNA functional similarity, and Gaussian interaction profile kernel similarity information of miRNAs and diseases to construct a heterogeneous network. NLPMMDA is a semi-supervised model which does not require verified negative samples. Leave-one-out cross validation (LOOCV) was implemented for four known types of miRNA-disease associations and demonstrated the reliable performance of our method. Moreover, case studies of lung cancer and breast cancer confirmed effective performance of NLPMMDA to predict novel miRNA-disease associations and their association types.
Collapse
Affiliation(s)
- Xiaotian Zhang
- School of Mechanical, Electrical and Information Engineering, Shandong University, Weihai 264209, China.
| | - Jian Yin
- School of Mechanical, Electrical and Information Engineering, Shandong University, Weihai 264209, China.
| | - Xu Zhang
- School of Mechanical, Electrical and Information Engineering, Shandong University, Weihai 264209, China.
| |
Collapse
|
112
|
Seok H, Lee H, Jang ES, Chi SW. Evaluation and control of miRNA-like off-target repression for RNA interference. Cell Mol Life Sci 2018; 75:797-814. [PMID: 28905147 PMCID: PMC11105550 DOI: 10.1007/s00018-017-2656-0] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 09/06/2017] [Accepted: 09/07/2017] [Indexed: 01/08/2023]
Abstract
RNA interference (RNAi) has been widely adopted to repress specific gene expression and is easily achieved by designing small interfering RNAs (siRNAs) with perfect sequence complementarity to the intended target mRNAs. Although siRNAs direct Argonaute (Ago), a core component of the RNA-induced silencing complex (RISC), to recognize and silence target mRNAs, they also inevitably function as microRNAs (miRNAs) and suppress hundreds of off-targets. Such miRNA-like off-target repression is potentially detrimental, resulting in unwanted toxicity and phenotypes. Despite early recognition of the severity of miRNA-like off-target repression, this effect has often been overlooked because of difficulties in recognizing and avoiding off-targets. However, recent advances in genome-wide methods and knowledge of Ago-miRNA target interactions have set the stage for properly evaluating and controlling miRNA-like off-target repression. Here, we describe the intrinsic problems of miRNA-like off-target effects caused by canonical and noncanonical interactions. We particularly focus on various genome-wide approaches and chemical modifications for the evaluation and prevention of off-target repression to facilitate the use of RNAi with secured specificity.
Collapse
Affiliation(s)
- Heeyoung Seok
- Division of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Korea
| | - Haejeong Lee
- Division of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Korea
| | - Eun-Sook Jang
- Division of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Korea
- EncodeGEN Co. Ltd, Seoul, 06329, Korea
| | - Sung Wook Chi
- Division of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Korea.
| |
Collapse
|
113
|
Zhang X, Yoon JY, Morley M, McLendon JM, Mapuskar KA, Gutmann R, Mehdi H, Bloom HL, Dudley SC, Ellinor PT, Shalaby AA, Weiss R, Tang WHW, Moravec CS, Singh M, Taylor AL, Yancy CW, Feldman AM, McNamara DM, Irani K, Spitz DR, Breheny P, Margulies KB, London B, Boudreau RL. A common variant alters SCN5A-miR-24 interaction and associates with heart failure mortality. J Clin Invest 2018; 128:1154-1163. [PMID: 29457789 DOI: 10.1172/jci95710] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 12/12/2017] [Indexed: 12/19/2022] Open
Abstract
SCN5A encodes the voltage-gated Na+ channel NaV1.5 that is responsible for depolarization of the cardiac action potential and rapid intercellular conduction. Mutations disrupting the SCN5A coding sequence cause inherited arrhythmias and cardiomyopathy, and single-nucleotide polymorphisms (SNPs) linked to SCN5A splicing, localization, and function associate with heart failure-related sudden cardiac death. However, the clinical relevance of SNPs that modulate SCN5A expression levels remains understudied. We recently generated a transcriptome-wide map of microRNA (miR) binding sites in human heart, evaluated their overlap with common SNPs, and identified a synonymous SNP (rs1805126) adjacent to a miR-24 site within the SCN5A coding sequence. This SNP was previously shown to reproducibly associate with cardiac electrophysiological parameters, but was not considered to be causal. Here, we show that miR-24 potently suppresses SCN5A expression and that rs1805126 modulates this regulation. We found that the rs1805126 minor allele associates with decreased cardiac SCN5A expression and that heart failure subjects homozygous for the minor allele have decreased ejection fraction and increased mortality, but not increased ventricular tachyarrhythmias. In mice, we identified a potential basis for this in discovering that decreased Scn5a expression leads to accumulation of myocardial reactive oxygen species. Together, these data reiterate the importance of considering the mechanistic significance of synonymous SNPs as they relate to miRs and disease, and highlight a surprising link between SCN5A expression and nonarrhythmic death in heart failure.
Collapse
Affiliation(s)
- Xiaoming Zhang
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Jin-Young Yoon
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Michael Morley
- Department of Internal Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jared M McLendon
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Kranti A Mapuskar
- Department of Radiation Oncology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Rebecca Gutmann
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Haider Mehdi
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Heather L Bloom
- Department of Medicine, Emory University Medical Center, Atlanta, Georgia, USA
| | - Samuel C Dudley
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Patrick T Ellinor
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Alaa A Shalaby
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Raul Weiss
- Department of Internal Medicine, The Ohio State University Medical Center, Columbus, Ohio, USA
| | - W H Wilson Tang
- Department of Cardiovascular Medicine, Cleveland Clinic Lerner College of Medicine, Cleveland, Ohio, USA
| | - Christine S Moravec
- Department of Molecular Cardiology, Cleveland Clinic Lerner College of Medicine, Cleveland, Ohio, USA
| | - Madhurmeet Singh
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Anne L Taylor
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | - Clyde W Yancy
- Division of Cardiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Arthur M Feldman
- Department of Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| | - Dennis M McNamara
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Kaikobad Irani
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Douglas R Spitz
- Department of Radiation Oncology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Patrick Breheny
- Department of Biostatistics, University of Iowa College of Public Heath, Iowa City, Iowa, USA
| | - Kenneth B Margulies
- Department of Internal Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Barry London
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Ryan L Boudreau
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| |
Collapse
|
114
|
Halade GV, Kain V, Serhan CN. Immune responsive resolvin D1 programs myocardial infarction-induced cardiorenal syndrome in heart failure. FASEB J 2018; 32:3717-3729. [PMID: 29455574 DOI: 10.1096/fj.201701173rr] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Resolvins are innate, immune responsive, bioactive mediators generated after myocardial infarction (MI) to resolve inflammation. The MI-induced bidirectional interaction between progressive left ventricle (LV) remodeling and kidney dysfunction is known to advance cardiorenal syndrome (CRS). Whether resolvins limit MI-induced cardiorenal inflammation is unclear. Thus, to define the role of exogenous resolvin D (RvD)-1 in post-MI CRS, we subjected 8- to 12-wk-old male C57BL/6 mice to coronary artery ligation. RvD1 was injected 3 h after MI. MI mice with no treatment served as MI controls (d 1 and 5). Mice with no surgery served as naive controls. In the injected mice, RvD1 promoted neutrophil (CD11b+/Ly6G+) egress from the infarcted LV, compared with the MI control group at d 5, indicative of neutrophil clearance and thereby resolved inflammation. Further, RvD1-injected mice showed higher reparative macrophages (F4/80+/Ly6Clow/CD206+) in the infarcted LV than did MI control mice at d 5 after MI. RvD1 suppressed the miRNA storm at d 1 and limited the MI-induced edematous milieu in a remote area of the LV compared with the MI control at d 5 after MI. Also, RvD1 preserved the nephrin expression that was diffuse in the glomerular membrane at d 5 and 28 in MI controls, indicating renal injury. RvD1 attenuated MI-induced renal inflammation, decreasing neutrophil gelatinase-associated lipocalin and proinflammatory cytokines and chemokines in the kidney compared with the MI control. In summary, RvD1 clears MI-induced inflammation by increasing resolving leukocytes and facilitates renoprotective mechanisms to limit CRS in acute and chronic heart failure.-Halade, G. V., Kain, V., Serhan, C. N. Immune responsive resolvin D1 programs myocardial infarction-induced cardiorenal syndrome in heart failure.
Collapse
Affiliation(s)
- Ganesh V Halade
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Vasundhara Kain
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
115
|
Zhao Y, Li Y, Tong L, Liang X, Zhang H, Li L, Fan G, Wang Y. Analysis of microRNA Expression Profiles Induced by Yiqifumai Injection in Rats with Chronic Heart Failure. Front Physiol 2018; 9:48. [PMID: 29467665 PMCID: PMC5808162 DOI: 10.3389/fphys.2018.00048] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 01/15/2018] [Indexed: 12/14/2022] Open
Abstract
Background: Yiqifumai Injection (YQFM) is clinically used to treat various cardiovascular diseases including chronic heart failure (CHF). The efficacy of YQFM for treating heart failure has been suggested, but the mechanism of action for pharmacological effects of YQFM is unclear. Methods: Echocardiography detection, left ventricular intubation evaluation, histopathology and immunohistochemical examination were performed in CHF rats to evaluate the cardioprotective effect of YQFM. Rat miRNA microarray and bioinformatics analysis were employed to investigate the differentially expressed microRNAs. In vitro models of AngII-induced hypertrophy and t-BHP induced oxidative stress in H9c2 myocardial cells were used to validate the anti-hypertrophy and anti-apoptosis effects of YQFM. Measurement of cell surface area, ATP content and cell viability, Real-time PCR and Western blot were performed. Results: YQFM significantly improved the cardiac function of CHF rats by increasing left ventricular ejection fraction and fractional shortening, decreasing left ventricular internal diameter and enhancing cardiac output. Seven microRNAs which have a reversible regulation by YQFM treatment were found. Among them, miR-21-3p and miR-542-3p are related to myocardial hypertrophy and cell proliferation, respectively and were further verified by RT-PCR. Target gene network was established and potential related signaling pathways were predicted. YQFM could significantly alleviate AngII induced hypertrophy in cellular model. It also significantly increased cell viabilities and ATP content in t-BHP induced apoptotic cell model. Western blot analysis showed that YQFM could increase the phosphorylation of Akt. Conclusion: Our findings provided scientific evidence to uncover the mechanism of action of YQFM on miRNAs regulation against CHF by miRNA expression profile technology. The results indicated that YQFM has a potential effect on alleviate cardiac hypertrophy and apoptosis in chronic heart failure.
Collapse
Affiliation(s)
- Yu Zhao
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yunfei Li
- State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tasly Academy, Tasly Holding Group Co., Ltd., Tianjin, China
| | - Ling Tong
- State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tasly Academy, Tasly Holding Group Co., Ltd., Tianjin, China
| | - Xinying Liang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Han Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lan Li
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guanwei Fan
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yi Wang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
116
|
Liu Y, Li H, Zhao C, Jia H. MicroRNA-101 inhibits angiogenesis via COX-2 in endometrial carcinoma. Mol Cell Biochem 2018; 448:61-69. [PMID: 29404887 DOI: 10.1007/s11010-018-3313-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 01/27/2018] [Indexed: 12/22/2022]
Abstract
Abnormal angiogenesis is critically involved in tumor progression and metastasis including endometrial cancer and is regulated by microRNAs such as microRNA-101 (miR-101). We hypothesize that miR-101 expression is disrupted in endometrial cancer and modulation of miR-101 levels is sufficient to regulate tumor growth through angiogenesis. We examined the expression levels of miR-101 and factors involved in angiogenesis in the patients with endometrial cancer. We also overexpressed or inhibited miR-101 in RL-95-2 cells and examined their effects on cell toxicity and tumor growth. Finally, we determined if miR-101 regulated tumorigenesis through cyclooxygenase-2 (COX-2). We found that miR-101 levels were significantly reduced. Factors involved in angiogenesis included vascular endothelial growth factor-A (VEGF-A), thrombospondin-1 (TSP-1), cyclooxygenase-2 (COX-2), prostaglandin E2 (PGE2), and aromatase (P450arom), which were increased in endometrial carcinoma. Modulation of miR-101 level was sufficient to affect tumor growth. Finally, we found that the effects of miR-101 inhibition on tumor growth were suppressed by COX-2 inhibition. Our results suggest that modulating miR-101 and COX-2 levels or their activity may be a potential therapeutic strategy for endometrial cancer.
Collapse
Affiliation(s)
- Ying Liu
- The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Shijiazhuang, 050000, Hebei, China
| | - Haiyan Li
- The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Shijiazhuang, 050000, Hebei, China.
| | - Congying Zhao
- The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Shijiazhuang, 050000, Hebei, China
| | - Hanbing Jia
- The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Shijiazhuang, 050000, Hebei, China
| |
Collapse
|
117
|
Abstract
The cardiac fibroblast has essential roles in production and maintenance of extracellular matrix. While its role in maladaptive myocardial remodeling has been a focus of many studies, the cardiac fibroblast has become a topic of great interest as a contributor to heart physiology and as a therapeutic target. Recent reports are changing how we view and study the cardiac fibroblast by providing greater insights into fibroblast biology using refined techniques for fibroblast identification and manipulation. Here, we briefly summarize some of these fundamental recent findings.
Collapse
Affiliation(s)
- Michelle D Tallquist
- University of Hawaii at Manoa, Center for Cardiovascular Research, Department of Medicine
| |
Collapse
|
118
|
Ji Y, Qiu M, Shen Y, Gao L, Wang Y, Sun W, Li X, Lu Y, Kong X. MicroRNA-327 regulates cardiac hypertrophy and fibrosis induced by pressure overload. Int J Mol Med 2018; 41:1909-1916. [PMID: 29393356 PMCID: PMC5810199 DOI: 10.3892/ijmm.2018.3428] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 01/19/2018] [Indexed: 12/24/2022] Open
Abstract
MicroRNA (miRNA/miR) dysregulation has been reported to be fundamental in the development and progression of cardiac hypertrophy and fibrosis. In the present study, miR-327 levels in fibroblasts were increased in response to cardiac hypertrophy induced by transverse aortic constriction with prominent cardiac fibrosis, particularly when compared with the levels in unstressed cardiomyocytes. In neonatal rat cardiac fibroblasts, induced expression of miR-327 upregulated fibrosis-associated gene expression and activated angiotensin II-induced differentiation into myofibroblasts, as assessed via α-smooth muscle actin staining. By contrast, miR-327 knockdown mitigated angiotensin II-induced differentiation. Cardiac fibroblast proliferation was not affected under either condition. In a mouse model subjected to transverse aortic constriction, miR-327 knockdown through tail-vein injection reduced the development of cardiac fibrosis and ventricular dysfunction. miR-327 was demonstrated to target integrin β3 and diminish the activation of cardiac fibroblasts. Thus, the present study supports the use of miR-327 as a therapeutic target in the reduction of cardiac fibrosis.
Collapse
Affiliation(s)
- Yue Ji
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Ming Qiu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yejiao Shen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Li Gao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yaqing Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Wei Sun
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Xinli Li
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yan Lu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Xiangqing Kong
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
119
|
Marotta P, Cianflone E, Aquila I, Vicinanza C, Scalise M, Marino F, Mancuso T, Torella M, Indolfi C, Torella D. Combining cell and gene therapy to advance cardiac regeneration. Expert Opin Biol Ther 2018; 18:409-423. [PMID: 29347847 DOI: 10.1080/14712598.2018.1430762] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION The characterization of multipotent endogenous cardiac stem cells (eCSCs) and the breakthroughs of somatic cell reprogramming to boost cardiomyocyte replacement have fostered the prospect of achieving functional heart repair/regeneration. AREAS COVERED Allogeneic CSC therapy through its paracrine stimulation of the endogenous resident reparative/regenerative process produces functional meaningful myocardial regeneration in pre-clinical porcine myocardial infarction models and is currently tested in the first-in-man human trial. The in vivo test of somatic reprogramming and cardioregenerative non-coding RNAs revived the interest in gene therapy for myocardial regeneration. The latter, together with the advent of genome editing, has prompted most recent efforts to produce genetically-modified allogeneic CSCs that secrete cardioregenerative factors to optimize effective myocardial repair. EXPERT OPINION The current war against heart failure epidemics in western countries seeks to find effective treatments to set back the failing hearts prolonging human lifespan. Off-the-shelf allogeneic-genetically-modified CSCs producing regenerative agents are a novel and evolving therapy set to be affordable, safe, effective and available at all times for myocardial regeneration to either prevent or treat heart failure.
Collapse
Affiliation(s)
- Pina Marotta
- a Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences , Magna Graecia University , Catanzaro , Italy
| | - Eleonora Cianflone
- a Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences , Magna Graecia University , Catanzaro , Italy
| | - Iolanda Aquila
- a Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences , Magna Graecia University , Catanzaro , Italy
| | - Carla Vicinanza
- a Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences , Magna Graecia University , Catanzaro , Italy
| | - Mariangela Scalise
- a Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences , Magna Graecia University , Catanzaro , Italy
| | - Fabiola Marino
- a Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences , Magna Graecia University , Catanzaro , Italy
| | - Teresa Mancuso
- a Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences , Magna Graecia University , Catanzaro , Italy
| | - Michele Torella
- b Department of Cardiothoracic Sciences , University of Campania "L. Vanvitelli" , Naples , Italy
| | - Ciro Indolfi
- a Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences , Magna Graecia University , Catanzaro , Italy
| | - Daniele Torella
- a Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences , Magna Graecia University , Catanzaro , Italy
| |
Collapse
|
120
|
Abstract
Despite recent advances in scientific knowledge and clinical practice, cardiovascular disease management and treatment remain a major burden. While several treatment strategies using drugs and surgeries are being developed for cardiovascular manifestations, gene-based therapies hold significant promise. Recent findings from our laboratory unveiled a novel mechanism that exosomes, secreted nanovesicles from stem cells, mediate cardiac repair via transferring their unique repertoire of microRNAs (miRNA) to recipient cells in the heart. Exosomes, unlike other vectors for gene delivery, present unique advantages such that exosomes are a cell-free natural system for ferrying RNA between cells, robust exosomal membrane can protect the RNA/gene of interest from digestion, and exosomes are rapidly taken up by target cells making them a more efficient vehicle for gene delivery. Here, we describe a stepwise protocol developed in our laboratory for generating exosomes from human CD34+ stem cells that carry exogenously applied Cy3 dye-labeled pre-miR miRNA precursors. We demonstrate that human CD34+ stem cell exosomes can rigorously enter into recipient cells and deliver Cy3 dye-labeled pre-miR miRNA precursors to regulate gene expression. Identification of key molecular targets to treat disease conditions is the foremost critical step and the novel approach presented here to generate exosomes carrying exogenous genetic information offers a valuable clinical tool for more effective treatment strategies.
Collapse
Affiliation(s)
- Prabhu Mathiyalagan
- Cardiovascular Research Center, Icahn School of Medicine, Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029-6574, USA
| | - Susmita Sahoo
- Cardiovascular Research Center, Icahn School of Medicine, Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY, 10029-6574, USA.
| |
Collapse
|
121
|
Dogan MV, Grumbach IM, Michaelson JJ, Philibert RA. Integrated genetic and epigenetic prediction of coronary heart disease in the Framingham Heart Study. PLoS One 2018; 13:e0190549. [PMID: 29293675 PMCID: PMC5749823 DOI: 10.1371/journal.pone.0190549] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 12/15/2017] [Indexed: 12/16/2022] Open
Abstract
An improved method for detecting coronary heart disease (CHD) could have substantial clinical impact. Building on the idea that systemic effects of CHD risk factors are a conglomeration of genetic and environmental factors, we use machine learning techniques and integrate genetic, epigenetic and phenotype data from the Framingham Heart Study to build and test a Random Forest classification model for symptomatic CHD. Our classifier was trained on n = 1,545 individuals and consisted of four DNA methylation sites, two SNPs, age and gender. The methylation sites and SNPs were selected during the training phase. The final trained model was then tested on n = 142 individuals. The test data comprised of individuals removed based on relatedness to those in the training dataset. This integrated classifier was capable of classifying symptomatic CHD status of those in the test set with an accuracy, sensitivity and specificity of 78%, 0.75 and 0.80, respectively. In contrast, a model using only conventional CHD risk factors as predictors had an accuracy and sensitivity of only 65% and 0.42, respectively, but with a specificity of 0.89 in the test set. Regression analyses of the methylation signatures illustrate our ability to map these signatures to known risk factors in CHD pathogenesis. These results demonstrate the capability of an integrated approach to effectively model symptomatic CHD status. These results also suggest that future studies of biomaterial collected from longitudinally informative cohorts that are specifically characterized for cardiac disease at follow-up could lead to the introduction of sensitive, readily employable integrated genetic-epigenetic algorithms for predicting onset of future symptomatic CHD.
Collapse
Affiliation(s)
- Meeshanthini V. Dogan
- Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa, United States of America
- Department of Psychiatry, University of Iowa, Iowa City, Iowa, United States of America
- Cardio Diagnostics LLC, Coralville, Iowa, United States of America
| | - Isabella M. Grumbach
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States of America
- Iowa City Veterans Affairs Healthcare System, Iowa City, Iowa, United States of America
| | - Jacob J. Michaelson
- Department of Psychiatry, University of Iowa, Iowa City, Iowa, United States of America
| | - Robert A. Philibert
- Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa, United States of America
- Department of Psychiatry, University of Iowa, Iowa City, Iowa, United States of America
- Behavioral Diagnostics LLC, Coralville, Iowa, United States of America
- * E-mail:
| |
Collapse
|
122
|
Qian LB, Jiang SZ, Tang XQ, Zhang J, Liang YQ, Yu HT, Chen J, Xu Z, Liu RM, Keller BB, Ji HL, Cai L. Exacerbation of diabetic cardiac hypertrophy in OVE26 mice by angiotensin II is associated with JNK/c-Jun/miR-221-mediated autophagy inhibition. Oncotarget 2017; 8:106661-106671. [PMID: 29290979 PMCID: PMC5739764 DOI: 10.18632/oncotarget.21302] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 09/18/2017] [Indexed: 12/31/2022] Open
Abstract
Both diabetes and angiotensin II (Ang II) excess trigger cardiac remodeling and dysfunction, and diabetic cardiomyopathy. We hypothesized that cardiac hypertrophy associated with the development of diabetic cardiomyopathy is worsened by increased Ang II. Male type 1 diabetic OVE26 and wild-type mice were given Ang II (sc., 1.15 mg/kg, twice a day) for 14 days. Diabetes-induced cardiac dysfunction and hypertrophy was exacerbated by Ang II treatment as determined by echocardiography, wheat germ agglutinin staining and atrial natriuretic peptide. Ang II treatment dramatically exacerbated diabetes-caused decreased LC3-II, a marker of autophagy, and increased p62, an indicator of cytosolic protein clearance. Ang II treatment also augmented diabetes-associated increased phosphorylated levels of c-Jun, JNK, mTOR, and miR-221, and decreased of p27 expression, a direct target of miR-221. Chromatin immunoprecipitation assay showed that Ang II elevated c-Jun binding to the promoter of miR-221 in diabetic mice. These results suggest that Ang II accelerates cardiac hypertrophy in the early stage of murine diabetes, probably through activation of the JKN/c-Jun/miR-221 axis and inhibition of downstream autophagy. Therefore, inhibition of Ang II or miR-221 in diabetic individuals may be a potential approach for delaying the onset and/or reducing the severity of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Ling-Bo Qian
- Cardiovascular Center, The First Hospital of Jilin University, Changchun 130021, China
- Department of Basic Medical Sciences, Hangzhou Medical College, Hangzhou 310053, China
- Pediatric Research Institute, Department of Pediatrics of the University of Louisville, Louisville, Kentucky 40202, USA
| | - Sai-Zhi Jiang
- Pediatric Research Institute, Department of Pediatrics of the University of Louisville, Louisville, Kentucky 40202, USA
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Xiao-Qiang Tang
- Cardiovascular Center, The First Hospital of Jilin University, Changchun 130021, China
- Pediatric Research Institute, Department of Pediatrics of the University of Louisville, Louisville, Kentucky 40202, USA
| | - Jian Zhang
- Cardiovascular Center, The First Hospital of Jilin University, Changchun 130021, China
- Pediatric Research Institute, Department of Pediatrics of the University of Louisville, Louisville, Kentucky 40202, USA
| | - Ya-Qin Liang
- Pediatric Research Institute, Department of Pediatrics of the University of Louisville, Louisville, Kentucky 40202, USA
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Hai-Tao Yu
- Cardiovascular Center, The First Hospital of Jilin University, Changchun 130021, China
- Pediatric Research Institute, Department of Pediatrics of the University of Louisville, Louisville, Kentucky 40202, USA
| | - Jing Chen
- Pediatric Research Institute, Department of Pediatrics of the University of Louisville, Louisville, Kentucky 40202, USA
| | - Zheng Xu
- Cardiovascular Center, The First Hospital of Jilin University, Changchun 130021, China
- Pediatric Research Institute, Department of Pediatrics of the University of Louisville, Louisville, Kentucky 40202, USA
| | - Rui-Ming Liu
- Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama 35294, USA
| | - Bradley B. Keller
- Cardiovascular Center, The First Hospital of Jilin University, Changchun 130021, China
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky 40202, USA
| | - Hong-Lei Ji
- Cardiovascular Center, The First Hospital of Jilin University, Changchun 130021, China
- Pediatric Research Institute, Department of Pediatrics of the University of Louisville, Louisville, Kentucky 40202, USA
| | - Lu Cai
- Cardiovascular Center, The First Hospital of Jilin University, Changchun 130021, China
- Pediatric Research Institute, Department of Pediatrics of the University of Louisville, Louisville, Kentucky 40202, USA
| |
Collapse
|
123
|
Lopez MS, Dempsey RJ, Vemuganti R. The microRNA miR-21 conditions the brain to protect against ischemic and traumatic injuries. CONDITIONING MEDICINE 2017; 1:35-46. [PMID: 34268484 PMCID: PMC8279043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Ischemic and traumatic injuries to CNS remain leading causes of death and disability worldwide, despite decades of research into risk factors, therapies, and preventative measures. Recent studies showed that CNS injuries significantly alter the cerebral microRNAome that impact the secondary brain damage as well as plasticity and recovery. Many microRNA based therapies are currently in various clinical trials for different pathologic conditions indicating their therapeutic potential. In the present review, we discuss the role of miR-21 in acute CNS injuries which is currently thought to be a potent neuroprotective microRNA. We emphasize on the potential of miR-21 in promoting cell and tissue survival and preventing inflammation and apoptosis. We also discussed the role of miR-21 in conditioning the brain to promote ischemic tolerance. Finally, we discussed some of the challenges and difficulties to develop miR-21 as a neuroprotective therapy in humans.
Collapse
Affiliation(s)
- Mary S Lopez
- Cellular and Molecular Pathology Program, University of Wisconsin, Madison, WI, USA
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Robert J Dempsey
- Cellular and Molecular Pathology Program, University of Wisconsin, Madison, WI, USA
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Raghu Vemuganti
- Cellular and Molecular Pathology Program, University of Wisconsin, Madison, WI, USA
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
- William S. Middleton Veteran's Administration Hospital, Madison, WI, USA
| |
Collapse
|
124
|
Smith DA, Newbury LJ, Drago G, Bowen T, Redman JE. Electrochemical detection of urinary microRNAs via sulfonamide-bound antisense hybridisation. SENSORS AND ACTUATORS. B, CHEMICAL 2017; 253:335-341. [PMID: 29200659 PMCID: PMC5614097 DOI: 10.1016/j.snb.2017.06.069] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Altered serum and plasma microRNA (miRNA) expression profiles have been observed in numerous human diseases, with a number of studies describing circulating miRNA biomarkers for cancer diagnosis, prognosis and response to treatment, and recruitment to clinical trials for miRNA-based drug therapy already underway. Electrochemical detection of biomarkers in urine has several significant advantages over circulating biomarker analysis including safety, cost, speed and ease of conversion to the point of care environment. Consequently, much current research is underway to identify urinary miRNA biomarkers for a variety of pathologies including prostate and bladder malignancies, and renal disorders. We describe here a robust method capable of electrochemical detection of human urinary miRNAs at femtomolar concentrations using a complementary DNA-modified glassy carbon electrode. A miR-21-specific DNA hybridisation probe was immobilised onto a glassy carbon electrode modified by sulfonic acid deposition and subsequent chlorination. In our pilot system, the presence of synthetic mature miR-21 oligonucleotides increased resistance at the probe surface to electron transfer from the ferricyanide/ferrocyanide electrolyte. Response was linear for 10 nM-10 fM miR-21, with a limit of detection of 20 fM, and detection discriminated between miR-21, three point-mutated miR-21 sequences, and miR-16. We then demonstrated similar sensitivity and reproducibility of miR-21 detection in urine samples from 5 human control subjects. Our protocol provides a platform for future high-throughput screening of miRNA biomarkers in liquid biopsies.
Collapse
Affiliation(s)
- Daniel A. Smith
- School of Chemistry, Cardiff University, Cardiff CF10 3AT, UK
- Wales Kidney Research Unit, Division of Infection and Immunity, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
- Cardiff Institute of Tissue Engineering and Repair, Cardiff University, Museum Place, Cardiff CF10 3BG, UK
| | - Lucy J. Newbury
- Wales Kidney Research Unit, Division of Infection and Immunity, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
- Cardiff Institute of Tissue Engineering and Repair, Cardiff University, Museum Place, Cardiff CF10 3BG, UK
| | - Guido Drago
- Gwent Electronic Materials Ltd, Monmouth House, Mamhilad Pk Est, Pontypool NP4 0HZ, UK
| | - Timothy Bowen
- Wales Kidney Research Unit, Division of Infection and Immunity, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
- Cardiff Institute of Tissue Engineering and Repair, Cardiff University, Museum Place, Cardiff CF10 3BG, UK
| | - James E. Redman
- School of Chemistry, Cardiff University, Cardiff CF10 3AT, UK
- Cardiff Institute of Tissue Engineering and Repair, Cardiff University, Museum Place, Cardiff CF10 3BG, UK
- Corresponding author at: School of Chemistry, Cardiff University, Cardiff CF10 3AT, UK.
| |
Collapse
|
125
|
Jeong D, Yoo J, Lee P, Kepreotis SV, Lee A, Wahlquist C, Brown BD, Kho C, Mercola M, Hajjar RJ. miR-25 Tough Decoy Enhances Cardiac Function in Heart Failure. Mol Ther 2017; 26:718-729. [PMID: 29273502 DOI: 10.1016/j.ymthe.2017.11.014] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 11/17/2017] [Accepted: 11/22/2017] [Indexed: 01/14/2023] Open
Abstract
MicroRNAs are promising therapeutic targets, because their inhibition has the potential to normalize gene expression in diseased states. Recently, our group found that miR-25 is a key SERCA2a regulating microRNA, and we showed that multiple injections of antagomirs against miR-25 enhance cardiac contractility and function through SERCA2a restoration in a murine heart failure model. However, for clinical application, a more stable suppressor of miR-25 would be desirable. Tough Decoy (TuD) inhibitors are emerging as a highly effective method for microRNA inhibition due to their resistance to endonucleolytic degradation, high miRNA binding affinity, and efficient delivery. We generated a miR-25 TuD inhibitor and subcloned it into a cardiotropic AAV9 vector to evaluate its efficacy. The AAV9 TuD showed selective inhibition of miR-25 in vitro cardiomyoblast culture. In vivo, AAV9-miR-25 TuD delivered to the murine pressure-overload heart failure model selectively decreased expression of miR-25, increased levels of SERCA2a protein, and ameliorated cardiac dysfunction and fibrosis. Our data indicate that miR-25 TuD is an effective long-term suppressor of miR-25 and a promising therapeutic candidate to treat heart failure.
Collapse
Affiliation(s)
- Dongtak Jeong
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jimeen Yoo
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Philyoung Lee
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sacha V Kepreotis
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ahyoung Lee
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Christine Wahlquist
- Department of Medicine and Cardiovascular Institute, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Brian D Brown
- Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Changwon Kho
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mark Mercola
- Department of Medicine and Cardiovascular Institute, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Roger J Hajjar
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
126
|
Wang M, Yu F, Wu W, Zhang Y, Chang W, Ponnusamy M, Wang K, Li P. Circular RNAs: A novel type of non-coding RNA and their potential implications in antiviral immunity. Int J Biol Sci 2017; 13:1497-1506. [PMID: 29230098 PMCID: PMC5723916 DOI: 10.7150/ijbs.22531] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 10/08/2017] [Indexed: 12/23/2022] Open
Abstract
Circular RNAs (circRNAs), a novel type of non-coding RNAs (ncRNAs), are ubiquitously expressed in eukaryotic cells during post-transcriptional processes. Unlike linear RNAs, circRNAs form covalent-closed continuous loops without 5' to 3' polarities and poly (A) tails. With advances in high-throughput sequencing technology, numerous circRNAs have been identified in plants, animals and humans. Notably, circRNAs display cell-type, tissue-type and developmental-stage specific expression patterns in eukaryotic transcriptome, which reveals their significant regulatory functions in gene expression. More importantly, circRNAs serve as microRNA (miRNA) sponges and crucial regulators of gene expression. Additionally, circRNAs modulate pre-mRNA alternative splicing and possess protein-coding capacity. CircRNAs exhibit altered expression under pathological conditions and are strongly associated with the development of various human diseases. Interestingly, circRNAs can also induce antiviral immune responses. A recent study found that the delivery of circRNAs generated in vitro activates RIG-I-mediated innate immune responses and provides protection against viral infection. The antiviral dsRNA-binding proteins, NF90/NF110, act as key regulators in circRNA biogenesis. NF90/NF110 are also functional in inhibiting viral replication through binding to viral mRNAs. In this review, we provide a comprehensive overview on the classification, biogenesis and functions of circRNAs. We also discuss the critical role of circRNAs in eliciting antiviral immunity, providing evidence for the potential implications of circRNAs in antiviral therapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Kun Wang
- Institute for Translational Medicine, Medical College of Qingdao University, Dengzhou Road 38, Qingdao 266021, China
| | - Peifeng Li
- Institute for Translational Medicine, Medical College of Qingdao University, Dengzhou Road 38, Qingdao 266021, China
| |
Collapse
|
127
|
Xiao J, Liu H, Cretoiu D, Toader DO, Suciu N, Shi J, Shen S, Bei Y, Sluijter JP, Das S, Kong X, Li X. miR-31a-5p promotes postnatal cardiomyocyte proliferation by targeting RhoBTB1. Exp Mol Med 2017; 49:e386. [PMID: 29053138 PMCID: PMC5668467 DOI: 10.1038/emm.2017.150] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 02/11/2017] [Accepted: 04/11/2017] [Indexed: 01/02/2023] Open
Abstract
A limited number of microRNAs (miRNAs, miRs) have been reported to control postnatal cardiomyocyte proliferation, but their strong regulatory effects suggest a possible therapeutic approach to stimulate regenerative capacity in the diseased myocardium. This study aimed to investigate the miRNAs responsible for postnatal cardiomyocyte proliferation and their downstream targets. Here, we compared miRNA profiles in cardiomyocytes between postnatal day 0 (P0) and day 10 (P10) using miRNA arrays, and found that 21 miRNAs were upregulated at P10, whereas 11 were downregulated. Among them, miR-31a-5p was identified as being able to promote cardiomyocyte proliferation as determined by proliferating cell nuclear antigen (PCNA) expression, double immunofluorescent labeling for α-actinin and 5-ethynyl-2-deoxyuridine (EdU) or Ki-67, and cell number counting, whereas miR-31a-5p inhibition could reduce their levels. RhoBTB1 was identified as a target gene of miR-31a-5p, mediating the regulatory effect of miR-31a-5p in cardiomyocyte proliferation. Importantly, neonatal rats injected with a miR-31a-5p antagomir at day 0 for three consecutive days exhibited reduced expression of markers of cardiomyocyte proliferation including PCNA expression and double immunofluorescent labeling for α-actinin and EdU, Ki-67 or phospho-histone-H3. In conclusion, miR-31a-5p controls postnatal cardiomyocyte proliferation by targeting RhoBTB1, and increasing miR-31a-5p level might be a novel therapeutic strategy for enhancing cardiac reparative processes.
Collapse
Affiliation(s)
- Junjie Xiao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Cardiac Regeneration and Ageing Lab, School of Life Science, Shanghai University, Shanghai, China
| | - Hui Liu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Dragos Cretoiu
- Victor Babes National Institute of Pathology, Bucharest, Romania.,Division of Cellular and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Daniela Oana Toader
- Department of Obstetrics and Gynecology, Polizu Clinical Hospital, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Nicolae Suciu
- Department of Obstetrics and Gynecology, Polizu Clinical Hospital, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania.,Alessandrescu-Rusescu National Institute of Mother and Child Health, Bucharest, Romania
| | - Jing Shi
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shutong Shen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yihua Bei
- Cardiac Regeneration and Ageing Lab, School of Life Science, Shanghai University, Shanghai, China.,Innovative Drug Research Center of Shanghai University, Shanghai, China
| | - Joost Pg Sluijter
- Laboratory of Experimental Cardiology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Saumya Das
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Xiangqing Kong
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xinli Li
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
128
|
Chodari L, Mohammadi M, Mohaddes G, Ghorbanzadeh V, Dariushnejad H. THE EFFECT OF TESTOSTERONE AND VOLUNTARY EXERCISE, ALONE OR TOGETHER, ON miRNA-126 EXPRESSION CHANGES IN HEART OF DIABETIC RATS. ACTA ENDOCRINOLOGICA-BUCHAREST 2017; 13:266-271. [PMID: 31149186 DOI: 10.4183/aeb.2017.266] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Methods Ninety Wistar male rats were used in this study. Type 1 diabetes was induced by i.p injection of 50 mg/kg of streptozotocin in all animals. After 42 days of treatment with testosterone (2mg/kg/day) or voluntary exercise alone or in combination, the heart of the rats has been removed and MicroRNA was extracted from the heart using miRCURYTM RNA isolation kit. Results Our results showed that either testosterone or exercise increased miRNA-126 expression levels in the heart of diabetic rats. Treatment of diabetic rats with testosterone and exercise at the same time had a synergistic effect on miRNA-126 levels in the heart. Furthermore, in castrated diabetes group, miRNA-126 levels were significantly decreased in heart, whereas either testosterone treatment or exercise training enhanced expression of this miRNA. Also, simultaneous treatment of castrated diabetic rats with testosterone and exercise had an additive effect on miRNA-126 expression levels. Conclusion This study showed that testosterone and exercise promote an increase in the expression of miRNA-126 in the heart tissue and this may be related to cardiac angiogenesis. These results may indicate that testosterone and exercise can help to prevent progression of diabetic cardiomyopathy due to impaired angiogenesis in the heart.
Collapse
Affiliation(s)
- L Chodari
- Department of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran.,Neurophysiology Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - M Mohammadi
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - G Mohaddes
- Neuroscience Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - V Ghorbanzadeh
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - H Dariushnejad
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
129
|
Choong OK, Lee DS, Chen CY, Hsieh PCH. The roles of non-coding RNAs in cardiac regenerative medicine. Noncoding RNA Res 2017; 2:100-110. [PMID: 30159427 PMCID: PMC6096405 DOI: 10.1016/j.ncrna.2017.06.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 06/03/2017] [Accepted: 06/05/2017] [Indexed: 02/06/2023] Open
Abstract
The emergence of non-coding RNAs (ncRNAs) has challenged the central dogma of molecular biology that dictates that the decryption of genetic information starts from transcription of DNA to RNA, with subsequent translation into a protein. Large numbers of ncRNAs with biological significance have now been identified, suggesting that ncRNAs are important in their own right and their roles extend far beyond what was originally envisaged. ncRNAs do not only regulate gene expression, but are also involved in chromatin architecture and structural conformation. Several studies have pointed out that ncRNAs participate in heart disease; however, the functions of ncRNAs still remain unclear. ncRNAs are involved in cellular fate, differentiation, proliferation and tissue regeneration, hinting at their potential therapeutic applications. Here, we review the current understanding of both the biological functions and molecular mechanisms of ncRNAs in heart disease and describe some of the ncRNAs that have potential heart regeneration effects.
Collapse
Affiliation(s)
- Oi Kuan Choong
- Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Desy S Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Chen-Yun Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Patrick C H Hsieh
- Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan.,Institute of Medical Genomics and Proteomics, Institute of Clinical Medicine and Department of Surgery, National Taiwan University & Hospital, Taipei 100, Taiwan
| |
Collapse
|
130
|
Abstract
PURPOSE OF REVIEW Work over the past decade has identified the important role of microRNAs (miRNAS) in regulating lipoprotein metabolism and associated disorders including metabolic syndrome, obesity, and atherosclerosis. This review summarizes the most recent findings in the field, highlighting the contribution of miRNAs in controlling LDL-cholesterol (LDL-C) and HDL-cholesterol (HDL-C) metabolism. RECENT FINDINGS A number of miRNAs have emerged as important regulators of lipid metabolism, including miR-122 and miR-33. Work over the past 2 years has identified additional functions of miR-33 including the regulation of macrophage activation and mitochondrial metabolism. Moreover, it has recently been shown that miR-33 regulates vascular homeostasis and cardiac adaptation in response to pressure overload. In addition to miR-33 and miR-122, recent GWAS have identified single-nucleotide polymorphisms in the proximity of miRNA genes associated with abnormal levels of circulating lipids in humans. Several of these miRNAs, such as miR-148a and miR-128-1, target important proteins that regulate cellular cholesterol metabolism, including the LDL receptor (LDLR) and the ATP-binding cassette A1 (ABCA1). SUMMARY MicroRNAs have emerged as critical regulators of cholesterol metabolism and promising therapeutic targets for treating cardiometabolic disorders including atherosclerosis. Here, we discuss the recent findings in the field, highlighting the novel mechanisms by which miR-33 controls lipid metabolism and atherogenesis, and the identification of novel miRNAs that regulate LDL metabolism. Finally, we summarize the recent findings that identified miR-33 as an important noncoding RNA that controls cardiovascular homeostasis independent of its role in regulating lipid metabolism.
Collapse
Affiliation(s)
- Binod Aryal
- Vascular Biology and Therapeutics Program, Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine, and Department of Pathology, Yale University School of Medicine, 10 Amistad St., New Haven, CT 06510. USA
| | - Abhishek K. Singh
- Vascular Biology and Therapeutics Program, Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine, and Department of Pathology, Yale University School of Medicine, 10 Amistad St., New Haven, CT 06510. USA
| | - Noemi Rotllan
- Vascular Biology and Therapeutics Program, Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine, and Department of Pathology, Yale University School of Medicine, 10 Amistad St., New Haven, CT 06510. USA
| | - Nathan Price
- Vascular Biology and Therapeutics Program, Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine, and Department of Pathology, Yale University School of Medicine, 10 Amistad St., New Haven, CT 06510. USA
| | - Carlos Fernández-Hernando
- Vascular Biology and Therapeutics Program, Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine, and Department of Pathology, Yale University School of Medicine, 10 Amistad St., New Haven, CT 06510. USA
- Corresponding author: Carlos Fernández-Hernando. Phone: +1 (203)-737-4615.
| |
Collapse
|
131
|
Ram TP, Fomison-Nurse I, Gandhi S, Coffey S, Saxena P, Galvin I, Bunton R, Williams MJ, Lamberts RR, Katare R. The diagnostic sensitivity of circulating cardio-enriched microRNAs is increased after normalization of high-density lipoprotein levels. Int J Cardiol 2017; 236:498-500. [DOI: 10.1016/j.ijcard.2017.01.119] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 01/15/2017] [Accepted: 01/24/2017] [Indexed: 10/20/2022]
|
132
|
Abstract
An increasing number of diseases are being newly closely associated with inflammation, where microRNAs seem to play a critical role in the whole disease process from initiation to development. MicroRNAs are small non-coding RNAs that govern gene expression and modulation by means of mRNA degradation or translational repression. After several profound research studies, new correlations between microRNA-155 and inflammation-related diseases are strongly emerging. Hence, we review in this paper the possible molecular mechanisms of microRNA-155 in inflammatory disorders. Furthermore, we also consider the feasibility of targeting it as a bright alternative to improve the early diagnose statistics and treatments in those diseases. MicroRNA-155 features a novel breakthrough in fine-tuning inflammatory responses and, thereby, in treating a wide spectrum of diseases with inflammation as a common denominator.
Collapse
|
133
|
Bertero T, Rezzonico R, Pottier N, Mari B. Impact of MicroRNAs in the Cellular Response to Hypoxia. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 333:91-158. [PMID: 28729029 DOI: 10.1016/bs.ircmb.2017.03.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In mammalian cells, hypoxia, or inadequate oxygen availability, regulates the expression of a specific set of MicroRNAs (MiRNAs), termed "hypoxamiRs." Over the past 10 years, the appreciation of the importance of hypoxamiRs in regulating the cellular adaptation to hypoxia has grown dramatically. At the cellular level, each hypoxamiR, including the master hypoxamiR MiR-210, can simultaneously regulate expression of multiple target genes in order to fine-tune the adaptive response of cells to hypoxia. This review addresses the complex molecular regulation of MiRNAs in both physiological and pathological conditions of low oxygen adaptation and the multiple functions of hypoxamiRs in various hypoxia-associated biological processes, including apoptosis, survival, proliferation, angiogenesis, inflammation, and metabolism. From a clinical perspective, we also discuss the potential use of hypoxamiRs as new biomarkers and/or therapeutic targets in cancer and aging-associated diseases including cardiovascular and fibroproliferative disorders.
Collapse
Affiliation(s)
- Thomas Bertero
- Université Côte d'Azur, CNRS, INSERM, IRCAN, FHU-OncoAge, Nice, France
| | - Roger Rezzonico
- Université Côte d'Azur, CNRS, IPMC, FHU-OncoAge, Sophia-Antipolis, France
| | | | - Bernard Mari
- Université Côte d'Azur, CNRS, IPMC, FHU-OncoAge, Sophia-Antipolis, France.
| |
Collapse
|
134
|
Taylor DA. The use of biologics in the management of cardiovascular diseases. Curr Opin Pharmacol 2017; 33:76-80. [DOI: 10.1016/j.coph.2017.03.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 03/27/2017] [Accepted: 03/28/2017] [Indexed: 01/14/2023]
|
135
|
Nassal DM, Wan X, Liu H, Maleski D, Ramirez-Navarro A, Moravec CS, Ficker E, Laurita KR, Deschênes I. KChIP2 is a core transcriptional regulator of cardiac excitability. eLife 2017; 6. [PMID: 28263709 PMCID: PMC5338919 DOI: 10.7554/elife.17304] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 02/19/2017] [Indexed: 11/18/2022] Open
Abstract
Arrhythmogenesis from aberrant electrical remodeling is a primary cause of death among patients with heart disease. Amongst a multitude of remodeling events, reduced expression of the ion channel subunit KChIP2 is consistently observed in numerous cardiac pathologies. However, it remains unknown if KChIP2 loss is merely a symptom or involved in disease development. Using rat and human derived cardiomyocytes, we identify a previously unobserved transcriptional capacity for cardiac KChIP2 critical in maintaining electrical stability. Through interaction with genetic elements, KChIP2 transcriptionally repressed the miRNAs miR-34b and miR-34c, which subsequently targeted key depolarizing (INa) and repolarizing (Ito) currents altered in cardiac disease. Genetically maintaining KChIP2 expression or inhibiting miR-34 under pathologic conditions restored channel function and moreover, prevented the incidence of reentrant arrhythmias. This identifies the KChIP2/miR-34 axis as a central regulator in developing electrical dysfunction and reveals miR-34 as a therapeutic target for treating arrhythmogenesis in heart disease. DOI:http://dx.doi.org/10.7554/eLife.17304.001 The heart pumps blood throughout the body to provide oxygen and nourishment. To do so, proteins in the heart create electrical signals that tell the heart muscles to contract in a coordinated manner. Heart disease can cause cells to lose control of the production or activity of these proteins, creating disorganized electrical signals called arrhythmias that interfere with the heart’s ability to pump. Sometimes these arrhythmias lead to sudden death. Researchers do not know exactly what triggers these changes in the heart’s normal electrical rhythms. This has made it difficult to develop strategies to prevent these disruptions or to fix them when they occur. By studying rat and human heart cells, Nassal et al. now show that a protein called KChIP2 stops working properly during heart disease. Most importantly, because of the decreased level of KChIP2 in heart disease, KChIP2 loses the ability to restrict the production of two microRNA molecules – a role that KChIP2 was not previously known to perform. This loss of activity sets off a cascade of signals that worsens the balance of electrical activity in the heart cells, creating arrhythmias. Treatments that restored proper levels of the fully working KChIP2 protein to the heart cells or that blocked the signals set off by a lack of KChIP2 returned the electrical activity of the cells back to normal. This also stopped the development of arrhythmias. Further studies are now needed to investigate whether these treatments have the same effects in living mammals. If effective, this could ultimately lead to new treatments for heart diseases and arrhythmias. DOI:http://dx.doi.org/10.7554/eLife.17304.002
Collapse
Affiliation(s)
- Drew M Nassal
- Heart and Vascular Research Center, Department of Medicine, Case Western Reserve University, Cleveland, United States.,Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, United States
| | - Xiaoping Wan
- Heart and Vascular Research Center, Department of Medicine, Case Western Reserve University, Cleveland, United States
| | - Haiyan Liu
- Heart and Vascular Research Center, Department of Medicine, Case Western Reserve University, Cleveland, United States
| | - Danielle Maleski
- Heart and Vascular Research Center, Department of Medicine, Case Western Reserve University, Cleveland, United States
| | - Angelina Ramirez-Navarro
- Heart and Vascular Research Center, Department of Medicine, Case Western Reserve University, Cleveland, United States
| | - Christine S Moravec
- Department of Molecular Cardiology, Cleveland Clinic, Cleveland, United States
| | - Eckhard Ficker
- Heart and Vascular Research Center, Department of Medicine, Case Western Reserve University, Cleveland, United States
| | - Kenneth R Laurita
- Heart and Vascular Research Center, Department of Medicine, Case Western Reserve University, Cleveland, United States
| | - Isabelle Deschênes
- Heart and Vascular Research Center, Department of Medicine, Case Western Reserve University, Cleveland, United States.,Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, United States
| |
Collapse
|
136
|
Raso A, Dirkx E. Cardiac regenerative medicine: At the crossroad of microRNA function and biotechnology. Noncoding RNA Res 2017; 2:27-37. [PMID: 30159418 PMCID: PMC6096413 DOI: 10.1016/j.ncrna.2017.03.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 02/13/2017] [Accepted: 03/13/2017] [Indexed: 12/21/2022] Open
Abstract
There is an urgent need to develop new therapeutic strategies to stimulate cardiac repair after damage, such as myocardial infarction. Already for more than a century scientist are intrigued by studying the regenerative capacity of the heart. While moving away from the old classification of the heart as a post-mitotic organ, and being inspired by the stem cell research in other scientific fields, mainly three different strategies arose in order to develop regenerative medicine, namely; the use of cardiac stem cells, reprogramming of fibroblasts into cardiomyocytes or direct stimulation of endogenous cardiomyocyte proliferation. MicroRNAs, known to play a role in orchestrating cell fate processes such as proliferation, differentiation and reprogramming, gained a lot of attention in this context the latest years. Indeed, several research groups have independently demonstrated that microRNA-based therapy shows promising results to induce heart tissue regeneration and improve cardiac pump function after myocardial injury. Nowadays, a whole new biotechnology field has been unveiled to investigate the possibilities for efficient, safe and specific delivery of microRNAs towards the heart.
Collapse
Affiliation(s)
| | - Ellen Dirkx
- Department of Cardiology, CARIM School for Cardiovascular Disease, Maastricht University, 6229ER Maastricht, The Netherlands
| |
Collapse
|
137
|
Eliasson L, Esguerra JLS, Wendt A. Lessons from basic pancreatic beta cell research in type-2 diabetes and vascular complications. Diabetol Int 2017; 8:139-152. [PMID: 30603317 DOI: 10.1007/s13340-017-0304-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 01/08/2017] [Indexed: 12/14/2022]
Abstract
The changes in life-style with increased access of food and reduced physical activity have resulted in the global epidemic of obesity. Consequently, individuals with type 2 diabetes and cardiovascular disease have also escalated. A central organ in the development of diabetes is the pancreas, and more specifically the pancreatic beta cells within the islets of Langerhans. Beta cells have been assigned the important task of secreting insulin when blood glucose is increased to lower the glucose level. An early sign of diabetes pathogenesis is lack of first phase insulin response and reduced second phase secretion. In this review, which is based on the foreign investigator award lecture given at the JSDC meeting in Sendai in October 2016, we discuss a possible cellular explanation for the reduced first phase insulin response and how this can be influenced by lipids. Moreover, since patients with cardiovascular disease and high levels of cholesterol are often treated with statins, we summarize recent data regarding effects on statins on glucose homeostasis and insulin secretion. Finally, we suggest microRNAs (miRNAs) as central players in the adjustment of beta cell function during the development of diabetes. We specifically discuss miRNAs regarding their involvement in insulin secretion regulation, differential expression in type 2 diabetes, and potential as biomarkers for prediction of diabetes and cardiovascular complications.
Collapse
Affiliation(s)
- Lena Eliasson
- Islet Cell Exocytosis, Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Lund University, Clinical Research Centre, SUS 91-11, Box 50332, 202 13 Malmö, Sweden
| | - Jonathan Lou S Esguerra
- Islet Cell Exocytosis, Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Lund University, Clinical Research Centre, SUS 91-11, Box 50332, 202 13 Malmö, Sweden
| | - Anna Wendt
- Islet Cell Exocytosis, Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Lund University, Clinical Research Centre, SUS 91-11, Box 50332, 202 13 Malmö, Sweden
| |
Collapse
|
138
|
Gu C, Liao B, Li X, Cai L, Chen H, Li K, Yang J. Network-based collaborative filtering recommendation model for inferring novel disease-related miRNAs. RSC Adv 2017. [DOI: 10.1039/c7ra09229f] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
According to the miRNA and disease similarity network, the unknown associations are predicted by combining the known miRNA-disease association network based on collaborative filtering recommendation algorithm.
Collapse
Affiliation(s)
- Changlong Gu
- College of Information Science and Engineering
- Hunan University
- Changsha
- China
| | - Bo Liao
- College of Information Science and Engineering
- Hunan University
- Changsha
- China
| | - Xiaoying Li
- College of Information Science and Engineering
- Hunan University
- Changsha
- China
| | - Lijun Cai
- College of Information Science and Engineering
- Hunan University
- Changsha
- China
| | - Haowen Chen
- College of Information Science and Engineering
- Hunan University
- Changsha
- China
| | - Keqin Li
- Department of Computer Science
- State University of New York
- New York 12561
- USA
| | - Jialiang Yang
- Department of Genetics and Gnomic Science
- Icahn School of Medicine at Mount Sinai
- New York 10029
- USA
| |
Collapse
|
139
|
Suttamanatwong S. MicroRNAs in bone development and their diagnostic and therapeutic potentials in osteoporosis. Connect Tissue Res 2017; 58:90-102. [PMID: 26963177 DOI: 10.3109/03008207.2016.1139580] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs approximately 22 nucleotides in length. miRNAs play an important role in the posttranscriptional regulation of gene expression via translational repression and targeting messenger RNA for degradation. In vivo and in vitro evidence has established the importance of miRNAs in physiology and developmental processes such as cell proliferation, differentiation, survival and apoptosis. miRNA dysregulation is associated with the pathogenesis of cardiovascular diseases, metabolic syndromes, and degenerative diseases. An increasing number of miRNAs have been found to play an important role in bone homeostasis. In this review, the roles of miRNAs in the regulation of bone formation and resorption as well as miRNAs that regulate key transcription factors of osteogenesis are discussed. A special emphasis is given to miRNAs whose direct targets have been identified. The miRNAs that contribute to the pathogenesis of osteoporosis and their therapeutic potential are also considered.
Collapse
Affiliation(s)
- Supaporn Suttamanatwong
- a Research Unit of Herbal Medicine, Biomaterial and Material for Dental Treatment, Department of Physiology, Faculty of Dentistry , Chulalongkorn University , Bangkok , Thailand
| |
Collapse
|
140
|
Balzano F, Deiana M, Dei Giudici S, Oggiano A, Pasella S, Pinna S, Mannu A, Deiana N, Porcu B, Masala AGE, Pileri PV, Scognamillo F, Pala C, Zinellu A, Carru C, Deiana L. MicroRNA Expression Analysis of Centenarians and Rheumatoid Arthritis Patients Reveals a Common Expression Pattern. Int J Med Sci 2017; 14:622-628. [PMID: 28824293 PMCID: PMC5562112 DOI: 10.7150/ijms.18972] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 03/29/2017] [Indexed: 01/21/2023] Open
Abstract
Micro-RNA (miRNA) are a family of small non-coding ribonucleic acids that inhibits post-transcriptionally the expression of their target messenger RNA (mRNA). We are interested in studying the involvement of miRNA in longevity and autoimmune diseases. In this study we compared the different expression of seven microRNAs between human plasma healthy controls, plasma samples of centenarians and samples from patients with rheumatoid arthritis. We used the Life Technologies' protocol to quantify seven miRNAs from 62 plasma samples: 20 healthy human controls, 14 centenarians, 28 patients with rheumatoid arthritis. TaqMan MicroRNA assays were used to analyze the expression profiles of miR-125b-5p, miR-425-5p, miR-200b5p, miR-200c-3p, miR-579-3p, miR-212-3p, miR-21-5p and miR-126-3p. The relative expression of mature miRNAs was analyzed using software REST. Our results show that miR-425-5p, miR-21 and miR-212 significantly decreased in centenarians and in patients with rheumatoid arthritis compared with controls. Furthermore in this work we highlight a connection between corticosteroid treatment and miRNAs expression.
Collapse
Affiliation(s)
- Francesca Balzano
- Department of Biomedical Sciences, University of Sassari, vl. San Pietro 43b, Sassari 07100, Italy
| | - Marta Deiana
- Associazione "L'Isola dei Centenari", Via Milano 4, Sassari 07100, Italy
| | - Silvia Dei Giudici
- Istituto Zooprofilattico Sperimentale della Sardegna, Via Vienna 2, Sassari 07100, Italy
| | - Annalisa Oggiano
- Istituto Zooprofilattico Sperimentale della Sardegna, Via Vienna 2, Sassari 07100, Italy
| | - Sara Pasella
- Department of Biomedical Sciences, University of Sassari, vl. San Pietro 43b, Sassari 07100, Italy
| | - Sara Pinna
- Department of Biomedical Sciences, University of Sassari, vl. San Pietro 43b, Sassari 07100, Italy
| | - Andrea Mannu
- Department of Biomedical Sciences, University of Sassari, vl. San Pietro 43b, Sassari 07100, Italy
| | - Nicola Deiana
- Associazione "L'Isola dei Centenari", Via Milano 4, Sassari 07100, Italy
| | - Baingio Porcu
- Department of Biomedical Sciences, University of Sassari, vl. San Pietro 43b, Sassari 07100, Italy
| | - Antonio G E Masala
- Istituto Zooprofilattico Sperimentale della Sardegna, Via Vienna 2, Sassari 07100, Italy
| | - Piera V Pileri
- Istituto Zooprofilattico Sperimentale della Sardegna, Via Vienna 2, Sassari 07100, Italy
| | | | - Carlo Pala
- Internal Medicine 1, A.O.U.University of Sassari, Sassari, Italy
| | - Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, vl. San Pietro 43b, Sassari 07100, Italy
| | - Ciriaco Carru
- Department of Biomedical Sciences, University of Sassari, vl. San Pietro 43b, Sassari 07100, Italy
| | - Luca Deiana
- Department of Biomedical Sciences, University of Sassari, vl. San Pietro 43b, Sassari 07100, Italy.,Associazione "L'Isola dei Centenari", Via Milano 4, Sassari 07100, Italy
| |
Collapse
|
141
|
Sapp RM, Shill DD, Roth SM, Hagberg JM. Circulating microRNAs in acute and chronic exercise: more than mere biomarkers. J Appl Physiol (1985) 2016; 122:702-717. [PMID: 28035018 DOI: 10.1152/japplphysiol.00982.2016] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 12/14/2016] [Accepted: 12/27/2016] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) are short, noncoding RNAs that influence biological processes by regulating gene expression after transcription. It was recently discovered that miRNAs are released into the circulation (ci-miRNAs) where they are highly stable and can act as intercellular messengers to affect physiological processes. This review provides a comprehensive summary of the studies to date that have investigated the effects of acute exercise and exercise training on ci-miRNAs in humans. Findings indicate that specific ci-miRNAs are altered in response to different protocols of acute and chronic exercise in both healthy and diseased populations. In some cases, altered ci-miRNAs correlate with fitness and health parameters, suggesting causal mechanisms by which ci-miRNAs may facilitate adaptations to exercise training. However, strong data supporting such mechanisms are lacking. Thus, a purpose of this review is to guide future studies by discussing current and novel proposed roles for ci-miRNAs in adaptations to exercise training. In addition, substantial, fundamental gaps in the field need to be addressed. The ultimate goal of this research is that an understanding of the roles of ci-miRNAs in physiological adaptations to exercise training will one day translate to therapeutic interventions.
Collapse
Affiliation(s)
- Ryan M Sapp
- Department of Kinesiology, University of Maryland, College Park, Maryland
| | - Daniel D Shill
- Department of Kinesiology, University of Maryland, College Park, Maryland
| | - Stephen M Roth
- Department of Kinesiology, University of Maryland, College Park, Maryland
| | - James M Hagberg
- Department of Kinesiology, University of Maryland, College Park, Maryland
| |
Collapse
|
142
|
Biglino G, Caputo M, Rajakaruna C, Angelini G, van Rooij E, Emanueli C. Modulating microRNAs in cardiac surgery patients: Novel therapeutic opportunities? Pharmacol Ther 2016; 170:192-204. [PMID: 27902930 DOI: 10.1016/j.pharmthera.2016.11.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
This review focuses on microRNAs (miRs) in cardiac surgery, where they are emerging as potential targets for therapeutic intervention as well as novel clinical biomarkers. Identification of the up/down-regulation of specific miRs in defined groups of cardiac surgery patients can lead to the development of novel strategies for targeted treatment in order to maximise therapeutic results and minimise acute, delayed or chronic complications. MiRs could also be involved in determining the outcome independently of complications, for example in relation to myocardial perfusion and fibrosis. Because of their relevance in disease, their known sequence and pharmacological properties, miRs are attractive candidates for therapeutic manipulation. Pharmacological inhibition of individual miRs can be achieved by modified antisense oligonucleotides, referred to as antimiRs, while miR replacement can be achieved by miR mimics to increase the level of a specific miR. MiR mimics can restore the function of a lost or down-regulated miR, while antimiRs can inhibit the levels of disease-driving or aberrantly expressed miRs, thus de-repressing the expression of mRNAs targeted by the miR. The main delivery methods for miR therapeutics involve lipid-based vehicles, viral systems, cationic polymers, and intravenous or local injection of an antagomiR. Local delivery is particularly desirable for miR therapeutics and options include the development of devices specific for local delivery, light-induced antimiR, and vesicle-encapsulated miRs serving as therapeutic delivery agents able to improve intracellular uptake. Here, we discuss the potential therapeutic use of miRNAs in the context of cardiac surgery.
Collapse
Affiliation(s)
| | - Massimo Caputo
- Bristol Heart Institute, University of Bristol, Bristol, UK; RUSH University Medical Center, Chicago, IL, USA
| | - Cha Rajakaruna
- Bristol Heart Institute, University of Bristol, Bristol, UK
| | | | | | - Costanza Emanueli
- Bristol Heart Institute, University of Bristol, Bristol, UK; National Heart and Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
143
|
Jones Buie JN, Goodwin AJ, Cook JA, Halushka PV, Fan H. The role of miRNAs in cardiovascular disease risk factors. Atherosclerosis 2016; 254:271-281. [PMID: 27693002 PMCID: PMC5125538 DOI: 10.1016/j.atherosclerosis.2016.09.067] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 08/31/2016] [Accepted: 09/22/2016] [Indexed: 12/12/2022]
Abstract
Coronary artery disease and atherosclerosis are complex pathologies that develop over time due to genetic and environmental factors. Differential expression of miRNAs has been identified in patients with coronary artery disease and atherosclerosis, however, their association with cardiovascular disease risk factors, including hyperlipidemia, hypertension, obesity, diabetes, lack of physical activity and smoking, remains unclear. This review examines the role of miRNAs as either biomarkers or potential contributors to the pathophysiology of these aforementioned risk factors. It is intended to provide an overview of the published literature which describes alterations in miRNA levels in both human and animal studies of cardiovascular risk factors and when known, the possible mechanism by which these miRNAs may exert either beneficial or deleterious effects. The intent of this review is engage clinical, translational, and basic scientists to design future collaborative studies to further elucidate the potential role of miRNAs in cardiovascular diseases.
Collapse
Affiliation(s)
- Joy N Jones Buie
- Medical University of South Carolina, Department of Pathology and Laboratory Medicine, 173 Ashley Avenue, Suite CRI 605B, Charleston, United States.
| | - Andrew J Goodwin
- Medical University of South Carolina, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Charleston, United States
| | - James A Cook
- Medical University of South Carolina, Department of Neurosciences, Charleston, United States
| | - Perry V Halushka
- Medical University of South Carolina, Department of Pharmacology, Charleston, United States
| | - Hongkuan Fan
- Medical University of South Carolina, Department of Pathology and Laboratory Medicine, 173 Ashley Avenue, Suite CRI 605B, Charleston, United States
| |
Collapse
|
144
|
Pacholewska A, Mach N, Mata X, Vaiman A, Schibler L, Barrey E, Gerber V. Novel equine tissue miRNAs and breed-related miRNA expressed in serum. BMC Genomics 2016; 17:831. [PMID: 27782799 PMCID: PMC5080802 DOI: 10.1186/s12864-016-3168-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 10/18/2016] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND MiRNAs regulate multiple genes at the post-transcriptional level and therefore play an important role in many biological processes. It has been suggested that miRNA exported outside the cells contribute to inter-cellular communication. Consequently, circulating miRNAs are of particular interest and are promising biomarkers for many diseases. The number of miRNAs annotated in the horse genome is much lower compared to model organisms like human and mouse. We therefore aimed to identify novel equine miRNAs for tissue types and breed in serum. RESULTS We analysed 71 small RNA-seq libraries derived from nine tissues (gluteus medius, platysma, masseter muscle, heart, liver, cartilage, bone, total blood and serum) using miRDeep2 and miRdentify tools. Known miRNAs represented between 2.3 and 62.9 % of the reads in 71 libraries. A total of 683 novel miRNAs were identified. Breed and tissue type affected the number of miRNAs detected and interestingly, affected its average intensity. A total of 50 miRNAs in serum proved to be potential biomarkers to differentiate specific breed types, of which miR-122, miR-200, miR-483 were over-expressed and miR-328 was under-expressed in ponies compared to Warmbloods. The different miRNAs profiles, as well as the differences in their expression levels provide a foundation for more hypotheses based on the novel miRNAs discovered. CONCLUSIONS We identified 683 novel equine miRNAs expressed in seven solid tissues, blood and serum. Additionally, our approach evidenced that such data supported identification of specific miRNAs as markers of functions related to breeds or disease tissues.
Collapse
Affiliation(s)
- Alicja Pacholewska
- Department of Clinical Veterinary Medicine, Swiss Institute of Equine Medicine, Vetsuisse Faculty, University of Bern, and Agroscope, Länggassstrasse 124, 3012, Bern, Switzerland. .,Department of Clinical Research and Veterinary Public Health, Institute of Genetics, Vetsuisse Faculty, University of Bern, Bremgartenstrasse 109A, 3012, Bern, Switzerland.
| | - Núria Mach
- Animal Genetics and Integrative Biology unit (GABI), INRA, AgroParis Tech, University of Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Xavier Mata
- Animal Genetics and Integrative Biology unit (GABI), INRA, AgroParis Tech, University of Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Anne Vaiman
- Animal Genetics and Integrative Biology unit (GABI), INRA, AgroParis Tech, University of Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Laurent Schibler
- Animal Genetics and Integrative Biology unit (GABI), INRA, AgroParis Tech, University of Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Eric Barrey
- Animal Genetics and Integrative Biology unit (GABI), INRA, AgroParis Tech, University of Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Vincent Gerber
- Department of Clinical Veterinary Medicine, Swiss Institute of Equine Medicine, Vetsuisse Faculty, University of Bern, and Agroscope, Länggassstrasse 124, 3012, Bern, Switzerland
| |
Collapse
|
145
|
Gu C, Liao B, Li X, Li K. Network Consistency Projection for Human miRNA-Disease Associations Inference. Sci Rep 2016; 6:36054. [PMID: 27779232 PMCID: PMC5078764 DOI: 10.1038/srep36054] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 10/11/2016] [Indexed: 11/20/2022] Open
Abstract
Prediction and confirmation of the presence of disease-related miRNAs is beneficial to understand disease mechanisms at the miRNA level. However, the use of experimental verification to identify disease-related miRNAs is expensive and time-consuming. Effective computational approaches used to predict miRNA-disease associations are highly specific. In this study, we develop the Network Consistency Projection for miRNA-Disease Associations (NCPMDA) method to reveal the potential associations between miRNAs and diseases. NCPMDA is a non-parametric universal network-based method that can simultaneously predict miRNA-disease associations in all diseases but does not require negative samples. NCPMDA can also confirm the presence of miRNAs in isolated diseases (diseases without any known miRNA association). Leave-one-out cross validation and case studies have shown that the predictive performance of NCPMDA is superior over that of previous method.
Collapse
Affiliation(s)
- Changlong Gu
- College of Information Science and Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Bo Liao
- College of Information Science and Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Xiaoying Li
- College of Information Science and Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Keqin Li
- Department of Computer Science, State University of New York, New Paltz, New York 12561, USA
| |
Collapse
|
146
|
Enquobahrie DA, Wander PL, Tadesse MG, Qiu C, Holzman C, Williams MA. Maternal pre-pregnancy body mass index and circulating microRNAs in pregnancy. Obes Res Clin Pract 2016; 11:464-474. [PMID: 27789200 DOI: 10.1016/j.orcp.2016.10.287] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 09/18/2016] [Accepted: 10/07/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND Maternal pre-pregnancy overweight and obese status has been associated with a number of pregnancy complications and adverse offspring outcomes. Mechanisms for observed associations, however, are largely unknown. We investigated associations of pre-pregnancy body mass index with early-mid pregnancy epigenetic biomarkers, circulating microRNAs. METHODS Peripheral blood was collected from participants (16-27 weeks gestation) of two multi-racial pregnancy cohorts, the Omega Study and the Pregnancy Outcomes and Community Health Study. Plasma miRNA expression was characterised using epigenome-wide (319 miRNAs) profiling among 20 pregnant women in each cohort. Cohort-specific linear regression models that included the predictor (pre-pregnancy body mass index), the outcome (microRNA expression), and adjustment factors (maternal age, gestational age at blood collection, and race) were fit. RESULTS Expression of 27 miRNAs was positively associated with pre-pregnancy body mass index in both cohorts (p-values <0.05). A number of these differentially expressed miRNAs have previously been associated with adipogenesis (e.g. let-7d*, miR-103-2*, -130b, -146b-5-p, -29c, and -26b). Identified miRNAs as well as their experimentally validated targets participate in pathways that involve organismal injury, reproductive system disease, connective tissue disorders, cancer, cellular development, growth and proliferation. CONCLUSION Pre-pregnancy body mass index is associated with circulating miRNAs in early-mid pregnancy.
Collapse
Affiliation(s)
- Daniel A Enquobahrie
- Center for Perinatal Studies, Swedish Medical Center, Seattle, WA, USA; Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Pandora L Wander
- Department of Medicine, University of Washington, Seattle, WA, USA.
| | - Mahlet G Tadesse
- Department of Mathematics and Statistics, Georgetown University, Washington DC, USA
| | - Chunfang Qiu
- Center for Perinatal Studies, Swedish Medical Center, Seattle, WA, USA
| | - Claudia Holzman
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA
| | - Michelle A Williams
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
147
|
Pinti MV, Hathaway QA, Hollander JM. Role of microRNA in metabolic shift during heart failure. Am J Physiol Heart Circ Physiol 2016; 312:H33-H45. [PMID: 27742689 DOI: 10.1152/ajpheart.00341.2016] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 10/07/2016] [Accepted: 10/08/2016] [Indexed: 12/16/2022]
Abstract
Heart failure (HF) is an end point resulting from a number of disease states. The prognosis for HF patients is poor with survival rates precipitously low. Energy metabolism is centrally linked to the development of HF, and it involves the proteomic remodeling of numerous pathways, many of which are targeted to the mitochondrion. microRNAs (miRNA) are noncoding RNAs that influence posttranscriptional gene regulation. miRNA have garnered considerable attention for their ability to orchestrate changes to the transcriptome, and ultimately the proteome, during HF. Recently, interest in the role played by miRNA in the regulation of energy metabolism at the mitochondrion has emerged. Cardiac proteome remodeling during HF includes axes impacting hypertrophy, oxidative stress, calcium homeostasis, and metabolic fuel transition. Although it is established that the pathological environment of hypoxia and hemodynamic stress significantly contribute to the HF phenotype, it remains unclear as to the mechanistic underpinnings driving proteome remodeling. The aim of this review is to present evidence highlighting the role played by miRNA in these processes as a means for linking pathological stimuli with proteomic alteration. The differential expression of proteins of substrate transport, glycolysis, β-oxidation, ketone metabolism, the citric acid cycle (CAC), and the electron transport chain (ETC) are paralleled by the differential expression of miRNA species that modulate these processes. Identification of miRNAs that translocate to cardiomyocyte mitochondria (miR-181c, miR-378) influencing the expression of the mitochondrial genome-encoded transcripts as well as suggested import modulators are discussed. Current insights, applications, and challenges of miRNA-based therapeutics are also described.
Collapse
Affiliation(s)
- Mark V Pinti
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia; and Mitochondria, Metabolism, and Bioenergentics Working Group, Morgantown, West Virginia
| | - Quincy A Hathaway
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia; and Mitochondria, Metabolism, and Bioenergentics Working Group, Morgantown, West Virginia
| | - John M Hollander
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia; and Mitochondria, Metabolism, and Bioenergentics Working Group, Morgantown, West Virginia
| |
Collapse
|
148
|
Beermann J, Piccoli MT, Viereck J, Thum T. Non-coding RNAs in Development and Disease: Background, Mechanisms, and Therapeutic Approaches. Physiol Rev 2016; 96:1297-1325. [PMID: 27535639 DOI: 10.1152/physrev.00041.2015] [Citation(s) in RCA: 1318] [Impact Index Per Article: 146.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Advances in RNA-sequencing techniques have led to the discovery of thousands of non-coding transcripts with unknown function. There are several types of non-coding linear RNAs such as microRNAs (miRNA) and long non-coding RNAs (lncRNA), as well as circular RNAs (circRNA) consisting of a closed continuous loop. This review guides the reader through important aspects of non-coding RNA biology. This includes their biogenesis, mode of actions, physiological function, as well as their role in the disease context (such as in cancer or the cardiovascular system). We specifically focus on non-coding RNAs as potential therapeutic targets and diagnostic biomarkers.
Collapse
Affiliation(s)
- Julia Beermann
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany; and National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Maria-Teresa Piccoli
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany; and National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Janika Viereck
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany; and National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany; and National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
149
|
Tao L, Bei Y, Chen P, Lei Z, Fu S, Zhang H, Xu J, Che L, Chen X, Sluijter JPG, Das S, Cretoiu D, Xu B, Zhong J, Xiao J, Li X. Crucial Role of miR-433 in Regulating Cardiac Fibrosis. Am J Cancer Res 2016; 6:2068-2083. [PMID: 27698941 PMCID: PMC5039681 DOI: 10.7150/thno.15007] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 08/06/2016] [Indexed: 12/11/2022] Open
Abstract
Dysregulation of microRNAs has been implicated in many cardiovascular diseases including fibrosis. Here we report that miR-433 was consistently elevated in three models of heart disease with prominent cardiac fibrosis, and was enriched in fibroblasts compared to cardiomyocytes. Forced expression of miR-433 in neonatal rat cardiac fibroblasts increased proliferation and their differentiation into myofibroblasts as determined by EdU incorporation, α-SMA staining, and expression levels of fibrosis-associated genes. Conversely, inhibition of miR-433 exhibited opposite results. AZIN1 and JNK1 were identified as two target genes of miR-433. Decreased level of AZIN1 activated TGF-β1 while down-regulation of JNK1 resulted in activation of ERK and p38 kinase leading to Smad3 activation and ultimately cardiac fibrosis. Importantly, systemic neutralization of miR-433 or adeno-associated virus 9 (AAV9)-mediated cardiac transfer of a miR-433 sponge attenuated cardiac fibrosis and ventricular dysfunction following myocardial infarction. Thus, our work suggests that miR-433 is a potential target for amelioration of cardiac fibrosis.
Collapse
|
150
|
Zhan C, Li C, Zhang H, Tang H, Ji F, Qiao SC, Xu WD, Wang ZW. MicroRNA-150 upregulation reduces osteosarcoma cell invasion and metastasis by downregulating Ezrin. Oncol Lett 2016; 12:3457-3462. [PMID: 27900020 DOI: 10.3892/ol.2016.5046] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Accepted: 08/05/2015] [Indexed: 12/12/2022] Open
Abstract
The present study aimed to investigate the effect of microRNA-150 (miRNA/miR-150) in osteosarcoma (OS) cell invasion and metastasis by the regulation of Ezrin. To compare the differences in the expression of miR-150 and Ezrin, cell models of OS metastasis were established by exogenous transfection of miR-150 on the basis of different expression levels of miR-150. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was used to estimate these expression levels. Ezrin expression was detected by western blot assay. Methylthiazolyldiphenyl-tetrazolium bromide assay was performed to determine cells proliferation. Cell invasion and migration were measured in vitro by Transwell migration assays. Detection of apoptosis adopted flow cytometry. The results of RT-qPCR showed that the miR-150 expression in OS F5M2 cells was significantly increased following exogenous transfection of miR-150 mimics, and the expression of miR-150 was positively correlated with the concentration of the miR-150 mimics. Western blot assay indicated that the Ezrin expression in the F5M2 cells was decreased with the exogenous overexpression of miR-150. Additionally, Transwell assays revealed that the overexpression of miR-150 significantly suppressed the invasion and metastasis ability of the F5M2 cells. miR-150 upregulation may reduce OS cell invasion and metastasis by downregulating the expression of Ezrin.
Collapse
Affiliation(s)
- Ce Zhan
- Department of Orthopedics, Changhai Hospital, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Cheng Li
- Department of Orthopedics, Changhai Hospital, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Hao Zhang
- Department of Orthopedics, Changhai Hospital, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Hao Tang
- Department of Orthopedics, Changhai Hospital, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Fang Ji
- Department of Orthopedics, Changhai Hospital, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Su-Chi Qiao
- Department of Orthopedics, Changhai Hospital, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Wei-Dong Xu
- Department of Orthopedics, Changhai Hospital, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Zhi-Wei Wang
- Department of Orthopedics, Changhai Hospital, The Second Military Medical University, Shanghai 200433, P.R. China
| |
Collapse
|