1
|
Wiyono AV, Ardinal AP, Raharjo PP. Unraveling the significance of innate inflammation in vascular disease. Int Rev Immunol 2025:1-16. [PMID: 40255209 DOI: 10.1080/08830185.2025.2489346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 02/06/2025] [Accepted: 03/31/2025] [Indexed: 04/22/2025]
Abstract
Atheroma formation is initiated by the activation of endothelial and smooth muscle cells, as well as immune cells, including neutrophils, lymphocytes, monocytes, macrophages, and dendritic cells. Monocytes, macrophages, and neutrophils are the innate immune cells that provide a rapid initial line of defence against vascular disease. These cells have a short lifespan and cannot retain memories, making them potential therapeutic targets for the inflammatory process associated with atherosclerosis. In addition, macrophages comprise the majority of vessel wall infiltrates and are, therefore, implicated in all stages of atherosclerosis progression. Neutrophils are the most common type of leukocyte found in circulation, and their high levels of matrix-degrading protease explain their significance in fibrous cap destabilization. However, the activation of immune cells becomes more complex by various microenvironmental stimuli and cytokines, which ultimately transform immune cells into their pro-inflammatory state. Different types of macrophage subsets with distinct functions in inflammation, such as M1 macrophages, cause an increase in pro-inflammatory cytokines and produce reactive oxygen species and nitric oxide, further worsening the disease. This review aims to shed light on immune-mediated inflammation in cardiovascular disease by focusing on the role of macrophage subsets in vascular inflammation and plaque stability, as well as the interaction between neutrophils and monocyte-macrophages.
Collapse
Affiliation(s)
- Alice Valeria Wiyono
- Faculty of Life Sciences & Medicine, King's College London, London, UK
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Padjadjaran, Rumah Sakit Umum Pusat Hasan Sadikin, Bandung, Indonesia
| | | | - Pradana Pratomo Raharjo
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Padjadjaran, Rumah Sakit Umum Pusat Hasan Sadikin, Bandung, Indonesia
| |
Collapse
|
2
|
Ardinal AP, Wiyono AV, Estiko RI. Unveiling the therapeutic potential of miR-146a: Targeting innate inflammation in atherosclerosis. J Cell Mol Med 2024; 28:e70121. [PMID: 39392102 PMCID: PMC11467738 DOI: 10.1111/jcmm.70121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 09/12/2024] [Accepted: 09/16/2024] [Indexed: 10/12/2024] Open
Abstract
Atherosclerosis is the foremost vascular disease, precipitating debilitating complications. Although therapeutic strategies have historically focused on reducing cholesterol deposition, recent insights emphasize the pivotal role of inflammation. Innate inflammation significantly contributes to plaque instability and rupture, underscoring the need for intervention across all disease stages. Numerous studies have highlighted the therapeutic potential of targeting innate immune pathways in atherosclerosis, revealing significant advancements in understanding the molecular mechanisms underlying inflammatory processes within arterial lesions. Notably, research has demonstrated that the modulation of microRNA-146a (miR-146a) expression impacts innate inflammation, effectively halts atherosclerosis progression, and enhances plaque stability by targeting interleukin-1 receptor-associated kinase (IRAK) and activating TNF receptor-associated factor 6 (TRAF6), a signalling pathway involving toll-like receptors (TLRs). Understanding the intricate mechanisms involved is crucial. This study provides a comprehensive analysis of the evidence and underlying mechanisms through which miR-146a exerts its effects. Integrating these findings into clinical practice may herald a transformative era in managing atherosclerotic cardiovascular disease.
Collapse
|
3
|
Xu T, Zhang Y, Liao G, Xuan H, Yin J, Bao J, Liu Y, Li D. Luteolin Pretreatment Ameliorates Myocardial Ischemia/Reperfusion Injury by lncRNA-JPX/miR-146b Axis. Anal Cell Pathol (Amst) 2023; 2023:4500810. [PMID: 38077523 PMCID: PMC10710365 DOI: 10.1155/2023/4500810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/03/2023] [Accepted: 11/13/2023] [Indexed: 12/18/2023] Open
Abstract
Background In the present study, we aimed to find out whether luteolin (Lut) pretreatment could ameliorate myocardial ischemia/reperfusion (I/R) injury by regulating the lncRNA just proximal to XIST (JPX)/microRNA-146b (miR-146b) axis. Methods We established the models in vitro (HL-1 cells) and in vivo (C57BL/6J mice) to certify the protection mechanism of Lut pretreatment on myocardial I/R injury. Dual luciferase reporter gene assay was utilized for validating that JPX could bind to miR-146b. JPX and miR-146b expression levels were determined by RT-qPCR. Western blot was utilized to examine apoptosis-related protein expression levels, including cleaved caspase-9, caspase-9, cleaved caspase-3, caspase-3, Bcl-2, Bax, and BAG-1. Apoptosis was analyzed by Annexin V-APC/7-AAD dualstaining, Hoechst 33342 staining, as well as flow cytometry. Animal echocardiography was used to measure cardiac function (ejection fraction (EF) and fractional shortening (FS) indicators). Results miR-146b was demonstrated to bind and recognize the JPX sequence site by dual luciferase reporter gene assay. The expression level of miR-146b was corroborated to be enhanced by H/R using RT-qPCR (P < 0.001 vs. Con). Moreover, JPX could reduce the expression of miR-146b, whereas inhibiting JPX could reverse the alteration (P < 0.001 vs. H/R, respectively). Western blot analysis demonstrated that Lut pretreatment increased BAG-1 expression level and Bcl-2/Bax ratio, but diminished the ratio of cleaved caspase 9/caspase 9 and cleaved caspase 3/caspase 3 (P < 0.001 vs. H/R, respectively). Moreover, the cell apoptosis change trend, measured by Annexin V-APC/7-AAD dualstaining, Hoechst 33342 staining, along with flow cytometry, was consistent with that of apoptosis-related proteins. Furthermore, pretreatment with Lut improved cardiac function (EF and FS) (P < 0.001 vs. I/R, respectively), as indicated in animal echocardiography. Conclusion Our results demonstrated that in vitro and in vivo, Lut pretreatment inhibited apoptosis via the JPX/miR-146b axis, ultimately improving myocardial I/R injury.
Collapse
Affiliation(s)
- Tongda Xu
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yuanyuan Zhang
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Gege Liao
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Haochen Xuan
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Jie Yin
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Jieli Bao
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yang Liu
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Dongye Li
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
4
|
The Emerging Role of Noncoding RNA Regulation of the Ferroptosis in Cardiovascular Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3595745. [PMID: 36187333 PMCID: PMC9519351 DOI: 10.1155/2022/3595745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 09/09/2022] [Indexed: 11/18/2022]
Abstract
Cardiovascular disease (CVD) is a significant public health issue due to its high prevalence and considerable contribution to the global disease burden. Recent studies suggest that genetic factors, including noncoding RNAs, have an important role in the progression of CVD. Noncoding RNA plays a critical role in genetic programming and gene regulation during development. Ferroptosis is a form of iron-dependent regulated cell death (RCD), which is mainly caused by increased lipid hydroperoxide and redox imbalance. Ferroptosis is essentially different from other forms of RCD in morphology and mechanism, such as apoptosis, autophagic cell death, pyroptosis, and necroptosis. Much evidence suggested ferroptosis is involved in the development of various CVDs, especially in cardiac ischemia/reperfusion injury, heart failure, and aortic dissection. Here, we review the latest findings based on noncoding RNA regulation of ferroptosis and its involvement in the pathogenesis of CVD and related treatments, aimed at providing insights into the impact of noncoding RNA regulation of ferroptosis for CVD.
Collapse
|
5
|
Wang X, Ren L, Chen S, Tao Y, Zhao D, Wu C. Long non-coding RNA MIR4435-2HG/microRNA-125a-5p axis is involved in myocardial ischemic injuries. Bioengineered 2022; 13:10707-10720. [PMID: 35475469 PMCID: PMC9208505 DOI: 10.1080/21655979.2022.2051259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
This study aimed to investigate whether and how long non-coding RNA (lncRNA) MIR4435-2 host gene (MIR4435-2HG) involved in acute myocardial ischemia/reperfusion (I/R). Blood samples were collected from acute myocardial infarction (AMI) patients to detect MIR4435-2HG expression. In vivo myocardial I/R mice model and in vitro H2O2-induced oxidative stress model were established. Echocardiography, TUNEL assay and lactate dehydrogenase (LDH) detection were performed to assess heart infarction and myocardium apoptosis. Relationship among microRNA-125a-5p (miR-125a-5p), MIR4435-2HG and Mitochondrial fission protein 1 (MTFP1) was predicted by Targetscan and verified by luciferase reporter assay. MIR4435-2HG was notably upregulated in AMI patients, myocardial I/R mice and H2O2-treated cells. Knockdown of MIR4435-2HG notably alleviated infraction volume, ejection fraction (EF) and fractional shortening (FS) levels, cell apoptosis portion and pro-apoptotic cleaved-caspase-3 and Cyt c expression caused by myocardial I/R and oxidative stress, as well as improved cardiomyocytes viability. Transfection with miR-125a-5p alleviated MIR4435-2HG-caused cardiomyocytes apoptosis during oxidative stress. MiR-125a-5p overexpression decreased luciferase activity of the wild-type MIR4435-2HG compared with the mutated MIR4435-2HG. The expression levels of MTFP1 were elevated in myocardium from MI mice model and H2O2-treated AC16 cardiomyocytes. In addition, miR-125a-5p overexpression inhibited MTFP1 expression, and could stimulate the wild-type MTFP1 promoter luciferase activity but not the mutated one. Our findings revealed the role of MIR4435-2HG in MI-induced myocardium injury and cardiomyocytes apoptosis, disclosed a novel MIR4435-2HG/miR-125a-5p regulatory axis during myocardial I/R, and thus identified a potential target for the therapy of myocardial IR injury.
Collapse
Affiliation(s)
- Xiuling Wang
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang110001, Liaoning Province, China
| | - Lina Ren
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang110001, Liaoning Province, China
| | - Shuai Chen
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang110001, Liaoning Province, China
| | - Yanli Tao
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang110001, Liaoning Province, China
| | - Dandan Zhao
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang110001, Liaoning Province, China
| | - Chunwei Wu
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang110001, Liaoning Province, China
| |
Collapse
|
6
|
Wang Y, Tan J, Wang L, Pei G, Cheng H, Zhang Q, Wang S, He C, Fu C, Wei Q. MiR-125 Family in Cardiovascular and Cerebrovascular Diseases. Front Cell Dev Biol 2021; 9:799049. [PMID: 34926475 PMCID: PMC8674784 DOI: 10.3389/fcell.2021.799049] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 11/11/2021] [Indexed: 02/05/2023] Open
Abstract
Cardiovascular and cerebrovascular diseases are a serious threaten to the health of modern people. Understanding the mechanism of occurrence and development of cardiovascular and cerebrovascular diseases, as well as reasonable prevention and treatment of them, is a huge challenge that we are currently facing. The miR-125 family consists of hsa-miR-125a, hsa-miR-125b-1 and hsa-miR-125b-2. It is a kind of miRNA family that is highly conserved among different species. A large amount of literature shows that the lack of miR-125 can cause abnormal development of the cardiovascular system in the embryonic period. At the same time, the miR-125 family participates in the occurrence and development of a variety of cardiovascular and cerebrovascular diseases, including myocardial ischemia, atherosclerosis, ischemia-reperfusion injury, ischemic stroke, and heart failure directly or indirectly. In this article, we summarized the role of the miR-125 family in the development and maturation of cardiovascular system, the occurrence and development of cardiovascular and cerebrovascular diseases, and its important value in the current fiery stem cell therapy. In addition, we presented this in the form of table and diagrams. We also discussed the difficulties and challenges faced by the miR-125 family in clinical applications.
Collapse
Affiliation(s)
- Yang Wang
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, China
| | - Jing Tan
- Department of Ultrasound Medicine, Binzhou People's Hospital, Binzhou, China
| | - Lu Wang
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, China
| | - Gaiqin Pei
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, China
| | - Hongxin Cheng
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, China
| | - Qing Zhang
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, China
| | - Shiqi Wang
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, China
| | - Chengqi He
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, China
| | - Chenying Fu
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,Aging and Geriatric Mechanism Laboratory, West China Hospital, Sichuan University, Chengdu, China
| | - Quan Wei
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, China
| |
Collapse
|
7
|
MicroRNA-590-3p relieves hypoxia/reoxygenation induced cardiomyocytes apoptosis and autophagy by targeting HIF-1α. Exp Ther Med 2021; 22:1077. [PMID: 34447470 PMCID: PMC8355641 DOI: 10.3892/etm.2021.10511] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 06/21/2021] [Indexed: 12/12/2022] Open
Abstract
Autophagy and apoptosis are key factors in myocardial ischemia/reperfusion (I/R) injury. MicroRNAs (miRNAs or miRs) participate in occurrence and development of myocardial I/R injury by regulating autophagy and apoptosis. The purpose of the present study was to investigate the role of miR-590-3p in the regulation of autophagy and apoptosis in hypoxia/reoxygenation (H/R)-treated cardiomyocytes. Following 6 h hypoxia and 6 h reoxygenation in primary rat cardiomyocytes, miR-590-3p was downregulated. Transfection of miR-590-3p mimic inhibited the increased autophagy and apoptosis following H/R treatment. Subsequent experiments demonstrated that miR-590-3p regulated induction of autophagy and apoptosis by targeting hypoxia inducible factor (HIF)-1α. Forced expression of HIF-1α rescued the protective effect of miR-590-3p on H/R-induced cardiomyocytes. In summary, the present study showed that miR-590-3p exhibited a protective effect on H/R-induced cardiomyocyte injury and may be a novel target for the treatment of myocardial ischemia disease.
Collapse
|
8
|
Wu Q, Shang Y, Bai Y, Wu Y, Wang H, Shen T. Sufentanil preconditioning protects against myocardial ischemia/reperfusion injury via miR-125a/DRAM2 axis. Cell Cycle 2021; 20:383-391. [PMID: 33475463 DOI: 10.1080/15384101.2021.1875668] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
This project aimed to investigate the protective mechanism of sufentanil pretreatment on myocardial ischemia-reperfusion injury (IRI). An in vivo rat model of myocardial IRI and an in vitro cultured cardiomyocyte model of hypoxia-reoxygenation (H/R) were used to confirm the anti-oxidation and anti-autophagy effects of sufentanil. The interaction between miR-125a and damage-regulated autophagy regulator 2 (DRAM2) was verified by luciferase reporter assay. We showed that pretreatment with sufentanil suppressed myocardial damage caused by IRI in rats by inhibiting oxidative stress and mitochondrial autophagy. Furthermore, the cardioprotective mechanism of sufentanil was mediated by miR-125a. MiR-125a targeted DRAM2 to ameliorate cardiomyocyte autophagy and oxidative injury following H/R treatment. In conclusion, our results demonstrated that sufentanil pretreatment produced a protective effect against myocardial IRI via regulating miR-125a/DRAM2 signaling axis.
Collapse
Affiliation(s)
- Qiaoling Wu
- Department of Anesthesiology, The First Hospital Affiliated to Jinzhou Medical University , Jinzhou, P.R. China
| | - You Shang
- Department of Anesthesiology, The First Hospital Affiliated to Jinzhou Medical University , Jinzhou, P.R. China
| | - Yanli Bai
- Department of Anesthesiology, The First Hospital Affiliated to Jinzhou Medical University , Jinzhou, P.R. China
| | - Yuanyuan Wu
- Department of Anesthesiology, The First Hospital Affiliated to Jinzhou Medical University , Jinzhou, P.R. China
| | - Hao Wang
- Department of Anesthesiology, The First Hospital Affiliated to Jinzhou Medical University , Jinzhou, P.R. China
| | - Tu Shen
- Department of Anesthesiology, The First Hospital Affiliated to Jinzhou Medical University , Jinzhou, P.R. China
| |
Collapse
|
9
|
Therapeutic Value of miRNAs in Coronary Artery Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8853748. [PMID: 33953838 PMCID: PMC8057887 DOI: 10.1155/2021/8853748] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 01/25/2021] [Accepted: 03/29/2021] [Indexed: 12/14/2022]
Abstract
Atherosclerotic ischemic coronary artery disease (CAD) is a significant community health challenge and the principal cause of morbidity and mortality in both developed and developing countries for all ethnic groups. The progressive chronic coronary atherosclerosis is the main underlying cause of CAD. Although enormous progress occurred in the last three decades in the management of cardiovascular diseases, the prevalence of CAD continues to increase worldwide, indicating the need for discovery of deeper molecular insights of CAD mechanisms, biomarkers, and innovative therapeutic targets. Recently, several research groups established that microRNAs essentially regulate various cardiovascular development and functions, and a deregulated cardiac enriched microRNA profile plays a vital role in the pathogenesis of CAD and its biological aging. Numerous studies established that over- or downregulation of a single miRNA gene by ago-miRNA or anti-miRNA is enough to modify the CAD disease process, significantly prevent age-dependent cardiac cell death, and markedly improve cardiac function. In the light of more recent experimental and clinical evidences, we briefly reviewed and discussed the involvement of miRNAs in CAD and their possible diagnostic/therapeutic values. Moreover, we also focused on the role of miRNAs in the initiation and progression of the atherosclerosis plaque as the strongest risk factor for CAD.
Collapse
|
10
|
Zhang J, Zhang T, Zhang W, Zou C, Zhang Q, Ma X, Zhu Y. Circular RNA-DENND4C in H9c2 cells relieves OGD/R-induced injury by down regulation of microRNA-320. Cell Cycle 2020; 19:3074-3085. [PMID: 33090893 DOI: 10.1080/15384101.2020.1831253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Ischemic heart disease (IHD) is one of the most deadly diseases worldwide. To detect the regulatory mechanism, the circular RNA (circRNA)-differentially expressed in normal cells and neoplasia domain containing 4 C (DENND4C) was explored in the H9c2 cells. The circRNA-DENND4C overexpressing plasmid, si-circRNA-DENND4C and miR-320 mimic were transfected into the H9c2 cells and treated with OGD/R stimulation. We took CCK-8 method, Annexin V-FITC/PI-flow cytometer to search for viability and apoptotic ability. With the help of qRT-PCR and western blot, the expression of circRNA-DENND4C and miR-320, as well as the Bax, Cleaved PARP/caspase 3 and signal proteins were separately determined. Regulation of circRNA-DENND4C and miR-320 was confirmed by dual-luciferase reporter assay. OGD/R induced suppression of cell viability, but enhancement of apoptosis and block of ERK and mTOR pathways. Moreover, circRNA-DENND4C was up-regulated after OGD/R stimulation and augmented OGD/R-stimulated damage while circRNA-DENND4C silencing displayed opposite influences. miR-320 was negatively controlled and targeted by the circRNA-DENND4C.The overexpressed miR-320 impeded the effects of circRNA-DENND4C. Besides, circRNA-DENND4C relieved the suppression of ERK and mTOR pathways caused by OGD/R stimulation, and all promoting impacts of circRNA-DENND4C were reversed by the miR-320 mimic. Overexpressed circRNA-DENND4C in H9c2 cells attenuated OGD/R-induced injuries by the down-regulation of miR-320 through the ERK and mTOR activation.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Cardiac Surgery, Shandong Provincial Hospital Affiliated to Shandong University , Jinan, China
| | - Tao Zhang
- Department of Cardiac Surgery, Shandong Provincial Hospital Affiliated to Shandong University , Jinan, China
| | - Wenlong Zhang
- Department of Cardiac Surgery, Shandong Provincial Hospital Affiliated to Shandong University , Jinan, China
| | - Chengwei Zou
- Department of Cardiac Surgery, Shandong Provincial Hospital Affiliated to Shandong University , Jinan, China
| | - Qian Zhang
- Department of Cardiac Surgery, Shandong Provincial Hospital Affiliated to Shandong University , Jinan, China
| | - Xiaochun Ma
- Department of Cardiac Surgery, Shandong Provincial Hospital Affiliated to Shandong University , Jinan, China
| | - Yanhui Zhu
- Department of Cardiac Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University , Jinan, China
| |
Collapse
|
11
|
RiPerC Attenuates Cerebral Ischemia Injury through Regulation of miR-98/PIK3IP1/PI3K/AKT Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6454281. [PMID: 33082912 PMCID: PMC7559836 DOI: 10.1155/2020/6454281] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 09/11/2020] [Accepted: 09/19/2020] [Indexed: 02/06/2023]
Abstract
Background Cerebral ischemic stroke is a refractory disease which seriously endangers human health. Remote ischemic perconditioning (RiPerC) by which the sublethal ischemic stimulus is administered during the ischemic event is beneficial after an acute stroke. However, the regulatory mechanism of RiPerC that relieves cerebral ischemic injury is still not completely clear. Methods In the present study, we investigated the regulatory mechanism of RiPerC in a rat model of ischemia induced by the middle cerebral artery occlusion (MCAO). Forty-eight adult male Sprague-Dawley (SD) rats were injected intracerebroventricularly with miR-98 agomir, miR-98 antagomir, or their negative controls (agomir-NC, antagomir-NC) 2 h before MCAO or MCAO+RiPerC followed by animal behavior tests and infraction volume measurement at 24 h after MCAO. The expression of miR-98, PIK3IP1, and tight junction proteins in rat hippocampus and cerebral cortex tissues was detected by quantitative polymerase chain reaction (qPCR) and Western blot (WB). Enzyme-linked immunosorbent assay (ELISA) was used to assess the IL-1β, IL-6, and TNF-α levels in the rat serum. Results The results showed that in MCAO group, the expression of PIK3IP1 was upregulated, but decreased after RiPerC treatment. Then, we found that PIK3IP1 was a potential target of miR-98. Treatment with miR-98 agomir decreased the infraction volume, reduced brain edema, and improved neurological functions compared to control rats. But treating with miR-98 antagomir in RiPerC group, the protective effect on cerebral ischemia injury was canceled. Conclusion Our finding indicated that RiPerC inhibited the MCAO-induced expression of PIK3IP1 through upregulated miR-98, thereby reducing the apoptosis induced by PIK3IP1 through the PI3K/AKT signaling pathway, thus reducing the cerebral ischemia-reperfusion injury.
Collapse
|
12
|
Niu S, Xu L, Yuan Y, Yang S, Ning H, Qin X, Xin P, Yuan D, Jiao J, Zhao Y. Effect of down-regulated miR-15b-5p expression on arrhythmia and myocardial apoptosis after myocardial ischemia reperfusion injury in mice. Biochem Biophys Res Commun 2020; 530:54-59. [PMID: 32828315 DOI: 10.1016/j.bbrc.2020.06.111] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 06/22/2020] [Indexed: 10/23/2022]
Abstract
In this study, the regulation of miR-15b-5p on myocardial ischemia reperfusion (I/R) injury-induced arrhythmia and myocardial apoptosis was investigated in mice. We observed the change in miR-15b-5p expression after mice suffered from myocardial I/R injury and the change in myocardial injury, infarct size, apoptosis, tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), superoxide dismutase (SOD) and malondialdehyde (MDA) after down-regulation of miR-15b-5p expression. The negative regulation of miR-15b-5p to KCNJ2 as well as whether cardioprotective effect formed by miR-15b-5p down-regulation relied on the increase of KNCJ2 expression were measured by dual-luciferase reporter assay system. miR-15b-5p expression increased and KCNJ2 mRNA and protein expressions decreased after myocardial ischemia reperfusion (all P < 0.05). miR-15b-5p negatively regulated KCNJ2 in a targeted way. Down-regulating miR-15b-5p expression or increasing KCNJ2 expression significantly decreased the incidence of arrhythmia, infarct size and apoptosis after myocardial I/R and lowered MDA content in the myocardial tissue as well as IL-6 and TNF-α content in the blood (all P < 0.05). KCNJ2 gene knockout reversed the above cardioprotective effect formed by miR-15b-5p down-regulation (P < 0.05). Down-regulating miR-15b-5p expression or up-regulating KCNJ2 expression improves arrhythmia after mice suffered from myocardial I/R injury and inhibits myocardial apoptosis.
Collapse
Affiliation(s)
- Siquan Niu
- Department of Cardiology, The 7th People's Hospital of Zhengzhou, Zhengzhou 450000, He'nan Province, China.
| | - Liang Xu
- Department of Cardiology, The 7th People's Hospital of Zhengzhou, Zhengzhou 450000, He'nan Province, China.
| | - Yiqiang Yuan
- Department of Cardiology, The 7th People's Hospital of Zhengzhou, Zhengzhou 450000, He'nan Province, China.
| | - Shaohua Yang
- Department of Cardiology, The 7th People's Hospital of Zhengzhou, Zhengzhou 450000, He'nan Province, China.
| | - Hongjie Ning
- Department of Cardiology, The 7th People's Hospital of Zhengzhou, Zhengzhou 450000, He'nan Province, China.
| | - Xutan Qin
- Department of Cardiology, The 7th People's Hospital of Zhengzhou, Zhengzhou 450000, He'nan Province, China.
| | - Pengcheng Xin
- Department of Cardiology, The 7th People's Hospital of Zhengzhou, Zhengzhou 450000, He'nan Province, China.
| | - Dongdong Yuan
- Department of Cardiology, The 7th People's Hospital of Zhengzhou, Zhengzhou 450000, He'nan Province, China.
| | - Jingmei Jiao
- Department of Cardiology, The 7th People's Hospital of Zhengzhou, Zhengzhou 450000, He'nan Province, China.
| | - Yujie Zhao
- Department of Cardiology, The 7th People's Hospital of Zhengzhou, Zhengzhou 450000, He'nan Province, China.
| |
Collapse
|
13
|
MiR-433 Regulates Myocardial Ischemia Reperfusion Injury by Targeting NDRG4 Via the PI3K/Akt Pathway. Shock 2020; 54:802-809. [PMID: 32187107 DOI: 10.1097/shk.0000000000001532] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND AND PURPOSE Myocardial ischemia reperfusion (IR) injury is a serious issue in the treatment of myocardial infarction. MiR-433 is upregulated in myocardial IR injury, but its specific effects remain unclear. In this study, we explored the effect and mechanism of miR-433 in myocardial IR injury. METHODS The expression of miR-433 was measured by qRT-PCR. H9c2 cells were transfected with miR-433 mimic and inhibitor after exposure to HR, respectively. Cell viability was detected by MTT. Cell apoptosis was measured by flow cytometry. Protein expression was assessed by western blot. Dual-luciferase reporter assay was performed to assess the target reaction between miR-433 and NDRG4. In vivo rat model of IR was used, and antagomiR-433 was injected to IR rats. RESULTS The qRT-PCR results showed that miR-433 expression increased in H9c2 cardiomyocytes after exposure to HR. Transfection with miR-433 inhibitor significantly increased cell viability, reduced LDH and apoptosis, downregulated Bax level, and upregulated Bcl-2 level. In contrast, the miR-433 mimic significantly augmented the HR-induced effects. Dual-luciferase reporter assay and western blot analysis suggested that miR-433 directly targeted NDRG4. NDRG4 silencing abrogated the protection of miR-433 inhibition on HR injury in H9c2 cells. It also reversed PI3K/Akt pathway activation that was induced by miR-433 inhibition. MiR-433 inhibition significantly decreased CK-MB and LDH serum level in IR rats. And NDRG4, p-PI3K, and p-Akt protein expression was elevated by antagomiR-433 injection in vivo. CONCLUSION MiR-433 regulated myocardial IR injury by targeting NDRG4 and modulating PI3K/Akt signal pathway.
Collapse
|
14
|
Li KS, Jiang WP, Li QC, Zhang HW, Bai Y, Zhang X, Li HY. MiR-29a in mesenchymal stem cells inhibits FSTL1 secretion and promotes cardiac myocyte apoptosis in hypoxia-reoxygenation injury. Cardiovasc Pathol 2019; 46:107180. [PMID: 31945680 DOI: 10.1016/j.carpath.2019.107180] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/07/2019] [Accepted: 11/11/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are under consideration for myocardial ischemia-reperfusion (I/R) injury therapy, but their mechanism remains to be evaluated. In this article, we aimed to study the effects of the miR-29a/follistatin-like 1 axis in bone marrow-derived mesenchymal stem cells on modulating myocyte apoptosis after hypoxia-reoxygenation (H/R) injury. METHODS An in vitro myocardial ischemia-reperfusion injury model of H9c2 cells was developed by hypoxia-reoxygenation injury. The mRNA levels of follistatin-like 1, Bcl-2, Bax, and miR-29a and the protein levels of Bcl-2, Bax, cleaved caspase-3, and components of the JAK2/STAT3 pathway were detected by qRT-PCR and western blotting, respectively. Secretion of follistatin-like 1 was evaluated by enzyme-linked immunosorbent assay. Cell apoptosis was evaluated by flow cytometry. The interaction between miR-29a and follistatin-like 1 was evaluated by dual luciferase reporter assay. RESULTS MiR-29a suppressed the expression and secretion of follistatin-like 1 in bone marrow-derived mesenchymal stem cells. Overexpression of follistatin-like 1 in bone marrow-derived mesenchymal stem cells decreased apoptosis of myocytes induced by hypoxia-reoxygenation. Cell apoptosis in myocytes was promoted by conditioned medium from bone marrow-derived mesenchymal stem cells with ectopic miR-29a expression. Conditioned medium of miR-29a-overexpressing bone marrow-derived mesenchymal stem cells inhibited the JAK2/STAT3 pathway in myocytes to promote apoptosis of myocytes. CONCLUSIONS MiR-29a in bone marrow-derived mesenchymal stem cells inhibits follistatin-like 1 secretion and promotes myocyte apoptosis by suppressing the JAK2/STAT3 pathway in hypoxia-reoxygenation injury.
Collapse
Affiliation(s)
- Kun-Sheng Li
- Department of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing 210008, Jiangsu Province, PR China
| | - Wei-Peng Jiang
- Department of Cardiology, Shenzhen University General Hospital, Shenzhen 518000, Guangdong Province, PR China
| | - Qiu-Chang Li
- Puyang Medical College, Shangyang Road and Wenyan Street, Puyang 457000, Henan Province, PR China
| | - Hao-Wen Zhang
- Second Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, PR China
| | - Yang Bai
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Road, Wuhan 430030, Hubei Province, PR China
| | - Xia Zhang
- Department of Geratology, The First Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou 310003, Zhejiang Province, PR China
| | - Hai-Ying Li
- Department of Cardiology, Shenzhen University General Hospital, Shenzhen 518000, Guangdong Province, PR China.
| |
Collapse
|
15
|
Neto-Neves EM, Pinheiro LC, Nogueira RC, Portella RL, Batista RI, Tanus-Santos JE. Sodium nitrite improves hypertension-induced myocardial dysfunction by mechanisms involving cardiac S-nitrosylation. J Mol Cell Cardiol 2019; 134:40-50. [DOI: 10.1016/j.yjmcc.2019.06.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 06/14/2019] [Accepted: 06/17/2019] [Indexed: 12/15/2022]
|
16
|
Li Y, Zhu X, Liu X, Du A, Yu B. miR-200a mediates protection of thymosin β-4 in cardiac microvascular endothelial cells as a novel mechanism under hypoxia-reoxygenation injury. J Cell Biochem 2019; 120:19098-19106. [PMID: 31265170 DOI: 10.1002/jcb.29237] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Revised: 08/22/2017] [Accepted: 08/24/2017] [Indexed: 12/29/2022]
Abstract
Thymosin β-4 (Tβ4) is a ubiquitous protein, which has been suggested to regulate multiple cell signal pathways and a variety of cellular functions. However, the role Tβ4 plays in the cardiac microvascular endothelial cells (CMECs) under myocardial ischemia/reperfusion injury is currently unknown. Here we investigated the effects of Tβ4 on hypoxia/reoxygenation (H/R) induced CMECs injury and its potential molecular mechanism. Cultured CMECs were positively identified by flow cytometry using antibody against CD31 and VWF/Factor VIII, which are constitutively expressed on the surface of CMECs. Then the reduced level of Tβ4 was detected in H/R-CMECs by a real-time quantitative polymerase chain reaction. To determine the effects of Tβ4 on H/R-CMECs, we transfected the overexpression or silence vector of Tβ4 into CMECs under H/R condition. Our results indicated that H/R treatment could reduce proliferation, increased apoptosis, adhesion, and reactive oxygen species (ROS) production in CMECs, which were attenuated by Tβ4 overexpression or aggravated by Tβ4 silencing, implying Tβ4 is able to promote CMECs against H/R-induced cell injury. Furthermore, the microRNA-200a (miR-200a) level was also increased by Tβ4 in H/R-CMECs or reduced by Tβ4 small interfering RNA. To investigated the mechanism of protective effects of Tβ4 on CMECs injury, the miR-200a inhibitor was transfected into H/R-CMECs. The results indicated that inhibition of miR-200a inversed the protection of Tβ4 on H/R-CMECs, specifically including cell proliferation, cell adhesion, cell apoptosis, and ROS production, as well as nuclear factor erythroid 2-related factor 2 (Nrf2) nuclear translocation. In conclusion, our results determined that Tβ4 attenuated H/R-induced CMECs injury by miR-200a-Nrf2 signaling.
Collapse
Affiliation(s)
- Yang Li
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Xiaolong Zhu
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Xiping Liu
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Aolin Du
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Bo Yu
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, PR China
| |
Collapse
|
17
|
miR-221 alleviates the inflammatory response and cell apoptosis of neuronal cell through targeting TNFAIP2 in spinal cord ischemia-reperfusion. Neuroreport 2019; 29:655-660. [PMID: 29596155 DOI: 10.1097/wnr.0000000000001013] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
This study aimed to examine the role of miR-221 in inflammatory response and apoptosis of neuronal cells after spinal cord ischemia/reperfusion (I/R) injury. Blood samples were obtained from 20 I/R patients and that of 20 healthy individuals were used as a control. AGE1.HN and SY-SH-5Y neuronal cell lines subjected to oxygen-glucose deprivation (OGD) stress were used in cell experiments. Real-time PCR and western blot were used to evaluate the expression of miR-221, tumor necrosis factor-α, and TNFAIP2. TUNEL assay analyzed cell apoptosis. I/R patients had lower serum levels of miR-221 than healthy controls. In OGD-AGE1.HN and SY-SH-5Y cells, miR-221 was significantly downregulated and TNFAIP2 mRNA and protein were upregulated; meanwhile, both proinflammatory cytokine tumor necrosis factor-α and anti-inflammation cytokine interleukin-6 were elevated and the percentage of apoptotic cells was increased. This inflammatory response and cell apoptosis induced by OGD stress were attenuated by miR-221 overexpression and enhanced by miR-221 knockdown. TNFAIP2 is a target gene for miR-221 and could be regulated negatively by the miR-221 mimic or the miR-221 inhibitor with or without OGD stress. Accordingly, TNFAIP2 overexpression reversed the inflammatory response and cell apoptosis induced by miR-221 under OGD stress. Downregulation of miR-221 occurs in spinal cord I/R injury and in cell lines subjected to oxygen-glucose deprivation. miR-221 regulates the inflammatory response and apoptosis of neuronal cells through its impact on TNFAIP2.
Collapse
|
18
|
Zhao L, Yang XR, Han X. MicroRNA-146b induces the PI3K/Akt/NF-κB signaling pathway to reduce vascular inflammation and apoptosis in myocardial infarction by targeting PTEN. Exp Ther Med 2018; 17:1171-1181. [PMID: 30679990 PMCID: PMC6328856 DOI: 10.3892/etm.2018.7087] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 07/13/2018] [Indexed: 02/06/2023] Open
Abstract
The aim of the present study was to investigate the function of microRNA-146b on myocardial infarction and the mechanism. An MTT assay, Annexin V/propidium iodide (PI) apoptosis assay, ELISA kits, western blot analysis and a caspase-3/8 activity assay were used to measure cell growth, vascular apoptosis inflammatory factors, and the B-cell lymphoma 2-associated X protein (Bax), phosphatase and tensin homolog (PTEN), phosphoinositide 3-kinase (PI3K)/Akt/nuclear factor (NF)-κB signaling pathway. The expression of microRNA-146b was downregulated in the myocardial infarction rat model, compared with the control group. In an in vitro model of myocardial infarction, the downregulation of microRNA-146b increased inflammatory factors, vascular apoptosis, caspase-3/8 activity and the protein expression of Bax. MicroRNA-146b reduced vascular apoptosis, caspase-3/8 activity and the protein expression of Bax. MicroRNA-146b also regulated the PI3K/Akt/NF-κB signaling pathway to mediate vascular inflammation and apoptosis in myocardial infarction by PTEN. A PI3K inhibitor decreased the effect of microRNA-146b on vascular inflammation and apoptosis following myocardial infarction. In conclusion, microRNA-146b mediated vascular inflammation and apoptosis in patients with myocardial infarction, which may be associated with activation of the PI3K/Akt/NF-κB signaling pathway by PTEN.
Collapse
Affiliation(s)
- Li Zhao
- Department of Internal Medicine, Jining No. 1 People's Hospital, Jining, Shandong 272111, P.R. China
| | - Xue Rong Yang
- Department of Nursing, Jining No. 1 People's Hospital, Jining, Shandong 272111, P.R. China
| | - Xu Han
- Health Care Unit, Jining No. 1 People's Hospital, Jining, Shandong 272111, P.R. China
| |
Collapse
|
19
|
MicroRNA miR-24-3p Reduces Apoptosis and Regulates Keap1-Nrf2 Pathway in Mouse Cardiomyocytes Responding to Ischemia/Reperfusion Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:7042105. [PMID: 30622671 PMCID: PMC6304907 DOI: 10.1155/2018/7042105] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 09/13/2018] [Accepted: 10/11/2018] [Indexed: 12/18/2022]
Abstract
In recent years, microRNAs (miRNAs) have received increasing attention for their role in ischemia/reperfusion injury (I/RI), and many miRNAs have been demonstrated to play a very important role in cardiac I/RI. The miRNA miR-24-3p is a tumor suppressor that regulates multiple tumors; however, it remains unclear whether the expression level of miR-24-3p is altered in cardiac cells under I/RI. In this study, we used mouse primary cardiomyocytes and the H9C2 cardiomyocyte cell line to perform in vitro stimulated ischemia/reperfusion (SI/R) and then detected miR-24-3p expression level using quantitative real-time PCR (qRT-PCR). We discovered that the expression of miR-24-3p was significantly increased in cardiomyocytes following SI/R, and that the miR-24-3p level was inversely correlated to the ischemia marker HIF-1a. Furthermore, we transfected cardiomyocytes with miR-24-3p mimic or inhibitor to explore the role of miR-24-3p in cardiomyocyte ischemia/reperfusion injury in vitro. We performed flow cytometry to detect the apoptotic rate of H9C2 cardiomyocytes and found that the transfection of miR-24-3p mimic resulted in the decrease of the apoptosis rate of cardiomyocytes after SI/R, whereas the transfection of miR-24-3p inhibitor increased the number of apoptotic cardiomyocytes. These data suggest that the overexpression of miR-24-3p could reduce in vitro myocardial cell apoptosis induced by I/R injury. Finally, we applied the dual luciferase reporter gene system to verify whether miR-24-3p targets the Keap1 gene, and found that the luciferase signal intensity from a vector carrying the Keap1 wild-type reporter gene was significantly reduced after transfection with miR-24-3p mimic. The Keap1 protein level was also reduced following the transfection of miR-24-3p. The results from this study suggest a novel function of miR-24-3p in protecting cardiomyocytes from ischemia/reperfusion injury by the activation of the Nrf2-Keap1 pathway.
Collapse
|
20
|
Zhai CL, Tang GM, Qian G, Hu HL, Wang SJ, Yin D, Zhang S. MicroRNA-98 attenuates cardiac ischemia-reperfusion injury through inhibiting DAPK1 expression. IUBMB Life 2018; 71:166-176. [PMID: 30419147 DOI: 10.1002/iub.1879] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Accepted: 03/07/2018] [Indexed: 02/04/2023]
Abstract
Cardiovascular ischemic disease is a large class of diseases that are harmful to human health. The significant role of microRNAs (miRNAs) in terms of controlling cardiac injury has been reported in latest studies. MiR-98 is very important in regulating the apoptosis, the differentiation, the growth as well as the metastasis of cells. Nevertheless, the effect of miR-98 in the cardiac ischemia reperfusion (I/R) injury has rarely been investigated. In the current research, we found that the miR-98 expression was down-regulated in the cardiomyocytes subjected to hypoxia/reoxygenation (H/R) and in the myocardium of the I/R rats. In addition, over-expression of miR-98 could significantly reduce the myocardial oxidative stress and ischemic injury as well as cell apoptosis. In agreement, similar findings were demonstrated in H9c2 cells subjected to H/R injury. Bioinformatic analysis using MiRanda and TargetScan and luciferase activity assay confirmed death-associated protein kinase 1 (DAPK1) as a direct target of miR-98. These findings suggest that miR-98 may be exploited as a novel molecular marker or therapeutic target for myocardial I/R injury. © 2018 IUBMB Life, 71(1):166-176, 2019.
Collapse
Affiliation(s)
- Chang-Lin Zhai
- Department of Cardiovascular Diseases, Xinhua Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, People's Republic of China.,Department of Cardiovascular Diseases, The Frist Affiliated Hospital of Jiaxing University, Jiaxing, People's Republic of China
| | - Guan-Min Tang
- Department of Cardiovascular Diseases, The Frist Affiliated Hospital of Jiaxing University, Jiaxing, People's Republic of China
| | - Gang Qian
- Department of Cardiovascular Diseases, The Frist Affiliated Hospital of Jiaxing University, Jiaxing, People's Republic of China
| | - Hui-Lin Hu
- Department of Cardiovascular Diseases, The Frist Affiliated Hospital of Jiaxing University, Jiaxing, People's Republic of China
| | - Shi-Jun Wang
- Department of Cardiovascular Diseases, The Frist Affiliated Hospital of Jiaxing University, Jiaxing, People's Republic of China
| | - Dong Yin
- Department of Cardiovascular Diseases, The Frist Affiliated Hospital of Jiaxing University, Jiaxing, People's Republic of China
| | - Song Zhang
- Department of Cardiovascular Diseases, Xinhua Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
21
|
MicroRNAs and histone deacetylase inhibition-mediated protection against inflammatory β-cell damage. PLoS One 2018; 13:e0203713. [PMID: 30260972 PMCID: PMC6160007 DOI: 10.1371/journal.pone.0203713] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 08/24/2018] [Indexed: 01/22/2023] Open
Abstract
Inflammatory β-cell failure contributes to type 1 and type 2 diabetes pathogenesis. Pro-inflammatory cytokines cause β-cell dysfunction and apoptosis, and lysine deacetylase inhibitors (KDACi) prevent β-cell failure in vitro and in vivo, in part by reducing NF-κB transcriptional activity. We investigated the hypothesis that the protective effect of KDACi involves transcriptional regulation of microRNAs (miRs), potential new targets in diabetes treatment. Insulin-producing INS1 cells were cultured with or without the broad-spectrum KDACi Givinostat, prior to exposure to the pro-inflammatory cytokines IL-1β and IFN-γ for 6 h or 24 h, and miR expression was profiled with miR array. Thirteen miRs (miR-7a-2-3p, miR-29c-3p, miR-96-5p, miR-101a-3p, miR-140-5p, miR-146a-5p, miR-146b-5p, miR-340-5p, miR-384-5p, miR-455-5p, miR-466b-2-3p, miR-652-5p, and miR-3584-5p) were regulated by both cytokines and Givinostat, and nine were examined by qRT-PCR. miR-146a-5p was strongly regulated by cytokines and KDACi and was analyzed further. miR-146a-5p expression was induced by cytokines in rat and human islets. Cytokine-induced miR-146a-5p expression was specific for INS1 and β-TC3 cells, whereas α-TC1 cells exhibited a higher basal expression. Transfection of INS1 cells with miR-146a-5p reduced cytokine signaling, including the activity of NF-κB and iNOS promoters, as well as NO production and protein levels of iNOS and its own direct targets TNF receptor associated factor 6 (TRAF6) and interleukin-1 receptor-associated kinase 1 (IRAK1). miR-146a-5p was elevated in the pancreas of diabetes-prone BB-DP rats at diabetes onset, suggesting that miR-146a-5p could play a role in type 1 diabetes development. The miR array of cytokine-exposed INS1 cells rescued by KDACi revealed several other miRs potentially involved in cytokine-induced β-cell apoptosis, demonstrating the strength of this approach.
Collapse
|
22
|
Zheng J, Li J, Kou B, Yi Q, Shi T. MicroRNA-30e protects the heart against ischemia and reperfusion injury through autophagy and the Notch1/Hes1/Akt signaling pathway. Int J Mol Med 2018. [PMID: 29532851 PMCID: PMC5881647 DOI: 10.3892/ijmm.2018.3548] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The aim of the present study was to determine the cardioprotective mechanisms by which micro (mi)RNA-30e protects the heart from myocardial ischemia/reperfusion injury (MI/R) and to explore the signaling pathways that may confer protection for the heart and be potential therapeutic targets. It was demonstrated that miRNA‑30e expression was decreased in patients with MI/R. In H9C2 cells, silencing (si)miRNA‑30e significantly inhibited cellular apoptosis, the expression of apoptosis regulator BAX (Bax) and caspase‑3 activity. It also significantly increased the expression of microtubule‑associated proteins 1A/1B light chain 3B, p62, Beclin‑1, neurogenic locus notch homolog protein‑1 (Notch1), Hes1 and phosphorylated‑protein kinase B (p‑Akt), and decreased the expression of inducible NO synthase (iNOS) and proteins associated with oxidative stress. The inhibition of autophagy following treatment with 3‑methyladenine significantly reversed the effect of si‑miRNA‑30e on apoptosis, Bax, caspase‑3, iNOS and oxidative stress in H9C2 cells. The promotion of Notch1 expression increased the effect of si‑miRNA‑30e on apoptosis, Bax, caspase‑3, iNOS, Notch1, Hes1 and p‑Akt protein expression and oxidative stress in H9C2 cells. Taken together, these results indicate that miRNA‑30e protects the heart from MI/R via autophagy and the Notch1/Hes1/Akt signaling pathway.
Collapse
Affiliation(s)
- Jianjie Zheng
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jing Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Bo Kou
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Qiuyue Yi
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Tao Shi
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
23
|
Ong SB, Katwadi K, Kwek XY, Ismail NI, Chinda K, Ong SG, Hausenloy DJ. Non-coding RNAs as therapeutic targets for preventing myocardial ischemia-reperfusion injury. Expert Opin Ther Targets 2018; 22:247-261. [DOI: 10.1080/14728222.2018.1439015] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Sang-Bing Ong
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Khairunnisa Katwadi
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Xiu-Yi Kwek
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Nur Izzah Ismail
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Medical School, Singapore, Singapore
- Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur, Malaysia
| | - Kroekkiat Chinda
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
- Biomedical Research Unit in Cardiovascular Sciences (BRUCS), Naresuan University, Phitsanulok, Thailand
| | - Sang-Ging Ong
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Derek J Hausenloy
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Medical School, Singapore, Singapore
- National Heart Research Institute of Singapore, National Heart CentreSingapore, Singapore
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, London, UK
- Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore
| |
Collapse
|
24
|
miR-125a, miR-139 and miR-324 contribute to Urocortin protection against myocardial ischemia-reperfusion injury. Sci Rep 2017; 7:8898. [PMID: 28827743 PMCID: PMC5566224 DOI: 10.1038/s41598-017-09198-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 07/21/2017] [Indexed: 12/20/2022] Open
Abstract
Urocortin 1 and 2 (Ucn-1 and Ucn-2) have established protective actions against myocardial ischemia-reperfusion (I/R) injuries. However, little is known about their role in posttranscriptional regulation in the process of cardioprotection. Herein, we investigated whether microRNAs play a role in urocortin-induced cardioprotection. Administration of Ucn-1 and Ucn-2 at the beginning of reperfusion significantly restored cardiac function, as evidenced ex vivo in Langendorff-perfused rat hearts and in vivo in rat subjected to I/R. Experiments using microarray and qRT-PCR determined that the addition of Ucn-1 at reperfusion modulated the expression of several miRNAs with unknown role in cardiac protection. Ucn-1 enhanced the expression of miR-125a-3p, miR-324-3p; meanwhile it decreased miR-139-3p. Similarly, intravenous infusion of Ucn-2 in rat model of I/R mimicked the effect of Ucn-1 on miR-324-3p and miR-139-3p. The effect of Ucn-1 involves the activation of corticotropin-releasing factor receptor-2, Epac2 and ERK1/2. Moreover, the overexpression of miR-125a-3p, miR-324-3p and miR-139-3p promoted dysregulation of genes expression involved in cell death and apoptosis (BRCA1, BIM, STAT2), in cAMP and Ca2+ signaling (PDE4a, CASQ1), in cell stress (NFAT5, XBP1, MAP3K12) and in metabolism (CPT2, FoxO1, MTRF1, TAZ). Altogether, these data unveil a novel role of urocortin in myocardial protection, involving posttranscriptional regulation with miRNAs.
Collapse
|