1
|
Sun S, Xue J, Guo Y, Li J. Bioinformatics analysis of genes related to ferroptosis in hepatic ischemia-reperfusion injury. Front Genet 2022; 13:1072544. [PMID: 36531223 PMCID: PMC9755192 DOI: 10.3389/fgene.2022.1072544] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 11/15/2022] [Indexed: 07/30/2023] Open
Abstract
Background: Primary liver cancer is the sixth most commonly diagnosed cancer and the third leading cause of cancer death worldwide in 2020, and it ranks fifth in global incidence. Liver resection or liver transplantation are the two most prominent surgical procedures for treating primary liver cancer. Both inevitably result in HIRI, causing severe complications for patients and affecting their prognosis and quality of survival. Ferroptosis, a newly discovered mode of cell death, is closely related to HIRI. We used bioinformatics analysis to explore the relationship between the two further. Methods: The GEO database dataset GSE112713 and the FerrDB database data were selected to use bioinformatic analysis methods (difference analysis, FRGs identification, GO analysis, KEGG analysis, PPI network construction and analysis, Hub gene screening with GO analysis and KEGG analysis, intergenic interaction prediction, drug-gene interaction prediction, miRNA prediction) for both for correlation analysis. The GEO database dataset GSE15480 was selected for preliminary validation of the screened Hub genes. Results: We analysed the dataset GSE112713 for differential gene expression before and after hepatic ischemia-reperfusion and identified by FRGs, yielding 11 genes. These 11 genes were subjected to GO, and KEGG analyses, and PPI networks were constructed and analysed. We also screened these 11 genes again to obtain 5 Hub genes and performed GO analysis, KEGG analysis, intergenic interaction prediction, drug-gene interaction prediction, and miRNA prediction on these 5 Hub genes. Finally, we obtained preliminary validation of all these 5 Hub genes by dataset GSE15480. Conclusion: There is a close relationship between HIRI and ferroptosis, and inhibition of ferroptosis can potentially be a new approach to mitigate HIRI treatment in the future.
Collapse
|
2
|
Wei X, Yuan Y, Yang Q. Long noncoding RNA PVT1 accelerates the growth of placental trophoblasts in preeclampsia through the microRNA-24-3p/HSD11B2 axis. Mol Reprod Dev 2022; 89:271-280. [PMID: 35735229 DOI: 10.1002/mrd.23575] [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: 12/03/2021] [Revised: 04/28/2022] [Accepted: 05/07/2022] [Indexed: 11/11/2022]
Abstract
Long noncoding RNA plasmacytoma variant translocation 1 (PVT1) is essential for the maintenance of normal functions of trophoblasts in preeclampsia (PE). This study aims to decipher the concrete mechanism of PVT1 with the microRNA-24-3p/Type-2 11β-hydroxysteroid dehydrogenase (miR-24-3p/HSD11B2) axis in PE. PVT1, miR-24-3p, and HSD11B2 expression levels in normal placental tissues and PE placental tissues were defined. HTR-8/SVneo cells were transfected to determine the effects of PVT1, miR-24-3p, and HSD11B2 on the growth of HTR-8/SVneo cells. The relationships among PVT1/miR-24-3p/HSD11B2 in HTR-8/SVneo cells were identified. PVT1 and HSD11B2 were downregulated, while miR-24-3p was upregulated in the placenta of PE. Upregulated/downregulated PVT1 promoted/impeded the growth of human placental trophoblast (HTR-8/SVneo) cells in PE. Restored/knocked down miR-24-3p impeded/enhanced the growth of HTR-8/SVneo cells in PE. PVT1 inhibited miR-24-3p to mediate HSD11B2. PVT1 sponges miR-24-3p to regulate HSD11B2; thereby, the growth of placental trophoblasts is promoted in PE.
Collapse
Affiliation(s)
- Xiaoying Wei
- Department of Obstetrics, Hubei Maternal and Child Health Hospital, Wuhan, Hubei, China
| | - Yichong Yuan
- Department of gynaecology, Hubei Maternal and Child Health Hospital, Wuhan, Hubei, China
| | - Qiong Yang
- Department of Obstetrics, Hubei Maternal and Child Health Hospital, Wuhan, Hubei, China
| |
Collapse
|
3
|
Wang XX, Wu LH, Ai L, Pan W, Ren JY, Zhang Q, Zhang HM. Construction of an HCC recurrence model based on the investigation of immune-related lncRNAs and related mechanisms. MOLECULAR THERAPY - NUCLEIC ACIDS 2021; 26:1387-1400. [PMID: 34900397 PMCID: PMC8626812 DOI: 10.1016/j.omtn.2021.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/10/2021] [Accepted: 11/03/2021] [Indexed: 01/27/2023]
Abstract
Long noncoding RNAs (lncRNAs) are emerging as critical regulators of gene expression and play fundamental roles in immune regulation. Growing evidence suggests that immune-related genes and lncRNAs can serve as markers to predict the prognosis of patients with cancers, including hepatocellular carcinoma (HCC). This study aimed to contract an immune-related lncRNA (IR-lncRNA) signature for prospective assessment to predict early recurrence of HCC. A total of 319 HCC samples under radical resection were randomly divided into a training cohort (161 samples) and a testing cohort (158 samples). In the training dataset, univariate, lasso, and multivariate Cox regression analyses identified a 9-IR-lncRNA signature closely related to disease-free survival. Kaplan-Meier analysis, principal component analysis, gene set enrichment analysis, and nomogram were used to evaluate the risk model. The results were further confirmed in the testing cohort. Furthermore, we constructed a competitive endogenous RNA regulatory network. The results of the present study indicated that this 9-IR-lncRNA signature has important clinical implications for improving predictive outcomes and guiding individualized treatment in HCC patients. These IR-lncRNAs and regulated genes may be potential biomarkers associated with the prognosis of HCC.
Collapse
Affiliation(s)
- Xiang-Xu Wang
- Department of Clinical Oncology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Li-Hong Wu
- Xijing 986 Hospital Department, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Liping Ai
- Department of Clinical Oncology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Wei Pan
- Department of Clinical Oncology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Jing-Yi Ren
- Department of Clinical Oncology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Qiong Zhang
- Department of Clinical Oncology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Hong-Mei Zhang
- Department of Clinical Oncology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
- Corresponding author: Hong-Mei Zhang, Department of Clinical Oncology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China.
| |
Collapse
|
4
|
Yang J, Xiang Z, Zhang J, Yang J, Zhai Y, Fan Z, Wang H, Wu J, Huang Y, Xiong M, Ma C. miR-24 Alleviates MI/RI by Blocking the S100A8/TLR4/MyD88/NF-kB Pathway. J Cardiovasc Pharmacol 2021; 78:847-857. [PMID: 34581696 DOI: 10.1097/fjc.0000000000001139] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 08/29/2021] [Indexed: 11/25/2022]
Abstract
ABSTRACT Although inflammation plays an important role in myocardial ischemia/reperfusion injury (MI/RI), an anti-inflammatory treatment with a single target has little clinical efficacy because of the multifactorial disorders involved in MI/RI. MicroRNAs (miR-24) can achieve multitarget regulation in several diseases, suggesting that this factor may have ideal effects on alleviation of MI/RI. In the present study, bioinformatics method was used to screen potential therapeutic targets of miR-24 associated with MI/RI. Three days before ischemia/reperfusion surgery, rats in the ischemia/reperfusion, miR-24, and adenovirus-negative control groups were injected with saline, miR-24, and adenovirus-negative control (0.1 mL of 5 × 109 PFU/mL), respectively. Myocardial enzymes, myocardial infarct size, cardiac function, and the possible molecular mechanism were subsequently analyzed. In contrast to the level of S100A8, the level of miR-24 in myocardial tissue was significantly reduced after 30 minutes of ischemia followed by reperfusion for 2 hours. Overexpression of miR-24 reduced the myocardial infarction area and improved the heart function of rats 3 days after MI/RI. Moreover, miR-24 inhibited infiltration of inflammatory cells in the peri-infarction area and decreased creatine kinase myocardial band and lactate dehydrogenase release. Interestingly, miR-24 upregulation reduced S100A8 expression, followed by inhibition of toll-like receptor 4/MyD-88/nuclear factor-k-gene binding signaling activation. In conclusion, miR-24 can alleviate MI/RI via inactivation of the S100A8/toll-like receptor 4/MyD-88/nuclear factor-k-gene binding signaling pathway.
Collapse
Affiliation(s)
- Jian Yang
- Department of Cardiology, the First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China; and
| | - Zujin Xiang
- Department of Cardiology, the First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, China
| | - Jing Zhang
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China; and
- Yichang Key Laboratory of Ischemic Cardiovascular and Cerebrovascular Disease Translational Medicine, Yichang, China
| | - Jun Yang
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China; and
| | - Yuhong Zhai
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China; and
| | - Zhixing Fan
- Yichang Key Laboratory of Ischemic Cardiovascular and Cerebrovascular Disease Translational Medicine, Yichang, China
| | - Huibo Wang
- Department of Cardiology, the First College of Clinical Medical Science, China Three Gorges University, Yichang, China
| | - Jingyi Wu
- Department of Cardiology, the First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Yichang Key Laboratory of Ischemic Cardiovascular and Cerebrovascular Disease Translational Medicine, Yichang, China
| | - Yifan Huang
- Department of Cardiology, the First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Yichang Key Laboratory of Ischemic Cardiovascular and Cerebrovascular Disease Translational Medicine, Yichang, China
| | - Mengting Xiong
- Department of Cardiology, the First College of Clinical Medical Science, China Three Gorges University, Yichang, China
| | - Cong Ma
- Department of Cardiology, the First College of Clinical Medical Science, China Three Gorges University, Yichang, China
| |
Collapse
|
5
|
Bland AR, Payne FM, Ashton JC, Jamialahmadi T, Sahebkar A. The cardioprotective actions of statins in targeting mitochondrial dysfunction associated with myocardial ischaemia-reperfusion injury. Pharmacol Res 2021; 175:105986. [PMID: 34800627 DOI: 10.1016/j.phrs.2021.105986] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/11/2021] [Accepted: 11/11/2021] [Indexed: 12/24/2022]
Abstract
During cardiac reperfusion after myocardial infarction, the heart is subjected to cascading cycles of ischaemia reperfusion injury (IRI). Patients presenting with this injury succumb to myocardial dysfunction resulting in myocardial cell death, which contributes to morbidity and mortality. New targeted therapies are required if the myocardium is to be protected from this injury and improve patient outcomes. Extensive research into the role of mitochondria during ischaemia and reperfusion has unveiled one of the most important sites contributing towards this injury; specifically, the opening of the mitochondrial permeability transition pore. The opening of this pore occurs during reperfusion and results in mitochondria swelling and dysfunction, promoting apoptotic cell death. Activation of mitochondrial ATP-sensitive potassium channels (mitoKATP) channels, uncoupling proteins, and inhibition of glycogen synthase kinase-3β (GSK3β) phosphorylation have been identified to delay mitochondrial permeability transition pore opening and reduce reactive oxygen species formation, thereby decreasing infarct size. Statins have recently been identified to provide a direct cardioprotective effect on these specific mitochondrial components, all of which reduce the severity of myocardial IRI, promoting the ability of statins to be a considerate preconditioning agent. This review will outline what has currently been shown in regard to statins cardioprotective effects on mitochondria during myocardial IRI.
Collapse
Affiliation(s)
- Abigail R Bland
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand
| | - Fergus M Payne
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand
| | - John C Ashton
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand
| | - Tannaz Jamialahmadi
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashad, Iran; School of Medicine, The University of Western Australia, Perth, Australia; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
6
|
Putative role of uncoupling proteins in mitochondria-nucleus communications and DNA damage response. J Biosci 2021. [DOI: 10.1007/s12038-021-00224-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
7
|
Rosuvastatin alleviated the liver ischemia reperfusion injury by activating the expression of peroxisome proliferator-activated receptor gamma (PPARγ). J Bioenerg Biomembr 2021; 53:573-583. [PMID: 34235609 DOI: 10.1007/s10863-021-09909-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/29/2021] [Indexed: 10/20/2022]
Abstract
Liver ischemia and reperfusion could cause serious damage to liver tissues. Abnormal liver function could induce serious damage and threaten human health. Evidence emerged to suggest that rosuvastatin could relieve cerebral ischemia-reperfusion injury and alleviate the disease related to vessels by activating the expression of PPARγ. However, whether rosuvastatin could relieve the liver ischemia reperfusion injury by enhancing the expression of PPARγ is unclear. For the strictness of experimental findings, we established both the rat models and the cell model of liver ischemia reperfusion injury by respectively treating rats and cells with rosuvastatin. PPARγ inhibitor was also used for the stimulation of these cells and rats. Reactive oxygen species (ROS) levels, apoptosis and related protein levels were determined with ROS staining, ROS staining and western blotting for the detection of injury induced by oxygen-glucose deprivation and re-oxygenation (OGD/R). Pretreatment of rosuvastatin promoted the expression of PPARγ in liver tissues and MIHA cells. It also inhibited the ischemia reperfusion and OGD/R induced production of ROS while promoted the release of SOD in liver tissues and MIHA cells. Furthermore, rosuvastatin also alleviated the ischemia reperfusion -induced apoptosis of liver tissues and OGD/R-induced MIHA cells apoptosis. However, application of PPARγ inhibitor abolished the restorative effects of rosuvastatin on the apoptosis and oxidative stress on liver tissues and MIHA cells. Rosuvastatin prevented the liver ischemia reperfusion injury of rats by activating PPARγ.
Collapse
|
8
|
He X, Kuang G, Zuo Y, Li S, Zhou S, Ou C. The Role of Non-coding RNAs in Diabetic Nephropathy-Related Oxidative Stress. Front Med (Lausanne) 2021; 8:626423. [PMID: 33959621 PMCID: PMC8093385 DOI: 10.3389/fmed.2021.626423] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 03/25/2021] [Indexed: 12/14/2022] Open
Abstract
Diabetic nephropathy (DN) is one of the main complications of diabetes and the main cause of diabetic end-stage renal disease, which is often fatal. DN is usually characterized by progressive renal interstitial fibrosis, which is closely related to the excessive accumulation of extracellular matrix and oxidative stress. Non-coding RNAs (ncRNAs) are RNA molecules expressed in eukaryotic cells that are not translated into proteins. They are widely involved in the regulation of biological processes, such as, chromatin remodeling, transcription, post-transcriptional modification, and signal transduction. Recent studies have shown that ncRNAs play an important role in the occurrence and development of DN and participate in the regulation of oxidative stress in DN. This review clarifies the functions and mechanisms of ncRNAs in DN-related oxidative stress, providing valuable insights into the prevention, early diagnosis, and molecular therapeutic targets of DN.
Collapse
Affiliation(s)
- Xiaoyun He
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Gaoyan Kuang
- Department of Orthopedics, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Yi Zuo
- Department of Endocrinology, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Shuangxi Li
- Department of Pathophysiology, Hunan University of Medicine, Huaihua, China
| | - Suxian Zhou
- Department of Endocrinology, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Chunlin Ou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
9
|
Kuai F, Zhou L, Zhou J, Sun X, Dong W. Long non-coding RNA THRIL inhibits miRNA-24-3p to upregulate neuropilin-1 to aggravate cerebral ischemia-reperfusion injury through regulating the nuclear factor κB p65 signaling. Aging (Albany NY) 2021; 13:9071-9084. [PMID: 33675584 PMCID: PMC8034910 DOI: 10.18632/aging.202762] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 12/29/2020] [Indexed: 12/19/2022]
Abstract
Purpose: The aim of this study was to investigate the role of the tumor necrosis factor and HNRNPL related immunoregulatory long non-coding RNA (THRIL) in cerebral ischemia-reperfusion injury. Methods: A rat middle cerebral artery occlusion/ischemia-reperfusion (MCAO/IR) model and an oxygen glucose deprivation/reoxygenation (OGD/R) cell model were constructed. THRIL was knocked down using siTHRIL. Neurological deficit score was detected based on the criteria of Zea-Longa. Brain region 2,3,5-Triphenyltetrazolium (TTC) staining and quantitative analysis of cerebral infarction volume, RT-qPCR, and fluorescence immunostaining were performed for assessing THRIL expression. MTT assay was used to detect the cell proliferation ability after transfection, TUNEL assay was applied to detect apoptosis, and western blot and ELISA detected related protein expression. A dual luciferase reporter system and RIP assay were used to confirm the target relationship. Results: THRIL was upregulated in both in vitro and in vivo models of brain ischemia-reperfusion injury. Knockdown of THRIL attenuated OGD/R neuronal apoptosis and OGD/R-induced inflammation. THRIL targeted and regulated the expression of miR-24-3p/neuropilin-1 (NRP1) axis. THRIL silencing significantly improved the neurological functioning of rats in the MCAO/R model by miR-24-3p/NRP1/NF-κB p65 signaling pathway. Conclusion: THRIL could aggravate cerebral ischemia-reperfusion injury by competitively binding to miR-24-3p to promote the upregulation of NRP1 and further promoted the activation of the NF-κB p65 signaling pathway.
Collapse
Affiliation(s)
- Feng Kuai
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China.,Department of Geriatrics, The First People's Hospital of Yancheng, The Forth Affiliated Hospital of Nantong University, Yancheng 224001, China
| | - Liang Zhou
- Department of orthopedic, The People's Hospital of Lianshui, Huai'an 223001, China
| | - Jianping Zhou
- Department of Geriatrics, The First People's Hospital of Yancheng, The Forth Affiliated Hospital of Nantong University, Yancheng 224001, China
| | - Xuemei Sun
- Department of Geriatrics, The First People's Hospital of Yancheng, The Forth Affiliated Hospital of Nantong University, Yancheng 224001, China
| | - Wanli Dong
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| |
Collapse
|
10
|
Yan X, Hou J. miR-22 Host Gene Enhances Nuclear Factor-kappa B Activation to Aggravate Hypoxia-induced Injury in AC16 Cardiomyocytes. Cell Transplant 2021; 30:963689721990323. [PMID: 33631962 PMCID: PMC7917431 DOI: 10.1177/0963689721990323] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Myocardial infarction (MI) is a severe life-threatening disease caused by acute and persistent ischemia and hypoxia and eventually leads to heart failure and sudden death. Long noncoding RNAs (lncRNAs) play significant roles in the pathology, diagnosis, and development of various cardiovascular diseases, including MI. This study aimed to explore the effect and molecular mechanism of lncRNA miR-22 host gene (MIR22HG) on hypoxia-induced injury in AC16 cardiomyocytes. The expression of MIR22HG and miR-24 in hypoxia-treated AC16 cardiomyocytes was detected by quantitative real-time polymerase chain reaction. Cell viability, lactate dehydrogenase release, levels of aspartate aminotransferase (AST) and creatine kinase-MB (CK-MB), and apoptosis were detected by Cell Counting Kit-8, lactate dehydrogenase (LDH) release assay, commercial enzyme-linked immune sorbent assay kits, and flow cytometry analysis, respectively. The protein levels of nuclear factor-kappa B (NF-κB) p65 and cytoplasmic inhibitor of kappa B alpha (IκBα) and phosphorylated IκBα were detected by western blot. Results showed that hypoxia treatment decreased viability and increased MIR22HG expression in AC16 cardiomyocytes. MIR22HG overexpression aggravated hypoxia-induced viability reduction, leakage of myocardial injury markers LDH, AST, and CK-MB, and apoptosis in AC16 cardiomyocytes, while MIR22HG knockdown elicited the reverse effects. MIR22HG overexpression enhanced NF-κB activation in hypoxia-treated AC16 cardiomyocytes. Inhibition of NF-κB pathway impaired the effects of MIR22HG overexpression on hypoxia-induced injury in AC16 cardiomyocytes. Moreover, MIR22HG knockdown inhibited the NF-κB pathway by upregulating miR-24 in AC16 cardiomyocytes. Inhibition of miR-24 resisted the effects of MIR22HG silencing on hypoxia-induced injury in AC16 cardiomyocytes. In conclusion, MIR22HG overexpression aggravated hypoxia-induced injury in AC16 cardiomyocytes via enhancing NF-κB activation by targeting miR-24.
Collapse
Affiliation(s)
- Xu Yan
- Department of Cardiology, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, PR China
| | - Jinlan Hou
- Department of Cardiology, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, PR China
| |
Collapse
|
11
|
Li T, Gu J, Yang O, Wang J, Wang Y, Kong J. Bone Marrow Mesenchymal Stem Cell-Derived Exosomal miRNA-29c Decreases Cardiac Ischemia/Reperfusion Injury Through Inhibition of Excessive Autophagy via the PTEN/Akt/mTOR Signaling Pathway. Circ J 2020; 84:1304-1311. [PMID: 32581152 DOI: 10.1253/circj.cj-19-1060] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Cardiac ischemia/reperfusion (I/R) injury will cause a large amount of cardiomyocyte loss and cascade reactions such as apoptosis, mitochondrial dysfunction, and excessive autophagy. Mesenchymal stem cells (MSCs) are promising therapeutic tools to replace damaged cardiomyocytes, but the underlying mechanism is still unknown. METHODS AND RESULTS Exosomes contain many microRNAs and protein, which are believed to have multiple biological functions. This study explored the role of bone marrow MSCs (BMMSCs)-derived exosomes under different oxidation levels in heart protection and miRNA-related mechanisms. Exosomes extracted from BMMSCs contained a high level of miR-29c, and its expression level changed after cells were treated under hypoxia/reoxygenation (H/R) conditions. In vivo I/R experiments also confirmed an expression change of miR-29c, and PTEN-Akt-mTOR is one of the predominant pathways that regulate autophagic change during this process. CONCLUSIONS This study highlighted the role of miR-29c in regulating autophagy under cardiac I/R injury, which also extended existing mechanisms of a stem cell and its derivative to explore potential therapeutic interventions in ischemic heart diseases.
Collapse
Affiliation(s)
- Te Li
- Department of Geriatrics, The First Hospital of Jilin University
| | | | - Ou Yang
- Department of Geriatrics, The First Hospital of Jilin University
| | - Jianmeng Wang
- Department of Geriatrics, The First Hospital of Jilin University
| | - Yonggang Wang
- Department of Cardiovascular Center, The First Hospital of Jilin University
| | - Jian Kong
- Department of Geriatrics, The First Hospital of Jilin University
| |
Collapse
|
12
|
Zhang H, Xue S, Feng Y, Shen J, Zhao J. MicroRNA-24-3p inhibition prevents cell growth of vascular smooth muscle cells by targeting Bcl-2-like protein 11. Exp Ther Med 2020; 19:2467-2474. [PMID: 32256723 PMCID: PMC7086294 DOI: 10.3892/etm.2020.8517] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 11/29/2019] [Indexed: 12/15/2022] Open
Abstract
Numerous reports have shown that dysfunction of vascular smooth muscle cells (VSMCs) serves a critical function in the development of cardiovascular disease, including coronary heart disease (CHD). microRNAs (miRNAs/miRs) have been reported to play important roles in regulating the function of VSMCs. The present study aimed to determine the role of miR-24-3p in VSMCs and to uncover the underlying mechanism. The expression of miR-24-3p in the peripheral blood samples of CHD patients was measured by reverse transcription-quantitative (RT-q)PCR. It was found that the level of miR-24-3p in the peripheral blood of patients with CHD was significantly upregulated compared with that in healthy controls. A dual luciferase reporter assay was performed to determine whether Bcl-2-like protein 11 (Bcl-2L11) was a target gene of miR-24-3p, and it was identified that Bcl-2L11 was a direct target of miR-24-3p. The mRNA level and protein expression of Bcl-2L11 in the peripheral blood of patients with CHD were measured by RT-qPCR and western blotting, respectively. The findings suggested that Bcl-2L11 was downregulated in the peripheral blood of patients with CHD. In addition, it was found that downregulation of miR-24-3p suppressed VSMC proliferation and promoted VSMC apoptosis, while the effects of the miR-24-3p inhibitor on cell viability and apoptosis were reversed by Bcl-2L11-small interfering (si)RNA. Additionally, downregulation of miR-24-3p increased the levels of Bcl-2L11, caspase-3 and Bax, and decreased Bcl-2 expression in VSMCs; these changes were abolished by Bcl-2L11-siRNA. In conclusion, the aforementioned results indicated that miR-24-3p was an important regulator in VSMC proliferation and apoptosis by targeting Bcl-2L11, which suggested that miR-24-3p might be a potential therapeutic target for the treatment of CHD.
Collapse
Affiliation(s)
- Huanxin Zhang
- Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Shizhen Xue
- Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Yi Feng
- Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Jun Shen
- Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Jixian Zhao
- Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| |
Collapse
|
13
|
Lin J, Lin H, Ma C, Dong F, Hu Y, Li H. MiR-149 Aggravates Pyroptosis in Myocardial Ischemia-Reperfusion Damage via Silencing FoxO3. Med Sci Monit 2019; 25:8733-8743. [PMID: 31741467 PMCID: PMC6880628 DOI: 10.12659/msm.918410] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND MicroRNAs (miRNAs), which modulate the expression of their target genes, are commonly involved in stimulating and adjusting of many processes that result in cardiovascular diseases, contain cardiac ischemia/reperfusion (I/R) damage. However, the expression and role of miR-149 in pyroptosis mediated myocardial I/R damage remains unclear. MATERIAL AND METHODS Real-time polymerase chain reaction was performed to measure the miR-149 and FoxO3 expression in I/R stimulated H9C2 cells. The cell proliferation, pyroptosis-related inflammatory genes in I/R-treated H9C2 cells transfected miR-149 mimics or miR-149 inhibitor were both explored. We predicted and confirmed miR-149 targets by using bioinformatics analyses and luciferase reporter assay. In addition, the potential relationship between miR-149 and FoxO3 in pyroptosis from I/R treated H9C2 cells was analyzed. RESULTS Our results showed that miR-149 was upregulated, while FoxO3 was downregulated in I/R stimulated H9C2 cells. Over-expression of miR-149 inhibited cell viability and promote pyroptosis, however, down-expression of miR-149 had an opposite effect in I/R treated H9C2 cells. Furthermore, miR-149 could negatively regulate FoxO3 expression by binding 3'UTR, whereas silencing of FoxO3 attenuated the effect of miR-149-mimics on cell proliferation and pyroptosis in I/R treated H9C2 cells. CONCLUSIONS Our study found that miR-149 played a critical role in pyroptosis during cardiac I/R injury, and thus, might provide a novel therapeutic target.
Collapse
Affiliation(s)
- Jie Lin
- Department of Cardiology, Shenzhen University General Hospital, Shenzhen, Guandong, China (mainland)
| | - Haihuan Lin
- Laboratory Medicine Humboldt University, Charité University Medicine, Berlin, China (mainland)
| | - Chao Ma
- Berlin-Brandenburg Center for Regenerative Therapies, Charité University Medicine, Berlin, China (mainland)
| | - Fengquan Dong
- Department of Cardiology, Shenzhen University General Hospital, Shenzhen, Guandong, China (mainland)
| | - Yingchun Hu
- Department of Cardiology, Shenzhen University General Hospital, Shenzhen, Guandong, China (mainland)
| | - Haiying Li
- Department of Cardiology, Shenzhen University General Hospital, Shenzhen, Guandong, China (mainland)
| |
Collapse
|