1
|
Guo D, Hu L, Xie P, Sun P, Yu W. Seipin is involved in oxygen-glucose deprivation/reoxygenation induced neuroinflammation by regulating the TLR3/TRAF3/NF-κB pathway. Int Immunopharmacol 2024; 134:112182. [PMID: 38703568 DOI: 10.1016/j.intimp.2024.112182] [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: 02/25/2024] [Revised: 04/25/2024] [Accepted: 04/27/2024] [Indexed: 05/06/2024]
Abstract
Seipin plays a crucial role in lipid metabolism and is involved in neurological disorders. However, the function and mechanism of action of seipin in acute ischemic stroke have not yet been elucidated. Here, we aimed to investigate the effect of seipin on neuroinflammation induced by oxygen-glucose deprivation/reoxygenation (OGD/R) and further explore the molecular mechanism by functional experiments. Our results revealed a significant decrease in seipin mRNA levels, accompanied by enhanced expression of TNF-α in patients with AIS, and a significant negative correlation between seipin and TNF-α was observed. Additionally, there was a negative correlation between seipin levels and the National Institutes of Health Stroke Scale (NIHSS) score. Furthermore, seipin levels were also decreased in middle cerebral artery occlusion/reperfusion (MCAO/R) mice and OGD/R-treated BV2 cells. RNA sequencing analysis showed that seipin knockdown altered the Toll-like receptor 3 (TLR3) signaling pathway. It was further confirmed in vitro that seipin knockdown caused significantly increased secretion of inflammatory factors including TNF-α, interleukin (IL)-1β, and interferon (IFN)-β. Meanwhile, seipin knockdown activated the Tlr3 signal pathway while this effect could be reversed by Tlr3 inhibitor in OGD/R treated BV2 cells. Furthermore, neuroinflammation induced by OGD/R was significantly reduced by seipin overexpression. Overall, our study demonstrate that seipin deficiency aggravates neuroinflammation by activating the TLR3/TRAF3/NF-κB signaling pathway after OGD/R stimuli, and suggest that seipin may be a potential therapeutic target for AIS.
Collapse
Affiliation(s)
- Dongfen Guo
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, School of Basic Medical Science, Guizhou Medical University, 09 Beijing Road, Guiyang 550004, Guizhou, China
| | - Lele Hu
- Department of Neurology, The Second People's Hospital of Guiyang, Guiyang 550023, Guizhou, China
| | - Peng Xie
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, School of Basic Medical Science, Guizhou Medical University, 09 Beijing Road, Guiyang 550004, Guizhou, China
| | - Ping Sun
- Department of Neurology, The Second People's Hospital of Guiyang, Guiyang 550023, Guizhou, China.
| | - Wenfeng Yu
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, School of Basic Medical Science, Guizhou Medical University, 09 Beijing Road, Guiyang 550004, Guizhou, China; Key Laboratory of Human Brain Bank for Functions and Diseases of Department of Education of Guizhou Province, Guizhou Medical University, Guiyang 550025, Guizhou, China.
| |
Collapse
|
2
|
Liang Y, Han D, Zhang S, Sun L. FOSL1 regulates hyperproliferation and NLRP3-mediated inflammation of psoriatic keratinocytes through the NF-kB signaling via transcriptionally activating TRAF3. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119689. [PMID: 38367916 DOI: 10.1016/j.bbamcr.2024.119689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 02/05/2024] [Accepted: 02/09/2024] [Indexed: 02/19/2024]
Abstract
Psoriasis is a common and immune-mediated skin disease related to keratinocytes hyperproliferation and inflammation. Fos-like antigen-1 (FOSL1) is an important transcription factor involved in various diseases. FOSL1 has been reported to be differentially expressed in psoriasis. However, the roles and mechanism of FOSL1 in psoriasis progression remain largely unknown. FOSL1 is an upregulated transcription factor in psoriasis and increased in M5-treated HaCaT cells. FOSL1 had a diagnostic value in psoriasis, and positively associated with PASI score, TNF-α and IL-6 levels in psoriasis patients. FOSL1 silencing attenuated M5-induced HaCaT cell hyperproliferation through decreasing cell viability and proliferative ability and increasing cell apoptosis. FOSL1 knockdown mitigated M5-induced NLRP3 inflammasome activation and it-mediated inflammatory cytokine (IL-6, IL-8 and CCL17) expression. TRAF3 expression was increased in psoriasis patients and M5-treated HaCaT cells. FOSL1 transcriptionally activating TRAF3 in HaCaT cells. TRAF3 overexpression reversed the suppressive effects of FOSL1 silencing on M5-induced hyperproliferation and NLRP3-mediated inflammation. FOSL1 knockdown attenuated M5-induced NF-κB signaling activation by reducing TRAF3. Activation of NF-κB signaling reversed the effects of FOSL1 knockdown on hyperproliferation and inflammation in M5-treated cells. FOSL1 silencing prevented M5-induced hyperproliferation and NLRP3-mediated inflammation of keratinocytes by inhibiting TRAF3-mediated NF-κB activity, indicating FOSL1 might act as a therapeutic target of psoriasis.
Collapse
Affiliation(s)
- Yan Liang
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China.
| | - Dan Han
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Shaojun Zhang
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Liang Sun
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| |
Collapse
|
3
|
Li J, Wang Z, Tan H, Tang M. ALKBH5-mediated m6A demethylation of pri-miR-199a-5p exacerbates myocardial ischemia/reperfusion injury by regulating TRAF3-mediated pyroptosis. J Biochem Mol Toxicol 2024; 38:e23710. [PMID: 38605440 DOI: 10.1002/jbt.23710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/22/2024] [Accepted: 03/29/2024] [Indexed: 04/13/2024]
Abstract
Myocardial ischemia‒reperfusion injury (MI/RI) is closely related to pyroptosis. alkB homolog 5 (ALKBH5) is abnormally expressed in the MI/RI models. However, the detailed molecular mechanism of ALKBH5 in MI/RI has not been elucidated. In this study, rats and H9C2 cells served as experimental subjects and received MI/R induction and H/R induction, respectively. The abundance of the targeted molecules was evaluated using RT-qPCR, Western blotting, immunohistochemistry, immunofluorescence, and enzyme-linked immunosorbent assay. The heart functions of the rats were evaluated using echocardiography, and heart injury was evaluated. Cell viability and pyroptosis were determined using cell counting Kit-8 and flow cytometry, respectively. Total m6A modification was measured using a commercial kit, and pri-miR-199a-5p m6A modification was detected by Me-RNA immunoprecipitation (RIP) assay. The interactions among the molecules were validated using RIP and luciferase experiments. ALKBH5 was abnormally highly expressed in H/R-induced H9C2 cells and MI/RI rats. ALKBH5 silencing improved injury and inhibited pyroptosis. ALKBH5 reduced pri-miR-199a-5p m6A methylation to block miR-199a-5p maturation and inhibit its expression. TNF receptor-associated Factor 3 (TRAF3) is a downstream gene of miR-199a-5p. Furthermore, in H/R-induced H9C2 cells, the miR-199a-5p inhibitor-mediated promotion of pyroptosis was reversed by ALKBH5 silencing, and the TRAF3 overexpression-mediated promotion of pyroptosis was offset by miR-199a-5p upregulation. ALKBH5 silencing inhibited pri-miR-199a-5p expression and enhanced pri-miR-199a-5p m6A modification to promote miR-199a-5p maturation and enhance its expression, thereby suppressing pyroptosis to alleviate MI/RI through decreasing TRAF3 expression.
Collapse
Affiliation(s)
- Jiarong Li
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha City, Hunan Province, P.R. China
| | - Zhirong Wang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha City, Hunan Province, P.R. China
| | - Huayi Tan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha City, Hunan Province, P.R. China
| | - Mi Tang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha City, Hunan Province, P.R. China
| |
Collapse
|
4
|
Fei Q, Liu J, Qiao L, Zhang M, Xia H, Lu D, Wu D, Wang J, Li R, Li J, Yang F, Liu D, Xie B, Hui W, Qian B. Mst1 attenuates myocardial ischemia/reperfusion injury following heterotopic heart transplantation in mice through regulating Keap1/Nrf2 axis. Biochem Biophys Res Commun 2023; 644:140-148. [PMID: 36646002 DOI: 10.1016/j.bbrc.2022.12.087] [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: 12/08/2022] [Revised: 12/17/2022] [Accepted: 12/30/2022] [Indexed: 01/09/2023]
Abstract
Ischemia reperfusion (I/R) injury remains a frequent adverse event that accompanies heart transplantation. Oxidative stress and aberrant production of free radicals were regarded as the culprit of cell death and tissue damage in post-transplant IR injury. Mst1 has been identified as a mediator of oxidative stress and Nrf2 regulates anti-oxidative enzymes, however, the interaction between Mst1 and Nrf2 anti-oxidative stress pathway remains to be clarified in the event of cardiac IR injury. Herein, the model of ischemia-reperfusion injury in heterotopic heart transplantation mice was firstly established.. We observed that cardiac IR induced upregulation of Mst1 and activation of Nrf2/HO-1pathway in mice receiving heterotopic heart transplantation. Further Cobalt dichloride-induced oxidative stress model of RAW264.7 macrophage cells were then established to mimic cardiac I/R injury, results showed that exposure to CoCl2 induced the upregulation of Mst1 and activation of Keap1/Nrf2 pathway, and genetic ablation of Mst-1 and inhibition of Keap1/Nrf2 pathway aggravated oxidative damage in those cells. Additional in vivo study showed that transfection of Mst1 shRNA spurred ROS generation and worsened cardiac damage in IR mice. Meanwhile, Mst1-KD mice receiving heart transplantation showed markedly downregulation of Nrf2, HO-1 yet upregulation of Keap1, indicating diminished protective effect against tissue damage caused by IR probably owing to the frustration of Keap1/Nrf2 pathway. Taken together, our findings demonstrated the protective effect of Mst1 from cardiac IR injury via triggering Keap1/Nrf2 axis and suppressing ROS generation, which shed light on the promising role of Mst1 in transitional management of IR injury resulted from cardiac transplantation.
Collapse
Affiliation(s)
- Qi Fei
- Department of Pulmonary and Critical Care Medicine, Peking University Shenzhen Hospital, Futian District, Shenzhent, 518036, Guangdong, People's Republic of China
| | - Justin Liu
- Department of Statistics, University of California, Riverside, 900 University Ave, Riverside, CA, 92521, USA
| | - Li Qiao
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei, 230601, People's Republic of China
| | - Meng Zhang
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei, 230601, People's Republic of China
| | - Haidong Xia
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei, 230601, People's Republic of China
| | - Daoqiang Lu
- School of Medicine, Foshan University, Foshan, 528000, Guangdong, People's Republic of China
| | - Di Wu
- School of Medicine, Foshan University, Foshan, 528000, Guangdong, People's Republic of China
| | - Jun Wang
- School of Medicine, Foshan University, Foshan, 528000, Guangdong, People's Republic of China
| | - Riwang Li
- School of Medicine, Foshan University, Foshan, 528000, Guangdong, People's Republic of China
| | - Jie Li
- School of Medicine, Foshan University, Foshan, 528000, Guangdong, People's Republic of China
| | - Fang Yang
- School of Medicine, Foshan University, Foshan, 528000, Guangdong, People's Republic of China
| | - Dahai Liu
- School of Medicine, Foshan University, Foshan, 528000, Guangdong, People's Republic of China.
| | - Baiyi Xie
- Department of Urology Surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi, China.
| | - Wenqiao Hui
- Anhui Province Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agriculture Sciences, Hefei, Anhui, 230031, China.
| | - Ban Qian
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei, 230601, People's Republic of China.
| |
Collapse
|
5
|
Wang J, Wang X, Cao M, Zhang L, Lin J. CircUSP39/miR-362-3p/TRAF3 Axis Mediates Hypoxia/Reoxygenation-Induced Cardiomyocyte Oxidative Stress, Inflammation, and Apoptosis. Int Heart J 2023; 64:263-273. [PMID: 37005320 DOI: 10.1536/ihj.22-232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/04/2023]
Abstract
Accumulating evidence suggested that aberrantly regulated circular RNA (circRNA) is a critical contributor to cardiovascular diseases, including acute myocardial infarction (AMI). However, the role and molecular mechanism of circUSP39 in AMI development remain unclear.Candidate circRNAs were screened from the Gene Expression Omnibus (GEO) database (GSE160717) and analyzed using the GEO2R tool. Hypoxia/reoxygenation (H/R) -induced AC16 cells were used to investigate the function of circUSP39 in H/R injury of cardiomyocytes. Quantitative real-time PCR (qRT-PCR) was employed to test RNA levels in H/R-induced AC16 cells. Cell Counting Kit-8, enzyme-linked immunosorbent assay, flow cytometry, and western blot (WB) assay were used to determine cell viability, oxidative stress, inflammatory factor levels, and cell apoptosis. RNA immunoprecipitation, RNA pull-down, and dual-luciferase reporter assay were conducted to validate the interactions between circRNA ubiquitin-specific peptidase 39 (circUSP39), miR-362-3p, and tumor necrosis factor receptor-associated factor 3 (TRAF3).In H/R-induced AC16 cells, the expression levels of circUSP39 and TRAF3 were upregulated whereas miR-362-3p expression was downregulated. CircUSP39 silencing markedly enhanced cell viability and superoxide dismutase activity but mitigated malondialdehyde level, secretion of inflammatory factors (IL-6, TNF-α, IL-1β, and MCP-1), and cell apoptosis in H/R-induced AC16 cells. CircUSP39 expedited H/R-induced AC16 cell injury by sponging miR-362-3p to increase the expression of TRAF3.CircUSP39 could facilitate H/R-induced cardiomyocyte oxidative stress, inflammation, and apoptosis by the miR-362-3p/TRAF3 axis, elucidating that it might be a therapeutic target for AMI.
Collapse
Affiliation(s)
| | - Xuan Wang
- Department of International Medical Center, Tianjin Hospital
| | - Mingying Cao
- Department of Cardiology, Tianjin Union Medical Center
| | - Lingli Zhang
- Department of Cardiology, Tianjin Union Medical Center
| | - Jingna Lin
- Department of Endocrinology, Tianjin Union Medical Center
| |
Collapse
|
6
|
Circ_0050908 up-regulates TRAF3 by sponging miR-324-5p to aggravate myocardial ischemia-reperfusion injury. Int Immunopharmacol 2022; 108:108740. [DOI: 10.1016/j.intimp.2022.108740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/25/2021] [Accepted: 03/28/2022] [Indexed: 11/21/2022]
|
7
|
Sun L, Liu J, Sun X, Zhang Y, Cui X. CircTRHDE knockdown protects WI-38 cells against LPS-induced inflammatory injury. Autoimmunity 2022; 55:233-242. [PMID: 35481453 DOI: 10.1080/08916934.2022.2062595] [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: 11/02/2022]
Abstract
BACKGROUND Circular RNAs (circRNAs) have been reported to be involved in the progression of infantile pneumonia. Here, we investigated the function of circTRHDE in lipopolysaccharide (LPS)-induced cell inflammatory injury to evaluate its role in infantile pneumonia progression. METHODS The circTRHDE, microRNA (miR)-381-3p and TNF-receptor associated factor 3 (TRAF3) expression were detected by quantitative real-time PCR. LPS-induced WI-38 cells were used to construct an inflammatory injury model. Cell viability, inflammation and apoptosis were measured by cell counting kit assay, ELISA assay and flow cytometry. Caspase3 activity, MDA level and SOD activity were analysed to assess cell apoptosis and oxidative stress. Protein levels were determined using western blot analysis. The interaction between miR-381-3p and circTRHDE or TRAF3 was confirmed by dual-luciferase activity assay and RNA pull-down assay. RESULTS CircTRHDE had increased expression in infantile pneumonia patients and LPS-induced WI-38 cells. LPS treatment inhibited WI-38 cell viability while promoting inflammation, apoptosis and oxidative stress. However, knockdown of circTRHDE remitted LPS-triggered WI-38 cell injury. CircTRHDE could sponge miR-381-3p to positively regulate TRAF3 expression. MiR-381-3p suppressed LPS-induced WI-38 cell inflammatory injury, and this effect was revoked by TRAF3 overexpression. Also, LPS-induced WI-38 cell inflammatory injury restrained by circTRHDE knockdown also were reversed by miR-318-3p inhibitor or TRAF3 overexpression. CONCLUSION Our findings demonstrated that circTRHDE might be a target for infantile pneumonia treatment, which relieved LPS-induced cell inflammatory injury by the regulation of the miR-318-3p/TRAF3 axis.
Collapse
Affiliation(s)
- Lifang Sun
- Department of Pediatrics, Zhengzhou Central Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Jingyan Liu
- Department of Pediatrics, Zhengzhou Central Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Xu Sun
- Department of Pediatrics, Zhengzhou Central Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Yanhua Zhang
- Department of Pediatrics, Zhengzhou Central Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Xinfang Cui
- Department of Pediatrics, Zhengzhou Central Hospital, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
8
|
Zhu S, Chen Z, Liang Q. TRAF3 promoted ROS-induced oxidative stress in model of cardiac infarction through the regulation of ULK1 ubiquitination. Clin Exp Hypertens 2022; 44:403-410. [PMID: 35318880 DOI: 10.1080/10641963.2022.2055766] [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: 11/03/2022]
Abstract
OBEJECTIVES Cardiac infarction is a dynamic, nonlinear and unpredictable course of disease, and who die of acute myocardial infarction, and coronary thrombosis. TRAF3 provide novel targets for the clinical prevention and treatment for tumors, viral infection, and so on.We investigated the mechanisms of TRAF3 gene, which plays a possible role in cardiac infarction and contributes to the pathogenesis of cardiac infarction-induced oxidative stress. METHODS Serum samples of patients with cardiac infarction and normal healthy volunteers were obtained from the 920 Hospital of PLA joint service support force. C57BL/6 mice were ligated and H9C2 cells were induced with 1% O2,5%CO2 and 94% N2. RESULTS The mRNA expression levels of TRAF3 in patients with cardiac infarction were increased, compared to healthy volunteers. Serum mRNA of TRAF3 was in positive correlation with serum CK levels in patients with cardiac infarction. Over-expression of TRAF3 heightened ROS-induced oxidative stress in vitro model of cardiac infarction. Then, TRAF3 recombinant protein could promote oxidative stress and aggravated cardiac infarction in mice model. Over-expression of TRAF3 induced ULK1 protein expression and reduced ULK1 ubiquitination in vitro model. The activation of ULK1 reduced the effects of TRAF3 on oxidative stress in vitro model of cardiac infarction. Meanwhile, the inhibition of ULK1 reversed the effects of si-TRAF3 on oxidative stress in vitro model of cardiac infarction. CONCLUSIONS This study identified that TRAF3 promoted ROS-induced oxidative stress in model of cardiac infarction through the regulation of ULK1 ubiquitination, which could potentially give rise to a new strategy for the treatment of cardiac infarction.
Collapse
Affiliation(s)
- Shaobing Zhu
- Department of Emergency, 920 Hospital of PLA joint service support force, Kunming, YN, China
| | - Zhenyu Chen
- Department of Emergency, 920 Hospital of PLA joint service support force, Kunming, YN, China
| | - Qilin Liang
- Department of Emergency, 920 Hospital of PLA joint service support force, Kunming, YN, China
| |
Collapse
|
9
|
Signaling pathways of inflammation in myocardial ischemia/reperfusion injury. CARDIOLOGY PLUS 2022. [DOI: 10.1097/cp9.0000000000000008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
10
|
Gissler MC, Stachon P, Wolf D, Marchini T. The Role of Tumor Necrosis Factor Associated Factors (TRAFs) in Vascular Inflammation and Atherosclerosis. Front Cardiovasc Med 2022; 9:826630. [PMID: 35252400 PMCID: PMC8891542 DOI: 10.3389/fcvm.2022.826630] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/27/2022] [Indexed: 12/20/2022] Open
Abstract
TNF receptor associated factors (TRAFs) represent a family of cytoplasmic signaling adaptor proteins that regulate, bundle, and transduce inflammatory signals downstream of TNF- (TNF-Rs), interleukin (IL)-1-, Toll-like- (TLRs), and IL-17 receptors. TRAFs play a pivotal role in regulating cell survival and immune cell function and are fundamental regulators of acute and chronic inflammation. Lately, the inhibition of inflammation by anti-cytokine therapy has emerged as novel treatment strategy in patients with atherosclerosis. Likewise, growing evidence from preclinical experiments proposes TRAFs as potent modulators of inflammation in atherosclerosis and vascular inflammation. Yet, TRAFs show a highly complex interplay between different TRAF-family members with partially opposing and overlapping functions that are determined by the level of cellular expression, concomitant signaling events, and the context of the disease. Therefore, inhibition of specific TRAFs may be beneficial in one condition and harmful in others. Here, we carefully discuss the cellular expression and signaling events of TRAFs and evaluate their role in vascular inflammation and atherosclerosis. We also highlight metabolic effects of TRAFs and discuss the development of TRAF-based therapeutics in the future.
Collapse
Affiliation(s)
- Mark Colin Gissler
- Cardiology and Angiology, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
- Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Peter Stachon
- Cardiology and Angiology, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
| | - Dennis Wolf
- Cardiology and Angiology, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
- Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
- *Correspondence: Dennis Wolf
| | - Timoteo Marchini
- Cardiology and Angiology, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
- Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
- Universidad de Buenos Aires, CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Buenos Aires, Argentina
| |
Collapse
|
11
|
Wang J, Wang Y. Circular RNA cerebellar degeneration-related protein 1 antisense RNA (Circ-CDR1as) downregulation induced by dexmedetomidine treatment protects hippocampal neurons against hypoxia/reoxygenation injury through the microRNA-28-3p (miR-28-3p)/tumor necrosis factor receptor-associated factor-3 (TRAF3) axis. Bioengineered 2021; 12:10512-10524. [PMID: 34787053 PMCID: PMC8810102 DOI: 10.1080/21655979.2021.1999369] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Cerebral ischemia/reperfusion (CI/R) injury results in serious brain tissue damage, thereby leading to long-term disability and mortality. It has been reported that dexmedetomidine (DEX) exerted neuroprotective effects in CI/R injury. Herein, we intended to investigate whether and how circular RNA (circRNA) cerebellar degeneration-related protein 1 antisense RNA (circ-CDR1as) was involved in the DEX-mediated protection on hippocampal neurons. In our work, the mouse hippocampal neuronal cells (HT-22) were used to construct a hypoxia/reperfusion (H/R) model for CI/R injury. Cell proliferation and apoptosis were evaluated by CCK-8 and flow cytometry. Gene expressions were detected by RT-qPCR. Levels of pro-inflammatory cytokines (TNF-α, IL-6, and IL-1β) were measured by ELISA. The association between miR-28-3p and circ-CDR1as or TRAF3 was verified by dual-luciferase assay. The results indicated that DEX alleviated HT-22 cell dysfunction induced by H/R treatment. In addition, circ-CDR1as was downregulated after DEX treatment and reversed the effects of DEX on the proliferation, apoptosis, and inflammatory responses of H/R-treated HT-22 cells. Circ-CDR1as positively regulated TRAF3 expression via interaction with miR-28-3p in HT-22 cells. Circ-CDR1as aggravated H/R-treated HT-22 cell dysfunction through targeting miR-28-3p. Furthermore, TRAF3 inhibition partly abolished the effect of circ-CDR1as overexpression on cellular activities of H/R-treated HT-22 cells. To sum up, our findings, for the first time, demonstrated that DEX exerted neuroprotective effects on hippocampal neurons against H/R treatment via the circ-CDR1as/miR-28-3p/TRAF3 regulatory network, providing novel therapeutic targets for DEX administration in CI/R treatment.
Collapse
Affiliation(s)
- Junhua Wang
- Department of Anesthesia, Liyang People's Hospital, Changzhou, P.R. China
| | - Ying Wang
- Department of Anesthesia, Liyang People's Hospital, Changzhou, P.R. China
| |
Collapse
|
12
|
Pan D, Lyu Y, Zhang N, Wang X, Lei T, Liang Z. RIP2 knockdown inhibits cartilage degradation and oxidative stress in IL-1β-treated chondrocytes via regulating TRAF3 and inhibiting p38 MAPK pathway. Clin Immunol 2021; 232:108868. [PMID: 34587513 DOI: 10.1016/j.clim.2021.108868] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 09/14/2021] [Accepted: 09/23/2021] [Indexed: 01/04/2023]
Abstract
Receptor-interacting protein 2 (RIP2) is a key mediator implicated in multiple cellular processes, and its dysregulation has been recently reported in colitis, asthma and other inflammatory diseases. However, the effects of RIP2 on osteoarthritis (OA) and the underlying mechanisms remain unclear. In this study, we found that RIP2 expression was upregulated in human articular cartilage tissues with OA and interleukin-1β (IL-1β)-treated chondrocytes. Knockdown of RIP2 inhibited IL-1β-induced extracellular matrix (ECM) and oxidative stress. Moreover, knockdown of TRAF3 reversed the effects of RIP2 silencing on cartilage degradation and oxidative stress in IL-1β-induced chondrocytes. In addition, p38 mitogen-activated protein kinase (MAPK) activator dehydrocorydalmine chloride (Dc) also reversed the effects of RIP2 silencing on IL-1β-induced chondrocytes. Taken together, our data reveal that RIP2 knockdown inhibits cartilage degradation and oxidative stress in IL-1β-treated chondrocytes by regulating TRAF3 expression and p38 MAPK pathway activation.
Collapse
Affiliation(s)
- DongSheng Pan
- Department of Orthopaedics, Xijing Hospital, Air Force Military Medical University, Xi'an 710032, Shaanxi, China
| | - Yanhong Lyu
- Department of Gynecology and Obstetrics, Xijing Hospital, Air Force Military Medical University, Xi'an 710032, Shaanxi, China
| | - Na Zhang
- Department of Orthopaedics, Xijing Hospital, Air Force Military Medical University, Xi'an 710032, Shaanxi, China
| | - Xuankang Wang
- Department of Orthopaedics, Xijing Hospital, Air Force Military Medical University, Xi'an 710032, Shaanxi, China
| | - Tao Lei
- School of Biomedical Engineering, Air Force Military Medical University, Xi'an 710032, Shaanxi, China.
| | - Zhuowen Liang
- Department of Orthopaedics, Xijing Hospital, Air Force Military Medical University, Xi'an 710032, Shaanxi, China.
| |
Collapse
|
13
|
MicroRNA MiR-27a-5p Alleviates the Cerulein-Induced Cell Apoptosis and Inflammatory Injury of AR42J Cells by Targeting Traf3 in Acute Pancreatitis. Inflammation 2021; 43:1988-1998. [PMID: 32647955 DOI: 10.1007/s10753-020-01272-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Acute pancreatitis (AP), a sudden inflammatory process of pancreas, is painful and may contribute to death. The aberrant expression of miR-27a-5p has been reported in many types of cancers and diseases including AP. Thus, it is urgent to manifest the functions and mechanism of miR-27a-5p in AP. The levels of miR-27a-5p, tumor necrosis factor (TNF) receptor-associated factor 3 (Traf3) in serum of AP patient, or cerulein-treated AR42J cells were detected by qRT-PCR. Functionally, the apoptotic rate, the protein levels of Bcl-2 and Bax, the caspase-3 activity, and the levels of IL-1β, IL-6, and TNF-α in cerulein-treated AR42J cells were measured by flow cytometry, Western blot, caspase-3 activity assay, and qRT-PCR and ELISA assay, respectively. In addition, the putative target of miR-27a-5p was predicted by TargetScan online database, and the dual luciferase reporter assay and RNA immunoprecipitation (RIP) assay were conducted to verify this interaction. Cerulein-treated mouse AP model was established to explore the role of miR-27a-5p in AP in vivo. The level of miR-27a-5p was notably downregulated in AP patients and cerulein-treated AR42J cells. The functional experiments indicated that miR-27a-5p mimics attenuated the promotion effects on cell apoptosis and the inflammatory response in AR42J cells caused by cerulein. The interaction between miR-27a-5p and Traf3 was predicted by TargetScan online database and validated by dual luciferase reporter assay and RIP assay. Following qRT-PCR results exhibited that Traf3 was apparently enhanced in cerulein-treated AR42J cells. The further functional experiments disclosed that Traf3 overexpression relieved the inhibitory effects on cell apoptosis and the inflammatory response induced by miR-27a-5p mimics in cerulein-treated AR42J cells. Moreover, miR-27a-5p alleviated cerulein-induced injury in vivo. In this study, we established the cerulein-treated AR42J cells as AP model in vitro. We validated that miR-27a-5p was significantly downregulated, and Traf3 was strikingly upregulated in AP patient and/or cerulein-treated AR42J cells. The further mechanistical and functional experiments unraveled that miR-27a-5p regulated Traf3 to relieve the cerulein-induced cell apoptosis and inflammatory injury of AR42J cells. Therefore, this novel regulatory network may provide therapeutic target for AP patients.
Collapse
|
14
|
LncRNA Gm4419 Regulates Myocardial Ischemia/Reperfusion Injury Through Targeting the miR-682/TRAF3 Axis. J Cardiovasc Pharmacol 2021; 76:305-312. [PMID: 32590403 DOI: 10.1097/fjc.0000000000000867] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Myocardial cell death during acute myocardial infarction occurs because of acute ischemia, persistent ischemia, reperfusion-associated injury, and the inflammatory infiltrate as a response to cell necrosis. In the present study, quantitative real-time PCR showed that lncRNA Gm4419 was highly upregulated in ischemia/reperfusion myocardial tissues and hypoxia/reoxygenation H9C2 cells, whereas miR-682 was downregulated. Knocking down Gm4419 with sh-Gm4419 resulted in the rescue of myocardial infarction and apoptosis induced by ischemia/reperfusion or hypoxia/reoxygenation. Our study further demonstrated that Gm4419 may bind with miR-682 directly. Moreover, in vitro experiments further demonstrated that miR-682 could bind to tumor necrosis factor receptor-associated factor 3 (TRAF3) directly. Most importantly, TRAF3 overexpression could counteract the effect of sh-Gm4419. Taken together, our study indicated that Gm4419 may target miR-682 via sponging to increase TRAF3 expression, thereby contributing to myocardial I/R injury. Therefore, the Gm4419/miR-682/TRAF3 axis may be an important regulatory mechanism in myocardial ischemia/reperfusion injury.
Collapse
|
15
|
Liu Y, Zhou P, Wang F, Zhang X, Yang D, Hong L, Ruan D. Inhibition of lncRNA SNHG8 plays a protective role in hypoxia-ischemia-reoxygenation-induced myocardial injury by regulating miR-335 and RASA1 expression. Mol Med Rep 2021; 24:597. [PMID: 34165173 PMCID: PMC8240175 DOI: 10.3892/mmr.2021.12236] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 05/12/2021] [Indexed: 12/24/2022] Open
Abstract
Long non-coding (lnc)RNAs serve a role in a number of diseases, including different types of cancer and acute myocardial infarction. The aim of the present study was to investigate the protective role of lncRNA small nucleolar RNA host gene 8 (SNHG8) in hypoxia-ischemia-reoxygenation (HI/R)-induced myocardial injury and its potential mechanism of action. Cell viability, proliferation, creatine kinase myocardial band, cell apoptosis and protein expression levels were determined by Cell Counting Kit-8 assay, EdU assay, ELISA, flow cytometry and western blotting, respectively. The association between SNHG8 and microRNA (miR)-335 was confirmed using a dual-luciferase reporter gene assay. The effects of the miR-335 inhibitor transfections had on increasing apoptosis and decreasing H9C2 cell viability were reversed in cells co-transfected with SNHG8 small interfering (si)RNA. Furthermore, it was found that miR-335 could regulate RAS p21 protein activator 1 (RASA1) expression and that transfection with SNHG8 siRNA downregulated RASA1 expression. Silencing of RASA1 protected against HI/R-induced H9C2 cell injury. However, SNHG8 siRNA did not further reduce apoptosis, demonstrating that SNHG8 may act through RASA1, and RASA1 may mediate the protection of SNHG8 siRNA in HI/R myocardial injury. Thus, inhibition of lncRNA SNHG8 alleviated HI/R-induced myocardial damage by regulating miR-335 and RASA1.
Collapse
Affiliation(s)
- Yanfeng Liu
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Ping Zhou
- Department of Cardiology, Yifeng County People's Hospital, Yichun, Jiangxi 336300, P.R. China
| | - Fengxiao Wang
- Department of Cardiology, Jiangxi Huimin Hospital, Nanchang, Jiangxi 330046, P.R. China
| | - Xuehong Zhang
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Dongmei Yang
- Department of Cardiovascular Surgery, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Lang Hong
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Dongyun Ruan
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
16
|
Zhou W, Lin D, Zhong Z, Ye Q. Roles of TRAFs in Ischemia-Reperfusion Injury. Front Cell Dev Biol 2020; 8:586487. [PMID: 33224951 PMCID: PMC7674171 DOI: 10.3389/fcell.2020.586487] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/05/2020] [Indexed: 12/13/2022] Open
Abstract
Tumor necrosis factor receptor-associated factor (TRAF) proteins are a family of signaling molecules that function downstream of multiple receptor signaling pathways, and they play a pivotal role in the regulation of intracellular biological progresses. These TRAF-dependent signaling pathways and physiological functions have been involved in the occurrence and progression of ischemia-reperfusion injury (IRI), which is a common pathophysiological process that occurs in a wide variety of clinical events, including ischemic shock, organ transplantation, and thrombolytic therapy, resulting in a poor prognosis and high mortality. IRI occurs in multiple organs, including liver, kidney, heart, lung, brain, intestine, and retina. In recent years, mounting compelling evidence has confirmed that the genetic alterations of TRAFs can cause subversive phenotype changes during IRI of those organs. In this review, based on current knowledge, we summarized and analyzed the regulatory effect of TRAFs on the IRI of various organs, providing clear direction and a firm theoretical basis for the development of treatment strategies to manipulate TRAF proteins or TRAF-dependent signaling pathways in IRI-related diseases.
Collapse
Affiliation(s)
- Wei Zhou
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
| | - Danni Lin
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China.,The First Affiliated Hospital, Zhejiang University School of Medicine, Department of Hepatobiliary and Pancreatic Surgery, Zhejiang Provincial Key Laboratory of Pancreatic Disease, Innovation Center for the Study of Pancreatic Diseases, Hangzhou, China
| | - Zibiao Zhong
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
| | - Qifa Ye
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China.,The Third Xiangya Hospital of Central South University, Research Center of National Health Ministry on Transplantation Medicine Engineering and Technology, Changsha, China
| |
Collapse
|
17
|
Gutiérrez-Venegas G, Fernández-Rojas B, Rosas-Martínez M, Sánchez-Carballido MA. Rutin Prevents LTA Induced Oxidative Changes in H9c2 Cells. Prev Nutr Food Sci 2020; 25:203-211. [PMID: 32676472 PMCID: PMC7333009 DOI: 10.3746/pnf.2020.25.2.203] [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: 11/07/2019] [Accepted: 03/20/2020] [Indexed: 12/29/2022] Open
Abstract
Lipoteichoic acid (LTA), a component of Gram-positive bacteria cell walls is involved in infective endocarditis (IE), a life-threatening disease. We evaluated for the first time, whether flavonoid rutin (quercetin-3-rutinoside) can block LTA-induced pro-inflammatory response and reactive oxygen species (ROS) production, and reduction of antioxidant enzymes. We found that rutin suppresses LTA effects on the antioxidant enzymes superoxide dismutase, catalase, and glutathione peroxidase, as well as the pro-inflammatory enzyme cyclooxygenase-2, preventing phosphorylation of the mitogen-activated protein kinases (MAPKs), p38, and c-Jun N-terminal kinase, and the increase of ROS production induced by LTA. Taken together, these findings suggest that rutin prevents oxidative damage, inflammation, and MAPKs activation induced by LTA. Rutin may exert a protective effect in IE. These data provide novel insights for future use of rutin to prevent the mechanisms of LTA-related pathogenesis, inflammatory processes, and antioxidant enzyme levels in diseases such as IE.
Collapse
Affiliation(s)
- Gloria Gutiérrez-Venegas
- Biochemistry Laboratory of the Division of Graduate Studies and Research, Faculty of Dentistry, National Autonomous University of Mexico, Mexico City 04510, Mexico
| | - Berenice Fernández-Rojas
- Biochemistry Laboratory of the Division of Graduate Studies and Research, Faculty of Dentistry, National Autonomous University of Mexico, Mexico City 04510, Mexico
| | - Marisol Rosas-Martínez
- Biochemistry Laboratory of the Division of Graduate Studies and Research, Faculty of Dentistry, National Autonomous University of Mexico, Mexico City 04510, Mexico
| | - Manuel Alejandro Sánchez-Carballido
- Biochemistry Laboratory of the Division of Graduate Studies and Research, Faculty of Dentistry, National Autonomous University of Mexico, Mexico City 04510, Mexico
| |
Collapse
|
18
|
Zhao Z, Tang Z, Zhang W, Liu J, Li B, Ding S. Inactivated pseudomonas aeruginosa protects against myocardial ischemia reperfusion injury via Nrf2 and HO-1. Exp Ther Med 2020; 19:3362-3368. [PMID: 32266034 DOI: 10.3892/etm.2020.8605] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 06/27/2019] [Indexed: 12/22/2022] Open
Abstract
The current study investigated the protective effects of inactivated pseudomonas aeruginosa (IPA) on myocardial ischemia reperfusion injury (MIR/I) and the mechanisms governing this interaction. Left anterior descending coronary artery ligation was performed on rats for 30 min and reperfusion was performed for a subsequent 2 h. Rat hearts were obtained and the myocardial infarction area was determined using nitroblue tetrazolium. Myocardial cell apoptosis was determined using flow cytometry. Malondialdehyde (MDA) content, lactate dehydrogenase (LDH) activity, superoxide dismutase (SOD) activity and catalase (CAT) activities were assayed using the corresponding kits. Additionally, nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase 1 (HO-1) were assayed using western blot and immunofluorescence analysis. When compared with the model group, the results of IPA treatment revealed improved heart function, reduced myocardial infarction area and reduced endothelial cell apoptosis, which led to decreased LDH and MDA levels, and increased SOD and CAT levels in serum, and decreased LDH and MDA levels and increased SOD and CAT in myocardial tissues. Moreover, increased Nrf2 and HO-1 expression levels in the myocardial tissues were also observed at all concentrations of IPA. It was concluded that IPA pretreatment ameliorated MIR/I and reduced endothelial apoptosis and oxidative stress via the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Zhigang Zhao
- Emergency Department, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430000, P.R. China
| | - Zhongzhi Tang
- Emergency Department, General Hospital of Central Theater Command, Wuhan, Hubei 430070, P.R. China
| | - Wenkai Zhang
- Emergency Department, General Hospital of Central Theater Command, Wuhan, Hubei 430070, P.R. China
| | - Jie Liu
- Emergency Department, General Hospital of Central Theater Command, Wuhan, Hubei 430070, P.R. China
| | - Bo Li
- Emergency Department, General Hospital of Central Theater Command, Wuhan, Hubei 430070, P.R. China
| | - Shifang Ding
- Cardiovascular Department, General Hospital of Central Theater Command, Wuhan, Hubei 430070, P.R. China
| |
Collapse
|
19
|
Wang Y, Wang D, Jin Z. miR‑27a suppresses TLR4‑induced renal ischemia‑reperfusion injury. Mol Med Rep 2019; 20:967-976. [PMID: 31173204 PMCID: PMC6625210 DOI: 10.3892/mmr.2019.10333] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 03/11/2019] [Indexed: 12/20/2022] Open
Abstract
Ischemia reperfusion injury (IRI) is one of the primary causes of acute renal injury and even acute renal failure. An increasing body of evidence has indicated that the aberrant expression of microRNAs (miRNA/miR) is closely associated with the progression of IRI. In the process of renal IRI, inflammatory reactions, cell adhesion and the death of renal tubular epithelial cells serve important roles. The present study investigated the expression of renal miRNAs following renal IRI in an attempt to identify the miRNAs that exert pivotal functions in renal IRI. The present study revealed that miR-27a, which was downregulated in IRI, and the 3′-untranslated region (UTR) of Toll-like receptor 4 (TLR4) have associated binding sites. The levels of TLR4, miR-27a and specific associated proteins in the renal tissues of gestational rats were determined by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis, immunohistochemistry and western blotting. The associations between miR-27a and TLR4 were analyzed, and the regulatory effect of miR-27a on TLR4 was detected via a luciferase reporter gene assay, western blotting and RT-qPCR in vivo and in vitro. In addition, the present study demonstrated that miR-27a suppressed the expression of TLR4 by binding to the 3′UTR of TLR4. The overexpression of miR-27a downregulated the expression of TLR4, which in turn inhibited the inflammation, cell adhesion and cell death in IRI. Therefore, miR-27a inhibited inflammation in IRI by decreasing the expression of TLR4.
Collapse
Affiliation(s)
- Yang Wang
- Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Dan Wang
- Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Zhen Jin
- Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|