1
|
Brown N, Roman M, Miller D, Murphy G, Woźniak MJ. A Systematic Review and Meta-Analysis of MicroRNA as Predictive Biomarkers of Acute Kidney Injury. Biomedicines 2024; 12:1695. [PMID: 39200160 PMCID: PMC11351452 DOI: 10.3390/biomedicines12081695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/23/2024] [Accepted: 07/28/2024] [Indexed: 09/01/2024] Open
Abstract
Acute kidney injury (AKI) affects 10-15% of hospitalised patients and arises after severe infections, major surgeries, or exposure to nephrotoxic drugs. AKI diagnosis based on creatinine level changes lacks specificity and may be delayed. MicroRNAs are short non-coding RNA secreted by all cells. This review of studies measuring miRNAs in AKI aimed to verify miRNAs as diagnostic markers. The study included data from patients diagnosed with AKI due to sepsis, ischaemia, nephrotoxins, radiocontrast, shock, trauma, and cardiopulmonary bypass. Out of 71 studies, the majority focused on AKI in sepsis patients, followed by cardiac surgery patients, ICU patients, and individuals receiving nephrotoxic agents or experiencing ischaemia. Studies that used untargeted assays found 856 differentially regulated miRNAs, although none of these were confirmed by more than one study. Moreover, 68 studies measured miRNAs by qRT-PCR, and 2 studies reported downregulation of miR-495-3p and miR-370-3p in AKI patients with sepsis after the AKI diagnosis. In three studies, upregulation of miR-21 was reported at the time of the AKI diagnosis with a significant pooled effect of 0.56. MiR-21 was also measured 19-24 h after cardiac surgery in three studies. However, the pooled effect was not significant. Despite the considerable research into miRNA in AKI, there is a knowledge gap in their applicability as diagnostic markers of AKI in humans.
Collapse
Affiliation(s)
| | | | | | | | - Marcin J. Woźniak
- Department of Cardiovascular Sciences and NIHR Cardiovascular Biomedical Research Unit, Glenfield Hospital, University of Leicester, Leicester LE3 9QP, UK; (N.B.); (M.R.); (D.M.); (G.M.)
| |
Collapse
|
2
|
Chen B, Dong L, Zhang J, Hao Y, Chi W, Song D. Exploring shared pathways and the shared biomarker ERRFI1 in Obstructive sleep apnoea and atherosclerosis using integrated bioinformatics analysis. Sci Rep 2023; 13:15103. [PMID: 37699925 PMCID: PMC10497545 DOI: 10.1038/s41598-023-42184-0] [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: 04/13/2023] [Accepted: 09/06/2023] [Indexed: 09/14/2023] Open
Abstract
Obstructive sleep apnea (OSA) is an upper airway disorder occurring during sleep and is associated with atherosclerosis (AS). AS is a cardiovascular disease caused by environmental and genetic factors, with a high global mortality rate. This study investigated common pathways and potential biomarkers of OSA and AS. Microarray data were downloaded from the Gene Expression Omnibus (GEO) database and used to screen for differentially expressed genes (DEGs) in the OSA and AS datasets. A weighted gene co-expression network analysis (WGCNA) was used to identify the co-expression modules of OSA and AS. The least absolute shrinkage and selection operators (LASSO) were used to determine critical biomarkers. Immune cell infiltration analysis was used to investigate the correlation between immune cell infiltration and common biomarkers of OSA and AS. Results revealed that differentially expressed genes may be involved in inflammatory processes, chemokine signaling pathways, and molecular changes in cell adhesion. ERBB receptor feedback inhibitor 1 (ERRFI1) was the best-shared biomarker for OSA and AS. Immune infiltration analysis showed that ERRFI1 expression was correlated with immune cell changes. Changes in immune pathways, inflammatory processes, and cell adhesion molecules may underlie the pathogenesis of both diseases, and ERRFI1 may be a potential diagnostic marker for patients with OSA and AS.
Collapse
Affiliation(s)
- Bowen Chen
- Clinical Biobank, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Liping Dong
- Clinical Biobank, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jihua Zhang
- Department of Otolaryngology, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ying Hao
- Clinical Biobank, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Weiwei Chi
- Clinical Biobank, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Dongmei Song
- Clinical Biobank, The First Hospital of Hebei Medical University, Shijiazhuang, China.
- Department of Otolaryngology, The First Hospital of Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
3
|
Tsuji K, Nakanoh H, Fukushima K, Kitamura S, Wada J. MicroRNAs as Biomarkers and Therapeutic Targets for Acute Kidney Injury. Diagnostics (Basel) 2023; 13:2893. [PMID: 37761260 PMCID: PMC10529274 DOI: 10.3390/diagnostics13182893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/04/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Acute kidney injury (AKI) is a clinical syndrome where a rapid decrease in kidney function and/or urine output is observed, which may result in the imbalance of water, electrolytes and acid base. It is associated with poor prognosis and prolonged hospitalization. Therefore, an early diagnosis and treatment to avoid the severe AKI stage are important. While several biomarkers, such as urinary L-FABP and NGAL, can be clinically useful, there is still no gold standard for the early detection of AKI and there are limited therapeutic options against AKI. miRNAs are non-coding and single-stranded RNAs that silence their target genes in the post-transcriptional process and are involved in a wide range of biological processes. Recent accumulated evidence has revealed that miRNAs may be potential biomarkers and therapeutic targets for AKI. In this review article, we summarize the current knowledge about miRNAs as promising biomarkers and potential therapeutic targets for AKI, as well as the challenges in their clinical use.
Collapse
Affiliation(s)
- Kenji Tsuji
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Hiroyuki Nakanoh
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Kazuhiko Fukushima
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
- Program in Membrane Biology, Center for Systems Biology, Department of Medicine, Division of Nephrology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Shinji Kitamura
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
- Department of Nursing Science, Faculty of Health and Welfare Science, Okayama Prefectural University, Okayama 719-1197, Japan
| | - Jun Wada
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| |
Collapse
|
4
|
Yu JT, Fan S, Li XY, Hou R, Hu XW, Wang JN, Shan RR, Dong ZH, Xie MM, Dong YH, Shen XY, Jin J, Wen JG, Liu MM, Wang W, Meng XM. Novel insights into STAT3 in renal diseases. Biomed Pharmacother 2023; 165:115166. [PMID: 37473682 DOI: 10.1016/j.biopha.2023.115166] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 07/07/2023] [Accepted: 07/11/2023] [Indexed: 07/22/2023] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is a cell-signal transcription factor that has attracted considerable attention in recent years. The stimulation of cytokines and growth factors can result in the transcription of a wide range of genes that are crucial for several cellular biological processes involved in pro- and anti-inflammatory responses. STAT3 has attracted considerable interest as a result of a recent upsurge in study because of their role in directing the innate immune response and sustaining inflammatory pathways, which is a key feature in the pathogenesis of many diseases, including renal disorders. Several pathological conditions which may involve STAT3 include diabetic nephropathy, acute kidney injury, lupus nephritis, polycystic kidney disease, and renal cell carcinoma. STAT3 is expressed in various renal tissues under these pathological conditions. To better understand the role of STAT3 in the kidney and provide a theoretical foundation for STAT3-targeted therapy for renal disorders, this review covers the current work on the activities of STAT3 and its mechanisms in the pathophysiological processes of various types of renal diseases.
Collapse
Affiliation(s)
- Ju-Tao Yu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Shuai Fan
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei 230032 China; Department of Urology, Institute of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032 China
| | - Xiang-Yu Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Rui Hou
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Xiao-Wei Hu
- Department of Clinical Pharmacy, Anhui Provincial Children's Hospital, Hefei 230051, China
| | - Jia-Nan Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Run-Run Shan
- School of Life Sciences, Anhui Medical University, Hefei 230032, China
| | - Ze-Hui Dong
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Man-Man Xie
- School of Life Sciences, Anhui Medical University, Hefei 230032, China
| | - Yu-Hang Dong
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Xiao-Yu Shen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Juan Jin
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei 230032, China
| | - Jia-Gen Wen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Ming-Ming Liu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Wei Wang
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei 230032 China; Department of Urology, Institute of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032 China.
| | - Xiao-Ming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
5
|
Fu D, Zhao H, Huang Y, Li J, Feng H, Li A, Liu Y, He L. Metformin regulates the effects of IR and IGF-1R methylation on mast cell activation and airway reactivity in diabetic rats with asthma through miR-152-3p/DNMT1 axis. Cell Biol Toxicol 2023; 39:1851-1872. [PMID: 36547818 DOI: 10.1007/s10565-022-09787-1] [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: 07/28/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND/AIM Metformin is a drug for treating type 2 diabetes mellitus (T2DM). Recently, metformin has been shown to reduce the risks of asthma-associated outcomes and asthma deterioration, thereby holding promise as a superior medicine for diabetic patients with asthma. However, the mechanism by which metformin reduces diabetic asthma is yet to be clarified. This study aimed at ascertaining the downstream molecules underlying the effect of metformin on the activation of mast cells (MCs) and airway reactivity in a concomitant diabetic and asthmatic rat model. METHODS A T2DM model was induced utilizing a high-fat diet and streptozotocin. Then, 10% ovalbumin was utilized to stimulate asthma-like pathology in the T2DM rats. RBL-2H3 cells were induced by anti-dinitrophenyl-specific immunoglobulin E for constructing an in vitro model. Luciferase assay and RNA immunoprecipitation (IP) assay were conducted to identify the interaction between microRNA-152-3p (miR-152-3p) and DNA methyltransferase 1 (DNMT1), while chromatin IP to identify the binding of DNMT1 to insulin receptor (IR) and insulin-like growth factor 1 receptor (IGF-1R) promoters. The effects of metformin on both pathological changes in vivo and biological behaviors of cells were evaluated. Using gain- and loss-of-function approaches, we assessed the role of the two interactions in the metformin-induced effect. RESULTS It was suggested that metformin could impede the MC activation and airway resistance in the concomitant diabetic and asthmatic rats. Additionally, metformin downregulated IR and IGF-1R through DNMT1-dependent methylation to repress MC activation and airway resistance. DNMT1 was testified to be a target gene of miR-152-3p. Furthermore, miR-152-3p-induced silencing of DNMT1 was blocked by metformin, hence restraining MC activation and airway resistance. CONCLUSION The findings cumulatively demonstrate that metformin downregulates IR/IGF-1R to block MC activation and airway resistance via impairing the binding affinity between miR-152-3p and DNMT1.
Collapse
Affiliation(s)
- Dan Fu
- Department of Endocrinology, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Hailu Zhao
- Diabetic Systems Center, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin, Guangxi, 541000, People's Republic of China
| | - Yan Huang
- Department of Anesthesiology, The Fifth Affiliated Hospital of Southern Medical University, No.566, Congcheng Ave, Guangzhou, Guangdong, 510900, People's Republic of China
| | - Jingjuan Li
- Department of Anesthesiology, The Fifth Affiliated Hospital of Southern Medical University, No.566, Congcheng Ave, Guangzhou, Guangdong, 510900, People's Republic of China
| | - Huafeng Feng
- Department of Anesthesiology, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, 541001, People's Republic of China
| | - Aiguo Li
- Department of Anesthesiology, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, 541001, People's Republic of China
| | - Yefen Liu
- Department of Anesthesiology, The Fifth Affiliated Hospital of Southern Medical University, No.566, Congcheng Ave, Guangzhou, Guangdong, 510900, People's Republic of China
| | - Liang He
- Department of Anesthesiology, The Fifth Affiliated Hospital of Southern Medical University, No.566, Congcheng Ave, Guangzhou, Guangdong, 510900, People's Republic of China.
| |
Collapse
|
6
|
Wang X, Liu Y, Lei P. LncRNA HOTAIRM1 promotes osteogenic differentiation of human bone marrow-derived mesenchymal stem cells by targeting miR-152-3p/ETS1 axis. Mol Biol Rep 2023:10.1007/s11033-023-08466-6. [PMID: 37171551 DOI: 10.1007/s11033-023-08466-6] [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: 04/02/2022] [Accepted: 04/18/2023] [Indexed: 05/13/2023]
Abstract
BACKGROUND Bone marrow mesenchymal stem cells (BMSCs) can differentiate into osteoblasts and thus present a tremendous therapeutic potential in osteoporosis. Here, we elucidated the involvement of long non-coding RNAs (lncRNAs) HOXA transcript antisense RNA, myeloid-specific 1 (HOTAIRM1) in the osteogenic differentiation of BMSCs. METHODS AND RESULTS The expression levels of HOTAIRM1, miR-152-3p, ETS proto-oncogene 1 (ETS1), runt-related transcription factor 2 (RUNX2), Osterix, and osteocalcin (OCN) were determined by a quantitative real-time polymerase chain reaction (qRT-PCR) or western blot method. Targeted relationship between miR-152-3p and HOTAIRM1 or ETS1 was confirmed by dual-luciferase reporter and RNA pull-down assays. The activity of alkaline phosphatase (ALP) was measured by the ALP Activity Assay Kit. The extent of the calcium deposition was assessed by Alizarin Red Staining. Our data showed that HOTAIRM1 and ETS1 levels were up-regulated and miR-152-3p expression was down-regulated during osteogenic differentiation of human BMSCs (HBMSCs). HOTAIRM1 overexpression enhanced osteogenic differentiation of HBMSCs, and decreased level of HOTAIRM1 suppressed osteogenic differentiation of HBMSCs. HOTAIRM1 directly targeted miR-152-3p. ETS1 was identified as a direct and functional target of miR-152-3p. Furthermore, HOTAIRM1 functioned as a post-transcriptional regulator of ETS1 expression by miR-152-3p. CONCLUSION The findings in this paper identify HOTAIRM1 as a novel regulator of osteogenic differentiation of BMSCs by the regulation of miR-152-3p/ETS1 axis, uncovering HOTAIRM1 as a promising therapeutic strategy for osteoporosis.
Collapse
Affiliation(s)
- Xuan Wang
- Department of Geriatrics, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin City, 300052, China
| | - Yan Liu
- Department of Orthopedics, Tianjin Union Medical Center, Tianjin City, China
| | - Ping Lei
- Department of Geriatrics, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin City, 300052, China.
| |
Collapse
|
7
|
SIRT7 silencing by miR-152-3p confers cell apoptosis and renal functional impairment induced by renal ischaemia/reperfusion injury. Int Urol Nephrol 2023; 55:367-376. [PMID: 35941302 DOI: 10.1007/s11255-022-03330-1] [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: 03/04/2022] [Accepted: 07/24/2022] [Indexed: 01/25/2023]
Abstract
PURPOSE Acute kidney injury (AKI) induced by renal ischaemia/reperfusion (I/R) during renal transplantation has been reported to be linked to the regulation of SIRT2, one of the members of SIRTUINS family. Current work is attempted to explore the influence and mechanism of SIRT7 in renal cell apoptosis controlled by miR-152-3p during renal I/R injury. METHODS Three databases were used to select the miRNAs regulating the expression of SIRT7. Overexpression and inhibition of miR-152-3p and Luciferase assay were employed to certify the modulation of miR-152-3p to SIRT7 in cells. RT-qPCR assay was used to measure the mRNA levels. Western blot assay was employed to determine the expression of proteins. TUNEL assay and Flow Cytometry were conducted to analyze cell apoptosis. RESULTS SIRT7 expression decreased in tissues of AKI patients and rats underwent renal I/R, which was associated with enhanced impairment of renal function. SIRT7 downregulation was attributed to the direct inhibition by miR-152-3p due to binding and inhibiting its seed sequence in 3'-UTR of SIRT7 mRNA. Consequently, the upregulation of miR-152-3p led to an inhibition of SIRT7 expression, an increase in expression of extrinsic apoptosis molecules containing FOXO3a, Bim, and caspase3, and apoptotic renal cells; while miR-152-3p inhibition abolished these phenotypes. CONCLUSION SIRT7 downregulation by miR-152-3p is a leading cause of renal cell apoptosis and functional impairment induced by renal I/R. Inhibition of miR-152-3p to restore SIRT7 expression can be a promising strategy against renal I/R injury.
Collapse
|
8
|
Cui H, Ren G, Hu X, Xu B, Li Y, Niu Z, Mu L. Suppression of lncRNA GAS6-AS2 alleviates sepsis-related acute kidney injury through regulating the miR-136-5p/OXSR1 axis in vitro and in vivo. Ren Fail 2022; 44:1070-1082. [PMID: 35793478 PMCID: PMC9272941 DOI: 10.1080/0886022x.2022.2092001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Acute kidney injury (AKI) is a common complication of sepsis and increase morbidity and mortality. Long non-coding RNA (LncRNA) GAS6-AS2 was related to inflammation and apoptosis in different diseases by regulating miRNAs and downstream genes, but its role in AKI remains unclear. Thus, we speculated that GAS6-AS2 might function in sepsis-related AKI via regulating target genes. Here, LPS or CLP was used to establish in vitro or in vivo sepsis-related AKI model. The interactions between GAS6-AS2 and miR-136-5p, and miR-136-5p and OXSR1, were validated by luciferase reporter assay, RNA pull-down, or RIP assay. Cell apoptosis was determined by flow cytometry, Western blotting, or IHC. The kidney injury was evaluated by H&E staining. The expression of GAS6-AS2, miR-136-5p, and OXSR1 was determined by qRT-PCR or Western blotting. We found that GAS6-AS2 was up-regulated in LPS-treated HK2 cells and the CLP-induced rat model. In vitro, GAS6-AS2 knockdown decreased cleaved caspase-3 and bax expression and increased bcl-2 expression. The levels of TNF-α, IL-1β, and IL-6 were reduced by GAS6-AS2 down-regulation. GAS6-AS2 knockdown ameliorated oxidative stress in the cells, as indicated by the reduced ROS and MDA levels and the elevated SOD level. In vivo, GAS6-AS2 down-regulation decreased urinary NGAL and Kim-1 levels and serum sCr and BUN levels, and H&E proved that the kidney injury was alleviated. GAS6-AS2 knockdown also reduced apoptosis, inflammation, and oxidation induced by CLP in vivo. Mechanically, GAS6-AS2 sponged miR-136-5p which targeted OXSR1. Overall, lncRNA GAS6-AS2 knockdown has the potential to ameliorate sepsis-related AKI, and the mechanism is related to miR-136-5p/OXSR1 axis.
Collapse
Affiliation(s)
- Hongrui Cui
- Department of Nephrology, The First Hospital of Hebei Medical University, Shijiazhuang, PR China
| | - Guangwei Ren
- Department of Nephrology, The First Hospital of Hebei Medical University, Shijiazhuang, PR China
| | - Xiuhong Hu
- Department of Nephrology, The First Hospital of Hebei Medical University, Shijiazhuang, PR China
| | - Baozhen Xu
- Department of Nephrology, The First Hospital of Hebei Medical University, Shijiazhuang, PR China
| | - Yuping Li
- Department of Nephrology, The First Hospital of Hebei Medical University, Shijiazhuang, PR China
| | - Zheli Niu
- Department of Nephrology, The First Hospital of Hebei Medical University, Shijiazhuang, PR China
| | - Liqin Mu
- Department of General Practice, The First Hospital of Hebei Medical University, Shijiazhuang, PR China
| |
Collapse
|
9
|
Li F, Zhou F, Yang B. MicroRNA152-3p Protects Against Ischemia/Reperfusion-Induced Bbb Destruction Possibly Targeting the MAP3K2/JNK/c-Jun Pathway. Neurochem Res 2022; 48:1293-1304. [PMID: 36445489 PMCID: PMC10066145 DOI: 10.1007/s11064-022-03828-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/28/2022] [Accepted: 11/14/2022] [Indexed: 11/30/2022]
Abstract
AbstractIn the current study, we reported that overexpression of miR-152-3p effectively ameliorated neurological deficits and protected blood-brain barrier(BBB) integrity in middle cerebral artery occlusion (MCAO) rats. In an in vitro model, the level of miR-152-3p was significantly decreased in bEnd.3 cells after oxygen–glucose deprivation/reperfusion (OGD/R) insult. miR-152-3p overexpressing bEnd.3 cell monolayers were protected from OGD/R-induced microvascular hyperpermeability. The miR-152-3p-mediated protective effect was associated with lower apoptosis of endothelia by negatively modulating the MAP3K2/JNK/c-Jun pathway.
Collapse
Affiliation(s)
- Fei Li
- Department of Neurology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Fangfang Zhou
- Department of Neurology, 2nd Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Binbin Yang
- Department of Neurology, 2nd Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
10
|
Mahtal N, Lenoir O, Tinel C, Anglicheau D, Tharaux PL. MicroRNAs in kidney injury and disease. Nat Rev Nephrol 2022; 18:643-662. [PMID: 35974169 DOI: 10.1038/s41581-022-00608-6] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2022] [Indexed: 11/09/2022]
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that regulate gene expression by degrading or repressing the translation of their target messenger RNAs. As miRNAs are critical regulators of cellular homeostasis, their dysregulation is a crucial component of cell and organ injury. A substantial body of evidence indicates that miRNAs are involved in the pathophysiology of acute kidney injury (AKI), chronic kidney disease and allograft damage. Different subsets of miRNAs are dysregulated during AKI, chronic kidney disease and allograft rejection, which could reflect differences in the physiopathology of these conditions. miRNAs that have been investigated in AKI include miR-21, which has an anti-apoptotic role, and miR-214 and miR-668, which regulate mitochondrial dynamics. Various miRNAs are downregulated in diabetic kidney disease, including the miR-30 family and miR-146a, which protect against inflammation and fibrosis. Other miRNAs such as miR-193 and miR-92a induce podocyte dedifferentiation in glomerulonephritis. In transplantation, miRNAs have been implicated in allograft rejection and injury. Further work is needed to identify and validate miRNAs as biomarkers of graft function and of kidney disease development and progression. Use of combinations of miRNAs together with other molecular markers could potentially improve diagnostic or predictive power and facilitate clinical translation. In addition, targeting specific miRNAs at different stages of disease could be a promising therapeutic strategy.
Collapse
Affiliation(s)
- Nassim Mahtal
- Paris Cardiovascular Research Center - PARCC, Inserm, Université Paris Cité, Paris, France
| | - Olivia Lenoir
- Paris Cardiovascular Research Center - PARCC, Inserm, Université Paris Cité, Paris, France.
| | - Claire Tinel
- Service de Néphrologie et Transplantation Adulte, Hôpital Necker-Enfants Malades, Université Paris Cité, Assistance Publique-Hôpitaux de Paris, Paris, France.,Institut Necker-Enfants Malades, Inserm, Université Paris Cité, Paris, France
| | - Dany Anglicheau
- Service de Néphrologie et Transplantation Adulte, Hôpital Necker-Enfants Malades, Université Paris Cité, Assistance Publique-Hôpitaux de Paris, Paris, France.,Institut Necker-Enfants Malades, Inserm, Université Paris Cité, Paris, France
| | - Pierre-Louis Tharaux
- Paris Cardiovascular Research Center - PARCC, Inserm, Université Paris Cité, Paris, France.
| |
Collapse
|
11
|
Abstract
Sepsis-associated AKI is a life-threatening complication that is associated with high morbidity and mortality in patients who are critically ill. Although it is clear early supportive interventions in sepsis reduce mortality, it is less clear that they prevent or ameliorate sepsis-associated AKI. This is likely because specific mechanisms underlying AKI attributable to sepsis are not fully understood. Understanding these mechanisms will form the foundation for the development of strategies for early diagnosis and treatment of sepsis-associated AKI. Here, we summarize recent laboratory and clinical studies, focusing on critical factors in the pathophysiology of sepsis-associated AKI: microcirculatory dysfunction, inflammation, NOD-like receptor protein 3 inflammasome, microRNAs, extracellular vesicles, autophagy and efferocytosis, inflammatory reflex pathway, vitamin D, and metabolic reprogramming. Lastly, identifying these molecular targets and defining clinical subphenotypes will permit precision approaches in the prevention and treatment of sepsis-associated AKI.
Collapse
Affiliation(s)
- Shuhei Kuwabara
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia
| | - Eibhlin Goggins
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia
| | - Mark D Okusa
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
12
|
Han J, Liu X, Wang L. Dexmedetomidine protects against acute lung injury in mice via the DUSP1/MAPK/NF-κB axis by inhibiting miR-152-3p. Pulm Pharmacol Ther 2022:102131. [PMID: 35551994 DOI: 10.1016/j.pupt.2022.102131] [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] [Received: 10/27/2021] [Revised: 04/07/2022] [Accepted: 05/01/2022] [Indexed: 10/18/2022]
Abstract
OBJECTIVE Acute lung injury (ALI) is a debilitating condition in clinics. Dexmedetomidine (Dex) is known for its anti-apoptotic and anti-inflammatory properties. This study attempted to investigate the protective mechanism of Dex in ALI mice. METHODS Mice were pretreated with Dex before model establishment by tracheal injection of lipopolysaccharide (LPS). Pulmonary function indexes and wet-to-dry (W/D) ratio were measured. Pulmonary pathological changes were observed through HE staining, CD31+-positive mouse pulmonary microvascular endothelial cells (MPMVECs) were counted through immunofluorescence staining, and apoptosis was detected through TUNEL staining. miR-152-3p mimic, sh-DUSP1, or p38 MAPK inhibitor was delivered into MPMVECs, followed by combined treatment of Dex and LPS. miR-152-3p expression, apoptosis, levels of apoptosis- and MAPK/NF-κB pathway-associated proteins, and inflammatory factors were measured through RT-qPCR, flow cytometry, Western blot, and ELISA. The binding relationship of miR-152-3p and DUSP1 was verified through bioinformatics software and dual-luciferase assay. ALI mouse model was established after injection of miR-152-3p antagomir. RESULTS Dex improved ALI mouse pulmonary function and mitigated injury in mice and MPMVECs. miR-125-3p overexpression or sh-DUSP1 partially abolished the protection of Dex on MPMVECs. miR-152-3p targeted DUSP1. sh-DUSP1 partially averted the protection of Dex on MPMVECs. Dex inhibited the activation of the MAPK/NF-κB pathway in MPMVECs mediated by LPS, which was partially reversed by sh-DUSP1. The p38 MAPK inhibitor SB203580 antagonized the protective effect of Dex on MPMVECs mediated by sh-DUSP1. Similarly, downregulation of miR-152-3p mitigated ALI via the DUSP1/MAPK/NF-κB axis in vivo. CONCLUSION Dex relieved ALI in mice via the DUSP1/MAPK/NF-κB axis by down-regulating miR-152-3p.
Collapse
Affiliation(s)
- Jieran Han
- Department of Anesthesiology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Xiao Liu
- Department of Anesthesiology, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, 471003, China
| | - Ling Wang
- Department of Anesthesiology, 989 Hospital of JOINT Logistic Support Force of PLA, Luoyang, 471031, China.
| |
Collapse
|
13
|
Chen Y, Jing H, Tang S, Liu P, Cheng Y, Fan Y, Chen H, Zhou J. Non-Coding RNAs in Sepsis-Associated Acute Kidney Injury. Front Physiol 2022; 13:830924. [PMID: 35464083 PMCID: PMC9024145 DOI: 10.3389/fphys.2022.830924] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/08/2022] [Indexed: 11/21/2022] Open
Abstract
Sepsis is a systemic inflammatory response caused by a severe infection that leads to multiple organ damage, including acute kidney injury (AKI). In intensive care units (ICU), the morbidity and mortality associated with sepsis-associated AKI (SA-AKI) are gradually increasing due to lack of effective and early detection, as well as proper treatment. Non-coding RNAs (ncRNAs) exert a regulatory function in gene transcription, RNA processing, post-transcriptional translation, and epigenetic regulation of gene expression. Evidence indicated that miRNAs are involved in inflammation and programmed cell death during the development of sepsis-associated AKI (SA-AKI). Moreover, lncRNAs and circRNAs appear to be an essential regulatory mechanism in SA-AKI. In this review, we summarized the molecular mechanism of ncRNAs in SA-AKI and discussed their potential in clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Yanna Chen
- Department of Anesthesiology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Huan Jing
- Department of Anesthesiology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Simin Tang
- Department of Anesthesiology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Pei Liu
- Department of Anesthesiology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Ye Cheng
- Department of Anesthesiology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Youling Fan
- Department of Anesthesiology, The First People’s Hospital of Kashgar, Xinjiang, China
- Department of Anesthesiology, The Second People’s Hospital of Panyu, Guangzhou, China
| | - Hongtao Chen
- Department of Anesthesiology, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jun Zhou
- Department of Anesthesiology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Jun Zhou,
| |
Collapse
|
14
|
KLF4 Affects Acute Renal Allograft Injury via Binding to MicroRNA-155-5p Promoter to Regulate ERRFI1. DISEASE MARKERS 2022; 2022:5845627. [PMID: 35340414 PMCID: PMC8947908 DOI: 10.1155/2022/5845627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/12/2022] [Accepted: 02/15/2022] [Indexed: 11/18/2022]
Abstract
Kruppel-like factor 4 (KLF4) owns the promising potential in treating kidney injury, which inevitably occurs during renal allograft. Given that, this research targets to unveil KLF4-oriented mechanism from microRNA-155-5p/ERBB receptor feedback inhibitor 1 (miR-155-5p/ERRFI1) axis in acute renal allograft injury. Mice were injected with miR-155-5p-related sequences before acute renal allograft modeling. Afterwards, serum inflammation, along with oxidative stress, renal tubular injury, and apoptosis in renal tissues were detected. HK-2 cells were processed by hypoxia/reoxygenation (H/R) and transfected with miR-155-5p- or ERRFI1-related sequences, after which cell proliferation and apoptosis were measured. KLF4, miR-155-5p, and ERRFI1 expressions and their interaction were tested. KLF4 and miR-155-5p levels were enhanced, and ERRFI1 level was repressed in mice after acute renal allograft and in H/R-treated HK-2 cells. KLF4 bound to the promoter of miR-155-5p. Depleting miR-155-5p reduced serum inflammation and attenuated oxidative stress, renal tubular injury, and apoptosis in mice with acute renal allograft injury. Downregulating miR-155-5p facilitated proliferation and repressed apoptosis of H/R-treated HK-2 cells. miR-155-5p targeted ERRFI1. Knocking down ERRFI1 antagonized the effects of downregulated miR-155-5p on acute renal allograft injury, as well as on H/R-treated HK-2 cell proliferation and apoptosis. A summary displays that silencing KLF4 suppresses miR-155-5p to attenuate acute renal allograft injury by upregulating ERRFI1, which provides a way to control acute renal allograft injury.
Collapse
|
15
|
Tong D, Zhao Y, Tang Y, Ma J, Wang Z, Li C. Circ-Usp10 promotes microglial activation and induces neuronal death by targeting miRNA-152-5p/CD84. Bioengineered 2021; 12:10812-10822. [PMID: 34753388 PMCID: PMC8809980 DOI: 10.1080/21655979.2021.2004362] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Spinal cord injury (SCI) is a traumatic disease resulting in neuronal injury. circRNAs are closely associated with human diseases. Nevertheless, the potential mechanism by which circRNAs impact SCI remains to be elucidated. The aim of this study was to investigate the regulatory roles of Circular RNAs (circRNAs) in SCI. The SCI mouse model and integrated bioinformatics analysis were used to identify the differentially expressed genes (DEGs). Functional enrichment analysis was conducted to study the related pathways. The circRNA-mediated ceRNA network and subnetwork was constructed based on circMir, TargetScan and miRanda. qRT–PCR, ELISA, flow cytometry, and luciferase reporter assays were carried out to validate the role of circ_0014637 (circ-Usp10) and microRNA(miR)-152-5p /CD84 in microglia. In all, 23 DE-circRNAs, 127 DE-miRNAs and 1327 DE-mRNAs were identified. We integrated these DEGs to construct a circRNA-miRNA-mRNA network. The circ-Usp10/miR-152-5p/CD84 axis was found to function in microglial activation. We also found that circ-Usp10 inhibited the secretion of proinflammatory factors in microglial BV2 cells. In addition, silencing circ-Usp10 significantly reduced the death of the neuronal cell line HT22. Taken together, we concluded that circ-Usp10 may function as a competing endogenous RNA (ceRNA) to promote microglial activation and induce neuronal death by targeting miR-152-5p/CD84. The circ-Usp10 may be a diagnostic biomarker and potential target for SCI therapy.
Collapse
Affiliation(s)
- Dake Tong
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, People's Republic of China
| | - Yanyin Zhao
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yang Tang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, People's Republic of China
| | - Jie Ma
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, People's Republic of China
| | - Zhiwei Wang
- Department of Orthopedic Surgery, The Third Affiliated Hospital of Naval Medical University, 700 North Moyu Road, Shanghai, 201805, China
| | - Cheng Li
- Department of Orthopedics, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, P.R. China
| |
Collapse
|
16
|
Xiong Y, Wang Y, Tian H, Li Y, Xu Q, He Z. Circ-PRKCI Alleviates Lipopolysaccharide-induced Human Kidney 2 Cell Injury by Regulating miR-106b-5p/GAB1 Axis. J Cardiovasc Pharmacol 2021; 78:523-533. [PMID: 34269703 DOI: 10.1097/fjc.0000000000001031] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 03/19/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT Circular RNAs act as vital regulators in diverse diseases. However, the investigation of circular RNAs in sepsis-engendered acute kidney injury remains dismal. We aimed to explore the effects of circular RNA protein kinase C iota (circ-PRKCI) in lipopolysaccharide (LPS)-mediated HK2 cell injury. Sepsis in vitro model was established by LPS treatment. Quantitative real-time polymerase chain reaction assay was conducted for determining the levels of circ-PRKCI, microRNA-106b-5p (miR-106b-5p), and growth factor receptor binding 2-associated binding protein 1 (GAB1). Cell viability and apoptosis were evaluated using Cell Counting Kit-8 assay and flow cytometry analysis, respectively. The concentrations of interleukin-6, interleukin-1β, and tumor necrosis factor-α were measured with enzyme-linked immunosorbent assay kits. The levels of oxidative stress markers were determined using relevant commercial kits. Western blot assay was conducted for B-cell lymphoma-2 (Bcl-2), BCL2-Associated X (Bax), and GAB1 protein levels. Dual-luciferase reporter assay and RNA immunoprecipitation assay were used to verify the association between miR-106b-5p and circ-PRKCI or GAB1. We found the Circ-PRKCI level was decreased in sepsis patients and LPS-induced human kidney 2 (HK-2) cells. LPS exposure inhibited cell viability and facilitated apoptosis, inflammation, and oxidative stress in HK-2 cells. Circ-PRKCI overexpression abrogated the effects of LPS on cell apoptosis, inflammation, and oxidative stress in HK-2 cells. Furthermore, circ-PRKCI was identified as the sponge for miR-106b-5p to positively regulate GAB1 expression. Overexpression of circ-PRKCI relieved LPS-mediated HK-2 cell damage by sponging miR-106b-5p. MiR-106b-5p inhibition ameliorated the injury of HK-2 cells mediated by LPS, whereas GAB1 knockdown reversed the effect. Collectively, Circ-PRKCI overexpression attenuated LPS-induced HK-2 cell injury by regulating miR-106b-5p/GAB1 axis.
Collapse
Affiliation(s)
- Yueli Xiong
- Department of Infectious Diseases, Huaihe Hospital of Henan University, Kaifeng, Henan, China; and
| | - Yang Wang
- Department of Ultrasound, Huaihe Hospital of Henan University, Kaifeng, Henan, China
| | - Hui Tian
- Department of Infectious Diseases, Huaihe Hospital of Henan University, Kaifeng, Henan, China; and
| | - Yuanyuan Li
- Department of Infectious Diseases, Huaihe Hospital of Henan University, Kaifeng, Henan, China; and
| | - Qingjie Xu
- Department of Infectious Diseases, Huaihe Hospital of Henan University, Kaifeng, Henan, China; and
| | - Zhenkun He
- Department of Infectious Diseases, Huaihe Hospital of Henan University, Kaifeng, Henan, China; and
| |
Collapse
|
17
|
The Role of microRNAs in Pulp Inflammation. Cells 2021; 10:cells10082142. [PMID: 34440911 PMCID: PMC8391605 DOI: 10.3390/cells10082142] [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: 08/01/2021] [Revised: 08/17/2021] [Accepted: 08/17/2021] [Indexed: 02/07/2023] Open
Abstract
The dental pulp can be affected by thermal, physical, chemical, and bacterial phenomena that stimulate the inflammatory response. The pulp tissue produces an immunological, cellular, and vascular reaction in an attempt to defend itself and resolve the affected tissue. The expression of different microRNAs during pulp inflammation has been previously documented. MicroRNAs (miRNAs) are endogenous small molecules involved in the transcription of genes that regulate the immune system and the inflammatory response. They are present in cellular and physiological functions, as well as in the pathogenesis of human diseases, becoming potential biomarkers for diagnosis, prognosis, monitoring, and safety. Previous studies have evidenced the different roles played by miRNAs in proinflammatory, anti-inflammatory, and immunological phenomena in the dental pulp, highlighting specific key functions of pulp pathology. This systematized review aims to provide an understanding of the role of the different microRNAs detected in the pulp and their effects on the expression of the different target genes that are involved during pulp inflammation.
Collapse
|
18
|
Hong X, Wang J, Li S, Zhao Z, Feng Z. RETRACTED: MicroRNA-375-3p in endothelial progenitor cells-derived extracellular vesicles relieves myocardial injury in septic rats via BRD4-mediated PI3K/AKT signaling pathway. Int Immunopharmacol 2021; 96:107740. [PMID: 34020393 DOI: 10.1016/j.intimp.2021.107740] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 04/08/2021] [Accepted: 04/27/2021] [Indexed: 02/04/2023]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Editor-in-Chief. Concern was raised about the reliability of the Western blot results in Figs. 1E, 4A+F, 5A+B and Supplementary Fig. 1O+P, which appear to have the same eyebrow shaped phenotype as many other publications tabulated here (https://docs.google.com/spreadsheets/d/149EjFXVxpwkBXYJOnOHb6RhAqT4a2llhj9LM60MBffM/edit#gid=0 [docs.google.com]). The journal requested the corresponding author comment on these concerns and provide the raw data. However, the authors were not responsive to the request for comment. Since original data could not be provided, the overall validity of the results could not be confirmed. Therefore, the Editor-in-Chief decided to retract the article.
Collapse
Affiliation(s)
- Xiaoyang Hong
- PICU, The Seventh Medical Center, PLA General Hospital, Beijing 100700, China
| | - Jie Wang
- Surgical Pediatric Intensive Care Unit, Children's Hospital Affiliated of Zhengzhou University, Zhengzhou, China
| | - Shuanglei Li
- Cardiovascular Surgery Department, PLA General Hospital, Beijing 100853, China
| | - Zhe Zhao
- PICU, The Seventh Medical Center, PLA General Hospital, Beijing 100700, China
| | - Zhichun Feng
- PICU, The Seventh Medical Center, PLA General Hospital, Beijing 100700, China.
| |
Collapse
|
19
|
Zhu L, Shi D, Cao J, Song L. LncRNA CASC2 Alleviates Sepsis-induced Acute Lung Injury by Regulating the miR-152-3p/PDK4 Axis. Immunol Invest 2021; 51:1257-1271. [PMID: 34165388 DOI: 10.1080/08820139.2021.1928693] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background: Acute lung injury (ALI) is an early complication of sepsis and it is also considered as an important cause of high mortality in sepsis patients. This research aimed to explore the potential role and mechanism of long non-coding RNA (lncRNA) cancer susceptibility candidate 2 (CASC2) in sepsis-induced ALI. Methods: The levels of CASC2, microRNA-152-3p (miR-152-3p) and pyruvate dehydrogenase kinase 4 (PDK4) in sepsis patients and LPS-treated HPAEpiC were detected by quantitative real-time PCR and western blot. Cell viability and apoptosis were assessed by Counting Kit-8 (CCK-8) assay and flow cytometry. The concentrations of inflammatory factors were tested by Enzyme-linked immunosorbent assay. Oxidative stress was evaluated by the levels of reactive oxygen species and superoxide dismutase using corresponding commercial kits. The targeting relationship between miR-152-3p and CASC2 or PDK4 was verified by dual-luciferase reporter, RNA immunoprecipitation (RIP) and RNA pull-down assays.Results: CASC2 and PDK4 were down-regulated, while miR-152-3p was up-regulated in sepsis patients and LPS-stimulated HPAEpiC. Overexpression of CASC2 relieved the LPS-resulted cell viability inhibition, apoptosis promotion, inflammatory and oxidative damages in HPAEpiC. In addition, miR-152-3p was a miRNA target of CASC2 and CASC2 alleviated cell injury in LPS-disposed HPAEpiC by sponging miR-152-3p. Moreover, miR-152-3p directly targeted PDK4 and CASC2 increased the PDK4 expression by depending on the sponge effect on miR-152-3p. Meanwhile, inhibition of miR-152-3p attenuated LPS-triggered HPAEpiC injury by upregulating the level of PDK4.Conclusion: These results suggested that CASC2 ameliorated the LPS-induced injury in HPAEpiC via regulating miR-152-3p/PDK4 pathway.
Collapse
Affiliation(s)
- Lili Zhu
- Department of Intensive Care Unit, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China
| | - Dongwu Shi
- Department of Intensive Care Unit, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China
| | - Jianghong Cao
- Department of Intensive Care Unit, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China
| | - Lu Song
- Department of Intensive Care Unit, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China
| |
Collapse
|
20
|
Han D, Fang R, Shi R, Jin Y, Wang Q. LncRNA NKILA knockdown promotes cell viability and represses cell apoptosis, autophagy and inflammation in lipopolysaccharide-induced sepsis model by regulating miR-140-5p/CLDN2 axis. Biochem Biophys Res Commun 2021; 559:8-14. [PMID: 33932903 DOI: 10.1016/j.bbrc.2021.04.074] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 04/17/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) play vital roles in human diseases, including sepsis-induced acute kidney injury (AKI). Here, we aimed to investigate the functions of lncRNA NKILA in sepsis-engendered AKI. METHODS HK2 cells stimulated with LPS were used to mimic sepsis-induced AKI in vitro. qRT-PCR was conducted for lncRNA NKILA and miR-140-5p levels. Cell Counting Kit-8 (CCK-8) assay and flow cytometry analysis were employed to analyze cell viability and apoptosis. Western blot assay was utilized to measured protein levels. ELISA kits were used to examine the concentrations of IL-6, IL-1β and TNF-α. Dual-luciferase reporter assay was utilized to analyze the relationships among lncRNA NKILA, miR-140-5p and claudin 2 (CLDN2). RESULTS LPS restrained HK2 cell viability and accelerated cell apoptosis and autophagy. LncRNA NKILA was increased in LPS-treated HK2 cells. LncRNA NKILA silencing reversed the promotional influence of LPS on cell progression in HK2 cells. miR-140-5p inhibition ameliorated lncRNA NKILA knockdown-mediated cell injury in LPS-mediated HK2 cells. CLDN2 was the target of miR-140-5p. MiR-140-5p elevation promoted cell viability and suppressed cell apoptosis, autophagy and inflammation in LPS-induced HK2 cells, with CLDN2 elevation overturned the effects. CONCLUSION LncRNA NKILA silencing protected HK2 cells from LPS-induced impairments by reducing CLDN2 through sponging miR-140-5p.
Collapse
Affiliation(s)
- Dan Han
- Department of Emergency Internal Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai City, China
| | - Rong Fang
- Department of Emergency Internal Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai City, China
| | - Rong Shi
- Department of Emergency Internal Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai City, China
| | - Yuanyuan Jin
- Department of Emergency Internal Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai City, China
| | - Qian Wang
- Department of Emergency Internal Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai City, China.
| |
Collapse
|
21
|
Tang Y, Yang X, Shu H, Yu Y, Pan S, Xu J, Shang Y. Bioinformatic analysis identifies potential biomarkers and therapeutic targets of septic-shock-associated acute kidney injury. Hereditas 2021; 158:13. [PMID: 33863396 PMCID: PMC8052759 DOI: 10.1186/s41065-021-00176-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/30/2021] [Indexed: 01/22/2023] Open
Abstract
Background Sepsis and septic shock are life-threatening diseases with high mortality rate in intensive care unit (ICU). Acute kidney injury (AKI) is a common complication of sepsis, and its occurrence is a poor prognostic sign to septic patients. We analyzed co-differentially expressed genes (co-DEGs) to explore relationships between septic shock and AKI and reveal potential biomarkers and therapeutic targets of septic-shock-associated AKI (SSAKI). Methods Two gene expression datasets (GSE30718 and GSE57065) were downloaded from the Gene Expression Omnibus (GEO). The GSE57065 dataset included 28 septic shock patients and 25 healthy volunteers and blood samples were collected within 0.5, 24 and 48 h after shock. Specimens of GSE30718 were collected from 26 patients with AKI and 11 control patents. AKI-DEGs and septic-shock-DEGs were identified using the two datasets. Subsequently, Gene Ontology (GO) functional analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, and protein-protein interaction (PPI) network analysis were performed to elucidate molecular mechanisms of DEGs. We also evaluated co-DEGs and corresponding predicted miRNAs involved in septic shock and AKI. Results We identified 62 DEGs in AKI specimens and 888, 870, and 717 DEGs in septic shock blood samples within 0.5, 24 and 48 h, respectively. The hub genes of EGF and OLFM4 may be involved in AKI and QPCT, CKAP4, PRKCQ, PLAC8, PRC1, BCL9L, ATP11B, KLHL2, LDLRAP1, NDUFAF1, IFIT2, CSF1R, HGF, NRN1, GZMB, and STAT4 may be associated with septic shock. Besides, co-DEGs of VMP1, SLPI, PTX3, TIMP1, OLFM4, LCN2, and S100A9 coupled with corresponding predicted miRNAs, especially miR-29b-3p, miR-152-3p, and miR-223-3p may be regarded as promising targets for the diagnosis and treatment of SSAKI in the future. Conclusions Septic shock and AKI are related and VMP1, SLPI, PTX3, TIMP1, OLFM4, LCN2, and S100A9 genes are significantly associated with novel biomarkers involved in the occurrence and development of SSAKI. Supplementary Information The online version contains supplementary material available at 10.1186/s41065-021-00176-y.
Collapse
Affiliation(s)
- Yun Tang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang Avenue, Wuhan, 430022, China
| | - Xiaobo Yang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang Avenue, Wuhan, 430022, China
| | - Huaqing Shu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang Avenue, Wuhan, 430022, China
| | - Yuan Yu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang Avenue, Wuhan, 430022, China
| | - Shangwen Pan
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang Avenue, Wuhan, 430022, China
| | - Jiqian Xu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang Avenue, Wuhan, 430022, China
| | - You Shang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang Avenue, Wuhan, 430022, China.
| |
Collapse
|
22
|
Yu B, Liang H, Zhou S, Ye Q, Wang Y. A novel genomic model for predicting the likelihood of delayed graft function in DCD kidney transplantation. Transl Androl Urol 2021; 10:1637-1646. [PMID: 33968652 PMCID: PMC8100846 DOI: 10.21037/tau-20-1533] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background The high incidence of delayed graft function (DGF) following kidney transplantation with donation after cardiac death allografts (DCD-KT) poses great challenges to transplant clinicians. This study aimed to explore the DGF-related biomarkers and establish a genomic model for DGF prediction specific to DCD KT. Methods By data mining a public dataset (GSE43974), the key DGF-related genes in DCD kidney biopsies taken after short-time reperfusion (45–60 min) were identified by differential expression analysis and a LASSO-penalized logistic regression model. Their coefficients for modeling were calculated by multivariate logistic regression. Receiver operating characteristic curves and a nomogram were generated to evaluate its predictive ability for DGF occurrence. Gene set enrichment analysis (GSEA) was performed to explore biological pathways underlying DGF in DCD KT. Results Five key DGF-related genes (CHST3, GOLPH3, ZBED5, AKR1C4, and ERRFI1) were first identified, all of which displayed good discrimination for DGF occurrence after DCD KT (all P<0.05). A five-mRNA-based risk score was further established and showed excellent predictive ability (AUC =0.9708, P<0.0001), which was obviously higher than that of the five genes alone. Eight DGF-related biological pathways in DCD kidneys, such as “arachidonic acid metabolism”, “lysosome”, “proximal tubule bicarbonate reclamation”, “glutathione metabolism”, were identified by GSEA (all P<0.05). Moreover, a convenient and visual nomogram based on the genomic risk score was also constructed and displayed high accuracy for DGF prediction specific to DCD KT. Conclusions The novel genomic model may effectively predict the likelihood of DGF immediately after DCD KT or even prior to transplantation in the context of normothermic machine perfusion in the future.
Collapse
Affiliation(s)
- Bin Yu
- 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, Wuhan, China
| | - Han Liang
- 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, Wuhan, China
| | - Shujun 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, 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, Wuhan, China.,The 3rd Xiangya Hospital of Central South University, Research Center of National Health Ministry on Transplantation Medicine Engineering and Technology, Changsha, China
| | - Yanfeng Wang
- 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, Wuhan, China
| |
Collapse
|