1
|
Jin Y, Jiang L, Wang Y, Huang Y, Yu W, Ma X. lncRNA PRR34-AS1 knockdown represses neuroinflammation and neuronal death in traumatic brain injury by inhibiting microRNA-498 expression. Brain Inj 2023; 37:611-620. [PMID: 36951415 DOI: 10.1080/02699052.2023.2192524] [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: 06/06/2022] [Revised: 10/19/2022] [Accepted: 03/03/2023] [Indexed: 03/24/2023]
Abstract
OBJECTIVE Traumatic brain injury (TBI) can result in motor and cognitive dysfunction and is a possible risk factor for the subsequent development of dementia. However, the pathogenesis of TBI remains largely unclear. This study investigated the roles of long non-coding RNAs (lncRNAs) and microRNAs (miRNAs) in inflammation and neuronal apoptosis following TBI. METHODS The lncRNA expression profiles in the cerebral cortices of TBI model mice and sham-operated mice were analyzed using microarray. We focused on an upregulated lncRNA, PRR34-AS1, because of its known modulatory role in apoptosis and inflammation. RESULTS Our findings indicated that the knockdown of PRR34-AS1 inhibited inflammation and neuronal apoptosis and improved long-term neurological function. Using an in vitro, cell-based model of etoposide-induced primary cortical neuronal injury, we demonstrated that PRR34-AS1 levels were higher in injured model cells than in untreated control cells. Silencing of PRR34-AS1 suppressed etoposide-induced apoptosis and the production of inflammatory mediators in primary cortical neurons. PRR34-AS1 directly targets microRNA-498 (miR-498) in primary cortical neurons. Importantly, the inhibition of miR-498 expression counteracted the effects of PRR34-AS1 silencing on neuronal apoptosis and inflammation. CONCLUSIONS These findings indicate that PRR34-AS1 may be a useful therapeutic target for TBI.
Collapse
Affiliation(s)
- Yue Jin
- Department of Neurology, The Fourth Affiliated Hospital Of Harbin Medical University, Harbin, Heilongjiang, China
| | - Lai Jiang
- Department of Neurology, The Second Affiliated Hospital Of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yifan Wang
- Department of Neurology, The Fourth Affiliated Hospital Of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yingxue Huang
- Department of Neurology, The Fourth Affiliated Hospital Of Harbin Medical University, Harbin, Heilongjiang, China
| | - Wei Yu
- Department of Neurology, The Fourth Affiliated Hospital Of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xueling Ma
- Department of Neurology, The Fourth Affiliated Hospital Of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
2
|
Yang J, Xie X. Tofacitinib protects intestinal epithelial cells against oxygen-glucose deprivation/reoxygenation injury by inhibiting the JAK/STAT3 signaling pathway. Exp Ther Med 2021; 22:1108. [PMID: 34504562 PMCID: PMC8383770 DOI: 10.3892/etm.2021.10542] [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: 03/03/2021] [Accepted: 06/30/2021] [Indexed: 12/14/2022] Open
Abstract
The present study aimed to investigate the role and potential mechanism of action of tofacitinib (Tofa) in intestinal ischemia/reperfusion (I/R) injury. The normal rat small intestine epithelial cell line, IEC-6, was used to establish an I/R injury model by inducing oxygen-glucose deprivation/reoxygenation (OGD/R). Cells were divided into the following five groups: Control, OGD/R, OGD/R with 50, 100 and 200 nM Tofa. Following Tofa administration, cell viability was measured using Cell Counting Kit-8 assay and a lactate dehydrogenase detection kit. The expression levels of cell apoptosis-related proteins, Bcl-2, cleaved-caspase-3 and cleaved-caspase-9 were detected using western blot analysis. Additionally, the levels of oxidative stress-related markers, such as reactive oxygen species (ROS), malondialdehyde (MDA) and superoxide dismutase (SOD), and inflammatory cytokines, TNF-α, IL-6 and IL-1β were assessed using the colorimetric method. Western blot analysis was also used to measure the expression levels of the Janus kinase (JAK)/STAT3 signaling pathway-related proteins, including phosphorylated (p)-JAK1, p-JAK3 and p-STAT3. Subsequently, colivelin, an agonist of the JAK/STAT3 pathway, was used to investigate whether the effects of Tofa on intestinal I/R injury were mediated by this signaling pathway. The results showed that Tofa dose-dependently elevated cell viability compared with that in the OGD/R group. By contrast, Tofa attenuated cell apoptosis, which was coupled with upregulated Bcl-2 expression, downregulated cleaved-caspase-3 and downregulated cleaved-caspase-9 levels, in OGD/R-induced IEC-6 cells. Furthermore, the contents of ROS and MDA were significantly increased following exposure to OGD/R, which were accompanied by the decreased activity of SOD. These effects were reversed following cell treatment with Tofa. Consistently, Tofa intervention reduced the secretion levels of TNF-α, IL-6 and IL-1β in a dose-dependent manner. Additionally, Tofa markedly downregulated the phosphorylation levels of JAK1, JAK3 and STAT3 in OGD/R-induced IEC-6 cells. However, treatment with colivelin markedly reversed the inhibitory effects of Tofa on cell viability, cell apoptosis, oxidative stress and inflammation. Overall, the findings of the present study suggested that Tofa could protect against intestinal I/R injury by inhibiting the JAK/STAT3 signaling pathway, which may hold promise as a therapeutic agent for intestinal I/R injury.
Collapse
Affiliation(s)
- Jing Yang
- Department of Pediatric Gastroenterology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610091, P.R. China
| | - Xiaoli Xie
- Department of Pediatric Gastroenterology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610091, P.R. China
| |
Collapse
|
3
|
Nan FY, Gu Y, Xu ZJ, Sun GK, Zhou JD, Zhang TJ, Ma JC, Leng JY, Lin J, Qian J. Abnormal expression and methylation of PRR34-AS1 are associated with adverse outcomes in acute myeloid leukemia. Cancer Med 2021; 10:5283-5296. [PMID: 34227248 PMCID: PMC8335806 DOI: 10.1002/cam4.4085] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 05/28/2021] [Accepted: 06/03/2021] [Indexed: 12/11/2022] Open
Abstract
It was previously reported that PRR34‐AS1 was overexpressed in some solid tumors. PRR34‐AS1 promoter was shown to have a differential methylation region (DMR), and was hypomethylated in acute myeloid leukemia (AML). Therefore, the present study used real‐time quantitative PCR (RQ‐PCR) to explore the expression characteristics of PRR34‐AS1 in AML. In addition, the correlation between the expression of PRR34‐AS1 and clinical prognosis of AML was determined. The findings of this study indicated that high PRR34‐AS1 expression was bound up with shorter overall survival (OS) in AML patients (p = 0.002). Moreover, patients with high expression of PRR34‐AS1 had significantly lower complete remission (CR) rate compared with those with low expression of PRR34‐AS1 after induction chemotherapy. Furthermore, multivariate analysis confirmed that PRR34‐AS1 expression was an independent factor affecting CR in whole‐AML, non‐APL‐AML, and CN‐AML patients (p = 0.032, 0.039, and 0.036, respectively). Methylation‐specific PCR (MSP) and bisulfite sequencing PCR (BSP) were used to explore the methylation status of PRR34‐AS1. PRR34‐AS1 promoter showed a pattern of hypomethylation in AML patients compared with normal controls (p = 0.122). Notably, of whole‐AML and non‐APL‐AML patients, PRR34‐AS1 hypomethylated patients presented a significantly shorter OS than those with a hypermethylated PRR34‐AS1 (p = 0.010 and 0.037, respectively). Multivariate analysis confirmed that the hypomethylation of PRR34‐AS1 served as an independent prognostic indicator in both whole‐cohort AML and non‐APL‐AML categories (p = 0.057 and 0.018, respectively). In summary, the findings of this study showed that abnormalities in PRR34‐AS1 are associated with poor prognosis in AML. Therefore, monitoring this index may be important in the prognosis of AML and can provide information on effective chemotherapy against the disease.
Collapse
Affiliation(s)
- Fang-Yu Nan
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China.,Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, People's Republic of China
| | - Yu Gu
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China.,Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, People's Republic of China
| | - Zi-Jun Xu
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China.,Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, People's Republic of China
| | - Guo-Kang Sun
- West China School of Public Health and China Fourth Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Jing-Dong Zhou
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China.,Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, People's Republic of China
| | - Ting-Juan Zhang
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China.,Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, People's Republic of China
| | - Ji-Chun Ma
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China.,Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, People's Republic of China
| | - Jia-Yan Leng
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China.,Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, People's Republic of China
| | - Jiang Lin
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China.,Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, People's Republic of China
| | - Jun Qian
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China.,Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, People's Republic of China
| |
Collapse
|
4
|
Wang Y, Zhang D, Hu X. A Three-Gene Peripheral Blood Potential Diagnosis Signature for Acute Rejection in Renal Transplantation. Front Mol Biosci 2021; 8:661661. [PMID: 34017855 PMCID: PMC8129004 DOI: 10.3389/fmolb.2021.661661] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 04/21/2021] [Indexed: 12/31/2022] Open
Abstract
Background: Acute rejection (AR) remains a major issue that negatively impacts long-term allograft survival in renal transplantation. The current study aims to apply machine learning methods to develop a non-invasive diagnostic test for AR based on gene signature in peripheral blood. Methods: We collected blood gene expression profiles of 251 renal transplant patients with biopsy-proven renal status from three independent cohorts in the Gene Expression Omnibus database. After differential expression analysis and machine learning algorithms, selected biomarkers were applied to the least absolute shrinkage and selection operator (LASSO) logistic regression to construct a diagnostic model in the training cohort. The diagnostic ability of the model was further tested in validation cohorts. Gene set enrichment analysis and immune cell assessment were also conducted for further investigation. Results: A novel diagnostic model based on three genes (TSEN15, CAPRIN1 and PRR34-AS1) was constructed in the training cohort (AUC = 0.968) and successfully verified in the validation cohort (AUC = 0.925) with high accuracy. Moreover, the diagnostic model also showed a promising value in discriminating T cell-mediated rejection (TCMR) (AUC = 0.786). Functional enrichment analysis and immune cell evaluation demonstrated that the AR model was significantly correlated with adaptive immunity, especially T cell subsets and dendritic cells. Conclusion: We identified and validated a novel three-gene diagnostic model with high accuracy for AR in renal transplant patients, and the model also performed well in distinguishing TCMR. The current study provided a promising tool to be used as a precise and cost-effective non-invasive test in clinical practice.
Collapse
Affiliation(s)
- Yicun Wang
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.,Institute of Urology, Capital Medical University, Beijing, China
| | - Di Zhang
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.,Institute of Urology, Capital Medical University, Beijing, China
| | - Xiaopeng Hu
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.,Institute of Urology, Capital Medical University, Beijing, China
| |
Collapse
|
5
|
Qin M, Meng Y, Luo C, He S, Qin F, Yin Y, Huang J, Zhao H, Hu J, Deng Z, Qiu Y, Hu G, Pan H, Qin Z, Huang Z, Yi T. lncRNA PRR34-AS1 promotes HCC development via modulating Wnt/β-catenin pathway by absorbing miR-296-5p and upregulating E2F2 and SOX12. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 25:37-52. [PMID: 34168917 PMCID: PMC8190132 DOI: 10.1016/j.omtn.2021.04.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 04/20/2021] [Indexed: 12/30/2022]
Abstract
Hepatocellular carcinoma (HCC) belongs to the most frequent cancer with a high death rate worldwide. Thousands of long non-coding RNAs (lncRNAs) have been confirmed to influence the development of human cancers, including HCC. Nevertheless, the biological role of PRR34 antisense RNA 1 (PRR34-AS1) in HCC remains obscure. Here, we observed via quantitative real-time reverse transcriptase polymerase chain reaction (quantitative real-time RT-PCR) that PRR34-AS1 was highly expressed in HCC cells. Functional assays revealed that PRR34-AS1 promoted HCC cell proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT) process in vitro and facilitated tumor growth in vivo. In addition, western blot analysis and TOP Flash/FOP Flash reporter assays verified that PRR34-AS1 stimulated Wnt/β-catenin pathway in HCC cells. Furthermore, RNA immunoprecipitation (RIP), RNA pull-down, and luciferase reporter assays uncovered that PRR34-AS1 sequestered microRNA-296-5p (miR-296-5p) to positively modulate E2F transcription factor 2 (E2F2) and SRY-box transcription factor 12 (SOX12) in HCC cells. Importantly, chromatin immunoprecipitation (ChIP) and luciferase reporter assays uncovered that E2F2 transcriptionally activated PRR34-AS1 in turn. Further, rescue experiments reflected that PRR34-AS1 affected HCC progression through targeting miR-296-5p/E2F2/SOX12/Wnt/β-catenin axis. Our findings found that PRR34-AS1 elicited oncogenic functions in HCC, which indicated that PRR34-AS1 might be a novel therapeutic target for HCC.
Collapse
Affiliation(s)
- Minzhen Qin
- Gastrointestinal Medicine, People’s Hospital of Baise, Baise, Guangxi 533000, P.R. China
| | - Yiliang Meng
- Department of Radiation Oncology, People’s Hospital of Baise, Baise, Guangxi 533000, P.R. China
| | - Chunying Luo
- Laboratory Medicine, Affiliated Hospital of YouJiang Medical University of Nationalities, Baise, Guangxi 533000, P.R. China
| | - Shougao He
- Gastrointestinal Medicine, Affiliated Hospital of YouJiang Medical University of Nationalities, Baise, Guangxi 533000, P.R. China
| | - Fengxue Qin
- Laboratory Medicine, Affiliated Hospital of YouJiang Medical University of Nationalities, Baise, Guangxi 533000, P.R. China
| | - Yixia Yin
- Gastrointestinal Medicine, Affiliated Hospital of YouJiang Medical University of Nationalities, Baise, Guangxi 533000, P.R. China
| | - Junling Huang
- Gastrointestinal Medicine, Affiliated Hospital of YouJiang Medical University of Nationalities, Baise, Guangxi 533000, P.R. China
| | - Hailiang Zhao
- Gastrointestinal Medicine, Affiliated Hospital of YouJiang Medical University of Nationalities, Baise, Guangxi 533000, P.R. China
| | - Jing Hu
- Gastrointestinal Medicine, Affiliated Hospital of YouJiang Medical University of Nationalities, Baise, Guangxi 533000, P.R. China
| | - Zhihua Deng
- Gastrointestinal Medicine, Affiliated Hospital of YouJiang Medical University of Nationalities, Baise, Guangxi 533000, P.R. China
| | - Yiying Qiu
- Gastrointestinal Medicine, Affiliated Hospital of YouJiang Medical University of Nationalities, Baise, Guangxi 533000, P.R. China
| | - Gaoyu Hu
- Gastrointestinal Medicine, Affiliated Hospital of YouJiang Medical University of Nationalities, Baise, Guangxi 533000, P.R. China
| | - Hanhe Pan
- Gastrointestinal Medicine, Affiliated Hospital of YouJiang Medical University of Nationalities, Baise, Guangxi 533000, P.R. China
| | - Zongshuai Qin
- Gastrointestinal Medicine, Affiliated Hospital of YouJiang Medical University of Nationalities, Baise, Guangxi 533000, P.R. China
| | - Zansong Huang
- Gastrointestinal Medicine, Affiliated Hospital of YouJiang Medical University of Nationalities, Baise, Guangxi 533000, P.R. China
- Corresponding author: Zansong Huang, Gastrointestinal Medicine, Affiliated Hospital of YouJiang Medical University of Nationalities, Baise, Guangxi 533000, P.R. China.
| | - Tingzhuang Yi
- Gastrointestinal Medicine, Affiliated Hospital of YouJiang Medical University of Nationalities, Baise, Guangxi 533000, P.R. China
- Corresponding author: Tingzhuang Yi, Gastrointestinal Medicine, Affiliated Hospital of YouJiang Medical University of Nationalities, Baise, Guangxi 533000, P.R. China.
| |
Collapse
|
6
|
Liu Z, Li Z, Xu B, Yao H, Qi S, Tai J. Long Noncoding RNA PRR34-AS1 Aggravates the Progression of Hepatocellular Carcinoma by Adsorbing microRNA-498 and Thereby Upregulating FOXO3. Cancer Manag Res 2020; 12:10749-10762. [PMID: 33154667 PMCID: PMC7606297 DOI: 10.2147/cmar.s263619] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 10/02/2020] [Indexed: 12/13/2022] Open
Abstract
Purpose Long noncoding RNAs are differentially expressed in hepatocellular carcinoma (HCC) and have been validated as essential regulators in HCC. However, there is limited knowledge regarding the detailed roles and mechanisms of most lncRNAs in HCC cells. In this study, the expression profiles of PRR34 antisense RNA 1 (PRR34-AS1) in HCC tissues and cell lines were determined. In addition, the detailed roles and underlying mechanisms of PRR34-AS1 in HCC cells were comprehensively elucidated. Methods Reverse transcription-quantitative polymerase chain reaction (PCR) was performed to measure PRR34-AS1 expression in HCC cells. Cell proliferation, apoptosis, and migration and invasion were evaluated in vitro using the cell counting kit-8 (CCK-8) assay, flow cytometric analysis, and transwell cell migration and invasion assays, respectively. In vivo tumor growth was determined using tumor xenograft experiments. The potential miRNA targets of PRR34-AS1 were predicted via bioinformatic analysis and further confirmed using the luciferase reporter assay, RNA immunoprecipitation assay, and reverse transcription-quantitative PCR. Results PRR34-AS1 was highly expressed in HCC tissues and cell lines, and its interference suppressed HCC cell proliferation, migration, and invasion but promoted cell apoptosis in vitro. In addition, loss of PRR34-AS1 decreased tumor growth in HCC cells in vivo. Mechanistically, PRR34-AS1 functions as a miR-498 sponge and subsequently increases forkhead box O3 (FOXO3) expression in HCC cells. Rescue experiments revealed that the suppressive effects triggered by PRR34-AS1 knockdown on the malignant characteristics of HCC cells could be abrogated by inhibiting miR-498 or restoring FOXO3 expression. Conclusion The depletion of PRR34-AS1 suppresses the oncogenicity of HCC cells by targeting the miR-498/FOXO3 axis. Therefore, the PRR34-AS1/miR-498/FOXO3 pathway may offer a basis for HCC treatment.
Collapse
Affiliation(s)
- Zhaoming Liu
- Department of Hepatobiliary Surgery, Harrison International Peace Hospital, Hengshui, Hebei 053000, People's Republic of China
| | - Zhen Li
- Department of Interventional Therapy, Harrison International Peace Hospital, Hengshui, Hebei 053000, People's Republic of China
| | - Binghui Xu
- Department of Hepatobiliary Surgery, Harrison International Peace Hospital, Hengshui, Hebei 053000, People's Republic of China
| | - Hao Yao
- Department of Hepatobiliary Surgery, Harrison International Peace Hospital, Hengshui, Hebei 053000, People's Republic of China
| | - Shuangyu Qi
- Department of Hepatobiliary Surgery, Harrison International Peace Hospital, Hengshui, Hebei 053000, People's Republic of China
| | - Jianxiong Tai
- Department of Hepatobiliary Surgery, Harrison International Peace Hospital, Hengshui, Hebei 053000, People's Republic of China
| |
Collapse
|