scholarly journals Silencing of microRNA-146a alleviates the neural damage in temporal lobe epilepsy by down-regulating Notch-1

2019 ◽  
Vol 12 (1) ◽  
Author(s):  
Hui Huang ◽  
Guiyun Cui ◽  
Hai Tang ◽  
Lingwen Kong ◽  
Xiaopeng Wang ◽  
...  

AbstractThis study aimed to evaluate the specific regulatory roles of microRNA-146a (miRNA-146a) in temporal lobe epilepsy (TLE) and explore the related regulatory mechanisms. A rat model of TLE was established by intraperitoneal injection of lithium chloride-pilocarpine. These model rats were injected intracerebroventricularly with an miRNA-146a inhibitor and Notch-1 siRNA. Then, neuronal damage and cell apoptosis in the cornu ammonis (CA) 1 and 3 regions of the hippocampus were assessed. SOD and MDA levels in the hippocampus were detected by chromatometry, and IL-1β, IL-6, and IL-18 levels were detected by ELISA. Then, we evaluated the expression levels of caspase-9, GFAP, Notch-1, and Hes-1 in the hippocampus. The interaction between Notch-1 and miRNA-146a was assessed by a dual luciferase reporter gene assay. A rat model of TLE was successfully established, which exhibited significantly increased miRNA-146a expression in the hippocampus. Silencing of miRNA-146a significantly alleviated the neuronal damage and cell apoptosis in the CA1 and CA3 regions of the hippocampus in TLE rats and decreased MDA, IL-1β, IL-6, and IL-18 levels and increased SOD levels in the hippocampus of TLE rats. In addition, silencing of miRNA-146a significantly decreased the expression levels of caspase-9, GFAP, Notch-1, and Hes-1 in the hippocampus of TLE rats. Notch-1 was identified as a target of miRNA-146a and silencing of Notch-1 aggravated the neuronal damage in the CA1 and CA3 regions. Silencing of miRNA-146a alleviated the neuronal damage in the hippocampus of TLE rats by down-regulating Notch-1.

2016 ◽  
Vol 36 (6) ◽  
Author(s):  
Fang He ◽  
Bei Liu ◽  
Qiang Meng ◽  
Yang Sun ◽  
Weiwen Wang ◽  
...  

Increasing evidence supports the involvement of inflammatory and immune processes in temporal lobe epilepsy (TLE). miRNAs represent small regulatory RNA molecules that have been shown to act as negative regulators of gene expression controlling different biological processes, including immune system homoeostasis and function. We investigated the expression and cellular distribution of miRNA-146a (miR-146a) in a rat model of TLE. Prominent up-regulation of miR-146a activation was evident in 1 week after status epilepticus (SE) and persisted in the chronic phase. The predicted miR-146a's target complement factor H (CFH) mRNA and protein expression was also down-regulated in TLE rat model. Furthermore, transfection of miR-146a mimics in neuronal and glial cells down-regulated CFH mRNA and protein levels respectively. Luciferase reporter assays demonstrated that miR-146a down-regulated CFH mRNA expression via 3′-UTR pairing. Down-regulating miR-146a by intracerebroventricular injection of antagomir-146a enhanced the hippocampal expression of CFH in TLE model and decreased seizure susceptibility. These findings suggest that immunopathological deficits associated with TLE can in part be explained by a generalized miR-146a-mediated down-regulation of CFH that may contribute to epileptogenesis in a rat model of TLE.


2021 ◽  
Author(s):  
Jianquan You ◽  
Fei Qian ◽  
Yu Huang ◽  
Yingxuan Guo ◽  
Yaqian Lv ◽  
...  

Abstract BackgroundCerebral ischemic stroke was a nervous system disease with high occurrence rate and mortality rate. This study aimed to investigate the role and mechanism of lncRNA WT1-AS in cerebral ischemic stroke. Materials and methodsStarbase and dual luciferase reporter gene assay were used to analyze the target relationship between lncRNA WT1-AS and miR-186-5p. qRT-PCR analysis was used to detect lncRNA WT1-AS and miR-186-5p expression. OGD-induced SH-SY5Y cells injury model was conducted, and cell viability and cell apoptosis were determined by MTT and flow cytometer assay. Caspase3 ability was determined using Caspase3 activity detection kit. ResultsmiR-186-5p was a target of lncRNA-WT1-AS. lncRNA WT1-AS was down-regulated and miR-186-5p was up-regulated in blood samples of patients with ischemic stroke and in OGD-induced SH-SY5Y cells. We found that WT1-AS-plasmid promoted OGD-induced cell viability, reduced cell apoptosis and decreased caspase3 ability, and these changes were reversed by miR-186-5p mimic. Subsequently, our results proved that XIAP was a target of miR-186-5p. Similarly, miR-186-5p inhibitor reduced OGD-induced neuronal damage by up-regulating XIAP expression. ConclusionlncRNA-WT1-AS/miR-186-5p/XIAP might be a new target for cerebral ischemic stroke treatment.


2020 ◽  
Author(s):  
Xiaowan Li ◽  
Fang Hao ◽  
Shuxin Tao ◽  
Weihua Wang ◽  
Xinxing Xiao ◽  
...  

Abstract Background Both long non-coding RNA (lncRNA) NEAT1 and microRNA (miR)-139 are crucial gene regulators in various disorders. This study aims to investigate their role in epilepsy and seizure-induced neuronal damage. Methods In this research, rat model of epilepsy was established by pilocarpine induction. The RNA and protein expression in hippocampal tissues and neurons were determined by RT-qPCR and Western blot analysis, respectively. Microarray analysis was used to predict the relationship between NEAT1 and miR-139 or between miR-139 and ROCK1, and dual luciferase reporter gene assay was performed to verify the interaction. The endogenous expression of related genes was modulated by recombinant plasmids and cell transfection. The cell apoptosis, levels of inflammatory factors and cell proliferation were detected by flow cytometry, ELISA and EdU assay. Results LncRNA NEAT1 and ROCK1 was upregulated, while the miR-139 was downregulated in hippocampal tissues and neurons of epileptic rats. Overexpression of NEAT1 decreased the activity of neurons, increased cell apoptosis, and increased the level of inflammatory factors. NEAT1 negatively targeted miR-139 to upregulate ROCK1. The RhoA/ROCK1 signaling pathway was activated by NEAT1 overexpression and miR-139 downregulation. Conclusion LncRNA NEAT1 suppressed pilocarpine-induced epilepsy by inhibiting apoptosis of hippocampal neurons through miR-139/RhoA/ROCK1 axis, and thereby inhibiting neuronal injury induced by seizure.


2013 ◽  
Vol 4 (3) ◽  
Author(s):  
Muhammad Ashhab ◽  
Ahmed Omran ◽  
Na Gan ◽  
Huimin Kong ◽  
Jing Peng ◽  
...  

AbstractBackground: Recently, microRNAs (miRNAs) have attracted much attention as novel players in the pathogenesis of mesial temporal lobe epilepsy (MTLE) in mature and developing brains. This study aimed to investigate the expression dynamics of miR-9, miR-138, miR-181a, miR-221, and miR-222 in the hippocampus of an immature rat model during the three stages of MTLE development and in children with MTLE. Methodology: qPCR was used to measure expression levels during the three stages of MTLE development (2 h, 3, and 8 weeks after induction of lithium-pilocarpine status epilepticus, representing the acute, latent, and chronic stages, respectively. Expression levels were also measured in hippocampi obtained from children with MTLE and normal controls. Results: In the rat model, miR-9 was significantly upregulated during the acute and chronic stages relative to controls, but not during the latent stage. MiR-138, miR-221 and miR-222 were all downregulated during all three stages of MTLE development. MiR-181a was downregulated during the acute stage, upregulated during the chronic stage, and unaltered during the latent stage. In children, miR-9 and miR-181a were upregulated, while miR-138, miR-221, and miR-222 were downregulated. Conclusion: Modulation of these miRNAs may be a new strategy in designing antiepileptic and anticonvulsant therapies for the developing brain.


2020 ◽  
Vol 65 (4) ◽  
pp. 187-201
Author(s):  
Shuai Shao ◽  
Hui Wang ◽  
Wei Shao ◽  
Na Liu

Polycystic ovary syndrome (PCOS) is a prevalent endocrine disorder and one of the most common causes of infertility in women. PCOS patients have been found with dysregulated miRNA, which is indicative of their roles as noninvasive biomarkers and novel therapeutic targets in PCOS. Herein, this study sets out to explore the mechanism of action of miR-199a-5p in PCOS in relation to the janus kinase/signal transducer and activator of transcription 3 (JAK/STAT3) pathway via Wilms’ tumor 1 (WT1) regulation in a rat model of PCOS. The expression of miR-199a-5p was highly expressed in ovarian cortical tissues and serum of PCOS patients, as examined by RT-qPCR. Ovarian granulosa cells (GCs) were harvested from PCOS rat model, followed by subsequent purification. Gain- and loss-of-function experiments of miR-199a-5p were performed to determine its functions in PCOS. Cell viability, cell apoptosis and serum hormone levels were assessed, the results of which showed that downregulation of miR-199a-5p contributed to the promotion of GC viability and inhibition of apoptosis, while simultaneously inducing the elevation of serum E2 level and reduction of serum AMH, PG, LH and FSH levels in the PCOS rat model. WT1 was identified as a target gene of miR-199a-5p by dual-luciferase reporter gene assay, and inhibition of miR-199a-5p resulted in the activation of WT1-mediated JAK/STAT3 pathway. The activated JAK/STAT3 pathway suppressed the development of PCOS by miR-199a-5p, indicating a mechanism by which miR-199a-5p could potentially prevent PCOS through the WT1-mediated JAK/STAT3 pathway.


2020 ◽  
Vol 15 (1) ◽  
pp. 1013-1023
Author(s):  
Lina Xing ◽  
Jinhai Ren ◽  
Xiaonan Guo ◽  
Shukai Qiao ◽  
Tian Tian

AbstractPrevious research has revealed the involvement of microRNA-212-5p (miR-212-5p) and cyclin T2 (CCNT2) in acute myeloid leukemia (AML). However, whether the miR-212-5p/CCNT2 axis is required for the function of decitabine in AML has not been well elucidated. Quantitative reverse transcription-polymerase chain reaction was used to examine enrichment of miR-212-5p. The relationship between CCNT2 and miR-212-5p was verified by the luciferase reporter assay. Cell apoptosis was evaluated by flow cytometry and western blot. CCK-8 assay was performed to determine cell viability. Decitabine significantly repressed cell viability, while promoted cell apoptosis. Meanwhile, the expression levels of cyclinD1, CDK4, and Bcl-2 were suppressed in cells with decitabine exposure, but Bax and caspase-3 expression levels were upregulated. Besides, miR-212-5p upregulation had the similar function with decitabine in AML cell proliferation and apoptosis. Subsequently, restoration of CCNT2 attenuated miR-212-5p overexpression-induced effects in Kasumi-1 and SKNO-1 cells. In addition, miR-212-5p depletion reversed decitabine-induced CCNT2 downregulation. The miR-212-5p/CCNT2 axis had an implication in the anti-leukemic effect of decitabine in AML.


Sign in / Sign up

Export Citation Format

Share Document