MicroRNA-26b-5p alleviates cerebral ischemia-reperfusion injury in rats via inhibiting the N-myc/PTEN axis by downregulating KLF10 expression

2021 ◽  
pp. 096032712199189
Author(s):  
Y Xiao ◽  
S Zheng ◽  
N Duan ◽  
X Li ◽  
J Wen

MicroRNAs plays important role in cerebral ischemia-reperfusion (CIR). However, the role of miR-26b-5p in CIR injury remains unclear. PC12 cells were treated with oxygen-glucose deprivation (OGD) for 0 h, 2 h, 4 h, 6 h, and then reoxygenated for 24 h to construct an in vitro I/R model. Then, miR-26b-5p mimic, small interfering RNA of KLF10 and KLF10 overexpression plasmid were transfected into cells respectively for mechanism study. Our results showed that miR-26b-5p was downregulated in OGD/R-induced PC12 cells. After overexpression of miR-26b-5p, cell proliferation ability was enhanced, apoptosis, ROS and inflammatory mediators were inhibited. Bioinformatics analysis indicated that miR-26b-5p was directly bound to the 3’ UTR of KLF10, and downregulated the expression of KLF10. KLF10 was upregulated in OGD/R cells, and transfection with si-KLF10 promoted cell proliferation and reduced apoptosis, NO concentration and inflammatory factor secretion. Moreover, pcDNA-KLF10 reversed the inhibitory effects of miR-26b-5p mimic on apoptosis, NO content and inflammatory factor secretion, as well as the downregulation of N-myc and PTEN expression. Meanwhile, I/R rat models were constructed and divided into sham operation group (femoral artery isolation only), model group (middle cerebral artery occlusion model of rats was prepared by thread embolization), treatment group (200 µL of miR-26b-5p mimic was injected into the brain of model rats). We observed that the infarct size of brain tissue was reduced, KLF10 expression was downregulated, and apoptosis and inflammatory response were reduced. These results suggest that miR-26b-5p had protective effects on CIRI and it may be a potential treatment target.

2021 ◽  
Author(s):  
Qi Gao ◽  
Yanfeng Wang

Abstract Background: Long non-coding RNAs (lnc-RNAs) and microRNAs (miRNAs) play key roles in the development of stroke. LncRNA FOXD2-AS1 has been reported to be important in many cancers. However, the role of lncRNA FOXD2-AS1 in stroke is limited known.Methods: Real-time polymerase chain reaction (real-time PCR) assays were used to measure expressions of lncRNA FOXD2-AS1, miR-3144-5p and KCTD15. Western blot assays were employed to examine Bax and Bcl2 protein expression. The cell viability was measured by cck8 assay. The cell apoptosis was detected by TUNEL staining assay The interaction between FOXD2-AS1, miR-3144-5p and KCTD was confirmed by site-directed mutagenesis and luciferase assays. Results: The expression of lncRNA FOXD2-AS1 was down-regulated in blood sample from stroke patients and MAO mice tissues. In addition, lncRNA FOXD2-AS1 was decreased on OGD/R treated cells. LncRNA FOXD2-AS1 overexpression promoted cell viability and reduced apoptosis in OGD/R-Induced PC12 Cells. LncRNA FOXD2-AS1 could interact with miR-3144-5p. Meanwhile, inhibition of miR-3144-5p alleviated OGD/ R-induced neuronal injury in PC12 Cells. Dual luciferase reporter assay verified that KCTD15 was a target of miR-3144-5p. And level of KCTD15 was positive with LncRNA FOXD2-AS1.Conclusion: Taken together, lncRNA FOXD2-AS1 protected against c cerebral ischemia-reperfusion injury by acting as a sponge of miR-3144-5p to modulate KCTD15 level.


2019 ◽  
Vol 17 (3) ◽  
pp. 322-328
Author(s):  
Luan Lan ◽  
Cao Lanxiu ◽  
Zhu Lei ◽  
Sun Jianhua

Diosmetin, a natural flavonoid, exhibits a variety of pharmacologic activities including inhibition of inflammation and oxidation. Therefore, its potential role in the management of cerebral ischemia/reperfusion (I/R) injury remains to be examined. In this study, we explored the underlying molecular mechanisms of diosmetin effects on cerebral ischemia/reperfusion injury in vitro. The results show that hypoxia/reoxygenation treatment of PC12 cells decreased cell viability and increased apoptosis, inflammation and oxidative stress. Diosmetin improved cellular viability, decreased lactate dehydrogenase release, and inhibited apoptosis in hypoxia-/reoxygenation-treated PC12 cells. Furthermore, diosmetin effectively inhibited the NF-kB signaling pathway to attenuate the inflammatory response. Also, diosmetin inhibited reactive oxygen species generation to attenuate I/R injury-induced oxidative stress in PC12 cells probably through the activation of Nrf 2/HO-1 pathway. Therefore, diosmetin effectively protected cells from I/R injury in nerve cells by scavenging reactive oxygen species by activating Nrf 2/HO-1 pathway and inhibiting inflammation by the suppression of NF-kB signaling pathway. Diosmetin can be regarded as a potential agent for cerebral ischemia/reperfusion injury treatment.


2016 ◽  
Vol 2016 ◽  
pp. 1-8 ◽  
Author(s):  
Shi Shu ◽  
Chun-Ming Li ◽  
Yan-Li You ◽  
Xiao-Lu Qian ◽  
Shuang Zhou ◽  
...  

Background. The therapeutic mechanisms of cerebral ischemia treatment by acupuncture are yet not well addressed.Objective. We investigated the effects of electroacupuncture (EA) at GV26 observing the expression of autophagy-related proteins Beclin-1 and LC3B and proportion of apoptotic cells and Bcl-2 positive cells in MCAO/R model rats.Methods. Sprague-Dawley (SD) male rats were randomly assigned to 7 groups: model groups (M6h, M24h, and M72h), EA treatment groups (T6h, T24h, and T72h), and sham operation group (S). Neurological deficit and cerebral infarction volume were measured to assess the improvement effect, while the expression of Beclin-1 and LC3B and proportion of Tunel-positive and Bcl-2 positive cells were examined to explore EA effect on autophagy and apoptosis.Results. EA significantly decreased neurological deficit scores and the volume of cerebral infarction. Beclin-1 was significantly decreased in T24h, while LC3B-II/LC3B-I ratio markedly reduced in 6th hour. EA groups markedly reduced the number of Tunel positive cells, especially in T24h. Meanwhile, the number of Bcl-2 positive cells obviously increased after EA treatment, especially in T6h and T24h.Conclusions. The alleviation of inadequate autophagy and apoptosis may be a key mechanism involved in the reflex regulation of EA at GV26 to treat cerebral ischemia.


Author(s):  
Lei Wang ◽  
Ying Tan ◽  
Ziyu Zhu ◽  
Jun Chen ◽  
Qiang Sun ◽  
...  

We aim to explore the expression and function of long non-coding RNA (lncRNA) ATP2B1-AS1 in a cerebral ischemia/reperfusion (I/R) injury. In this study, we established a middle cerebral artery occlusion/reperfusion (MCAO/IR) rat model and an OGD/R PC12 cell model to evaluate the expression and role of ATP2B1-AS1 in the cerebral I/R injury. We found that the expression of ATP2B1-AS1 was upregulated in both in vitro and in vivo cerebral I/R injury models. Knockdown of ATP2B1-AS1 increased the cell viability, inhibited apoptosis, and decreased the expressions of inflammation cytokines. The target of ATP2B1-AS1 was predicted and validated to be miR-330-5p. MiR-330-5p abrogated the regulatory effect of ATP2B1-AS1 on cell viability, apoptosis, and cytokines of OGD/R PC12 cells. Furthermore, the results showed that miR-330-5p targeted TLR4, which was also upregulated in the infarcted area of MCAO/IR rats and OGD/R PC12 cells. Overexpression of ATP2B1-AS1 increased the expressions of TLR4, MyD88, and NF-κB p65 of OGD/R PC12 cells, while the effect of ATP2B1-AS1 was abrogated by miR-330-5p. In addition, knockdown of ATP2B1-AS1 decreased the latency time, increased the time of passing the platform position, reduced the cerebral infarct volume, decreased neurological deficit scores, and reduced the number of damaged neurons of MCAO/IR rats that were subjected to the Morris water maze test. Taken together, our study indicates that ATP2B1-AS1 may be an attractive therapeutic target for the treatment of cerebral ischemic injuries.


2020 ◽  
Vol 48 (9) ◽  
pp. 030006052094585 ◽  
Author(s):  
Chengli Ling ◽  
Chang Lei ◽  
Manshu Zou ◽  
Xiong Cai ◽  
Yun Xiang ◽  
...  

Objective The therapeutic efficacy of apigenin in PC12 cells and rats remains uncertain. The aim of this study was to investigate the neuroprotective effects of apigenin against cerebral ischemia/reperfusion injury, both in vitro and in vivo. Methods We first treated PC12 cells with cobalt chloride (CoCl2) to create a model of oxidative stress injury. Cell viability was then determined using a multifunctional microplate reader. In addition, reactive oxygen species (ROS) levels, apoptosis, and mitochondrial membrane potentials (MMPs) were examined using high-content cytometer analysis. The efficacy of apigenin treatment was also analyzed in a rat middle cerebral artery occlusion (MCAO) model using TTC staining and neurological deficit scores. Results The half-inhibitory concentration of CoCl2 was 1.2 mM. Pretreatment with 10 µg ⋅ mL−1 apigenin significantly enhanced cell viability, reduced ROS levels, alleviated apoptosis, and improved MMP in PC12 cells with CoCl2-induced injury in vitro. In addition, apigenin treatment in vivo significantly improved neurological deficit scores and reduced infarct areas in MCAO rats. These results suggest that the neuroprotective mechanisms of apigenin may be related to mitochondrial activation. Conclusions Apigenin had excellent neuroprotective effects for the treatment of cerebral ischemia/reperfusion injury in vitro and in vivo.


2021 ◽  
Vol 20 (1) ◽  
pp. 23-28
Author(s):  
Chunying Deng ◽  
Peilan Zhang ◽  
Yun Zhang

Purpose: To investigate the effect of FOXO4 on cerebral ischemia/reperfusion (CIR) injury and the underlying mechanism.Methods: An in vitro ischemia/reperfusion (IR) model was achieved using oxygen-glucose deprivation/reoxygenation (OGD/R). Expression of RNA and protein was determined using quantitative real time polymerase chain reaction (qRT-PCR) and western blotting, respectively. Cell viability and apoptosis were determined using MTT assay and flow cytometry, respectively. Commercial kits were used to measure lactate dehydrogenase (LDH), reactive oxygen species (ROS), chloramphenicol acetyltransferase (CAT), malondialdehyde (MDA), and superoxide dismutase (SOD).Results: Following OGD/R, FOXO4 mRNA and protein expressions were upregulated in SH-SY5H human neuroblastoma cells. ODG/R reduced cell proliferation and increased the proportion of apoptotic cells, and these effects were inhibited by knockdown of FOXO4 (p < 0.05). Levels of cleaved caspase 3 and cleaved poly(ADP-ribose) polymerases (PARPs) were increased after ODG/R and these increaseswere inhibited by FOXO4 knockdown. ROS content and levels of LDH and MDA were increased after ODG/R and decreased by knockdown of FOXO4 (p < 0.05). Levels of CAT and SOD were reduced after ODG/R, and this reduction was reversed by knockdown of FOXO4 (p < 0.05).Conclusion: The results demonstrate that knockdown of FOXO4 promotes cell proliferation and inhibits cellular apoptosis via reduction of oxidative stress after CIR injury, indicating a new therapeutic target for the treatment of CIR injury. Keywords: FOXO4, Neuronal survival, Oxidative stress, Cerebral ischemia/reperfusion injury


2019 ◽  
Vol 22 (04) ◽  
pp. 122-130
Author(s):  
Rihab H Al-Mudhaffer ◽  
Laith M Abbas Al-Huseini ◽  
Saif M Hassan ◽  
Najah R Hadi

Sign in / Sign up

Export Citation Format

Share Document