logo logo
Silencing of Long Noncoding RNA GAS5 Blocks Experimental Cerebral Ischemia-Reperfusion Injury by Restraining AQP4 Expression via the miR-1192/STAT5A Axis. Molecular neurobiology The long noncoding RNA (lncRNA) GAS5 has been shown to affect disease development in stroke. This study aimed to elucidate the regulatory mechanism of the lncRNA GAS5 on STAT5A in cerebral ischemia/reperfusion (I/R) injury. First, GAS5 and STAT5A levels in the blood of patients with stroke were determined. Then, a middle cerebral artery occlusion and reperfusion rat model was established in which short hairpin RNAs targeting GAS5 or STAT5A were intracranially injected, followed by the assessment of neurological function, cerebral injury and water content, and inflammation. Primary rat astrocytes were induced with oxygen-glucose deprivation/reoxygenation (OGD/R), and cell proliferation, apoptosis, and inflammation were evaluated. Moreover, the interplay between GAS5, miR-1192, and STAT5A and the binding of STAT5A to the AQP4 promoter were identified. GAS5 and STAT5A were strongly expressed in stroke patients, and inhibition of GAS5 or STAT5A in model rats improved neurological function, reduced infarction and neuronal apoptosis, and diminished cerebral water content and astrocyte activation. Furthermore, GAS5 or STAT5A downregulation restored proliferation and restrained apoptosis and inflammation in OGD/R-induced astrocytes. Mechanistically, GAS5 targeted miR-1192, which negatively regulated STAT5A. Astrocytes showed perturbed proliferation and strengthened apoptosis and inflammation when miR-1192 was inhibited despite the silencing of GAS5, while these unfavorable effects were abolished by STAT5A silencing. STAT5A binds to the AQP4 promoter and regulates its expression. Silencing of GAS5 and overexpresion of AQP4 led to lower cell viability and higher apoptosis and inflammation than GAS5 silencing alone. Overall, GAS5 silencing inhibited AQP4 through the miR-1192/STAT5A axis, thus alleviating cerebral I/R injury. 10.1007/s12035-022-03045-5
Formononetin protects against inflammation associated with cerebral ischemia-reperfusion injury in rats by targeting the JAK2/STAT3 signaling pathway. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie BACKGROUND:Formononetin is a type of phytoestrogen obtained from the Chinese medical herb Red Clover. It exhibits anti-neoplastic hepatoprotective, and neuroprotective properties. However, the anti-inflammatory effect of formononetin in cerebral ischemia-reperfusion injury has not been reported. OBJECTIVE:To explore the potential mechanism of action of formononetin in cerebral ischemia-reperfusion injury with regard to the JAK2/STAT3 signaling pathway. METHODS:Male SD rats were used to establish a middle cerebral artery occlusion (MCAO) model and randomly divided into 5 groups: Sham, MCAO, JAK2 Inhibitor (Ag490), Formononetin, Inhibitor + Formononetin. The protective effect of formononetin in MCAO rats was detected by performing neurological deficit testing, TTC staining, H&E staining, Nissl staining, ELISA, RT-PCR, western blotting and immunofluorescence. RESULTS:Formononetin significantly alleviated the neurological deficit and the pathological state of brain tissues, and reduced the volume of cerebral infarction, levels of IL-18 and TNF-α inflammatory factors in plasma, mRNA levels of IL-6 and IL-1β in rat brain tissue, and the protein levels of p-JAK2, p-STAT3, NLRP3, ASC, cl-Caspase-1, and cl-IL-1β in the MCAO rat brain tissue. CONCLUSION:Formononetin has anti-inflammatory effects. It may inhibit the relevant targets in the JAK2/STAT3 signaling pathway, thereby having a certain protective effect against cerebral ischemia-reperfusion injury. 10.1016/j.biopha.2022.112836
Anti-IL-23 exerted protective effects on cerebral ischemia-reperfusion injury through JAK2/STAT3 signaling pathway. Fan Lichao,Zhou Lichun Molecular biology reports Ischemia-reperfusion frequently occurs in ischemic cerebral vascular disease, during which the inflammatory signaling plays essential roles. The aim of this study was to discover the efficacy of the antibody to a key immune cytokine IL-23 (anti-IL-23) for the therapy of cerebral ischemia-reperfusion injury. We established the cerebral ischemia-reperfusion injury model by middle cerebral artery occlusion (MCAO). Anti-IL-23 injection attenuated lesions indicated by histology study. RT-PCR and Western blot were employed to detect the mRNA and protein expression of JAK2 and STAT3 after anti-IL-23 treatment. ELISA was utilized to measure the levels of MDA (malondialdehyde) and superoxide dismutase (SOD). Moreover, curcumin and IL-6 were implicated in the endogenous intervention of IL-23 signaling in vivo. Our data demonstrated that the treatment of anti-IL-23 might transcriptionally activate the classic immune pathway in the brain. Anti-IL-23 augmented phosphorylation levels of both JAK2 and STAT3, suggesting the amplification signaling of JAK/STAT after exogenous IL-23 intervention. Anti-IL-23 reduced ROS molecules of STAT downstream in the serum and brain. It also alleviated the injury by bringing down levels of MDA and SOD in the serum. JAK2 inhibitor could abolish the effect of anti-IL-23 whereas JAK3 ameliorated the injury. The combination of anti-IL-23 and JAK3i could reduce infarct volume more effectively. In summary, this study indicated that anti-IL-23 had protective effects against cerebral ischemia-reperfusion injury by targeting the immune specific JAK2-STAT3 in JAK/STAT pathway. 10.1007/s11033-021-06339-4
Narirutin-rich fraction from grape fruit peel protects against transient cerebral ischemia reperfusion injury in rats. Nutritional neuroscience Ischemic stroke is one of the leading causes of disability in adults worldwide. The present study was aimed to evaluate the efficacy of Narirutin-rich fraction (NRF), obtained from grape fruit peel, on cerebral ischemia/reperfusion injury in rats. Male Wistar rats (180-200 g) were subjected to bilateral carotid artery occlusion for 30 min followed by reperfusion for 24 h to induce cerebral ischemia/reperfusion injury. NRF (150, 300 mg/kg, oral) was administered for 7 days continuously before animals were subjected to ischemia/reperfusion injury. Various behavioral tests (for measurement of motor coordination, locomotor activity, and spatial memory), biochemical parameters (lipid peroxidation, superoxide dismutase, and catalase activity), and histopathological alterations were assessed. Seven-day NRF (150 and 300 mg/kg) pretreatment significantly improved neurobehavioral alterations and histological findings as compared to the disease control group. Further NRF treatment significantly reduced oxidative damage as indicated by improved lipid peroxidation, superoxide dismutase, and catalase activity as compared to disease control animals. The present study demonstrated the protective effect of NRF against cerebral ischemia/reperfusion injury in rats. The results suggest that NRF can be a potential pretreatment option against cerebral ischemia/reperfusion injury. 10.1080/1028415X.2020.1821518
Enhanced expression of P2X4 purinoceptors in pyramidal neurons of the rat hippocampal CA1 region may be involved ischemia-reperfusion injury. Xiang Zhenghua,Jiang Xin,Ji Rihui,Yuan Hongbin Purinergic signalling Ischemic stroke is the most serious disease that harms human beings. In principle, its treatment is to restore blood flow supply as soon as possible. However, after the blood flow is restored, it will lead to secondary brain injury, that is, ischemia-reperfusion injury. The mechanism of ischemia-reperfusion injury is very complicated. This study showed that P2X4 receptors in the pyramidal neurons of rat hippocampus were significantly upregulated in the early stage of ischemia-reperfusion injury. Neurons with high expression of P2X4 receptors are neurons that are undergoing apoptosis. Intraventricular injection of the P2X4 receptor antagonist 5-(3-bromophenyl)-1,3-dihydro-2H-benzofuro[3,2-e]-1,4-diazepin-2-one (5-BDBD) and PSB-12062 can partially block neuronal apoptosis, to promote the survival of neurons, indicating that ATP through P2X4 receptors is involved in the process of cerebral ischemia-reperfusion injury. Therefore, identifying the mechanism of neuronal degeneration induced by extracellular ATP via P2X4 receptors after ischemia-reperfusion will likely find new targets for the treatment of ischemia-reperfusion injury, and will provide a useful theoretical basis for the treatment of ischemia-reperfusion injury. 10.1007/s11302-021-09780-z
By targeting apoptosis facilitator BCL2L13, microRNA miR-484 alleviates cerebral ischemia/reperfusion injury-induced neuronal apoptosis in mice. Liu Xindong,Wang Xin,Zhang Lijuan,Zhou Yi,Yang Le,Yang Minghao Bioengineered Neuronal apoptosis was considered as one of the main factors of cerebral ischemia/reperfusion injury. Understanding the molecular regulatory mechanism of neuronal apoptosis under the cerebral ischemia/reperfusion injury may provide the novel therapeutic targets for cerebral ischemia/reperfusion injury. However, the molecular regulatory mechanism of neurons fate determination under the cerebral ischemia/reperfusion injury remains poorly understood. This study was aimed to delve into the related molecular mechanism of miR-484 on the regulation of cerebral ischemia/reperfusion injury-induced neuronal apoptosis in mice. In this study, quantitative real-time polymerase chain reaction assays revealed that the expression level of miR-484 was down-regulated in neurons following OGD. Then, CCK8 assay western blot assay, and flow cytometry assay verified that upregulation of miR-484 increased viability and inhibited apoptosis of neurons following OGD. Further bioinformatics methods and dual-luciferase reporter assay were applied together to anticipate and certify the interaction between miR-484 and BCL2L13. Finally, cerebral infarct size assessment and TUNEL staining confirmed that overexpression of miR-484 alleviated cerebral ischemia/reperfusion injury in mice, and overexpression of BCL2L13 could abolish the effect of miR-484-suppressed cell apoptosis. All these results suggested that miR-484 alleviates cerebral ischemia/reperfusion injury-induced neuronal apoptosis in mice by targeting apoptosis facilitator BCL2L13. 10.1080/21655979.2021.1898134
[Mechanism of Guanxinning against cerebral ischemia-reperfusion injury in mice based on transcriptomic analysis]. Zhongguo Zhong yao za zhi = Zhongguo zhongyao zazhi = China journal of Chinese materia medica Guanxinning, a modern Chinese medicine preparation composed of Salviae Miltiorrhizae Radix et Rhizoma and Chuanxiong Rhizoma, has the activities of activating blood circulation, resolving blood stasis, dredging vessels, and nourishing the heart. Clinical studies have demonstrated that Guanxinning has therapeutic effect on ischemic stroke, while the specific mechanism remains to be clarified. In this study, the potential mechanism of Guanxinning against cerebral ischemia-reperfusion injury in mice was explored and then verified in vitro. The mouse model of cerebral ischemia-reperfusion injury was established with middle cerebral artery embolization(MCAO) method. The pharmacological effects of Guanxinning on the model mice were investigated based on neurological function score, cerebral infarction area, pathological morphology, neuron injury, and apoptosis. The results showed that Guanxinning lowered neurological functional score, reduced cerebral infarction area, and ameliorated the histopathological morphology, neuronal damage, and apoptosis in the model mice. RNA samples were extracted from brain tissues and subjected to RNA sequencing(RNA-seq). The differentially expressed genes(DEGs) were screened with the thresholds of ■. GO function enrichment analysis and KEGG pathway enrichment analysis were performed for the 297 common DEGs, which indicated that Guanxinning may regulate the inflammatory response, oxidative stress response, energy metabolism, and apoptosis to treat cerebral ischemia-reperfusion injury in mice. Guanxinning exerted protective effect through inhibiting inflammation and reducing oxidative stress in hypoxia/reoxygenation injured SH-SY5 Y cells. Furthermore, Western blot indicated that Guanxinning down-regulated the protein levels of p-NF-κB p65 and p-p38 MAPK and up-regulated those of PPARγ and PGC-1α. The findings suggested that Guanxinning may inhibit inflammation and reduce oxidative stress by suppressing TNF signaling pathway and activating PPAR signaling pathway, thereby exerting the therapeutic effect on cerebral ischemia-reperfusion injury in mice. This study preliminarily reveals the mechanism of Guanxinning against cerebral ischemia-reperfusion injury and provides a basis for clinical application of Guanxinning. 10.19540/j.cnki.cjcmm.20220322.402