Abstract

Ischemic stroke is a kind of disease with high mortality and high disability, which brings a huge burden to the public health system (Hu et al. (2017)), and it poses a serious threat to the quality of life of patients. Cerebral ischemia/reperfusion injury is an important pathophysiological mechanism. This study aims to assess the mechanism of SNHG15 in the occurrence and development of cerebral ischemia/reperfusion injury of nerve cells and to investigate its potential value for diagnosis and treatment. SNHG15 targeted miRNA molecules and target genes were predicted with bioinformatics tools such as StarBase and TargetScan. The process of ischemic reperfusion in cerebral apoplexy in normal cultured and oxygen-glucose-deprived and reoxygenated neurons was simulated with RT-PCR and western blot technique. The expressions of SNHG15 and miR-141 were detected with qPCR, and the expressions of SIRT1 and p65, TNF-α, ROS, iNOS, and IL-6 were detected with western blot. Meanwhile, SNHG15 siRNAs and miR-141 mimics were transfected for SH-SY5Y, with western blot testing. And the expressions of miR-141, SIRT1, and p65, TNF-α, ROS, iNOS, and IL-6 were tested. According to the prediction with bioinformatics tools of StarBase and TargetScan, miR-141 is the target of lncSNHG15. In the luciferase reporter plasmid double-luciferase assay, miR-141 and SIRT1 were defined as the target relationship. In the oxygen-glucose-deprived reoxygenation model group, SNHG15 expression increased, miR-141 expression decreased, SIRT1 expression increased, and the expressions of p65, TNF-α, ROS, iNOS, and IL-6 decreased. In the SNHG15-siRNA-transfected oxygen-glucose-deprived reoxygenation cell model group, miR-141 expression increased, SIRT1 expression decreased, and the expressions of p65, TNF-α, and IL-6 increased compared with the si-NC group. In the miR-141-mimic-transfected oxygen-glucose-deprived reoxygenation cell model, SNHG15 expression decreased, SIRT1 expression decreased, and the expressions of p65, TNF-α, IL-1β, and IL-6 increased. In conclusion, SNHG15 expression increased during the process of oxygen-glucose-deprived reoxygenation, and the oxidative stress process was reduced by miR-141/SIRT1.

Highlights

  • Ischemic stroke is a kind of disease with high mortality and high disability, which brings a huge burden to the public health system [1]

  • Cerebral ischemia/reperfusion injury (CIR) is a kind of neurological injury caused by ischemia and hypoxia after cerebral ischemia, which may lead to microvascular dysfunction or even aggravation of neurological injury when blood perfusion is restored in a short period of time. erefore, we need to find safe and effective drugs to intervene in CIR, which may effectively improve the prognosis of patients. e oxygen-glucose-deprived (OGD) reoxygenation model can effectively simulate the pathophysiological process of brain CIR in vitro by simulating the glucose-free environment with a hypoxia device combined with a glucose-free medium

  • SH-SY5Y cells are human neuroblastoma, which can be used in neuronal injury in vitro models because they are similar to neurons in morphology, neurochemistry, and electrophysiological characteristics. erefore, OGD reoxygenation treatment on SHSY5Y cells can be used as a recognized cell model in the study of CIR, so as to be widely used [2, 3]

Read more

Summary

Introduction

Ischemic stroke is a kind of disease with high mortality and high disability, which brings a huge burden to the public health system [1]. E oxygen-glucose-deprived (OGD) reoxygenation model can effectively simulate the pathophysiological process of brain CIR in vitro by simulating the glucose-free environment with a hypoxia device combined with a glucose-free medium. After CIR, lncRNA H19 expression will increase to further promote the death of neurons by regulating miR-21 expression and initiating programmed necrosis of microglial cells [7]. Small nucleolar RNA host gene 14 (SNHG14) may contribute to the inflammatory response to CIR by regulating miR-136 expression, so as to aggravate its nerve injury [8]. After CIR, SNHG12 expression can increase and activate autophagy by regulating the functions of mesenchymal stem cells, increasing the death of neurons. Downregulation of SNHG12 expression can increase the functions of mesenchymal stem cells, reducing autophagy and apoptosis in cerebral microvascular endothelial cells, protecting the neurons [9]

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call