Identification of differentially expressed miRNA 48 h after cerebral ischemia–reperfusion injury in mice by the technique of miRNA microarray

2020 ◽  
Vol 98 (12) ◽  
pp. 855-860
Author(s):  
Yaping Zhang ◽  
Nan Ding ◽  
Hanlu Yi ◽  
Yudong Zhao ◽  
Zankai Ye ◽  
...  

The objective was to identify the differential expressed miRNA during cerebral ischemia–reperfusion injury (CIRI) process, thereby assisting in elucidating the mechanism of CIRI development and providing a potential target for CIRI prevention and treatment. Six mice were randomly assigned to two groups: control group and CIRI model group. A global cerebral IR model by four-vessel occlusion was prepared among the CIRI model group. Brain tissues were collected 48 h after reperfusion. Total RNA was extracted for each sample. miRNA microarrays were employed to detect the differentially expressed miRNA between the CIRI group and the control group. One differentially expressed miRNA was selected for verification by PCR. Compared with the control group, 69 miRNAs were significantly differential expressed in samples of the CIRI group, among which 50 miRNAs were upregulated and 19 miRNAs were downregulated. The real-time qPCR results indicated that the results of the miRNA microarray were reliable. A number of miRNAs were significantly regulated in the CIRI model, which suggested that miRNA was closely associated with the pathological alterations after ischemia. These identified miRNAs may provide directions and targets for the future pathological research of CIRI.

Stroke ◽  
2012 ◽  
Vol 43 (suppl_1) ◽  
Author(s):  
Qian G He ◽  
Lihua Yu ◽  
Wenming Xu ◽  
Jiachuan Duan ◽  
Jian Guo ◽  
...  

Background: Ischemic neuronal cell apoptosis is a principal neuropathological feature of stroke. The p75 neurotrophin receptor (p75NTR) induces apoptosis associated with JNK-p53-BAX pathway, p53 is the substrate of the HECT domain-containing E3 ubiquitin ligase Huwe1. Recent studies suggest that the precursor form of NGF (proNGF) binds to p75NTR, and withhold the interaction of proNGF/p75NTR is efficacious in reducing neuronal apoptosis. Studies on tumor and phylogeny, show that Huwe1 highly expressed in CNS, playing a role in the regulation of cell apoptosis and a variety of injury types. Our aim is to examine whether Huwe1 modulates proNGF/p75NTR in cerebral ischemia-reperfusion injury. Methods: Eight male rhesus monkey were randomly divided into two groups: sham(n=2) and model group(n=6). The model group was administered equal volume of PBS, or silencing huwe1 Lentiviral Vector or empty Vector in right caudatum and putamen using brain stereotaxic technology and subjected to transient right middle cerebral artery occlusion (MCAO) a month later. A battery of neurological evaluation and magnetic resonance imaging (MRI) were employed to evaluate animals. Animals were sacrificed 3 days after MCAO and brains were processed for testing transfection efficiency using GFP fluorescence and evaluating cell apoptosis using TUNEL staining. The related factors in caudatum, putamen, temporal lobe and hippocampus was analyzed with QPCR, western blotting with loading control GADPH, and Immunohistochemistry. Results: The model group showed significant functional deficit than sham group with neurological evaluation (p<0.05), whereas the silencing Huwe1 group’s was the most serious. In right caudatum and putament, ischemia-reperfusion injury increased the number of TUNEL+cells(p<0.05 vs sham group) and upregulation of huwe1, proNGF and p75NTR in protein and nucleotides level (p<0.05 vs sham group), but silencing Huwe1 group increased TUNEL+cells most significantly, produced profound modulation with decreased expression of Huwe1 and obvious upregulation of proNGF and p75NTR(p<0.05 vs PBS or empty Vector group) ( Figure 1 ). However, there is no significant difference in other positions (data not show). Conclusions: Huwe1 modulates proNGF/p75NTR in the cerebral ischemia-reperfusion injury, and p53 may be as a indirect fator involved in this process. Our findings provide a novel mechanism in regulating proNGF/p75NTR signaling, suggesting its potential therapeutic target in ischemic stroke.


2021 ◽  
Vol 15 ◽  
Author(s):  
Dazhuang Yi ◽  
Qunhui Wang ◽  
Yuhao Zhao ◽  
Yu Song ◽  
Hong You ◽  
...  

AimThis study was conducted in order to reveal the alterations in the N6-methyladenosine (m6A) modification profile of cerebral ischemia–reperfusion injury model rats.Materials and MethodsRats were used to establish the middle cerebral artery occlusion and reperfusion (MCAO/R) model. MeRIP-seq and RNA-seq were performed to identify differences in m6A methylation and gene expression. The expression of m6A methylation regulators was analyzed in three datasets and detected by quantitative real-time polymerase chain reaction, western blot, and immunofluorescence.ResultsWe identified 1,160 differentially expressed genes with hypermethylated or hypomethylated m6A modifications. The differentially expressed genes with hypermethylated m6A modifications were involved in the pathways associated with inflammation, while hypomethylated differentially expressed genes were related to neurons and nerve synapses. Among the m6A regulators, FTO was specifically localized in neurons and significantly downregulated after MCAO/R.ConclusionOur study provided an m6A transcriptome-wide map of the MACO/R rat samples, which might provide new insights into the mechanisms of 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