Elsevier

Brain Research Bulletin

Volume 156, March 2020, Pages 33-42
Brain Research Bulletin

Cerebral ischemia-reperfusion aggravated cerebral infarction injury and possible differential genes identified by RNA-Seq in rats

https://doi.org/10.1016/j.brainresbull.2019.12.014Get rights and content

Highlights

  • Cerebral I/R aggravates neurological function injury and neuroinflammation.

  • TLR4/MyD88 inflammatory signaling pathway plays an important role after cerebral I/R.

  • HRAS/RAF1 neurotrophic signaling pathway plays an important roles after cerebral I/R.

Abstract

Numerous studies have shown that local excessive inflammatory response in brain tissue was an important pathogenesis of secondary injury following cerebral ischemia-reperfusion (I/R). However, the inflammatory-related targets and pathways after cerebral I/R injury are still unclear. This study was to investigate possible targets and mechanisms after cerebral I/R injury. Rats were subjected to transient or permanent middle cerebral artery occlusion (MCAO). Neurological deficit scores test was used to evaluate neurological function. Cerebral infarction was evaluated by MRI, TTC staining and Nissl staining. Microglia activation was detected by immunofluorescence using Iba-1 antibody. Inflammatory factors were detected by ELISA assay. RNA-sequencing transcriptome analysis was processed and the differential genes were verified by real-time quantitative PCR (qPCR) and western blotting. The results showed that neurological function of rats in I/R group was more severe than that in I group on the 7th after cerebral I/R. Therefore, the differences between cerebral ischemia and cerebral I/R for 7 days were studied in further study. The results showed that the levels of pro-inflammatory factors in I/R group were higher and the levels of anti-inflammatory factors were lower than those in I group. KEGG pathway and gene network enrichment analysis revealed that some common differential up- and down-regulated genes were involved in most of significant pathways. These common differential up-regulated genes belonged to TLR4/MYD88 inflammatory signaling pathway and common differential down-regulated genes belonged to HRAS/RAF1 neurotrophic signaling pathway. Interestingly, according to the genetic interaction analysis of string database, these up-regulated differential genes might promote the development of inflammation, while the down-regulated differential genes might inhibit the development of inflammation. Furthermore, qPCR and WB results verified that these pro-inflammatory genes in the I/R group were higher than those in the I group, while possible anti-inflammatory genes in the I/R group were lower than those in the I group. It is concluded that TLR4/MYD88 inflammatory signaling pathway and HRAS/RAF1 neurotrophic signaling pathway may play different roles after cerebral I or I/R and may be therapeutic targets for stroke recovery.

Introduction

Cerebral ischemia-reperfusion (I/R) injury refers to the recanalization of blood flow after cerebral ischemia, leading to further tissue damage and dysfunction, which is a complex pathophysiological process involving multiple factors (Feigin et al., 2009; Patel et al., 2013; Huang et al., 2006). Cerebral I/R can cause a wide range of microvascular dysfunction and changes in tissue barrier function. The inflammatory response caused by cerebral I/R injury can cause systemic inflammatory response or multiple organ dysfunction syndrome, accounting for 30–40 % of severe mortality (Perego et al., 2011; Eltzschig and Eckle, 2011; Zhang et al., 2018a, b). Therefore, it is urgent to prevent and treat cerebral I/R by exploring the mechanisms of cerebral I/R injury.

Numerous studies have shown that inflammatory immune response is widely involved in cerebral I/R injury, and local excessive inflammatory response in brain tissue is an important pathogenesis of secondary injury following cerebral I/R (Gelderblom et al., 2009; Zhou et al., 2013). Neuroinflammation is like a double-edged sword, which on the one hand aggravates neuronal damage and on the other hand promotes tissue repair and reincarnation (Patel et al., 2013). Therefore, exploring the mechanism of neuroinflammation and finding new targets will be of great significance in the prevention and treatment of cerebral I/R injury.

Neuroinflammation caused by activated microglia and astrocytes is a key determinant in the short-term and long-term prognosis after cerebral I/R (Kyritsis et al., 2012; Macrez et al., 2011; Tobin et al., 2014). Microglia, as a resident macrophage in the brain, directly participates in neuroinflammation after polarization activation (Peruzzotti-Jametti et al., 2014; Nowicka et al., 2008). There are two major types of macrophage/microglia phenotypes: classical activation (Ml) and alternative activation (M2). Although M1/M2 does not fully reflect the activation of macrophage/microglia diversity, it represents two extreme phenotypes of microglia activation and its mediated inflammatory response (Morioka et al., 1993; Kettenmann et al., 2011). Therefore, it has important value to regulate M1/M2 microglial activation to minimize the detrimental effect and/or maximize the protective role. In this study, RNA-sequencing was used to explore the key inflammatory targets which affecting reperfusion injury by comparing the reperfusion and non-reperfusion after cerebral ischemia in rats.

Section snippets

Animals

Male Sprague-Dawley rats (240−280 g) were purchased from Beijing Sibefu Biotechnology Co., Ltd., license number: SCXK (Beijing 2012-0001). All animals were housed in a room with controlled temperature of 23 ± 2 °C and humidity of 55 ± 5 % with a regular 12 h light-dark cycle and allowed to have water and food ad libitum. After acclimatization for 3 days, rats were subjected to transient middle cerebral artery occlusion (MCAO) for 90 min or permanent MCAO. The MCAO rats were inserted a suture

Differences of survival rate and neurobehavioral function between cerebral I/R and I in rats

As shown in Fig. 2A and B, there was no animal death in the sham group, but the mortality rate of the I/R group (23.3 %) was higher than that of the I group (13.3 %). These indicated that cerebral I/R had severe secondary damage. In addition, the body weight of normal rats increased steadily with time, and the body weight of rats in cerebral I/R group was slightly lower than that in cerebral I group (Fig. 2C).

The neurological behavior of the rats in the I/R group and the I group was both

Discussion

The pathophysiological process of cerebral I/R injury was complex and highly interactive (Lansberg et al., 2012; Gauberti et al., 2013, 2014). Inflammatory response played a key role in cerebral I/R injury (Smith et al., 2012; Li et al., 2017; Nagy and Nardai, 2017). The inflammatory response mediated microvascular dysfunction was an important reason why cerebral ischemia reperfusion caused secondary brain damage (Gulyas et al., 2012; Fernandez-Lizarbe et al., 2009).

The inflammatory response

Author statement

Xiao Cheng, Ying-Lin Yang, Wei-Han Li and Man Liu performed the animal experiments; Xiao Cheng and Yue-Hua Wang analyzed the data and wrote the manuscript; Yue-Hua Wang and Guan-Hua Du designed the experiments; and all authors read and approved the final manuscript.

Declaration of Competing Interest

The authors declare that there are no conflicts of interest.

Acknowledgements

This research was supported by the National Key Research & Development Plan (2018YFC0311005); the National Natural Science Foundation of China (81473383), the Drug Innovation Major Projects (2018ZX09711001-003-019), the Medical and Health Innovation Project of Chinese Academy of Medical Sciences (2016-I2M-3-007), and Innovation Fund for Graduate of Beijing Union Medical College (2017-1007-02 & 2018-1007-04).

References (40)

  • Y.R. Yang et al.

    Early and late treadmill training after focal brain ischemia in rats

    Neurosci. Lett.

    (2003)
  • W. Zhang et al.

    Salvianolic acid A attenuates ischemia reperfusion induced rat brain damage by protecting the blood brain barrier through MMP-9 inhibition and anti-inflammation

    Chin. J. Nat. Med.

    (2018)
  • W.W. Zhang et al.

    Buyang Huanwu Decoction ameliorates ischemic stroke by modulating multiple targets with multiple components: in vitro evidences

    Chin. J. Nat. Med.

    (2018)
  • G.J. Del Zoppo

    Acute anti-inflammatory approaches to ischemic stroke

    Ann. N. Y. Acad. Sci.

    (2010)
  • H.K. Eltzschig et al.

    Ischemia and reperfusion-from mechanism to translation

    Nat. Med.

    (2011)
  • S. Fernandez-Lizarbe et al.

    Critical role of TLR4 response in the activation of microglia induced by ethanol

    J. Immunol.

    (2009)
  • J.H. Garcia et al.

    Effects of CD11b/18 monoclonal antibody on rats with permanent middle cerebral artery occlusion

    Am. J. Pathol.

    (1996)
  • M. Gauberti et al.

    Ultra-sensitive molecular MRI of vascular cell adhesion molecule-1 reveals a dynamic inflammatory penumbra after strokes

    Stroke

    (2013)
  • M. Gauberti et al.

    Molecular magnetic resonance imaging of brainimmune interactions

    Front. Cell. Neurosci.

    (2014)
  • M. Gelderblom et al.

    Temporal and spatial dynamics of cerebral immune cell accumulation in stroke

    Stroke

    (2009)
  • Cited by (22)

    • Analysis of the mechanism of Buyang Huanwu Decoction against cerebral ischemia-reperfusion by multi-omics

      2023, Journal of Ethnopharmacology
      Citation Excerpt :

      Cerebral ischemia-reperfusion (I/R) injury is a pathophysiological disease involving multiple factors. It is after cerebral ischemia, the subsequent perfusion restores blood flow and recanalization, which further cause severe brain tissue damage and dysfunction (Cheng et al., 2020; Ding et al., 2020). Cerebral injury mechanisms remain unclear and complex, but existing studies believe that they involve the production of oxidative free radicals, neuroinflammation, Ca2+ overload, cell apoptosis, etc.(Guan and Chen, 2021; He et al., 2020; Shi et al., 2021).

    • A deuterohemin peptide protects cerebral ischemia-reperfusion injury by preventing oxidative stress in vitro and in vivo

      2023, Experimental Cell Research
      Citation Excerpt :

      CIRI can damage cerebral microvascular endothelial cells and the blood-brain barrier, causing nerve-cell infiltration and brain edema [4], leading to the necrosis and apoptosis of nerve cells in the penumbra of the brain [5]. After CIRI, the aerobic metabolism of glucose in brain cells is weakened, the synthesis of adenosine-triphosphate is reduced, and ROS are abundantly generated [6], resulting in a large release of excitatory amino acids (EAAs) [7], calcium overload [8] and neuroinflammation [9], ultimately leading to increased inflammation and brain damage. The accumulation of ROS causes oxidative stress, which in turn causes biomolecular damages, such as protein oxidation, lipid peroxidation, and oxidative DNA damage.

    • Xiao-Xu-Ming decoction extracts promotes mitochondrial biogenesis and improves neurobehavioral deficits in cerebral ischemia/reperfusion rats

      2022, Pharmacological Research - Modern Chinese Medicine
      Citation Excerpt :

      The water layer is gradient eluted by HP-20 macroporous resin chromatographic column, combined with 40% and 60% ethanol eluent (the mass ratio is 1.7:1), the solvent is recovered under reduced pressure, concentrated to an appropriate amount, dried in vacuum, crushed into powder, and then mixed with the middle layer powder to form XXM [14]. After acclimatization, rats were subjected to transient middle cerebral artery occlusion (MCAO) for 90 min and following reperfusion [17]. Briefly, the cerebral I/R rats were inserted a suture (0.36 ± 0.02 mm, 2636A2, Beijing Cinontech, China) into the middle cerebral artery for 90 min and then reperfusion.

    • Ovatodiolide protects ischemia-reperfusion-induced neuronal injury via microglial neuroinflammation via mediating SIRT1/NF-κB pathway

      2022, Brain Research Bulletin
      Citation Excerpt :

      Microglia are the main immune cell in CNS, and they become rapidly activated following cerebral ischemia injury (Szalay et al., 2016). Though activated microglia can remove cell debris and restore tissue integrity (Lalancette-Hébert et al., 2007), microglia cause detrimental effects, such as expanding ischemic infarction and aggravating neurological deficits by releasing neurotoxic substances (Cheng et al., 2020; Surinkaew et al., 2018; Jiang et al., 2021; Jiang et al., 2020). Thus, mitigating microglia-mediated neuroinflammation make much sense in treating stroke (Ma et al., 2017; Liu et al., 2016).

    View all citing articles on Scopus
    View full text