Elsevier

Neuroscience Letters

Volume 627, 3 August 2016, Pages 92-99
Neuroscience Letters

Research article
An apoE-derived mimic peptide, COG1410, alleviates early brain injury via reducing apoptosis and neuroinflammation in a mouse model of subarachnoid hemorrhage

https://doi.org/10.1016/j.neulet.2016.05.058Get rights and content

Highlights

  • An apoE-derived peptide, COG1410, alleviated neurological deficits in SAH mice.

  • Anti-apoptotic and anti-inflammatory effects of COG1410 contribute to the neuroprotective effect of COG1410.

  • COG1410 regulates apoptotic and inflammatory signals, and suppresses microglial activation in the acute phase of SAH.

Abstract

This study investigated the neuroprotective effects of COG1410, an apoliporotein E (apoE)-derived mimic peptide, against early brain injury (EBI) after subarachnoid hemorrhage (SAH). SAH was induced in C57BL/6 J mice (n = 68) by endovascular perforation. Mice received intravenous injection of COG1410 (2 mg/kg) or equal volume of vehicle (saline). The mortality rate, neurological score, rotarod latencies, cell apoptosis, microglial activation, pro-inflammatory cytokines production and protein levels of apoptotic and inflammatory markers were assessed at 24 h after sham operation or SAH. Results showed that COG1410 alleviated the neurological deficits associated with SAH. Compared with vehicle treatment group, the number of apoptotic cells and activated microglia decreased significantly in the COG1410 treated group. COG1410 enhanced Akt activation and suppressed caspase-3 cleavage. The imbalance of Bax and Bcl-2 induced by SAH was regulated by COG1410. Additionally, COG1410 attenuated cytokines production of IL-1β, IL-6 and TNF-α and suppressed the activation of JNK/c-Jun and NF-κB. Taken together, COG1410 protected against EBI via reducing apoptosis and neuroinflammation, through mechanisms that involve the regulation of apoptotic signaling and microglial activation. COG1410 is a potential neuroprotective agent for SAH treatment.

Introduction

Subarachnoid hemorrhage (SAH) is a fatal neurovascular disease with high mortality and morbidity [1]. Traditional explanations for the poor prognosis of SAH include delayed cerebral ischemia caused by cerebral vasospasm (CVS). In recent years, the direct brain injury induced by the initial bleeding in the acute phase of SAH, termed as early brain injury (EBI), has attracted more attention [2]. Considering intact neurons as the basis of normal neurological functions, SAH has long been reported to cause neuronal apoptosis leading to declines in neurological functions [3], [4]. Indeed, inhibition of neuronal apoptosis alleviates neurological deficits in SAH animal models [4], [5]. Neuroinflammation is another major pathological process in EBI following a SAH event [6], and anti-neuroinflammation has been proven to be beneficial for SAH animal models [7]. Therefore, targeting apoptosis and neuroinflammation is a promising therapeutic strategy for EBI after SAH.

Apolipoprotein E (apoE) is the major apolipoprotein in the central nervous system (CNS). ApoE not only participates in lipid metabolism, but also exerts anti-neuroinflammatory and anti-apoptotic effects via binding to its functional receptors [8], [9]. Like most other proteins, apoE holoprotein has not been found to cross the blood brain barrier (BBB) for reasons that include its 34-kDa molecular weight. COG1410 is an apoE-mimic peptide derived from amino acids 138–149 of the receptor region of the apoE holoprotein with aminoisobutyric acid (Aib) substitutions at positions 140 and 145 (acetyl-AS-Aib-LRKL-Aib-KRLL-amide). COG1410 has been shown to penetrate the BBB [10]. The therapeutic window for the treatment of traumatic brain injury (TBI) animals expands to 120 min [11], making it a potential neuroprotective agent for SAH treatment via intravenous administration. COG1410 has been proven to protect against neurological deficits and cerebral-vasospasm (CVS) after SAH [12]. However, the detailed mechanisms and the role of COG1410 in neural apoptosis and neuroinflammation in the acute phase of SAH are poorly understood. The present study aimed to investigate the anti-apoptotic and anti-inflammatory effects of COG1410 against EBI after SAH.

Section snippets

Animals and experimental design

All experiments were conducted in strict accordance with the recommendations of the Guide for the Care and Use of Laboratory Animals of China. The protocol was approved by the committee on the Ethics of Animal Experiments of the Chongqing Medical University. Adult (10–12 weeks) male C57BL/6 J mice were obtained from the Laboratory Animal Center of Chongqing Medical University. Mice (n = 68) were randomly divided in three groups: (1) the sham group (n = 15): mice underwent a sham operation and

Statistical analysis

All data was expressed as the mean ± standard deviation (SD) and compared with repeated measures of analysis of variance (ANOVA). Bonferroni’s post hoc method was applied for comparison among groups. The difference of mortality rate was analyzed using chi-square test. All statistic values were calculated using SPSS 19.0 (SPSS, Inc. Chicago, USA). Significance was assumed at P < 0.05.

SAH grade and mortality

The severity of SAH based upon bleeding scale scoring was divided into 3 levels [15]: mild SAH, 0–7; moderate SAH, 8–12; severe SAH, 13–18. It is reported that the behavioral deficits of mild SAH animals is similar to that of the sham [18]. A total of 6 mice with mild SAH were excluded from this study. The group severity scores were 0, 13.3 ± 2.8, and 12.8 ± 3.3 in the sham group, SAH + vehicle group and SAH + COG1410 group, respectively (Fig. 1A). There was no significant difference between the two

Discussion

Experimentally applied SAH induced evident mortality and neurological deficits accompanied by neuronal apoptosis and neuroinflammation in mice. COG1410 alleviated apoptosis in the cortex and modulated the apoptosis-related signaling pathways. Microglial activation and pro-inflammatory cytokines production was efficiently suppressed by COG1410 treatment. The attenuation of these pathological changes may contribute to the improvement of neurological function of SAH mice treated with COG1410.

The

Author contributions

Y.J. and XC.S. conceived and designed the experiments. Y.W., JW.P. and JH.P. performed the experiments. Y.W. interpreted the data and drafted the manuscript. F.C., M.P.V. and FQ.L contributed to the data analysis and manuscript revision.

Author disclosure statement

No conflicting interests exist.

Acknowledgement

This work was supported by grants from the National Natural Science Foundation of China (No. 81371319 and No. 81571159), the Program for New Century Excellent Talents in University (NCET-12-1057), the Foundation for Outstanding Youth Academic Technology Leaders of Sichuan province (2014JQ0022), and the National Key Clinical Specialty Construction Project of China (No.{2011}170).

References (37)

  • L. Jiang et al.

    Effects of ApoE on intracellular calcium levels and apoptosis of neurons after mechanical injury

    Neuroscience

    (2015)
  • A. Pocivavsek et al.

    Microglial low-density lipoprotein receptor-related protein 1 modulates c-Jun N-terminal kinase activation

    J. Neuroimmunol.

    (2009)
  • M. Gotthardt et al.

    Interactions of the low density lipoprotein receptor gene family with cytosolic adaptor and scaffold proteins suggest diverse biological functions in cellular communication and signal transduction

    J. Biol. Chem.

    (2000)
  • S. Lin et al.

    Heme activates TLR4-mediated inflammatory injury via MyD88/TRIF signaling pathway in intracerebral hemorrhage

    J. Neuroinflammation

    (2012)
  • J. van Gijn et al.

    Subarachnoid haemorrhage: diagnosis causes and management

    Brain

    (2001)
  • H. Endo et al.

    Akt/GSK3beta survival signaling is involved in acute brain injury after subarachnoid hemorrhage in rats

    Stroke

    (2006)
  • Y. Hong et al.

    Neuroprotective effect of hydrogen-rich saline against neurologic damage and apoptosis in early brain injury following subarachnoid hemorrhage: possible role of the Akt/GSK3beta signaling pathway

    PLoS One

    (2014)
  • G.F. Prunell et al.

    Inflammation in the brain after experimental subarachnoid hemorrhage

    Neurosurgery

    (2005)
  • Cited by (49)

    • Therapeutic potential of ApoE-mimetic peptides in CNS disorders: Current perspective

      2022, Experimental Neurology
      Citation Excerpt :

      COG1410 showed neuroprotection in Subarachnoid hemorrhage (SAH) which was mediated by its anti-inflammatory and anti-apoptotic action (increased activation of Akt and suppressed caspase-3 cleavage). It reduced the level of the cytokines (IL-1β, IL-6, and TNF-α) by suppressing the NF-κB and JNK/c-Jun (Wu et al., 2016). In early brain injury (EBI) followed by the SAH, COG1410 has raised the level of Beclin-1, LC3B-II and p-GSK-3β proteins (makers for the activation of the autophagy) thus, suggesting COG1410 acts as an autophagy inducer (Li et al., 2018; Wu et al., 2016).

    • Behavioral tests in rodent models of stroke

      2020, Brain Hemorrhages
      Citation Excerpt :

      Consistent with this report, rats with neural stem cell transplantation exhibited improved performance in rotarod test at day 60 after ICH.103 Similarly, SAH mice showed reduced latency before falling compared to sham controls at 24 hours after injury.104 In another study, SAH rats displayed faster falling in the rotarod test at 48 hours after injury, and Nrf2 activator improved their performance.105

    • Protein phosphatase 2A as a therapeutic target in inflammation and neurodegeneration

      2019, Pharmacology and Therapeutics
      Citation Excerpt :

      The possible involvement of PP2A in therapeutic effects of FTY720 has not been explored in these contexts. However, protective effects of PP2A-activating peptides have been demonstrated in experimental models of ischemic or physical brain injury (Cao et al., 2016; Hoane, Kaufman, Vitek, & McKenna, 2009; Kaufman et al., 2010; Laskowitz et al., 2012; Pang et al., 2017; Qin et al., 2017; Tukhovskaya, Yukin, Khokhlova, Murashev, & Vitek, 2009; Wu et al., 2016). In some of these studies, protection of neurological function was accompanied by prevention of BBB dysfunction (Cao et al., 2016; Pang et al., 2017; Qin, You, et al., 2017), suggesting at least some degree of overlap between therapeutic effects of FTY720 and PADs.

    View all citing articles on Scopus
    View full text