Elsevier

Brain Research

Volume 1768, 1 October 2021, 147588
Brain Research

GSK-3β inhibitor TWS119 alleviates hypoxic-ischemic brain damage via a crosstalk with Wnt and Notch signaling pathways in neonatal rats

https://doi.org/10.1016/j.brainres.2021.147588Get rights and content

Highlights

  • TWS119 improves the tissue structure at 7 d after hypoxic-ischemic brain damage.

  • TWS119 attenuates cell apoptosis at 7 d after hypoxic-ischemic brain damage.

  • TWS119 up-regulates synaptic protein expression at 7 d after hypoxic-ischemic brain damage.

  • TWS119 activates Wnt signaling pathway at 24 h after hypoxic-ischemic brain damage.

  • TWS119 suppresses Notch signaling pathway at 7 d after hypoxic-ischemic brain damage.

Abstract

Preterm infant brain injury is a leading cause of morbidity and disability in survivors of preterm infants. Unfortunately, the effective treatment remains absent. Recent evidence suggests that GSK-3β inhibitor TWS119 has a neuroprotective role in adult brain injury by activation of Wnt/β-catenin signaling pathway. However, the role on neonatal brain injury is not yet explored. The study aims to evaluate the effect of TWS119 at 7 d after hypoxic-ischemic brain damage and investigate the mechanism that it regulates Wnt and Notch signaling pathways at 24 h after hypoxic-ischemic brain damage in neonatal rats. Three-day-old rats were randomly divided into 3 groups: sham group, HI group and TWS119 group. The neonatal rats were subjected to left carotid artery ligation followed by 2 h of hypoxia (8.0% O2). A single dose of TWS119 (30 mg/kg) was intraperitoneally injected 20 min prior to hypoxia-ischemia (HI). At 7 d after HI, TWS119 improved the tissue structure, reduced cell apoptosis, up-regulated bcl-2 expression, up-regulated the expression of PSD-95 and Synapsin-1. At 24 h after HI, it activated Wnt/β-catenin signaling pathway by up-regulation of β-catenin protein expression and wnt3a/wnt5a/wnt7a mRNA expression. Simultaneously, it suppressed Notch signaling pathway by down-regulation of Notch1 and HES-1 proteins expression. Our study suggested that TWS119 performed a neuroprotective function at 7 d after hypoxic-ischemic brain damage via a crosstalk with Wnt/β-catenin and Notch signaling pathways at 24 h after hypoxic-ischemic brain damage in neonatal rats.

Introduction

Preterm infant brain injury is a leading cause of disability in survivors of preterm infants. It has been reported that 5–10% of preterm survivors may suffer from severe neurological disability, such as cerebral palsy, and 25%-50% of survivors manifest with milder cognitive disabilities and behavioral problems (Back, 2015, Back, 2017). It places economic burden on the society and family. Previous evidence suggests that neuron death (Zhu et al., 2003, Zhu et al., 2005, Thornton et al., 2017) and synaptic injury (Wang et al., 2018, Liu et al., 2019) are involved in the pathogenesis of Preterm infant brain injury. However, the specific mechanism is still unclear and the effective therapy is very limited.

The Wnt and Notch pathways are two of highly conserved signaling pathways with a crosstalk in development and diseases (Collu et al., 2014). Evidence suggests that the activation of the Wnt signaling pathway may play an important role in the neuroprotective response after hypoxic-ischemic brain damage by diverse protective mechanisms, such as neurogenesis, neuroplasticity and angiogenesis (Lambert et al., 2016, Shruster et al., 2012, Sun et al., 2014). On the contrary, Notch signaling pathway is activated and plays a negative function in vivo and in vitro in hypoxic-ischemic brain damage (Xu et al., 2018). Evidence suggests that Notch pathway plays a role in neuronal apoptosis (Park et al., 2013). Moreover, Notch pathway interplays with other signaling pathways to induce cell death, such as Pin-1 (Baik et al., 2015) and p53 (Balaganapathy et al., 2018) pathways.

Evidence suggests that Wnt/β-catenin pathway is down-regulated by Glycogen synthase kinase-3β (GSK-3β) (Oliva et al., 2018), while Notch pathway is up-regulated by GSK-3β (Foltz et al., 2002). GSK-3β is a serine/threonine kinase and involved in the processes of neuronal plasticity and neurodegeneration (Jaworski et al., 2019). Inhibitors of GSK-3β perform a neuroprotective function on hypoxic-ischemic brain damage. Recently, TWS119, as one of GSK-3β inhibitors, plays an important neuroprotective role in adult stroke animal model by various mechanisms, such as anti-inflammatory activation (Song et al., 2019), attenuating hemorrhagic transformation (Wang et al., 2016), improving the blood-brain barrier (Wang et al., 2017). However, the role on neonatal brain injury is yet not explored. GSK-3β inhibitor SB216763 modulates Wnt and Notch pathways to equilibrate neurogenesis and gliogenesis in a rat model of Parkinson’s disease (Singh et al., 2018). Whether TWS119 plays a protective role in hypoxic-ischemic brain damage via modulation of Wnt and Notch pathways is unclear.

Some evidence suggests that the rat brain at postnatal days 10 is comparable to that of a term infant (Semple et al., 2013) which is the key period to assess brain maturation and injury of preterm infants (Duerden and Thompson, 2020). Therefore, in this study, postnatal day 10 (7 d after hypoxic-ischemic brain damage) is considered to assess the protective effect of TWS119 after hypoxic-ischemic brain damage. The aims of this study is to evaluate the neuroprotective effect of TWS119 at 7 d after hypoxic-ischemic brain damage and investigate the mechanism that it regulates Wnt and Notch signaling pathways at 24 h after hypoxic-ischemic brain damage in neonatal rats.

Section snippets

TWS119 improves the tissue structure at 7 d after hypoxic-ischemic brain damage

As shown in Fig. 1, hematoxylin-eosin staining results revealed that the sham group had normal neuron morphology, clear cytoplasm, and uniform and clear nucleus in the cortex and hippocampus (Fig. 1, A/D/G/J), while HI group exhibited abnormal, disordered and loose neurons arrangement, and Pyknotic neurons which were darkly stained pyknotic nuclei, cell body shrinkage, and intense eosinophilic cytoplasm (Fig. 1, B/E/H/K). Compared with HI group, TWS119 group showed the denser and ordered neuron

Discussion

In this study, we found that TWS119, as one of inhibitors of GSK-3β, improved tissue structure, reduced cell apoptosis and increased synaptic protein expression at 7 d after hypoxic-ischemic brain damage. During our research, it simultaneously activated Wnt signaling pathway and suppressed Notch signaling pathway at 24 h after hypoxic-ischemic brain damage in neonatal rats.

Cell death plays an important role in hypoxic-ischemic brain damage, which can be divided into four forms: necrosis,

Drug administration and experimental design

TWS119 (Sigma, SML 1271, dissolved in 1 % DMSO) was administrated to the neonatal rats by intraperitoneal injection 20 min prior to HI procedure (30 mg/kg) (Wang et al., 2016). The neonatal rats were randomly divided into 3 groups: Sham group: sham-operated rats; HI group: rats were subjected to HI and vehicle (1% DMSO) treatment; TWS119 group: rats were subjected to HI and TWS119 treatment. The neonatal rats were decapitated at 7 d after HI for investigating the functions of TWS119 on tissue

Declaration of Competing Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgments

This work was supported by the National Natural Science Foundation of China (No. 81771622 to Hong Cui) and the Natural Science Foundation of Beijing Municipality (No. 7202035 to Lijun Yang).

References (41)

  • B.D. Semple et al.

    Brain development in rodents and humans: Identifying benchmarks of maturation and vulnerability to injury across species

    Prog. Neurobiol.

    (2013)
  • A. Stadlin et al.

    Development of a postnatal 3-day-old rat model of mild hypoxic-ischemic brain injury

    Brain Res.

    (2003)
  • C. Thornton et al.

    Role of mitochondria in apoptotic and necroptotic cell death in the developing brain

    Clin. Chim. Acta

    (2015)
  • W. Wang et al.

    GSK-3beta as a target for protection against transient cerebral ischemia

    Int J Med Sci.

    (2017)
  • Z. Xu et al.

    Expression changes of the notch signaling pathway of PC12 cells after oxygen glucose deprivation

    Int. J. Biol. Macromol.

    (2018)
  • N. Zhang et al.

    Inhibition of miR-200b-3p alleviates hypoxia-ischemic brain damage via targeting Slit2 in neonatal rats

    Biochem. Biophys. Res. Commun.

    (2020)
  • T.V. Arumugam et al.

    Evidence that γ-Secretase-Mediated Notch Signaling Induces Neuronal Cell Death via the Nuclear Factor-κB-Bcl-2-Interacting Mediator of Cell Death Pathway in Ischemic Stroke

    Mol. Pharmacol.

    (2011)
  • S.A. Back

    White matter injury in the preterm infant: pathology and mechanisms

    Acta Neuropathol.

    (2017)
  • S. Baik et al.

    Pin1 promotes neuronal death in stroke by stabilizing Notch intracellular domain

    Ann. Neurol.

    (2015)
  • P. Balaganapathy et al.

    Interplay between Notch and p53 promotes neuronal cell death in ischemic stroke

    J. Cereb. Blood Flow Metab.

    (2018)
  • Cited by (10)

    • Exploring the potential of ginseng glycoprotein to improve learning and memory in mice via Notch signaling pathway and structural analysis using multi-information fusion based on liquid chromatography-mass spectrometry

      2023, Journal of Ethnopharmacology
      Citation Excerpt :

      Li et al. have revealed that Botch could alleviate microglial activation and prevent the development of neuroinflammation by antagonizing the activation of the Notch signaling pathway (H. Li et al., 2019). The glycogen synthase kinase-3β inhibitor reportedly inhibits the Notch signaling pathway by downregulating the expression of Notch1 and HES-1 proteins, exerts neuroprotective functions 7 days after hypoxic-ischemic brain damage, and inhibits apoptosis (Gao et al., 2021). Additionally, the Notch pathway is directly associated with inflammation.

    View all citing articles on Scopus
    View full text