Elsevier

Brain Research Bulletin

Volume 157, April 2020, Pages 26-36
Brain Research Bulletin

Research report
Sodium aescinate provides neuroprotection in experimental traumatic brain injury via the Nrf2-ARE pathway

https://doi.org/10.1016/j.brainresbull.2020.01.019Get rights and content

Highlights

  • Sodium aescinate provides neuroprotection after TBI.

  • Sodium aescinate decreases TBI-induced oxidative stress.

  • Sodium aescinate reduces TBI-induced neuron cell death and apoptosis.

  • Sodium aescinate provides neuroprotection via the Nrf2-ARE pathway after TBI.

Abstract

Sodium aescinate (SA), a natural plant extract, has been proven to provide neuroprotection in neurological diseases. However, its role and the underlying pathophysiological mechanisms in traumatic brain injury (TBI) are still not well understood. The present study was aimed to investigate the protective effects of SA in both in vivo and in vitro TBI models. Mice or neurons were randomly divided into control, TBI, TBI + vehicle and TBI + SA groups. Neurologic severity score (NSS) was used to evaluate the neurological impairment. Brain water content and lesion volume were used to assess the brain injury degree. Malondialdehyde (MDA) and glutathione peroxidase (GPx) levels were used to estimate oxidative stress. Western blot was used to determine the protein levels. Nissl and terminal deoxynucleotidyl transferase-mediated dUTP nick 3’-end labeling (TUNEL) staining were used to measure cell death and apoptosis. Our results revealed that treatment of SA could improve neurological function, decrease cerebral edema and attenuate brain lesion after TBI. Furthermore, administration of SA suppressed TBI-induced oxidative stress, neuron cell death and apoptosis. In addition, SA activated the nuclear factor erythroid 2-related factor 2 (Nrf2)-antioxidant response element (ARE) pathway after TBI. However, SA failed to provide neuroprotection following TBI in Nrf2−/− mice. Taken together, our results provided the first evidence that SA treatment played a key role in neuroprotection after TBI through the Nrf2-ARE pathway.

Introduction

Traumatic brain injury (TBI) is one of the leading causes of disability and death in modern society, resulting in high medical costs (Brooks et al., 2013). It is defined as any head injury with traumatic etiology, such as penetrating or blunt trauma and non-accidental injury. The pathological process of TBI includes both primary and secondary brain injury. Although the primary brain damage is the major factor determining the patients’ outcomes, the secondary brain damage induced by multiple pathological processes, such as inflammation, cell death, apoptosis, oxidative stress, provides the possibility for clinical intervention (Ding et al., 2014; Zhang and Wang, 2017). Despite the efforts on searching effective methods to attenuate the secondary brain injury, patients suff ;ering with TBI always end up with poor prognosis (Sun et al., 2015). Therefore, new and effective strategies of treatment are urgently needed to reduce the heavy disease and economic burden.

Nuclear factor erythroid 2-related factor 2 (Nrf2) is a member of the basic leucine zipper (bZIP) family of transcription factors (Villeneuve et al., 2010). Under basal conditions, Nrf2 is retained in the cytoplasm by its inhibitor, Kelch-like ECH-associated protein 1 (Keap1) (Kobayashi et al., 2004). Once activated, Nrf2 dissociates from Keap1, translocates into the nucleus and promotes the transcription of antioxidative Phase II enzymes such as heme oxygenase-1 (HO-1) and nicotinamide adenine dinucleotide phosphate, quinine oxidoreductase-1 (NQO-1) by binding to antioxidant response element (ARE) (de Vries et al., 2008).

Sodium aescinate (SA) is a triterpene saponin that extracted from the seeds of chestnut (Chen et al., 2017). The therapeutic properties of SA such as anti-inflammatory, relieving tissue edema and recovering vascular permeability have been confirmed in previous studies (Zhang et al., 2015). In addition, SA has been suggested to obliterate the excessive reactive oxygen species (ROS) when cellular homeostasis was disturbed in the field of spinal cord injury (SCI) (Cheng et al., 2016), hepatocellular carcinoma (HCC) (Wang et al., 2012), and so on. However, the role of SA in TBI is seldom illustrated. The purpose of the present study was to determine whether SA administration after TBI could attenuate brain injury in TBI models.

Section snippets

Animal preparation

This study was carried out in accordance with the principles of the Basel Declaration and recommendations of the National Institute of Health Guide for the Care and Use of Laboratory Animals (NIH Publications No. 8023, revised 1978). The protocol was approved by the Institutional Animal Care and Use Committee of Nanjing University (Nanjing, China). Male ICR mice (28−32 g) were obtained from Animal Center of Jinling hospital (Nanjing, China). Mice were housed on a 12 h light/dark cycle at

SA provided neuroprotection after TBI

To determine whether SA protected mice against TBI, we firstly used the NSS to evaluate the motor performance of mice after TBI. As showed in Fig. 1A, SA-treated mice showed better motor performance than that of the vehicle-treated mice at 1 day. In addition, a significant difference was still detectable at 3 days. However, there was no significant difference between the TBI + vehicle and TBI + SA groups at 7 days (P > 0.05).

We further used brain water content to confirm the neuroprotective

Discussion

To the best of our knowledge, this is the first study exploring the role of SA in TBI models. The main findings of our study were as follows: (1) SA could improve neurological function, decreased cerebral edema and attenuated lesion volume after TBI. (2) SA suppressed TBI-induced oxidative stress, neuron cell death and apoptosis. (3) SA activated the Nrf2-ARE pathway after TBI. (4) SA lost its neuroprotective effects in Nrf2−/− mice.

Recently, there is growing evidence showing that oxidative

Conclusion

Our studies indicated that SA could provide neuroprotection against TBI by combating oxidative stress, cell death and apoptosis through the Nrf2-ARE pathway. These results demonstrated that SA may be a promising drug for the treatment of TBI.

Author statement

Li Zhang: Conceptualization, Methodology, Software. Maoxing Fei: Data curation, Writing- Original draft preparation. Yihao Zhu: Conduct the experiment. Handong Wang: Writing- Reviewing, Editing, Validation.

Declaration of Competing Interest

The authors declare no potential conflicts of interest.

Acknowledgments

This work was supported by Grants from the National Natural Science Foundation of China (No. 81672503) from Handong Wang and the National Natural Science Foundation of China (No. 81702484) from Li Zhang.

References (33)

  • L. Zhang et al.

    Fisetin alleviates oxidative stress after traumatic brain injury via the Nrf2-ARE pathway

    Neurochem. Int.

    (2018)
  • Y. Zhao et al.

    Interactions between SIRT1 and MAPK/ERK regulate neuronal apoptosis induced by traumatic brain injury in vitro and in vivo

    Exp. Neurol.

    (2012)
  • C. Angeloni et al.

    Traumatic brain injury and NADPH oxidase: a deep relationship

    Oxid. Med. Cell. Longev.

    (2015)
  • L. Chen et al.

    Effect of sodium aescinate treatment on PCOS rat model with insulin resistance

    Bratisl. Lek. Listy

    (2017)
  • X.J. Fan et al.

    Effects of sodium aescinate on bcl-2 and caspase-3 expression and apoptosis after focal cerebral ischemia reperfusion injury in rats

    Zhong nan da xue xue bao. Yi xue ban = Journal of Central South University. Medical sciences

    (2005)
  • J. Fang et al.

    Baicalin provides neuroprotection in traumatic brain injury mice model through Akt/Nrf2 pathway

    Drug Des. Dev. Ther.

    (2018)
  • Cited by (0)

    1

    Li Zhang and Maoxing Fei equally contributed to this work.

    View full text