Elsevier

Brain Research

Volume 1701, 15 December 2018, Pages 137-145
Brain Research

Research report
A novel Nrf2 activator, RS9, attenuates secondary brain injury after intracerebral hemorrhage in sub-acute phase

https://doi.org/10.1016/j.brainres.2018.08.021Get rights and content

Highlights

  • RS9 decreased brain edema and neurological deficit after ICH.

  • RS9 up-regulated HO-1 and SOD-1 expression after ICH.

  • RS9 decreased neuronal cell death after in vitro hemin injury.

  • RS9 has a protective effect for SBI after ICH.

Abstract

The poor prognosis of intracranial hemorrhage (ICH) is attributed to secondary brain injury (SBI), which is caused by oxidative stress. Blood components induce reactive oxygen species (ROS) over-production and cause cytotoxicity. We focused on the antioxidant system and investigated nuclear factor-erythroid 2-related factor 2 (Nrf2), which is a transcription factor that controls several antioxidant enzymes. We examined the effects of a novel Nrf2 activator, RS9, on SBI after ICH. ICH was induced by injecting autologous blood collected from the jugular vein (25 µL) into the striatum of mice. RS9 (0.2 mg/kg, i.p.) was administrated 0, 24, and 48 h after the induction of ICH. Using the ICH model, we measured brain edema, neurological function, neuronal damage and antioxidant proteins expression. We then investigated the mechanisms responsible for the effects of RS9 in vitro using the SH-SY5Y cell line. We used zinc protoporphyrin (ZnPP), a heme oxygenase-1 (HO-1) inhibitor, to elucidate the relationship between HO-1 expression and cell death in vitro in a hemin injury model. RS9 decreased brain edema, improved neurological deficits, decreased neuronal damage area and up-regulated HO-1 and superoxide dismutase 1 (SOD) expressions in the ICH mouse model. RS9 also suppressed neuronal cell death and ROS over-production in vitro. These protective effects were cancelled by the ZnPP co-treatment. Our results suggest that the activation of Nrf2 by RS9 exerts neuroprotective effects that are mediated by the attenuation of oxidative stress, and also that RS9 is an effective therapeutic candidate for the treatment for SBI after ICH.

Introduction

The central nervous system (CNS) is highly sensitive to oxidative stress, and the disruption of the redox system is known to induce neuronal damage (Liu et al., 2017). A main factor in oxidative stress is reactive oxygen species (ROS), which cause neuronal cell death and accelerate several neurological disorders in Parkinson’s disease, amyotrophic lateral sclerosis (ALS), and Alzheimer’s disease (D'Ambrosi et al., 2017, Dias et al., 2013). The removal of excess ROS produced in CNS disorders represents a potential approach to improve patient outcomes.

Nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor that controls antioxidant responses and plays important roles in attenuating oxidative stress in order to maintain homeostasis (Itoh et al., 2010, Suzuki et al., 2013, Yamazaki et al., 2015). Nrf2 induces the expression of cytoprotective and antioxidant genes, such as heme oxygenase 1 (HO-1), nicotinamide adenine dinucleotide phosphate quinone oxidoreductase 1 (NQO-1), and glutathione s-transferase (GST) (Zhang et al., 2013). Under normal conditions, Nrf2 binds the cytoskeletal protein, Kelch-like ECH-associated protein 1 (Keap1) with the N-terminal Nrf2-ECH homology 2 (Neh2) domain of Nrf2. After exposure to oxidative stress, Nrf2 dissociates from Keap1 and the complex is translocated into the nucleus. Nrf2 subsequently binds to the antioxidant response element (ARE) and activates antioxidant proteins, which exert cytoprotective and anti-inflammatory effects (Itoh et al., 2010).

A recent study demonstrated that oxidative stress is involved in stroke (Lee and Won, 2014), particularly in intracerebral hemorrhage (ICH) (Duan et al., 2016). ICH is a devastating disease that accounts for 20–30% of stroke cases. The mortality rate in one month is approximately 40–50% (Feigin et al., 2003). The main cause of ICH is the vascular burden caused by persistently high blood pressure conditions. Vascular fragility due to arteriosclerosis and congenital cerebral arteriovenous malformation is also considered to contribute to ICH (Sutherland and Auer, 2006, Wang and Dore, 2007).

Brain injury after ICH is divided into two types: primary brain injury (PBI) and secondary brain injury (SBI). PBI is mainly caused by the mass effect of a hematoma within the brain parenchyma, and induces neuronal cell death via mechanical damage within a few hours of the onset of ICH (Aronowski and Hall, 2005, Siaw-Debrah et al., 2017). SBI is caused by hematoma components within a few days (Wang and Dore, 2007). Hematoma components induce neuronal damage and are one of the factors aggravating SBI (Hua et al., 2007, Liu et al., 2010, Liu et al., 2017). In the pathology of SBI, blood components, such as hemoglobin (Hb), heme, and hemin, induce neuronal injury by accelerating the production of ROS (Duan et al., 2016, Qu et al., 2016). Previous studies showed that hemin induced oxidative stress and neuronal injury (Siaw-Debrah et al., 2017, Zhou et al., 2017). Heme is degraded into iron (Fe2+), bilirubin, and carbon monoxide. Fe2+ generates ROS, such as the hydroxyl radical, through the Fenton reaction, which leads to oxidative stress (Satoh et al., 2006, Thomas et al., 2009).

The activation of Nrf2 induces the up-regulation of downstream antioxidant enzymes, including HO-1 (Inoue et al., 2017). In recent years, the Nrf2 pathway has been attracting increasing attention as a therapeutic target for oxidative stress in CNS disorders. Dimethyl fumarate (DMF), an Nrf2 activator, has been applied to the treatment of multiple sclerosis in the clinical stage (Linker and Haghikia, 2016). We also reported that the Nrf2 activator, bardoxolone methyl (BARD) exerted several protective effects, and ameliorated cerebral ischemia reperfusion injury and hemorrhagic infract under the administration of warfarin (Imai et al., 2016, Takagi et al., 2014). The mechanism of activating Nrf2 involves BARD interacting with Keap1 in order to prevent Nrf2 ubiquitination and accelerating translocation into the nucleus (Ichikawa et al., 2009). RS9 (C32H43NO6: (1a, 2a, 21b)-2-cyano-21-hydroxy-3, 12-dioxo-1, 2-epoxyolean-9(11)-en-28-oate) is a novel Nrf2 activator that is biotransformed from BARD (Nakagami et al., 2015). A previous study demonstrated that RS9 exerts cytoprotective effects by suppressing blood-retinal barrier hyper-permeability in mice and rabbits (Nakagami et al., 2016). In addition, our findings showed that RS9 exerted protective effects in a light-induced retinal damage model and ischemic stroke model (Inoue et al., 2017, Yamauchi et al., 2016).

Some studies in the ICH field reported that DMF and nicotinamide improved ICH injury in vivo (Iniaghe et al., 2015, Wei et al., 2017). These findings indicate that the activation of Nrf2 protects against SBI in ICH. Therefore, we investigated whether RS9 exerts protective effects against SBI in vivo and in vitro.

Section snippets

RS9 protected the brain from SBI in the autologous blood injection ICH model

The protocol of this experiment was shown in Fig. 1A. In order to investigate the effects of RS9 in the autologous blood injection model, we initially measured brain edema volumes using the water content test. The water content of the ipsilateral side, particularly in the striatum, was significantly lower in the RS9 treatment group than in the vehicle group (Fig. 1B). We also performed the Garcia test and grid walk test to investigate the effects of RS9 on neurological deficits. In both tests,

Discussion

A few days after ICH, heme and hemin derived from the hematoma induce oxidative stress and cause neuronal cell death (Duan et al., 2016, Wang and Dore, 2007). This damage has been suggested to aggravate functional outcomes after ICH (Chen-Roetling et al., 2015a). We focused on the Nrf2 pathway as an approach to improve patient outcomes. Previous studies showed that the activation of Nrf2 exerts protective effects on neurons and glial cells (Chen-Roetling et al., 2015b, Iizumi et al., 2016).

Animals and experiments

All experimental designs and procedures were performed in accordance with the ARRIVE (Animal Research; Reporting In Vivo Experiments) guidelines, basic experiment guidelines (Hemorrhagic Stroke Academia Industry Roundtable, 2018) and approved by the animal experiment committees of Gifu Pharmaceutical University, Japan (Ethic nos. 2015-245, 2015-302, 2016-036, 2017-104). In all experiments, we used male ddY mice (12 weeks old, body weight; 40–50 g) purchased from Japan SLC, Ltd. (Hamamatsu,

Acknowledgments

We would like to thank Yasuhiro Nakagami (Daiichi Sankyo Co., Ltd.) for suggesting instructions regarding this work.

References (60)

  • S. Tsujii

    Zonisamide suppresses endoplasmic reticulum stress-induced neuronal cell damage in vitro and in vivo

    Eur. J. Pharmacol.

    (2015)
  • Y. Xin

    Sulforaphane prevents angiotensin II-induced cardiomyopathy by activation of Nrf2 via stimulating the Akt/GSK-3ss/Fyn pathway

    Redox Biol.

    (2018)
  • K. Yamauchi

    A novel nuclear factor erythroid 2-related factor 2 (Nrf2) activator RS9 attenuates brain injury after ischemia reperfusion in mice

    Neuroscience

    (2016)
  • M. Zhang

    Emerging roles of Nrf2 and phase II antioxidant enzymes in neuroprotection

    Prog. Neurobiol.

    (2013)
  • Y. Zhao

    Paeonol pretreatment attenuates cerebral ischemic injury via upregulating expression of pAkt, Nrf2, HO-1 and ameliorating BBB permeability in mice

    Brain Res. Bull.

    (2014)
  • Y.F. Zhou

    Hepcidin protects neuron from hemin-mediated injury by reducing iron

    Front. Physiol.

    (2017)
  • J.A. Araujo et al.

    Heme oxygenase-1, oxidation, inflammation, and atherosclerosis

    Front. Pharmacol.

    (2012)
  • J. Aronowski et al.

    New horizons for primary intracerebral hemorrhage treatment: experience from preclinical studies

    Neurol. Res.

    (2005)
  • A. Bir

    alpha-Synuclein-induced mitochondrial dysfunction in isolated preparation and intact cells: implications in the pathogenesis of Parkinson's disease

    J. Neurochem.

    (2014)
  • J. Chen-Roetling et al.

    Targeting heme oxygenase after intracerebral hemorrhage

    Ther. Targets Neurol. Dis.

    (2015)
  • J. Chen-Roetling

    Astrocyte overexpression of heme oxygenase-1 improves outcome after intracerebral hemorrhage

    Stroke

    (2015)
  • Z. Chen

    Osthole, a natural coumarin improves cognitive impairments and BBB dysfunction after transient global brain ischemia in C57 BL/6J mice: involvement of Nrf2 pathway

    Neurochem. Res.

    (2015)
  • N. D'Ambrosi et al.

    Neuroinflammation in amyotrophic lateral sclerosis: role of redox (dys)regulation

    Antioxid Redox Signal

    (2017)
  • V. Dias et al.

    The role of oxidative stress in Parkinson’s disease

    J. Parkinsons Dis.

    (2013)
  • X. Duan

    Intracerebral hemorrhage, oxidative stress, and antioxidant therapy

    Oxid. Med. Cell Longev.

    (2016)
  • J.H. Garcia

    Neurological deficit and extent of neuronal necrosis attributable to middle cerebral artery occlusion in rats. Statistical validation

    Stroke

    (1995)
  • Hemorrhagic Stroke Academia Industry Roundtable, P.

    Basic and translational research in intracerebral hemorrhage: limitations, priorities, and recommendations

    Stroke

    (2018)
  • Y. Hua

    Brain injury after intracerebral hemorrhage: the role of thrombin and iron

    Stroke

    (2007)
  • T. Ichikawa

    Dihydro-CDDO-trifluoroethyl amide (dh404), a novel Nrf2 activator, suppresses oxidative stress in cardiomyocytes

    PLoS ONE

    (2009)
  • T. Iizumi

    A possible role of microglia-derived nitric oxide by lipopolysaccharide in activation of astroglial pentose-phosphate pathway via the Keap1/Nrf2 system

    J. Neuroinflamm.

    (2016)
  • Cited by (32)

    • Levetiracetam, an Antiepileptic Drug has Neuroprotective Effects on Intracranial Hemorrhage Injury

      2020, Neuroscience
      Citation Excerpt :

      The protocol for the bICH model is shown in Fig. 1A, and the sampling area used to evaluate the cerebral edema is shown in Fig. 1B. Firstly, to investigate the effect of LEV on cerebral edema formation in the bICH model, brain water content was measured at 3 days after ICH induction. A previous report showed that brain water content was increased after ICH (Sugiyama et al., 2018). In the current study, both doses of LEV (50 or 150 mg/kg) significantly decreased the brain water content around the hematoma [F(2, 21) = 5.960, p = 0.009] (Fig. 1C).

    • Ghrelin attenuates secondary brain injury following intracerebral hemorrhage by inhibiting NLRP3 inflammasome activation and promoting Nrf2/ARE signaling pathway in mice

      2020, International Immunopharmacology
      Citation Excerpt :

      In regard of the effect of NLRP3 inflammasome on neuroinflammation and SBI post-ICH, interference with NLRP3 inflammasome may have neuroprotective effects against ICH-induced SBI. Studies have indicated that oxidative stress is a necessary contributor of SBI following ICH [10,11]. Oxidative stress can trigger or enhance inflammatory response, which in turn aid in enhanced oxidative stress [12].

    • Glutathione alleviates acute intracerebral hemorrhage injury via reversing mitochondrial dysfunction

      2020, Brain Research
      Citation Excerpt :

      Detailed information about each group is included in the figure legend descriptions below. Intracerebral hemorrhage was induced by autologous blood injection into the caudate nucleus according to a method described previously (Sugiyama et al., 2018; Tan et al., 2019a; Xiong et al., 2016). Briefly, mice were anesthetized by intraperitoneal injection of pentobarbital (40 mg/kg) and fixed on a stereotaxic brain locator.

    View all citing articles on Scopus
    1

    Contributed equally.

    View full text