Journal List > Korean J Physiol Pharmacol > v.29(3) > 1516090425

Li, Yi, Qian, Xu, Min, Wang, Guo, Yu, and Zhang: Shikonin attenuates blood–brain barrier injury and oxidative stress in rats with subarachnoid hemorrhage by activating Sirt1/Nrf2/HO-1 signaling

Abstract

Subarachnoid hemorrhage (SAH) is a serious intracranial hemorrhage characterized by acute bleeding into the subarachnoid space. The effects of shikonin, a natural compound from the roots of Lithospermum erythrorhizon, on oxidative stress and blood–brain barrier (BBB) injury in SAH was evaluated in this study. A rat model of SAH was established by endovascular perforation to mimic the rupture of intracranial aneurysms. Rats were then administered 25 mg/kg of shikonin or dimethylsulfoxide after surgery. Brain edema, SAH grade, and neurobehavioral scores were measured after 24 h of SAH to evaluate neurological impairment. Concentrations of the oxidative stress markers superoxide dismutase (SOD), glutathione (GSH), and malondialdehyde (MDA) in the brain cortex were determined using the corresponding commercially available assay kits. Evans blue staining was used to determine BBB permeability. Western blotting was used to quantify protein levels of tight junction proteins zonula occludens-1, Occludin, and Claudin-5. After modeling, the brain water content increased significantly whereas the neurobehavioral scores of rats with SAH decreased prominently. MDA levels increased and the levels of the antioxidant enzymes GSH and SOD decreased after SAH. These changes were reversed after shikonin administration. Shikonin treatment also inhibited Evans blue extravasation after SAH. Furthermore, reduction in the levels of tight junction proteins after SAH modeling was rescued after shikonin treatment. In conclusion, shikonin exerts a neuroprotective effect after SAH by mitigating BBB injury and inhibiting oxidative stress in the cerebral cortex.

INTRODUCTION

Subarachnoid hemorrhage (SAH) refers to the cerebrovascular condition in which blood vessels in the spinal cord or brain rupture and the subarachnoid space is subsequently filled with blood [1]. Most cases (~85%) of SAH are caused by the rupture of intracranial aneurysms. The risk of death after SAH increases with age in elderly patients [2]. Secondary brain injury after SAH, which can present as blood–brain barrier (BBB) breakdown, neuroinflammation, oxidative stress, brain edema, and neural cell death, has been attracting increasing research attention [3]. Secondary brain injury within a short time (72 h) after SAH is defined as early brain injury (EBI) [4,5] and is closely associated with poor outcomes in patients with SAH [6]. Therefore, there is an urgent need to identify effective drugs and develop novel therapeutic strategies to improve patient outcomes.
Shikonin is a natural compound obtained from the roots of Lithospermum erythrorhizon [7]. It is called “Zicao” in traditional Chinese medicine [8]. Shikonin exerts numerous pharmacological effects including antioxidant, anti-inflammatory, antitumor, and sepsisalleviating effects [9,10]. It arrests PD-L1 (an immune checkpoint protein) expression in sepsis by reducing pyruvate kinase M2 levels [7]. Recently, shikonin has been demonstrated to attenuate levels of inflammatory mediators (TNFα, IL-4, IL-6, and IL-17) in blood samples of Sjögren syndrome mouse model by inactivating the MAPK signaling [11]. Furthermore, it mitigates neuronal apoptosis by suppressing neuroinflammation and oxidative stress in Parkinson’s disease by regulating Akt, NF-κB, and MAPK signaling [12]. These findings highlight the significance of shikonin in neuron-related disorders. More importantly, shikonin exerts a protective effect on neuronal cells and facilitates the absorption of hemorrhages in the brain [13]. However, its precise mechanism in alleviating SAH is unclear.
Current evidence shows that activation of SIRT1/Nrf2/HO-1 signaling is an essential cellular defense mechanism that protects against reactive oxygen species (ROS) [14,15]. Therefore, activation of the SIRT1/Nrf2/HO-1 pathway can counteract oxidative damage in many animal models of organ injury. Shikonin can protect against renal oxidative stress in rats by activating the SIRT1/Nrf2/HO-1 pathway [16]. Moreover, SIRT1 can significantly induce antioxidation and play an important role in protecting the nervous system after SAH [17,18]. Therefore, whether shikonin exerts an antioxidant effect in the brain tissues of rats after SAH by regulating the SIRT1/Nrf2/HO-1 pathway was explored in this study. Accordingly, a rat model of SAH was established by endovascular perforation to mimic the rupture of intracranial aneurysms. Our findings highlight the protective role of shikonin in cerebral injury and suggest its potential in treating brain injury after SAH.

METHODS

Animals

A total of 60 male Sprague–Dawley rats (300 ± 20 g, 9–10 weeks old) were purchased from Vital River Animal Technology. All animals were housed in a standard environment at a temperature of 24°C and humidity of 52%–60%, and subjected to a 12-h/12-h light/dark cycle. All rats were provided free access to food and water. All animal experiments were approved by Wuhan Myhalic Biotechnology Co., Ltd (HLK-202311169) and were conducted according to the Guide for the Care and Use of Laboratory Animals.

Establishment of a rat model of SAH

Endovascular perforation was conducted following a previously published protocol to mimic the rupture of intracranial aneurysms [19-21]. Rats were anesthetized with 3% isoflurane through tracheal intubation using a rodent ventilator (Shanghai Ranger Apparatus). Next, a sharpened 4-0 nylon suture was inserted into the left internal carotid artery from the cut of the external carotid artery stump. The suture was put out after perforating the bifurcation of the left anterior and middle cerebral artery, followed by suture of the incision. Sham-operated rats were subjected to a similar surgery except artery perforation.

Grouping and drug administration

Rats were randomly assigned into the following 4 groups: (a) sham + dimethyl sulfoxide (DMSO) group (n = 10): sham-operated rats received equal volumes of phosphate-buffered saline (PBS) + 1% DMSO; (b) sham + shikonin group (n = 10): rats underwent sham operation and received 25 mg/kg shikonin; (c) SAH + DMSO group (n = 20, 5 rats were excluded due to death): rats were subjected to SAH and administered equal volumes of PBS + 1% DMSO; (d) sham + shikonin group (n = 20, 2 rats were excluded due to death): rats were subjected to SAH and administered 25 mg/kg shikonin. Shikonin was obtained from Santa Cruz Biotechnology (517-89-5) and dissolved in DMSO. Shikonin concentration used in this study was determined based on that reported previously [22]. Rats were intraperitoneally administered 25 mg/kg shikonin after 1 h of SAH.

SAH grading

SAH severity was evaluated by 2 performers based on the SAH grading scale [20]. Rats were anesthetized and sacrificed after 24 h of SAH induction. The ventral side of the brain was divided into 6 areas. The degree of SAH in each area was scored from 0–3 points based on the volume of blood clots on the basal surface. The specific explanation of each grade was described as reported previously [23]. Rats were scored on 6 segments, and those with an average score of < 7 were excluded from subsequent experiments.

Neurological scoring

The neurological function of rats was assessed using a beam balance test after 24 h of SAH. Briefly, rats were placed on a beam (590 cm) and allowed to walk. Next, the distance that each rat could walk within 1 min was measured and scored. The scoring criteria were as follows:
(a) 4: rats could walk more than 20 cm; (b) 3: rats could walk a distance less than 20 cm; (c) 2: rats could walk but fell from the beam; (d) 1: rats could not walk but could stay on the beam; (e) 0: rats could not walk and fell from the beam. The assay was performed in triplicate.
The Garcia scoring system was also used to measure rats’ neurological functions after 24 h of SAH. Rats were scored from 6 aspects, and the score for each aspect was graded on 0–3 points. The 6 aspects were spontaneous activity, autonomous moving of each limb, forelimb outstretching, climbing, proprioception of tentacles, and body proprioception.

Assessment of brain water content

Brain edema in rats was determined by measuring water content in the brain. Rats were anesthetized and sacrificed 24 h after SAH, and their brains were collected. After removal of clotted blood, the initial weight of the wet brains was obtained. Next, the brains were dried in an oven at 100°C for 24 h, and the dry weight was obtained. The brain water content was defined as (wet weight − dry weight)/wet weight × 100%.

Determination of malondialdehyde (MDA), glutathione (GSH), and superoxide dismutase (SOD) contents

The concentrations of MDA and antioxidant enzymes (SOD and GSH) in the brain cortex were measured using the corresponding assay kits following the manufacturers’ recommendations. The kits included a lipid peroxidation (MDA) assay kit (ab118970, Abcam), SOD assay kit (ab65354, Abcam), and GSH assay kit (ab239727, Abcam). A microplate reader (Thermo Fisher Scientific) was used to determine MDA, SOD, and GSH concentrations at a wavelength of 535 nm, 450 nm, and 420 nm, respectively.

Determination of ROS levels

An ROS assay kit (Jiancheng Bioengineering Institute) was used to determine ROS production in brain tissues following the manufacturer’s recommendations. ROS content was determined by measuring fluorescence intensity/mg protein.

Evans blue staining

Evans blue extravasation was measured as previously described [24] to determine BBB permeability. After 24 h of SAH, the left jugular veins of rats were injected with Evans blue dye (2% in saline, 4 ml/kg; Sigma-Aldrich). The procedure lasted for more than 2 min. The dye was allowed to circulate for 1 h. To clear the intravascular dye, the rats were anesthetized and a transcardial perfusion with PBS was performed. The brains were homogenized in PBS and centrifuged at 14,000 ×g for 30 min at 4°C. Next, the supernatant was mixed with the same volume of trichloroacetic acid, incubated for 1 h at 25°C, and centrifuged at 14,000 ×g for 30 min at 4°C. The absorbance of the supernatant at 620 nm was determined using a spectrophotometer (Thermo Fisher Scientific), and BBB permeability was analyzed.

Western blotting

Total protein in brain samples was extracted using a protein extraction kit (Beyotime). A bicinchoninic acid kit (Beyotime) was used to determine protein concentration. Next, the proteins (50 μg) were isolated using 10%–12% sodium dodecyl sulfate–polyacrylamide gel electrophoresis and loaded on a nitrocellulose membrane (Millipore). The membrane was blocked with 5% nonfat milk and incubated overnight at 4°C with primary antibodies against zonula occludens-1 (ZO-1; sc-33725, 1:1,000, Santa Cruz Biotechnology), Occludin (ab167161, 1:1,000, Abcam), Claudin-5 (35-2500, 1:1,000, Thermo Fisher Scientific), Sirt1 (ab110304, 1:1,000), Nrf2 (ab313825, 1:500), HO-1 (ab305290, 1:1,000), and GAPDH (loading control, ab9485, 1:2,500, Abcam). Next, the membranes were washed and incubated with secondary antibodies. Enhanced chemiluminescence reagent (Millipore) was used to visualize the protein bands, and the intensity of the bands was quantified using ImageJ software (National Institutes of Health). The uncropped images of bands are provided in Supplementary Fig. 1, 2.

Statistical analyses

Data were processed using GraphPad Prism 8 (GraphPad Software) and are presented as mean ± standard deviation. Shapiro–Wilk test was used to assess whether the means of the variables were the same or different. One-way analysis of variance followed by Tukey’s post-hoc analysis were used for comparisons among the 4 groups. p < 0.05 was considered statistically significant. The detailed numerical results of the statistical analysis are provided in Supplementary Table 1.

RESULTS

Chemical structure of shikonin and experimental design

The chemical structure of shikonin is presented in Fig. 1A and the design idea is shown in Fig. 1B. Sprague–Dawley rats were used to establish an animal model of SAH and were primarily assigned into the following 2 groups: a sham-operated group and an SAH-induction group. After the establishment of the rat model of SAH, DMSO or shikonin was administered to rats in the sham + DMSO, sham + shikonin, SAH + DMSO, and SAH + shikonin groups. SAH grade and neurological score were determined after 24 h of SAH induction. Next, western blotting was performed and levels of the biochemical markers were determined.

Shikonin alleviates neurological deficits and brain edema in rats after SAH

SAH can cause neurologic complications, including brain edema and neurological deficits. Therefore, behavior tests were conducted to evaluate the neurological functions of rats. In the beam balance test, rats in the SAH + DMSO group had a low score of 1 or even 0 with significantly reduced neurological behavior scores compared with those in the sham + DMSO group (Fig. 2A). According to the modified Garcia scoring system, model rats showed obvious neurological deficits versus those in the sham-operated group (Fig. 2B). The scores of rats in the sham + shikonin group were similar to those in the sham + DMSO group, indicating that shikonin did not cause brain damage to rats (Fig. 2A, B). Shikonin administration prominently alleviated the neurological functions of model rats, as evidenced by the increased scores of rats in the SAH + shikonin group compared with those in the SAH + DMSO group (Fig. 2A, B). Additionally, the brain water content increased markedly after SAH modeling, which was effectively alleviated after shikonin treatment (Fig. 2C). Sixty rats were used for this study; 7 rats were excluded as they died within 24 h of inducing SAH. The mortality rate of rats with SAH was 17.5% (7/40), and none of the rats died in the sham groups (0/20) (Table 1). The brain samples of rats from the 4 groups are shown in Fig. 2D. Subarachnoid blood clots can be seen on the ipsilateral side, within the surface of the brain stem and around the circle of Willis after SAH modeling (Fig. 2D). These changes induced by SAH were mitigated after shikonin treatment (Fig. 2D). The SAH grade showed a significant increase in the SAH + DMSO group compared with that in the sham + DMSO group (Fig. 2E). There was no notable difference in SAH grade between the SAH + DMSO group and the SAH + shikonin group (Fig. 2E). Overall, shikonin could alleviate neurological deficits in rats with SAH but hardly change the extent of SAH.

Shikonin attenuates oxidative stress in the brain cortex of rats after SAH

Changes in the levels of oxidative stress markers in the brain tissues of rats were measured to determine the effect of shikonin on oxidative stress after SAH. Shikonin treatment led to a minimal change in MDA and GSH levels and SOD production in rats in the sham group (Fig. 3A-C). On the other hand, SAH modeling led to a marked elevation in MDA content while decreasing GSH content and SOD production. Shikonin treatment exerted a rescuing effect by reversing the increase in MDA content and the reduction of GSH and SOD levels (Fig. 3A-C). Furthermore, ROS content was markedly elevated in response to SAH induction, and this trend was markedly counteracted after shikonin administration (Fig. 3D). These findings collectively indicated that shikonin could attenuate oxidative stress after SAH by improving the dysfunction of oxidative indicators and reducing ROS levels.

Shikonin attenuates BBB permeability and upregulates tight junction proteins after SAH

The effect of shikonin on BBB permeability after SAH was determined based on Evans blue extravasation. Extravasation was significantly promoted after 24 h of SAH versus that in the sham + DMSO group (9.78 ± 1.1 versus 3.95 ± 0.49) and the alteration in rats with SAH was reversed after shikonin treatment (6.22 ± 0.71) (Fig. 4A). The protein levels of the tight junction proteins of the BBB (ZO-1, Occludin, and Claudin5) in brain tissues were quantified using western blotting. ZO-1, Occludin, and Claudin-5 levels were noticeably decreased in the SAH + DMSO group, whereas shikonin treatment reversed this change (Fig. 4B, C). These results suggested that shikonin attenuated BBB permeability after SAH by reducing the loss of tight junctions.

Shikonin activates Sirt1/Nrf2/HO-1 signaling in the brain tissues of rats with SAH

Western blotting was performed to measure the protein levels of Sirt1, Nrf2, and HO-1 in the cerebral cortex of rats to determine the mechanism related to the protective role of shikonin against SAHinduced brain injury. SAH induction prominently reduced Sirt1, Nrf2, and HO-1 levels compared with those in sham-operated rats (p < 0.001, Fig. 5A-C). Shikonin treatment significantly rescued the reduction in Sirt1, Nrf2, and HO-1 levels in the brain tissues of rats with SAH (p < 0.001, Fig. 5A-C). These findings indicated that shikonin activated Sirt1/Nrf2/HO-1 signaling in rats with SAH.

DISCUSSION

EBI is a main determinant of the prognosis and survival of SAH [25]. The degree of EBI is estimated based on the extent of neurological deficits, brain edema, BBB injury, and oxidative stress response [26,27]. In this study, an intraperitoneal injection of shikonin (25 mg/kg) in rats was found to be effective in alleviating EBI following SAH. Brain edema and damage to the BBB occurred in rats 24 h after SAH along with a prominent decrease in neurobehavior score, suggesting EBI in the acute phase after SAH. Shikonin treatment markedly alleviated neurological dysfunction, brain edema, and BBB injury in rats, suggesting the neuroprotective role of shikonin against EBI following SAH. Shikonin exerts antioxidant, anti-inflammatory, and antitumor effects [28,29]. A previous study revealed that shikonin could improve neurological deficits, ameliorate BBB permeability, and reduce infarct volume and edema in mice 24 h after a transient ischemic stroke [30], which is consistent with our findings. Compared with the previous study by Wang et al. [30], the novelty of our study lies in that the antioxidant potential of shikonin in brain tissues was experimentally validated and the related mechanism of SIRT1/Nrf2/HO-1 signaling was elucidated. Shikonin alleviates irradiated brain injury in mice by ameliorating memory deficits and reducing oxidative stress [31], holding the view that shikonin can treat brain injury. This finding is in line with the conclusions of the present study. Moreover, shikonin exerts a protective effect in rabbits with SAH by repressing the inflammatory response at 24 h and oxidative stress at 72 h [32]. These studies conducted using different animal models further support our findings.
Numerous studies have confirmed that oxidative stress is closely correlated to EBI [33]. Oxidative stress is characterized by excessive ROS production, lipid peroxidation, and protein inactivation [34-36]. The increase in ROS in EBI after SAH results in neuronal necrosis and BBB destruction, which can result in brain edema and finally lead to neurological exacerbation [37,38]. Antioxidant enzymes such as SOD and GSH can effectively eliminate ROS [39]. MDA is generated by the reaction of lipids with oxygen free radicals [40]. In this study, after 24 h of SAH, MDA concentration increased while GSH and SOD contents decreased in the brain tissues of rats. However, shikonin treatment significantly reduced excessive MDA production and increased GSH and SOD contents. Consistently, shikonin has been reported to increase catalase and SOD activities and the ratio of GSH/oxidized GSH in the brain of mice [31]. The antioxidant mechanism of shikonin in SAH was not explored in our study. Previous studies suggest that shikonin may exert its neuroprotective and antioxidant effects by activating the Nrf2/ARE pathway and inhibiting the MAPK and NF-κB pathways [12,41,42].
SIRT1 is involved in various biological functions in cells [43,44]. A recent study revealed that the activation of SIRT1 contributes to the suppression of neuroinflammation, neuronal cell death, and ferroptosis following SAH [45]. In addition, SIRT1 plays a crucial role in oxidative stress by regulating Nrf2 activation [46-48]. Consistent with these previous findings, our study revealed that shikonin activated the SIRT1/Nrf2/HO-1 pathway in the brain tissues of rats after SAH, suggesting that shikonin may exert a protective role in SAH-induced brain injury by activating SIRT1/Nrf2/HO-1 signaling. SIRT1 inhibitors such as EX527 [43,44] can be evaluated in future studies to verify the role of shikonin in brain injury after SAH. As reported by Ma et al. [49], EX527 successfully inactivated SIRT1 in human renal tubular epithelial cells and thereby reversed the inhibitory effects of sweroside on high glucose-induced cell injury. Therefore, further experiments can be carried out using the specific SIRT1 inhibitor (EX527) to investigate whether inhibition of SIRT1 can abolish the protective effect of shikonin on SAH-induced brain injury.
This study demonstrated that shikonin attenuated BBB injury and oxidative stress in rats after SAH, indicating its therapeutic potential in alleviating SAH. A limitation of this study is the lack of further research on the molecular mechanisms underlying the neuroprotective role of shikonin, which will be studied subsequently.

SUPPLEMENTARY MATERIALS

Supplementary data including two figures and one table can be found with this article online at https://doi.org/10.4196/kjpp.24.182

ACKNOWLEDGEMENTS

None.

Notes

FUNDING

None to declare.

CONFLICTS OF INTEREST

The authors declare no conflicts of interest.

REFERENCES

1. Alwageed HS. 2022; Detection of subarachnoid hemorrhage in computed tomography using association rules mining. Comput Intell Neurosci. 2022:1133819. DOI: 10.1155/2022/1133819. PMID: 36093508. PMCID: PMC9451997.
crossref
2. Goldberg J, Schoeni D, Mordasini P, Z'Graggen W, Gralla J, Raabe A, Beck J, Fung C. 2018; Survival and outcome after poor-grade aneurysmal subarachnoid hemorrhage in elderly patients. Stroke. 49:2883–2889. DOI: 10.1161/STROKEAHA.118.022869. PMID: 30571422.
crossref
3. Lauzier DC, Jayaraman K, Yuan JY, Diwan D, Vellimana AK, Osbun JW, Chatterjee AR, Athiraman U, Dhar R, Zipfel GJ. 2023; Early brain injury after subarachnoid hemorrhage: incidence and mechanisms. Stroke. 54:1426–1440. DOI: 10.1161/STROKEAHA.122.040072. PMID: 36866673. PMCID: PMC10243167.
crossref
4. Rass V, Helbok R. 2019; Early brain injury after poor-grade subarachnoid hemorrhage. Curr Neurol Neurosci Rep. 19:78. DOI: 10.1007/s11910-019-0990-3. PMID: 31468197. PMCID: PMC6715808.
crossref
5. Li Y, Wu P, Bihl JC, Shi H. 2020; Underlying mechanisms and potential therapeutic molecular targets in blood-brain barrier disruption after subarachnoid hemorrhage. Curr Neuropharmacol. 18:1168–1179. DOI: 10.2174/1570159X18666200106154203. PMID: 31903882. PMCID: PMC7770641.
crossref
6. Sehba FA, Hou J, Pluta RM, Zhang JH. 2012; The importance of early brain injury after subarachnoid hemorrhage. Prog Neurobiol. 97:14–37. DOI: 10.1016/j.pneurobio.2012.02.003. PMID: 22414893. PMCID: PMC3327829.
crossref
7. Yuan L, Wang Y, Chen Y, Chen X, Li S, Liu X. 2023; Shikonin inhibits immune checkpoint PD-L1 expression on macrophage in sepsis by modulating PKM2. Int Immunopharmacol. 121:110401. DOI: 10.1016/j.intimp.2023.110401. PMID: 37302371.
crossref
8. Guo C, He J, Song X, Tan L, Wang M, Jiang P, Li Y, Cao Z, Peng C. 2019; Pharmacological properties and derivatives of shikonin-A review in recent years. Pharmacol Res. 149:104463. DOI: 10.1016/j.phrs.2019.104463. PMID: 31553936.
crossref
9. Sun Q, Gong T, Liu M, Ren S, Yang H, Zeng S, Zhao H, Chen L, Ming T, Meng X, Xu H. 2022; Shikonin, a naphthalene ingredient: therapeutic actions, pharmacokinetics, toxicology, clinical trials and pharmaceutical researches. Phytomedicine. 94:153805. DOI: 10.1016/j.phymed.2021.153805. PMID: 34749177.
crossref
10. Mu Z, Guo J, Zhang D, Xu Y, Zhou M, Guo Y, Hou Y, Gao X, Han X, Geng L. 2021; Therapeutic effects of shikonin on skin diseases: a review. Am J Chin Med. 49:1871–1895. DOI: 10.1142/S0192415X21500889. PMID: 34961421.
crossref
11. Guo W, Wang X, Sun C, Wang J, Wang T. 2023; Shikonin ameliorates salivary gland damage and inflammation in a mouse model of Sjögren's syndrome by modulating MAPK signaling pathway. Korean J Physiol Pharmacol. 27:357–364. DOI: 10.4196/kjpp.2023.27.4.357. PMID: 37386833. PMCID: PMC10316193.
crossref
12. Guo L, Li Y, Li W, Qiu J, Du J, Wang L, Zhang T. 2022; Shikonin ameliorates oxidative stress and neuroinflammation via the Akt/ERK/JNK/NF-κB signalling pathways in a model of Parkinson's disease. Clin Exp Pharmacol Physiol. 49:1221–1231. DOI: 10.1111/1440-1681.13709. PMID: 36054693.
crossref
13. Nie S, Zhao J, Huang Y, Zhou C, Gong F, Zhao F. 2021; Study on the effect of shikonin on CD36 expression and phagocytic ability of microglia in the isolated cerebral haemorrhage model. Folia Neuropathol. 59:198–204. DOI: 10.5114/fn.2021.107110. PMID: 34284547.
crossref
14. Mahmoud AM, Abd El-Ghafar OAM, Alzoghaibi MA, Hassanein EHM. 2021; Agomelatine prevents gentamicin nephrotoxicity by attenuating oxidative stress and TLR-4 signaling, and upregulating PPARγ and SIRT1. Life Sci. 278:119600. DOI: 10.1016/j.lfs.2021.119600. PMID: 33984362.
crossref
15. Arab HH, Ashour AM, Eid AH, Arafa EA, Al Khabbaz HJ, Abd El-Aal SA. 2022; Targeting oxidative stress, apoptosis, and autophagy by galangin mitigates cadmium-induced renal damage: role of SIRT1/Nrf2 and AMPK/mTOR pathways. Life Sci. 291:120300. DOI: 10.1016/j.lfs.2021.120300. PMID: 34999115.
crossref
16. Balaha MF, Alamer AA, Eisa AA, Aljohani HM. 2023; Shikonin alleviates gentamicin-induced renal injury in rats by targeting renal endocytosis, SIRT1/Nrf2/HO-1, TLR-4/NF-κB/MAPK, and PI3K/Akt cascades. Antibiotics (Basel). 12:826. DOI: 10.3390/antibiotics12050826. PMID: 37237729. PMCID: PMC10215741.
crossref
17. Lin F, Li R, Tu WJ, Chen Y, Wang K, Chen X, Zhao J. 2021; An update on antioxidative stress therapy research for early brain injury after subarachnoid hemorrhage. Front Aging Neurosci. 13:772036. DOI: 10.3389/fnagi.2021.772036. PMID: 34938172. PMCID: PMC8686680.
crossref
18. Yuan B, Zhao XD, Shen JD, Chen SJ, Huang HY, Zhou XM, Han YL, Zhou LJ, Lu XJ, Wu Q. 2022; Activation of SIRT1 alleviates ferroptosis in the early brain injury after subarachnoid hemorrhage. Oxid Med Cell Longev. 2022:9069825. DOI: 10.1155/2022/9069825. PMID: 35855863. PMCID: PMC9288286.
crossref
19. Wang Y, Pan XF, Liu GD, Liu ZH, Zhang C, Chen T, Wang YH. 2021; FGF-2 suppresses neuronal autophagy by regulating the PI3K/Akt pathway in subarachnoid hemorrhage. Brain Res Bull. 173:132–140. DOI: 10.1016/j.brainresbull.2021.05.017. PMID: 34023434.
crossref
20. Shao J, Meng Y, Yuan K, Wu Q, Zhu S, Li Y, Wu P, Zheng J, Shi H. 2023; RU.521 mitigates subarachnoid hemorrhage-induced brain injury via regulating microglial polarization and neuroinflammation mediated by the cGAS/STING/NF-κB pathway. Cell Commun Signal. 21:264. Erratum in: Cell Commun Signal. 2024;22:390. DOI: 10.1186/s12964-023-01274-2. PMID: 37770901. PMCID: PMC10537158.
crossref
21. Zhang T, Xu S, Wu P, Zhou K, Wu L, Xie Z, Xu W, Luo X, Li P, Ocak U, Ocak PE, Travis ZD, Tang J, Shi H, Zhang JH. 2019; Mitoquinone attenuates blood-brain barrier disruption through Nrf2/PHB2/OPA1 pathway after subarachnoid hemorrhage in rats. Exp Neurol. 317:1–9. DOI: 10.1016/j.expneurol.2019.02.009. PMID: 30779914.
crossref
22. Jia Y, Li Z, Wang T, Fan M, Song J, Lv P, Jin W. 2021; Shikonin attenuates chronic cerebral hypoperfusion-induced cognitive impairment by inhibiting apoptosis via PTEN/Akt/CREB/BDNF signaling. Evid Based Complement Alternat Med. 2021:5564246. DOI: 10.1155/2021/5564246. PMID: 34211568. PMCID: PMC8205575.
crossref
23. Shi SS, Zhang HB, Wang CH, Yang WZ, Liang RS, Chen Y, Tu XK. 2015; Propofol attenuates early brain injury after subarachnoid hemorrhage in rats. J Mol Neurosci. 57:538–545. DOI: 10.1007/s12031-015-0634-2. PMID: 26342279.
crossref
24. Zhu C, Wang D, Chang C, Liu A, Zhou J, Yang T, Jiang Y, Li X, Jiang W. 2024; Dexmedetomidine alleviates blood-brain barrier disruption in rats after cerebral ischemia-reperfusion by suppressing JNK and p38 MAPK signaling. Korean J Physiol Pharmacol. 28:239–252. DOI: 10.4196/kjpp.2024.28.3.239. PMID: 38682172. PMCID: PMC11058545.
crossref
25. Xu P, Tao C, Zhu Y, Wang G, Kong L, Li W, Li R, Li J, Zhang C, Wang L, Liu X, Sun W, Hu W. 2021; TAK1 mediates neuronal pyroptosis in early brain injury after subarachnoid hemorrhage. J Neuroinflammation. 18:188. DOI: 10.1186/s12974-021-02226-8. PMID: 34461942. PMCID: PMC8406585.
crossref
26. Xu W, Yan J, Ocak U, Lenahan C, Shao A, Tang J, Zhang J, Zhang JH. 2021; Melanocortin 1 receptor attenuates early brain injury following subarachnoid hemorrhage by controlling mitochondrial metabolism via AMPK/SIRT1/PGC-1α pathway in rats. Theranostics. 11:522–539. DOI: 10.7150/thno.49426. PMID: 33391490. PMCID: PMC7738864.
crossref
27. Zeng H, Chen H, Li M, Zhuang J, Peng Y, Zhou H, Xu C, Yu Q, Fu X, Cao S, Cai J, Yan F, Chen G. 2021; Autophagy protein NRBF2 attenuates endoplasmic reticulum stress-associated neuroinflammation and oxidative stress via promoting autophagosome maturation by interacting with Rab7 after SAH. J Neuroinflammation. 18:210. DOI: 10.1186/s12974-021-02270-4. PMID: 34530854. PMCID: PMC8447596.
crossref
28. Wang F, Yao X, Zhang Y, Tang J. 2019; Synthesis, biological function and evaluation of Shikonin in cancer therapy. Fitoterapia. 134:329–339. DOI: 10.1016/j.fitote.2019.03.005. PMID: 30858045.
crossref
29. Zhang Y, Zhang H, Wang M, Gao S, Hong L, Hou T, Zhang Y, Zhu Y, Qian F. 2021; Shikonin ameliorates lipoteichoic acidinduced acute lung injury via promotion of neutrophil apoptosis. Mol Med Rep. 23:133. DOI: 10.3892/mmr.2020.11772. PMID: 33313945.
crossref
30. Wang L, Li Z, Zhang X, Wang S, Zhu C, Miao J, Chen L, Cui L, Qiao H. 2014; Protective effect of shikonin in experimental ischemic stroke: attenuated TLR4, p-p38MAPK, NF-κB, TNF-α and MMP-9 expression, up-regulated claudin-5 expression, ameliorated BBB permeability. Neurochem Res. 39:97–106. DOI: 10.1007/s11064-013-1194-x. PMID: 24248858.
crossref
31. Gan L, Wang ZH, Zhang H, Zhou R, Sun C, Liu Y, Si J, Liu YY, Wang ZG. 2015; Protective effects of shikonin on brain injury induced by carbon ion beam irradiation in mice. Biomed Environ Sci. 28:148–151.
32. Shi X, Zhen L, Ding H, Chen J, Zhang S, Fu Y. 2019; Role of ATP-sensitive potassium channels and inflammatory response of basilar artery smooth muscle cells in subarachnoid hemorrhage of rabbit and immune-modulation by shikonin. Food Chem Toxicol. 134:110804. DOI: 10.1016/j.fct.2019.110804. PMID: 31505234.
crossref
33. Sies H. 2015; Oxidative stress: a concept in redox biology and medicine. Redox Biol. 4:180–183. DOI: 10.1016/j.redox.2015.01.002. PMID: 25588755. PMCID: PMC4309861.
crossref
34. Jones DP. 2008; Radical-free biology of oxidative stress. Am J Physiol Cell Physiol. 295:C849–C868. DOI: 10.1152/ajpcell.00283.2008. PMID: 18684987. PMCID: PMC2575825.
crossref
35. Pizzino G, Irrera N, Cucinotta M, Pallio G, Mannino F, Arcoraci V, Squadrito F, Altavilla D, Bitto A. 2017; Oxidative stress: harms and benefits for human health. Oxid Med Cell Longev. 2017:8416763. DOI: 10.1155/2017/8416763. PMID: 28819546. PMCID: PMC5551541.
crossref
36. Newsholme P, Cruzat VF, Keane KN, Carlessi R, de Bittencourt PI Jr. 2016; Molecular mechanisms of ROS production and oxidative stress in diabetes. Biochem J. 473:4527–4550. DOI: 10.1042/BCJ20160503C. PMID: 27941030.
crossref
37. Zhang Y, Lei Y, Yao X, Yi J, Feng G. 2021; Pinoresinol diglucoside alleviates ischemia/reperfusion-induced brain injury by modulating neuroinflammation and oxidative stress. Chem Biol Drug Des. 98:986–996. DOI: 10.1111/cbdd.13956. PMID: 34546621. PMCID: PMC9293043.
crossref
38. Han Y, Zhang T, Su J, Zhao Y, Chenchen , Wang , Li X. 2017; Apigenin attenuates oxidative stress and neuronal apoptosis in early brain injury following subarachnoid hemorrhage. J Clin Neurosci. 40:157–162. DOI: 10.1016/j.jocn.2017.03.003. PMID: 28342702.
crossref
39. Sies H. 1997; Oxidative stress: oxidants and antioxidants. Exp Physiol. 82:291–295. DOI: 10.1113/expphysiol.1997.sp004024. PMID: 9129943.
crossref
40. Tsikas D. 2017; Assessment of lipid peroxidation by measuring malondialdehyde (MDA) and relatives in biological samples: analytical and biological challenges. Anal Biochem. 524:13–30. DOI: 10.1016/j.ab.2016.10.021. PMID: 27789233.
crossref
41. Du H, Zhou X, Shi L, Xia M, Wang Y, Guo N, Hu H, Zhang P, Yang H, Zhu F, Teng Z, Liu C, Zhao M. 2022; Shikonin attenuates cochlear spiral ganglion neuron degeneration by activating Nrf2-ARE signaling pathway. Front Mol Neurosci. 15:829642. DOI: 10.3389/fnmol.2022.829642. PMID: 35283722. PMCID: PMC8908960.
crossref
42. Zhong J, Wang Z, Xie Q, Li T, Chen K, Zhu T, Tang Q, Shen C, Zhu J. 2020; Shikonin ameliorates D-galactose-induced oxidative stress and cognitive impairment in mice via the MAPK and nuclear factor-κB signaling pathway. Int Immunopharmacol. 83:106491. DOI: 10.1016/j.intimp.2020.106491. PMID: 32279045.
crossref
43. Xia DY, Yuan JL, Jiang XC, Qi M, Lai NS, Wu LY, Zhang XS. 2021; SIRT1 promotes M2 microglia polarization via reducing ROS-mediated NLRP3 inflammasome signaling after subarachnoid hemorrhage. Front Immunol. 12:770744. DOI: 10.3389/fimmu.2021.770744. PMID: 34899720. PMCID: PMC8653696.
crossref
44. Zhang Z, Fang J, Zhou J, Ding F, Zhou G, Zhao X, Zhuang Z, Lu Y. 2022; Pterostilbene attenuates subarachnoid hemorrhage-induced brain injury through the SIRT1-dependent Nrf2 signaling pathway. Oxid Med Cell Longev. 2022:3550204. DOI: 10.1155/2022/3550204. PMID: 36506933. PMCID: PMC9729048.
crossref
45. Chen L, Xu H, Zhang C, He J, Wang Y. 2024; Semaglutide alleviates early brain injury following subarachnoid hemorrhage by suppressing ferroptosis and neuroinflammation via SIRT1 pathway. Am J Transl Res. 16:1102–1117. DOI: 10.62347/IZGJ1332. PMID: 38715815. PMCID: PMC11070367.
crossref
46. Mao H, Wang L, Xiong Y, Jiang G, Liu X. 2022; Fucoxanthin attenuates oxidative damage by activating the Sirt1/Nrf2/HO-1 signaling pathway to protect the kidney from ischemia-reperfusion injury. Oxid Med Cell Longev. 2022:7444430. DOI: 10.1155/2022/7444430. PMID: 35126819. PMCID: PMC8816562.
crossref
47. Ma W, Guo W, Shang F, Li Y, Li W, Liu J, Ma C, Teng J. 2020; Bakuchiol alleviates hyperglycemia-induced diabetic cardiomyopathy by reducing myocardial oxidative stress via activating the SIRT1/Nrf2 signaling pathway. Oxid Med Cell Longev. 2020:3732718. DOI: 10.1155/2020/3732718. PMID: 33062139. PMCID: PMC7545423.
crossref
48. Zhang X, Wu Q, Lu Y, Wan J, Dai H, Zhou X, Lv S, Chen X, Zhang X, Hang C, Wang J. 2018; Cerebroprotection by salvianolic acid B after experimental subarachnoid hemorrhage occurs via Nrf2- and SIRT1-dependent pathways. Free Radic Biol Med. 124:504–516. DOI: 10.1016/j.freeradbiomed.2018.06.035. PMID: 29966698. PMCID: PMC6286712.
crossref
49. Ma X, Guo Z, Zhao W, Chen L. 2023; Sweroside plays a role in mitigating high glucose-induced damage in human renal tubular epithelial HK-2 cells by regulating the SIRT1/NF-κB signaling pathway. Korean J Physiol Pharmacol. 27:533–540. DOI: 10.4196/kjpp.2023.27.6.533. PMID: 37884285. PMCID: PMC10613573.
crossref

Fig. 1

Chemical structure of shikonin and experimental design.

(A) Chemical structure of shikonin. (B) Experimental design. SAH, subarachnoid hemorrhage; DMSO, dimethyl sulfoxide.
kjpp-29-3-283-f1.tif
Fig. 2

Shikonin alleviates neurological deficits and brain edema in rats after SAH.

(A, B) Beam balance test (A) and modified Garcia scoring system (B) were used to determine the neurological function of rats in the sham + DMSO group, sham + shikonin group, SAH + DMSO group, and SAH + shikonin group. (C) Brain water content of rats in the 4 groups was determined to evaluate the extent of brain edema. (D) Representative image of rat brains from the 4 groups. (E) SAH grade in the 4 groups. Values are shown as mean ± standard deviation. SAH, subarachnoid hemorrhage; DMSO, dimethyl sulfoxide. ***p < 0.001.
kjpp-29-3-283-f2.tif
Fig. 3

Shikonin attenuates oxidative stress in the brain cortex of rats after SAH.

(A-C) Contents of MDA, GSH, and SOD in the brain cortex of rats were determined using the corresponding assay kits in the sham + DMSO group, sham + shikonin group, SAH + DMSO group, and SAH + shikonin group. (D) ROS content in the brain tissues of rats from each group was determined using a commercial assay kit. Values are shown as mean ± standard deviation. SAH, subarachnoid hemorrhage; MDA, malondialdehyde; GSH, glutathione; SOD, superoxide dismutase; DMSO, dimethyl sulfoxide; ROS, reactive oxygen species. ***p < 0.001.
kjpp-29-3-283-f3.tif
Fig. 4

Shikonin attenuates BBB permeability and upregulates tight junction proteins after SAH.

(A) Evans blue staining assay was used to determine the BBB permeability of rats in the sham + DMSO group, sham + shikonin group, SAH + DMSO group, and SAH + shikonin group. (B, C) Protein levels of tight junctions (ZO-1, Occludin, and Claudin5) in the 4 groups were determined using Western blotting. Values are shown as mean ± standard deviation. BBB, blood–brain barrier; SAH, subarachnoid hemorrhage; DMSO, dimethyl sulfoxide; ZO-1, zonula occludens-1. ***p < 0.001.
kjpp-29-3-283-f4.tif
Fig. 5

Shikonin activates the Sirt1/Nrf2/HO-1 signaling in the brain tissues of rats with SAH.

(A) Western blotting to determine the protein levels of Sirt1, Nrf2, and HO-1 in the cerebral cortex of rats from the sham + DMSO, sham + shikonin, SAH + DMSO, and SAH + shikonin groups. (B-D) Quantification of the relative protein levels of Sirt1, Nrf2, and HO-1 with normalization to GAPDH. Values are shown as mean ± standard deviation. SAH, subarachnoid hemorrhage; DMSO, dimethyl sulfoxide. ***p < 0.001.
kjpp-29-3-283-f5.tif
Table 1
Animal usage and mortality
Groups Mortality
Sham + DMSO 0% (0/10)
Sham + Shikonin 0% (0/10)
SAH + DMSO 25% (5/20)
SAH + Shikonin 10% (2/20)

SAH, subarachnoid hemorrhage; DMSO, dimethyl sulfoxide.

TOOLS
Similar articles