Protective effect 3,4-dihydroxyphenylethanol in subarachnoid hemorrhage provoked oxidative neuropathy

  • Authors:
    • Yu‑Wen Zhong
    • Juan Wu
    • Hua‑Long Hu
    • Wei‑Xin Li
    • Yong Zhong
  • View Affiliations

  • Published online on: July 14, 2016     https://doi.org/10.3892/etm.2016.3526
  • Pages: 1908-1914
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Clinical studies have indicated that early brain injury (EBI) following subarachnoid hemorrhage (SAH) is associated with fatal outcomes. Oxidative stress and brain edema are the characteristic pathological events in occurrence EBI following SAH. The present study aimed to examine the effect of 3,4‑dihydroxyphenylethanol (DOPET) against SAH‑induced EBI, and to demonstrate whether the effect is associated with its potent free radical scavenging property. SAH was induced in rats using an endovascular perforation technique, and 24 h later the rats displayed diminished neurological scores and brain edema. Furthermore, elevated malondialdehyde (an index of lipid peroxidation) and depleted levels of antioxidants were observed in the rat cerebral cortex tissue. Quantitative polymerase chain reaction analysis indicated the upregulated mRNA expression of the apoptotic markers caspase‑3 and ‑9 in the cerebral cortex. Furthermore, the protein expression levels of the proinflammatory cytokines tumor necrosis factor‑α, interleukin (IL)‑1β and IL‑6 were significantly upregulated in SAH‑induced rats. By constrast, treatment with DOPET significantly attenuated EBI by reducing brain edema, elevation of antioxidant status, inhibition of apoptosis and inflammation. In this context, DOPET may be a potent agent in the treatment of EBI following SAH, as a result of its free radical scavenging capacity.

Introduction

Globally, subarachnoid hemorrhage (SAH) is a prominent pathological occurrence which is involved in the etiology of 5–7% of all strokes cases that involve high mortality and functional loss (1). Despite advances in medical treatment and diagnosis, the mortality and morbidity rates of SAH have not been decreased significantly (2). Furthermore, the outcome of treatment in SAH patients remains poor, with a mortality rate of >50% and high morbidity among the survivors (35). Due to its complex pathology, researchers have deepened their interest in understanding the mechanisms underlying SAH at molecular level (6).

In the event of SAH, two clinical scenarios have been addressed primarily; vasospasm and early brain injury (EBI) (7). Arterial narrowing during SAH elicits fatal complications such as cerebral ischemia, and hence targeting the vasospasm has been a key target in the treatment of SAH among neurosurgeons in the past years (4). However, little success has been achieved in improving outcome following SAH (8,9). Additionally, accumulating studies have indicated that the administration of clazosentan in SAH patients does not improve patient outcome, despite reducing vasoconstriction (8,10). Therefore, studies have investigated the involvement of cardinal factors such as ischemia, disruption of the blood brain barrier (BBB), inflammatory reactions and cortical spreading depression in the early stages following SAH (11,12).

Previous studies have suggested that the EBI period (24–72 h) following SAH elicits a series of events that may lead to poor prognosis (13,14). Furthermore, prior reports indicate that oxidative stress and brain edema are involved in EBI after SAH (15,16). Furthermore, reactive oxygen species (ROS) and reactive nitrogen species have been implicated in the occurrence of brain injury after SAH (17).

During EBI, brain edema occurs due to the disruption of the BBB (18,19) more than as a consequence of vasospasm (16). Thus, in clinical diagnosis, global edema has been proposed as a sole independent risk factor for fatal complications after SAH (16). Furthermore, noxious oxidative assault has been closely associated with brain edema (19). Therapeutic approaches have focussed on inhibiting ROS-induced apoptosis and inflammation as reasonable choices for the treatment of brain injury (20). Among an array of therapeutic interventions, a potential approach to boost or combat endogenous defense against oxidative stress is through dietary or pharmacological intake of antioxidants (21).

3,4-Dihydroxyphenylethanol (DOPET) is a phenol extracted from olive oil and grape juice, and is an endogenous metabolite of dopamine (22). DOPET has a good safety margin (23), and has been suggested to exert neuroprotective (24,25), cardioprotective (26), uroprotective (27), renoprotective (28), hepatoprotective (29), anti-diabetic and antiobesity (30), anti-osteoporotic (31), anti-inflammatory (32), anti-atherosclerotic (33), anticarcinogenic (34) and anti-virus effects (35) in animal studies. Notably, earlier reports indicate the neuroprotective potential of DOPET in rats and in vitro (3638). On the basis of these preliminary findings, we investigated whether that DOPET may be an effective molecule in the in the mitigation of SAH in a rat model.

Materials and methods

Animals

A total of 21 male Sprague-Dawley rats (weight, 170–200 g; age, 9 weeks) were obtained from the animal facility of Tongcheng People's Hospital (Xianning, China). The animals were maintained under standard laboratory conditions of relative humidity (55±5%), temperature (25±2°C) and light (12-h light/dark). The rats were fed standard diet pellets and water was provided ad libitum.

Animal grouping

Sprague-Dawley rats were divided into three groups (n=7 per group): Sham-operated rats (sham group); SAH rats treated with saline (SAH group); and SAH rats treated with DOPET (10 mg/kg) orally (SAH + DOPET group).

Administration of DOPET

DOPET was purchased from Sigma-Aldrich (St. Louis, MO, USA) and dissolved in 0.9% saline at a concentration of 3%. In the SAH + DOPET rats, DOPET (10 mg/kg) was injected intraperitoneally at 5 min and 6 h after SAH induction. In the SAH group, the rats underwent SAH-induction and were treated with an equal volume of 0.9% saline. No treatment was applied in the sham-operated animals.

Induction of SAH

SAH in rats was induced using an endovascular perforation technique, as described previously (39). Briefly, in anesthetized rats (5% isoflurane; Sigma-Aldrich) the left carotid artery and its branches were exposed and transected distally and reflected caudally in line with the internal carotid artery (ICA). Then, a blunted 4-0 monofilament nylon suture (Ethicon, San Angelo, TX, USA) was placed in the external carotid artery and advanced through the ICA until resistance was detected at 18–20 mm from the common carotid artery bifurcation. Next, the suture was advanced for ~3 mm to perforate the ICA near its intracranial bifurcation and removed after 15 sec.

Neurological test

The neurological evaluation was performed at 24 h after SAH surgery using the Garcia scoring method (40). In this evaluation, spontaneous activity, symmetry in the movement of four limbs, forepaw outstretching, climbing, body proprioception and response to vibrissae touch were assessed. These six tests were each scored from 0 to 3. Overall scores were graded as a minimum of 0 and the maximum as 18.

Brain water content

Rats were sacrificed by CO2 inhalation 24 h after SAH. The whole brain was removed and immediately weighed to obtain the wet weight, and then dried at 105°C for 24 h to obtain the dry weight. The brain water content was calculated as: [(Wet weight-dry weight)/wet weight] × 100% (41).

Tissue harvesting

Following the evaluation of neurological score, the rats (n=7) were anesthetized using 5% isoflurane and the brains were removed for biochemical analysis. The olfactory bulb, pons and medulla were discarded and the cerebral cortex was dissected, weighed and chilled using liquid nitrogen until homogenization. These procedures lasted up to 3 min. The cerebral cortex was homogenized in 10 volumes (1:10 w/v) of cold saline. Brain samples were homogenized and centrifuged at 4,000 × g at 4°C for 10 min. Supernatant aliquots were used to assay various biochemical parameters.

Estimation of lipid peroxidation and oxidative stress

The activities of malondialdehyde (MDA; A003-1), glutathione (GSH; A006), glutathione peroxidase (GPx; A007) and superoxide dismutase (SOD; A001-1) in the cerebral cortex homogenate were measured respectively using commercial kits (Nanjing Jiancheng Bioengineering Institute, Nanjing, China), according to the manufacturer's instructions. Briefly, lipid peroxidation was estimated using the level of MDA (ε=155 mM−1cm−1), which was determined spectrophotometrically at A532. A yellow complex is produced during the reaction between 5,5′-dithio-bis-(2-nitrobenzoic acid) and a sulfhydryl compound. Through spectrophotometry, GSH levels were detected. Activity of GPx was calculated by the reduction of GSH. The color of 5-thio-dinitrobenzoic acid anion produced by the reaction between GSH and 5,5′-dithio-bis-(2-nitrobenzoic acid) is yellow and the absorbance is measured at 412 nm via spectrophotometry. The method of SOD determination involves generation of superoxide radical by photoreduction of riboflavin and its detection by nitrite formation from hydroxylamine hydrochloride at 543 nm. One unit of SOD activity was defined as the amount of enzyme capable of inhibiting 50% of nitrite formation under assay conditions. All standards and samples were run in duplicate. Tissue protein concentrations were determined using a BCA Protein Assay kit (Bio-Rad Laboratories, Inc., Hercules, CA, USA).

Reverse transcription quantitative polymerase chain reaction

Total RNA was extracted from cerebral cortex tissue using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc., Carlsbad, CA, USA) according to manufacturer's instructions. A total of 10 µl RNA was reverse transcribed using Moloney murine leukemia virus RT (Thermo Fisher Scientific, Inc.) in a 30 µl reaction mixture. The resultant cDNA (20 ng) was amplified using an iCycler IQ real-time detection system (Bio-Rad Laboratories, Inc) using IQ Supermix with 0.5X SYBR-Green (Bio-Rad Laboratories, Inc.). β-actin served as an endogenous control. Rat-specific primers for caspase-3 and caspase-9 were synthesized by Shanghai Shine Gene Molecular Biotech, Inc., (Shanghai, China), and the sequences were as follows: Caspase-3, forward 5′-GGTATTGAGACAGACAGTGG-3′ and reverse 5′-CATGGGATCTGTTTCTTTGC-3′; caspase-9, forward 5′-ACAAGGCCTTCGACAGTG-3′ and reverse 5′-GTACCAGGAACCGCTCTT-3′; and β-actin, forward 5′-ATCTGGCACCACACCTTC-3′ and reverse 5′-AGCCAGGTCCAGACGCA-3′. Thermocycling conditions were as follows: Initial denaturation at 94°C for 2 min, followed by 35 cycles of denaturation at −95°C for 15 sec, annealing at −58°C for 45 sec and extension at −60°C for 30–45 sec, with final extension at 72°C for 5 min. mRNA expression levels were normalized to the β-actin internal reference gene and the relative expression levels were calculated using the 2−ΔΔCq method (42) and CFX Manager software (Bio-Rad Laboratories, Inc). Reactions were performed in triplicate.

Western blot analysis

Western blotting was performed as described previously (43). Briefly, the left basal cortical sample facing the blood clot was weighed, homogenized, and centrifuged at 1,000 × g for 10 min at 4°C. The resulting supernatants were further centrifuged. Samples were transferred to sterile tubes containing cold TCAAEB [acetone containing 10% (w/v) TCA and 0.07% mercaptoethanol], and the proteins were precipitated for 1 h at −20°C, followed by centrifugation at 18,900 × g for 15 min at 4°C. The supernatant was decanted, and the pellet was washed twice with chilled wash buffer (acetone containing 0.07% mercaptoethanol, 2 mM EDTA and EDTA-free proteinase inhibitor cocktail tablets (Roche Diagnostics GmbH, Mannheim, Germany), followed by the removal of the acetone. The pellet was subsequently solubilized in LB-TT [7 M urea, 2 M thiourea, 4% (w/v) CHAPS, 18 mM Tris-HCl (pH 8.0), 14 mM trizma base, EDTA-free proteinase inhibitor cocktail, 0.2% (v/v) Triton X-100 (R), containing 50 mM dithiothreitol]. The protein content was measured using a DC protein assay kit (Bio-Rad Laboratories, Inc.) prior to electrophoresis. An equal quantity of protein (60 µg) from each sample was resuspended in loading buffer (Bio-Rad Laboratories, Inc.), denatured at 95°C for 5 min, separated by 10–15% sodium dodecyl sulfate polyacrylamide gel electrophoresis, and transferred onto polyvinylidene fluoride membranes (both Bio-Rad Laboratories, Inc.). The membranes were blocked with non-fat dry milk buffer for 2 h and incubated overnight at 4°C with primary antibodies against interleukin (IL)-1β (cat. no. sc-7884; 1:500), IL-6 (cat. no. sc-13026; 1:800), tumor necrosis factor (TNF)-α (cat. no. sc-1351; 1:800) and β-actin (cat. no. sc-47778; 1:2,000; Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA). The membranes were processed with horseradish peroxidase-conjugated chicken anti-rabbit IgG secondary antibodies (1:500; Santa Cruz Biotechnology, Inc.) at room temperature for 3 h.

Statistic analysis

Data are presented as the mean ± standard error of the mean. SPSS, version 12.0 (SPSS, Inc., Chicago, IL, USA) was used for statistical analysis of the data. All data were subjected to one-way analysis of variance followed by the Tukey test for multiple comparisons. P<0.05 was considered to be statistically significant.

Results

Effect of SAH and DOPET on neurological score

The neurological score was significantly (P<0.05) decreased in the SAH group compared to the sham group. After DOPET treatment, neurological deficits were reduced compared to that of the SAH (P<0.05) (Fig. 1).

Effect of SAH and DOPET on brain water content

As shown in Fig. 2, brain water content was significantly (P<0.05) elevated in SAH group compared to the sham group at 24 h after SAH. Brain edema was attenuated significantly (P<0.05) reduced in the SAH + DOPET group compared with the SAH group.

Effect of SAH and DOPET on lipid peroxidation in brain cortex homogenate

Lipid peroxidation in the cerebral cortex was quantified by measuring MDA levels (Fig. 3). The level of MDA in the brain of SAH rats was significantly higher compared with in the sham rat group (P<0.05). The increase in lipid peroxidation indicates an elevated in vivo oxidative stress in the brain of SAH rats, which was significantly decreased by treatment with DOPET compared to SAH rats (P<0.05).

Effect of SAH and DOPET on oxidative stress

The concentration of ROS is determined by the balance between the rate of production and the rate of clearance by various antioxidant compounds and enzymes. In the present study, post SAH there was a significant (P<0.05) decline in the level of antioxidants (GSH, SOD, and GPx) when compared to the sham rats. Treatment with DOPET significantly (P<0.05) increased the level of antioxidant in brain through its anti lipid peroxidative effect (Fig. 4).

Effect of SAH and DOPET on caspase-3 and caspase-9 mRNA expression

In the experimental SAH model, the caspase-3 and caspase-9 mRNA expression levels in the cerebral cortex were significantly increased (P<0.05) when compared with the sham-operated rats. However, therapeutic intervention with DOPET downregulated the mRNA levels of caspase-3 and caspase-9 when compared with the SAH rats, and thus attenuated the apoptosis (Fig. 5).

Effect of SAH and DOPET on protein expression of proinflammatory cytokines

Western blot analysis was used to evaluate the protein expression levels of TNF-α, IL-6 and IL-1β. Compared with the sham group, levels of the three inflammatory cytokines were significantly increased 24 h after SAH in the SAH group (P<0.05), whereas DOPET administration significantly reduced the levels of TNF-α, IL-6 and IL-1β compared with the SAH group (P<0.05). These results show that administration of DOPET downregulates the cortical expressions of pro-inflammatory cytokines 24 h after SAH (Fig. 6).

Discussion

Oxidative stress is a biological event which emerges from the potent cellular oxidizing ability of abundant ROS or free radicals (44,45). Following SAH, increased generation of oxidative stress occurs and prior results suggest that oxidative stress is a prime mediator of brain injury (15). During SAH, clot derived hemoglobin (Hb) triggers free radicals, including O−2•, H2O2 and OH, which subsequently react. Auto-oxidation of Hb produces O−2• and dismutation of two O−2• forms H2O2, which is the source of highly reactive OH in the reaction catalyzed by ferric ion (46). Amongst these oxidants, OH is highly potent and attacks the nucleic acids, lipids and proteins to produce a marked cytotoxic effect (47,48). Thus, the generation of OH free radicals from extravasated Hb (49), loss of mitochondrial integrity (50) and depletion of endogenous antioxidant system (51) have been elsewhere reported in experimental or human SAH.

Lipid peroxidation (LPO) is a noxious biological event induced by the free radicals such as OH, ONOO and H2O2 resulting in structural alterations of membranes and functional impairment of cellular components. MDA, the end product of LPO, attacks the polyunsaturated fatty acids of the cell membrane and thus serves as an effective marker of free radical damage (52). Similarly, in the present study, elevated MDA levels were observed in the cortex of SAH rats, which is in corroboration with a previous report (53). Treatment with DOPET significantly mitigated the elevated MDA level. The anti-lipid peroxidative effect of DOPET may be due to its lipophilic and hydrophilic nature (54). The phenolic group may imbed in the membrane, acting as a chain-breaking inhibitor of lipid peroxidation (55).

Furthermore, the downregulation of antioxidant defense system may be crucially involved in the pathology of SAH (56). In the antioxidant defense mechanism, the primary protection is performed by SOD against oxidative stress and LPO (15). In the oxidative stress cascade, the superoxide radical is initially generated and converted into H2O2 and molecular oxygen by catalase or GPx (57). Thus, vital organs and tissues are more prone to oxidative stress attack, which may be due to reduced antioxidant levels (58). The non-enzymic antioxidant reduced GSH, terminates the vicious cycle of ROS by reacting with the single oxygen and hydroxide radical and thus prevents tissue damage (59). In the present investigation, SAH rats displayed diminished glutathione, SOD, GPx and levels in cortex tissue. However, treatment with DOPET restored the altered antioxidant status to normal which may be due to the scavenging of free radicals and inhibition of LPO (60).

Delayed global edema has been displayed as an independent predictor of mortality (16). Furthermore, post SAH provoked cerebral edema may prelude elevated intracranial pressure (ICP) and brain herniation, leading to irreversible brain damage or mortality (61). Clinically, brain edema are underscored as cytotoxic or vasogenic edema (62). The characteristic features of cytotoxic edema include swelling with intracellular fluid accumulation which resembles astrocyte swelling (63). In cases of vasogenic edema, disruption of BBB occurs which may lead to the accumulation of fluid surrounding the cells (64). Furthermore, studies suggest that the altered expression levels of aquaporins, BBB disruption, clot derived substances, secondary noxious events like elevated ICP and hypertension are actively involved in the progression of brain edema after SAH, and hypertension are involved in the pathogenesis of brain edema (65,66). Turbulence in the BBB permeability is a key event during the brain injury after SAH (66). Furthermore, in SAH patients with vasogenic edema, a direct noxious effect after BBB rupture have been proved clinically, as well as in experimental studies (67). Furthermore, the edema increases the brain volume and thus extends the elevated ICP after SAH (68). Consequently, there is an elevation in ICP, which further reduces cerebral blood flow, leading to increased ischemia (65). In the present study, it was found that the brain water content increased obviously after SAH and administration of DOPET abated brain edema significantly. Previous reports suggest that DOPET mitigates brain edema in ischemic rats by reducing of BBB permeability (69).

However, oxidative stress can induce changes of enzymes which are apoptosis-related, including p53, caspase-3 and caspase-9 (70). Caspase-9 is an essential protein involved in the breakdown of procaspase-3 to caspase-3 (71). During SAH, caspase-3 was overexpressed in the cortical neurons and the upregulation of caspase-3 led to the apoptosis of neural cells and brain edema (72). The present data showed that the expression of caspase-3 and caspase-9 increased significantly in the experimental SAH group, while these expression levels may be reversed by DOPET administration. These results suggest that DOPET could inhibit proapoptotic enzymes via its antioxidant activity and exertion of a neuroprotection effect.

In conclusion, DOPET treatment significantly attenuated the toxic manifestation of SAH by preserving BBB integrity, inhibition of lipid peroxidation and restoration of antioxidant levels. Furthermore, the mRNA expression levels of the apoptotic markers caspase-3 and caspase-9 and the protein expression of proinflammatory cytokines TNF-α, IL-6 and IL-1β were downregulated DOPET intervention. Further studies on DOPET are required to elucidate the neuroprotective mechanism involved in its protective effect against SAH trauma.

Acknowledgements

The present study was supported by Tongcheng People's Hospital (grant no. TCYY-20140402).

References

1 

Ansar S, Maddahi A and Edvinsson L: Inhibition of cerebrovascular raf activation attenuates cerebral blood flow and prevents upregulation of contractile receptors after subarachnoid hemorrhage. BMC Neurosci. 12:1072011. View Article : Google Scholar : PubMed/NCBI

2 

Lantigua H, Ortega-Gutierrez S, Schmidt JM, Lee K, Badjatia N, Agarwal S, Claassen J, Connolly ES and Stephan A: Mayercorresponding author. Subarachnoid hemorrhage: Who dies, and why? Crit Care. 19:3092015.

3 

Schievink WI, Riedinger M, Jhutty TK and Simon P: Racial disparities in subarachnoid hemorrhage mortality: Los Angeles county, california, 1985–1998. Neuroepidemiology. 23:299–305. 2004. View Article : Google Scholar : PubMed/NCBI

4 

Van Gijn J and Rinkel GJ: Subarachnoid haemorrhage: Diagnosis, causes and management. Brain. 124:249–278. 2001. View Article : Google Scholar : PubMed/NCBI

5 

Hop JW, Rinkel GJ, Algra A and van Gijn J: Changes in functional outcome and quality of life in patients and caregivers after aneurysmal subarachnoid hemorrhage. J Neurosurg. 95:957–963. 2001. View Article : Google Scholar : PubMed/NCBI

6 

Chen S, Feng H, Sherchan P, Klebe D, Zhao G, Sun X, Zhang J, Tang J and Zhang JH: Controversies and evolving new mechanisms in subarachnoid hemorrhage. Prog Neurobio. 115:64–91. 2014. View Article : Google Scholar

7 

Pluta RM, Hansen-Schwartz J, Dreier J, Vajkoczy P, Macdonald RL, Nishizawa S, Kasuya H, Wellman G, Keller E, Zauner A and Dorsch N: Cerebral vasospasm following subarachnoid hemorrhage: Time for a new world of thought. Neurol Res. 31:151–158. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Cahill J, Calvert JW and Zhang JH: Mechanisms of early brain injury after subarachnoid hemorrhage. J Cereb Blood Flow Metab. 26:1341–1353. 2006. View Article : Google Scholar : PubMed/NCBI

9 

Vajkoczy P, Meyer B, Weidauer S, Raabe A, Thome C, Ringel F, Breu V and Schmiedek P: Clazosentan (AXV-034343), a selective endothelin A receptor antagonist, in the prevention of cerebral vasospasm following severe aneurysmal subarachnoid hemorrhage: Results of a randomized, double-blind, placebo-controlled, multicenter phase IIa study. J Neurosurg. 103:9–17. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Macdonald RL, Kassell NF, Mayer S, Ruefenacht D, Schmiedek P, Weidauer S, Frey A, Roux S and Pasqualin A: CONSCIOUS-1 Investigators: Clazosentan to overcome neurological ischemia and infarction occurring after subarachnoid hemorrhage (CONSCIOUS-1): Randomized, double-blind, placebo-controlled phase 2 dose-finding trial. Stroke. 39:3015–3021. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Hansen-Schwartz J, Vajkoczy P, Macdonald RL, Pluta RM and Zhang JH: Cerebral vasospasm: Looking beyond vasoconstriction. Trends Pharmacol Sci. 28:252–256. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Rabinstein AA: Secondary brain injury after aneurysmal subarachnoid haemorrhage: More than vasospasm. Lancet Neurol. 10:593–595. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Broderick JP, Brott TG, Duldner JE, Tomsick T and Leach A: Initial and recurrent bleeding are the major causes of death following subarachnoid hemorrhage. Stroke. 25:1342–1347. 1994. View Article : Google Scholar : PubMed/NCBI

14 

Sehba FA and Bederson JB: Mechanisms of acute brain injury after subarachnoid hemorrhage. Neurol Res. 28:381–398. 2006. View Article : Google Scholar : PubMed/NCBI

15 

Gaetani P, Pasqualin A, Rodriguezy Baena R, Borasio E and Marzatico F: Oxidative stress in the human brain after subarachnoid hemorrhage. J Neurosurg. 89:748–754. 1998. View Article : Google Scholar : PubMed/NCBI

16 

Claassen J, Carhuapoma JR, Kreiter KT, Du EY, Connolly ES and Mayer SA: Global cerebral edema after subarachnoid hemorrhage: frequency, predictors and impact on outcome. Stroke. 33:1225–1232. 2002. View Article : Google Scholar : PubMed/NCBI

17 

Cahill J and Zhang JH: Subarachnoid hemorrhage: Is it time for a new direction? Stroke. 40(Suppl 3): S86–S87. 2009. View Article : Google Scholar : PubMed/NCBI

18 

László FA, Varga C and Dóczi T: Cerebral oedema after subarachnoid haemorrhage. Pathogenetic significance of vasopressin. Acta Neurochir (Wien). 133:122–133. 1995. View Article : Google Scholar : PubMed/NCBI

19 

Dóczi T, Joó F, Adám G, Bozóky B and Szerdahelyi P: Blood-brain barrier damage during the acute stage of subarachnoid hemorrhage, as exemplified by a new animal model. Neurosurgery. 18:733–739. 1986. View Article : Google Scholar : PubMed/NCBI

20 

Palade C, Ciurea AV, Nica DA, Savu R and Moisa HA: Interference of apoptosis in the pathophysiology of subarachnoid hemorrhage. Asian J Neurosurg. 8:106–111. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Gilgun-Sherki Y, Rosenbaum Z, Melamed E and Offen D: Antioxidant therapy in acute central nervous system injury: Current state. Pharmacol Rev. 54:271–284. 2002. View Article : Google Scholar : PubMed/NCBI

22 

de la Torre R, Covas MI, Pujadas MA, Fitó M and Farré M: Is dopamine behind the health benefits of red wine? Eur J Nutr. 45:307–310. 2006. View Article : Google Scholar : PubMed/NCBI

23 

Auñon-Calles D, Canut L and Visioli F: Toxicological evaluation of pure hydroxytyrosol. Food Chem Toxicol. 55:498–504. 2013. View Article : Google Scholar : PubMed/NCBI

24 

González-Correa JA, Navas MD, Lopez-Villodres JA, Trujillo M, Espartero JL and De La Cruz JP: Neuroprotective effect of hydroxytyrosol and hydroxytyrosol acetate in rat brain slices subjected to hypoxia-reoxygenation. Neurosci Lett. 446:143–146. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Ristagno G, Fumagalli F, Porretta-Serapiglia C, Orrù A, Cassina C, Pesaresi M, Masson S, Villanova L, Merendino A, Villanova A, et al: Hydroxytyrosol attenuates peripheral neuropathy in streptozotocin-induced diabetes in rats. J Agric Food Chem. 60:5859–5865. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Granados-Principal S, El-Azem N, Pamplona R, Ramirez-Tortosa C, Pulido-Moran M, Vera-Ramirez L, Quiles JL, Sanchez-Rovira P, Naudí A, Portero-Otin M, et al: Hydroxytyrosol ameliorates oxidative stress and mitochondrial dysfunction in doxorubicin-induced cardiotoxicity in rats with breast cancer. Biochem Pharmacol. 90:25–33. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Rouissi K, Hamrita B, Kouidi S, Messai Y, Jaouadi B, Hamden K, Medimegh I, Ouerhani S, Cherif M and Elgaaied AB: In vivo prevention of bladder urotoxicity: Purified hydroxytyrosol ameliorates urotoxic effects of cyclophosphamide and buthionine sulfoximine in mice. Int J Toxicol. 30:419–427. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Capasso G, Di Gennaro CI, Della Ragione F, Manna C, Ciarcia R, Florio S, Perna A, Pollastro RM, Damiano S, Mazzoni O, et al: In vivo effect of the natural antioxidant hydroxytyrosol on cyclosporine nephrotoxicity in rats. Nephrol Dial Transplant. 23:1186–1195. 2008. View Article : Google Scholar : PubMed/NCBI

29 

Pan S, Liu L, Pan H, Ma Y, Wang D, Kang K, Wang J, Sun B, Sun X and Jiang H: Protective effects of hydroxytyrosol on liver ischemia/reperfusion injury in mice. Mol Nutr Food Res. 57:1218–1227. 2003. View Article : Google Scholar

30 

Cao K, Xu J, Zou X, Li Y, Chen C, Zheng A, Li H, Li H, Szeto IM, Shi Y, et al: Hydroxytyrosol prevents diet-induced metabolic syndrome and attenuates mitochondrial abnormalities in obese mice. Free Radic Biol Med. 67:396–407. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Hagiwara K, Goto T, Araki M, Miyazaki H and Hagiwara H: Olive polyphenol hydroxytyrosol prevents bone loss. Eur J Pharmacol. 662:78–84. 2011. View Article : Google Scholar : PubMed/NCBI

32 

de la Puerta R, Ruiz Gutierrez V and Hoult JR: Inhibition of leukocyte 5-lipoxygenase by phenolics from virgin olive oil. Biochem Pharmacol. 57:445–449. 1999. View Article : Google Scholar : PubMed/NCBI

33 

González-Santiago M, Martín-Bautista E, Carrero JJ, Fonollá J, Baró L, Bartolomé MV, Gil-Loyzaga P and López-Huertas E: One month administration of hydroxytyrosol, phenolic antioxidant present in olive oil, to hyperlipemic rabbits improves blood lipid profile, antioxidant status and reduces atherosclerosis development. Atherosclerosis. 188:35–42. 2006. View Article : Google Scholar : PubMed/NCBI

34 

Zhao B, Ma Y, Xu Z, Wang J, Wang F, Wang D, Pan S, Wu Y, Pan H, Xu D, et al: Hydroxytyrosol, a natural molecule from olive oil, suppresses the growth of human hepatocellular carcinoma cells via inactivating AKT and nuclear factor-kappa B pathways. Cancer Lett. 347:79–87. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Lee-Huang S and Huang PL, Zhang D, Lee JW, Bao J, Sun Y, Chang YT, Zhang J and Huang PL: Discovery of small-molecule HIV-1 fusion and integrase inhibitors oleuropein and hydroxytyrosol: Part I. fusion (corrected) inhibition. Biochem Biophys Res Commun. 354:872–878. 2007. View Article : Google Scholar : PubMed/NCBI

36 

Cabrerizo S, De La Cruz JP, López-Villodres JA, Muñoz-Marín J, Guerrero A, Reyes JJ, Labajos MT and González-Correa JA: Role of the inhibition of oxidative stress and inflammatory mediators in the neuroprotective effects of hydroxytyrosol in rat brain slices subjected to hypoxia reoxygenation. J Nutr Biochem. 24:2152–2157. 2013. View Article : Google Scholar : PubMed/NCBI

37 

St-Laurent-Thibault C, Arseneault M, Longpré F and Ramassamy C: Tyrosol and hydroxytyrosol, two main components of olive oil, protect N2a cells against amyloid-β-induced toxicity. Involvement of the NF-κB signaling. Curr Alzheimer Res. 8:543–551. 2011. View Article : Google Scholar : PubMed/NCBI

38 

Schaffer S, Podstawa M, Visioli F, Bogani P, Müller WE and Eckert GP: Hydroxytyrosol-rich olive mill wastewater extract protects brain cells in vitro and ex vivo. J Agric Food Chem. 55:5043–5049. 2007. View Article : Google Scholar : PubMed/NCBI

39 

Park S, Yamaguchi M, Zhou C, Calvert JW, Tang J and Zhang JH: Neurovascular protection reduces early brain injury after subarachnoid hemorrhage. Stroke. 35:2412–2417. 2004. View Article : Google Scholar : PubMed/NCBI

40 

Garcia JH, Wagner S, Liu KF and Hu XJ: Neurological deficit and extent of neuronal necrosis attributable to middle cerebral artery occlusion in rats. Statistical validation. Stroke. 26:627–634; discussion 635. 1995. View Article : Google Scholar : PubMed/NCBI

41 

Xi G, Hua Y, Keep RF, Younger JG and Hoff JT: Brain edema after intracerebral Hemorrhage: The effects of systemic complement depletion. Acta Neurochir Suppl. 81:253–256. 2002.PubMed/NCBI

42 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) Method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

43 

Tsubokawa T, Jadhav V, Solaroglu I, Shiokawa Y, Konishi Y and Zhang JH: Lecithinized superoxide dismutase improves outcomes and attenuates focal cerebral ischemic injury via antiapoptotic mechanisms in rats. Stroke. 38:1057–1062. 2007. View Article : Google Scholar : PubMed/NCBI

44 

Wallace DC: A mitochondrial paradigm of metabolic and degenerative diseases, aging and cancer: A dawn for evolutionary medicine. Annu Rev Genet. 39:359–407. 2005. View Article : Google Scholar : PubMed/NCBI

45 

Winterbourn CC: Biological reactivity and biomarkers of the neutrophil oxidant, hypochlorous acid. Toxicology. 181-182:223–227. 2002. View Article : Google Scholar : PubMed/NCBI

46 

Macdonald RL and Weir BK: Cerebral vasospasm and free radicals. Free Radic Biol Med. 16:633–643. 1994. View Article : Google Scholar : PubMed/NCBI

47 

Ohsawa I, Ishikawa M, Takahashi Watanabe M, Nishimaki K, Yamagata K, Katsura K, Katayama Y, Asoh S and Ohta S: Hydrogen acts as a therapeutic antioxidant by selectively reducing cytotoxic oxygen radicals. Nat Med. 13:688–694. 2007. View Article : Google Scholar : PubMed/NCBI

48 

Floyd RA and Carney JM: Free radical damage to protein and DNA: mechanisms involved and relevant observations on brain undergoing oxidative stress. Ann Neurol. 32(Suppl): S22–S27. 1992. View Article : Google Scholar : PubMed/NCBI

49 

Asano T: Oxyhemoglobin as the principal cause of cerebral vasospasm: A holistic view of its actions. Crit Rev Neurosurg. 9:303–318. 1999. View Article : Google Scholar : PubMed/NCBI

50 

Rodriguez y Baena R, Gaetani P, Silvani V, Spanu G and Marzatico F: Effect of nimodipine on mitochondrial respiration in different rat brain areas after subarachnoid haemorrhage. Acta Neurochir Suppl (Wien). 43:177–181. 1988.PubMed/NCBI

51 

Kaynar MY, Tanriverdi T, Kafadar AM, Kacira T, Uzun H, Aydin S, Gumustas K, Dirican A and Kuday C: Detection of soluble intercellular adhesion molecule-1 and vascular cell adhesion molecule-1 in both cerebrospinal fluid and serum of patients after aneurysmal subarachnoid hemorrhage. J Neurosurg. 101:1030–1036. 2004. View Article : Google Scholar : PubMed/NCBI

52 

Rossi R, Dalle-Donne I, Milzani A and Giustarini D: Oxidized forms of glutathione in peripheral blood as biomarkers of oxidative stress. Clin Chem. 52:1406–1414. 2006. View Article : Google Scholar : PubMed/NCBI

53 

Erşahin M, Ozsavcı D, Sener A, Ozakpınar OB, Toklu HZ, Akakin D, Sener G and Yeğen BÇ: Obestatin alleviates subarachnoid haemorrhage-induced oxidative injury in rats via its anti-apoptotic and antioxidant effects. Brain Inj. 27:1181–1189. 2013. View Article : Google Scholar : PubMed/NCBI

54 

Faine LA, Rodrigues HG, Galhardi CM, Ebaid GM, Diniz YS, Padovani CR and Novelli EL: Effects of olive oil and its minor constituents on serum lipids, oxidative stress, and energy metabolism in cardiac muscle. Can J Physiol Pharml. 84:239–245. 2006. View Article : Google Scholar

55 

Deiana M, Incani A, Rosa A, Corona G, Atzeri A, Loru D, Paola Melis M and Assunta Dessì M: Protective effect of hydroxytyrosol and its metabolite homovanillic alcohol on H(2)O(2) induced lipid peroxidation in renal tubular epithelial cells. Food Chem Toxicol. 46:2984–2990. 2008. View Article : Google Scholar : PubMed/NCBI

56 

Erşahin M, Toklu HZ, Erzik C, Cetinel S, Akakin D, Velioğlu-Oğünç A, Tetik S, Ozdemir ZN, Sener G and Yeğen BC: The anti-inflammatory and neuroprotective effects of ghrelin in subarachnoid hemorrhage-induced oxidative brain damage in rats. J Neurotrauma. 27:1143–1155. 2010. View Article : Google Scholar : PubMed/NCBI

57 

Uttara B, Singh AV, Zamboni P and Mahajan RT: Oxidative stress and neurodegenerative diseases: A review of upstream and downstream antioxidant therapeutic options. Curr Neuropharmacol. 7:65–74. 2009. View Article : Google Scholar : PubMed/NCBI

58 

Kandhare AD, Raygude KS, Ghosh P, Ghule AE and Bodhankar SL: Neuroprotective effect of naringin by modulation of endogenous biomarkers in streptozotocin induced painful diabetic neuropathy. Fitoterapia. 83:650–659. 2012. View Article : Google Scholar : PubMed/NCBI

59 

Pastore A, Federici G, Bertini E and Piemonte F: Analysis of glutathione: Implication in redox and detoxification. Clinica Chim Acta. 333:19–39. 2003. View Article : Google Scholar

60 

Gutierrez VR, de la Puerta R and Catalá A: The effect of tyrosol, hydroxytyrosol and oleuropein on the non-enzymatic lipid peroxidation of rat liver microsomes. Mol Cell Biochem. 217:35–41. 2001. View Article : Google Scholar : PubMed/NCBI

61 

Graham DI, McIntosh TK, Maxwell WL and Nicoll JA: Recent advances in neurotrauma. J Neuropathol Exp Neurol. 59:641–651. 2000. View Article : Google Scholar : PubMed/NCBI

62 

Katzman R, Clasen R, Klatzo I, Meyer JS, Pappius HM and Waltz AG: Report of joint committee for stroke resources. IV. Brain edema in stroke. Stroke. 8:512–540. 1977. View Article : Google Scholar : PubMed/NCBI

63 

Kimelberg HK: Current concepts of brain edema. Review of laboratory investigations. J Neurosurg. 83:1051–1059. 1995. View Article : Google Scholar : PubMed/NCBI

64 

Feillet-Coudray C, Sutra T, Fouret G, Ramos J, Wrutniak-Cabello C, Cabello G, Cristol JP and Coudray C: Oxidative stress in rats fed a high-fat high-sucrose diet and preventive effect of polyphenols: Involvement of mitochondrial and NAD(P)H oxidase systems. Free Radic Biol Med. 46:624–632. 2009. View Article : Google Scholar : PubMed/NCBI

65 

Fukuhara T, Douville CM, Eliott JP, Newell DW and Winn HR: Relationship between intracranial pressure and the development of vasospasm after aneurysmal subarachnoid hemorrhage. Neurol Med Chir (Tokyo). 38:710–715; discussion 716–717. 1998. View Article : Google Scholar : PubMed/NCBI

66 

Imperatore C, Germanò A, d'Avella D, Tomasello F and Costa G: Effects of the radical scavenger AVS on behavioral and BBB changes after experimental subarachnoid hemorrhage. Life Sci. 66:779–790. 2000. View Article : Google Scholar : PubMed/NCBI

67 

Dóczi T: The pathogenetic and prognostic significance of blood-brain barrier damage at the acute stage of aneurysmal subarachnoid haemorrhage. Clinical and experimental studies. Acta Neurochir (Wien). 77:110–132. 1985. View Article : Google Scholar : PubMed/NCBI

68 

Fornezza U, Carraro R, Demo P, Zamperetti N, Volpin L, Landi A, De Luca GP and Benedetti A: The transcranial Doppler ultrasonography in the evaluation of vasospasm and of intracranial hypertension after subarachnoid hemorrhage. Agressologie. 31:259–261. 1990.PubMed/NCBI

69 

Mohagheghi F, Bigdeli MR, Rasoulian B, Zeinanloo AA and Khoshbaten A: Dietary virgin olive oil reduces blood brain barrier permeability, brain edema and brain injury in rats subjected to ischemia-reperfusion. Scientific World Journal. 10:1180–1191. 2010. View Article : Google Scholar : PubMed/NCBI

70 

Ayer RE and Zhang JH: Oxidative stress in subarachnoid haemorrhage: Signifi-cance in acute brain injury and vasospasm. Acta Neurochir Suppl. 104:33–41. 2008. View Article : Google Scholar : PubMed/NCBI

71 

Cregan SP, MacLaurin JG, Craig C, Robertson GS, Nicholson DW, Park DS and Slack RS: Bax-dependent caspase-3 activation is a key determinant in p53-induced apoptosis in neurons. J Neurosci. 19:7860–7869. 1999.PubMed/NCBI

72 

Simard JM, Geng Z, Woo SK, Ivanova S, Tosun C, Melnichenko L and Gerzanich V: Glibenclamide reduces inflammation, vasogenic edema and caspase-3 activation after subarachnoid hemorrhage. J Cereb Blood Flow Metab. 29:317–330. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2016
Volume 12 Issue 3

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhong YW, Wu J, Hu HL, Li WX and Zhong Y: Protective effect 3,4-dihydroxyphenylethanol in subarachnoid hemorrhage provoked oxidative neuropathy. Exp Ther Med 12: 1908-1914, 2016
APA
Zhong, Y., Wu, J., Hu, H., Li, W., & Zhong, Y. (2016). Protective effect 3,4-dihydroxyphenylethanol in subarachnoid hemorrhage provoked oxidative neuropathy. Experimental and Therapeutic Medicine, 12, 1908-1914. https://doi.org/10.3892/etm.2016.3526
MLA
Zhong, Y., Wu, J., Hu, H., Li, W., Zhong, Y."Protective effect 3,4-dihydroxyphenylethanol in subarachnoid hemorrhage provoked oxidative neuropathy". Experimental and Therapeutic Medicine 12.3 (2016): 1908-1914.
Chicago
Zhong, Y., Wu, J., Hu, H., Li, W., Zhong, Y."Protective effect 3,4-dihydroxyphenylethanol in subarachnoid hemorrhage provoked oxidative neuropathy". Experimental and Therapeutic Medicine 12, no. 3 (2016): 1908-1914. https://doi.org/10.3892/etm.2016.3526