Open Access

Ameliorative effects of 6‑gingerol in cerebral ischemia are mediated via the activation of antioxidant and anti‑inflammatory pathways

  • Authors:
    • Ratchaniporn Kongsui
    • Jinatta Jittiwat
  • View Affiliations

  • Published online on: February 15, 2023     https://doi.org/10.3892/br.2023.1608
  • Article Number: 26
  • Copyright: © Kongsui et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Focal ischemia occurs when an embolus or thrombus occludes an artery, causing the rapid obstruction of cerebral blood flow. Although stroke represents a main cause of disability and mortality in developing countries, therapeutic approaches available for this condition remain very limited. The aim of the present study was to examine the effects of the phytochemical, 6‑gingerol, on the brain infarct volume, neuronal loss and on the oxidative stress parameters, cyclooxygenase‑2 (COX‑2) and interleukin (IL)‑6, in an animal model of focal ischemic stroke. Male Wistar rats, weighing 250‑300 g, were divided into the following six groups: i) The control; ii) right middle cerebral artery occlusion (Rt.MCAO) + vehicle; iii) Rt.MCAO + piracetam; iv) Rt.MCAO + 6‑gingerol (6‑Gin) at 5 mg/kg body weight (BW); v) Rt.MCAO + 6‑Gin at 10 mg/kg BW; and vi) the Rt.MCAO + 6‑Gin at 20 mg/kg BW group. The rats in each group received the vehicle or piracetam or 6‑gingerol intraperitoneally for 7 days following Rt.MCAO. The brain infarct volume, neuronal loss and alterations in antioxidant and anti‑inflammatory levels were assessed in the cortex and hippocampus. The results revealed that the brain infarct volume, malondialdehyde level and the density ratio of COX‑2 and IL‑6 to β‑actin were significantly decreased following treatment with 6‑gingerol. In addition, neuronal density and superoxide dismutase activity in the cortex and hippocampus were increased. On the whole, the findings of the present study suggest that 6‑gingerol exerts antioxidant and anti‑inflammatory effects in vivo, which effectively ameliorate the brain damage induced by focal cerebral ischemic stroke.

Introduction

Focal ischemia occurs when an embolus or thrombus occludes an artery, causing the rapid obstruction of cerebral blood flow (CBF). There is increasing evidence to indicate that stroke is a main cause of disability and mortality in developing countries (1). Moreover, the global economic burden of stroke on health care is enormous. At present, stroke accounts for ~34% of total global healthcare-related costs (2). In addition, effective therapeutic strategies for stroke remain very limited. Novel treatment options are thus required.

Normally, the average CBF for an adult is ~50 ml/100 g/min, which is crucial for brain function (3). In focal infarcts, the CBF decreases to <10 ml/100 g/min, causing irreversible neuronal damage (4). Folioing the onset of complete ischemia, the levels of adenosine triphosphate (ATP) are depleted, disrupting ionic homeostasis. Radical species then form and these are involved in mediating damage. Cells have enzymatic and non-enzymatic defense mechanisms to protect themselves from this type of damage (5). Enzymatic mechanisms include neutralization by superoxide dismutase (SOD), glutathione peroxidase (GSH-Px) and catalase (CAT). Previous studies have demonstrated that increasing the levels of these antioxidant enzymes can mitigate brain damage caused by ischemic stroke (5-7).

Neuronal inflammation and apoptosis are also induced by the oxidative stress described above (8). Arachidonic acid derivatives and prostaglandins are the major inflammatory mediators. With the onset of ischemia, the levels of intracellular calcium increase, with the consequent activation of phospholipase. The metabolites of arachidonic acid are the most critical contributors to the pathophysiology of ischemic stroke. A number of studies have found that interleukin (IL)-1 and IL-6 are also mediators of ischemic damage (9-11). Cyclooxygenase-2 (COX-2) is also a mediator of inflammation (12,13), and there is accumulating evidence to indicate that COX-2 suppression can attenuate ischemic injury following middle cerebral artery occlusion (14). Herbal bioactive components exhibit anti-inflammatory, antioxidant and anti-apoptotic properties that may have therapeutic potential against the neuronal injury caused by ischemic stroke.

The main phytochemical present in ginger (Zingiber officinale; Zingiberaceae family) is 6-gingerol. This compound exhibits a variety of pharmacological properties, including anti-inflammatory, antioxidant (15,16), anti-apoptotic (17) and anticancer activities (18). Additionally, a previous study demonstrated that 6-gingerol significantly reduced the infarct volume and brain damage following ischemia/reperfusion injury by inhibiting NLR family pyrin domain containing 3 inflammasome-induced inflammation and neuronal apoptosis (19). However, the effects of 6-gingerol against focal ischemic stroke-induced brain damage have not yet been evaluated, at least to the best of our knowledge. Thus, the present study examined the effects of 6-gingerol on the brain infarct volume, neuronal loss and on the oxidative stress parameters, COX-2 and IL-6, in rats following focal ischemic stroke.

Materials and methods

Test treatments

The test compound, 6-gingerol (98.7% purity; chemical structure illustrated in Fig. 1), was purchased from Chengdu Biopurify Phytochemicals Ltd. Piracetam, the positive control, was obtained from Glaxosmithkline (Thailand) Ltd. and DMSO, the vehicle, was obtained from Thermo Fisher Scientific, Inc. (product code: D/4121/PB15).

Study design

A total of 90 healthy male Wistar rats (weighing 250-300 g, 8 weeks old, from the Northeastern Laboratory Animal Center, Khon Kaen University, Khon Kaen, Thailand) were randomly divided into six groups (n=15 per group) as follows: i) The control; ii) right middle cerebral artery occlusion (Rt.MCAO) + vehicle; iii) Rt.MCAO + piracetam at 250 mg/kg body weight (BW); iv) Rt.MCAO + 6-gingerol (6-Gin) at 5 mg/kg BW; v) Rt.MCAO + 6-Gin at 10 mg/kg BW; and vi) the Rt.MCAO + 6-Gin at 20 mg/kg BW group. The animals in all the groups received their treatments by intraperitoneal (i.p.) injection once daily for 7 consecutive days following Rt.MCAO. All rats were housed in groups of five in standard metal cages, maintained under standard conditions with a 12-h on/off light/dark cycle, relative humidity controlled at ~30-60%, and a temperature controlled at 23±2˚C. The rats had ad libitum access to water and commercial pellets (24 h/day). The piracetam at 250 mg/kg BW and 6-gingerol at 5, 10 and 20 mg/kg BW were selected based on previous research by the authors, preliminary studies and literature reviews (6,16,20). Moreover, as previously reported, the median lethal dose of gingerol (i.p. administration) has been determined in mice and rats. The dose required to kill half the members of a tested population following a specified test duration value (LD50) has been reported to be ~58.1 mg/kg BW (21). Thus, the doses used in the present study were noted to be non-toxic. The infarct volume was examined for 5 rats in each group using 2,3,5-triphenyltetrazolium chloride (TTC) staining. Another 5 rats per group were used to measure cerebral cortex and the hippocampus neuronal density using cresyl violet staining. The remaining 5 animals per group were used to examine the malondialdehyde (MDA) levels and SOD activities in the cortex and hippocampus mitochondria using biochemical assays. IL-6 and COX-2 expression levels were also measured in the cortex and hippocampus of the rats treated with those doses of 6-gingerol that produced optimum changes in infarct volume, neuronal density and oxidative stress markers (Fig. 2). Two replicates were performed for each test.

Model of Rt.MCAO

All animals were fasted overnight, but were allowed free access to water before their surgery. The animals were anesthetized using isoflurane (5% for induction and 1-3% for maintenance) delivered in 100% oxygen. The model of focal ischemia was established by the permanent intraluminal occlusion of the right middle cerebral artery, as previously described (22). Briefly, a 4-0 silicone-coated monofilament (USS DGTM Division of United States Surgical; Tyco Healthcare Group LP, Norwalk, CT, USA) was inserted into the internal carotid artery ~17 mm or until a slight resistance was detected. The wound was then sutured and 10% povidone iodine solution was applied at the incision site for antiseptic postoperative care. In the sham operation, all the arteries were exposed as described above, but monofilament insertion was not performed. The criteria for humane endpoints was defined as the inability to move, wound infection following surgery, a weight loss of >20%, dehydration, dyspnea, progressive pain, lack of response to external stimuli and bleeding from any orifice. However, all animals in the present study survived to the end of the study period (8 days).

Determination of brain infarct volume

At the end of the study period, the rats were anesthetized with thiopental sodium (80 mg/kg BW; i.p. administration) prior to cardiac perfusion with cold normal saline solution. The brains were then removed from the skull and the 2-mm-thick coronal sections were stained with 2% TTC (MilliporeSigma) in normal saline for 30 min at 37˚C. Images were then obtained using a digital camera and the infarct volume was determined using Image J® software (version 1.53e, National Institutes of Health). The infarct volumes were then calculated using the formula described in a previous study by the authors (6).

Cresyl violet staining for neuronal density determination

Serial coronal sections of the cortex and hippocampus (30-µm-thick) were stained with cresyl violet acetate solution (MilliporeSigma) for 16 min at 60˚C to determine the neuronal density. Regions of the cortex and hippocampus (CA1, CA2 and CA3) were then examined using an Olympus light microscope (model CX23; Olympus Corporation) at x40 magnification. Images of the cortex and hippocampus at stereotaxic co-ordinates, selected as described in a previous study by the authors (23), were used to measure neuronal density. This was achieved by blinded analysis, and the data are expressed as a percentage of the control.

Isolation of brain mitochondria for biochemical assays

Following the completion of perfusion, brain tissues from the cerebral cortex and hippocampal regions were isolated and prepared for mitochondrial extraction, using a protocol described in a previous study by the authors (24). The brain tissues were stored at -80˚C until use.

Protein determination

The method described in the study by Lowry et al (25) was used to determine the mitochondrial protein concentrations in the brain areas aforementioned, using bovine serum albumin (MilliporeSigma) as a standard.

Determination of the MDA level

The lipid peroxidation product, MDA, was used as an indicator of oxidative stress. Its levels were measured using the thiobarbituric acid (MilliporeSigma) reaction in all samples, according to the method described in the study by Ohkawa et al (26). The results are reported as nmol/mg protein mitochondria.

Determination of SOD activity

A SOD assay kit from MilliporeSigma (19160-1K-F) was used to determine the SOD activity. Data are expressed as U/mg protein mitochondria.

Western blot analysis

At the end of the study period, COX-2 and IL-6 expression levels were measured in the cortex and hippocampus of the rats using western blot analysis as described in a previous study by the authors (22). The brain tissues were homogenized in lysis buffer (cat. no. 87792, Thermo Fisher Scientific, Inc.) and the method of Lowry et al (25) was used to determine the total protein concentrations. Equal quantities of protein (40 µg protein) were subjected to 10% SDS-polyacrylamide gel electrophoresis, transferred to a Hybond-P (PVDF) membrane (Cytiva) and then incubated with rabbit monoclonal anti-COX-2 (1:1,000, cat. no. ab179800, Abcam), mouse monoclonal anti-IL-6 (1:2,000, cat. no. ab9324, Abcam) and rabbit monoclonal anti-β-actin (1:5,000, cat. no. AC026, ABclonal Biotech Co., Ltd.) antibodies at 4˚C overnight. The membranes were then incubated with anti-rabbit (1:2,000, cat. no. AS063, ABclonal Biotech Co., Ltd.) or anti-mouse (1:2,000, cat. no. 12-349, MilliporeSigma) secondary antibodies for 1 h at room temperature. The immunoreactive proteins on the blots were visualized using chemiluminescent substrate (Supersignal West Pico; Pierce; Thermo Fisher Scientific, Inc.). The density of the COX-2 and IL-6 bands were normalized to β-actin, and the protein expression was calculated using a ChemiDocTM MP imaging system with Image Lab software (version 6.0.0 build 25, Bio-Rad Laboratories Inc.).

Statistical analysis

The data are expressed as the mean ± the standard error of the mean. Statistical analysis was assessed using one-way analysis of variance (ANOVA), followed by a Tukey’s post hoc test using SPSS® software (version 25, SPSS-IBM Inc.). A P-value <0.05 was considered to indicate a statistically significant difference.

Results

Effects of 6-gingerol on brain injury in rats subjected to Rt.MCAO

The present study measured the brain infarct volume using TTC staining following treatment of the rats with 6-gingerol. Rats undergoing permanent occlusion (Rt.MCAO) and who received the vehicle exhibited a significantly increased infarct volume (P<0.05) compared to the rats in the control group (Fig. 3). However, the rats treated with piracetam and 6-gingerol exhibited a marked decrease in their infarct volumes compared to the vehicle-treated ischemic stroke group (P<0.05).

Effects of 6-gingerol on neuronal damage in the cortex and hippocampus of rats subjected to Rt.MCAO

Cerebral ischemia induced major neuronal damage in the cortex and all subregions of the hippocampus (CA1, CA2 and CA3). The Rt.MCAO + vehicle (DMSO) group exhibited a significant decrease in neuronal survival in the cortex or CA1, CA2 or CA3 regions of the hippocampus compared to the control. By contrast, piracetam (at a dose of 250 mg/kg BW) and 6-gingerol (at doses of 10 and 20 mg/kg BW) markedly reduced ischemic stroke-induced neuronal loss within the cortex and all subregions of the hippocampus (P<0.05) compared to the Rt.MCAO + vehicle group (Fig. 4). At the lower dose 5 mg/kg BW), 6-gingerol did not lead to any significant difference in neuronal damage in all areas compared to the Rt.MCAO + vehicle group.

Effects of 6-gingerol on oxidative stress in the mitochondria from the cortex and hippocampus of rats subjected to Rt.MCAO

As oxidative stress plays a key role in the pathogenesis of ischemic stroke, the present study also examined the effects of 6-gingerol on oxidative stress markers, including MDA levels and activity of the scavenging enzyme, SOD, in the mitochondria. Rats that underwent permanent Rt.MCAO exhibited increased mitochondrial MDA levels (Fig. 5A) and significantly reduced mitochondrial SOD activities (Fig. 5B) compared to the control group. By contrast, the rats treated with piracetam or 6-gingerol (5, 10 and 20 mg/kg BW) exhibited mitochondrial SOD activities that were significantly less diminished and mitochondrial MDA levels that were significantly less elevated than the Rt.MCAO + vehicle group both in the cerebral cortex and hippocampus (P<0.05; Fig. 5).

Effects of 6-gingerol on COX-2 and IL-6 expression in the cortex and hippocampus of rats subjected to Rt.MCAO

To assess the anti-inflammatory activity of 6-gingerol, the present study measured COX-2 and IL-6 protein expression in the cortex and hippocampus using western blot analysis. As 20 mg/kg BW 6-gingerol led to optimum changes in infarct volume, neuronal density and oxidative stress markers, this dose was selected to investigate the effects of 6-gingerol on COX-2 and IL-6 levels. Bands showing positive immunoreactivities against COX-2, β-actin and IL-6 were detected at 74, 41 and 25 kDa, respectively (Fig. 6A). Treatment with 6-gingerol at a dose of 20 mg/kg BW markedly decreased the density ratios of COX-2 and IL-6 to the β-actin band compared to the Rt.MCAO + vehicle group (P<0.05, Fig. 6B and C).

Discussion

Animal models are widely used to gain understanding of the pathophysiology of cerebral ischemic injury. In such studies, MCAO is one of the most commonly employed surgical procedures to induce ischemic stroke (27). This model, known as the permanent MCAO model, has been developed to mimic human ischemic stroke, and generates cerebral infarction in both cortical and subcortical areas (28). The present study used the MCAO model to investigate 6-gingerol as an experimental therapy. As was expected, cortical and subcortical region infarct volumes were large in rats treated with only DMSO, whereas these volumes were significantly smaller in the rats treated with 6-gingerol and piracetam.

In addition to the infarcts described above, cortical and hippocampal neuronal loss and brain damage are reportedly induced in the MCAO model (29-31), with apoptotic cell death occurring after 24 h (32-34). Data from the present study are in agreement with these previous findings. Herein, the positive control drug, piracetam, which increases CBF, was found to enhance the density of neurons within the cortex and hippocampus. This result is also in agreement with those of previous studies, which reported the nootropic action of this drug and its ability to ameliorate cerebral ischemia-induced brain damage (35-37). In the present study, the experimental treatment agent, 6-gingerol, markedly alleviated ischemic stroke-induced neuronal damage within the cortex and hippocampus regions CA1, CA2 and CA3. Although this has not been previously demonstrated, at least to the best of our knowledge, it does correspond well with other reported activities of 6-gingerol. For example, 6-gingerol has previously been shown to reduce the in vitro apoptosis of PC12 cells (38). It has also been previously demonstrated that 6-gingerol protects rats from lipopolysaccharide-induced brain injury by improving the expression of brain-derived neurotrophic factor (BDNF) within the cerebral cortex and hippocampus (20). In another in vivo study, 6-gingerol improved the hippocampal levels of BDNF and nerve growth factor in rats exposed to gold nanoparticles (17). Moreover, during pathophysiological processes in the subacute phase of ischemic stroke (up to 7 days following complete ischemia onset), ATP levels are depleted, disrupting ionic homeostasis. Radical species then form and these are involved in mediating damage. Cells have enzymatic and non-enzymatic defense mechanisms to protect themselves from this type of damage (5). In addition, the initiation of the inflammatory response followed by the release of mediators exacerbates the effects of neuronal inflammation and primary damage (39). Therefore, the present study selected the duration of 6-gingerol treatment to be 7 days.

Reactive oxygen species (ROS) contribute to the progression of numerous diseases (40-42), and both ROS and lipid peroxidation (LPO) are known to mediate tissue damage in ischemic stroke. Cells have enzymatic and non-enzymatic defense mechanisms to protect themselves from some of this damage (5). Enzymatic mechanisms include neutralization by SOD, GSH-Px and CAT. Previous studies have found that increasing these antioxidant enzymes can mitigate brain damage from ischemic stroke (5-7). Another study demonstrated that 6-gingerol is a potent antioxidant, with the potential to treat and prevent chronic diseases (43). As mitochondrial dysfunction and SOD deficiency contribute to increased neuronal death, increased numbers of superoxide anions and increased cerebral infarction (44,45), the present study examined the effects of 6-gingerol on LPO (by examining the MDA levels) and SOD activity. The results revealed that both the positive control, piracetam, and the experimental treatment agent, 6-gingerol (5, 10 and 20 mg/kg BW), decreased the LPO product and increased SOD activity in cortical and hippocampal mitochondria compared to the Rt.MCAO + vehicle group. Several lines of evidence demonstrate that 6-gingerol has antioxidant, anti-inflammatory (15,16), anti-apoptotic (17) and anticancer properties (18). In addition, a 6-gingerol rich fraction from Z. officinale has been shown to prevent acrylonitrile-induced cerebral cortex damage partly via its antioxidant and anti-inflammatory activities (46).

In the development of ischemic damage, the production of free radicals is markdly elevated, and oxidative stress causes neuronal inflammation and apoptosis (8). Arachidonic acid derivatives and prostaglandins are two of the major inflammatory mediators involved. With the onset of ischemia, intracellular calcium increases, with the consequent activation of phospholipase. Arachidonic acid metabolites are then formed, contributing to the pathophysiological process. A number of studies have found that IL-1 and IL-6 are also inflammatory mediators of ischemic damage (9-11). COX-2 is a key inflammatory mediator in cerebral ischemia and neurodegenerative disorders too (12,13). Recent evidence indicates that the suppression of COX-2 attenuates ischemic injury following MCAO (14). In the present study, significant decreases in the density ratio of COX-2 and IL-6 to the β-actin band were detected in rats subjected to Rt.MCAO receiving 20 mg/kg BW 6-gingerol for 7 days. This is in accordance with the findings of previous studies demonstrating that the inhibition of inflammation can decrease the brain infarct volume in experimental stroke (47) and 6-gingerol can alleviate inflammatory damage by inhibiting the production of pro-inflammatory cytokines (48-51). Consequently, 6-gingerol can attenuate focal cerebral ischemic stroke-induced neuronal injury by suppressing oxidative stress and inflammatory mediators.

In animal studies, it has been demonstrated that the mitochondria are the main source of free radical generation following focal cerebral ischemia. It is possible that mutations in nuclear genes following cerebral ischemia alter the apoptotic process. Gene mutations in mitochondria may be an initiating event that leads to apoptotic neuronal death. Therefore, drugs or other interventions that enhance repair efficiency or reduce apoptosis in the brain offer the possibility of improved outcomes following ischemic stroke. An imbalance between mitochondrial fusion and fission can also enhance free radical generation and has been detected in stroke and other diseases. Mitogen-activated protein kinase (MAPK) is associated with mitochondrial dynamics. Bioinformatics research has demonstrated that some miRNAs are involved in the modulation of MAPK, which is crucial for the alleviation of inflammation and apoptosis in ischemic stroke (52-54). Furthermore, research using rat models of MCAO has demonstrated that reductions in mitofusin (Mfn)1 and Mfn2 induce Ca2+ overload mitochondrial Bax translocation (55) and promote neuronal apoptosis. Additionally, Mfn2 has been reported to decrease caspase-3 and increase the Bcl-2/Bax ratio, these two factors reducing cellular susceptibility to apoptosis after cerebral ischemic stroke (54). Further studies are t required in order better understand the effects of 6-gingerol on these pathways and its potential as a supportive treatment for patients with ischemic stroke.

The present study has a few limitations. Firstly, a weakness of the MCAO model is that the filament may not always be inserted far enough to completely occlude the middle cerebral artery. To address this issue, some researchers use laser doppler flowmetry (LDF) to measure the reduction in CBF and confirm successful occlusion. The present study did not use LDF or micro computed tomography (micro-CT), although the authors did always check that the filament was of optimal size and that the point of occlusion was associated with the infarct size after the rats were euthanized. A second limitation is that the present study did not measure LPO or SOD activity in the blood or other tissues. LPO and SOD activity were measured in just the cerebral cortex and hippocampus mitochondria. Lastly, the small number of replicates used in the biochemical assays represents another study limitation. Three or more replicates would have been preferable to two; however, pipetting was performed by an experienced laboratorian and the standard curves had high R2 values.

In conclusion, 6-gingerol (10 and 20 mg/kg BW for 7 days) exerted antioxidant and anti-inflammatory effects, which effectively reduced brain damage in a rat model of focal cerebral ischemic stroke. However, further investigations are warranted to determine whether any additional mechanism(s) contribute to the ameliorative effects detected in the present study.

Acknowledgements

The authors would like to thank Dr Tim Cushnie (Faculty of Medicine, Mahasarakham University, Mahasarakham, Thailand) for language-editing assistance. As Mahasarakham University does not have any animal housing facility, the animal experiments were performed at the Northeastern Laboratory Animal Center, Khon Kaen University, Khon Kaen, Thailand, which is close to Mahasarakham University.

Funding

Funding: The present study received funding from the Mahasarakham University Faculty of Medicine (Med MSU 2/65).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

RK was involved in the study methodology, and in the writing, reviewing and editing of the manuscript. JJ was involved in the conception and design of the study, in funding acquisition, data curation, in the study methodology, as well as in the writing of the original draft and in the writing, reviewing and editing of the manuscript and in project administration. Both authors have read and approved the final manuscript and confirmed the authenticity of all the raw data.

Ethics approval and consent to participate

All animal-related protocols were designed to minimize animal suffering, and were performed according to the approval of the Institutional Animal Care and Use Committee at Khon Kaen University, Khon Kaen, Thailand (Record No. IACUC-KKU-6/65).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Gebreyohannes EA, Bhagavathula AS, Abebe TB, Seid MA and Haile KT: In-Hospital mortality among ischemic stroke patients in Gondar University Hospital: A retrospective cohort study. Stroke Res Treat. 2019(7275063)2019.PubMed/NCBI View Article : Google Scholar

2 

Rochmah TN, Rahmawati IT, Dahlui M, Budiarto W and Bilqis N: Economic burden of stroke disease: A systematic review. Int J Environ Res Public Health. 18(7552)2021.PubMed/NCBI View Article : Google Scholar

3 

Fantini S, Sassaroli A, Tgavalekos KT and Kornbluth J: Cerebral blood flow and autoregulation: current measurement techniques and prospects for noninvasive optical methods. Neurophotonics. 3(031411)2016.PubMed/NCBI View Article : Google Scholar

4 

Jaffer H, Morris VB, Stewart D and Labhasetwar V: Advances in stroke therapy. Drug Deliv Transl Res. 1:409–419. 2011.PubMed/NCBI View Article : Google Scholar

5 

Davis SM and Pennypacker KR: Targeting antioxidant enzyme expression as a therapeutic strategy for ischemic stroke. Neurochem Int. 107:23–32. 2017.PubMed/NCBI View Article : Google Scholar

6 

Jittiwat J, Suksamrarn A, Tocharus C and Tocharus J: Dihydrocapsaicin effectively mitigates cerebral ischemia-induced pathological changes in vivo, partly via antioxidant and anti-apoptotic pathways. Life Sci. 283(119842)2021.PubMed/NCBI View Article : Google Scholar

7 

Jittiwat J, Chonpathompikunlert P and Sukketsiri W: Neuroprotective effects of Apium graveolens against focal cerebral ischemia occur partly via antioxidant, anti-inflammatory, and anti-apoptotic pathways. J Sci Food Agric. 101:2256–2263. 2021.PubMed/NCBI View Article : Google Scholar

8 

Wu L, Xiong X, Wu X, Ye Y, Jian Z, Zhi Z and Gu L: Targeting oxidative stress and inflammation to prevent ischemia-reperfusion injury. Front Mol Neurosci. 13(28)2020.PubMed/NCBI View Article : Google Scholar

9 

Vidale S, Consoli A, Arnaboldi M and Consoli D: Postischemic Inflammation in Acute Stroke. J Clin Neurol. 13:1–9. 2017.PubMed/NCBI View Article : Google Scholar

10 

Kawabori M and Yenari MA: Inflammatory responses in brain ischemia. Curr Med Chem. 22:1258–1277. 2015.PubMed/NCBI View Article : Google Scholar

11 

Jin R, Yang G and Li G: Inflammatory mechanisms in ischemic stroke: Role of inflammatory cells. J Leukoc Biol. 87:779–789. 2010.PubMed/NCBI View Article : Google Scholar

12 

Fathali N, Ostrowski RP, Lekic T, Jadhav V, Tong W, Tang J and Zhang JH: Cyclooxygenase-2 inhibition provides lasting protection against neonatal hypoxic-ischemic brain injury. Crit Care Med. 38:572–578. 2010.PubMed/NCBI View Article : Google Scholar

13 

Minghetti L: Cyclooxygenase-2 (COX-2) in inflammatory and degenerative brain diseases. J Neuropathol Exp Neurol. 63:901–910. 2004.PubMed/NCBI View Article : Google Scholar

14 

Yan W, Ren D, Feng X, Huang J, Wang D, Li T and Zhang D: Neuroprotective and Anti-Inflammatory Effect of pterostilbene against cerebral ischemia/reperfusion injury via suppression of COX-2. Front Pharmacol. 12(70329)2021.PubMed/NCBI View Article : Google Scholar

15 

Alsahli MA, Almatroodi SA, Almatroudi A, Khan AA, Anwar S, Almutary AG, Alrumaihi F and Rahmani AH: 6-Gingerol, a major ingredient of ginger attenuates Diethylnitrosamine-Induced liver injury in rats through the modulation of oxidative stress and anti-inflammatory activity. Mediators Inflamm. 2021(6661937)2021.PubMed/NCBI View Article : Google Scholar

16 

Almatroodi SA, Alnuqaydan AM, Babiker AY, Almogbel MA, Khan AA and Husain Rahmani A: 6-Gingerol, a bioactive compound of ginger attenuates renal damage in Streptozotocin-Induced diabetic rats by regulating the oxidative stress and inflammation. Pharmaceutics. 13(317)2021.PubMed/NCBI View Article : Google Scholar

17 

Majdi Yazdi G, Vaezi G, Hojati V and Mohammad-Zadeh M: The Effect of 6-gingerol on Growth factors and apoptosis indices in rats exposed to gold nanoparticles. Basic Clin Neurosci. 12:301–308. 2021.PubMed/NCBI View Article : Google Scholar

18 

Wang S, Zhang C, Yang G and Yang Y: Biological properties of 6-gingerol: A brief review. Nat Prod Commun. 9:1027–1030. 2014.PubMed/NCBI

19 

Luo J, Chen J, Yang C, Tan J, Zhao J, Jiang N and Zhao Y: 6-Gingerol protects against cerebral ischemia/reperfusion injury by inhibiting NLRP3 inflammasome and apoptosis via TRPV1/FAF1 complex dissociation-mediated autophagy. Int Immunopharmacol. 100(108146)2021.PubMed/NCBI View Article : Google Scholar

20 

Adetuyi BO and Farombi EO: 6-Gingerol, an active constituent of ginger, attenuates lipopolysaccharide-induced oxidation, inflammation, cognitive deficits, neuroplasticity, and amyloidogenesis in rat. J Food Biochem. 45(e13660)2021.PubMed/NCBI View Article : Google Scholar

21 

Suekawa M, Ishige A, Yuasa K, Sudo K, Aburada M and Hosoya E: Pharmacological studies on ginger. I. Pharmacological actions of pungent constitutents, (6)-gingerol and (6)-shogaol. J Pharmacobiodyn. 7:836–848. 1984.PubMed/NCBI View Article : Google Scholar

22 

Jittiwat J: Baihui point laser acupuncture ameliorates cognitive impairment, motor deficit, and neuronal loss partly via antioxidant and anti-inflammatory effects in an animal model of focal ischemic stroke. Evid Based Complement Alternat Med. 2019(1204709)2019.PubMed/NCBI View Article : Google Scholar

23 

Wattanathorn J, Jittiwat J, Tongun T, Muchimapura S and Ingkaninan K: Zingiber officinale Mitigates Brain Damage and Improves Memory Impairment in Focal Cerebral Ischemic Rat. Evid Based Complement Alternat Med. 2011(429505)2011.PubMed/NCBI View Article : Google Scholar

24 

Jittiwat J: Laser Acupuncture at GV20 Improves brain damage and oxidative stress in animal model of focal ischemic stroke. J Acupunct Meridian Stud. 10:324–330. 2017.PubMed/NCBI View Article : Google Scholar

25 

Lowry OH, Rosebrough NJ, Farr AL and Randall RJ: Protein measurement with the Folin phenol reagent. J Biol Chem. 193:265–275. 1951.PubMed/NCBI

26 

Ohkawa H, Ohishi N and Yagi K: Assay for lipid peroxides in animal tissues by thiobarbituric acid reaction. Anal Biochem. 95:351–358. 1979.PubMed/NCBI View Article : Google Scholar

27 

Rossmeisl JH Jr, Rohleder JJ, Pickett JP, Duncan R and Herring IP: Presumed and confirmed striatocapsular brain infarctions in six dogs. Vet Ophthalmol. 10:23–36. 2007.PubMed/NCBI View Article : Google Scholar

28 

Wayman C, Duricki DA, Roy LA, Haenzi B, Tsai SY, Kartje G, Beech JS, Cash D and Moon L: Performing permanent distal middle cerebral with common carotid artery occlusion in aged rats to study cortical ischemia with sustained disability. J Vis Exp. (53106)2016.PubMed/NCBI View Article : Google Scholar

29 

Teertam SK and Prakash Babu P: Differential role of SIRT1/MAPK pathway during cerebral ischemia in rats and humans. Sci Rep. 11(6339)2021.PubMed/NCBI View Article : Google Scholar

30 

Wang C, Ma Z, Wang Z, Ming S, Ding Y, Zhou S and Qian H: Eriodictyol attenuates MCAO-Induced brain injury and neurological deficits via reversing the autophagy dysfunction. Front Syst Neurosci. 15(655125)2021.PubMed/NCBI View Article : Google Scholar

31 

Lee B, Choi EJ, Lee EJ, Han SM, Hahm DH, Lee HJ and Shim I: The neuroprotective effect of methanol extract of gagamjungjihwan and fructus euodiae on ischemia-induced neuronal and cognitive impairment in the rat. Evid Based Complement Alternat Med. 2011(685254)2011.PubMed/NCBI View Article : Google Scholar

32 

Shah FA, Li T, Kury LTA, Zeb A, Khatoon S, Liu G, Yang X, Liu F, Yao H, Khan AU, et al: Pathological comparisons of the hippocampal changes in the transient and permanent middle cerebral artery occlusion rat models. Front Neurol. 10(1178)2019.PubMed/NCBI View Article : Google Scholar

33 

Chung JY, Yi JW, Kim SM, Lim YJ, Chung JH and Jo DJ: Changes in gene expression in the rat hippocampus after focal cerebral ischemia. J Korean Neurosurg Soc. 50:173–178. 2011.PubMed/NCBI View Article : Google Scholar

34 

Genovese T, Mazzon E, Paterniti I, Esposito E, Bramanti P and Cuzzocrea S: Modulation of NADPH oxidase activation in cerebral ischemia/reperfusion injury in rats. Brain Res. 1372:92–102. 2011.PubMed/NCBI View Article : Google Scholar

35 

Paliwal P, Dash D and Krishnamurthy S: Pharmacokinetic study of piracetam in focal cerebral ischemic rats. Eur J Drug Metab Pharmacokinet. 43:205–213. 2018.PubMed/NCBI View Article : Google Scholar

36 

Muley MM, Thakare VN, Patil RR, Bafna PA and Naik SR: Amelioration of cognitive, motor and endogenous defense functions with silymarin, piracetam and protocatechuic acid in the cerebral global ischemic rat model. Life Sci. 93:51–57. 2013.PubMed/NCBI View Article : Google Scholar

37 

He Z, Liao Y, Zheng M, Zeng FD and Guo LJ: Piracetam improves cognitive deficits caused by chronic cerebral hypoperfusion in rats. Cell Mol Neurobiol. 28:613–627. 2008.PubMed/NCBI View Article : Google Scholar

38 

Rezazadeh-Shojaee FS, Ramazani E, Kasaian J and Tayarani-Najaran Z: Protective effects of 6-gingerol on 6-hydroxydopamine-induced apoptosis in PC12 cells through modulation of SAPK/JNK and survivin activation. J Biochem Mol Toxicol. 36(e22956)2022.PubMed/NCBI View Article : Google Scholar

39 

Zhao M, Yao Y, Du J, Kong L, Zhao T, Wu D, Man L and Zhou W: 6-Gingerol Alleviates neonatal hypoxic-ischemic cerebral and white matter injury and contributes to functional recovery. Front Pharmacol. 12(707772)2021.PubMed/NCBI View Article : Google Scholar

40 

Liu Z, Ren Z, Zhang J, Chuang CC, Kandaswamy E, Zhou T and Zuo L: Role of ROS and Nutritional antioxidants in human diseases. Front Physiol. 9(477)2018.PubMed/NCBI View Article : Google Scholar

41 

Gorjão R, Takahashi HK, Pan JA and Massao Hirabara S: Molecular mechanisms involved in inflammation and insulin resistance in chronic diseases and possible interventions. J Biomed Biotechnol. 2012(841983)2012.PubMed/NCBI View Article : Google Scholar

42 

Liou GY and Storz P: Reactive oxygen species in cancer. Free Radic Res. 44:479–496. 2010.PubMed/NCBI View Article : Google Scholar

43 

Mohd Yusof YA: Gingerol and its role in chronic diseases. Adv Exp Med Biol. 929:177–207. 2016.PubMed/NCBI View Article : Google Scholar

44 

El-Senousey HK, Chen B, Wang JY, Atta AM, Mohamed FR and Nie QH: Effects of dietary vitamin C, vitamin E, and alpha-lipoic acid supplementation on the antioxidant defense system and immune-related gene expression in broilers exposed to oxidative stress by dexamethasone. Poult Sci. 97:30–38. 2018.PubMed/NCBI View Article : Google Scholar

45 

Watts LT, Lloyd R, Garling RJ and Duong T: Stroke neuroprotection: Targeting mitochondria. Brain Sci. 3:540–560. 2013.PubMed/NCBI View Article : Google Scholar

46 

Farombi EO, Abolaji AO, Adetuyi BO, Awosanya O and Fabusoro M: Neuroprotective role of 6-Gingerol-rich fraction of Zingiber officinale (Ginger) against acrylonitrile-induced neurotoxicity in male Wistar rats. J Basic Clin Physiol Pharmacol. 30:2018.PubMed/NCBI View Article : Google Scholar

47 

Whiteley W, Jackson C, Lewis S, Lowe G, Rumley A, Sandercock P, Wardlaw J, Dennis M and Sudlow C: Inflammatory markers and poor outcome after stroke: A prospective cohort study and systematic review of interleukin 6. PLoS Med. 6(e1000145)2009.PubMed/NCBI View Article : Google Scholar

48 

Ju SA, Nguyen QT, Nguyen TT, Suh JH, An WG, Callaway Z, Joe Y, Chung HT and Kim BS: Pretreatment with 6-Gingerol Ameliorates Sepsis-Induced Immune Dysfunction by Regulating the Cytokine Balance and Reducing Lymphocyte Apoptosis. Oxid Med Cell Longev. 2021(5427153)2021.PubMed/NCBI View Article : Google Scholar

49 

Hwang YH, Kim T, Kim R and Ha H: The Natural Product 6-Gingerol Inhibits Inflammation-Associated Osteoclast Differentiation via Reduction of Prostaglandin E2 Levels. Int J Mol Sci. 19(2068)2018.PubMed/NCBI View Article : Google Scholar

50 

Tripathi S, Maier KG, Bruch D and Kittur DS: Effect of 6-gingerol on pro-inflammatory cytokine production and costimulatory molecule expression in murine peritoneal macrophages. J Surg Res. 138:209–213. 2007.PubMed/NCBI View Article : Google Scholar

51 

Kim SO, Chun KS, Kundu JK and Surh YJ: Inhibitory effects of [6]-gingerol on PMA-induced COX-2 expression and activation of NF-kappaB and p38 MAPK in mouse skin. Biofactors. 21:27–31. 2004.PubMed/NCBI View Article : Google Scholar

52 

Gugliandolo A, Silvestro S, Sindona C, Bramanti P and Mazzon E: MiRNA: Involvement of the MAPK Pathway in Ischemic Stroke. A Promising Therapeutic Target. Medicina (Kaunas). 57(1053)2021.PubMed/NCBI View Article : Google Scholar

53 

Safa A, Abak A, Shoorei H, Taheri M and Ghafouri-Fard S: MicroRNAs as regulators of ERK/MAPK pathway: A comprehensive review. Biomed Pharmacother. 132(110853)2020.PubMed/NCBI View Article : Google Scholar

54 

Yang M, He Y, Deng S, Xiao L, Tian M, Xin Y, Lu C, Zhao F and Gong Y: Mitochondrial Quality Control: A pathophysiological mechanism and therapeutic target for stroke. Front Mol Neurosci. 14(786099)2021.PubMed/NCBI View Article : Google Scholar

55 

Vongsfak J, Pratchayasakul W, Apaijai N, Vaniyapong T, Chattipakorn N and Chattipakorn SC: The alterations in mitochondrial dynamics following cerebral ischemia/reperfusion injury. Antioxidants (Basel). 10(1384)2021.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

April-2023
Volume 18 Issue 4

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Kongsui R and Kongsui R: Ameliorative effects of 6‑gingerol in cerebral ischemia are mediated via the activation of antioxidant and anti‑inflammatory pathways. Biomed Rep 18: 26, 2023
APA
Kongsui, R., & Kongsui, R. (2023). Ameliorative effects of 6‑gingerol in cerebral ischemia are mediated via the activation of antioxidant and anti‑inflammatory pathways. Biomedical Reports, 18, 26. https://doi.org/10.3892/br.2023.1608
MLA
Kongsui, R., Jittiwat, J."Ameliorative effects of 6‑gingerol in cerebral ischemia are mediated via the activation of antioxidant and anti‑inflammatory pathways". Biomedical Reports 18.4 (2023): 26.
Chicago
Kongsui, R., Jittiwat, J."Ameliorative effects of 6‑gingerol in cerebral ischemia are mediated via the activation of antioxidant and anti‑inflammatory pathways". Biomedical Reports 18, no. 4 (2023): 26. https://doi.org/10.3892/br.2023.1608