Effect of Gua Lou Gui Zhi decoction on focal cerebral ischemia‑reperfusion injury through regulating the expression of excitatory amino acids and their receptors

  • Authors:
    • Xianwen Chen
    • Huang Li
    • Mingqing Huang
    • Mei Huang
    • Wei Xu
    • Kedan Chu
    • Lidian Chen
    • Yuqin Zhang
  • View Affiliations

  • Published online on: April 28, 2014     https://doi.org/10.3892/mmr.2014.2189
  • Pages: 248-254
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Gua Lou Gui Zhi decotion (GLGZD) has been reported to be an effective treatment for post‑apoplectic limb spasm in the clinic. The present study aimed to investigate whether GLGZD had an affect on cerebral injuries induced by middle cerebral artery occlusion (MCAO) in rats and its possible mechanism. High‑performance liquid chromatography was performed to analyze GLGZD. Furthermore, a model was established to assess the efficacy of GLGZD. Neurological defect scores and screen tests were analyzed. Brain ischemic infarct volume was measured using 2,3,5‑triphenyl tetrazolium chloride staining and glutamic acid (Glu), aspartic acid (Asp) and glycine (Gly) levels in the cerebrospinal fluid were measured using the Hitachi automatic amino acid analyzer. Immunohistochemistry was performed to determine the expression of the α‑amino‑3‑hydroxy‑5‑methyl‑4‑isoxazole‑propionic acid (AMPA) and N‑methyl‑D‑aspartic acid (NMDA) glutamate receptors, and to analyze histopathological change. GLGZD was found to improve neurological performance and reduce infarct volumes in MCAO rats. In addition, GLGZD was observed to enhance motor performance, which was assessed using the screen test. Furthermore, GLGZD was found to reduce Glu, Asp and Gly levels in the cerebrospinal fluid and downregulate the protein expression of the AMPA and NMDA glutamate receptors. Thus, it was demonstrated that GLGZD may exert neuroprotective effects through the modulation of excitatory amino acids, and AMPA and NMDA receptor expression.

Introduction

Stroke, a cerebrovascular disease, is defined as a sudden neurological deficiency in the brain, which is caused by either hemorrhage or a lack of blood supply in the brain, termed ischemia. A total of 80% of strokes are caused by arterial blockage and consequent ischemia and 20% are caused by hemorrhage. At present, stroke represents one of the three most dangerous diseases threatening human health (1). It has been reported that 70–80% of stroke survivors have a level of disability, particularly post-apoplectic limb spasm, which has become an increasingly serious social problem (2). Post-apoplectic limb spasm is one of the key difficulties, which is faced during rehabilitation therapy following stroke, due to its high rate of motor impairment, difficulty of treatment and importance for limb function (3). The identification of effective therapeutic approaches to treat post-apoplectic limb spasm is important for stroke rehabilitation.

It is well established that excitotoxicity is the major pathophysiological mechanism underlying ischemic stroke-induced brain injury (4). Furthermore, excitatory amino acid (EAA)-induced excitotoxicity has been proposed to be involved in post-apoplectic limb spasm. Ischemia-reperfusion (I/R)-induced injury has been reported to be associated with numerous biochemical events, a number of which may result from the release of EAAs and the activation of their receptors (5). When EAAs reach toxic levels, they contribute to increased excitability, which leads to injury (6). The associated ion channels open and cause excessive Ca2+ influx, which stimulates a cascade of enzymatic processes, leading to irreversible injury (7). EAAs, including glutamic acid (Glu) and aspartic acid (Asp) are important neurotransmitters in the central nervous system (CNS) and have a major role in synaptic transmission. It is well established that EAAs are involved in the transmission of sensory information and motion instruction (8).

A large number of pharmacological agents have been used in attempt to improve motor impairment. These agents work through a variety of neurotransmitter- and receptor-associated mechanisms (9). A number of these agents have been used in clinical trials with varying results; however, many of these agents exhibit considerable side effects. Traditional Chinese medicine has advantages compared with these other pharmacological agents, including lower side effects, thus more regular doses may be taken. Traditional Chinese medicines, including Buyang Huanwu Decotion (10) and Xiao-Xu-Ming decotion (11), have been used for stroke and muscular therapy to treat post-apoplectic limb spasm with the advantages of easy administration and few adverse reactions.

Gua Lou Gui Zhi decotion (GLGZD) is a traditional Chinese medicine that consists of the following extracts: Radix Trichosanthis, Radix Paeoniae Alba, Ramulus Cinnamomi, Rhizoma Zingiberis Recens, Radix Glycyrrhizae and Fructus Ziziphi Jujubae. Clinical trials have found that GLGZD improves limb muscle tension, benefiting locomotive function and quality of life in patients with stroke (1214). However, the effect of GLGZD on the release of EAAs in the brain during I/R and the underlying mechanisms have yet to be elucidated.

Middle cerebral artery occlusion (MCAO) followed by reperfusion is a model of focal ischemia in rats, which resembles human ischemic stroke (15). The present study aimed to investigate the levels of EAAs and their receptors within the brain and whether GLGZD had a protective effect against I/R through modulating the release of EAAs and the expression of EEA receptors. Infarct volumes and neurological deficits were measured and analyzed in five groups.

Materials and methods

Chemicals, reagents and animals

Specific pathogen-free male Sprague-Dawley rats (weight, 180–220 g) were obtained from the Laboratory Animal Center of Fujian University of Traditional Chinese Medicine (Fuzhou, China). The principles of laboratory animal care were followed and the study was approved by the Ethics Committee of Fujian University of Traditional Chinese Medicine.

Dried drugs were purchased from Tongchun Drugstore (Fuzhou, China) and were identified by Professor Yang (College of Pharmacy, Fujian University of Traditional Chinese Medicine). Voucher specimens were deposited at the College of Pharmacy of Fujian University of Traditional Chinese Medicine. Peoniflorin, liquiritigenin, liquiritin, cinnamaldehyde, cinnamic acid and glycyrrhizin were purchased from the National Institute for the Control of Pharmaceutical and Biological Products (Beijing, China). Methanol and acetonitrile were of high performance liquid chromatography (HPLC)-grade and purchased from Merck KGaA (Darmstadt, Germany). Phosphoric acid, hydrochloric acid, petroleum, chloroform, acetoacetate and N-butanol were used as analytical reagents and were purchased from Aladdin Reagents Co., Ltd. (Shanghai, China). Other reagents were all of analytical grade. The deionized water used throughout the experiments was generated using a Millipore water purification system (Billerica, MA, USA). A CX31 microscope was purchased from Olympus Corporation (Tokyo, Japan).

Preparation of GLGZD

According to ‘Jinkui Yaolue’ (16), GLGZD consists of Radix Trichosanthis, Radix Paeoniae Alba, Ramulus Cinnamomi, Rhizoma Zingiberis Recens, Radix Glycyrrhizae and Fructus Ziziphi Jujubae. In order to obtain the GLGZD water extract, volatile oils were extracted from Ramulus Cinnamomi and Rhizoma Zingiberis Recens. Other medical materials were decocted through boiling in distilled water twice for 1 h. The solution was then dried under vacuum to obtain a final concentration of 1.08 g/ml. GLGZD was then stored for further analysis.

HPLC analysis of GLGZD

The GLGZD extract was subjected to HPLC analysis. Sample solutions were injected into the HPLC system (Shimadzu Corporation, Kyoto, Japan) for analysis in triplicate. The HPLC system was equipped with a LC-20A pump system, SPD-M20A photodiode array detector and Diamonsil® C18 reversed-phase column (I.D. 4.6×250 mm, 5 μm). Separation was achieved using a linear gradient program for mobile phase A (acetonitrile) and mobile phase B (water containing 0.1% phosphoric acid). The elution was initiated with a gradient of 95% B for 45 min, followed by 68% for 15 min and 52% for 5 min. Flow rate and injection volume were 1.0 ml/min and 10 μl, respectively.

Focal cerebral I/R model

An intraluminal suture method was used for the induction of focal cerebral ischemia. Rats were anesthetized using 10% chloral hydrate solution (0.3 ml/100 g body weight; intraperitoneal injection). MCAO was induced using an intraluminal suture method as described previously, but with certain modifications (15). In brief, the left common carotid artery (CCA) and the external carotid artery (ECA) were exposed. A 3-0 surgical monofilament nylon suture was then inserted from the ECA into the internal carotid artery (ICA) and was used to occlude the origin of the left MCA until light resistance was felt (18–20 mm from the CCA bifurcation). After 2 h of MCAO, the nylon suture was withdrawn, followed by 2 h of reperfusion.

Experimental grouping and treatment

Sixty rats were divided into the following five experimental groups and underwent the following treatments: Sham-operated group (n=12), the rats were subjected to surgical procedure, but MCAO was not induced, except for exposure of the right ICA and the right ECA; MCAO model group (n=12), rats received normal saline and underwent MCAO surgery; positive control group (n=12), rats received piracetam (6 g/kg body weight) and underwent MCAO surgery; GLGZD low-dose group (n=12), rats received GLGZD (3.6 g/kg body weight) and underwent MCAO surgery; and GLGZD high-dose group (n=12), rats received GLGZD (14.2 g/kg body weight) and underwent MCAO surgery. In the GLGZD treatment groups, GLGZD was administered once a day for seven days.

Scoring neurological defects

Rats were scored based on a five-point scale (17). The scale ratings were as follows: 0, no neurological symptoms; 1, unable to completely extend the front jaw on the other side; 2, rotating while crawling and falling to the contralateral side; 3, unable to walk without assistance; and 4, unconsciousness. Rats with a score of 1–3 were considered successful models and were included in the study. A sample of the ipsilateral cortex was taken at the indicated time and used for sample preparation.

Behavioral tests were performed on the rats after 60 min of ischemia followed by seven days of exercise. Each rat was scored based on a five-point scale as described previously (18).

Screen tests

In order to investigate motor function recovery in the rats subjected to ischemia, motor performance was measured prior to surgery and on days one and eight following surgery. In order to measure the muscle strength of the forelimbs, a net screen was used (19). The trial commenced subsequent to placing the rat on the horizontal screen on the ground. The screen was turned over 90° within 2 sec by raising one side gradually. The screen was maintained in this position for 5 sec.

The duration of time for which rats held on to the net screen was recorded in seconds. The scoring criterion was as follows: 5, holding on to the screen and climbing upward; 4, holding on to the screen with forelimbs and not falling down; 3, holding on to the screen temporally, but slipping a certain distance; 2, falling down to the ground within the test period; 1, falling down to the ground as soon as the screen was at 90°.

2,3,5-triphenyltetrazolium chloride (TTC) staining and measurement of brain ischemic infarct volume

The six rats in each group were sacrificed by chloral hydrate and decapitated in order to remove the brain to measure the infarct volume following I/R. The brain was placed at 20°C for ~10 min, then cut into six coronal slices continuously from front to back using a blade. The brain tissues were immersed into 2% TTC solution (T8877; Sigma-Aldrich, St. Louis, MO, USA) in phosphate-buffered saline (PBS; pH 7.4) and stained at 37°C for 1 h and turned over several times. Subsequent to staining, the viable cerebral tissue was stained red while the infarcted cerebral tissue remained pale. Images were captured using a high-resolution digital camera (IXUS130; Canon, Tokyo, Japan) and analyzed using Image-Pro Plus (Media Cybernetics, Inc., Rockville, MD, USA). Infarct volume was quantified using the Motic Med 6.0 Digital Medical Image Analysis system (Motic Instruments Inc., Richmond, Canada), The infarct size was calculated as a percentage of the viable cerebral tissue of the whole brain (20).

Analysis of Glu, Asp and glycine (Gly) levels in the cerebrospinal fluid

Cerebrospinal fluid was collected and the Glu, Asp and Gly levels were analyzed using a Hitachi automatic amino acid analyzer (21).

Cerebral histopathology

Brain samples were collected from six rats in each group for cerebral histopathological analysis. The brain samples were paraffin-embedded, sliced and stained with hematoxylin and eosin. Histopathological changes were observed using a light microscope.

Immunohistochemistry

Paraffin-embedded brain tissue samples (0.5×0.5×0.1 cm) were used for immunohistochemical analysis of N-methyl-D-aspartic acid receptor (NMDAR) and glutamate receptor (GluR) 1/2/3/4 expression. Briefly, the paraffin sections were dewaxed, repaired using citric acid, incubated with 3% H2O2, washed using PBS and blocked with normal goat serum. Sections were then incubated with primary antibodies (polyclonal rabbit Anti-NMDA-NR and Anti-GluR1/2/3/4; Beijing Biosynthesis Co., Ltd., Beijing, China) at room temperature for 2 h, washed using PBS and incubated with biotinylated secondary antibodies. Sections were then washed, incubated with horseradish peroxidase-labeled streptavidin and stained with 3,3′-diaminobenzidine. PBS was used to replace the primary antibody as a negative control. Five high-power fields (magnification, ×400) were randomly selected in each slide.

Statistical analysis

Data are presented as the mean ± standard deviation. Analysis of variance was performed to determine significant differences between the groups. SPSS 16.0 statistical software (SPSS, Inc., Chicago, IL, USA) was used for statistical analyses. P<0.05 was considered to indicate a statistically significant difference.

Results

HPLC of GLGZD

Various mobile phase systems were used to resolve the active constituents of GLGZD. The proposed method of analysis gave simultaneous quantification of six compounds. The individual constituents were identified by comparing their peaks, UV spectra and retention times against their corresponding reference standards (Fig. 1). The percentage content of each compound was estimated using a calibration curve. The compounds present in the GLGZD extract were peoniflorin (74.2 mg/g), liquiritigenin (0.92 mg/g), liquiritin (0.09 mg/g), cinnamic acid (0.20 mg/g), cinnamaldehyde (2.13 mg/g) and glycyrrhizic acid (1.14 mg/g).

Effect of GLGZD on infarct volume

As shown in Fig. 2, the infarct volume in the MCAO model group was found to be significantly higher than that in the sham-operated group. Furthermore, seven days following ischemia, infarct volumes were observed to be decreased in the GLGZD groups compared with the MCAO model group. The infarct volumes in the MCAO model group vs. the GLGZD groups were as follows: 0.77±0.13 vs. 0.29±0.12% for the GLGZD low-dose group and 0.77±0.13 vs. 0.18±0.12% (P<0.05) for the GLGZD high-dose group. Infarct volume was more greatly reduced in the GLGZD high-dose group than the GLGZD low-dose group. The infarct volumes in the MCAO model group vs. the piracetum group was 0.77±0.13 vs. 0.56±0.07% (P<0.05), showing that the results were significantly different between the two groups.

Effect of GLGZD on neurological behavior and motor performance

The effect of GLGZD on neurological and motor function were assessed by measuring neurological and motor performance. The induction of MCAO for 60 min followed by reperfusion for 2 h caused marked changes in rat behavior. The neurological behavior of the rats that were subjected to MCAO was measured using the scoring method described by Longa et al (15). Motor performance was measured using a screen test scoring method in order to investigate the motor function recovery of the rats subjected to MCAO.

As shown in Fig. 3A and B, the rats in the MCAO model group that were subjected to I/R injury, exhibited severe neurological deficit (score: 2) and motor disorder, and showed circling towards the contralateral side with a reduced mobility compared with the rats in the sham-operated group (score: 0). Rats in the GLGZD groups showed significant improvements in behavior between days 1 and 7, particularly those in the high-dose group. Rats in the piracetum group showed improvements in behavior between days 5 and 7, while no changes in neurological function or motor performance were observed in the rats in the sham-operated group.

Effect of GLGZD on levels of the EAAs Glu, Asp and Gly in the cerebrospinal fluid

The excessive release of EAAs in the brain following I/R injury is closely associated with post-apoplectic limb spasm. As shown in Table I, MCAO induced significant increases in the levels of the excitatory neurotansmitters Glu, Asp and Gly. The levels of Glu, Asp and Gly in the low- and high-dose GLGZD groups were lower than those in the MCAO model group, but higher than those in the sham-operated group. Similarly, compared with the MCAO model group, the levels of Glu, Asp and Gly in the piracetum group decreased and were significantly different (P<0.05). These findings show that GLGZD had a modulatory effect on EAA levels.

Table I

Levels of the excitatory amino acids Glu, Asp and Gly in the cerebrospinal fluid.

Table I

Levels of the excitatory amino acids Glu, Asp and Gly in the cerebrospinal fluid.

GroupAspGluGly
Sham-operation3.00±0.12.4.27±0.72.1.02±0.04.
MCAO model4.84±0.23a6.34±0.66a1.65±0.02a
Piracetam3.25±0.15b4.27±0.68b0.85±0.06b
GLGZD-L3.36±0.26.4.64±0.81b0.96±0.04.
GLGZD-H3.34±0.11b4.43±0.75b0.72±0.03b

a P<0.05, vs. the sham-operation group;

b P<0.05, vs. the MCAO model group.

{ label (or @symbol) needed for fn[@id='tfn3-mmr-10-01-0248'] } Glu, glutamic acid; ASP, aspartic acid; Gly, glycine; GLGZD, Gua Lou Gui Zhi decotion; L, low-dose; H, high-dose; MCAO, middle cerebral artery occlusion.

Cerebral histopathology

As shown in Fig. 4, the cortical neural cells in the sham-operated group were observed to be arranged orderly and to exhibit normal cell morphology, with clearly visible structures and integrity. The cell membranes and nuclei were normal, as was the tissue interspace, which exhibited no edema or inflammatory cell infiltration. The cortical region of the ischemic side of the brain in the rats in the MCAO model group exhibited visible disorder and high levels of cell necrosis, with cells showing nucleolar shrinkage and breakdown, as well as vacuolar degeneration. Furthermore, the mesenchymal cells demonstrated high levels of edema and inflammatory cell infiltration compared with those in the MCAO model group. The cortical neural cells in the rats in the GLGZD groups showed decreased pathological changes compared with those in the MCAO model group. Neuronal degeneration and necrosis, as well as disorderly cell arrangement, tissue edema, nuclear dissolution and nucleolar shrinkage were observed in the cells in the GLGZD treatment groups; however, the quantity and extent was decreased compared with that in the MCAO model group. Similarly, compared with the MCAO model group, the cortical neural cells in the rats in the piracetum groups exhibited reduced pathological changes

Immunohistochemistry

Semi-quantitative scoring was performed according to the ratio of positively stained cells and the staining intensity (22,23). Immunohistochemical scoring revealed a significant increase in NMDAR and GluR1/3/4, and a significant decrease in GluR2 in the hippocampus of the MCAO model rats compared with the sham rats (Fig. 5; Table II). After seven days of high-dose GLGZD administration, the scores were further reduced to 4.4±0.6 for NMDAR (P<0.05), 4.4±0.7 for GluR1 (P<0.05), 3.6±0.3 for GluR3 (P<0.01) and 3.6±0.7 for GluR4, while they were upregulated to 3.6±0.3 for GluR2 (P<0.01). For the piracetum groups, the scores were reduced to 4.0±0.1 for NMDAR (P<0.05), 4.2±0.6 for GluR1 (P<0.05), 3.6±0.4 for GluR3 (P<0.01) and 2.8±0.5 for GluR4, while they were upregulated to 4.0±0.0 for GluR2 (P<0.01). Alteration in the expression of NMDAR and GluR1/2/3/4 was neutralized with GLGZD treatment.

Table II

Immunohistochemical scores of NMDAR and GluR1/2/3/4 expression in brain tissue.

Table II

Immunohistochemical scores of NMDAR and GluR1/2/3/4 expression in brain tissue.

GroupNMDARGluR1GluR2GluR3GluR4
Sham-operation1.8±0.4.2.0±0.6.5.6±0.4.4.8±0.53.2±0.5
MCAO model5.6±0.2a5.2±0.7a1.6±0.6a5.6±0.44.0±0.9
Piracetam4.0±0.1b4.2±0.6b4.0±0.0c3.6±0.4c2.8±0.5
GLGZD-L4.8±0.5.4.6±0.1.3.2±0.4c4.4±0.74.4±0.7
GLGZD-H4.4±0.6b4.4±0.7b3.6±0.3c3.6±0.73.6±0.7

a P<0.01, vs. the sham-operation group;

b P<0.05 and

c P<0.01, vs. the MCAO model group.

{ label (or @symbol) needed for fn[@id='tfn7-mmr-10-01-0248'] } GluR, glutamate receptor; NMDAR, N-methyl-D-aspartic acid receptor; GLGZD, Gua Lou Gui Zhi decotion; L, low-dose; H, high-dose; MCAO, middle cerebral artery occlusion.

Discussion

Stroke often causes motor impairment, which may be defined as a velocity-dependent increase in resistance to passive stretch or movement, resulting in abnormal signaling between the CNS and the muscles, leading to muscle excitability (24). In the CNS, EAAs and their receptors have been shown to have a crucial role in motor impairment following cerebral I/R injury. Glu is the predominant excitatory neurotransmitter in the brain, and the subsequent activation of its receptors induces an increase in intracellular calcium and the generation of free radicals, leading to cell death, necrosis or apoptosis (25). Asp is present in high levels in the CNS within the hypothalamus and cerebellum, and is an excitatory neurotransmitter of intermediate neurons, which causes neuron depolarization and generates excitatory postsynaptic potential with slow and persistent function (26). It is well established that Gly has an important role in inhibitory synaptic transmission in the spinal cord and the brain stem. Furthermore, it has been reported that Gly is an important facilitator of glumate function in the cerebrum, through an allosteric site on the NMDA receptor (27). Therefore, Gly may have excitatory and inhibitory roles in the CNS.

Ionotropic glutamate receptors consist of three subclasses: NMDA, AMPA and kainate receptors, termed based on their characteristic agonists. AMPA and kainate receptors are often known as non-NMDA receptors. NMDA and non-NMDA glutamate receptors have a role in mediating motor impairment (28). NMDAR toxicity is dependent on extracellular Ca2+ and occurs as a consequence of high Ca2+ influx directly through the receptor-gated ion channel in the receptor (29). However, the majority of AMPA receptor (AMPAR) channels have poor Ca2+ permeability (30). AMPA-type GluRs are heteromeric channels comprised of a combination of GluR1-4 subunits (31), with changes in the expression of GluR1-4 being associated with development, aging, epilepsy and alterations in synaptic efficacy. The presence of the GluR2 subunit determines the permeability of the AMPAR to Ca2+ and other divalent cations. The majority of neurons exhibit high levels of GluR2 expression, which renders these cells impermeable to calcium influx thorugh AMPARs (32). Thus, AMPAR Ca2+ permeability is considered to be determined by the relative abundance of GluR2 subunits in the heteromeric AMPA receptors. It has been reported that ischemic conditions may alter the subunit composition of AMPA receptors towards a more Ca2+-permeable combination, which may promote neuronal degeneration (33).

GLGZD has been used for the treatment of post-apoplectic limb spasm. Yang et al (14) showed that GLGZD had a significant therapeutic effect on post-apoplectic limb spasm. The present study aimed to investigate the effect of GLGZD on focal I/R insult-induced motor impairment and behavioral disturbance, by inducing cerebral I/R injury in rats. GLGZD was found to improve neurological and motor performance. Furthermore, the present study identified a large infarct volume mainly in the cortex and striatum (or in the transient focal cerebral ischaemic brains) of the ipsilateral hemisphere of the brain in the ischemic rats. Pretreatment with GLGZD at high- and low-doses was observed to reduce the infarct volume compared with the sham-operated group. This finding suggests that oral administration of GLGZD extracts significantly inhibits reperfusion-induced neuronal cell death. In the present study, behavioral tests were used to assess the success of MCAO. Screen tests were used to assess behavioral dyskinesia caused by cerebral infarction. The findings suggest that GLGZD improves stroke-induced neurobehavioral abnormalities in rats.

In the present study, the quantity of Glu and Asp were found to increase markedly in the brain tissue of the rats in the MCAO model group, which indicates that the cytotoxicity of EAAs is involved in the establishment of ischemic brain injury and delayed nerve damage. The level of Gly in the brain tissue was observed to increase significantly in the MCAO model group, which may have a role in cerebral ischemia injury and a synergistic toxic effect with Glu. GLGZD treatment was found to inhibit the release of Glu and Asp during I/R.

Immunohistochemistry of the NMDAR revealed that the mice in the low- and high-dose GLGZD groups exhibited decreased positive NMDA expression, which may reduce excessive activation of the NMDA receptor, as well as reduce nerve cell cytotoxicity, necrosis and apoptosis. Immunohistochemistry of AMPAR subunits revealed that the the mice in the low- and high-dose GLGZD groups exhibited increased GluR2 expression, but decreased GluR1, 3 and 4 expression, which may prevent Ca2+ influx and have a protective effect against nerve injury. Thus, the neuroprotective effects of GLGZD may be partially due to its capacity to attenuate excitotoxic Glu and Asp accumulation. The effect of Gly and its receptors on cerebral I/R injury requires further investigation.

In conclusion, the present study established a HPLC method to qualitatively and quantitatively analyze the effect of GLGZD on EEAs and their receptors in the brains of MCAO model rats. The findings provides important insight into GLGZD as a neuroprotective agent.

Acknowledgements

This research was done in state key laboratory of Chinese pharmacies of Fujian Provincial Department of Science and Technology, Collaborative Innovation Center for Rehabilitation Technology and TCM Rehabilitation Research Center of SATCM. It was funded by the Important Subject of Fujian Province Science and technology hall of China (2012Y0041) and the important subject of Fujian province Education Hall of China (JA12176).

Abbreviations:

GLGZD

Gua Lou Gui Zhi decotion

MCAO

middle cerebral artery occlusion

CSF

cerebrospinal fluid

NMDA

N-methyl-D-aspartic acid

AMPA

α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid

TTC

2,3,5-triphenyl tetrazolium chloride

EAAs

excitatory amino acids

CNS

central nervous system

References

1 

Zeng MF, Pan LM, Zhu HX, Zhang QC and Guo LW: Comparative pharmacokinetics of baicalin in plasma after oral administration of Huang-Lian-Jie-Du-Tang or pure baicalin in MCAO and sham-operated rats. Fitoterapia. 81:490–496. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Riddoch MJ, Glyn WH and Bateman A: Stroke issues in recovery and rehabilitation. Physiotherapy. 81:689–694. 1995.

3 

Lamontagne A, Stephenson JL and Fung J: Physiological evaluation of gait disturbances post stroke. Clin Neurophysiol. 118:717–729. 2007. View Article : Google Scholar : PubMed/NCBI

4 

Wang HW, Liou KT, Wang YH, et al: Deciphering the neuroprotective mechanisms of Bu-yang Huan-wu decotion by an integrative neurofunctional and genomic approach in ischemic stroke mice. J Ethnopharmacol. 138:22–23. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Li C, Yan Z, Yang J, Chen H, Li H, Jiang Y and Zhang Z: Neuroprotective effects of resveratrol on ischemic injury mediated by modulating the release of neurotransmitter and neuromodulator in rats. Neurochem Int. 56:495–500. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Herz RC, Gaillard PJ, Wildt DJ and Versteeg DH: Differences in striatal extracellular amino acid concentrations between Wistar and Fischer 344 rats after middle cerebral artery occlusion. Brain Res. 715:163–171. 1996. View Article : Google Scholar

7 

Phillis JW and O’Regan MH: Characterization of modes of release of amino acids in the ischemic/reperfused rat cerebral cortex. Neurochem Int. 43:461–467. 2003. View Article : Google Scholar : PubMed/NCBI

8 

Doble A: The role of excitotoxicity in neurodenenerative disease: implications for therapy. Pharmacol Ther. 81:163–221. 1999. View Article : Google Scholar : PubMed/NCBI

9 

Van HA, Sarre S, Smolders I and Michotte Y: Determination of amino acids associated with cerebral ischaemia in rat brain microdialysates using narrowbore liquid chromatography and fluorescence detection. J Neurosci Methods. 144:63–71. 2005. View Article : Google Scholar

10 

Chern CM, Liao JF, Wang YH and Shen YC: Melatonin ameliorates neural function by promoting endogenous neurogenesis through the MT2 melatonin receptor in ischemic-stroke mice. Free Radic Biol Med. 52:1634–1647. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Zhu XH, Li SJ, Hu HH, Sun LR, Das M and Gao TM: Neuroprotective effects of Xiao-Xu-Ming decotion against ischemic neuronal injury in vivo and in vitro. J Ethnopharmacol. 37:38–46. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Sun X: Research on formula treating paralysis and spasticity from ‘treatise on febrile and miscellaneous diseases’. Zhongguo Zhong Yi Ji Chu Yi Xue Za Zhi. 8:644–645. 2010.(In Chinese).

13 

Zhang L and Ai H: Effects of Gua Lou Gui Zhi decotion on c-fos and c-jun in epileptic rats. Sichuan Hua xi Zhong Yi Yao Yan Jiu Suo. 23:21–22. 2005.(In Chinese).

14 

Yang C, Chen L and Tao J: New usage of a classical formula-Gua Lou Gui Zhi decotion. Liaoning Zhong Yi Za Zhi. 8:166–167. 2012.(In Chinese).

15 

Longa EZ, Weinstein PR, Carlson S and Cummins R: Reversible middle cerebral artery occlusion without craniectomy in rats. Stroke. 20:84–91. 1989. View Article : Google Scholar : PubMed/NCBI

16 

Zhang Z: Jinkui Yaolue. Lin Y, Yang P, Hou X and Yang Y: Synopsis of Golden Chamber. Macmillan Press; Beijing: pp. 203–204. 2008, (In Chinese).

17 

Guo J, Liu L, Ma C, Xu B, Duan X and Wang B: Effect of restraint stress on depression-like behaviors in rats after transient focal cerebral ischemic injury. Neural Regen Res. 2:390–394. 2007. View Article : Google Scholar

18 

Yin J, Tu C, Zhao J, Ou D, Chen G, Liu Y and Xiao X: Exogenous hydrogen sulfide protects against global cerebral ischemia/reperfusion injury viaitsanti-oxidative, anti-inflammatory and anti-apoptotic effects in rats. Brain Res. 1491:188–196. 2013. View Article : Google Scholar

19 

Benedek A, Moricz K, Juranyi Z, Gigler G, Levay G, Harsing LG Jr, Matyus P, Szénási G and Albert M: Use of TTC staining for the evaluation of tissue injury in the early phases of reperfusion after focal cerebral ischemia in rats. Brain Res. 1116:159–165. 2006. View Article : Google Scholar : PubMed/NCBI

20 

Gao WJ, Qian T and Cong B: Effect of Yishen Jiangzhuo Decotion on the EAA content of hippocampus in cerebral ischemia reperfusion rats. Chin J Intergrative Med Cardio/Cerebrovasc Dis. 1:319–321. 2003.

21 

Guo C, Tong L, Xi MM, Yang HF, Dong H and Wen AD: Neuroprotective effect of calycosin on cerebral ischemia and reperfusion injury in rats. J Ethnopharmacol. 144:768–774. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Soslow RA, Dannenberg AJ, Rush D, Woerner BM, Khan KN, Masferrer J and Koki AT: Cox-2 is expressed in human pulmonary, colonic, and mammary tumors. Cancer. 89:2637–2645. 2000. View Article : Google Scholar : PubMed/NCBI

23 

Zhang Y, Xu W, Li H, Zhang X, Xia Y, Chu K and Chen L: Therapeutic effects of total alkaloids of Tripterygium wilfordii Hook f. on collagen-induced arthritis in rats. J Ethnopharmacol. 45:699–705. 2013.

24 

Pisano F, Miscio G, Del Conte C, Pianca D, Candeloro E and Colombo R: Quantitative measures of spasticity in post-stroke patients. Clin Neurophysiol. 111:1015–1022. 2000. View Article : Google Scholar : PubMed/NCBI

25 

Sucher NJ: Insights from molecular investigations of traditional Chinese herbal stroke medicines: implications for neuroprotective epilepsy therapy. Epilepsy Behav. 8:350–362. 2006. View Article : Google Scholar

26 

D’Aniello A: D-Aspartic acid: an endogenous amino acid with an important neuroendocrine role. Brain Res Rev. 53:215–234. 2007.PubMed/NCBI

27 

Yang ZZ, Li J, Li SX, Feng W and Wang H: Effect of ginkgolide B on striatal extracellular amino acids in middle cerebral artery occluded rats. J Ethnopharmacol. 136:117–122. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Kocsis P, Tarnawa I, Szombathelyi Z and Farkas S: Participation of AMPA-and NMDA-type excitatory amino acid receptors in the spinal reflex transmission, in rat. Brain Res Bull. 60:81–91. 2003. View Article : Google Scholar : PubMed/NCBI

29 

Tu W, Xu X, Peng L, et al: DAPK1 Interaction with NMDA receptor NR2B subunits mediates brain damage in stroke. Cell. 140:222–234. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Xiong ZG, Zhu XM, Chu XP, et al: Neuroprotection in ischemia: blocking calcium-permeable acid-sensing ion channels. Cell. 118:687–698. 2004. View Article : Google Scholar : PubMed/NCBI

31 

Kamphuis W, Klooster J and Dijk F: Expression of AMPA-type glutamate receptor subunit (GluR2) in ON-bipolar neurons in the rat retina. J Comp Neurol. 455:172–186. 2003. View Article : Google Scholar : PubMed/NCBI

32 

Dijk F and Kamphuis W: Ischemia-induced alterations of AMPA-type glutamate receptor subunit expression patterns in the rat retina - an immunocytochemical study. Brain Res. 997:207–221. 2004. View Article : Google Scholar : PubMed/NCBI

33 

Cebers G, Cebere A and Liljequist S: Metabolic inhibition potentiates AMPA-induced Ca2+ fluxes and neurotoxicity in rat cerebellar granule cells. Brain Res. 779:194–204. 1998. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2014
Volume 10 Issue 1

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen X, Li H, Huang M, Huang M, Xu W, Chu K, Chen L and Zhang Y: Effect of Gua Lou Gui Zhi decoction on focal cerebral ischemia‑reperfusion injury through regulating the expression of excitatory amino acids and their receptors. Mol Med Rep 10: 248-254, 2014
APA
Chen, X., Li, H., Huang, M., Huang, M., Xu, W., Chu, K. ... Zhang, Y. (2014). Effect of Gua Lou Gui Zhi decoction on focal cerebral ischemia‑reperfusion injury through regulating the expression of excitatory amino acids and their receptors. Molecular Medicine Reports, 10, 248-254. https://doi.org/10.3892/mmr.2014.2189
MLA
Chen, X., Li, H., Huang, M., Huang, M., Xu, W., Chu, K., Chen, L., Zhang, Y."Effect of Gua Lou Gui Zhi decoction on focal cerebral ischemia‑reperfusion injury through regulating the expression of excitatory amino acids and their receptors". Molecular Medicine Reports 10.1 (2014): 248-254.
Chicago
Chen, X., Li, H., Huang, M., Huang, M., Xu, W., Chu, K., Chen, L., Zhang, Y."Effect of Gua Lou Gui Zhi decoction on focal cerebral ischemia‑reperfusion injury through regulating the expression of excitatory amino acids and their receptors". Molecular Medicine Reports 10, no. 1 (2014): 248-254. https://doi.org/10.3892/mmr.2014.2189