ROCK inhibitor Y‑27632 protects rats against cerebral ischemia/reperfusion‑induced behavioral deficits and hippocampal damage

  • Authors:
    • Lihe Li
    • Baoyang Liu
  • View Affiliations

  • Published online on: August 9, 2019     https://doi.org/10.3892/mmr.2019.10584
  • Pages: 3395-3405
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cerebral ischemic injury is a major cause of death and long‑term disability worldwide that leads to neurological and behavioral deficits, and for which successful treatments are still lacking. Ras homolog family member A (RhoA) and Rho‑associated coiled‑coil containing protein kinase (ROCK) are associated with the growth of neurons and the movement of neuronal growth cones. RhoA/ROCK inhibitors have been demonstrated to promote the recovery of motor function following nerve injury, but the underlying mechanism requires further investigation. The present study aimed to investigate the effects of the ROCK inhibitor Y‑27632 on middle cerebral artery occlusion (MCAO)‑induced cerebral ischemic injury. Rats were randomly assigned to the Control, Y‑27632, MCAO + Vehicle or MCAO + Y‑27632 group. Firstly, infarct volume, cognitive ability and cerebral injury were assessed. Secondly, indicators of cerebral inflammation, oxidative stress and apoptosis were evaluated. Finally, the expression of recombinant glial fibrillary acidic protein (GFAP) and allograft inflammatory factor 1 (AIF1) in the brain were measured to assess the activation of astrocytes and microglia, respectively. The results showed that Y‑27632 effectively increased the survival rate and behavioral performance of rats, and attenuated the cerebral injury, oxidative stress and cerebral inflammation levels following MCAO. The disturbance in hippocampal neurons caused by MCAO was also alleviated following treatment with Y‑27632. Neuronal apoptosis was also decreased following Y‑27632 treatment, as demonstrated by the TUNEL assay and the expression levels of Caspases‑3, 8 and 9 and Bax/Bcl‑2 ratio. The levels of GFAP and AIF1 were increased by MCAO and further promoted by Y‑27632, indicating the activation of astrocytes and microglia. In conclusion, the present study offered evidence of a protective effect of Y‑27632 administration on cerebral ischemia/reperfusion induced behavioral and hippocampal damage by activating astrocytes and microglia.

Introduction

Cerebrovascular disease is becoming a prominent public health concern. Due to its high rates of disability, morbidity and mortality, cerebrovascular disease not only has a strong impact on a patient's quality of life, but is also accompanied by heavy economic burdens for the patients' families and society (1). Currently, treatment of cerebrovascular disease consists in the surgical removal of blocked blood vessels in order to improve the blood supply in and around the lesion site (2). Once the blood supply improves, the infarcted area is prone to develop severe cerebral ischemia/reperfusion (CIR) injury. There are several pathological mechanisms involved in the process of CIR damage, including glutamate-induced excitotoxicity, loss of ionic homeostasis, energy failure, inflammatory response, increased oxidative stress and apoptosis (3). The accumulation of reactive oxygen species (ROS) in cells can directly lead to cell necrosis and indirectly induce cell senescence, apoptosis and necrosis by activating oxidative signaling pathways (4). Local excessive inflammatory reactions after cerebral ischemia-reperfusion also cause tissue damage. In recent years, calcium ion antagonist, radical scavengers and other neuroprotective drugs have been developed and used for the treatment of CIR injury (5). However, the possible side effects of these treatments, including drug resistance, cerebral hemorrhage and gastrointestinal irritation, may exceed the clinical benefits of long-term therapy.

Ras homolog family member A (RhoA) has GTPase activity and acts as a signal transducer or as a molecular switch in cell signaling pathways. It may also act on the cytoskeleton and regulates the response of actin to extracellular signals. Rho associated coiled-coil containing protein kinase (ROCK) can catalyze the conversion of inactive RhoA to active RhoA (6). In neurons, activation of RhoA leads to axonal retraction; conversely, inactivation of RhoA or ROCK promotes the growth of neurons and the movement of neuron growth cones. Therefore, RhoA/ROCK pathway is closely associated with the growth of neuron and their axons (7). Y-27632 is a type of synthetic pyridine complex that can be introduced in cells via vectors. Y-27632 is a cell-permeable, highly potent and selective inhibitor of Rho-associated, coiled-coil containing protein kinase (ROCK). It binds intracellularly to the catalytic site of the upstream effector of the Rho protein ROCK (both ROCK1 and 2), thereby inhibiting its kinase activity. Y-27632 inhibits both ROCK1 (Ki=220 nM) and ROCK2 (Ki=300 nM) by competing with ATP for binding to the catalytic site. Y-27632 has been shown to inhibit ROCK kinase activity in epithelial cells, endothelial cells, smooth muscle cells and neurons (8). It has also been found that Y-27632 promotes neurite outgrowth in PC12 cells (9). Y-27632 has also been used in treatments following spinal cord injury, for it is known to promote neurite outgrowth and axonal regeneration in neurons (10). However, the effect of Y-27632 on CIR injury needs to be studied. IR injury is also known to stimulate the activation and proliferation of astrocytes and microglia to protect brain tissues (11,12). Glial fibrillary acidic protein (GFAP), found in astrocytes and ependymal cells, is a type of cellular microfilament with multiple functions in cell activity, and its expression may be affected following CIR.

Therefore, the present study, aimed to investigate the effects of the ROCK inhibitor Y-27632 on CIR injury and to explore its underlying mechanism of action. The cognitive ability, cerebral infarct, inflammation, oxidative stress and apoptosis in rats with middle cerebral artery occlusion (MCAO) were used to evaluate the effect of Y-27632 on CIR. The levels GFAP and allograft inflammatory factor 1 (AIF1) in neurons were assessed by western blotting to evaluate the activation of astrocytes and microglia in the context of CIR.

Materials and methods

Animals

A total of 310 male Sprague-Dawley rats, weighing 230–250 g, were purchased from Tianjin Medical University animal center. Animals were housed at 25°C with a 12-h day/night cycle in the Animal Center of Baodi District People's Hospital with food and water available ad libitum. The procedures of the present study followed international guidelines of animal care (NIH publication No. 92-3415, revised 1999) and were approved by the Animal Care Committee in Baodi District People's Hospital.

Experimental protocol

The experimental protocol is summarized in Fig. 1. For biochemical evaluation, hematoxylin and eosin (H&E) staining, western blotting and behavioral assessment, 80 rats were randomly grouped into Control, Y-27632, MCAO + Vehicle and MCAO + Y-27632 groups, with 20 animals in each group. Rats in the Control group were operated on like the experimental MCAO ones, but the nylon filament was not introduced to the carotid artery. Rats in the Y-27632 group received Y-27632 injection and the same operation as Control. Rats in MCAO + Vehicle group received MCAO surgery and were injected with saline 120 min later. Rats in MCAO + Y-27632 group received MCAO surgery and were injected with Y-27632 120 min later. After MCAO, animals were allowed to rest for 24 h. Then, half the animals in each group were sacrificed and used in for biochemical, H&E and western blot analyses, while the remaining half was used for neurological deficit scoring. After 48 h, these animals were tested in the water maze. For survival rate studies, 64 rats were randomly grouped into the Control group (n=12), Y-27632 group (n=12), MCAO + Vehicle group (n=20) and MCAO + Y-27632 group (n=20). Survival checks were performed at 24, 48 h and 7 days after MCAO surgery. The animal health and behavior were monitored twice a day during the experiments. Survival checks were performed at 1, 2 and 7 days following MCAO surgery; the inability of rats to right themselves in 30 sec after being placed on their side was considered a humane endpoint and were euthanized by overdose of sodium pentobarbital (150 mg/kg). All Rats were euthanatized with intraperitoneal injection of an overdose of sodium pentobarbital (150 mg/kg) at the end of the study. Death was verified by checking the heartbeat. Rats would be euthanatized if serious infection occurred and remained uncontrollable. However, no rat was euthanized prior to the end of the study due to ill health.

MCAO procedure

The MCAO procedure was similar to that of Yang et al (13). Briefly, rats were anesthetized with ketamine (100 mg/kg, intra-muscularly) and xylazine (7.5 mg/kg, intra-muscularly) and mounted on a wooden plate. A 3 cm long incision was made in the center of neck to expose the common carotid and external carotid arteries. After these two arteries were ligated, a nylon filament (diameter, 0.25 mm) was inserted into the internal carotid artery and advanced for 20 mm. MCAO lasted for 60 min to generate ischemia, and the nylon filament was pulled out to allow reperfusion of the injured site. The body temperature of animals was kept at 37±1°C with a heating blanket during the IR process.

Intracerebroventricular injection

After 30 min since reperfusion, rats were anesthetized with ketamine (100 mg/kg, intra-muscularly) and xylazine (7.5 mg/kg, intra-muscularly). After the head of each rat was mounted in a stereotaxic instrument, the skull was revealed by a midline incision. A craniotomy was drilled in the right lateral cerebral ventricle at the following stereotaxic coordinates relative to the bregma: 1.00 mm anteroposterior, 1.5 mm mediolateral and 2.00 mm dorsoventral. In the MCAO + Vehicle group, 10 µl artificial cerebrospinal fluid was injected into the right lateral cerebral ventricle with a microinjector. In the Y-27632 and MCAO + Y-27632 groups, 10 µl Y-27632 (10−6 mol/l) was injected in the same manner. Afterwards, the incision was closed with a wound clip. Ampicillin (20 mg/kg, twice per day) and meloxicam (0.2 mg/kg, once per day) were injected intra-muscularly to prevent wound infection and pain.

2,3,5-Triphenyltetrazolium chloride (TTC) staining

Following experiments, the brains of animals were collected for histological analysis. After 20 min freezing at −20°C, 5 to 6–1.7 mm thick coronal sections were obtained and placed in 2% TTC phosphate buffer solution (23.9 mM TTC in 0.05 M phosphate buffer solution, pH 7.5). These sections were stained at 37°C for 30 min in the dark, and then images were capture with a digital camera. The normal tissue was stained red, while the infarcted tissue exhibited white coloration. The picture analyzed using a pathological image analysis computer system, and the infarct size of each layer was measured to calculate the infarct volume.

Neurological deficit scoring

Neurological deficit scoring was performed as previously described (14). At 48 h after MCAO, the potential deficit points of animals were evaluated as follows: no nerve injury symptom, 0 points; contralateral papillary flexion, 1 point; reduced contralateral forepaw clenched grip, 2 points; spontaneous movement in all directions and circle to the opposite side when pulling their tails, 3 points; loss of consciousness, 4 points.

Water maze study

The water maze study was performed at 48 h after MCAO following the methodology described by Ahmadi et al (15). Briefly, a black circular tank (120 cm diameter, 80 cm high) was divided into four equal quadrants and used as the water maze. The depth and the temperature of water were kept at 30 cm and 25±1°C, respectively. A circular platform was placed 1.5 cm under the water in a quadrant. A video camera was used to track the travel path and duration of rats. Beginning at 48 h after MCAO, the animals were trained for five consecutive days with the platform located in the same position. The escape latency, traveled length and mean speed of animals were calculated from the recorded video. There was no significant difference between groups in the mean speed, indicating that physical deficit was not caused by the MCAO procedure. On the sixth day, the platform was removed. The rats were allowed to swim in the water maze for 60 s, and the number of times they crossed the platform location (crosses) and the duration they spent in the target quadrant were recorded by the video camera.

Measurement of oxidative and pro-inflammatory factors in the brain tissue

At 24 h after MCAO, animals were sacrificed and brains were harvested and sliced into 1.0 mm thick slices. A total 100 mg brain tissue of the infarcted area was collected and homogenized in PBS (10 mM, pH=7.4). Next, they were centrifuged at 12,000 × g at 4°C for 30 min before the supernatant was collected. The levels of oxidative markers [malondialdehyde (MDA), (8-hydroxy-2-deoxyguanosine (8-OHdG), 3-nitrotyrosine (3-NT)] and pro-inflammatory factors [tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6] in the supernatant were measured using corresponding assay kits (Sigma-Aldrich; Merck KGaA), following the manufacturer's protocol. Their concentration was calculated with absorbance values which were detected by a microplate reader (Azure Biosystems, Inc.). The protein levels were measured with a protein assay kit (Nanjing Jiancheng Bioengineering Institute) using the bicinchoninic acid method. All the concentrations were normalized to the protein level.

Hematoxylin and eosin (H&E) staining

A total of 24 h following MCAO, rats were anesthetized with ketamine (100 mg/kg, intra-muscularly) and xylazine (7.5 mg/kg, intra-muscularly) and perfused with cold saline containing heparin (40 mg/l) and 4% paraformaldehyde. After the rats were fixed on a wooden plate, the needle used for perfusion was inserted into the aortic root of the rats from the left apex. After the needle was fixed, the perfusion with cold saline containing heparin started. When no blood was seen in the perfusion solution, which indicated that the blood in the animal was completely replaced by cold saline containing heparin, the perfusion solution was changed to 4% paraformaldehyde. 1 h later, the brains were removed, fixed in 4% paraformaldehyde and embedded in paraffin at room temperature. Coronal sections (6 µm thick) were taken every millimeter from −1.3 to −6.3 mm relative to the bregma. Hippocampal sections of the ipsilateral side of the MCAO from eight rats in each group were collected and stained with H&E at room temperature for 5 min to analyze the neuronal injury. The numbers of nucleoli and cytoplasmic Nissl bodies and pyknosis-positive neurons were compared within groups. A VS120 Virtual Slide Microscope (Olympus Corporation) and cellSens Imaging Software (version 2.2; Olympus Corporation) were used to capture and analyze the images.

Immunohistochemical (IHC) analysis

IHC analysis of neuron specific enolase (NSE), a marker of cerebral damage, was performed to evaluate CIR. Hippocampal sections of the ipsilateral side of the MCAO from eight rats in each group were collected. Dewaxing and hydration were first carried out with xylene and alcohol. Endogenous peroxidase activity was then blocked by the addition of 0.3% hydrogen peroxide at room temperature for 30 min, and antigen retrieval was performed using 0.01 M citrate buffer. Next, non-specific antibodies were blocked by incubation with 5% goat serum (Beyotime Institute of Biotechnology) at 37°C for 45 min. They were then incubated with primary antibody against NSE (cat. no. SAB4500768; 1:1,000; Sigma-Aldrich; Merck KGaA) at 4°C overnight and with the goat anti-rabbit IgG secondary antibody conjugated to biotin (cat. no. SAB4600006, 1:5,000, Sigma-Aldrich; Merck KGaA) at 37°C for 1 h. Afterwards, the sections were washed with PBS three times and incubated with streptavidin conjugated to horseradish peroxidase (cat. no. 18-152, 10 µg/ml, Sigma-Aldrich; Merck KGaA) at room temperature for 1 h. The color was developed using DAB coloring solution, then counterstained with hematoxylin at room temperature for 10 min. Finally, the slices were dehydrated and sealed with xylene and alcohol. Representative images were taken from the hippocampal region in a total of four different sections per animal using a VS120 Virtual Slide Microscope (Olympus Corporation) at a magnification of ×200. The number of immunoreactive signals of NSE was counted in four randomly selected fields per section.

Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining

Hippocampal sections of the ipsilateral side of the MCAO from eight rats in each group were collected. Dewaxing and hydration were first carried out with xylene and alcohol. Afterwards, the paraffin sections were digested with 20 µg/ml proteinase K (Sigma-Aldrich; Merck KGaA) at room temperature for 10 min. Next, they were incubated with TUNEL assay solution (Promega Corporation) at 37°C for 90 min, according to the manufacturer's protocol. Finally, representative images were taken from the hippocampal region in a total of four different sections per animal using a VS120 Virtual Slide Microscope (Olympus Corporation) at a magnification of ×200. Five randomly selected fields in each image were examined. The brown colored apoptotic cells were identified as ‘TUNEL positive cells’ and the percentage of the TUNEL positive cells were calculated.

Western blotting

A total of 100 mg infarcted cerebral cortex tissue was cut into small pieces, and 1 ml RIPA lysis buffer (Beyotime Institute of Biotechnology) and 1 ml PMSF (final concentration, 1 mM; Beyotime Institute of Biotechnology) were added to the tissue, which was then homogenized until fully lysed. After centrifugation at 30,000 × g for 3 min at 4°C, the supernatant was collected. The protein concentration in supernatants was determined using a standard BCA protein assay kit (Beyotime Institute of Biotechnology), then 40 µg protein in the supernatant was transferred to polyvinylidene difluoride (PVDF) membranes using Mini-Protean Tetra Electrophoresis System (Bio-Rad Laboratories, Inc.). Afterwards, PVDF membranes were blocked in 5% skimmed milk at room temperature for 2 h. They were incubated with primary antibodies against Caspase-3 (cat. no. sc-271759; 1:1,000; Santa Cruz Biotechnology, Inc.), Caspase-8 (cat. no. sc-5263; 1:1,000; Santa Cruz Biotechnology, Inc.), Caspase-9 (cat. no. sc-133109; 1:1,000; Santa Cruz Biotechnology, Inc.), Bax (sc-20067; 1:1,000; Santa Cruz Biotechnology, Inc.), Bcl-2 (cat. no. sc-56015; 1:1,000; Santa Cruz Biotechnology, Inc.), GFAP (cat. no. sc-33673; 1:1,000, Santa Cruz Biotechnology, Inc.) and AIF1 (cat. no. sc-32725; 1:1,000; Santa Cruz Biotechnology, Inc.) at 4°C overnight. The next day, they were incubated with a horseradish peroxidase-conjugated secondary antibody (cat. no. G-21040; 1:10,000; Thermo Fisher Scientific, Inc.) at 37°C for 2 h. The enhanced chemiluminescent substrate was applied to the blot membrane and incubated at room temperature for 5 min. The blot membrane was then placed in a chemiluminescence imager (Bio-Rad Laboratories, Inc.) for image acquisition, and densitometry analysis was performed using ImageJ 1.43 software (National Institutes of Health).

Statistical analysis

Statistical analysis was performed using two-way analysis of variance method followed by Turkey post hoc test with SPSS software 17.0 (SPSS, Inc.). Data were represented as the mean ± standard error of the mean. The number of experimental repeats was 12. P<0.05 was considered to indicate a statistically significant difference.

Results

Treatment with the ROCK inhibitor increases survival following MCAO

Survival checks were performed at 24 and 48 h and 7 days after MCAO surgery. As shown in Table I, the survival rates at 24 and 48 h and 7 days in Control and Y-27632 groups were 100%, suggesting that Y-27632 alone did not affect overall survival. In the MCAO + Vehicle group, the survival rates at 24 and 48 h and 7 days were 60, 50 and 35%, respectively, with only seven animals surviving until the seventh day after the MCAO surgery. In the MCAO + Y-27632 group, however, the survival rates at 24, 48 h and 7 days were significantly higher at 90, 80 and 50%, respectively, with 10 rats surviving until the seventh day after the MCAO surgery.

Table I.

Effect of Y-27632 on rat survival rate.

Table I.

Effect of Y-27632 on rat survival rate.

Survival

24 h48 h7 days



n%n%N%
Control12/1210012/1210012/12100
Y-2763212/1210012/1210012/12100
MCAO+Vehicle12/20a6010/20a507/20a35
MCAO+Y-2763218/20b9016/20b8010/20b50

{ label (or @symbol) needed for fn[@id='tfn1-mmr-20-04-3395'] } n, number of survivors/number of rats in the group. MCAO, middle cerebral artery occlusion.

a P<0.05 compared to Control

b P<0.05 compared to MCAO+Vehicle.

Y-27632 reduces the MCAO-induced infarct volume in the brain

To assess the effect of ROCK inhibitor Y-27632 on the infarct caused by MCAO, the infarcted brain area was stained with TTC and compared between groups. As shown in Fig. 2, the infarct volume in the MCAO group was 31.5±3.6%, MCAO + Vehicle group was 28.3±5.1%, while the infarct volume in the MCAO + Y-27632 group was 18.2±3.3%. There was no significant difference between MCAO group and MCAO + Vehicle group. The infarct size difference between MCAO + Vehicle and MCAO + Y-27632 group was statistically significant (P<0.05).

Y-27632 prevents neurological impairments caused by CIR

As shown in Fig. 3A, MCAO induced significant neurological deficits. The deficit score in the MCAO + Vehicle group was 3.1±0.23 (P<0.05 compared to Control). There was no significant difference between MCAO group and MCAO + Vehicle group. However, the application of the ROCK inhibitor Y-27632 significantly decreased the neurological deficit score to 2.2±0.31 (P<0.05 compared to MCAO + Vehicle). The parameters evaluated with the water maze test are presented in Fig. 3B-F. The mean swimming speed (Fig. 3B) was not different between these four groups, suggesting that the motor ability of rats were not altered by MCAO or Y-27632. Following treatment with Y-27632, the escape latency and travel length during training days (Fig. 3C and D) were significantly decreased compared to MCAO + Vehicle (P<0.05). Rats in MCAO + Vehicle group on the test day (sixth day; Fig. 3E) crossed the hidden platform significantly less, while Y-27632 treated animals significantly increased the number of crosses (P<0.05 compared to MCAO + Vehicle). There was no significant difference between MCAO group and MCAO + Vehicle group. The time rats spent in target quadrant on the test day (sixth day; Fig. 3F) was significantly longer in the MCAO + Y-27632 group compared with the MCAO + Vehicle group (P<0.05). Y-27632 alone did not affect all these behavior test results (P>0.05 compared to Control).

ROCK inhibitor attenuates the histological changes caused by CIR

The histological changes to the hippocampus across the four groups are shown in Fig. 4. In the MCAO and MCAO + Vehicle groups, the brain tissue became less compact, the extracellular space was widened. In the MCAO + Y-27632 group, these observations were significantly less common.

Treatment with the ROCK inhibitor attenuates the oxidative injury and inflammation

The changes in oxidative injury indicators across groups are displayed in Fig. 5A-C. The levels of MDA and 3-NT (Fig. 5A and C) in the MCAO + Vehicle group were greatly increased compared to Control, and were significantly inhibited by co-treatment with Y-27632. The levels of 8-OHdG (Fig. 5B) was greatly increased compared to Control, but not changed by Y-27632. The changes in inflammatory marker expression across groups are shown in Fig. 5D-F. The levels of TNF-α and IL-1β (Fig. 5D and E) in the MCAO + Vehicle group were significantly increased compared to Control, and were inhibited by co-treatment with Y-27632 (P<0.05 compared to MCAO + Vehicle). The IL-6 levels (Fig. 5F) were also increased compared to Control, but not significantly changed by Y-27632. Y-27632 alone did not affect oxidative injury or inflammation (P>0.05 compared to Control).

Y-27632 attenuates the expression of NSE in hippocampus tissue

The expression of NSE, a marker of CIR-induced damage, in the hippocampus of Control, Y-27632, MCAO + Vehicle and MCAO + Y-27632 groups is shown in Fig. 6. Representative images of IHC staining for NSE are shown in Fig. 6A, while Fig. 6B shows the quantification results of NSE expression. The results indicated that the expression of NSE was significantly increased following MCAO, but inhibited by the treatment with Y-27632 (P<0.05 compared to MCAO + Vehicle).

Y-27632 attenuates neuron apoptosis caused by CIR

To analyze the effect of ROCK inhibition on neuron apoptosis caused by CIR, the apoptosis rate in the hippocampus tissue and the protein levels of Caspase-3, Caspase-8 and Caspase-9 and the Bax/Bcl-2 ratio in the infarcted brain tissues were evaluated (Fig. 7). Representative images of the TUNEL assay are shown in Fig. 7A, while Fig. 7B indicates the quantification of apoptotic numbers in the hippocampus. The rate of apoptosis was greatly increased by MCAO, but reduced with Y-27632 treatment (P<0.05 compared to MCAO + Vehicle). Y-27632 alone did not change the rate of apoptosis. As illustrated in Fig. 8, the protein levels of Caspase-3, Caspase-8 and Caspase-9 were also increased following MCAO, but reduced with Y-27632 treatment. Furthermore, Y-27632 significantly decreased the Bax/Bcl-2 ratio in infarcted brain tissues. Y-27632 alone did not affect neuron apoptosis (P>0.05 compared to Control).

Y-27632 increases the levels of GFAP and AIF1 following MCAO

To explore the effect of ROCK inhibition on astrocytes and microglia function, the levels of GFAP and AIF1 were evaluated in infarcted brain tissues by western blotting. Representative images of western blotting for GFAP and the quantification of its expression are shown in Fig. 9A, while Fig. 9B presents representative images of AIF1 western blot analysisAIF1 and respective quantification of its expression. The results demonstrated that the protein levels of GFAP and AIF1 were both increased by MCAO, and the treatment of Y-27632 further increased these protein levels to a higher level following MCAO. Y-27632 alone did not affect GFAP and AIF1 protein levels (P>0.05 compared to Control).

Discussion

The present study investigated the effects of the ROCK inhibitor Y-27632 on CIR injury and explored its underlying mechanism of action. It demonstrated that ROCK inhibition by Y-27632 significantly improved the cognitive ability that was impaired following CIR damage. Y-27632 also reduced the expression of oxidative injury and inflammation markers in the brain and attenuated the histological changes in the hippocampus caused by the injury. The level of neuronal apoptosis in CIR-injured rats was also significantly decreased by Y-27632 treatment. Lastly, the protein levels of GFAP and AIF1 were further increased following Y-27632 exposure and Y-27632 significantly increased the survival rates at 24, 48 h and 7 days after CIR.

ROCK is an important kinase involved in cell mitosis, adhesion, cytoskeleton regulation, muscle cell contraction, tumor cell infiltration/migration and other cell functions (16). ROCK participates in the signaling pathway that regulates the presence of its respective receptors in the cytoplasm and is also involved in the regulation of the cytoskeletal structure (17). In recent years, ROCK has been increasingly studied in context of IR injury (1820). The pretreatment of heart tissue with fusadil may prevent the endothelial dysfunction and significantly reduce the degree of myocardial infarction (21). In the early stages of reperfusion, upregulation of Rho expression in the ischemic myocardium activates ROCK (22). To evaluate the potential beneficial effect of ROCK inhibition on CIR, rats with MCAO were treated with the ROCK inhibitor Y-27632 and their survival rates and neurological performance were examined. The survival rate of animals treated with MCAO and Vehicle at 24, 48 h and 7 days was 60, 50 and 35%, respectively. In contrast, MCAO animals treated with Y-27632 group exhibited significantly higher survival rates (90, 80 and 50% at 24 and 48 h and 7 days respectively). Furthermore, Y-27632 significantly decreased the neurological deficit score and enhanced the performance of rats in the water maze tests compared to MCAO + Vehicle animals. These results indicated that ROCK inhibition may effectively prevent death and the neurological impairment caused by CIR.

Different factors, including ischemia, hypoxia, hyperglycemia and infection, may activate tyrosine kinases and G protein-coupled receptors, which in turn activate RhoA. Activation of the RhoA/ROCK signaling pathway may have detrimental effects during IR. Firstly, activation of the RhoA/ROCK signaling pathway in the vascular endothelium is known to inhibit P13K/AKT activation, decrease nitric oxide synthase 3 phosphorylation and nitric oxide production, which may reduce regional cerebral blood flow following CIR (23). Secondly, activation of RhoA/ROCK signaling pathway may increase blood flow viscosity. The exact mechanism remains unclear, but is hypothesized to be associated with erythrocyte deformability (24). Thirdly, activation of the RhoA/ROCK signaling pathway promotes the expression of serpin family E member 2 in vascular endothelial cells and worsens intracapillary circulation (25). Phosphorylation of myosin light-chain in vascular endothelial cells can lead to a decrease in tight junctions between cells and increased endothelial cell permeability (26). Lastly, activation of the RhoA/ROCK signaling pathway may increase the infiltration of inflammatory (neutrophils) into ischemic tissue, aggravating the inflammatory response during IR, and reducing the integrity of the blood-brain barrier (27). The mechanism underlying fusadil's protection against cerebral infarction includes increased local cerebral blood flow by vasodilation and reduced inflammatory response (28). To evaluate the effect of ROCK inhibition on inflammatory response induced following CIR injury, rats with MCAO were treated with Y-27632 and the levels of pro-inflammatory factors TNF-α, IL-1β and IL-6 in the infarcted brain tissue were examined (2930). The levels of TNF-α and IL-1β in MCAO + Vehicle group were significantly increased compared to Control, and treatment with Y-27632 significantly inhibited their increase, suggesting the involvement of ROCK signaling pathway in the inflammatory response induced by CIR. MCAO also induced significant histological changes in the hippocampus, and these were significantly attenuated following treatment with the ROCK inhibitor. Overall, these results indicated that Y-27632 may have directly attenuated the inflammatory response and reduced hippocampal injury induced by CIR.

Free oxygen radicals are an important product in IR injuries. These may increase the activity of arginase in endothelial cells through Protein kinase C-activated RhoA/ROCK signaling pathway, reduce the production of nitric oxide, increase superoxide production and disturb normal endothelial function (31). Moreover, the RhoA/ROCK signaling pathway has been shown to reduce the production of free oxygen radicals following cerebral infarction (32). As the present study showed, the levels of MDA and 3-NT in MCAO + Vehicle group were greatly increased compared to Control, and treatment of Y-27632 prevented this increase. This result indicated that the protective effect of Y-27632 against CIR injury may also be associated with reduced oxidative stress.

When the central nervous system is injured, astrocytes will transform from a resting state to an activated state (and are hence termed ‘reactive astrocytes’). Reactive gliosis results in the hypertrophy of glial cells and in the proliferation of microglia (33). The response of astrocytes to injury is thought to promote the protection of the nervous system and the isolation of the injury area from the surrounding healthy tissue. After the injury, activated astrocytes and microglia may increase the secretion of neurotrophic factors (including nerve growth factor, neurotrophin-3, brain-derived neurotrophic factor), which can maintain the survival of neurons and accelerate the growth of neurites (34). The release of these factors also allows activated glial cells to provide neurons with ‘scaffolds’ that help restore disrupted neural connections. In the infarcted area, the expression level of a variety of growth factors secreted by astrocytes increases significantly, which provides direct nutritional support for neurons and oligodendrocytes and limits secondary damage (35). Hypoxia, inflammatory response and trauma can promote activated astrocytes to secrete vascular endothelial growth factor (VEGF). VEGF may also protect neurons and promote angiogenesis and formation of new blood vessels, which is beneficial for the repair of damaged areas (36). By eliminating glial cells or by selectively blocking a signal pathway, a study has demonstrated that the key to barrier remodeling is to activate proliferation and migration of astrocytes (37). Therefore, to explore the potential role of astrocytes and microglia in the Y-27632-mediate protection against CIR injury, the protein levels of GFAP (a marker of astrocyte) and AIF1 (a marker of microglia) in the infarcted area were evaluated. The results demonstrated that the protein levels of GFAP and AIF1 were greatly increased by CIR, as expected considering the damage caused to the neuronal tissue. However, the treatment of Y-27632 further increased these protein levels. These results confirmed that astrocytes and microglia would are more active in response to CIR. It also revealed that Y-27632 may stimulate their response and exert protective effects via activation of both astrocytes and microglia. Previous studies have found that the inhibition of the ROCK pathway attenuated methylmercury-induced astrocyte death (38). It was also revealed that ROCK inhibitors Y-27632 and fasudil promote microglial migration in the spinal cord via the ERK signaling pathway (39). Consistent with these studies, the present study confirmed that activation of astrocytes and microglia may be a possible mechanism by which ROCK inhibition may protect brain tissue against CIR injury.

However, the present study had some limitations. Firstly, multiple mechanisms may be associated with Y-27632-induced protection against CIR. The present study focused on the activation of astrocytes and microglia, but this may not be the only mechanism. Secondly, this is a primary animal study on the protective effect of Y-27632 in CIR injury. Many factors should be investigated when considering its clinical application against CIR, including, but not limited to, its effects on human beings, toxicity and dosage. Thirdly, further mechanism should be investigated in future studies, such as how Y-27632 regulates the activation of astrocytes and microglia or the interaction between ROCK and astrocytes and microglia. Finally, the study would be more perfect if the formation of the glial scar was shown by immunohistochemical examination, but because of the limitation of time and the technology, we are unable to perform it.

In conclusion, the present study confirmed that Y-27632, a ROCK inhibitor, effectively increased the survival rate and behavioral performance of rats with CIR damage. It also decreased the oxidative stress, cerebral inflammation, neuron apoptosis and hippocampal damage in animals with MCAO. The activated proliferation of astrocytes and microglia may be responsible for this beneficial effect.

Acknowledgements

Not applicable.

Funding

No funding was received.

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

LL conceived and designed the study and revised the manuscript; BL performed the experiments and performed the statistical analysis. All authors read and approved the manuscript.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of Tianjin Baodi District People's Hospital.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Iadecola C and Anrather J: Stroke research at a crossroad: Asking the brain for directions. Nat Neurosci. 14:1363–1368. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Breuer L, Knott M, Struffert T, Kloska S, Kurka N, Schwab S, Dörfler A, Köhrmann M and Engelhorn T: Limited versus whole-brain perfusion for the indication of thrombolysis in the extended time window of acute cerebral ischemia. J Stroke Cerebrovasc Dis. 24:2491–2496. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Xiong XY, Liu L and Yang QW: Refocusing neuroprotection in cerebral reperfusion era: New challenges and strategies. Front Neurol. 9:2492018. View Article : Google Scholar : PubMed/NCBI

4 

Panieri E, Gogvadze V, Norberg E, Venkatesh R, Orrenius S and Zhivotovsky B: Reactive oxygen species generated in different compartments induce cell death, survival, or senescence. Free Radic Biol Med. 57:176–187. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Xue J, Zhang X, Zhang C, Kang N, Liu X, Yu J, Zhang N, Wang H, Zhang L, Chen R, et al: Protective effect of Naoxintong against cerebral ischemia reperfusion injury in mice. J Ethnopharmacol. 182:181–189. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Julian L and Olson MF: Rho-associated coiled-coil containing kinases (ROCK): Structure, regulation, and functions. Small GTPases. 5:e298462014. View Article : Google Scholar : PubMed/NCBI

7 

Schmandke A, Schmandke A and Strittmatter SM: ROCK and Rho: Biochemistry and neuronal functions of Rho-associated protein kinases. Neuroscientist. 13:454–469. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Gong H and Yang CY: Morphological and hydrodynamic correlations with increasing outflow facility by rho-kinase inhibitor Y-27632. J Ocul Pharmacol Ther. 30:143–153. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Oda T, Kume T, Izumi Y, Ishihara K, Sugmimoto H and Akaike A: Na+/Ca2+ exchanger inhibitors inhibit neurite outgrowth in PC12 cells. J Pharmacol Sci. 116:128–131. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Wang J, Li H, Yao Y, Ren Y, Lin J, Hu J, Zheng M, Song X, Zhao T, Chen YY, et al: β-elemene enhances GAP-43 expression and neurite outgrowth by inhibiting RhoA kinase activation in rats with spinal cord injury. Neuroscience. 383:12–21. 2018. View Article : Google Scholar : PubMed/NCBI

11 

Pang Y, Chai CR, Gao K, Jia XH, Kong JG, Chen XQ, Vatcher G, Chen JG and Yu AC: Ischemia preconditioning protects astrocytes from ischemic injury through 14-3-3γ. J Neurosci Res. 93:1507–1518. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Zhao JJ, Hu JX, Lu DX, Ji CX, Qi Y, Liu XY, Sun FY, Huang F, Xu P and Chen XH: Soluble cpg15 from astrocytes ameliorates neurite outgrowth recovery of hippocampal neurons after mouse cerebral ischemia. J Neurosci. 37:1628–1647. 2017. View Article : Google Scholar : PubMed/NCBI

13 

Yang C, Zhang X, Fan H and Liu Y: Curcumin upregulates transcription factor Nrf2, HO-1 expression and protects rat brains against focal ischemia. Brain Res. 28:133–141. 2009. View Article : Google Scholar

14 

Jia D, Deng Y, Gao J, Liu X, Chu J and Shu Y: Neuroprotective effect of Panax notoginseng plysaccharides against focal cerebral ischemia reperfusion injury in rats. Int J Biol Macromol. 63:177–180. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Ahmadi M, Rajaei Z, Hadjzadeh MA, Nemati H and Hosseini M: Crocin improves spatial learning and memory deficits in the Morris water maze via attenuating cortical oxidative damage in diabetic rats. Neurosci Lett. 642:1–6. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Mueller BK, Mack H and Teusch N: Rho kinase, a promising drug target for neurological disorders. Nat Rev Drug Discov. 4:387–398. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Tanaka T, Nishimura D, Wu RC, Amano M, Iso T, Kedes L, Nishida H, Kaibuchi K and Hamamori Y: Nuclear Rho kinase, ROCK2, targets p300 acetyltransferase. J Biol Chem. 281:15320–15329. 2006. View Article : Google Scholar : PubMed/NCBI

18 

Wen JY, Gao SS, Chen FL, Chen S, Wang M and Chen ZW: Role of CSE-produced H2S on cerebrovascular relaxation via RhoA-ROCK inhibition and cerebral ischemia-reperfusion injury in Mice. ACS Chem Neurosci. 10:1565–1574. 2019. View Article : Google Scholar : PubMed/NCBI

19 

Sun XQ, Chen S, Wang LF and Chen ZW: Total flavones of Rhododendron simsii Planch flower protect isolated rat heart from ischaemia-reperfusion injury and its mechanism of UTR-RhoA-ROCK pathway inhibition. J Pharm Pharmacol. 70:1713–1722. 2018. View Article : Google Scholar : PubMed/NCBI

20 

Chen F, Liu Z, Peng W, Gao Z, Ouyang H, Yan T, Ding S, Cai Z, Zhao B, Mao L and Cao Z: Activation of EphA4 induced by EphrinA1 exacerbates disruption of the blood-brain barrier following cerebral ischemia-reperfusion via the Rho/ROCK signaling pathway. Exp Ther Med. 16:2651–2658. 2018.PubMed/NCBI

21 

Yada T, Shimokawa H, Hiramatsu O, Kajita T, Shigeto F, Tanaka E, Shinozaki Y, Mori H, Kiyooka T, Katsura M, et al: Beneficial effect of hydroxyfasudil, a specific Rho-kinase inhibitor, on ischemia/reperfusion injury in canine coronary microcirculation in vivo. J Am Coll Cardiol. 45:599–607. 2005. View Article : Google Scholar : PubMed/NCBI

22 

Hamid SA, Bower HS and Baxter GF: Rho kinase activation plays a major role as a mediator of irreversible injury in reperfused myocardium. Am J Physiol Heart Circ Physiol. 292:H2598–H2606. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Shin HK, Salomone S, Potts EM, Lee SW, Millican E, Noma K, Huang PL, Boas DA, Liao JK, Moskowitz MA and Ayata C: Rho-kinase inhibition acutely augments blood flow in focal cerebral ischemia via endothelial mechanisms. J Cereb Blood Flow Metab. 27:998–1009. 2007. View Article : Google Scholar : PubMed/NCBI

24 

Tiftik RN, Baskurt OK, Kul S and Buyukafsar K: The functional significance of the rho/rho-kinase pathway in human erythrocytes. Turk J Haematol. 31:168–174. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Iwasaki H, Okamoto R, Kato S, Konishi K, Mizutani H, Yamada N, Isaka N, Nakano T and Ito M: High glucose induces plasminogen activator inhibitor-1 expression through Rho/Rho-kinase-mediated NF-kappaB activation in bovine aortic endothelial cells. Atherosclerosis. 196:22–28. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Shen L, Black ED, Witkowski ED, Lencer WI, Guerriero V, Schneeberger EE and Turner JR: Myosin light chain phosphorylation regulates barrier function by remodeling tight junction structure. J Cell Sci. 119:2095–2106. 2006. View Article : Google Scholar : PubMed/NCBI

27 

Satoh S, Utsunomiya T, Tsurui K, Kobayashi T, Ikegaki I, Sasaki Y and Asano T: Pharmacological profile of hydroxy fasudil as a selective rho kinase inhibitor on ischemic brain damage. Life Sci. 69:1441–1453. 2001. View Article : Google Scholar : PubMed/NCBI

28 

Rikitake Y, Kim HH, Huang Z, Seto M, Yano K, Asano T, Moskowitz MA and Liao JK: Inhibition of Rho kinase (ROCK) leads to increased cerebral blood flow and stroke protection. Stroke. 36:2251–2257. 2005. View Article : Google Scholar : PubMed/NCBI

29 

Lu Z, Cheng D, Yin J, Wu R, Zhang G, Zhao Q, Wang N, Wang F and Liang M: Antithrombin III protects against contrast-induced nephropathy. EBioMedicine. 17:101–107. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Yin J, Wang F, Kong Y, Wu R, Zhang G, Wang N, Wang L, Lu Z and Liang M: Antithrombin III prevents progression of chronic kidney disease following experimental ischaemic-reperfusion injury. J Cell Mol Med. 21:3506–3514. 2017. View Article : Google Scholar : PubMed/NCBI

31 

Chandra S, Romero MJ, Shatanawi A, Alkilany AM, Caldwell RB and Caldwell RW: Oxidative species increase arginase activity in endothelial cells through the RhoA/Rho kinase pathway. Br J Pharmacol. 165:506–519. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Kahles T, Luedike P, Endres M, Galla HJ, Steinmetz H, Busse R, Neumann-Haefelin T and Brandes RP: NADPH oxidase plays a central role in blood-brain barrier damage in experimental stroke. Stroke. 38:3000–3006. 2007. View Article : Google Scholar : PubMed/NCBI

33 

Gao Z, Zhu Q, Zhang Y, Zhao Y, Cai L, Shields CB and Cai J: Reciprocal modulation between microglia and astrocyte in reactive gliosis following the CNS injury. Mol Neurobiol. 48:690–701. 2013. View Article : Google Scholar : PubMed/NCBI

34 

Cragnolini AB, Montenegro G, Friedman WJ and Masco DH: Brain-region specific responses of astrocytes to an in vitro injury and neurotrophins. Mol Cell Neurosci. 88:240–248. 2018. View Article : Google Scholar : PubMed/NCBI

35 

Williams A, Piaton G and Lubetzki C: Astrocytes-friends or foes in multiple sclerosis? Glia. 55:1300–1312. 2007. View Article : Google Scholar : PubMed/NCBI

36 

Guo D, Murdoch CE, Xu H, Shi H, Duan DD, Ahmed A and Gu Y: Vascular endothelial growth factor signaling requires glycine to promote angiogenesis. Sci Rep. 7:147492017. View Article : Google Scholar : PubMed/NCBI

37 

Okada S, Nakamura M, Katoh H, Miyao T, Shimazaki T, Ishii K, Yamane J, Yoshimura A, Iwamoto Y, Toyama Y and Okano H: Conditional ablation of Stat3 or Socs3 discloses a dual role for reactive astrocytes after spinal cord injury. Nat Med. 12:829–834. 2006. View Article : Google Scholar : PubMed/NCBI

38 

Dos Santos AA, Lopez-Granero C, Farina M, Rocha JBT, Bowman AB and Aschner M: Oxidative stress, caspase-3 activation and cleavage of ROCK-1 play an essential role in MeHg-induced cell death in primary astroglial cells. Food Chem Toxicol. 113:328–336. 2018. View Article : Google Scholar : PubMed/NCBI

39 

Fu PC, Tang RH, Yu ZY, Xie MJ, Wang W and Luo X: The Rho-associated kinase inhibitors Y27632 and fasudil promote microglial migration in the spinal cord via the ERK signaling pathway. Neural Regen Res. 13:677–683. 2018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2019
Volume 20 Issue 4

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
Li L and Liu B: ROCK inhibitor Y‑27632 protects rats against cerebral ischemia/reperfusion‑induced behavioral deficits and hippocampal damage. Mol Med Rep 20: 3395-3405, 2019.
APA
Li, L., & Liu, B. (2019). ROCK inhibitor Y‑27632 protects rats against cerebral ischemia/reperfusion‑induced behavioral deficits and hippocampal damage. Molecular Medicine Reports, 20, 3395-3405. https://doi.org/10.3892/mmr.2019.10584
MLA
Li, L., Liu, B."ROCK inhibitor Y‑27632 protects rats against cerebral ischemia/reperfusion‑induced behavioral deficits and hippocampal damage". Molecular Medicine Reports 20.4 (2019): 3395-3405.
Chicago
Li, L., Liu, B."ROCK inhibitor Y‑27632 protects rats against cerebral ischemia/reperfusion‑induced behavioral deficits and hippocampal damage". Molecular Medicine Reports 20, no. 4 (2019): 3395-3405. https://doi.org/10.3892/mmr.2019.10584