Cannabinoid receptor 2 agonist attenuates blood‑brain barrier damage in a rat model of intracerebral hemorrhage by activating the Rac1 pathway

  • Authors:
    • Zhe Wang
    • Yongfu Li
    • Shuangyong Cai
    • Rui Li
    • Guanbo Cao
  • View Affiliations

  • Published online on: August 22, 2018     https://doi.org/10.3892/ijmm.2018.3834
  • Pages: 2914-2922
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Blood‑brain barrier (BBB) disruption and consequent edema formation are the most common brain injuries following intracerebral hemorrhage (ICH). Endocannabinoid receptors can alter the permeability of various epithelial barriers and have potential neuroprotective effects. The present study aimed to explore whether the selective cannabinoid receptor 2 (CNR2) agonist, JWH133, can ameliorate BBB integrity and behavioral outcome by activating Ras‑related C3 botulinum toxin substrate 1 (Rac1) following ICH. Autologous arterial blood was injected into the basal ganglia of rats to induce ICH. Animals were randomly divided into the following groups: Sham‑operated, ICH+vehicle, ICH+JWH133, ICH+JWH13+vehicle, ICH+JWH133+AM630 (a selective CNR2 antagonist), ICH+AM630, ICH+JWH133 +NSC23766 (a Rac1 antagonist) and ICH+NSC23766. JWH133 and AM630 were independently intraperitoneally administrated at 1 h prior to ICH. NSC23766 was intracerebroventricularly (ICV) administered 30 min prior to ICH. A modified Garcia test, corner test, Evans blue extravasation and brain water content analysis were performed at 24 and 72 h following ICH. Western blotting and pull‑down assays were performed at 24 h following ICH. The results demonstrated that JWH133 treatment improved neurofunctional deficits, reduced perihematomal brain edema and alleviated BBB damage at 24 and 72 h following ICH. In addition, JWH133 treatment increased the protein expression levels of guanosine‑5'‑triphosphate‑Rac1 and of the adherens junction proteins occludin, zonula occludens‑1 and claudin‑5. However, these effects were reversed by AM630 and NSC23766 treatment. In conclusion, the present findings revealed that JWH133 treatment attenuated brain injury in a rat model of ICH via activation of the Rac1 signaling pathway, thus preserving BBB integrity.

Introduction

Intracerebral hemorrhage (ICH) is a fatal stroke subtype, associated with high mortality worldwide, and it accounts for 10–15% of all hospitalized stroke patients (1,2). Blood-brain barrier (BBB) damage and angioedema occur in the early stages of injury. BBB breakdown leads to brain injury progression and long-term neurological deficits (3). Destruction of the BBB directly aggravates vasogenic brain edema, and has been identified as an important contributor of secondary brain damage (4). Endothelial cells are interconnected by tight junctions (TJs) and mainly composed of zonula occludens-1 (ZO-1), occludin and claudin proteins (5). ZO-1 anchors the transmembrane protein occludin to the actin cytoskeleton, which confers the capacity of BBB to preclude permeation of blood substances (6). Changes in TJ components result in loss of BBB integrity and BBB decomposition (7). A variety of signal transduction pathways mediate the opening of tight junctions, including Ras-related C3 botulinum toxin substrate 1 (Rac1) (8). Therefore, regulating BBB permeability and reducing the degradation of TJ proteins may allow for a more effective ICH treatment strategy.

The endocannabinoid system has been studied as a potential therapeutic target for neuroprotection (9). There are two major subtypes of cannabinoid receptor, cannabinoid receptor 1 (CNR1) and cannabinoid receptor 2 (CNR2) (10,11). CNR2 is highly expressed in immune cells and mediates inflammatory responses (12). Previous studies have reported that cannabinoid receptors serve an important regulatory role in normal BBB physiology and protect BBB during ischemic stroke (13). Pharmacological data has suggested that CNR2 agonists increase the presence of tight junction proteins in membrane fractions in endothelial resistance and neuroinflammation (14). JWH133 (a specific CNR2 agonist), which exhibits a very high affinity for CNR2 (Ki=3.4 nmol/l) but low affinity for CNR1 (Ki=677 nmol/l), has been reported to attenuate brain edema in rat models of germinal matrix hemorrhage (15), and to improve functional outcomes and reduce brain edema following experimental subarachnoid hemorrhage in rats. In addition, JWH-133 treatment attenuates BBB damage in traumatic brain injury (16). These results indicate that CNR2 activation has potential as a practical and therapeutic intervention.

Rac1 is an important and well-studied member of a small-molecular-weight guanosine-5′-triphosphate (GTP) binding protein located inside the cell membrane. It serves an important role in cell proliferation and differentiation (17) and in regulation of cytoskeletal organization in many cell types. Rac1 maintains and stabilizes the barrier function of microvascular endothelial cells (18). Rac1 activation inhibits RhoA, which may preserve BBB integrity and, therefore, reduce the development of vasogenic brain edema after ICH (19). CNR2 concurrently or sequentially affects TJ protein stability via the Rho GTPase pathway (20). CNR2 agonists reduce integrin activation and lamellipodia formation in primary monocytes via inhibition of small GTPases (RhoA and Rac1) and affects cytoskeletal proteins (21). However, whether CNR2 activation can prevent BBB disruption and brain edema by inhibiting Rac1 activation in ICH, remains unknown.

Therefore, the present study investigated the effects of the CNR2 agonist, JWH133, on a rat model of ICH-induced BBB damage, and the role of Rac1 in the neuroprotective process. The present study demonstrated that JWH133, a selective CNR2 agonist, improved neurofunctional deficits, reduced brain edema and alleviated BBB damage following ICH, and these effects were reversed by AM630 (CNR2 antagonist) and NSC23766 (Rac1 antagonist) treatment.

Materials and methods

Chemicals

JWH133 (CNR2 agonist), and AM630 (CNR2 antagonist) were purchased from Tocris Bioscience (Bristol, UK). JWH133 and AM630 were dissolved in dimethyl sulfoxide (DMSO) and then diluted with sterile PBS. NSC23766 (Rac1 inhibitor) was purchased from Abcam (Cambridge, UK) and also dissolved in DMSO. Rac1 Pull-down Activation Assay kit was obtained from Cytoskeleton Inc. (Denver, CO, USA). Rat anti-Rac1 monoclonal antibody (cat. no. PA1-091; 1:1,000) was purchased from Thermo Fisher Scientific, Inc. (Waltham, MA, USA). Anti-occludin (cat. no. sc-133256; 1:800), anti-ZO-1 (cat. no. sc-33725; 1:800) and anti-claudin-5 (cat. no. sc-374221; 1:800) antibodies were purchased from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA). An antibody targeting β-actin (cat. no. 3700; 1:1,000) was purchased from Cell Signaling Technology, Inc. (Danvers, MA, USA). Goat anti-rabbit (cat. no. sc-2004; 1:2,000) and goat anti-mouse secondary antibodies (cat. no. sc-2005; 1:2,000) were purchased from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA).

Animals and treatments

A total of 180 male Sprague-Dawley rats (275–300 g) were purchased from the Laboratory Animal Center, Third Military Medical University (Chongqing, China). Rats were housed in a light and temperature controlled environment at 20–22°C. Food and water were available ad libitum. Experimental protocols were approved by the Medical Ethics Committee of the Third Military Medical University.

The experiments were divided into two parts. In experiment I, animals were randomly divided into the following groups (15 rats per group): Sham-operated (Sham group), ICH+vehicle (Vehicle group), ICH+JWH133 (JWH133 group), ICH+JWH133+vehicle (JWH133+vehicle group), ICH+ JWH133+AM630 (JWH133+AM630 group) and ICH+AM630 (AM630 group). JWH133 (5.0 mg/kg) and AM630 (1.0 mg/kg) were intraperitoneally (i.p.) injected 1 h prior to ICH. In experiment II, animals were randomly divided into the following groups (15 rats per group): Sham-operated (Sham group), ICH+vehicle (Vehicle group), ICH+JWH133 (JWH133 group), ICH+J WH133+vehicle (J WH133+vehicle group), ICH+JWH133+NSC23766 (JWH133+NSC23766 group) and ICH+NSC23766 (NSC23766 group). JWH133 (5.0 mg/kg) was injected i.p. at 1 h prior to IHC, and NSC23766 (0.1 mg/kg) was administered intracerebroventricularly (ICV) at 30 min prior to ICH. Animals were sacrificed 24 and 72 h following surgery. The control rats received an equal volume of vehicle. The physiological parameters of rats were measured at 24 and 72 h following the ICH procedure. The experimental design is presented in Fig. 1.

Rat ICH model

The rat ICH model was established as previously described (22). Briefly, animals were anesthetized with an intraperitoneal injection of pentobarbital (40 mg/kg) and placed on a stereotaxic apparatus. A 1-mm cranial burr hole was drilled, and a needle was inserted into the right basal ganglia under stereotactic guidance (coordinates: 0.2-mm anterior, 3.5-mm lateral to the midline and 5.5-mm ventral), and 100 µl of autologous arterial blood extracted from the femoral artery was slowly infused at a rate of 5 µl/min using a microinfusion pump. After the injection was complete, the needle was left in place for 20 min before withdrawal. Control animals were injected with 100 µl of normal saline. Bone wax was used to close the burr hole, and the skin incision was closed with sutures. In order to avoid infection, all procedures were performed under aseptic conditions.

Western blot analysis

Rats were sacrificed at 24 and 72 h post-ICH, and brain tissues surrounding the hemorrhagic region were isolated. Total protein was isolated from brain tissues using RIPA lysis buffer (Beyotime Institute of Biotechnology, Beijing, China). The protein concentrations were determined by bicinchoninic acid (BCA) assay. Equal amounts of protein (40 µg) were separated on 10% sodium dodecyl sulfate-polyacrylamide gels and transferred into PVDF membranes (EMD Millipore, Billerica, MA, USA). The membrane was blocked in 5% non-fat milk at room temperature for 1 h and incubated overnight at 4°C with the following primary antibodies: Anti-occludin, anti-ZO-1, anti-claudin-5 and β-actin. Then the membranes were incubated with secondary antibody for 1 h at room temperature. (1:2,000; Santa Cruz Biotechnology, Inc.) and immunoblots were visualized using an ECL Plus chemiluminescence kit (Amersham; GE Healthcare, Chicago, IL, USA). Blot bands were semi-quantitatively analyzed using ImageJ (version 4.0; National Institutes of Health, Bethesda, MD, USA).

Functional behavioral test

The sensorimotor Garcia (23) and Corner test (24) were conducted in a blinded fashion, and used to assess neurofunctional deficits in rats at 24 and 72 h post-ICH. The Garcia Test was modified to examine spontaneous activity, axial sensation, vibrissae touch and limb symmetry, as well as the animal's ability to turn laterally, outstretch forelimbs and climb. The worst performance was awarded 0 points, and the best performance, 3 points, for each subtest. The sum point score was calculated to determine neurological function (maximum score, 21).

The corner test was conducted as described by Li et al (24). The rat was placed between two boards, each with dimension of 30×20×1 cm3. The edges of the two boards were connected at an angle of 30°. A small opening along the joint between the two boards encouraged entry into the corner. The rat was placed between the two angled boards facing the corner and half way to the corner. When the rat entered far into the corner both sides of the vibrissae were stimulated together. Rearing forwards and upwards was observed, followed by turning to face the open end. The turns in each direction were recorded for each test. A total of 10 trials were performed for each rat, with a break of ≥30 sec between trials. The % of right turns was calculated. Only turns involving full rearing along either board were included.

Analysis of the brain water content

The brain water content was examined in rats at 24 and 72 h post-ICH, according to a previous study (25). Animals were anesthetized with pentobarbital (40 mg/kg; i.p.), sacrificed and the brains were surgically removed. The cerebrum was divided into 5 parts: Ipsilateral cortex (Ipsi-CX) and contralateral cortex (Cont-CX), ipsilateral basal ganglia (Ipsi-BG) and contralateral basal ganglia (Cont-BS), and cerebellum (cerebel). Each part was immediately weighed using an analytical microbalance (APX-60; Denver Instrument, Bohemia, NY, USA) to measure the wet weight, and then they were dried at 100°C for 24 h to measure the dry weight. The brain water content was calculated using the formula: Brain water content (%) = (wet weight−dry weight)/wet weight ×100.

Evans blue staining

Evans blue staining was performed at 24 and 72 h post-ICH to evaluate blood-brain barrier permeability, as previously described by Ma et al (26). The rats were anesthetized and received femoral vein injection of 2% Evans blue solution (5 ml/kg; Sigma-Aldrich; Merck KGaA, Darmstadt, Germany), allowing the dye to circulate for 3 h prior to transcranial perfusion with PBS pH 7.4. Following transcranial perfusion, the brains were surgically removed, divided into right and left brain hemispheres and stored at −80°C until use. Brain specimens were homogenized in PBS, sonicated and centrifuged at 12,000 × g at 4°C for 30 min. The supernatant was collected and an equal amount of trichloroacetic acid (50%) was added. The supernatant was incubated overnight at 4°C and centrifuged at 15,000 × g for 30 min at 4°C. The absorbance of the sample was measured at 620 nm using a spectrophotometer (Multiskan GO; Thermo Fisher Scientific, Inc.) and quantified according to a standard curve.

Rac1-GTPase pull-down assay

Rac1 activation assays were performed as described previously (27). In brief, brain tissues surrounding the hemorrhagic region underwent homogenization in RIPA buffer (Santa Cruz Biotechnology, Inc.) supplemented with protease and phosphatase inhibitors (Sigma-Aldrich; Merck KGaA) at 4°C. The samples were incubated in a 4°C water bath for 1.5 h. Samples were centrifuged for 10 min at 10,000 × g at 4°C and the supernatants were collected. The protein concentration was determined using a BCA kit. A portion of each supernatant was added to Pak-GST-conjugated beads (10 µl) for detection of total Rac1 GTPases. The solution was continuously shaken for 2 h at 4°C, and centrifuged for 1 min at 3,000 × g at 4°C. The samples were washed three times with lysis buffer by centrifugation at 4,500 rpm at 4°C for 5 min. Proteins bound to the beads were rinsed with distilled water and the Rac1-GTPase activity was determined by western blotting analysis.

Statistical analysis

The data was analyzed using SPSS 13.0 software (SPSS Inc., Chicago, IL, USA). Data were expressed as the mean ± standard deviation. One-way analysis of variance was used to compare the differences among the multiple groups of data, followed by Tukey's post hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

JWH133 treatment reduces neurofunctional deficit and brain edema at 24 and 72 h following ICH

A modified method of autologous blood injection was used to establish the ICH model, as described by Yang et al (22). Neurofunctional deficits and brain water content were evaluated at 24 and 72 h following surgery. As illustrated in Fig. 2A and B, rats performed significantly lower scores during the Garcia and Corner tests at 24 and 72 h following surgery. In addition, the results demonstrated increased brain water content in the ipsilateral basal ganglia of the brain in the vehicle group, compared with the sham group (Fig. 2C and D). JWH133 administration post-surgery significantly ameliorated the neurofunctional deficits and reduced the brain water content compared with the vehicle group (Fig. 2). However, the selective CNR2 antagonist, AM630, reversed this treatment effect (Fig. 2).

JWH133 protects against ICH-induced BBB destruction

Evans blue staining was used to detect BBB integrity at 24 and 72 h following ICH. The results demonstrated that Evans blue leakage was significantly increased at 24 and 72 h post-ICH in the vehicle group compared with the sham group (Fig. 3). JWH133 treatment significantly reduced Evans blue leakage at 24 and 72 h post-ICH compared with the vehicle group (Fig. 3). However, the effect of JWH133 on decreasing the Evans blue leakage was reversed by AM630 treatment (Fig. 3).

JWH133 administration upregulates GTP-Rac1 protein expression following ICH

To elucidate the potential mechanism of JWH133 on the CNR2/Rac1/TJ signaling pathway following ICH, a pull-down method was used to analyze the effect of JWH133 on the expression of GTP-Rac1 protein at 24 h post-ICH. As illustrated in Fig. 4, the GTP-Rac1 protein levels were decreased in the ipsilateral basal ganglia at 24 h post-ICH compared with the sham group. However, the GTP-Rac1 protein expression levels were higher in the JWH133-treated group and JWH133+vehicle group, compared with the vehicle group (Fig. 4). The combination treatment of JWH133 with AM630 reversed the GTP-Rac1 protein upregulation, compared with the JWH133 group (Fig. 4).

JWH133 administration prevents ICH-induced TJ-related protein attenuation

The expression levels of the TJ proteins, ZO-1, claudin-5 and occludin, were measured by western blotting at 24 h post-ICH. As illustrated in Fig. 5, the results demonstrated that the protein expression levels of ZO-1, claudin-5 and occludin in the ipsilateral basal ganglia of rats were significantly decreased at 24 h post-ICH compared with the sham group. JWH133 administration significantly upregulated the protein expression levels of ZO-1, claudin-5 and occludin (Fig. 5). The combination treatment of JWH133 with AM630 reversed ZO-1, claudin-5 and occludin protein levels compared with the JWH133 group (Fig. 5).

Rac1 inhibition reverses the neuroprotective effect of JWH133

In order to confirm that the neuroprotection effect of JWH133 was mediated by Rac1, the Rac1 inhibitor, NSC23766, was injected in the rats at 0.5 h prior to ICH. As illustrated in Fig. 6A, NSC23766 administration significantly decreased the JWH133-induced upregulation of GTP-Rac1 protein expression. Next, neurofunctional deficits, brain water content, and Evans Blue permeability were detected at 24 and 72 h post-ICH. NSC23766 administration reversed the protective effect of JWH133 treatment, as demonstrated by significantly lower scores in the Garcia and Corner test (Fig. 6B and C), as well as significantly increased brain water content in the ipsilateral basal ganglia of the brain (Fig. 6D and E). In addition, the combination treatment of JWH133 with NSC23766 increased Evans Blue leakage compared with the JWH133 group (Fig. 6F).

Rac1 inhibition reverses the effect of JWH133 on TJ-related protein expression post-ICH

The results demonstrated that JWH133 treatment increased ZO-1, claudin-5 and occludin protein expression levels at 24 h post-ICH, compared with the vehicle group (Fig. 7). However, NSC23766 administration, alone or in combination with JWH133, efficiently decreased ZO-1, claudin-5 and occludin protein expression compared with the JWH133 group (Fig. 7).

Discussion

In the present study, autologous blood was infused into the caudate nucleus of rats to establish a ICH model, similar to models that are widely used to study the mechanisms of brain injury. This rat model was then used to investigate the effects of CNR2 activation in ICH. The results demonstrated that JWH133 administration attenuated neurofunctional deficits and brain edema in rats at 24 and 72 h following experimental ICH. In addition, JWH133 treatment decreased BBB permeability at 24 and 72 h following experimental ICH. These results suggested that cannabinoid receptor agonists may serve a protective role following ICH. However, whether CNR2 activation can prevent BBB disruption and brain edema by inhibiting Rac1 activation in ICH has not been clarified.

In addition, in the present study, the expression levels of GTP-Rac1 and the tight junction proteins ZO-1, occludin and claudin-5 were detected at 24 h following treatment with JWH133 alone or in combination with AM630 or NSC23766. The results demonstrated that JWH133 treatment increased the expression levels of ZO-1, occludin and claudin proteins, and induced activation of Rac1 (GTP-Rac1) at 24 h following ICH. Combination treatment with the CNR2 antagonist, AM630, or the Rac1 inhibitor, NSC23766, reversed all the treatment effects of JWH133. These results suggest that the neuroprotection was mediated by CNR2-induced activation of the Rac1 pathway. However, the expression of GTP-Rac1 and the tight junction proteins at 72 h post-treatment of JWH133 alone or combination of AM630 or NSC23766 require further study.

Increased BBB permeability and consequent vasogenic edema formation occurred following ICH. BBB disruption results from ICH by influx of blood-borne cells and substances into the brain parenchyma. This leads to early brain edema and sensorimotor dysfunction (28,29) and ultimately results in high morbidity and mortality rates (30). It has been previously reported that CNR2 activation has an important role in preventing brain edema and neuroinflammation. CNR2 agonists diminish inflammation-induced activation of brain endothelial cells and BBB disruption in conjunction with increased TJ protein expression (31). A previous study reported that JWH133 treatment increased ZO-1 expression and attenuated neurological outcome and brain edema, protecting the BBB following subarachnoid hemorrhage (14). In addition, JWH133 attenuated BBB dysfunction and preserved TJ protein expression in the rodent brain following LPS-induced encephalitis (32). The present findings are consistent with previous reports in which CNR2 activation by JWH133 significantly reduced BBB permeability and increased ZO-1, occludin and claudin expression.

Rac1 is a small GTPase that belongs to the Ras superfamily. Rac1 has been previously reported to maintain and stabilize barrier functions of microvascular endothelial cells (18). CNR2 agonists attenuate the activation of ras homolog family member A (RhoA) and Rac1 during migration (33). CNR2 agonists reduce integrin activation and lamellipodia formation in primary monocytes via inhibition of small GTPases (RhoA and Rac1) and effects on cytoskeletal proteins (34). JWH133 also activates the extracellular signal-regulated kinase (ERK) and phosphoinositide 3-kinase (PI3K) pathways. A previous study has reported that JWH133 induced a greater and sustained activation of ERK1/2 kinase in lipopolysaccharide (LPS)/interferon (IFN)-γ-induced interleukin (IL)-12p40 release from murine macrophages (35). JWH-133 treatment activates ERK1/2 and signal transducer and activator of transcription (STAT)-3 in a mouse model of ischemia/reperfusion (36). JWH-133 induced PI3K/AKT activity in osteoclast precursors and osteoblasts of breast cancer-induced osteolysis (37). JWH133 increased oligodendrocyte progenitor cell proliferation by activating PI3K/AKT (38). In the present study, it was demonstrated that administration of JWH133 increased the GTP-Rac-1/total-Rac-1 protein expression ratio, and the barrier protective effect of JWH133 was reversed by NSC23766 (Rac1 inhibitor). These findings suggest that Rac1 participates in the improvement of BBB destruction and brain edema by JHH133 in experimental ICH. In the present study, only one selective CNR2 agonist, JWH133, was selected to investigate the effects on a rat model of ICH-induced BBB damage and its potential mechanism. The effects of other CNR2 agonists, such as AM1241, on a rat model of ICH require further investigation.

In conclusion, the present results demonstrated that the CNR2 agonist JWH133 attenuated neurofunctional deficits and brain edema, and improved the BBB dysfunction induced by ICH, through the Rac1 pathway. Therefore, it is likely that targeting CNR2 may be a clinically feasible approach in the future to protect against ICH.

Funding

The present study was supported by the National Key Basic Research Program of China, 973 Program (grant no. 2014CB541000) and the National Natural Science Foundation of China (grant no. 31071036).

Availability of data and materials

The analyzed datasets generated during the study are available from the corresponding author on reasonable request.

Authors' contributions

ZW wrote the manuscript and interpreted the data. YL and SC analyzed the data and revised the manuscript. RL searched the literature and collected the data. GC designed the study. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Experimental protocols involving animals were approved by the Medical Ethical Committee of the Third Military Medical University.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

References

1 

Broderick JP, Adams HP Jr, Barsan W, Feinberg W, Feldmann E, Grotta J, Kase C, Krieger D, Mayberg M, Tilley B, et al: Guidelines for the management of spontaneous intracerebral hemorrhage: A statement for healthcare professionals from a special writing group of the stroke council, American Heart Association. Stroke. 30:905–915. 1999. View Article : Google Scholar : PubMed/NCBI

2 

Qureshi AI, Mendelow AD and Hanley DF: Intracerebral haemorrhage. Lancet. 373:1632–1644. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Sweeney MD, Sagare AP and Zlokovic BV: Blood-brain barrier breakdown in Alzheimer disease and other neurodegenerative disorders. Nat Rev Neurol. 14:133–150. 2018. View Article : Google Scholar : PubMed/NCBI

4 

Guan JX, Sun SG, Cao XB, Chen ZB and Tong ET: Effect of thrombin on blood brain barrier permeability and its mechanism. Chin Med J (Engl). 117:1677–1681. 2004.

5 

Ballabh P, Braun A and Nedergaard M: The blood-brain barrier: An overview: Structure, regulation, and clinical implications. Neurobiol Dis. 16:1–13. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Bazzoni G and Dejana E: Endothelial cell-to-cell junctions: Molecular organization and role in vascular homeostasis. Physiol Rev. 84:869–901. 2004. View Article : Google Scholar : PubMed/NCBI

7 

Luh C, Kuhlmann CR, Ackermann B, Timaru-Kast R, Luhmann HJ, Behl C, Werner C, Engelhard K and Thal SC: Inhibition of myosin light chain kinase reduces brain edema formation after traumatic brain injury. J Neurochem. 112:1015–1025. 2010. View Article : Google Scholar

8 

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

9 

Xu JY and Chen C: Endocannabinoids in synaptic plasticity and neuroprotection. Neuroscientist. 21:152–168. 2015. View Article : Google Scholar

10 

Devane WA, Dysarz FR III, Johnson MR, Melvin LS and Howlett AC: Determination and characterization of a cannabinoid receptor in rat brain. Mol Pharmacol. 34:605–613. 1988.PubMed/NCBI

11 

Matsuda LA, Lolait SJ, Brownstein MJ, Young AC and Bonner TI: Structure of a cannabinoid receptor and functional expression of the cloned cDNA. Nature. 346:561–564. 1990. View Article : Google Scholar : PubMed/NCBI

12 

Haugh O, Penman J, Irving AJ and Campbell VA: The emerging role of the cannabinoid receptor family in peripheral and neuro-immune interactions. Curr Drug Targets. 17:1834–1840. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Chi OZ, Barsoum S, Grayson J, Hunter C, Liu X and Weiss HR: Effects of cannabinoid receptor agonist WIN 55,212-2 on blood-brain barrier disruption in focal cerebral ischemia in rats. Pharmacology. 89:333–338. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Fujii M, Sherchan P, Krafft PR, Rolland WB, Soejima Y and Zhang JH: Cannabinoid type 2 receptor stimulation attenuates brain edema by reducing cerebral leukocyte infiltration following subarachnoid hemorrhage in rats. J Neurol Sci. 342:101–106. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Tao Y, Tang J, Chen Q, Guo J, Li L, Yang L, Feng H, Zhu G and Chen Z: Cannabinoid CB2 receptor stimulation attenuates brain edema and neurological deficits in a germinal matrix hemorrhage rat model. Brain Res. 1602:127–135. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Amenta PS, Jallo JI, Tuma RF and Elliott MB: A cannabinoid type 2 receptor agonist attenuates blood-brain barrier damage and neurodegeneration in a murine model of traumatic brain injury. J Neurosci Res. 90:2293–2305. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Etienne-Manneville S and Hall A: Rho GTPases in cell biology. Nature. 420:629–635. 2002. View Article : Google Scholar : PubMed/NCBI

18 

Gerhard R, John H, Aktories K and Just I: Thiol-modifying phenylarsine oxide inhibits guanine nucleotide binding of Rho but not of Rac GTPases. Mol Pharmacol. 63:1349–1355. 2003. View Article : Google Scholar : PubMed/NCBI

19 

Huang B, Krafft PR, Ma Q, Rolland WB, Caner B, Lekic T, Manaenko A, Le M, Tang J and Zhang JH: Fibroblast growth factors preserve blood-brain barrier integrity through RhoA inhibition after intracerebral hemorrhage in mice. Neurobiol Dis. 46:204–214. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Persidsky Y, Heilman D, Haorah J, Zelivyanskaya M, Persidsky R, Weber GA, Shimokawa H, Kaibuchi K and Ikezu T: Rho-mediated regulation of tight junctions during monocyte migration across the blood-brain barrier in HIV-1 encephalitis (HIVE). Blood. 107:4770–4780. 2006. View Article : Google Scholar : PubMed/NCBI

21 

Tang J, Chen Q, Guo J, Yang L, Tao Y, Li L, Miao H, Feng H, Chen Z and Zhu G: Minocycline attenuates neonatal germin al-matrix-hemorrhage-induced neuroinflammation and brain edema by activating cannabinoid receptor 2. Mol Neurobiol. 53:1935–1948. 2016. View Article : Google Scholar

22 

Yang F, Wang Z, Zhang JH, Tang J, Liu X, Tan L, Huang QY and Feng H: Receptor for advanced glycation end-product antagonist reduces blood-brain barrier damage after intracerebral hemorrhage. Stroke. 46:1328–1336. 2015. View Article : Google Scholar : PubMed/NCBI

23 

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

24 

Li X, Blizzard KK, Zeng Z, DeVries AC, Hurn PD and McCullough LD: Chronic behavioral testing after focal ischemia in the mouse: Functional recovery and the effects of gender. Exp Neurol. 187:94–104. 2004. View Article : Google Scholar : PubMed/NCBI

25 

Manaenko A, Chen H, Kammer J, Zhang JH and Tang J: Comparison Evans Blue injection routes: Intravenous versus intraperitoneal, for measurement of blood-brain barrier in a mice hemorrhage model. J Neurosci Methods. 195:206–210. 2011. View Article : Google Scholar

26 

Ma Q, Huang B, Khatibi N, Rolland WN II, Suzuki H, Zhang JH and Tang J: PDGFR-alpha inhibition preserves blood-brain barrier after intracerebral hemorrhage. Ann Neurol. 70:920–931. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Cheng CC, Lai YC, Lai YS, Hsu YH, Chao WT, Sia KC, Tseng YH and Liu YH: Transient knockdown-mediated deficiency in plectin alters hepatocellular motility in association with activated FAK and Rac1-GTPase. Cancer Cell Int. 15:292015. View Article : Google Scholar : PubMed/NCBI

28 

Wang L, Fan W, Cai P, Fan M, Zhu X, Dai Y, Sun C, Cheng Y, Zheng P and Zhao BQ: Recombinant ADAMTS13 reduces tissue plasminogen activator-induced hemorrhage after stroke in mice. Ann Neurol. 73:189–198. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Vose LR, Vinukonda G, Jo S, Miry O, Diamond D, Korumilli R, Arshad A, Zia MT, Hu F, Kayton RJ, et al: Treatment with thyroxine restores myelination and clinical recovery after intra-ventricular hemorrhage. J Neurosci. 33:17232–17246. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Zhou Y, Wang Y, Wang J, Anne Stetler R and Yang QW: Inflammation in intracerebral hemorrhage: From mechanisms to clinical translation. Prog Neurobiol. 115:25–44. 2014. View Article : Google Scholar

31 

Yang MC, Zhang HZ, Wang Z, You FL and Wang YF: The molecular mechanism and effect of cannabinoid-2 receptor agonist on the blood-spinal cord barrier permeability induced by ischemia-reperfusion injury. Brain Res. 1636:81–92. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Aijaz S, Balda MS and Matter K: Tight junctions: Molecular architecture and function. Int Rev Cytol. 248:261–298. 2006. View Article : Google Scholar : PubMed/NCBI

33 

Kurihara R, Tohyama Y, Matsusaka S, Naruse H, Kinoshita E, Tsujioka T, Katsumata Y and Yamamura H: Effects of peripheral cannabinoid receptor ligands on motility and polarization in neutrophil-like HL60 cells and human neutrophils. J Biol Chem. 281:12908–12918. 2006. View Article : Google Scholar : PubMed/NCBI

34 

Rom S, Zuluaga-Ramirez V, Dykstra H, Reichenbach NL, Pacher P and Persidsky Y: Selective activation of cannabinoid receptor 2 in leukocytes suppresses their engagement of the brain endothelium and protects the blood-brain barrier. Am J Pathol. 183:1548–1558. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Correa F, Mestre L, Docagne F and Guaza C: Activation of cannabinoid CB2 receptor negatively regulates IL-12p40 production in murine macrophages: Role of IL-10 and ERK1/2 kinase signaling. Br J Pharmacol. 145:441–448. 2005. View Article : Google Scholar : PubMed/NCBI

36 

Montecucco F, Lenglet S, Braunersreuther V, Burger F, Pelli G, Bertolotto M, Mach F and Steffens S: CB cannabinoid receptor activation is cardioprotective in a mouse model of ischemia/reperfusion. J Mol Cell Cardiol. 46:612–620. 2009. View Article : Google Scholar : PubMed/NCBI

37 

Sophocleous A, Marino S, Logan JG, Mollat P, Ralston SH and Idris AI: Bone cell-autonomous contribution of type 2 cannabinoid receptor to breast cancer-induced osteolysis. J Biol Chem. 290:22049–22060. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Gomez O, Sanchez-Rodriguez MA, Ortega-Gutierrez S, Vazquez-Villa H, Guaza C, Molina-Holgado F and Molina-Holgado E: A basal tone of 2-arachidonoylglycerol contributes to early oligodendrocyte progenitor proliferation by activating phosphatidylinositol 3-Kinase (PI3K)/AKT and the mammalian target of rapamycin (MTOR) pathways. J Neuroimmune Pharmacol. 10:309–317. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2018
Volume 42 Issue 5

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang Z, Li Y, Cai S, Li R and Cao G: Cannabinoid receptor 2 agonist attenuates blood‑brain barrier damage in a rat model of intracerebral hemorrhage by activating the Rac1 pathway. Int J Mol Med 42: 2914-2922, 2018
APA
Wang, Z., Li, Y., Cai, S., Li, R., & Cao, G. (2018). Cannabinoid receptor 2 agonist attenuates blood‑brain barrier damage in a rat model of intracerebral hemorrhage by activating the Rac1 pathway. International Journal of Molecular Medicine, 42, 2914-2922. https://doi.org/10.3892/ijmm.2018.3834
MLA
Wang, Z., Li, Y., Cai, S., Li, R., Cao, G."Cannabinoid receptor 2 agonist attenuates blood‑brain barrier damage in a rat model of intracerebral hemorrhage by activating the Rac1 pathway". International Journal of Molecular Medicine 42.5 (2018): 2914-2922.
Chicago
Wang, Z., Li, Y., Cai, S., Li, R., Cao, G."Cannabinoid receptor 2 agonist attenuates blood‑brain barrier damage in a rat model of intracerebral hemorrhage by activating the Rac1 pathway". International Journal of Molecular Medicine 42, no. 5 (2018): 2914-2922. https://doi.org/10.3892/ijmm.2018.3834