Protective effects of geniposide and ginsenoside Rg1 combination treatment on rats following cerebral ischemia are mediated via microglial microRNA‑155‑5p inhibition

  • Authors:
    • Jun Wang
    • Dan Li
    • Jincai Hou
    • Hongtao Lei
  • View Affiliations

  • Published online on: December 7, 2017     https://doi.org/10.3892/mmr.2017.8221
  • Pages: 3186-3193
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Geniposide, an active component of Gardenia, has been reported to protect against cerebral ischemia in animals. Ginsenoside Rg1, a component of Panax notoginseng, is usually administered in combination with Gardenia for the treatment of acute ischemic stroke; however, there are unknown effects of ginsenoside Rg1 that require further investigation. In the present study, the effects of geniposide and ginsensoide Rg1 combination treatment on focal cerebral ischemic stroke were investigated. For in vivo analysis, male rats were separated into three groups, including the (control), model and geniposide + ginsenoside Rg1 groups (n=8 per group). A middle cerebral artery occlusion model was established as the model group. The treatment group was treated with geniposide (30 mg/kg, tail vein injection) + ginsenoside Rg1 (6 mg/kg, tail vein injection), and the model group received saline instead. Neurobehavioral deficits, infarct volume, brain edema, and the expression of microRNA (miR)‑155‑5p and CD11b by reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) and immunohistochemistry, were assessed following 24 h of ischemia. For in vitro analysis, BV2 mouse microglial cells were cultured and exposed to geniposide (40 µg/ml) + ginsenoside Rg1 (8 µg/ml) during various durations of oxygen‑glucose deprivation (OGD). The expression levels of miR‑155‑5p, pri‑miR‑155 and pre‑miR‑155 were detected by RT‑qPCR. The results demonstrated that increases in brain infarct volume, edema volume, CD11b‑positive cells and miR‑155‑5p levels were alleviated following geniposide + ginsenoside administration in rats exposed to ischemia. Furthermore, geniposide + ginsenoside Rg1 treatment suppressed the miR‑155‑5p, pri‑miR‑155 and pre‑miR‑155 expression levels in OGD‑injured BV2 microglial cells. The results of the present study demonstrated that tail vein administration of geniposide in combination with ginsenoside Rg1 protected against focal cerebral ischemia in rats through inhibition of microglial miR‑155‑5p following ischemic injury, which may serve as a novel therapeutic agent for the treatment of strokes.

Introduction

Ischemic stroke caused by blood vessel occlusion is a major cause of morbidity and mortality in humans. Numerous pathological processes are associated with ischemic neuronal damage, including inflammation, energy metabolism disturbance, necrotic and apoptotic cell death, and excitotoxicity (1). Previous studies have aimed to improve the understanding of the underlying mechanisms of ischemic stroke; however, further investigation is required to identify a potential target for the effective prevention and treatment of stroke. Recent clinical and experimental studies have demonstrated that alternative complementary medicine may exhibit a unique efficacy in the treatment of stroke (2,3).

MicroRNA (miRNA/miR), which are non-coding RNAs, are frequently employed in research and are characterized by their regulatory effects on gene expression by inhibiting mRNA translation and/or mediating mRNA degradation (4,5). As numerous pathways are implicated in cerebral ischemic injury, including oxidative stress, excitotoxicity, mitochondrial dysfunction and inflammation, miRNAs have been associated with cerebral ischemic injury due to their roles in ischemic processes, including neuronal cell differentiation (6,7), dendritic plasticity and neuronal outgrowth modulation (8). As a multifunctional miRNA, miR-155 expression is affected by cerebral ischemia (911) and has been reported to be involved in the regulation of a variety of physiological and pathological processes, including inflammation and immunity (12,13). Several inflammatory and cytotoxic factors, including tumor necrosis factor-α, interleukin (IL)-1β and IL-6, secreted by activated microglia may exert an effect on neuronal cell survival (14). miR-155 may regulate the immune response mediated by microglia in various pathological states (1517).

Gardenia is the fruit of Gardenia jasminoides Ellis (Rubiaceae), which is a commonly used Chinese herb. Gardenia has been reported to exhibit therapeutic effects on hepatic disorders, brain diseases, inflammatory diseases and contusions (1820). A previous study reported that geniposide, an active component of Gardenia, reduces neuronal cell death and increases cell viability following oxygen and glucose deprivation (OGD) (21). Our previous study also revealed that geniposide exhibits protective effects against infarction volume, microglial activation and inflammatory cytokine release in middle cerebral artery occlusion (MCAO) model rats (22). In clinical and experimental research, Gardenia has previously been used in combination with Panax notoginseng to treat brain diseases, including cerebrovascular disease and Alzheimer's disease (23,24). For rats injured by local ischemia, ginsenoside Rg1, a component of Panax notoginseng, was reported to alleviate neurological function deficits, as determined by improvements in the learning and memory abilities, in addition to a reduction in the apoptosis of neurons in the hippocampus CA1 zone (25,26). The present study aimed to investigate whether tail vein-administered geniposide in combination with ginsenoside Rg1 may protect against ischemic stroke via miR-155 inhibition. An in vivo investigation into miR-155-5p expression, and the effects of geniposide + ginsenoside Rg1 on miR-155-5p expression within the brain and activation of microglia, as determined by immunohistochemistry for CD11b, was performed. In addition, an in vitro study was performed to analyze the effects of geniposide + ginsenoside Rg1 on miR-155-5p expression in BV2 microglial cells.

Materials and methods

Animals and grouping

Specific pathogen and virus antibody-free grade male Sprague-Dawley rats (n=56; 253.4±20.3 g; 7–8 weeks-old) were obtained from Beijing Vital River Laboratory Animal Technology Co., Ltd. (Beijing, China). In accordance with the UK Animals (Scientific Procedures) Act of 1986 (27), the animal experiments were performed with the approval of the ethics committee of the Institute of Basic Theory, China Academy of Chinese Medical Sciences (Beijing, China). The rats were housed in rooms maintained at 23±2°C in a 12 h light/dark cycle and fed a rodent standard diet with free access to water. The animals were randomly divided into three groups (n=8 per group): Sham, model and geniposide (30 mg/kg) + ginsenoside Rg1 (6 mg/kg) groups. For the sham group, the rats underwent similar surgery to the model group, without nylon filament insertion; rats in the model group were administrated saline via the tail vein immediately following MCAO. Rats in the geniposide + ginsenoside Rg1 group were administered geniposide (30 mg/kg) + ginsenoside Rg1 (6 mg/kg) via the tail vein immediately following MCAO. Geniposide (purity, >95%; cat. no. 110749-201718) and ginsenoside Rg1 (purity, >95%; cat. no. 110703-201731) were purchased from the National Institutes for Food and Drug Control (Beijing, China).

MCAO model establishment

The MCAO model was established as described by Longa et al (28). Rats were anesthetized with 10% chloral hydrate (350 mg/kg body weight, intraperitoneal injection) (29,30). Subsequently, a midline incision was made to expose the left common carotid artery and the external carotid artery (ECA), including the occipital artery branches. Following this, blood from the occipital artery and superior thyroid artery branches of the ECA was coagulated using a bipolar electrocoagulator. The internal carotid artery (ICA) was isolated and carefully separated from the adjacent vagus nerve. Following ICA exposure, the origin of the left middle cerebral artery was occluded using a 0.25 mm diameter nylon filament coated with poly-L-lysine, which was inserted into the ICA via the ECA. Rectal temperature was maintained at 37°C during and following surgery with a temperature-controlled heating pad (CMA 150 Carnegie Medicin AB, Solna, Sweden). After 2, 6 and 24 h, the ischemic procedure was complete.

Infarct size and brain edema detection

Rats of the 24 h ischemia group were selected for in vivo analysis; statistical calculations demonstrated no significant difference when comparing the 2 and 6 h ischemia group rats with the control group. Following the performance of ischemic injury for different time periods, all animals were sacrificed. Brain tissues were removed and prepared into 2 mm coronal sections. The sections were stained with 1.5% 2,3,5-triphenyltetrazolium chloride at 37°C for 10 min and fixed in 4% paraformaldehyde for 24 h at room temperature. The images of the stained slices were analyzed using Image-Pro Plus 6.0 (Media Cybernetics, Inc., Rockville, MD, USA) to determine the infarct size, which was expressed as the percentage of the whole brain tissue. Brain edema was calculated according to the following equation: [(volume of ipsilateral hemisphere-volume of contralateral hemisphere)/volume of contralateral hemisphere] ×100. The experiment was independently repeated three times.

Cell culture and in vitro model

Mouse BV2 microglial cells were purchased from the Institute of Basic Medical Sciences of the China Science Academy (Beijing, China) and cultured in Dulbecco's modified Eagle medium/Ham's F12 (DMEM/F12; Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) culture medium containing 10% fetal bovine serum (Thermo Fisher Scientific, Inc.) and 1% penicillin and streptomycin (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) in a humidified 5% CO2/95% air atmosphere at 37°C. The microglial cells were passaged at 90% confluence, with two to three passages per week. To mimic the OGD pathological process, glucose-free DMEM (Gibco; Thermo Fisher Scientific, Inc.) was used instead of the culture medium and oxygen was restricted in the flasks by keeping the culture flasks in a sealed tank with a persistent low-flow (1.5 l/min) of 95% N2 and 5% CO2 mixture. The inlet and outlet ends of the tubes were then clipped and the sealed tank was returned to the incubator. DMEM was used to dissolve geniposide and ginsenoside Rg1 and prepare the storage solution. A total of 40 µg/ml geniposide + 8 µg/ml ginsenoside Rg1 was dissolved within the culture media. After 4, 8 and 16 h of OGD exposure at 37°C, with or without geniposide (40 µg/ml) + ginsenoside Rg1 (8 µg/ml), BV2 cells were collected for the analysis of miR-155-5p expression. The experiment was independently repeated three times.

miR-155-5p, pri-miR-155 and pre-miR-155 quantification by reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA of brain tissue and cultured BV2 microglial cells was isolated and analyzed using a Reverse Transcriptase kit (Suzhou GenePharma, Co., Ltd., Suzhou, China). The temperature and duration of reverse transcription was performed at 30°C for 10 min, 42°C for 30 min and 99°C for 5 min. A stem-loop real-time PCR system was used to determine the level of miRNAs using the Maxima SYBR Green qPCR Master Mix (Fermentas; Thermo Fisher Scientific, Inc., Waltham MA, USA) and a StepOne Sequence Detector (Applied Biosystems; Thermo Fisher Scientific, Inc.). qPCR was performed at 95°C for 10 min followed by 40 cycles at 95°C for 15 sec and 60°C for 60 sec. The following primer sequences were used: miR-155 forward, 5′-GCTTCGGTTAATGCTAATCGTG-3′; miR-155 reverse, 5′-AGAGCAGGGTCCGAGGTA-3′; U6 forward, 5′-ATTGGAACGATACAGAGAAGATT-3′; U6 reverse, 5′-GGAACGCTTCACGAATTTG-3′; pre-miR-155 forward, 5′-TGCTAATTGTGATAGGGGTTTT-3′; pre-miR-155 reverse, 5′-TATGGTTGTTCACGACTCCTTCAC-3′; pri-miR-155 forward, 5′-AGGCTTTTCCTGGGCACC-3′; and pri-miR-155 reverse, 5′-CATGAACAAACCACAACGAGC-3′. RT-qPCR was performed in duplicate and relative expression of miR-155-5p, pri-miR-155 and pre-miR-155 were normalized to U6 expression and determined by the 2−ΔΔCq method (31). The experiment was independently repeated three times.

Immunohistochemistry

Brain tissues from rats were removed for immunohistochemistry analysis at the termination of ischemia. Brain tissues were fixed in 4% paraformaldehyde overnight at room temperature. Sections (5 µm) embedded in paraffin were deparaffinized in Histo-Clear (HS-200; National Diagnostics, Atlanta, GA, USA) rehydrated and washed with 0.01 M PBS; 3% hydrogen peroxide in methanol was used at room temperature for 30 min to inhibit the endogenous peroxidase. Sections were incubated with rabbit anti-CD11b (1:500) antibody (cat. no. ab133357; Abcam, Cambridge, MA) at 4°C overnight following blocking of nonspecific antigens with goat serum (Gibco; Thermo Fisher Scientific, Inc.) at 37°C for 20 min. Subsequently, sections were incubated with horseradish peroxidase-conjugated secondary antibodies at 37°C for 2 h. Peroxidase detection was performed using a mixture containing 0.05% 3,3′-diaminobenzidine. Following staining, all sections were coverslipped and analyzed. The immunohistochemistry images were acquired using an Olympus Fluoview FV1000 microscope (LSI3-FV1000-Inverted; Olympus Corporation, Tokyo, Japan) and Image-Pro Plus 6.0 software (Media Cybernetics, Inc.). The experiment was independently repeated three times.

Statistical analysis

Data are presented as the mean + standard deviation. The statistical analysis was conducted with SPSS software (SPSS, Inc. Chicago, IL, USA). Significant differences among the groups were determined by one-way analysis of variance, followed by a Student-Newman-Keuls post-hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

miR-155-5p expression in rats

In the present study, a rat MCAO model was established. miR-155-5p expression levels increased in a time-dependent manner; miR-155-5p expression levels were significantly increased in the ipsilateral brain tissue of rats in the 24 h ischemia group compared with the sham group (P<0.01; Fig. 1A). The Cq values of 2, 6 and 24 h ischemia groups increased by 1.42-, 1.62- and 2.94-fold, respectively, compared with the sham group (Fig. 1A). To investigate whether the increase of miR-155-5p in the brain was region specific, miR-155-5p expression levels were observed within various brain regions at 24 h post-ischemia. The results demonstrated that miR-155-5p expression was significantly increased in the cortex and basal ganglia of the ipsilateral brain in MCAO models, compared with in the sham group (P<0.05; Fig. 1B and C). However, a significant increase was not observed in the expression levels of miR-155-5p within the hippocampus of MCAO model rats, compared with in the sham group (Fig. 1D). Treatment with geniposide + ginsenoside Rg1 attenuated the miR-155-5p Cq value markedly compared with the model group (1.67±0.52 and 2.93±0.87, respectively; P<0.05; Fig. 1E). However, geniposide + ginsenoside Rg1 had no effect on the miR-155-5p expression levels in the basal ganglia of MCAO model rats (Fig. 1F).

Geniposide + ginsenoside Rg1 protects brain tissue against ischemic injury

To assess the in vivo protective effect of geniposide + ginsenoside Rg1 against ischemic injury, the percentage of cerebral infarction and edema was investigated within an MCAO rat model. Infarction was not observed in the sham group. Cerebral infarction within the model group was observed primarily in the frontal and parietal lobe of the cortex, the caudate nucleus and posterior limbs of the internal capsule (Fig. 2A). The results revealed that geniposide + ginsenoside Rg1 treatment significantly reduced brain infarction size and edema volume compared with the model group (P<0.05). In particular, the percentage of cerebral infarction and brain edema of rats subjected to geniposide + ginsenoside Rg1 was decreased by 34.2 and 42.6%, respectively, compared with the MCAO model group (Fig. 2B and C).

Geniposide + ginsenoside Rg1 inhibits microglia stimulation

Immunohistochemistry demonstrated low levels of CD11b expression within the sham group (Fig. 3A). However, after 24 h of ischemia, CD11b immunoreactivity was visibly increased in the model group (Fig. 3B), which was primarily present in the cortex region, indicating a marked activation of microglial cells within ischemic penumbra. The positive staining of CD11b was primarily located within the cell membranes, while the microglial morphology altered from branch- to amoeba-like, indicating a marked stimulation. Following treatment with geniposide + ginsenoside Rg1, the expression of CD11b was suppressed in the brain parenchyma, compared with the model group (Fig. 3C), as indicated by a reduced number of CD11b-positive cells in the ischemic penumbra region. Semi-quantitative analysis revealed the relative gray values of immunohistochemical staining (Fig. 3D), which indicated that the number of CD11b-positive cells was markedly reduced by geniposide + ginsenoside Rg1 treatment in MCAO model rats (P<0.05).

miR-155-5p expression within BV2 microglial cells

To further investigate the protective effects of geniposide + ginsenoside Rg1 against ischemic injury in vitro, BV2 microglial cells were cultured and the levels of immature forms of miR-155 (pri-miR-155 and pre-miR-155) and mature miR-155-5p were observed by RT-qPCR. As presented in Fig. 4A, miR-155-5p expression levels within the OGD 4 h group were not markedly altered compared with in control group. However, when the duration of ischemic was extended to 8 or 16 h, miR-155-5p expression levels within BV2 microglial cells were significantly increased compared with the control group (P<0.001). Consequently, OGD 8 h was selected as the duration for stimulation for subsequent analysis of BV2 cells. Treatment of cells with geniposide + ginsenoside Rg1 for 8 h significantly decreased the expression levels of the immature and mature forms of miR-155. In particular, miR-155-5p expression within BV2 microglial cells incubated with geniposide + ginsenoside Rg1 was reduced by 93.9% compared with the model group (Fig. 4B; P<0.01). In addition, following treatment with geniposide + ginsenoside Rg1, the expression levels of pri-miR-155 and pre-miR-155, the immature forms of miR-155, were significantly inhibited by 97.9 and 97%, respectively, compared with the model group (P<0.01; Fig. 4C and D).

Discussion

Ischemic stroke leads to a series of complex, multipart-cascade events that contribute to irreversible brain injury and neurovascular network dysfunction within the ischemic core (32). Activation of microglia and astrocytes following stroke leads to enhanced release of proinflammatory factors, which may persist for several weeks (33). Although the immune responses mediated by microglia serve an important role in pathogen clearance and tissue regeneration, without treatment, the inflammatory state may worsen neuronal injury (16). miRNAs are a type of small non-coding RNA that regulate target gene expression and are highly conserved in various eukaryotes (34). Numerous miRNAs have been reported to be involved in the regulation of several inflammation phases and responses to cerebral ischemia (3537). Of these miRNAs, miR-155 has been demonstrated to be associated with the regulation of various physiological and pathological processes, including inflammation, immunity, cancer and cardiovascular diseases (12,13). As a critical inflammation-associated miRNA, miR-155 regulates the host immune response by repressing the expression of target genes (38). The majority of target genes of miR-155 are reported to be associated with the inflammatory regulation process. For example, it has been demonstrated that inflammatory responses mediated by miR-155 in ischemic cerebral tissues were modulated by Toll-like receptor (TLR)4/myeloid differentiation primary response gene 88 and suppressor of cytokine signaling 1 expression, while acetylbritannilactone may suppress miR-155 expression and inhibit inflammatory actions (39). In addition, an association was reported between the expression of TLRs 3, 7 and 9, and miR-155 transcription, in the olfactory bulbs (40). Based on the reported associations between miR-155 and post-ischemic brain inflammation, therapeutic approaches that target miR-155 may be promising for the treatment of cerebral ischemic damage.

Gardenia, the fruit of Gardenia jasminoides Ellis, exhibits varying concentrations of iridoid glycosides. The root of Panax notoginseng (Burk.) F. H. Chen is a widely used traditional Chinese medicine that belongs to the genus Panax and family Araliaceae. Panax notoginseng is also termed Radix notoginseng or Sanchi. In clinical and experimental research, Gardenia is usually used in combination with Panax notoginseng to treat central nervous system diseases, including cerebrovascular disease and Alzheimer's disease. Geniposide and ginsenoside Rg1 are the active components of Gardenia and Panax notoginseng, respectively. In a carrageenan-induced rat paw edema model, Gardenia and Panax notoginseng have been reported to exhibit anti-inflammatory effects via the inhibition of nitric oxide production (41). These compounds were also demonstrated to ameliorate neurological injury, brain infarct volume and the permeability of the blood-brain barrier in MCAO rat models by reducing the expression of proteinase-activated receptor 1 (42). However, studies concerning the role of geniposide + ginsenoside Rg1 have predominantly been performed in neurons; to the best of our knowledge, investigation regarding their combined effects on microglial cells is limited.

The results of the present study indicated that ischemia caused severe damage in cerebral tissue with a large area of cerebral infarction and brain edema. Geniposide combined with ginsenoside Rg1 reduced the percentage of cerebral infarction size and brain edema in rats subjected to MCAO, which demonstrated a protective effect on ischemia-induced cerebral injury in vivo. RT-qPCR revealed that miR-155 expression levels were unaltered in the 2 and 6 h ischemia groups; however, when the duration of ischemia was prolonged to 24 h, miR-155 expression levels increased within the brain tissue, compared with the sham group. Notably, miR-155 expression levels were increased in the cortex and basal ganglia in MCAO rats, compared with the sham group, indicating the potential of miR-155 overexpression as a target for the treatment of cerebral ischemic injury. Rats treated with geniposide combined with ginsenoside Rg1 demonstrated significantly attenuated expression levels of miR-155 expression levels. Based on the results for infarction size and brain edema, the neuroprotective effects of geniposide + ginsenoside Rg1 may be associated with the downregulation of miR-155. In the present study, 10% chloral hydrate was employed for anesthesia during the course of MCAO surgery, according to the previously described method (29,30) in which all mice or rats were treated with 10% chloral hydrate for anesthesia; however, no symptoms of peritonitis were determined in the present study. In the present study, only the brain tissue was dissected for observation and the abdominal cavity was not observed as there were no obvious symptoms of peritonitis in rats. However, a previous study reported that intraperitoneal administration of 10% chloral hydrate resulted in adynamic ileus and peritonitis in rats, gastric ulcers in rats and peritonitis in swine (43). Therefore, in future experiments, we plan to change to gas anesthesia with isoflurane.

To the best of our knowledge, activated microglia become hypertrophic, rapidly proliferate and migrate to the inflammatory area where they generate excessive amounts of proinflammatory and neurotoxic cytokines that cause neuronal damage (44,45). In the current study, the results of immunohistochemistry demonstrated that microglial cells increased in number and appeared to possess an activated morphology within ischemic-injured regions. Geniposide + ginsenoside Rg1 markedly reversed the CD11b expression, indicating an inhibitory effect of geniposide + ginsenoside Rg1 on microglial cells. As microglial miR-155 expression serves an important role in the pathological process following ischemic stroke, an in vitro experiment was performed to observe the effects of geniposide + ginsenoside Rg1 on microglial cells. The results demonstrated that miR-155 expression levels were elevated and microglial cells were activated following OGD injury for 8 h, compared with the control group, indicating microglial activation and high inflammatory potential following OGD. However, geniposide + ginsenoside Rg1 reduced the expression of mature miR-155 and the immature precursors, pri-miR-155 and pre-miR-155, indicating a potential protective role of geniposide + ginsenoside Rg1 in microglial activation. Geniposide + ginsensoside Rg1 treatment may activate microglial cells to release inflammatory factors. miR-155 is an important miRNA that regulates the mRNA expression of inflammatory factors; based on the inhibitory effect on miR-155 in the current study, the anti-inflammatory effect of geniposide + ginsenoside Rg1 may be achieved by inhibiting miR-155 expression.

In conclusion, the in vivo and in vitro experiments of the present study demonstrated that the administration of geniposide in combination with ginsenoside Rg1 via tail vein injection inhibited ischemia-induced microglial cell activation, which may be associated with the inhibition of microglial miR-155-5p expression. The results of the present study indicated that geniposide in combination with ginsenoside Rg1 may serve as a potential alternative for the prevention of microglial cell injury in ischemic diseases.

Acknowledgements

The present study was supported by the Natural Science Foundation in China (grant no. 81473449).

References

1 

Xu F, Li J, Ni W, Shen YW and Zhang XP: Peroxisome proliferator-activated receptor-gamma agonist 15d-prostaglandin J2 mediates neuronal autophagy after cerebral ischemia-reperfusion injury. PLoS One. 8:e550802013. View Article : Google Scholar : PubMed/NCBI

2 

Cheng CY and Lee YC: Anti-Inflammatory effects of traditional chinese medicines against ischemic injury in in vivo models of cerebral ischemia. Evidence Based Complement Alternat Med. 2016:57394342016. View Article : Google Scholar

3 

Liu X, Tao Y, Wang F, Yao T, Fu C, Zheng H, Yan Y, Liang X, Jiang X and Zhang Y: Kudiezi injection mitigates myocardial injury induced by acute cerebral ischemia in rats. BMC Complement Alternat Med. 17:82017. View Article : Google Scholar

4 

Ambros V: The functions of animal microRNAs. Nature. 431:350–355. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Bartel DP: MicroRNAs: Target recognition and regulatory functions. Cell. 136:215–233. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Lau P and Hudson LD: MicroRNAs in neural cell differentiation. Brain Res. 1338:14–19. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Rybak A, Fuchs H, Smirnova L, Brandt C, Pohl EE, Nitsch R and Wulczyn FG: A feedback loop comprising lin-28 and let-7 controls pre-let-7 maturation during neural stem-cell commitment. Nat Cell Biol. 10:987–993. 2008. View Article : Google Scholar : PubMed/NCBI

8 

White RE and Giffard RG: MicroRNA-320 induces neurite outgrowth by targeting ARPP-19. Neuroreport. 23:590–595. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Lim KY, Chua JH, Tan JR, Swaminathan P, Sepramaniam S, Armugam A, Wong PT and Jeyaseelan K: MicroRNAs in Cerebral Ischemia. Transl Stroke Res. 1:287–303. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Liu DZ, Tian Y, Ander BP, Xu H, Stamova BS, Zhan X, Turner RJ, Jickling G and Sharp FR: Brain and blood microRNA expression profiling of ischemic stroke, intracerebral hemorrhage, and kainate seizures. J Cereb Blood Flow Metab. 30:92–101. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Liu Y, Zhang J, Han R, Liu H, Sun D and Liu X: Downregulation of serum brain specific microRNA is associated with inflammation and infarct volume in acute ischemic stroke. J Clin Neurosci. 22:291–295. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Faraoni I, Antonetti FR, Cardone J and Bonmassar E: miR-155 gene: A typical multifunctional microRNA. Biochim Biophys Acta. 1792:497–505. 2009. View Article : Google Scholar : PubMed/NCBI

13 

O'Connell RM, Kahn D, Gibson WS, Round JL, Scholz RL, Chaudhuri AA, Kahn ME, Rao DS and Baltimore D: MicroRNA-155 promotes autoimmune inflammation by enhancing inflammatory T cell development. Immunity. 33:607–619. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Dheen ST, Kaur C and Ling EA: Microglial activation and its implications in the brain diseases. Curr Med Chem. 14:1189–1197. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Butovsky O, Jedrychowski MP, Cialic R, Krasemann S, Murugaiyan G, Fanek Z, Greco DJ, Wu PM, Doykan CE, Kiner O, et al: Targeting miR-155 restores abnormal microglia and attenuates disease in SOD1 mice. Ann Neurol. 77:75–99. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Cardoso AL, Guedes JR, Pereira de Almeida L and Pedroso de Lima MC: miR-155 modulates microglia-mediated immune response by down-regulating SOCS-1 and promoting cytokine and nitric oxide production. Immunology. 135:73–88. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Pareek S, Roy S, Kumari B, Jain P, Banerjee A and Vrati S: MiR-155 induction in microglial cells suppresses Japanese encephalitis virus replication and negatively modulates innate immune responses. J Neuroinflammation. 11:972014. View Article : Google Scholar : PubMed/NCBI

18 

Chen QC, Zhang WY, Kim H, Lee IS, Ding Y, Youn UJ, Lee SM, Na M, Min BS and Bae K: Effects of Gardeniae Fructus extract and geniposide on promoting ligament cell proliferation and collagen synthesis. Phytother Res. 24 Suppl 1:S1–S5. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Wang J, Li PT, Du H, Hou JC, Li WH, Pan YS and Chen HC: Tong Luo Jiu Nao injection, a traditional Chinese medicinal preparation, inhibits MIP-1β expression in brain microvascular endothelial cells injured by oxygen-glucose deprivation. J Ethnopharmacol. 141:151–157. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Wang SW, Lai CY and Wang CJ: Inhibitory effect of geniposide on aflatoxin B1-induced DNA repair synthesis in primary cultured rat hepatocytes. Cancer Lett. 65:133–137. 1992. View Article : Google Scholar : PubMed/NCBI

21 

Lee P, Lee J, Choi SY, Lee SE, Lee S and Son D: Geniposide from Gardenia jasminoides attenuates neuronal cell death in oxygen and glucose deprivation-exposed rat hippocampal slice culture. Biol Pharm Bull. 29:174–176. 2006. View Article : Google Scholar : PubMed/NCBI

22 

Wang J, Hou J, Zhang P, Li D, Zhang C and Liu J: Geniposide reduces inflammatory responses of oxygen-glucose deprived rat microglial cells via inhibition of the TLR4 signaling pathway. Neurochem Res. 37:2235–2248. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Zhang Y, Lin SX, Hua Q, Yang KY, Yao N, Yi LS, Wang AM, Chen WJ and Chen JY: Research on the mechanism of the active ingredients of Sanchi and Gardenia removing the early amyloid in the AD transgenic mouse brain. Chin Pharmacol Bull. 28:179–84. 2012.(In Chinese).

24 

Zhai YS, Du SY, Lu Y, Wang Y, Xu B and Gao Y: Impact of notoginseng total saponin on intestinal absorption kinetics of jasminoidin. China J Trad Chin Med Pharm. 25:459–462. 2010.

25 

Lanou W, Heqin Z, Wenjun H and Wenfeng C: Analysis of behavioral index of ginsenoside_RG 1 intervention on brain ischemia. Chin J Lab Ani Sci. 14:283–285. 2004.

26 

Xia L, Cuifen B, Jia W, Jia L and Shujian Q: The protective role and mechanism of Ginsenoside Rg1 on the ischemic-reperfusion injuried neurons in hippocampus CA1 of rat. Progress Anat Sci. 16:177–180. 2010.

27 

Hollands C: The animals (scientific procedures) act 1986. Lancet. 2:32–33. 1986. View Article : Google Scholar : PubMed/NCBI

28 

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

29 

Liu JH, Feng D, Zhang YF, Shang Y, Wu Y, Li XF and Pei L: Chloral hydrate preconditioning protects against ischemic stroke via upregulating annexin A1. CNS Neuroscience Ther. 21:718–726. 2015. View Article : Google Scholar

30 

Chang CF, Lin SZ, Chiang YH, Morales M, Chou J, Lein P, Chen HL, Hoffer BJ and Wang Y: Intravenous administration of bone morphogenetic protein-7 after ischemia improves motor function in stroke rats. Stroke. 3:558–564. 2003. View Article : Google Scholar

31 

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

32 

Brouns R and De Deyn PP: The complexity of neurobiological processes in acute ischemic stroke. Clin Neurol Neurosurg. 111:483–495. 2009. View Article : Google Scholar : PubMed/NCBI

33 

Liguz-Lecznar M and Kossut M: Influence of inflammation on poststroke plasticity. Neural Plast. 2013:2585822013. View Article : Google Scholar : PubMed/NCBI

34 

Taganov KD, Boldin MP and Baltimore D: MicroRNAs and immunity: Tiny players in a big field. Immunity. 26:133–137. 2007. View Article : Google Scholar : PubMed/NCBI

35 

O'Connell RM, Rao DS, Chaudhuri AA and Baltimore D: Physiological and pathological roles for microRNAs in the immune system. Nat Rev Immunol. 10:111–122. 2010. View Article : Google Scholar : PubMed/NCBI

36 

Xiao C and Rajewsky K: MicroRNA control in the immune system: Basic principles. Cell. 136:26–36. 2009. View Article : Google Scholar : PubMed/NCBI

37 

Zhu J, Yue H, Qiao C and Li Y: Association between single-nucleotide polymorphism (SNP) in miR-146a, miR-196a2, and miR-499 and risk of ischemic stroke: A meta-analysis. Med Sci Monit. 21:3658–3663. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Wang J, Yang K, Zhou L, Minhaowu, Wu Y, Zhu M, Lai X, Chen T, Feng L, Li M, et al: MicroRNA-155 promotes autophagy to eliminate intracellular mycobacteria by targeting Rheb. PLoS Pathog. 9:e10036972013. View Article : Google Scholar : PubMed/NCBI

39 

Wen Y, Zhang X, Dong L, Zhao J, Zhang C and Zhu C: Acetylbritannilactone modulates MicroRNA-155-mediated inflammatory response in ischemic cerebral tissues. Mol Med. 21:197–209. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Oliveira BRSM, Vieira FV, De S Vieira D, da Silva SEL, Gameiro R, Flores EF and Cardoso TC: Expression of miR-155 associated with Toll-like receptors 3, 7, and 9 transcription in the olfactory bulbs of cattle naturally infected with BHV5. J Neurovirol. 23:772–778. 2017. View Article : Google Scholar : PubMed/NCBI

41 

Zheng X, Yang D, Liu X, Wang N, Li B, Cao H, Lu Y, Wei G, Zhou H and Zheng J: Identification of a new anti-LPS agent, geniposide, from Gardenia jasminoides Ellis, and its ability of direct binding and neutralization of lipopolysaccharide in vitro and in vivo. Int Immunopharmacol. 10:1209–1219. 2010. View Article : Google Scholar : PubMed/NCBI

42 

Xie CL, Li JH, Wang WW, Zheng GQ and Wang LX: Neuroprotective effect of ginsenoside-Rg1 on cerebral ischemia/reperfusion injury in rats by downregulating protease-activated receptor-1 expression. Life Sci. 121:145–151. 2015. View Article : Google Scholar : PubMed/NCBI

43 

Silverman J and Muir WW III: A review of laboratory animal anesthesia with chloral hydrate and chloralose. Lab Anim Sci. 43:210–216. 1993.PubMed/NCBI

44 

Smith JA, Das A, Ray SK and Banik NL: Role of pro-inflammatory cytokines released from microglia in neurodegenerative diseases. Brain Res Bull. 87:10–20. 2012. View Article : Google Scholar : PubMed/NCBI

45 

Vilhardt F: Microglia: Phagocyte and glia cell. Int J Biochem Cell Biol. 37:17–21. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2018
Volume 17 Issue 2

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
Wang J, Li D, Hou J and Lei H: Protective effects of geniposide and ginsenoside Rg1 combination treatment on rats following cerebral ischemia are mediated via microglial microRNA‑155‑5p inhibition. Mol Med Rep 17: 3186-3193, 2018
APA
Wang, J., Li, D., Hou, J., & Lei, H. (2018). Protective effects of geniposide and ginsenoside Rg1 combination treatment on rats following cerebral ischemia are mediated via microglial microRNA‑155‑5p inhibition. Molecular Medicine Reports, 17, 3186-3193. https://doi.org/10.3892/mmr.2017.8221
MLA
Wang, J., Li, D., Hou, J., Lei, H."Protective effects of geniposide and ginsenoside Rg1 combination treatment on rats following cerebral ischemia are mediated via microglial microRNA‑155‑5p inhibition". Molecular Medicine Reports 17.2 (2018): 3186-3193.
Chicago
Wang, J., Li, D., Hou, J., Lei, H."Protective effects of geniposide and ginsenoside Rg1 combination treatment on rats following cerebral ischemia are mediated via microglial microRNA‑155‑5p inhibition". Molecular Medicine Reports 17, no. 2 (2018): 3186-3193. https://doi.org/10.3892/mmr.2017.8221