更全的杂志信息网

Ginkgolide B promotes the proliferation and differentiation of neural stem cells following cerebral ischemia/reperfusion injury, both in vivo and in vitro

更新时间:2016-07-05

Introduction

Neural stem cells (NSCs) are promising for the development of novel therapies for nervous system diseases. Their presence in the central nervous system indicates that it has the capacity for self-repair. Brain ischemia activates the proliferative potential of resident NSCs (Zhu et al., 2005; Ryu et al., 2016). Recent studies have shown that NSCs are present in the brains of adult humans and animals, and that following brain injury, the proliferation of endogenous NSCs is induced (Alagappan et al., 2009; Xie et al., 2016; Chen et al.,2017). However, the proliferation of endogenous NSCs following brain ischemia is insufficient for central nervous system self-repair. Thus, the use of exogenous drugs to induce self-repair by endogenous NSCs in the brain is a promising therapeutic strategy for promoting neurological recovery and reconstruction after stroke.

Neuronal development, axonal growth, neurotransmitter synthesis and apoptosis in the brain are regulated by multiple factors via signaling pathways. In the absence of exogenous drugs, only a small proportion of proliferative NSCs differentiate into mature neurons (Yu et al., 2016). Thus, inducing the proliferation of endogenous NSCs with exogenous growth factors should promote the migration of NSCs to the site of injury and their differentiation into functional neurons. Nerve growth factor and other growth factors, such as brain-derived neurotrophic factor (BDNF), promote the proliferation and differentiation of NSCs and the formation of protrusions in newly formed neurons (Ochi et al., 2016). Epidermal growth factor (EGF) regulates the proliferation, migration and differentiation of NSCs, and plays a critical role in the maintenance of central nervous system homeostasis (Huang et al., 2016). In addition, a variety of signaling pathways are involved in the regulation of neurite growth, including growth factor-mediated signaling pathways and suppressors of cytokine signaling (SOCS) (Barnat et al., 2016).

中国银行董事长陈四清表示,中国银行作为进口博览会银行类唯一综合服务支持企业,长期坚持“开放促进发展、合作实现共赢”的理念,致力于推动中国与世界各国的互利合作和共同繁荣。

Traditional Chinese medicine has recently been found to enhance neural plasticity in the central nervous system(Maclennan et al., 2002; Wang et al., 2015). For centuries,extracts from leaves of the Ginkgo biloba tree have been used in China for the treatment of a variety of diseases (Gu et al., 2012). Ginkgolide B (GKB) is one of several major terpene lactone components that have been identi fi ed in Ginkgo biloba extracts (Qin et al., 2014), and it has a molecular weight of 424.4 Da (Cui et al., 2012). GKB is a potent platelet activating factor antagonist (Hu et al., 2011). A potentially important property of GKB is that it can pass through the brain-blood barrier, particularly following cerebral ischemia/reperfusion injury (Fang et al., 2010). A number of in vitro and in vivo studies have demonstrated that GKB has marked neuroprotective effects against ischemia-induced impairments (Maclennan et al., 2002; Xia and Fang, 2007;Hu et al., 2011; Gu et al., 2012; Qin et al., 2014). However,the mechanisms underlying the neuroprotective effects of GKB have yet to be clari fi ed (Gu et al., 2012; Qin et al.,2014). The neuroprotective actions of GKB might be related to its anti-inflammatory effects, scavenging of oxygen free radicals, inhibition of thrombosis, and platelet activating factor antagonism (Hu et al., 2011; Gu et al., 2012; Qin et al.,2014).

Tang et al. (2011) found that GKB promotes the proliferation and functional activities of bone marrow-derived endothelial progenitor cells. We hypothesized that GKB might increase the proliferation and differentiation of NSCs in rats with ischemic/reperfusion injury. Therefore, in the present study, we investigate the effects of GKB on the proliferation and morphology of NSCs in rats with focal cerebral ischemia.

Materials and Methods

In vitro experiments

Cell culture

A NSC line was purchased by Cyagen Biological Technology Co., Ltd., China. For differentiation, NSCs were subcultured in Dulbecco’s modified Eagle’s medium (DMEM) (Gibco,New York, NY, USA) containing 20 ng/mL EGF and basic fi broblast growth factor (bFGF). Passage 2—3 neurospheres were harvested by centrifugation and seeded onto coverslips in 12-well plates pre-coated with 100 mg/L polylysine. Cells were randomly assigned to the following four groups: control, 20 mg/L GKB, 40 mg/L GKB and 60 mg/L GKB (6 wells per group). In the control group, cells were maintained in DMEM/F12 (Gibco) containing 10% fetal bovine serum. In the 20, 40 and 60 mg/L GKB groups, cells were maintained in DMEM/F12 containing 10% fetal bovine serum and treated with GKB (lyophilized powder, dissolved in 0.9% saline;China Pharmaceutical and Biological Products, China) at 20, 40 or 60 mg/L. Cells were grown at 37 °C in a humidi fi ed environment with 5% CO2. NSC morphology was observed at 3, 7 and 14 days.

Cell morphology

After cell culture, the coverslips were collected, and the morphology of the NSCs was observed under a phase contrast microscope (Changfang Optical Instrument Co., Shanghai,China). SPOT Advance software (Informer Technologies,Dominica, NY, USA) was used to measure the length of cell protrusions (3 days) and cell body area (14 days) as previously described (Mousavi and Doweidar, 2015).

Rats were sacri fi ced after anesthesia, and their brains were removed. Total RNA was extracted from the penumbra and reverse-transcribed into cDNA. The cDNA was used for quantitative RT-PCR (Ba et al., 2014) for BDNF (forward primer: 5′-TTG TGG TTT GTT GCC GTT GC-3′; reverse primer: 5′-TCC CCA TCC CCT AAG CCA GT-3′) and EGF(forward primer: 5′-CGC CGC AGA CTT ACC CAG A-3′;reverse primer: 5′-CGG AGA CGT ACC CTG TTT TGA C-3′). The thermocycling parameters were as follows: 50°C for 3 minutes, 95°C for 3 minutes; 45 cycles of 95°C for 10 seconds (denaturation), 59°C for 20 seconds (annealing) and 72°C for 20 seconds (extension); and 72°C for 10 minutes(final extension). Data were analyzed with BIO-RAD CFX Manager (Bio-Rad, Hercules, CA, USA) and output into PDF and EXCEL documents. Actin served as an internal reference.Relative expression of the target gene was determined as the ratio of expression of the target gene to that of β-actin.

GKB treatment

为了适应学科教学改革的新形势,也为了推动学校发展提升,寻找到自身力量——内动力、内驱力。就是一种必然的选择。这种自身力量或内动力、内驱力就是校本教研。

cytochemistry was performed for SOCS2 protein. Cells were observed using a fl uorescence microscope (Olympus).

Figure 1 Effect of ginkgolide B (GKB) on the morphology of neural stem cells.

(A—F) Cell morphology of neural stem cells (arrows) at different time points (× 200). (A—C) Neural stem cells after induced differentiation for 3 (A), 7 (B) and 14 (C) days in the control group; (D—F) neural stem cells after induced differentiation for 3 (D), 7 (E) and 14 (F) days in the 40 mg/L GKB group. (G, H) The length of neural stem cell processes 3 days after differentiation. (G) Body area of neural stem cells 14 days after differentiation. (H) Data are presented as the mean ± SD and were tested with analysis of variance (n = 3 per group). *P < 0.05, vs.control group; #P < 0.05, vs. 20 mg/L GKB group.

Figure 2 Effect of ginkgolide B (GKB) on differentiation of neural stem cells.

Figure 3 Effect of ginkgolide B (GKB) on neurological function in rats with cerebral ischemia/reperfusion injury.

Data are presented as the mean ± SD (n = 3 per group; independent t-test). †P < 0.05, vs. day 3; §P < 0.05, vs. middle cerebral artery occlusion (MCAO) group.

Figure 4 Effect of ginkgolide B (GKB) on nestin immunoreactivity in the penumbra of rats with cerebral ischemia/reperfusion injury.

(A) Nestin expression (arrows) in the brain of rats at 3, 7 and 14 days after middle cerebral artery occlusion (MCAO) (immunohistochemistry; scale bars: 50 μm). (B) Quantitative analysis of nestin immunoreactivity. Data are presented as the mean ± SD (n = 6 per group; two-way analysis of variance with Bonferroni correction for pairwise comparison). ‡P < 0.05, vs. sham group; $P < 0.05, vs. day 7.

Figure 5 Effect of ginkgolide B (GKB) on neuron-speci fi c enolase(NSE) immunoreactivity in the penumbra of rats with cerebral ischemia/reperfusion injury.

(A) NSE expression (arrows) in the brain of rats at 3, 7 and 14 days after reperfusion (immunohistochemistry; scale bars: 50 μm). (B) Quantitative analysis of NSE immunoreactivity. Data are presented as the mean± SD (n = 6 per group; two-way analysis of variance with Bonferroni correction for pairwise comparison). ‡P < 0.05, vs. sham group; §P <0.05, vs. middle cerebral artery occlusion (MCAO) group; †P < 0.05, vs.day 3; $P < 0.05, vs. day 7.

Figure 6 Effect of ginkgolide B (GKB) on glial fi brillary acid protein(GFAP) immunoreactivity in the penumbra of rats with cerebral ischemia/reperfusion injury

(A) GFAP expression (arrows) in the brain of rats at 3, 7 and 14 days after reperfusion (immunohistochemistry; scale bars: 50 μm). (B) Quantitative analysis of GFAP immunoreactivity. Data are presented as the mean ± SD (n = 6 per group; two-way analysis of variance with Bonferroni correction for pairwise comparison). ‡P < 0.05, vs. sham group; §P< 0.05, vs. middle cerebral artery occlusion (MCAO) group; $P < 0.05,vs. day 7; *P < 0.05, vs. day 3 within the same group; #P < 0.05, vs. day 7 within the same group.

Figure 7 Effect of ginkgolide B (GKB) on bromodeoxyuridine (BrdU)immunoreactivity in rats with cerebral ischemia/reperfusion injury.

Figure 8 Effect of ginkgolide B (GKB) on brain-derived neurotrophic factor (BDNF) and epidermal growth factor (EGF)mRNA expression in rats with cerebral ischemia/reperfusion injury.

Relative expression of BDNF mRNA (A) and EGF mRNA (B) 3, 7 and 14 days after intervention. Data are presented as the mean ± SD (n = 3 per group; two-way analysis of variance with Bonferroni correction for pairwise comparison). ‡P < 0.05, vs. sham group; §P < 0.05, vs. middle cerebral artery occlusion (MCAO) group.

In vivo experiments

In the sham group, BrdU-positive cells were only visible in the subgranular and subventricular zones of the dentate gyrus (Figure 7A). At 3 days after reperfusion, the number of BrdU-positive cells was reduced, and these cells were sparsely distributed. At 7 days after reperfusion, the number of BrdU-positive cells increased, and some dark-stained cells were aggregated. No signi fi cant temporal trend was seen for the BrdU-positive cells. A signi fi cant increase in BrdU-positive cells was observed in the MCAO + GKB group compared with the sham and MCAO groups at all time points (all P < 0.05; Figure 7B).

1.3.1 对照组 32例患者,仅采用口服用药治疗法进行治疗,口服药物包括泼尼松片(辰欣药业股份),30毫克/次,晨顿服(5 d后减半量,逐减至停药);标准桃金娘油胶囊(德国GeloMyrtol) 0.3克/次,2次/天(15 d);酌情使用头孢羟氨苄片(清远华能制药),0.5克/次,2次/天(<7 d)。

This study was approved by the Ethics Committee of Zhejiang University of China (approval No. 2015-152), and all procedures were conducted according to the Experimental Animal Use and Welfare guidelines of Zhejiang University. A total of 150 male clean healthy Sprague-Dawley rats,weighing 250—280 g and aged 8 weeks, were purchased from the Zhejiang Academy of Medical Sciences of China (license No. SCXK2014-0001). The rats were randomly assigned to three groups: sham, middle cerebral artery occlusion(MCAO) and MCAO + GKB (n = 50 per group).

A cerebral ischemia/reperfusion model was established by MCAO (Tang et al., 2011). In brief, a 4-0 mono fi lament nylon thread was inserted into the right common carotid artery to obstruct the middle cerebral artery (MCA) for 90 minutes, and then the thread was withdrawn. In the sham group, the thread was inserted into the right common carotid artery without reaching the bifurcation between the MCA and anterior cerebral artery. The investigators who performed the procedures were blind to treatments.

After the cells were cultured for 3, 7 and 14 days, the coverslips were collected, and the medium was carefully removed.The coverslips were rinsed three times with 0.01 M PBS (pH 7.3) and fi xed in 4% paraformaldehyde in 0.1 M PBS (pH 7.3)at room temperature for 30 minutes. The residual paraformaldehyde was removed, and cells were washed three times in PBS. Cells were incubated with 10% goat serum in 0.01 M PBS (pH 7.3) at room temperature for 30 minutes. The medium was removed, and cells were incubated with primary antibodies (1:200; 100 μL), including mouse anti-neuron specific enolase (NSE) polyclonal (Abcam, Hong Kong,China), mouse anti-glial fibrillary acidic protein (GFAP)polyclonal (Santa Cruz Biotechnology, Dallas, TX, USA) and mouse anti-SOCS-2 polyclonal (Bioworld, Nanjing, China),at 4°C overnight. On day 2, after removal of the medium,cells were washed three times in PBS and incubated with biotinylated goat anti-mouse secondary antibody (1:200, 100 μL/well; Bioworld, Visalia, CA, USA) at room temperature for 2 hours. The cells were washed three times in PBS andincubated with avidin conjugated to Texas red (1:100, 100 μL/well; Thermo Fisher Scientific, Waltham, MA, USA) in the dark at room temperature for 2 hours. After washing in PBS, cells were mounted with glycerin buffer. Cells were then observed under a fl uorescence microscope (Olympus,Tokyo, Japan), and positive cells were counted. There were five wells (coverslips) in each group, and five fields were randomly selected from each coverslip. The total number of cells and positive cells were independently counted,followed by calculation of the proportion of positive cells.NSE-positive cells (neurons) and GFAP-positive cells (astrocytes) were counted. After cell culture for 7 days, immuno-

GKB at 20 mg/kg was intraperitoneally administered immediately and 6 hours after ischemia, and thereafter once daily.In the sham and MCAO groups, an equal volume of normal saline (2 mL) was intraperitoneally administered with the same schedule. In each group, rats received intraperitoneal injection of bromodeoxyuridine (BrdU) at 50 mg/kg three times (once every 4 hours) within 12 hours. Then, 4 hours after the last injection of BrdU, the rats were sacri fi ced for assessing the proliferation of NSCs.

At 3, 7 and 14 days after surgery, neurological function was evaluated (n = 5 per time point for each of the three groups). These rats were then sacrificed for immunohistochemistry, real-time quantitative polymerase chain reaction(RT-PCR) and western blot assay (n = 5 per group, for a total of 45 rats for each of the three assays).

Neurological function assessment

Neurological function was assessed using the Zea Longa method (Longa et al., 1989). The higher the neurological de fi cit score, the greater the impairment. Neurological function was evaluated 3, 7 and 14 days after surgery. To reduce variation caused by differences in impairment, rats with a score of 1—3 within 24 hours after surgery were used in the following experiments.

Immunohistochemistry

Immunohistochemistry was performed for nestin, NSE,GFAP and BrdU. After sacri fi ce, brains were collected, dehydrated in a graded ethanol series, and permeabilized in xylene. Brains from the ischemic penumbra were embedded in paraffin and cut into 4-μm sections, followed by drying at 37°C for 12—24 hours. The sections were stored at room temperature. Immunohistochemistry was performed using the SP immunohistochemical staining kit (Fuzhou Maixin Biotechnology Development Co., Ltd., Fuzhou, China) according to the manufacturer’s protocol to detect nestin (a neural stem cell marker; mouse polyclonal antibody, 1:200;Santa Cruz Biotechnology), NSE (a neuronal marker; mouse polyclonal antibody, 1:200; Abcam), GFAP (a marker for astrocytes; mouse polyclonal antibody, 1:200; Santa Cruz Biotechnology) and BrdU (mouse polyclonal antibody, 1:100;Santa Cruz Biotechnology). The slide was incubated with primary antibodies at 4°C overnight, followed by incubation with goat anti-mouse IgG (1:100; Bioworld) at 37°C for 20 minutes. The sections were observed under a light microscope (Shanghai Optical Instrument Factory, Shanghai, China). Six fi elds were randomly selected at the penumbra from each section at 400× magni fi cation, and at least six sections were observed in each group. Positive cells were counted.

RT-PCR

Immuno fl uorescence

Western blot assay

The brains were removed after sacrifice, and the penumbra was collected for protein extraction. Mouse polyclonal SOCS-2 (1:100; Bioworld), mouse polyclonal BDNF (1:200;Santa Cruz Biotechnology) and mouse polyclonal β-actin(1:200; Santa Cruz Biotechnology) were used as primary antibodies. Goat anti-rabbit IgG-horseradish peroxidase (1:200;Bioworld) and goat anti-mouse IgG-horseradish peroxidase(1:200; Bioworld) were used as secondary antibodies. The membrane was incubated with the primary antibody at 4°C overnight, followed by incubation with the secondary antibody at room temperature for 2 hours. The optical density of each band was measured with ImageJ software (NIH,Bethesda, MD, USA). β-Actin served as an internal reference. The level of the target protein was normalized to that of β-actin to obtain relative expression.

Statistical analysis

Figure 9 Effect of ginkgolide B (GKB) on brain-derived neurotrophic factor (BDNF) and suppressor of cytokine signaling 2(SOCS2) protein expression in rats with cerebral ischemia/reperfusion injury after middle cerebral artery occlusion (MCAO).

(A) Expression of BDNF, SOCS2 and β-actin in the brain of rats at 3, 7 and 14 days after reperfusion (western blot assay). (B, C) Quantitative analysis of BDNF protein (B) and SOCS2 protein (C) expression. Relative protein expression was calculated as the ratio of the optical density of the target protein to that of β-actin. Data are presented as the mean± SD (n = 3 per group; two-way analysis of variance with Bonferroni correction for pairwise comparison). ‡P < 0.05, vs. sham group.

Three days after injury, neurological de fi cits were observed in rats, and the neurological scores continued to decline at 7 and 14 days. The neurological de fi cit score for rats in the sham group was 0. The score in the MCAO group decreased over time, and the score was signi fi cantly lower in rats given treatment compared with rats in the MCAO group from days 3 through 14 (all P < 0.05; Figure 3).

Results

Effects of GKB on NSC proliferation and differentiation in vitro

Cell morphological changes

Twelve hours after differentiation was induced, most neurospheres in each group were adherent to the wall. Cells exhibited radial growth with the neurosphere at the center. The round cells became spindle-shaped and displayed some protrusions. After 3 days of differentiation, many cells migrated out of the neurosphere, and the processes were further lengthened. In the GKB groups, the growth of processes was rapid. A signi fi cant difference in the length of the processes was found 3 days after differentiation. Figure 1A–F show the cell morphologies at 3, 7 and 14 days after 40 mg/L GKB treatment. Compared with the 40 or 60 mg/L GKB group,process length was shorter in the control group and the 20 mg/L GKB group (both P < 0.05; Figure 1G). The mean cell body area of the NSCs was signi fi cantly smaller in the control and 20 mg/L GKB groups compared with the 40 and 60 mg/L GKB groups on day 14 (all P < 0.05; Figure 1H).

Differentiation of NSCs

那么,“物”的权力来自何处?换言之,它为什么会表现为一种异化的力量呢?无疑,“物”的权力不是来自于“物”自身,因为它只是一个客观的存在,那就只能来自于一种外在的力量。马克思在不同场合谈到了这种力量形成的机制。

After 7 days of differentiation, the cells were fi xed for immunofluorescence. Neuron-like cells (NSE-positive cells) had a large cell body, were round or oval, and displayed 1 or 2 long processes. Astrocytes (GFAP-positive cells) had some processes. The SOCS2-positive cells had a large cell body and became oval or spindle-shaped. The sizes of NSE-, GFAP-and SOCS2-positive cells were higher in the 40 mg/L group compared with the control group (all P < 0.05; Figure 2).

Effects of GKB on NSC proliferation and differentiation in the brain of rats with cerebral ischemia/reperfusion injury Neurological function

All data are presented as the mean ± SD and were analyzed using SPSS 22.0 software (IBM, Armonk, NY, USA). Differences in protrusion length and cell body area were evaluated by one-way analysis of variance (ANOVA). Independent t-test was used for testing the difference in percentage of positive NSCs between the control and 40 mg/L GKB groups.The effects of time and the three treatments were examined by two-way ANOVA. The two-way ANOVA model included interaction terms comprising time and group. If a signi fi cant interaction was identi fi ed, strati fi ed analyses by time or group were then performed. Multiple comparisons were carried out with the Bonferroni correction method if a signi fi cant difference was revealed by one-way ANOVA and two-way ANOVA. A two-sided signi fi cance level of 0.05 was set.

Nestin-positive cells

刑事立法政策是刑事政策在刑法立法上的体现,属于最狭义的刑事政策。然究竟何为刑事政策,目前学界对此的表述可谓林林总总尚无统一的定义,但通说认为刑事政策的概念有广义、狭义和最狭义之分。其中最狭义的刑事政策是指国家或社会公共团体以特别预防为目的,对不同的个别犯罪者或具有犯罪危险者,采取个别的有针对性的抑制活动和强制措施。[1]

[1]陈新谦,金有豫主编 .新编药物学[M].第13版.北京:人民卫生出版社,1992:181-184.

In the sham group, nestin-positive cells were observed in the ventricular ependyma and the subependymal zone. Nestin protein expression in the MCAO and MCAO + GKB groups was slightly increased on day 7, but then significantly reduced on day 14 (P = 0.013, vs. day 7; Figure 4A). Compared with the sham group, the proportion of nestin-positive cells was significantly higher in the MCAO + GKB group (P <0.05; Figure 4B).

NSE-positive cells

In the sham group, approximately 10% of the cells were positive for NSE at all three time points. In the model and MCAO + GKB groups, down-regulation of NSE protein was found at day 14 compared with day 3 or 7 (Figure 5A). In addition, although NSE expression was higher in the MCAO group than in the sham group at day 3, the proportions of NSE-positive cells in the two groups were comparable and signi fi cantly lower than those in the MCAO + GKB group after day 7 (all P ≤ 0.009; Figure 5B).

GFAP-positive cells

诚然,融合教学法对教师综合素质要求也相应的提高了,首先教师不断提高专业理论水平,经常查阅相关资料,掌握最前沿的科研理论动态,在知识的深度和广度上不断地充实自己[12]。其次,在临床病理和病理解剖方面也积累了丰富的实践经验。再者更要精心策划和设计教学中设置的问题,研究引导学生自主学习的方法及如何提高学生分析问题和解决问题综合能力和学习热情。

宏观经济学中的“杠杆率”定义仍然存在一定的争议[3]。有些学者认为宏观经济中的“杠杆率”应当是微观经济中所有个体“杠杆率”的加总,应当反映的是一个地区的资产负债率[5];由于在使用这一定义时,各个国家和地区的杠杆率无法进行比较,所以国际清算银行(BIS)和国际货币基金组织(IMF)往往采用“债务总额 /GDP”定义和计算一个地区及其各个部门的杠杆率。

In the sham group, GFAP-positive cells were not observed in the various brain regions examined, including the cortex(Figure 6A). Throughout the whole observation period,the number of GFAP-positive cells was not significantly changed, except for an apparent decrease in the MCAO +GKB group on day 14 compared with day 7 (P = 0.002). The number of GFAP-positive cells was significantly higher in the MCAO + GKB group than in the sham or MCAO group(all P ≤ 0.015; Figure 6B).

综上可见,敦煌文献数据库的建设随着计算机技术和数字化技术的发展而方兴未艾,但已取得了显著的成绩,在文献保护和研究方面做出了巨大贡献。但通过调研也发现,敦煌数据库的建设尚缺乏系统理论语言学原则指导下建立的、面向敦煌文献语言文字研究而创建的深加工研究型语料库。现有的敦煌文献电子化、数字化工作取得的显著成果,为建设这种深加工多模态语料库提供了有利条件。

Animal model

BDNF and EGF mRNA expression

RT-PCR results are shown in Figure 8. Seven days after injury, mRNA expression of BDNF and EGF was highest in the MCAO + GKB group, followed by the MCAO group and the sham group.

导致混合汽过浓的可能原因有:喷油嘴滴漏、水温传感器信号失真、空气流量计信号失真和空燃比传感器信号错误等。据此检查:喷油嘴无滴漏;水温传感器信号未见异常;空气流量计动态和静态数值均正常、且电热丝没有异物附着;空燃比传感器信号可以真实地反映尾气浓度变化,且主动测试结果也正常。至此常规的疑点已经确认完毕,均未发现异常。

BDNF and SOCS2 protein expression

The protein bands were scanned, and their optical densities were determined and compared. Western blot assay results are shown in Figure 9. BDNF protein expression was not changed over time, and no difference was detected among the sham, MCAO, MCAO + GKB groups (Figure 9B). A similar result was obtained for SOCS2 protein expression,except downregulation was found in the MCAO and MCAO+ GKB groups compared with the sham group at day 3 (all P< 0.05; Figure 9C).

Discussion

Our results indicate that GKB in an ischemia/reperfusion rat model increases the numbers of NSCs, neurons and astrocytes, and promotes the proliferation and self-renewal of NSCs. These effects of the herbal medicine might be related to the upregulation of EGF and SOCS2 expression, which serve to improve neurological function. These fi ndings provide insight into the mechanisms underlying the neuroprotective effectiveness of GKB after ischemia/reperfusion. It should be noted that GKB can be absorbed by the gastrointestinal system and easily traverses the blood-brain barrier.The mean half-life of GKB in the free form is approximately 140 minutes (Mauri et al., 2001). GKB appears to have therapeutic potential for promoting the proliferation and differentiation of NSCs (Nabavi et al., 2015).

The adult central nervous system microenvironment is less than optimal for NSC survival and differentiation (Cai et al., 2015). To improve the microenvironment, researchers have co-transplanted NSCs with astrocytes and brain microvascular endothelial cells and found that this improved memory in a rat model of ischemic stroke (Cai et al., 2015).In a study to investigate the protective effects of NSCs, the cells were transplanted in a cerebral ischemia rat model and the results suggest that the mechanism underlying neuroprotection might involve the regulation of early in fl ammatory events (Watanabe et al., 2016). Our study showed that GKB increases the proliferation of NSCs and improves neurological function in the MCAO rat model, consistent with the results of these previous studies.

The mechanism underlying the GKB-induced differentiation of NSCs is poorly understood. In our study, NSE (a marker of neurons) and GFAP (a marker of astrocytes) were both expressed. GKB treatment significantly increased NSE and GFAP expression in the brains of rats with MCAO. This suggests that NSCs in the brain are pluripotent. Further experiments are needed to determine if GKB treatment enhances the differentiation of NSCs in the brains of rats with MCAO.

A number of factors may promote the growth of neurites,including growth factors, neurotrophic factors and neurite growth factors (Liu et al., 2016). SOCS has been shown to be involved in the regulation of neurite growth. In our study,SOCS2 was mainly expressed in the cell bodies of NSCs. After GKB treatment, SOCS2 expression increased signi fi cantly, as demonstrated by immunofluorescence and western blot assay. This was accompanied by the improvement of neurological function. This suggests that GKB upregulates SOCS2 expression, thereby inhibiting the JAK/STAT signaling pathway(Yang et al., 2012; Letellier and Haan, 2016) to block growth hormone and cytokine signaling. The upregulation of SOCS2 by GKB also likely promotes the binding of SOCS2 to and the activation of the EGF receptor, resulting in the differentiation of NSCs into neurons and rapid neurite growth.

(3)在研究方法上,现有研究主要使用探索性的理论分析,实证分析和案例分析使用较少。需要指出的是,实证分析能对理论研究起到深化和补充作用,有些内容,例如内创业绩效,使用实证分析才能彻底理顺内创业绩效的影响因素及因素之间的相互关系;而对于某一具体对象的案例分析更具有客观性。

We also found that nestin expression and BrdU levels were at a low level in the periphery of the ischemic region in the sham group. At 3, 7 and 14 days after reperfusion, cells positive for nestin or BrdU were seen in the periphery of the ischemic region. Nestin and BrdU levels peaked at 7 and 14 days after reperfusion in the MCAO group. In the MCAO +GKB group, the number of cells positive for Nestin or BrdU increased markedly. GKB treatment signi fi cantly increased the number of cells positive for NSE or GFAP, suggesting that the migratory NSCs had differentiated into neurons and glial cells.

BrdU-positive cells

A number of studies on the neuroprotective effects of GKB in ischemia have provided evidence for several different mechanisms of action. Gu et al. (2012) found that GKB inhixited NF-κB in an ischemia/reperfusion mouse model, perhaps accounting for the anti-inflammatory and anti-apoptotic effects of the herbal medicine. Qin et al. (2014)reported that in mice with ischemia/reperfusion injury,GKB prevented cathepsin-mediated cell death. Zhou et al.(2014) carried out an in vitro study to investigate the effect of hydrolyzed ginkgolides on cytochrome P-450s (CYPs),and found a marked increase in CYP34A mRNA expression.Wang et al. (2014) found that the neuroprotective effects of GKG in focal ischemia in rats might involve inhibition of stress-activated protein kinase/c-Jun N-terminal kinase activation, which would block the mitochondrial apoptotic pathway. Finally, Wu et al. (2015) found that GKB provides neuroprotection in ischemic injury by inhibiting the expression of the stress-related protein RTP801. Therefore, future studies should investigate whether these mechanisms are involved in the neuroprotective effects of GKB in the rodent model of ischemic stroke. It should be noted that we only investigated the short-term (at 3, 7 and 14 days after reperfusion) neuroprotective effects of GKB and that the intermediate and long-term effects remain to be elucidated. Thus,future studies should also examine the longer-term effects of the herbal medicine.

In conclusion, GKB promoted the proliferation and differentiation of NSCs in the rat brain following cerebral ischemia, thereby increasing the proportion of neurons to improve neurological outcome. GKB might accomplish these effects, at least in part, by enhancing expression of BDNF,EGF and SOCS2.

Author contributions: PDZ and HJZ performed experiments. MH and RM participated in paper preparation and editing. SNJ was in charge of statistical analysis. JSZ was responsible for manuscript review. All authors approved the fi nal version of this paper.

Con fl icts of interest: The authors declare no competing fi nancial interests.

Financial support: This work was supported by the National Natural science Foundation of China, No. 81073082 to JSZ. The funding body played no role in the study design, in the collection, analysis and interpretation of data,in the writing of the paper, and in the decision to submit the paper for publication.

Institutional review board statement: This study was approved by the Ethics Committee of Zhejiang University of China (approval No. 2015-152). The experimental procedure followed the National Institutes of Health Guide for the Care and Use of Laboratory Animals (NIH Publications No. 8023, revised 1985).

水稻是我国重要的粮食作物,水稻产业发展影响着我国粮食产业发展。我国水稻育种要紧跟时代的发展步伐,培育出适应新时代的新品种。将杂交育种与现代技术进行有效结合,引进新的种质资源,培育出满足大众和生态环保需要的健康绿色、高产、优质水稻品种是大势所趋。

Copyright license agreement: The Copyright License Agreement has been signed by all authors before publication.

Data sharing statement: Datasets analyzed during the current study are available from the corresponding author on reasonable request.

Plagiarism check: Checked twice by iThenticate.

Peer review: Externally peer reviewed.

云梦站在一边,目瞪口呆。她做梦都想不到母亲竟会产生如此疯癫的想法做出如此疯癫的举动,事情发展到现在,似乎母亲终于对呼伦一直以来对她的不满进行了反击。然母亲的反击是温柔的,平和的,甚至对呼伦和云梦,充满了慈爱的关怀。母亲就像一位武功高深莫测的老尼,面对呼伦的花拳秀腿,不躲不避,不急不恼,身形稳健,笑容可掬,只需如此轻轻一击,呼伦和她,立刻一败涂地,毫无还手之力了。

Open access statement: This is an open access journal, and articles are distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as appropriate credit is given and the new creations are licensed under the identical terms.

References

Alagappan D, Lazzarino DA, Felling RJ, Balan M, Kotenko SV, Levison SW (2009) Brain injury expands the numbers of neural stem cells and progenitors in the SVZ by enhancing their responsiveness to EGF. ASN Neuro 1:e00009.

Ba YY, Wang H, Ning XJ, Luo L, Li WS (2014) Construction and identifi cationof human glial cell-derived neurotrophic factor gene-modi fi ed Schwann cells from rhesus monkeys. Hum Gene Ther Methods 25:339-344.

Barnat M, Benassy MN, Vincensini L, Soares S, Fassier C, Propst F, Andrieux A, von Boxberg Y, Nothias F (2016) The GSK3-MAP1B pathway controls neurite branching and microtubule dynamics. Mol Cell Neurosci 72:9-21.

Cai Q, Chen Z, Song P, Wu L, Wang L, Deng G, Liu B, Chen Q (2015)Co-transplantation of hippocampal neural stem cells and astrocytes and microvascular endothelial cells improve the memory in ischemic stroke rat. Int J Clin Exp Med 8:13109-13117.

Chen T, Yu Y, Tang LJ, Kong L, Zhang CH, Chu HY, Yin LW, Ma HY (2017)Neural stem cells over-expressing brain-derived neurotrophic factor promote neuronal survival and cytoskeletal protein expression in traumatic brain injury sites. Neural Regen Res 12:433-439.

Cui Y, Yi D, Bai X, Sun B, Zhao Y, Zhang Y (2012) Ginkgolide B produced endophytic fungus (Fusarium oxysporum) isolated from Ginkgo biloba.Fitoterapia 83:913-920.

Fang W, Deng Y, Li Y, Shang E, Fang F, Lv P, Bai L, Qi Y, Yan F, Mao L (2010)Blood brain barrier permeability and therapeutic time window of Ginkgolide B in ischemia-reperfusion injury. Eur J Pharm Sci 39:8-14.

Gu JH, Ge JB, Li M, Wu F, Zhang W, Qin ZH (2012) Inhibition of NF-kappaB activation is associated with anti-in fl ammatory and anti-apoptotic effects of Ginkgolide B in a mouse model of cerebral ischemia/reperfusion injury. Eur J Pharm Sci 47:652-660.

Hu YY, Huang M, Dong XQ, Xu QP, Yu WH, Zhang ZY (2011) Ginkgolide B reduces neuronal cell apoptosis in the hemorrhagic rat brain: possible involvement of Toll-like receptor 4/nuclear factor-kappa B pathway. J Ethnopharmacol 137:1462-1468.

Huang F, Wu Y, Wang H, Chang J, Ma G, Yin Z (2016) Effect of controlled release of brain-derived neurotrophic factor and neurotrophin-3 from collagen gel on neural stem cells. Neuroreport 27:116-123.

Letellier E, Haan S (2016) SOCS2: physiological and pathological functions.Front Biosci (Elite Ed) 8:189-204.

Liu D, Liu Z, Liu H, Li H, Pan X, Li Z (2016) Brain-derived neurotrophic factor promotes vesicular glutamate transporter 3 expression and neurite outgrowth of dorsal root ganglion neurons through the activation of the transcription factors Etv4 and Etv5. Brain Res Bull 121:215-226.

Longa EZ, Weinstein PR, Carlson S, Cummins R (1989) Reversible middle cerebral artery occlusion without craniectomy in rats. Stroke 20:84-91.

Maclennan KM, Darlington CL, Smith PF (2002) The CNS effects of Ginkgo biloba extracts and ginkgolide B. Prog Neurobiol 67:235-257.

Mauri P, Simonetti P, Gardana C, Minoggio M, Morazzoni P, Bombardelli E,Pietta P (2001) Liquid chromatography/atmospheric pressure chemical ionization mass spectrometry of terpene lactones in plasma of volunteers dosed with Ginkgo biloba L. extracts. Rapid Commun Mass Spectrom 15:929-934.

Mousavi SJ, Doweidar MH (2015) Three-dimensional numerical model of cell morphology during migration in multi-signaling substrates. PLoS One 10:e0122094.

Nabavi SM, Habtemariam S, Daglia M, Braidy N, Loizzo MR, Tundis R,Nabavi SF (2015) Neuroprotective effects of Ginkgolide B against ischemic stroke: a review of current literature. Curr Top Med Chem 15:2222-2232.

Ochi T, Nakatomi H, Ito A, Imai H, Okabe S, Saito N (2016) Temporal changes in the response of SVZ neural stem cells to intraventricular administration of growth factors. Brain Res 1636:118-129.

Qin XF, Lu XJ, Ge JB, Xu HZ, Qin HD, Xu F (2014) Ginkgolide B prevents cathepsin-mediated cell death following cerebral ischemia/reperfusion injury. Neuroreport 25:267-273.

Ryu S, Lee SH, Kim SU, Yoon BW (2016) Human neural stem cells promote proliferation of endogenous neural stem cells and enhance angiogenesis in ischemic rat brain. Neural Regen Res 11:298-304.

Tang Y, Huang B, Sun L, Peng X, Chen X, Zou X (2011) Ginkgolide B promotes proliferation and functional activities of bone marrow-derived endothelial progenitor cells: involvement of Akt/eNOS and MAPK/p38 signaling pathways. Eur Cell Mater 21:459-469.

Wang NQ, Wang LY, Zhao HP, Liu P, Wang RL, Song JX, Gao L, Ji XM,Luo YM (2015) Luoyutong treatment promotes functional recovery and neuronal plasticity after cerebral ischemia-reperfusion injury in rats.Evid Based Complement Alternat Med 2015:369021.

Wang X, Jiang CM, Wan HY, Wu JL, Quan WQ, Wu KY, Li D (2014) Neuroprotection against permanent focal cerebral ischemia by ginkgolides A and B is associated with obstruction of the mitochondrial apoptotic pathway via inhibition of c-Jun N-terminal kinase in rats. J Neurosci Res 92:232-242.

Watanabe T, Nagai A, Sheikh AM, Mitaki S, Wakabayashi K, Kim SU, Kobayashi S, Yamaguchi S (2016) A human neural stem cell line provides neuroprotection and improves neurological performance by early intervention of neuroin fl ammatory system. Brain Res 1631:194-203.

Wu X, Su J, Chen L, Ma B, Gu X, Zhu L (2015) Ginkgolide B protects neurons from ischemic injury by inhibiting the expression of RTP801. Cell Mol Neurobiol 35:943-952.

Xia SH, Fang DC (2007) Pharmacological action and mechanisms of ginkgolide B. Chin Med J (Engl) 120:922-928.

Xie Q, Wang F, Zhou GP, Zhang H, Ma JX (2016) Comparative analysis of the proliferation and differentiation of neural stem cells in the hippocampal dentate gyrus of rats with different ages. Zhongguo Zuzhi Gongcheng Yanjiu 20:5426-5431.

Yang HL, Sun C, Sun C, Qi RL (2012) Effect of suppressor of cytokine signaling 2 (SOCS2) on fat metabolism induced by growth hormone (GH)in porcine primary adipocyte. Mol Biol Rep 39:9113-9122.

Yu JH, Seo JH, Lee JY, Lee MY, Cho SR (2016) Induction of neurorestoration from endogenous stem cells. Cell Transplant 25:863-882.

Zhou XW, Ma Z, Geng T, Wang ZZ, Ding G, Yu-an B, Xiao W (2014) Evaluation of in vitro inhibition and induction of cytochrome P450 activities by hydrolyzed ginkgolides. J Ethnopharmacol 158 Pt A:132-139.

Zhu LL, Wu LY, Yew DT, Fan M (2005) Effects of hypoxia on the proliferation and differentiation of NSCs. Mol Neurobiol 31:231-242

Pei-DongZheng,RajneeshMungur,Heng-JunZhou,MuhammadHassan,Sheng-NanJiang,Jie-ShengZheng
《Neural Regeneration Research》2018年第7期文献

服务严谨可靠 7×14小时在线支持 支持宝特邀商家 不满意退款

本站非杂志社官网,上千家国家级期刊、省级期刊、北大核心、南大核心、专业的职称论文发表网站。
职称论文发表、杂志论文发表、期刊征稿、期刊投稿,论文发表指导正规机构。是您首选最可靠,最快速的期刊论文发表网站。
免责声明:本网站部分资源、信息来源于网络,完全免费共享,仅供学习和研究使用,版权和著作权归原作者所有
如有不愿意被转载的情况,请通知我们删除已转载的信息 粤ICP备2023046998号