用户名: 密码: 验证码:
胶质细胞谷氨酸转运体亚型GLT-1调制对大鼠脑缺血预处理脑保护作用的影响
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
脑缺血疾病是严重危害人类健康的疾病之一。神经细胞对缺血性危害极为敏感,如果缺血时间较长,神经细胞会大量死亡,即使恢复血液供应,神经细胞也难以再生,甚至会留下严重的后遗症。随着对脑缺血疾病研究的深入,脑缺血耐受(brain ischemic tolerance, BIT)现象越来越普遍受到关注。实验发现,给动物突然造成较严重的脑缺血后,海马CA1区的神经元会大量死亡。若在此之前,预先给动物造成轻微、短时、不至于引起神经元死亡的脑缺血,间隔一定时间后,再给动物造成较严重的、通常会引起大量神经元死亡的脑缺血,此时海马CA1区的神经元则基本不死亡,即神经元对缺血性损害产生了抵抗力,这一现象被称为脑缺血耐受。预先给予的轻微、短时脑缺血称为脑缺血预处理(cerebral ischemic preconditioning,CIP)。阐明脑缺血预处理脑保护作用的发生机制,对临床上研究、开发提高神经元对缺血缺氧耐受性的治疗方法具有重要意义。
     各种疾病引起的脑组织缺血、缺氧均可引起神经元能量代谢障碍,抑制Na+/K+依赖式ATP酶的活动,使细胞外K+浓度明显增高,Na+浓度相应降低,导致神经元去极化,引起兴奋性神经末梢释放谷氨酸;此外,细胞外高K+能够逆转高亲合力谷氨酸转运体的活动,把谷氨酸从细胞内转运至细胞外。这些原因引起细胞外谷氨酸等兴奋性氨基酸增多。这些增多的谷氨酸与神经细胞膜相应的受体结合,引起Na+、Ca2+内流以及细胞内Ca2+释放,导致Na+、Ca2+超载,进而使神经细胞死亡。因此,将这些氨基酸称为兴奋性神经毒素。降低脑缺血时细胞外液中谷氨酸的浓度,减少其与突触后膜特异性受体的结合,是防治其兴奋性毒性作用、减轻缺血时神经元损伤的重要手段。兴奋性氨基酸转运体(excitatory amino acid transports, EAATs)是调控脑内细胞外液谷氨酸浓度的重要机制。其中星形胶质细胞谷氨酸转运体亚型GLT-1在终止谷氨酸能神经传递,维持细胞外液谷氨酸浓度处于正常水平方面发挥重要作用。一些研究对星形胶质细胞GLT-1在缺血缺氧耐受中的作用给予了关注。例如,Douen等研究表明,皮层扩散性抑制预处理可以下调星形胶质细胞谷氨酸转运体(EAAT1和EAAT2)的表达,防止缺血引起的谷氨酸逆转运;Romera等应用离体(in vitro)模型发现,缺血或缺氧预处理可以使谷氨酸转运体表达增加。这些结果提示GLT-1可能与脑缺血耐受有关。但星形胶质细胞GLT-1在整体情况下(in vivo)是否参与CIP的脑保护作用?调制GLT-1是否可增强脑细胞对缺血性损害的耐受性?等尚未见报道。因此,本实验应用大鼠全脑缺血模型、GLT-1抑制剂Dihydrokainate (DHK)、GLT-1反义及正义寡核苷酸,在整体水平探讨调制GLT-1对脑缺血耐受诱导的影响,为阐明GLT-1在脑缺血耐受形成中的作用提供实验依据。
     1 DHK抑制GLT-1的功能阻断CIP诱导的脑缺血耐受
     应用大鼠四血管闭塞(4-vessel occlusion,4VO)全脑缺血模型,观察GLT-1特异性抑制剂DHK对CIP脑保护作用的影响,探讨GLT-1在脑缺血耐受中的作用。
     将96只凝闭双侧椎动脉2d后的Wistar大鼠随机分为8组:①sham组(n=6):只暴露双侧颈总动脉,不阻断血流;②CIP组(n=6):夹闭双侧颈总动脉3 min;③脑缺血打击组(n=6):夹闭双侧颈总动脉8 min;④CIP+脑缺血打击组(n=6):夹闭双侧颈总动脉3 min作为CIP,再灌注2 d后再夹闭双侧颈总动脉8 min;⑤双蒸水组(n=6):右侧脑室注射双蒸水20μl;⑥DHK组(n=30):右侧脑室注射DHK溶液20μl,根据DHK的剂量进一步分为10、100、200、500和1000 nmol 5个亚组,每组6只动物;⑦DHK+CIP组(n=18):右侧脑室注射DHK溶液20μl,20 min后夹闭双侧颈总动脉3 min,根据DHK的剂量进一步分为10、100和200 nmol 3个亚组,每组6只动物;⑧DHK+CIP+脑缺血打击组(n=18):右侧脑室注射DHK溶液20μl ,20 min后夹闭双侧颈总动脉3 min,2 d后夹闭双侧颈总动脉8 min。根据DHK剂量不同分为10、100、200 nmol 3个亚组,每组6只动物。以上各组动物于sham手术、侧脑室注射或末次脑缺血后7 d断头取脑,常规脑组织切片(5μm厚),硫堇染色,光学显微镜下观察海马CA1区组织学形态,确定锥体神经元迟发性死亡(delayed neuronal death, DND)情况。计数海马CA1区每1 mm区段内细胞膜完整、胞核饱满、核仁清晰的锥体细胞数目,每张切片双侧海马各计数3个区段取平均数为神经元密度(Neuronal density, ND)。根据以下标准确定组织学分级(Histological grade, HG):0级,无神经元死亡;Ⅰ级,散在神经元死亡;Ⅱ级,成片神经元死亡;Ⅲ级,几乎全部的神经元死亡。
     结果发现,4VO过程中,大鼠翻正反射消失,瞳孔散大,脑电波频率变慢,波幅逐渐变小甚至成等电位线,说明产生了全脑缺血。
     硫堇结果显示,Sham组和CIP组大鼠海马CA1区无明显的DND,HG为0~Ⅰ级,ND值分别为208.25±5.97和202.93±4.32。脑缺血打击组大鼠海马CA1区出现明显的DND,与sham组和CIP组相比,HG(Ⅱ~Ⅲ级)明显升高(P<0.01),ND值(45.86±21.93)显著降低(P<0.01)。CIP+脑缺血打击组海马CA1区DND不明显,与脑缺血打击组相比,HG(0~Ⅰ级)明显降低(P<0.01),ND值(208.07±5.87)显著升高(P<0.01),表明CIP可以诱导海马CA1区神经元产生缺血性耐受,对抗缺血打击引起的DND。右侧脑室注射双蒸水后海马CA1区组织形态与Sham组和CIP组基本一致。单纯侧脑室给予DHK组中,10、100、200 nmol剂量组海马CA1区神经元少量死亡,三组的HG均为Ⅰ级,ND值分别为172.76±17.31、162.64±6.12和155.43±9.82;而500、1000 nmol剂量组的HG分别为为Ⅱ级和Ⅲ级,ND值分别为105.35±3.84和6±1.39;与10、100、200 nmol组相比,HG和ND的变化具有显著性(P<0.05),表明大剂量的DHK可引起显著的海马CA1区锥体神经元死亡。因此,以下各组中,仅观察DHK 10、100和200 nmol的影响。DHK+CIP组中海马CA1区组织形态与Sham组和CIP组基本一致(10、100和200 nmol组HG均为为0~Ⅰ级,ND值分别为192.15±6.25、188.65±3.90和191.35±3.88)。DHK+CIP+脑缺血打击组中,大鼠海马CA1区出现了显著的DND,随着DHK剂量的增加,其HG级逐渐升高(10 nmol组为Ⅰ级,100 nmol组为Ⅰ~Ⅱ级,200 nmol组为Ⅲ级),ND逐渐下降(10 nmol组为160.87±13.55,100 nmol组为117.07±10.11,200 nmol组为4.87±2.02);与CIP+脑缺血打击组相比,上述HG和ND的变化具有显著性(P<0.01);此外,与单独DHK组相比,DHK+CIP+脑缺血打击组的DHK的量效曲线明显左移,并且斜率变陡。这些结果表明,DHK+CIP+脑缺血打击组中,除去DHK本身引起的少量神经元死亡以外,DHK还阻断CIP抗脑缺血打击的作用,从而引起更多的锥体细胞死亡。
     以上结果表明, GLT-1特异性抑制剂DHK可剂量依赖性地阻断CIP抗脑缺血打击的作用,提示GLT-1参与CIP诱导的脑缺血耐受。
     2 GLT-1反义寡核苷酸抑制GLT-1表达减弱CIP抗脑缺血打击的作用
     应用GLT-1反义寡核苷酸(antisense oligodeoxynucleotides,AS-ODNs)抑制GLT-1蛋白的表达,观察其对CIP抗脑缺血打击作用的影响,进一步探讨GLT-1在脑缺血耐受中的作用。
     2.1 GLT-1 AS-ODNs对GLT-1蛋白表达的影响
     将42只凝闭双侧椎动脉36 h的Wistar大鼠随机分为3组:①对照组(n=6):右侧脑室注射双蒸水5μl,注射后12 h取材;②AS-ODNs 9 nmol组(n=18):右侧脑室注射AS-ODNs溶液5μl(9 nmol),根据AS-ODNs注射后取材的时间又分为三个亚组:12 h、24 h、36 h组;③AS-ODNs 18 nmol组(n=18):右侧脑室注射AS-ODNs溶液5μl(18 nmol),根据AS-ODNs注射后取材的时间又分为三个亚组:12 h、24 h、36 h组。所有大鼠在预定时间断头取材,低温条件下分离海马CA1区,采用Western blotting方法测定GLT-1的蛋白表达,应用凝胶图像分析系统对Western免疫反应阳性条带进行积分光密度(integrated optical density,IOD)测定,以GLT-1的IOD值与β-actin的IOD值的比值代表GLT-1表达的相对水平。
     结果显示,双蒸水对照组的IOD的比值为0.65±0.22。注射AS-ODNs 9 nmol组中,12 h组的IOD比值为0.37±0.07,24 h组为0.20±0.05, 36 h组为0.25±0.07。注射AS-ODNs 18 nmol组中,12 h组的IOD比值为0.11±0.05,24 h组为0.05±0.02,36 h组为0.17±0.16。与双蒸水对照组相比,注射AS-ODNs后IOD的比值显著降低(P<0.05),且呈现一定的剂量依赖性。
     以上结果表明,侧脑室注射AS-ODNs溶液可以剂量依赖性地抑制GLT-1蛋白表达,这种抑制作用在注射AS-ODNs后24 h最为显著。
     2.2 GLT-1 AS-ODNs抑制CIP对海马CA1区锥体神经元的保护作用
     将54只凝闭双侧椎动脉的Wistar大鼠随机分为7组:①sham组(n=6):只暴露双侧颈总动脉,不阻断血流;②CIP组(n=6):夹闭双侧颈总动脉3 min;③脑缺血打击组(n=6):夹闭双侧颈总动脉8 min;④CIP+脑缺血打击组(n=6):夹闭双侧颈总动脉3 min作为CIP,再灌注2 d后再夹闭双侧颈总动脉8 min。⑤双蒸水组(n=6):于分离暴露双侧颈总动脉(但不夹闭)前12 h、后12 h及后36 h右侧脑室注射双蒸水,每次5μl,其他同sham组;⑥AS-ODNs组(n=12):于分离暴露双侧颈总动脉(但不夹闭)前12 h、后12 h及后36 h右侧脑室注射AS-ODNs溶液,每次5μl,其他同sham组。根据AS-ODNs的剂量进一步分为9和18 nmol 2个亚组,每组6只动物;⑦AS-ODNs+CIP+脑缺血打击组(n=12):于CIP前12 h、后12 h及后36 h右侧脑室注射AS-ODNs溶液,每次5μl,其它同CIP+脑缺血打击组。根据AS-ODNs的剂量进一步分为9 nmol和18 nmol 2个亚组,每组6只动物。以上各组动物均在sham手术、侧脑室注射或末次脑缺血后7d断头取材,常规脑组织切片(5μm厚),硫堇染色下观察海马CA1区DND的发生情况(方法同前)。
     结果显示,Sham组和CIP组大鼠海马CA1区无明显的DND,HG为0~Ⅰ级,ND值分别为208.25±5.97和202.86±4.28。脑缺血打击组大鼠海马CA1区出现明显的DND,HG(Ⅱ~Ⅲ级)明显升高(P<0.01),ND值(45.86±21.93)显著降低(P<0.01)。CIP+脑缺血打击组海马CA1区DND不明显,与脑缺血组相比,HG(0~Ⅰ级)明显降低(P<0.01),ND值(208.98±5.9)显著升高(P<0.01),表明CIP可以诱导海马CA1区神经元产生缺血性耐受,对抗缺血打击引起的DND。侧脑室注射双蒸水和AS-ODNs (9 nmol and 18 nmol)后海马CA1区组织形态与Sham组和CIP组基本一致,HG为0~Ⅰ级,ND值略有减少(分别为176.22±6.29、176.89±2.88和175.32±2.77(P<0.01)),表明侧脑室注射以及单纯AS-ODNs不会对海马CA1区神经元产生明显损伤。AS-ODNs+CIP+脑缺血打击组中,与CIP+脑缺血打击组相比,其HG显著升高(Ⅱ~Ⅲ级)(P<0.01),ND值显著下降(分别为133.56±3.42、70.94±7.38)(P<0.01),表明AS-ODNs抑制了GLT-1表达从而减弱了CIP对海马CA1区锥体神经元的保护作用。
     以上结果显示,单纯侧脑室注射AS-ODNs可引起海马CA1区GLT-1表达下调,但不引起明显的DND;而预先侧脑室注射AS-ODNs可减弱CIP对海马CA1区锥体神经元的保护作用。这些结果表明AS-ODNs通过抑制GLT-1表达从而减弱了CIP对海马CA1区锥体神经元的保护作用,进一步证实了GLT-1在CIP对脑缺血耐受诱导中的作用。
     3 GLT-1正义寡核苷酸上调GLT-1的表达增强脑对缺血打击的耐受
     应用GLT-1正义寡核苷酸(sense oligodeoxynucleotides,S-ODNs)上调GLT-1的表达后,观察海马CA1区锥体神经元对脑缺血打击的耐受情况,进一步确定GLT-1在脑缺血耐受中的作用。
     3.1 GLT-1 S-ODNs上调GLT-1的表达
     将42只凝闭双侧椎动脉36 h的Wistar大鼠随机分为3组:①对照组(n=6):右侧脑室注射双蒸水5μl,注射后12 h取材;②S-ODNs 9 nmol组(n=18):右侧脑室注射S-ODNs溶液5μl(9 nmol),根据S-ODNs注射后取材的时间又分为三个亚组:12 h、24 h、36 h组;③S-ODNs 18 nmol组(n=18):右侧脑室注射S-ODNs溶液5μl(18 nmol),根据S-ODNs注射后取材的时间又分为三个亚组:12 h、24 h、36 h组。所有大鼠在预定时间断头取材,低温条件下分离海马CA1区,采用Western blotting方法测定GLT-1的蛋白表达(方法同前)。
     结果显示,双蒸水对照组的IOD的比值为0.64±0.19。注射S-ODNs 9 nmol组中,12 h组的IOD比值为0.78±0.02,24 h组为1.01±0.04, 36 h组为0.68±0.06。注射S-ODNs 18 nmol组中,12 h组的IOD比值为0.84±0.02,24 h组为1.23±0.03,36 h组为0.62±0.02。与双蒸水对照组相比,注射S-ODNs后IOD的比值显著增加(P<0.01),且呈现剂量依赖性。以上结果表明,侧脑室注射S-ODNs溶液可以剂量依赖性地上调GLT-1蛋白表达,这种上调作用在注射S-ODNs后24 h最为显著。
     3.2 GLT-1 S-ODNs增强海马CA1区锥体神经元对缺血打击的耐受性
     将凝闭双侧椎动脉的Wistar大鼠54只随机分为7组:①sham组(n=6):只暴露双侧颈总动脉,不阻断血流;②CIP组(n=6):夹闭双侧颈总动脉3 min;③脑缺血打击组(n=6):夹闭双侧颈总动脉8 min;④CIP+脑缺血打击组(n=6):夹闭双侧颈总动脉3 min作为CIP,再灌注2 d后再夹闭双侧颈总动脉8 min;⑤双蒸水组(n=6):于分离暴露双侧颈总动脉(但不夹闭)前12 h、后12 h及后36 h右侧脑室注射双蒸水,每次5μl,其他同sham组;⑥S-ODNs组(n=12):于分离暴露双侧颈总动脉(但不夹闭)前12 h、后12 h及后36 h右侧脑室注射S-ODNs溶液,每次5μl,其他同sham组。根据S-ODNs的剂量进一步分为9和18 nmol 2个亚组,每组6只动物;⑦S-ODNs+脑缺血打击组(n=12):于分离暴露双侧颈总动脉(但不夹闭)前12 h、后12 h及后36 h右侧脑室注射S-ODNs溶液,每次5μl,并于暴露双侧颈总动脉2 d后夹闭双侧颈总动脉8 min。根据S-ODNs的剂量进一步分为9和18 nmol 2个亚组,每组6只动物。以上各组动物均在sham手术或末次手术后7d断头取材,常规脑组织切片(5μm厚),硫堇染色下观察海马CA1区DND的发生情况(方法同前)。
     结果显示,Sham组和CIP组大鼠海马CA1区无明显的DND,HG为0~Ⅰ级,ND值分别为208.25±5.97和202.86±4.28。脑缺血打击组大鼠海马CA1区出现明显的DND,HG(Ⅱ~Ⅲ级)明显升高(P<0.01),ND值(45.86±21.93)显著降低(P<0.01)。CIP+脑缺血打击组海马CA1区DND不明显,与脑缺血组相比,HG(0~Ⅰ级)明显降低(P<0.01),ND值(208.98±5.9)显著升高(P<0.01),表明CIP可以诱导海马CA1区神经元产生缺血性耐受,对抗缺血打击引起的DND。侧脑室注射双蒸水和S-ODNs后海马CA1区组织形态与Sham组和CIP组基本一致,HG为0~Ⅰ级,ND值为176.22±6.29(双蒸水)、186.53±5.64(9 nmol S-ODNs)和204.4±12.99(18 nmol S-ODNs),与sham组或CIP组相比无显著性差异(P>0.05)。S-ODNs +脑缺血打击组的HG为Ⅰ级,与脑缺血打击组相比显著降低(P<0.01);ND值分别为148.53±9.1(9 nmol S-ODNs组)和166.67±8.31(18 nmol S-ODNs组),与脑缺血打击组相比显著增加(P<0.01);表明S-ODNs通过上调GLT-1蛋白表达从而减弱了脑缺血对海马CA1区锥体神经元的损伤。
     以上结果显示,侧脑室注射S-ODNs引起海马CA1区GLT-1表达明显上调,预先注射S-ODNs减弱了脑缺血打击对海马CA1区锥体神经元的损伤。这些结果证实S-ODNs通过增加GLT-1的蛋白表达从而减弱了脑缺血对海马CA1区锥体神经元的损伤,产生了与CIP相同的效果。
     4结语
     (1)谷氨酸转运体GLT-1抑制剂DHK通过抑制GLT-1的功能而减弱了CIP对海马CA1区锥体神经元的保护作用;
     (2)侧脑室注射谷氨酸转运体GLT-1 AS-ODNs可抑制GLT-1的表达,同时减弱了CIP对海马CA1区锥体神经元的保护作用;
     (3)侧脑室注射GLT-1 S-ODNs上调GLT-1表达的同时,增强了海马CA1区锥体神经元对脑缺血打击的耐受性,产生了与CIP相同的效果。
     (4)以上结果表明GLT-1参与了CIP诱导的BIT。
Ischemic cerebrovascular disease is one kind of diseases which severely impair human health. Since neuronal cells are very sensitive to ischemia, long time brain ischemic insult results in neuronal death, which usually can not revive and frequently causes severe sequelae even if blood supply recovers. With the progress of the studies on the ischemic cerebrovascular disease, more and more attention has been paid to the phenomenon of brain ischemic tolerance (BIT). It is found in animal experiment that suddenly severe cerebral ischemic insult causes severe delayed neuronal death (DND) in the CA1 hippocampus. But when the ischemic insult was preceded by a transient sublethal cerebral ischemic preconditioning, the DND normally induced by the ischemic insult could be significantly prevented. This phenomenon was called brain ischemic tolerance (BIT), and the sublethal cerebral ischemia given in advance was called cerebral ischemic preconditioning (CIP). It is important to clarify the mechanism of the neuro-protection induced by CIP for developing new therapeutic methods to enhance the tolerance of neurons to ischemia and hypoxia.
     Ischemia and hypoxia in brain tissue induced by various kinds of diseases can result in dysmetabolism in energy and inhibit the action of Na+-K+-ATPase, which would lead to an obvious increase of extracellular K+ concentration and intracellular Na+ concentration. The changes facilitate depolarization of neurons and release of glutamate in the nerve terminal. In addition, the high concentration of extracellular K+ and intracellular Na+ can reverse the activity of high-affinity glutamate transporter, which would lead to reversal transporting of glutamate from intracellular to extracellular space. These reasons result in an increase in extracellular glutamate concentration. The increased extracellular glutamate binds with and activates NMDA or non-NMDA receptors, and then induces influx of Na+ and Ca2+ and release of intracellular Ca2+ to cytoplasm. The neurons die of the overload of Na+ and Ca2+. Thus, the glutamate has been referred to as excitotoxin. Therefore, it is an impotant strategy to decrease the concentration of extracellular glutamate or to reduce the combination of glutamate with its specific receptor for preventing the excitotoxicity of the increased glutamate in brain ischemia. Excitatory amino acid transports (EAATs) are important mechanism for regulating the extracellular concentration of glutamate. It has been well-known that the glial glutamate transporter subtype GLT-1 plays a dominant role in terminating glutamate neurotransmission, and maintaining the extracellular glutamate below neurotoxic levels. Some studies paid attention to roles of GLT-1 in induction of tolerance of brain to ischemic or hypoxic insults. For example, Douen et al reported that preconditioning with cortical spreading depression could down-regulate the expression of glutamate transporters EAAT1 and EAAT2 of astrocytes to prevent the reversal transporting of glutamate during cerebral ischemia. Romera et al found that preconditioning with ischemia or hypoxia in vitro could increase the expression of glutamate transports. These results suggested that GLT-1 might associate with the induction of BIT. But there is no report yet about whether GLT-1 of astrocytes participates in the neuro-protection of CIP in vivo and whether the modulation of GLT-1 may enhance the tolerance of neuronal cells to ischemic insult. Therefore, the present study was undertaken to explore in vivo the effect of modulation of GLT-1 by Dihydrokainate (DHK), a selective inhibitor of GLT-1, GLT-1 antisense oligodeoxynucleotides (AS-ODNs) or sense oligodeoxynucleotides (S-ODNs) on the induction of BIT using rat global cerebral ischemic model. The results to be obtained would provide experimental evidence for clarifying the role of GLT-1 in the induction of BIT.
     1 Inhibiting function of GLT-1 with DHK blocked BIT induced by CIP
     The role of GLT-1 in BIT was investigated in rat 4-vessel occlusion (4VO) global brain ischemia model by observing the effect of Dihydrokainate (DHK), a selective inhibitor of GLT-1, on the neuro-protection of CIP against delayed pyramidal neuronal death in the CA1 hippocampus normally induced by severe brain ischemic insult.
     Ninety-six Wistar rats with permanently occluded bilateral vertebral arteries for 2d were randomly assigned to eight groups:①Sham group (n=6): the bilateral common carotid arteries (BCCA) were separated, but without occluding the blood flow;②CIP group (n=6): the BCCA were clamped for 3 min;③Brain ischemic insult group (n=6): the BCCA were clamped for 8 min;④CIP+Brain ischemic insult group (n=6): a CIP for 3 min was preformed first, and then a brain ischemic insult for 8 min was given 2d after the CIP;⑤Distilled water group (n=6): 20μl distilled water was injected into the right lateral ventricle;⑥DHK group(n=30): 20μl DHK solution was injected into the right lateral ventricle. This group was further divided into 10 nmol, 100 nmol, 200 nmol, 500 nmol and 1000 nmol subgroups according to the doses of DHK;⑦DHK+CIP group(n=18): 20μl DHK solution was injected into the right lateral ventricle 20 min before CIP. This group was further divided into 10 nmol, 100 nmol and 200 nmol subgroups according to the doses of DHK;⑧DHK+CIP+brain ischemic insult group (n=18): 20μl DHK solution was injected into the right lateral ventricle 20 min before CIP. Other procedures were the same as those in CIP+brain ischemic insult group. According to the doses of DHK used, this group was also further divided into 10 nmol, 100 nmol and 200 nmol subgroups. The animals were sacrificed by decapitation at 7d after the sham operations or the last operations. The brain was sectioned in a thickness of 5μm and stained with thionin. Histological changes of the CA1 hippocampus were examined under light microscope to determine delayed neuronal death (DND) by neuronal density (ND) and histological grade (HG) of the CA1 hippocampus. The ND was determined by counting the number of surviving pyramidal neurons with intact cell membrane, full nucleus and clear nucleolus within 1 mm linear length of the CA1 hippocampus. The average of the number in 3 areas of the CA1 hippocampus was calculated as value of ND. HG was divided into the following 4 grades: grade 0, no neuron death; grade Ⅰ, scattered single neuron death; gradeⅡ, death of many neurons; gradeⅢ, death of almost complete neurons. The average HG of the bilateral hippocampus was counted as statistical data.
     The results showed that during the 4VO, the pupils of rats enlarged. EEG showed decreases in frequency and amplitude, even approached isoelectric level, and the righting reflex disappeared. These manifestations indicated that the rats were really subjected to global cerebral ischemia.
     It was found by thionine staining that there was no significant neuronal damage in the CA1 subfield of the hippocampus in the sham and CIP groups. HG was 0~Ⅰ, and values of ND were 208.25±5.97 and 202.93±4.32 in the two groups, respectively. Obvious destruction of the CA1 subfield was found in brain ischemic insult for 8 min group. The value of ND was 45.86±21.93, which was much low compared with that in the sham and CIP groups (P<0.01). HG was gradeⅡ~Ⅲ, which was much high compared with that in the sham and CIP. In CIP+ischemic insult group, no obvious neuronal damage in the hippocampal CA1 region was found. HG (grade 0~Ⅰ) was significantly lower than that in brain ischemic insult group (P<0.01), and the value of ND (208.07±5.87) was much higher than that in brain ischemic insult group (P<0.01). The results indicated that CIP protected pyramidal neurons of the CA1 hippocampus against DND induced by brain ischemic insult. There was no apparent DND in distilled water group, in which histological characteristics such as HG and ND were similar with those in the sham group. DHK in doses of 10, 100 and 200 nmol just caused slight DND of pyramidal neurons in the CA1 hippocampus. HG was gradeⅠin each dose, and the values of ND were 172.76±17.31, 162.64±6.12 and 155.43±9.82, respectively. While obvious DND was found in 500 and 1000 nmol groups; the value of ND was significantly decreased (105.35±3.84 and 6±1.39, respectively); HG was significantly increased (ⅡandⅢ, respectively) compared with 10, 100 and 200 nmol DHK groups (P<0.05). These results indicated that large dose DHK could lead to significant pyramidal neuron damage in the hippocampal CA1 region. Thus, we only observed effects of 10, 100 and 200 nmol DHK in the subsequent experiment. There was no apparent DND was found in DHK+CIP group in each dose of 10, 100 and 200 nmol DHK, in which histological characteristics such as HG and ND were similar with those in the sham group. Obvious DND was found in DHK+CIP+brain ischemic insult group. HG wereⅠ,Ⅰ~ⅡandⅢ, and the values of ND were 160.87±13.55, 117.07±10.11 and 4.87±2.02, respectively, in the doses of 10 nmol, 100 nmol and 200 nmol. The changes above in DHK+CIP+brain ischemic insult group were significant compared with CIP+brain ischemic insult group (P<0.01). Additionally, the correlative curve between the doses and effectiveness in DHK+CIP+brain ischemic insult groups shifted to left siginifantly, and the slope rate became sharper than that in DHK group. These results indicated that administration of DHK blocked in a dose dependent manner the neuroprotection of CIP against DND normally induced by severe brain ischemic insult in the CA1 hippoacmpus.
     The above results suggested the involvement of GLT-1 in the neuroprotection of CIP against DND normally induced by severe brain ischemic insult in the CA1 hippoacmpus.
     2 Inhibition of the expression of GLT-1 with GLT-1 AS-ODNs attenuated the neuro-protection of CIP against brain ischemic insult
     The role of GLT-1 in BIT was investigated by observing the effect of antisense oligodeoxynucleotides (AS-ODNs) of GLT-1, which can inhibit the protein expression of GLT-1, on the neuro-protection of CIP against brain ischemic insult.
     2.1 The effect of AS-ODNs on the protein expression of GLT-1
     Forty-two Wistar rats with permanently occluded bilateral vertebral arteries for 36 h were randomly assigned to three groups.①control group (n=6): 5μl distilled water was injected into the right lateral ventricle and the animals were sacrificed by decapitation 12 h later.②AS-ODNs 9 nmol group(n=18):5μl AS-ODNs (9 nmol) solution was injected into the right lateral ventricle. This group was further divided into 12h, 24h and 36h subgroups according to the time of reperfusion after injection.③AS-ODNs 18 nmol group(n=18):5μl AS-ODNs (18 nmol) solution was injected into the right lateral ventricle. This group was also further divided into 12h, 24h and 36h subgroups according to the time of reperfusion after injection. The animals were sacrificed by decapitation at the determined endpoint of the experiment. The hippocampal CA1 was dissected out quickly on ice and determine the expression of GLT-1 using Western blotting. Integrated optical density (IOD) of each band was measured using image analysis system. The changes of GLT-1 expression were represented with the ratio of IOD of the aim protein band toβ-actin.
     The results of Western blotting showed that IOD in the control group was 0.65±0.22. In 9 nmol AS-ODNs groups, IOD were 0.37±0.07, 0.20±0.05 and 0.25±0.07, respectively. In 18 nmol AS-ODNs groups, IOD were 0.11±0.05, 0.05±0.02 and 0.17±0.16, respectively. IOD in AS-ODNs groups were decreased significantly compared with control group (P<0.05), and these changes were in a dose dependent manner.
     The results indicatedthatAS-ODNs injected into the lateral ventricle inhibited expression of GLT-1 in a dose dependent manner, and the inhibiting effect was much significant at 24h.
     2.2 AS-ODNs of GLT-1 inhibit the neuro-protection on pyramidal neurons of the CA1 hippocampus induced by CIP
     Fifty-four Wistar rats with permanently occluded bilateral vertebral arteries were randomly assigned to seven groups.①Sham group (n=6): the bilateral common carotid arteries (BCCA) were separated, but without occluding the blood flow.②CIP group (n=6): the BCCA were clamped for 3 min.③Brain ischemic insult group (n=6): the BCCA were clamped for 8 min.④CIP+Brain ischemic insult group (n=6): 3 min CIP was preformed 2d prior to 8 min ischemic insult.⑤D istilled water group (n=6): 5μl distilled water was injected into the right lateral ventricle 12 h before, 12 h and 36 h after the BCCA were separated (but without occluding the blood flow), respectively.⑥AS-ODNs group(n=12): 5μl AS-ODNs solution was injected into the right lateral ventricle 12 h before, 12 h and 36 h after the BCCA were separated (but without occluding the blood flow), respectively. This group was further divided into 9 nmol and 18 nmol subgroups according to the doses of AS-ODNs (n=6 in each group).⑦A S-ODNs+CIP+Ischemic insult group (n=12): 5μl AS-ODNs solution was injected into the right lateral ventricle 12h before, 12h and 36h after CIP, respectively. This group was also further divided into 9 nmol and 18 nmol subgroups according to the doses of AS-ODNs. The other treatment was the same as CIP+Brain ischemic insult group, with six rats in each group. The animals were sacrificed by decapitation 7 d after sham operations or the last operations. Histological changes of the CA1 region of the hippocampus were examined using thionin staining (the methods were the same as that in part 1).
     Thionin staining showed that there was no significant neuronal damage in the CA1 subfield of the hippocampus in the sham and CIP groups. HG was 0~Ⅰ, and values of ND were 208.25±5.97 and 202.86±4.28 in the two groups, respectively. Obvious DND in the hippocampal CA1 region was found in ischemic insult for 8 min group. The value of ND was 45.86±21.93, which was much low compared with that in the sham and CIP groups (P<0.01). HG was gradeⅡ~Ⅲ, which was much high compared with that in the sham and CIP groups. In CIP+ischemic insult group, no obvious neuronal damage in the hippocampal CA1 region was found. HG (grade 0~Ⅰ) was significantly lower than that in 8 min brain ischemia group (P<0.01), and the value of ND was 208.98±5.9, which was much higher than that in 8 min brain ischemic insult group (P<0.01). The results indicated that CIP induced BIT of pyramidal neurons of the CA1 hippocampus against DND caused by brain ischemic insult. There was no apparent DND was found when distilled water and AS-ODNs (9 nmol and 18 nmol) were administrated into right lateral cerebral ventricle, and the histological changes were the same as those in sham and CIP groups. HG was grade 0~Ⅰand the values of ND were 176.22±6.29, 176.89±2.88 and 175.32±2.77, respectively. These results indicated that the injection of distilled water and AS-ODNs into the right lateral cerebral ventricle could not lead to apparent pyramidal neuron damage in the hippocampal CA1 region. In AS-ODNs +CIP + brain ischemic insult groups, DND were significantly obvious compared with that in CIP+ ischemic insult group. HG were gradeⅡ~Ⅲ, and ND (9 nmol and 18 nmol groups) were 133.56±3.42 and 70.94±7.38, respectively (P<0.01), Which indicated that AS-ODNs attenuated the neuro-protection on pyramidal neurons of the CA1 hippocampus induced by CIP via inhibiting the expression of GLT-1.
     The results indicated that the injection of AS-ODNs into the lateral cerebral ventricle induced the down-regulation of GLT-1 but no obvious DND in hippocampal CA1 region. However, the protective effect of CIP on pyramidal neurons of the CA1 hippocampus was attenuated by the injection of AS-ODNs. These results further suggested the association of GLT-1 with the induction of BIT by CIP in rats.
     3 Up-regulating the expression of GLT-1 with GLT-1 S-ODNs enhanced the tolerance to brain ischemic insult in the hippocampal CA1 region
     The role of GLT-1 in the brain ischemic tolerance was investigated by observing the effect of sense oligodeoxynucleotides (S-ODNs) of GLT-1 on tolerance to brain ischemic insult in the hippocampal CA1 region via up-regulating the protein expression of GLT-1.
     3.1 The effect of S-ODNs on the protein expression of GLT-1
     Forty-two Wistar rats with permanently occluded bilateral vertebral arteries for 36 h were randomly assigned to three groups.①control group (n=6): 5μl distilled water was injected into the right lateral cerebral ventricle and the animals were sacrificed by decapitation 12 h later.②S-ODNs 9 nmol group(n=18):5μl S-ODNs (9 nmol) solution was injected into the right lateral cerebral ventricle. This group was further divided into 12 h, 24 h and 36 h subgroups according to the time of reperfusion after injection.③S-ODNs 18 nmol group (n=18):5μl S-ODNs (18 nmol) solution was injected into the right lateral cerebral ventricle. This group was further divided into 12 h, 24 h and 36 h subgroups according to the time of reperfusion after injection. The animals were sacrificed by decapitation at the determined endpoint of the experiment. The hippocampal CA1 was dissected out quickly on ice and detected the expression of GLT-1 using Western blotting. IOD of each band was measured using image analysis system. The changes of GLT-1 expression were represented with the ratio of IOD of the aim protein toβ-actin.
     The results of Western blotting showed that IOD in the control group was 0.64±0.19. In 9 nmol S-ODNs groups, IOD were 0.78±0.02, 1.01±0.04 and 0.68±0.06, respectively. In 18 nmol S-ODNs groups, IOD were 0.84±0.02, 1.23±0.03 and 0.62±0.02, respectively. IOD in S-ODNs groups were up-regulated significantly compared with control group (P<0.05), and these changes were in a dose dependent manner.
     The results indicated that S-ODNs injected into the lateral ventricle up-regulated the protein expression of glutamate transporter subtype GLT-1 in a dose dependent manner, and this effect was much significant at 24 h. 3.2 S-ODNs of GLT-1 attenuated the injury of pyramidal neurons in the CA1 hippocampus induced by brain cerebral ischemia
     Fifty-four Wistar rats with permanently occluded bilateral vertebral arteries were randomly assigned to seven groups.①Sham group (n=6): the bilateral common carotid arteries (BCCA) were separated, but without occluding the blood flow.②CIP group (n=6): the BCCA were clamped for 3 min.③Brain ischemic insult group (n=6): the BCCA were clamped for 8 min.④CIP+Brain ischemic insult group (n=6): 3 min CIP was preformed 2d prior to 8 min ischemic insult.⑤D istilled water group (n=6): 5μl distilled water was injected into the right lateral cerebral ventricle 12h before, 12h and 36h after the BCCA were separated (but without occluding the blood flow) , respectively.⑥S-ODNs group(n=12): 5μl S-ODNs solution was injected into the right lateral cerebral ventricle 12h before, 12h and 36h after the BCCA were separated (but without occluding the blood flow), respectively. This group was further divided into 9 nmol and 18 nmol subgroups according to the doses of S-ODNs (n=6 in each group).⑦S -ODNs + Ischemic insult group (n=12): 5μl S-ODNs solution was injected into the right lateral cerebral ventricle 12 h before, 12 h and 36 h after the BCCA were separated (but without occluding the blood flow), respectively. The BCCA were clamped for 8 min after the BCCA were separated for 2 d. This group was also further divided into 9 nmol and 18 nmol subgroups according to the doses of S-ODNs. The animals were sacrificed by decapitation 7 d after sham operations or the last operations. Histological changes of the CA1 hippocampus were examined using thionin staining (the method was the same as that in part 1).
     Thionin staining showed that there was no significant neuronal damage in the CA1 subfield of the hippocampus in the sham and CIP groups. HG was 0~Ⅰ, and values of ND were 208.25±5.97 and 202.86±4.28 in the two groups, respectively. Obvious DND in the hippocampal CA1 region was found in ischemic insult for 8 min group and the value of ND was 45.86±21.93, which was much low compared with that in the sham and CIP groups (P<0.01). HG was gradeⅡ~Ⅲ, which was much high compared with that in the sham and CIP groups. In CIP+ischemic insult group, no obvious neuronal damage in the hippocampal CA1 region was found. HG (grade 0~Ⅰ) was significantly lower than that in 8 min brain ischemia group (P<0.01) and the value of ND was 208.98±5.9, which was much higher than that in 8 min brain ischemic insult group (P<0.01). The results indicated that CIP induced BIT of pyramidal neurons of the CA1 hippocampus against DND caused by brain ischemic insult. There was no apparent DND when distilled water and S-ODNs (9 nmol and 18 nmol) were administrated into right lateral cerebral ventricle, and the histological changes were the same as those in sham and CIP groups. HG was grade 0~Ⅰand the values of ND were 176.22±6.29, 186.53±5.64 and 204.4±12.99, respectively (P>0.05). In S-ODNs + Ischemic insult group, ND (they were 148.53±9.1 in 9 nmol group and 166.67±8.31 in 18 nmol group) was significantly increased compared with that in brain ischemic insult group (P<0.01). HG was gradeⅠwhich was significantly decreased compared with that in brain ischemic insult group. These results indicated that S-ODNs attenuate the damage of pyramidal neurons in the CA1 hippocampus induced by brain ischemic insult through up-regulating the expression of GLT-1.
     The results indicated that the injection of S-ODNs into the lateral cerebral ventricle led to the up-regulation of GLT-1 in hippocampal CA1 region. The damage induced by brain ischemic insult on pyramidal neurons of the CA1 hippocampus was attenuated by the injection of S-ODNs. These results further suggested the role of GLT-1 in the induction of BIT by CIP in rats.
     4 Conclusions
     (1) DHK, an inhibitor selective for glial glutamate transporter GLT-1, inhibited the protective effect induced by CIP on pyramidal neurons in the CA1 subfield of the hippocampus via inhibiting the function of GLT-1 in rats.
     (2) The injection of AS-ODNs of GLT-1 into the lateral cerebral ventricle inhibited the expression of glutamate transporter subtype GLT-1. At the same time, AS-ODNs of GLT-1 attenuated the neuro-protection on pyramidal neurons of the CA1 hippocampus induced by CIP.
     (3) The injection of S-ODNs of GLT-1 into the lateral cerebral ventricle enhanced the tolerance to brain ischemic insult in the hippocampal CA1 region as wll as up-regulating the protein expression of GLT-1.
     (4) These results indicated that GLT-1 participates in BIT induced by CIP.
引文
1 Matsumoto Y, Yamamoto S, Suzuki Y, et al. Na+/H+ exchanger inhibitor, SM-20220, is protective against excitotoxicity in cultured cortical neurons. Stroke, 2004, 35(1): 185~190
    2 Katsuki H, Akaike A. Excitotoxic degeneration of hypothalamic orexin neurons in slice culture. Neurobiol Dis, 2004, 15(1): 61~69
    3 O’Shea RD. Roles and regulation of glutamate transporters in the central nervous system. Clin Exp Pharmacol Physiol, 2002, 29(11): 1018~1023
    4 Sullivan R, Rauen T, Fischer F, et al.Cloning, transport properties, and differential localization of two splice variants of GLT-1 in the rat CNS: Implications for CNS glutamate homeostasis. Glia, 2004, 45(2): 155~169
    5 Kanai Y, Hediger MA. The glutamate and neutral amino acid transporter family: physiological and pharmacological implications. Eur J Pharmacol, 2003, 479 (1-3): 237~247
    6 Hu WH, Walters WM, Xia XM, et al. Neuronal glutamate transporter EAAT4 is expressed in astrocytes. Glia, 2003, 44(1): 13~25
    7 Rothstein JD, Patel S, Regan MR, et al. Beta-lactam antibiotics offer neuroprotection by increasing glutamate transporter expression. Nature, 2005, 433(7021): 73~77
    8 Pulsinelli WA, and Brierley JB. A new model of bilateral hemispheric ischemia in the unanesthetized rat. Stroke, 1979, 10(3): 267~272
    9 耿进霞,张敏,李文斌,等。凝闭双侧椎动脉预处理对大鼠全脑缺血损伤的防护作用。中国应用生理学杂志,2007,23(1):24~29
    10 Sun JL, Duan SM, Wang J, et al. Effects of MK-801 or NBQX, NMDA by intracerebroventricular injection on the lowest anesthesia effective concentration of isoflurane in rats. Chin J Anesthesiol, 2002, 22(5): 315~316
    11 Feng RF, Li WB, Liu HQ, et al. Effects of α-methyl-(4-tetrazolyl-phenyl) glycine on the induction of hippocampal ischemic tolerance in the rat. Acta Physiol Sin, 2003, 55(3): 303~310
    12 Fukui M, Nakagawa T, Minami M, et al. Inhibitory role of supraspinal P2X3/P2X2/3 subtypes on nociception in rats. Mol Pain, 2006, 5(2): 19
    13 Kato H,Liu Y,Araki T,et al. Temporal profile of the effects of pretreatment with brief cerebral ischemic on the neuronal damage following secondary ischemic insult in the gerbil:cumulative damage and protective effects.Brain Res,1991,553:238~242
    14 Li WB, Zhou AM, Li QJ, et al. Experimental research on brain ischemic tolerance in the hippocampus induced by cerebral ischemic preconditioning in rats. Chin J App Physiol, 2002, 18(2): 109~113
    15 Meller R, Minami M, Cameron JA, et al. CREB-mediated Bcl-2 protein expression after ischemic preconditioning. J Cereb Blood Flow Metab, 2005, 25(2): 234~246
    16 Kirino T. Ischemic tolerance. J Cereb Blood Flow Metab, 2002, 22(11): 1283~1296
    17 Bickler PE, Fahlman CS.Moderate increases in intracellular calcium activate neuroprotective signals in hippocampal neurons. Neuroscience, 2004, 127(3): 673~683
    18 Yellon DM,Baxter GF,Garcia-Dorado D,et al. Ischemic preconditioning: present position and future directions. Cardiovasc Res, 1998, 37(1): 21~33
    19 Zhou AM, Li QJ, Chen XL, et al. Increase in amount and affinity of adenosine receptor in rat hippocampal cellular membrane sinduced by cerebral ischemic preconditioning and its protective effects on the neurons. Acta Physiol Sin, 2001, 53(4): 265~269
    20 Real MF. Mechanism of exitotoxicity in neurologic disease. FASEBJ, 1992, 6(15): 3338~3344
    21 Bruhn T, Levy LM, Nielsen M, et al. Ischemia induced changes in expression of the astrocyte glutamate transporter GLT-1 in hippocampus of the rat. Neurochem Int, 2000, 37(2-3): 277~285
    22 Torp R, Lekieffre D, Levy LM, et al. Reduced postischemic expression of a glial glutamate transporter, GLT-1, in the rat hippocampus. Exp BrainRes, 1995, 103(1): 51~58
    23 Rao VL, Dogan A, Todd KG, et al. Antisense knockdown of the glial glutamate transporter GLT-1, but not the neuronal glutamate transporter EAAC1, exacerbates transient focal cerebral ischemia-induced neuronal damage in rat brain. J Neurosci, 2001, 21(6): 1876~1883
    24 Chen W, Aoki C, Mahadomrongkul V, et al. Expression of a variant form of the glutamate transporter GLT-1 in neuronal cultures and in neurons and astrocytes in the rat brain. J Neurosci, 2002, 22(6): 2142~2152
    25 Li JQ, Zhang LF, Wang YS, et al. Isoflurane preconditioning induces neuroprotection against global cerebral ischemia reperfusion in rat. Chin J Anesthesiol, 2003, 23(7): 515~518
    26 Sakai F, Amaha K. Midazolam and ketamine inhibit glutamate release via a cloned human brain glutamate transporter. Can J Anaesth, 2000, 47(8): 800~806
    27 Alkire MT, Haier RJ, Barker SJ, et al. Cerebral metabolism during propofol anesthesia in humans studied with positron emission tomography. Anesthesiology, 1995, 82(2): 393~403
    28 Rasool N, Faroqui M, Rubinstein EH. Lidocaine accelerates neuroelectical recovery after incomplete global ischemia in rabbits. Stroke, 1990, 21: 929
    1 Matsumoto Y, Yamamoto S, Suzuki Y, et al. Na+/H+ exchanger inhibitor, SM-20220, is protective against excitotoxicity in cultured cortical neurons. Stroke, 2004, 35(1): 185~190
    2 Katsuki H, Akaike A. Excitotoxic degeneration of hypothalamic orexin neurons in slice culture. Neurobiol Dis, 2004, 15(1): 61~69
    3 Rintoul GL,Filiano AJ,Brocard JB, etal.Glutamate decrease mitochondrial size and movement in primary forebrain neurons.J Neurosci, 2003, 23 (21): 7881~7888
    4 Nicotera P, Bano D.The enemy at the gates.Ca2+ entry through TRPM7 channels and anoxic neuronal death. Cell,2003,115(7):768~770
    5 Zacco A,Togo J,Spence K, et al.3-hydroxy-3-methylglutaryl conenzyme A reductase inhibitors protect cortical neurons from excitotoxicity. Neurosci, 2003,23(35):11104~11111
    6 Sanchez-Gomez MV, Alberdi E, Ibarretxe G, et al.Caspase-dependent and caspase-independent oligodendrocyte death mediated by AMPA andkainate receptors. Neurosci,2003,23(29):9519~9528
    7 Whetsell WO Jr.J. Current concepts of excitotoxicity. Neuropathol Exp Neurol,1996,55(1):1~13
    8 Kitagawa K, Matsumoto M, Tagaya M, et al. “ischemic tolerance” phenomenon found in the brain. Brain Res, 1990, 528: 21~24
    9 Kato H, Liu Y, Araki T, et al. Temporal profile of the effects of pretreatment with brief cerebral ischemia on the neuronal damage following secondary ischemic insult in the gerbil: cumulative damage and protective effects. Brain Res, 1991, 553: 238~242
    10 Kitagawa K, Matsumoto M, Kuwabara K, et al. ‘Ischemic tolerance’ phenomenon detected in various brain regions. Brain Res, 1991, 561: 203~211
    11 Kirino T, Tsujita Y, Tamura A, et al. Induced tolerance to ischemia in gerbil hippocampal neurons. J Cereb Blood Flow Metab, 1991, 11(2): 299~307
    12 Chen J, Graham SH, Zhu RL, et al. Stress proteins and tolerance to focal cerebral ischemia. J Cereb Blood Flow Metab, 1996, 16: 566~577
    13 Glazier SS, O'Rourke DM, Graham DI, et al. Induction of ischemic tolerance following brief focal ischemia in rat brain. J Cereb Blood Flow Metab, 1994, 14(4): 545~553
    14 Kato H, Kogure K, Arake T, et al. Astroglial and microglial reactions in the gerbil hippocampus with induced ischemic tolerance. Brain Res, 1994, 664(1-2): 69~76
    15 张敬军,陈青,孙思琴. 海马区星形胶质细胞的活化状态与缺血耐受性关系的研究. 中国病理生理杂志,2000,16(2): 160~162
    16 汪长胜,霍正禄,杨瑞和,等. 缺血预处理后海马 CA1 区反应性星形胶质细胞增生与迟发性神经元缺血耐受性的关系. 临床神经病学杂志, 2001, 14(3): 152~154
    17 Rothstein JD, Patel S, Regan MR, etal. Beta-lactam antibiotics offer neuroprotection by increasing glutamate transporter expression. Nature, 2005, 433(7021): 73~77
    18 张敬军,李天富,夏作理。cGMP对脑缺血再灌流后海马区星形胶质细胞活化的作用机制研究。神经解剖学杂志,2002,18(2):181~183
    19 Rothstein JD, Dykes-Hoberg M, Pardo CA, etal. Knockout of glutamate transporters reveals a major role for astroglial transport in excitotoxicity and clearance of glutamate. Neuron,1996 16:675~686
    20 Vemuganti L, Raghavendra Rao, Aclan Dogan, etal. Antisense knockdown of the glial glutamate transporter GLT-1, but not the neuronal glutamate transporter EAAC1, exacerbates transient focal cerebral ischemia-induced neuronal damage in rat brain. The Journal of Neuroscience, 2001, 21(6):1876~1883
    21 Rao VL, Dogan A, Bowen KK, etal. Antisense knockdown of the glial glutamate transporter GLT-1 exacerbates hippocampal neuronal damage following traumatic injury to rat brain. Eur J Neurosci, 2001, 13(1): 119~128
    22 Romera C,Hurtado O,Botella SH, etal. In vitro ischemic tolerance involves upregulation of glutamate transport partly mediated by the TACE/ADAM 17-tumor necrosis factor-alpha pathway.J Neurosci,2004,24(6):1350~1357
    23 Payet O,Maurin L, Bonne C, etal. Hypoxia stimulates glutamate uptake in whole rat retinal cells in vitro. Neurosci Lett, 2004,356(2):148~150
    1 Rothstein JD, Patel S, Regan MR, et al. Beta-lactam antibiotics offer neuroprotection by increasing glutamate transporter expression. Nature, 2005, 433(7021): 73~77
    2 Petito CK, Halaby IA. Relationship between ischemia and ischemic neuronal necrosis to astrocyte expression of glial fibrillary acidic protein. Int J Dev Neurosci, 1993,11(2):239~247
    3 Vemuganti L , Raghavendra Rao, Aclan Dogan, et al. Antisense knockdown of the glial glutamate transporter GLT-1, but not the neuronal glutamate transporter EAAC1, exacerbates transient focal cerebral ischemia-induced neuronal damage in rat brain. The Journal of Neuroscience, 2001, 21(6):1876~1883
    4 Rao VL, Dogan A, Bowen KK, et al. Antisense knockdown of the glial glutamate transporter GLT-1 exacerbates hippocampal neuronal damage following traumatic injury to rat brain. Eur J Neurosci, 2001, 13(1): 119~128
    5 Guo H, Lai L, Butchbach ME, et al. Increased expression of the glial glutamate transport EAAT2 modulates excitotoxicity and delays the onset but not the outcome of ALS in mice. Hum Mol Genet, 2003, 12 (19): 2519~2532
    6 O’Shea RD. Roles and regulation of glutamate transporters in the central nervous system. Clin Exp Pharmacol Physiol, 2002, 29(11): 1018-1023.
    7 Kitagawa K, Matsumoto M, Tagaya M, et al.“ischemic tolerance” phenomenon found in the brain. Brain Res, 1990, 528: 21~24
    8 Kato H, Liu Y, Araki T, et al. Temporal profile of the effects of pretreatment with brief cerebral ischemia on the neuronal damage following secondary ischemic insult in the gerbil: cumulative damage andprotective effects. Brain Res, 1991, 553: 238~242
    9 Kitagawa K, Matsumoto M, Kuwabara K, et al. ‘Ischemic tolerance’ phenomenon detected in various brain regions. Brain Res, 1991, 561: 203~211
    10 Kirino T, Tsujita Y, Tamura A, et al. Induced tolerance to ischemia in gerbil hippocampal neurons. J Cereb Blood Flow Metab, 1991, 11(2): 299~307
    11 Chen J, Graham SH, Zhu RL, et al. Stress proteins and tolerance to focal cerebral ischemia. J Cereb Blood Flow Metab, 1996, 16: 566~577
    12 Glazier SS, O'Rourke DM, Graham DI, et al. Induction of ischemic tolerance following brief focal ischemia in rat brain. J Cereb Blood Flow Metab, 1994 , 14 (4): 545~553
    13 Shimizu S, Nagayama T, Jin KL, et al. bcl-2 Antisense treatment prevents induction of tolerance to focal ischemia in the rat brain. J Cereb Blood Flow Metab, 2001, 21(3):233~243
    14 Shamloo M, Rytter A, Wieloch T. Activation of the extracellular signal-regulated protein kinase cascade in the hippocampal CA1 region in a rat model of global cerebaral ischemic preconditioning. Neuroscience, 1999, 93(1):81~88
    15 Hiraide T, Katsura K, Muramatsu H, et al. Adenosine receptor antagonists cancelled the ischemic tolerance phenomenon in gerbil. Brain Res, 2001, 910(1-2):94~98
    16 Zhou AM,Li WB,Li QJ,等. Increase in amount and affinity of adenosine receptor in rat hippocampal cellular membranes induced by cerebral ischemic preconditioning and its protective effects on the neurons. Acta Physiol Sin, 2001, 53(4): 265~269
    17 Zhou AM, Li WB, Li QJ, et al. A short cerebral ischemic preconditioning up-regulates adenosine receptors in the hippocampal CA1 region of rats. Neurosci Res, 2004, 48(4):397~404
    18 Kato H, Liu Y, Araki T, Kogure K. Temporal profile of the effects of pretreatment with brief cerebral ischemia on the neuronal damagefollowing secondary ischemic insult in the gerbil:cumulative damage and protective effects. Brain Res, 1991, 12;553(2):238~242
    19 Miyashita K, Abe H, Nakajima T, et al. Induction of ischaemic tolerance in gerbil hippocampus by pretreatment with focal ischaemia. Neuroreport, 1994, 30;6(1):46~48
    20 刘惠卿, 李文斌, 冯荣芳, 等. 一氧化氮合酶抑制剂 L-NAME 对大鼠脑缺血耐受诱导的影响. 生理学报, 2003, 55(2):219~224
    21 Kurkinen K, Busto R, Goldsteins G, et al. Isoform-specific membrane translocation of protein kinase C after ischemic preconditioning. Neurochem Res, 2001, 26(10):1139~1144
    22 Gu Z, Jiang Q, Zhang G, et al. Diphosphorylation of extracellular signal-regulated kinases and c-Jun N-terminal protein kinases in brain ischemic tolerance in rat. Brain Res, 2000, 860(1-2):157~160
    23 Sommer C, Gass P, Kiessling M. Selective c-JUN expression in CA1 neurons of the gerbil hippocampus during and after acquisition of an ischemia-tolerant state. Brain Pathol, 1995,5(2):135~144
    24 Belayev L, Ginsberg MD, Alonso OF, et al. Bilateral ischemic tolerance of rat hippocampus induced by prior unilateral transient focal ischemia: relationship to c-fos mRNA expression. Neuroreport, 1996, 20,8(1): 55~59
    25 Perdrizet GA, Lena CJ, Shapiro DS, et al. Preoperative stress conditioning prevents paralysis after experimental aortic surgery: increased heat shock protein content is associated with ischemic tolerance of the spinal cord. J Thorac Cardiovasc Surg, 2002, 124(1):162~170
    26 Lepore DA, Knight KR, Anderson RL, et al. Role of priming stresses and Hsp70 in protection from ischemia-reperfusioninjury in cardiac and skeletal muscle. Cell Stress Chaperones, 2001, 6(2):93~96
    27 Hampton CR, Shimamoto A, Rothnie CL, et al. HSP70.1 and HSP70.3 are required for late-phase protection induced by ischemic preconditioning of mouse hearts. Am J Physiol Heart Circ Physiol, 2003, 285(2):H866~874
    28 Mori T, Muramatsu H, Matsui T, et al. Possible role of the superoxideanion in the development of neuronal tolerance following ischemic preconditioning in rats. Neuropathol Appl Neurobiol, 2000,26(1):31~44
    1 Matsumoto Y, Yamamoto S, Suzuki Y, et al. Na+/H+ exchanger inhibitor, SM-20220, is protective against excitotoxicity in cultured cortical neurons. Stroke, 2004, 35(1): 185~190
    2 Katsuki H, Akaike A. Excitotoxic degeneration of hypothalamic orexin neurons in slice culture. Neurobiol Dis, 2004, 15(1): 61~69
    3 Rintoul GL,Filiano AJ,Brocard JB, et al.Glutamate decrease mitochondrial size and movement in primary forebrain neurons.J Neurosci, 2003, 23 (21): 7881~7888
    4 Nicotera P, Bano D.The enemy at the gates.Ca2+ entry through TRPM7 channels and anoxic neuronal death. Cell,2003,115(7):768~770
    5 Zacco A, Togo J, Spence K, et al. 3-hydroxy-3-methylglutaryl conenzyme A reductase inhibitors protect cortical neurons from excitotoxicity. Neurosci, 2003,23(35):11104~11111
    6 Sanchez-Gomez MV, Alberdi E, Ibarretxe G, et al.Caspase-dependent and caspase-independent oligodendrocyte death mediated by AMPA and kainate receptors. Neurosci,2003,23(29):9519~9528
    7 Whetsell WO Jr.J. Current concepts of excitotoxicity. Neuropathol Exp Neurol,1996,55(1):1~13
    8 韩济生. 神经科学原理,第二版. 北京: 北京医科大学出版社,1999:534~538
    9 Rothstein JD, Patel S, Regan MR, et al. Beta-lactam antibiotics offerneuroprotection by increasing glutamate transporter expression. Nature, 2005, 433(7021): 73~77
    10 O’Shea RD. Roles and regulation of glutamate transporters in the central nervous system. Clin Exp Pharmacol Physiol, 2002, 29(11): 1018~1023
    11 Chaudhry FA, Lehre KP, Campagne MV, et al. Glutamate transporters in glia lplasma membranes: highly differentiated localizations revealed by quantitative ultrastructural immunocytochemistry. Neuron, 1995, 15: 711~720
    12 Rothstein JD, Martin L, LeveyAI, et al. Localization of neuronal and glial glutamate transpoters. Neuron, 1994,13: 713~725
    13 Watase K, Hashimoto K, Kano M, et al. Motor discoordination and increased susceptibility to cerebral injury in GLAST mutant mice. Eur. J Neurosci, 1998,10: 976~988
    14 Sullivan R, Rauen T, Fischer F, et al.Cloning, transport properties, and differential localization of two splice variants of GLT-1 in the rat CNS: Implications for CNS glutamate homeostasis. Glia, 2004, 45(2): 155~169
    15 Kanai Y, Hediger MA. The glutamate and neutral amino acid transporter family: physiological and pharmacological implications. Eur J Pharmacol, 2003, 479 (1-3): 237~247
    16 Hu WH, Walters WM, Xia XM, et al. Neuronal glutamate transporter EAAT4 is expressed in astrocytes. Glia, 2003, 44(1): 13~25
    17 Nagao S, Kwak S, Kanazawa I. EAAT4, a glutamate transporter with properties of achloride channel, is predominantly localized in Purkinje cell dendrites, and forms parasagittal compartments in rat cerebellum. Neuroscience, 1997,78: 929~933
    18 Schluter K, Figiel M, Rozyczka J,et al. CNS region-specific regulation of glial glutamate transporter expression . Eur J Neurosci, 2002,16: 836~842
    19 韩济生.神经科学原理,第二版. 北京: 北京医科大学出版社,1999:306~315
    20 韩济生.神经科学原理,第二版. 北京: 北京医科大学出版社,1999:525~526
    21 Auger C, Attwell D. Fast removal of synaptic glutamate by postsynaptic transporters. Neuron, 2000, 28: 547~558
    22 韩济生.神经科学原理,第二版. 北京: 北京医科大学出版社,1999:536~537
    23 Nicholas J, Maragakis, Jeffrey D, et al. Glutamate transporters in neurologic disease. Basic Sci Semin Neurol, 2001,58: 365~370
    24 Maragakis NJ, Rothstein JD. Glutamate transporters: animal models to neurologic disease. Neurobiol Dis, 2004, 15(3):461~73.
    25 Shaw PJ. Calcium, glutamate, and amyotrophic lateral sclerosis: more evidence but on certainties. Ann. Neurol,1999,46:803~805
    26 Rothstein JD, Van Kammen M, Levey, et al. Selective loss of glial glutamate transporter GLT-1 in amyotrophic lateral sclerosis. Ann. Neurol, 1995,38:73~84
    27 Tian G, Lai L, Guo H,et al. Translational control of glial glutamate transporter EAAT2 expression. J Biol Chem, 2007, 19, 282 (3): 1727 ~1737
    28 Okada K, Yamashita U, Tsuji S. Modulation of Na(+)-dependent glutamate transporter of murine astrocytes by inflammatory mediators. J UOEH,2005, 27(2):161~170
    29 Boston-Howes W, Gibb SL, Williams EO, et al. Caspase-3 cleaves and inactivates the glutamate transporter EAAT2. J Biol Chem, 2006, 19,281 (20):14076~14084.
    30 韩济生.神经科学原理,第二版. 北京: 北京医科大学出版社,1999:1111~1113
    31 Holthoff VA, Vieregge P, Kessler J,et al. Discordant twins with Parkinson’s disease: positron emission tomography and early signs of impaired cognitive circuits . Ann Neurol, 1994, 36 (2): 176~182
    32 Yoshino H, Hattori Y, Imai H, et al. Sparteine oxidation by hepatic cytochrome P2450 in patients with Parkinson’s disease. Rinsho Shinkeigaku, 1993, 33(3): 261~265
    33 Iacopino AM, Christakos S. Specific reduction of calcium-binding protein(28-kilodaltoncal bindin-D) gene expression in aging and neurodegenerative diseases. Proc Natl Acad Sci USA, 1990, 87 (11): 4078~4082
    34 Zhang P, Land W, Lee S, et al. Electron tomography of degenerating neurons in mice with abnormal regulation of iron metabolism .J Struct Biol, 2005,150 (2): 144~153
    35 Defazio G, Dal Toso R, Benvegnu D, et al. Parkinsonian serum carries complement –dependent toxicity for rat mesencephalic dopaminergic neurons in culture. Brain Res, 1994, 633(1/2): 206~212
    36 McGeer PL, Itagaki S, Boyes BE. Reactive microglias are positive for HLA-DR in the substantia nigra of Parkinson’s and Alzheimercs disease brains. Neurology, 1988, 38(8): 1285~1291
    37 Le WD, Colom LV, Xie WJ, et al. Cell death induced by beta-amy-loid 1-40 in MES 23.5 hybrid clone: the role of nitric oxide and NMDA-gated channel activation leading to apoptosis . Brain Res, 1995, 686(1) :49~60
    38 Tamas A, Lubics A, Szalontay L, et al. Age and gender differences in behavioral and morphological out come after 6-hydroxydopamine-induced lesion of the substantia nigra in rats. Behav Brain Res, 2005, 158(2): 221~229
    39 Plaitakis A, Shashidharan P. Glutamate transport and metabolism in dopaminergic neurons of substantia nigra: implications for the pathogenesis of Parkonson’s disease. J .Neurol, 2000,247: II 25~35
    40 Zipp F, Demisch L, Derouiche A, et al. Glutamine synthetase activity in patients with Parkinson’s disease . Acta Neurol Scand, 1998,97:300~302
    41 Berman SB, Hastings TG. Inhibition of glutamate transport in synaptosomes by dopamine oxidation and reactive oxygen species. J Neurochem,1997,69:1185~1195
    42 Dabir DV, Robinson MB, Swanson E, et al. Impaired glutamate transport in a mouse model of tau pathology in astrocytes.J Neurosci,2006, 26(2): 644~654
    43 Wilson JM, Khabazian I, Pow DV, et al. Decrease in glial glutamatetransporter variants and excitatory amino acid receptor down-regulation in a murine model of ALS-PDC. Neuromolecular Med,2003,3(2):105~118
    44 Wilson JM, Petrik MS, Moghadasian MH, et al. Examining the interaction of apo E and neurotoxicity on a murine model of ALS-PDC. Can J Physiol Pharmacol,2005,83(2):131~141
    45 韩济生.神经科学原理,第二版. 北京: 北京医科大学出版社,1999:1104~1111
    46 韩济生. 神经科学原理,第二版. 北京: 北京医科大学出版社,1999:1108-1111.
    47 Stuart A. Lipton, Paul A. Rosenberg. Excitatory amino acids as a final common pathway for neurologic disordes. New Engl J Med, 1994, 330(9): 613~622
    48 Beveniste H, Drejer J, Schousboe A, et al. Elevation of the extracellular concentration of glutamate and aspartate in rat hippocampus during transient ischemia monitored by intracerebral microdialysis. J Neurochem, 1984, 43(5): 1369~1374
    49 李军,曾邦雄. 脑缺血性损伤级联反应的研究进展. 国外医学麻醉学与复苏分册, 2001,6(22): 364~368
    50 Krenz NR, Weaver LC. Effect of spinal cord transection on N-methy-D-aspartate receptors in the cord. J Neurotraum,1998, 15(12):1027~1036
    51 Jacob CP, Koutsilieri E, Bartl J, et al. Alterations in expression of glutamatergic transporters and receptors in sporadic Alzheimer's disease. J Alzheimers Dis,2007,11(1):97~116
    52 韩济生.神经科学原理,第二版. 北京: 北京医科大学出版社,1999:1085~1102
    53 Campiani G, Fattorusso C, De Angelis M, et al. Neuronal high-affinity sodium-dependent glutamate transporters (EAATs): targets for the development of novel therapeutics against neurodegenerative diseases. Curr Pharm Des, 2003, 9 (8):599~625
    54 Sullivan R, Rauen T, Fischer F, et al. Cloning, transport properties, anddifferential localization of two splice variants of GLT-1 in the rat CNS: implications for CNS glutamate homeostasis. Glia,2004, 45(2):155~169
    55 Danbolt NC. Glutamate uptake. Prog Neurobiol,2001 ,65(1):1~10
    56 Tanaka K, Watase K, Manabe T. Epilepsy and exacerbation of brain injury in mice lacking the glutamate transporter EAA'T2-1.Science, 1997,276(5319):1699~1702
    57 Watanabe T, Morimoto K, Hirao T.Amygdala-kindledand pentylenetetra- zole-induced seizures in glutamate transporters EAATl-deficient mice. Brain Res, 1999, 845(1):92~96
    58 邱永明,陆兆丰,江基尧,等. 谷氨酸转运体在全脑缺血性癫痫中作用的研究. 立体定向和功能性神经外科杂志,2005,18 (5):275~279
    59 李林繁,麦洁文,周迩. 各种谷氨酸转运体亚型在颞叶癫痫患者病变部位的表达. 海南医学,2005,16(6):51~53
    60 韩济生.神经科学原理,第二版. 北京: 北京医科大学出版社,1999:1064~1069
    61 Mitani A, Tanaka K. Functional changes of glial glutamate transporter GLT-1 during ischemia: An in vivo study in the hippocampal CA1 of normal mice and mutant mice lacking GLT-1. J Neurosci,2003,23(18): 7176~7182
    62 Velly LJ, Guillet BA, Masmejean FM, et al. Neuroprotective effects of propofol in model of ischemic cortical cell cultures: role of glutamate and its transporters. Anesthesiology,2003,99(2):368~375
    63 Kanai Y, Hediger MA. The glutamate and neutral amino acid transporter family: physiological and pharmacological implications. Eur J Pharmacol, 2003, 479 (1-3): 237~247
    64 Vemuganti L , Raghavendra Rao, Aclan Dogan, et al. Antisense knockdown of the glial glutamate transporter GLT-1, but not the neuronal glutamate transporter EAAC1, exacerbates transient focal cerebral ischemia-induced neuronal damage in rat brain. The Journal of Neuroscience, 2001, 21(6):1876~1883
    65 Rao VL, Dogan A, Todd KG, et al. Antisense knockdown of the glialglutamate transporter GLT-1, but not the neuronal glutamate transporter EAAC1, exacerbates transient focal cerebral ischemia-induced neuronal damage in rat brain. J Neurosci, 2001, 21(6): 1876~1883
    66 Rao VL, Dogan A, Bowen KK, et al. Antisense knockdown of the glial glutamate transporter GLT-1 exacerbates hippocampal neuronal damage following traumatic injury to rat brain. Eur J Neurosci, 2001, 13(1): 119~128
    67 张敬军,李天富, 夏作理. cGMP 对脑缺血再灌流后海马区星形胶质细胞活化的作用机制研究. 神经解剖学杂志,2002,18(2):181~183
    68 Hinoi E, Takarada T, Tsuchihashi Y, et al.Glutamate transporters as drug targets. Curr Drug Targets CNS Neurol Disord,2005,4(2):211~220
    1 Woodburne VL, Hayward NU, Poat JA. The effect of dizocilpine and inadoline on immediate early gene expression in the gerbil global ischemia model. Neuropharmacology,1993,32(10):1047
    2 Mudrick LA, Baimbridge KG. Long-term changes in the rat hippocampal formation following cerebral ischemia. Brain Res,1989,493(1):179
    3 Juurlink BH, Sweeney MI. Mechanisms that result in damage during andfollowing cerebral ischemia. Neuro Bio Res,1997,21(2):121
    4 Kitagawa K, Matsumoto M, Tagaya M, et al. “ischemic tolerance” phenomenon found in the brain. Brain Res, 1990, 528: 21~24
    5 王尧,杜子威.神经生物化学与分子生物学.北京:人民卫生出版社,1997,390
    6 Rosenberg PA, Azenman E. Hundred-fold increase in neuronal vulnerability to glutamate toxicity in astrocyte-poorcultures of rat cerebral cortex.Neuro Lett, 1998,103(2):162
    7 Matsumoto Y, Yamamoto S, Suzuki Y, et al. Na+/H+ exchanger inhibitor, SM-20220, is protective against excitotoxicity in cultured cortical neurons. Stroke, 2004, 35(1): 185~190
    8 Katsuki H, Akaike A. Excitotoxic degeneration of hypothalamic orexin neurons in slice culture. Neurobiol Dis, 2004, 15(1): 61~69
    9 Rintoul GL,Filiano AJ,Brocard JB, et al.Glutamate decrease mitochondrial size and movement in primary forebrain neurons.J Neurosci, 2003, 23 (21): 7881~7888
    10 Nicotera P, Bano D.The enemy at the gates.Ca2+ entry through TRPM7 channels and anoxic neuronal death. Cell,2003,115(7):768~770
    11 Zacco A,Togo J,Spence K, et al. 3-hydroxy-3-methylglutaryl conenzyme A reductase inhibitors protect cortical neurons from excitotoxicity. Neurosci, 2003,23(35):11104~11111
    12 Sanchez-Gomez MV, Alberdi E, Ibarretxe G, et al.Caspase-dependent and caspase-independent oligodendrocyte death mediated by AMPA and kainate receptors. Neurosci,2003,23(29):9519~9528
    13 Whetsell WO Jr.J. Current concepts of excitotoxicity. Neuropathol Exp Neurol,1996,55(1):1~13
    14 韩济生. 神经科学原理,第二版. 北京: 北京医科大学出版社,1999:534~538
    15 Mayer B, Hemmens B. Biosynthesis and action of NO in ammalian cells. Trends Biochem Sci, 1997, 22(12):477
    16 Siliver IA, Erencinska M. Ion homeostasis in rat brain in vivo intra-andextracellular Ca2+ and H+ in the hippocampus during recovery from short-term transient ischemia. J Cereb Blood Flow Metab, 1992,12(5):722
    17 Zaidan E,Sims N. The calcium content of mitchondria from brain subregions following short-term forebrain ischemia and recirculation in the rat. J Neur, 1994,63(5):1812
    18 Beckman JS, Ye Yz, Chen J. The interaction of nitric oxide with oxygen radicals and scavenger in cerebral ischemic injury. Adv Neurol, 1996,71(1):339
    19 Zini I, Tomasi A, Grimaldi R, et al. Detection of free radicals during brain ischemia and reperfusion by spintrapping and microdialysis. Neurosci Lett, 1992,138(2):279
    20 Kitagawa K, Matsumoto M, Kuwabara K, et al. Ischemic tolerance phenomenon detected in various brain regions. Brain Aes. 1991,561:203
    21 Dowden J, Corbett D. Ischemic preconditioning in 18- to 20-month-old gerbils: long-term survival with functional outcome measures. Stroke, 1999, 30(6):240~246
    22 Kirino T, Tsujita Y, Tamura A. Induced tolerance to ischemia in gerbil hippocampal neurons. J Cereb Blood Flow Metab, 1991, 11(2):299~307
    23 Kato H, Liu Y, Araki T, et al. Temporal profile of the effects of pretreatment with brief cerebral ischemia on the neuronal damage following secondary ischemic insult in the gerbil: cumulative damage and protective effects. Brain Res, 1991, 553(2):238~242
    24 Liu Y, Kato H, Nakata N, et al. Protection of rat hippocampus against ischemic neuronal damage by pretreatment with sublethal ischemia. Brain Res, 1992, 586(1):121~124
    25 Nishi S, Taki W, Uemura Y, et al. Ischemic tolerance due to the induction of HSP70 in a rat ischemic recirculation model. Brain Res, 1993, 615(2):281~288
    26 Wu C, Zhan RZ, Qi S, et al. A forebrain ischemic preconditioning model established in C57Black/Crj6 mice. J Neurosci Methods, 2001, 107(1-2):101~106
    27 Lin Y, Kato H, Nakata N, et al. Protection of rat hippocampus against ischemic neuronal damage by pretreatment with subletha ischemia. Brain Res,1992,586:121
    28 Matsushima K, Hakim AM. Transient forebrain protects against subsequent focal cerebral ischemia without changing cerebral perfusion. Stroke, 1995, 26:1047
    29 Kitagawa K, Matsumoto M, Matsushita K, et al. Ischemic tolerance in moderately symptomatic gerbils after unilateral carotid occlusion. Brain Res, 1996, 716(1-2):39~46
    30 Glazier SS, O'Rourke DM, Graham DI, et al. Induction of ischemic tolerance following brief focal ischemia in rat brain. J Cereb Blood Flow Metab, 1994, 14(4):545~553
    31 Mullins PG, Reid DG, Hockings PD, et al. Ischemic preconditioning in the rat brain: a longitudinal magnetic resonance imaging (MRI) study. NMR Biomed, 2001, 14(3):204~209
    32 Masada T, Hua Y, Xi G, et al. Attenuation of ischemic brain edema and cerbrovascular injury after ischemic preconditioning in the rat. J Cereb Blood Flow Metab, 2001, 21(1):22~33
    33 Belayev L, Ginsberg MD, Alonso OF, et al. Bilateral ischemic tolerance of rat hippocampus induced by prior unilateral transient focal ischemia: relationship to c-fos mRNA expression. Neuroreport, 1996, 20,8(1): 55~59
    34 Simon RP, Niiro M, Gwinn R. Prior ischemic stress protects against experimental stroke. Neurosci Lett, 1993, 163(2):135~137
    35 Takahata Y, Shimoji K. Brain injury improves survival of mice following brain ischemia. Brain Res, 1986, 381(2):368~371
    36 Faden AI, Demediuk P, Panter SC, et al, The role of excitatory amino acids and NMDA receptors in traumatic brain injury. Science, 1989, 244(4906):798~800
    37 Brown IR, Rush S, Ivy GO. Induction of a heat shock gene at the site of tissue injury in the rat brain. Neuron, 1989, 2(6):1559~1564
    38 Tanno H, Nockels RP, Pitts LH, et al. Immunolocalization of heat shock protein after fluid percussive brain injury and relationship to breakdown of the blood-brain barrier. J Cereb Blood Flow Metab, 1993, 13(1):116~124
    39 Jenkins LW, Moszynski K, Lyeth BG, et al. Increased vulnerability of the mildly traumatized rat brain to cerebral ischemia: the use of controlled secondary ischemia as a research tool to identify common or different mechanisms contributing to mechanical and ischemic brain injury. Brain Res, 1989, 477(1-2):211~224
    40 Przyklenk K, Bauer B, Ovize M, et al. Regional ischemic 'preconditioning' protects remote virgin myocardium from subsequent sustained coronary occlusion. Circulation, 1993, 87(3):893~899
    41 McClanahan TB, Nao BS, Wolke LJ, et al. Brief renal occlusion and reperfusion induces myocardial infarct size in rabbits(Abstract). FASEB J, 1993, 7: A118
    42 Gho BC, Schoemaker RG, van den Doel MA, et al. Myocardial protection by brief ischemia in noncardiac tissue. Circulation, 1996, 94(9):2193~2200
    43 Takaoka A, Nakae I, Mitsunami K, et al. Renal ischemia/ reperfusion remotely improves myocardial energy metabolism during myocardial ischemia via adenosine receptors in rabbits: effects of ”remote preconditioning”. J Am Coll Cardiol, 1999, 33(2):556~564
    44 Tang ZL, Dai W, Li YJ, et al. Involvement of capsaicin-sensitive sensory nerves in early and delayed cardioprotection induced by a brief ischaemia of the small intestine. Naunyn Schmiedebergs Arch Pharmacol, 1999, 359(3):243~247
    45 Schoemaker RG, van Heijningen CL. Bradykinin mediates cardiac preconditioning at a distance. Am J Physiol, 2000, 278 (2):H1571~H1576
    46 Kharbanda RK, Mortensen UM, White PA, et al. Transient limb ischemia induces remote ischemic preconditioning in vivo. Circulation,2002,106(23):2881~2883
    47 赵红岗,李文斌,刘惠卿,等. 肢体缺血预处理减轻大鼠海马缺血/再灌注损伤. 中国应用生理学杂志, 2004,20(1):50~53
    48 Zhao Hong-Gang, Li Wen-Bin, Li Qing-Jun, et al. Limb ischemic preconditioning attenuates apoptosis of pyramidal neurons in the CA1 hippocampus induced by cerebral ischemia-reperfusion in rats. Acta Physiol Sin,2004, 3
    49 周爱民, 李清君, 陈晓玲, 等. 脑预缺血引起大鼠海马细胞膜腺苷受体数量和亲和力升高及其对神经元的保护作用. 生理学报, 2001, 53(4):265~269
    50 Zhou AM, Li WB, Li QJ, et al. A short cerebral ischemic preconditioning up-regulates adenosine receptors in the hippocampal CA1 region of rats. Neurosci Res, 2004, 48(4):397~404
    51 Reshef A, Sperling O, Zoref-Shani E. Opening of ATP-sensitive potassium channels by cromakalim confers tolerance against chemical ischemia in rat neuronal cultures. Neurosci Lett, 1998, 250(2):111~114
    52 Reshef A, Sperling O, Zoref-Shani E. Opening of K(ATP) channels is mandatory for acquisition of ischemic tolerance by adenosine. Neuroreport, 2000, 11(3):463~465
    53 刘惠卿, 李文斌, 冯荣芳, 等. 一氧化氮合酶抑制剂 L-NAME 对大鼠脑缺血耐受诱导的影响.生理学报, 2003, 55(2):219~224
    54 Atochin DN, Clark J, Demchenko IT, et al. Rapid cerebral ischemic preconditioning in mice deficient in endothelial and neuronal nitric oxide synthases. Stroke, 2003,34(5):1299~1303
    55 Kuntscher MV, Kastell T, Altmann J, et al. Acute remote ischemic preconditioning II: the role of nitric oxide. Microsurgery, 2002,22(6):227~231
    56 Kurkinen K, Busto R, Goldsteins G, et al. Isoform-specific membrane translocation of protein kinase C after ischemic preconditioning. Neurochem Res, 2001, 26(10):1139~1144
    57 Gu Z, Jiang Q, Zhang G, et al. Diphosphorylation of extracellularsignal-regulated kinases and c-Jun N-terminal protein kinases in brain ischemic tolerance in rat. Brain Res, 2000, 860(1-2):157~160
    58 Furuta S, Ohta S, Hatakeyama T, et al. Recovery of protein synthesis in tolerance-induced hippocampal CA1 neurons after transient forebrain ischemia.Acta Neuropathol (Berl), 1993, 86(4):329~336
    59 Gabai VL, Sherman MY. Invited review: Interplay between molecular chaperones and signaling pathways in survival of heat shock. J Appl Physiol. 2002 Apr;92(4):1743~1748
    60 Perdrizet GA, Lena CJ, Shapiro DS, et al. Preoperative stress conditioning prevents paralysis after experimental aortic surgery: increased heat shock protein content is associated with ischemic tolerance of the spinal cord. J Thorac Cardiovasc Surg, 2002, 124(1):162~70
    61 Lepore DA, Knight KR, Anderson RL, et al. Role of priming stresses and Hsp70 in protection from ischemia-reperfusioninjury in cardiac and skeletal muscle. Cell Stress Chaperones, 2001, 6(2):93~96
    62 Hampton CR, Shimamoto A, Rothnie CL, et al. HSP70.1 and HSP70.3 are required for late-phase protection induced by ischemic preconditioning of mouse hearts. Am J Physiol Heart Circ Physiol, 2003, 285(2):H866~874
    63 Nishi S, Taki W, Uemura Y, et al. Ischemic tolerance due to the induction of HSP70 in a rat ischemic recirculation model. Brain Res, 1993, 615(2):281~288
    64 Nakata N, Kato H, Kogure K. Inhibition of ischemic tolerance in the gerbil hippocampus by quercetin and anti-heat shock protein-70 antibody. Neuroreport, 1993, 4(6):695~698
    65 Mori T, Muramatsu H, Matsui T, et al. Possible role of the superoxide anion in the development of neuronal tolerance following ischemic preconditioning in rats. Neuropathol Appl Neurobiol, 2000,26(1):31~44
    66 Currie RW, Ellison JA, White RF, et al. Benign focal ischemic preconditioning induces neuronal Hsp70 and prolonged astrogliosis with expression of Hsp27. Brain Res, 2000, 863(1-2):169~181
    67 Ohtsuki T, Ruetzler CA, Tasaki K, et al. Interleukin-1 mediates inductionof tolerance to global ischemia in gerbil hippocampal CA1 neurons. J Cereb Blood Flow Metab, 1996,16(6):1137~1142
    68 Nawashiro H, Tasaki K, Ruetzler CA, et al. TNF-alpha pretreatment induces protective effects against focal cerebral ischemia in mice. J Cereb Blood Flow Metab, 1997,17(5):483~490
    69 Barone FC, White RF, Spera PA, et al. Ischemic preconditioning and brain tolerance: temporal histological and functional outcomes, protein synthesis requirement, and interleukin-1 receptor antagonist and early gene expression. Stroke, 1998, 29(9):1937~1950; discussion 1950~1951
    70 Ginis I, Jaiswal R, Klimanis D, et al. TNF-alpha-induced tolerance to ischemic injury involves differential control of NF-kappa B transactivation: J Cereb Blood Flow Metab, 2002, 22(2):142~152
    71 Nawashiro H, Tasaki K, Ruetzler CA, et al. TNF-alpha pretreatment induces protective effects against focal cerebral ischemia in mice. J Cereb Blood Flow Metab, 1997,17(5):483~490
    72 Ohtsuki T, Matsumoto M, Kuwabara K, et al. Influence of oxidative stress on induced tolerance to ischemia in gerbil hippocampal neurons. Brain Res, 1992, 599(2):246~252
    73 Toyoda T, Kassell NF, Lee KS. Induction of ischemic tolerance and antioxidant activity by brief focal ischemia. Neuroreport, 1997 ,8(4):847~851
    74 Mitchell K, Kariko K, Harris VA, et al. Preconditioning with cortical spreading depression does not upregulate Cu/Zn-SOD or Mn-SOD in the cerebral cortex of rats. Brain Res Mol Brain Res, 2001,96(1-2):50~58
    75 Sugawara T, Noshita N, Lewen A, et al. Neuronal expression of the DNA repair protein Ku 70 after ischemic preconditioning corresponds to tolerance to global cerebral ischemia. Stroke, 2001, 32(10):2388~2393
    76 Stephenson DT, Schober DA, Smalstig EB, et al. Peripheral benzodiazepine receptors are colocalized with activated microglia following transient global forebrain ischemia in the rat. J Neurosci, 1995, 15(7 Pt 2):5263~5274
    77 Petito CK, Halaby IA. Relationship between ischemia and ischemic neuronal necrosis to astrocyte expression of glial fibrillary acidic protein. Int J Dev Neurosci, 1993,11(2):239~247
    78 Yu AC, Lee YL, Fu WY,et al. Gene expression in astrocytes during and after ischemia. Prog Brain Res, 1995, 105:245~253
    79 雷万龙, 袁群芳, 姚志彬. 局灶性脑缺血区星形胶质细胞可塑性变化的实验研究. 神经解剖学杂志, 2000, 16(2):123~126
    80 O’Shea RD. Roles and regulation of glutamate transporters in the central nervous system. Clin Exp Pharmacol Physiol, 2002, 29(11): 1018~1023
    81 Min Zhang, Wen-Bin Li, Jin-Xia Geng, et al.The upregulation of glial glutamate transporter-1participates in the induction of brain ischemic tolerance in rats. J Cereb Blood Flow Metab, 2007 Jan 17; Epub ahead of print
    82 Zhong LT, Sarafian T, Kane DJ, et al. bcl-2 inhibits death of central neural cells induced by multiple agents. Proc Natl Acad Sci U S A, 1993, 90(10):4533~4537
    83 Linnik MD, Zahos P, Geschwind MD, et al. Expression of bcl-2 from a defective herpes simplex virus-1 vector limits neuronal death in focal cerebral ischemia. Stroke, 1995, 26(9):1670~1674
    84 Zhang S, Wang W. Altered expression of bcl-2 mRNA and Bax in hippocampus with focal cerebral ischemia model in rats. Chin Med J (Engl), 1999, 112(7):608~611
    85 Kato H, Liu Y, Araki T, Kogure K. Temporal profile of the effects of pretreatment with brief cerebral ischemia on the neuronal damage following secondary ischemic insult in the gerbil:cumulative damage and protective effects. Brain Res, 1991, 12;553(2):238~242
    86 Nakata N, Kato H, Kogure K. Ischemic tolerance and extracellular amino acid concentrations in gerbil hippocampus measured by intracerebral microdialysis. Brain Res Bull, 1994, 35(3):247~151
    87 Sommer C, Gass P, Kiessling M. Selective c-JUN expression in CA1 neurons of the gerbil hippocampus during and after acquisition of anischemia-tolerant state. Brain Pathol, 1995,5(2):135~144
    88 Yoneda Y, Kuramoto N, Azuma Y, et al. Possible involvement of activator protein-1 DNA binding in mechanisms underlying ischemic tolerance in the CA1 subfield of gerbil hippocampus. Neuroscience, 1998,86(1):79~97
    89 Herrera DG, Robertson HA. Activation of c-fos in the brain. Prog Neurobiol, 1996, 50(2-3):83~107
    90 Tomasevic G, Shamloo M, Israeli D, et al. Activation of p53 and its target genes p21(WAF1/Cip1) and PAG608/Wig-1 in ischemic preconditioning. Brain Res Mol Brain Res, 1999, 70(2):304~313
    91 Shimazaki K, Nakamura T, Nakamura K, et al. Reduced calcium elevation in hippocampal CA1 neurons of ischemia-tolerant gerbils. Neuroreport, 1998, 9(8):1875~1878
    92 Sapolsky RM. Cellualr defenses agsinst excitotoxic insults. J Neurochem, 2001, 76(6):1601~1611
    93 Gong L, Gao TM, Li X, et al. Enhancement in activities of large conductance calcium-activated potassium channels in CA1 pyramidal neurons of rat hippocampus after transient forebrain ischemia. Brain Res, 2000, 884(1-2):147~154
    94 Huang H, Gao TM, Gong L, et al. Potassium channel blocker TEA prevents CA1 hippocampal injury following transient forebrain ischemia in adult rats. Neurosci Lett, 2001, 305(2):83~86

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700