用户名: 密码: 验证码:
大鼠局灶性脑缺血后继发性脑损伤机制及替米沙坦脑保护作用的研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
脑血管病是造成中国居民死亡的第一原因和成年年人残疾的第一原因,以缺血性脑血管病占80%。研究表明,脑缺血触发一系列复杂的病理级联反应,包括能量代谢异常、炎症反应、氧化应激、自由基生成、兴奋性氨基酸的毒性作用等,直接或者间接导致神经细胞凋亡/死亡,破坏血脑屏障形成脑水肿,并最终导致神经功能缺失。在脑缺血后复杂的病理损伤机制中,炎症反应是继发性脑损伤的最重要原因之一。尽管目前对缺血性脑损伤机制有了比较深入的了解,但仍缺乏有效的治疗手段。除了极少数脑缺血患者在超早期经过溶栓治疗取得满意效果外,绝大多数患者将面临残疾甚至死亡的厄运。探讨有效阻止缺血性脑损伤机制,保护神经细胞的方法,是目前神经科学研究的热点和难点。
     尽管已经有多种药物经过实验研究,证明具有抗炎、抗氧化、抗凋亡等作用,但多数未能在临床得到广泛应用,使理论和实践大大脱节。替米沙坦是一种临床广泛应用的血管紧张素受体阻断剂类(angiotensin type1receptor blocker,ARB)降血压药物。近年来,动物和临床实验发现,替米沙坦不仅能够有效治疗高血压、保护靶器官和减轻心室重构,还能够对脑缺血具有良好的抗炎作用,能够显著减轻脑梗死体积、脑水肿和继发性脑损伤,其机制多与激活过氧化物酶体增殖物激活受体γ(peroxisome prolixferator-activated receptors, PPARy)有关。国内外多项研究证明,PPARγ是一个关键性核转录因子,与其激动剂或者配体结合后活化,具有抗炎、抗氧化、抗凋亡、神经保护等多重作用。替米沙坦是唯一一种具有活化PPARγ功能的ARB类药物。结合基础研究和临床应用的实际需要,替米沙坦可能是治疗缺血性脑血管病的主要药物之一,但替米沙坦的作用机制仍需深入研究。
     应用线栓法建立大鼠永久性局灶脑缺血模型,分别对炎症反应代表性因子PPARγ、 NF-κB、Egr-1和MMP-9,神经保护因子PEDF,和以Claudin-5为代表的血脑屏损伤程度等方面进行动态观察,并应用PPARy激动剂替米沙坦和PPARy特异性拮抗剂GW9662进行干预,观察上述各因子的变化,以及评价神经功能缺失、脑水肿和梗死体积等。以PPARy为切入点,深入探讨脑缺血后脑损伤机制及替米沙坦的脑保护作用机制。
     本研究将分三部分对上述内容进行探讨,各部分内容概述如下。
     第一部分大鼠脑缺血后炎症反应及替米沙坦抗炎作用机制研究
     目的:动态观察PPARγ、NF-κB、Egr-1和MMP-9等在大鼠脑缺血后的表达变化,探讨替米沙坦的抗炎作用机制。
     方法:应用Longa等的线栓法建立MCAO大鼠模型,采用成年健康雄性Sprague-Dawley大鼠,体重230g-250g。实验1:将大鼠随机分为正常组(Normal)和手术组(MCAO),手术组按照MCAO术后不同时间点分为3h、6h、24h、48h、72h5个亚组。实验2:将大鼠随机分为假手术组(Sham)、溶剂对照组(Vehicle)、替米沙坦组(Telmisartan)及替米沙坦+GW9662组(Telmisartan+GW9662)。替米沙坦30mg/kg于MCAO手术前1h以灌胃方式给予,以防止大鼠麻醉时呕吐;假手术组和对照组给予等体积的生理盐水;替米沙坦+GW9662组于MCAO手术前1h以灌胃方式给予30mg/kg替米沙坦,术后立即腹腔注射4mg/kgGW9662。将各组大鼠分别于相应时间点进行神经功能评分后断头处死,采用免疫组化、western blot和qR-T PCR来观察脑缺血后PPARγ、NF-κB、Egr-1和MMP-9等:mRNA和蛋白水平的动态变化,TTC染色评价脑梗死体积,干湿重法评价患侧脑水肿。
     结果:MCAO组大鼠脑缺血后自24h开始,出现明显的神经功能缺失,持续并进行性加重至所观察到的72h,与正常组比较差异有统计学意义(P<0.05)。同时,自脑缺血24h开始,PPARy表达呈明显减少趋势,而NF-κB.Egr-1、MMP-9等呈明显增加趋势,且持续至本研究所观察到的72h,与正常组比较,差异有统计学意义(P<0.05)。术后24h,替米沙坦干预组与溶剂对照组比较,神经功能缺失有明显改善,脑水肿明显减轻、梗死体积明显减小,保护性因子PPARγ等表达明显增加,而NF-κB、Egr-1、MMP-9等明显减少,差异有统计学意义(P<0.05)。替米沙坦+GW9662组与替米沙坦组比较,神经功能缺失有明显加重、脑水肿和梗死体积增加,并且PPARy表达明显减少,而NF-κB、Egr-1、MMP-9等明显增加,差异有统计学意义(P<0.05)。溶剂对照组和替米沙坦+GW9662组比较无差别。
     结论:大鼠脑缺血早期炎症反应迅速出现,脑缺血后24h开始明显出现,并持续至所观察的72h,炎症反应程度与神经功能缺损程度具有相关性。替米沙坦单独和联合GW9662干预表明,PPARγ是炎症反应的关键性调控基因。替米沙坦通过激活PPARγ,下调NF-кB、Egr-1和MMP-9,显著抑制了炎症反应,减轻脑缺血体积和脑水肿,改善了神经功能缺失。
     第二部分大鼠脑缺血后PEDF表达变化及替米沙坦调节机制的研究
     目的:动态观察大鼠脑缺血后PEDF的表达变化,探讨替米沙坦的脑保护作用机制。方法:应用Longa等的线栓法建立MCAO大鼠模型,采用成年健康雄性Sprague-Dawley大鼠,体重230g~250g。实验1:将大鼠随机分为正常组(Normal)和手术组(MCAO),手术组按照MCAO术后不同时间点分为3h、6h、24h、48h、72h5个亚组。实验2:将大鼠随机分为假手术组(Sham)、溶剂对照组(Vehicle)、替米沙坦组(Telmisartan)和替米沙坦+GW9662组(Telmisartan+GW9662)。替米沙坦30mg/kg于MCAO手术前1h以灌胃方式给予,以防止大鼠麻醉时呕吐;假手术组和对照组给予等体积的生理盐水;替米沙坦+GW9662组于MCAO手术前1h以灌胃方式给予30mg/kg替米沙坦,术后立即腹腔注射4mg/kgGW9662。将各组大鼠分别于相应时间点进行神经功能评分后断头处死,采用免疫组化、western blot和qR-T PCR来观察脑缺血后PEDF的mRNA和蛋白水平动态变化,TTC染色评价脑梗死体积,干湿重法评价患侧脑水肿。
     结果:MCAO组大鼠脑缺血后自24h开始,出现明显的神经功能缺失,持续并进行性加重至所观察到的72h,与正常组比较差异有统计学意义(P<0.05)。脑缺血自24h开始,PEDF表达呈明显减少趋势,持续至本研究所观察到的72h,与正常组比较,差异有统计学意义(P<0.05)。术后24h,替米沙坦干预组与对照组比较,神经功能缺失有明显改善,脑水肿明显减轻、梗死体积明显减小,神经保护因子PEDF表达明显增加,差异有统计学意义(P<0.05)。替米沙坦+GW9662组与替米沙坦组比较,神经功能缺失有明显加重、脑水肿和梗死体积增加,并且PEDF表达明显减少,差异有统计学意义(P<0.05)。溶剂对照组和替米沙坦+GW9662组比较无差别。
     结论:首次证明局灶性脑缺血早期,神经保护因子PEDF开始显著减少,可能是神经功能缺失、脑水肿等缺血性脑损伤的原因之一。首次通过大鼠脑缺血模型证明,核转录因子PPARy是PEDF的上游调控基因,上调PEDF是是替米沙坦脑保护机制之一。替米沙坦干预通过激活PPARγ上调PEDF的表达,明显改善了脑缺血的神经功能缺失、脑水肿,减小了梗死体积。
     第三部分大鼠脑缺血后血脑屏障通透性变化及替米沙坦血脑屏障保护作用的研究
     目的:动态观察大鼠脑缺血后血脑屏障通透性变化,评价替米沙坦对血脑屏障的保护作用及机制。
     方法:应用Longa等的线栓法建立MCAO大鼠模型,采用成年健康雄性Sprague-Dawley大鼠,体重230g-250g。实验1:将大鼠随机分为正常组(Normal)和手术组(MCAO),手术组按照MCAO术后不同时间点分为3h、6h、24h、48h、72h5个亚组。实验2:将大鼠随机分为假手术组(Sham)、溶剂对照组(Vehicle)、替米沙坦组(Telmisartan)和替米沙坦+GW9662组(Telmisartan+GW9662).替米沙坦30mg/kg于MCAO手术前1h以灌胃方式给予,以防止大鼠麻醉时呕吐;假手术组和对照组给予等体积的生理盐水;替米沙坦+GW9662组于MCAO手术前1h以灌胃方式给予30mg/kg替米沙坦,术后立即腹腔注射4mg/kgGW9662。血脑屏障通透性通过检测血管内皮细胞紧密连接蛋白成分Claudin-5进行评价。将各组大鼠分别于相应时间点进行神经功能评分后断头处死,采用免疫组化、western blot和qR-T PCR来观察脑缺血后Claudin-5以及MMP-9的mRNA和蛋白水平动态变化,TTC染色评价脑梗死体积,干湿重法评价患侧脑水肿。
     结果:MCAO组大鼠脑缺血后自24h开始,出现明显的神经功能缺失,持续并进行性加重至所观察到的72h,与正常组比较差异有统计学意义(P<0.05)。脑缺血自24h开始,Cludin-5表达呈明显减少趋势,持续至本研究所观察到的72h,与正常组比较,差异有统计学意义;此时,MMP-9表达显著升高(P<0.05)。术后24h,替米沙坦干预组与对照组比较,神经功能缺失有明显改善,脑水肿明显减轻、梗死体积明显减小,Cludin-5表达明显增加,MMP-9表达减少,差异有统计学意义(P<0.05)。替米沙坦+GW9662组与替米沙坦组比较,神经功能缺失有明显加重、脑水肿和梗死体积增加,Cludin-5表达明显减少,差异有统计学意义(P<0.05)。溶剂对照组和替米沙坦+GW9662组比较无差别。
     结论:局灶性脑缺血早期,血脑屏障通透性显著增加是导致明显的神经功能缺失、脑水肿形成的直接原因。替米沙坦干预通过激活PPARγ、抑制MMP-9、上调Cludin-5的表达,保护了血脑屏障的完整性,因此改善了脑缺血的神经功能缺失、脑水肿,减小了梗死体积。
Stroke is the second commonest cause of death and the leading cause of adult disability worldwide and ischemic stroke accounts for about80%.
     Studies showed that cerebral ischemia triggered massive physiopathologic processes including energy metabolism, inflammatory responses, oxidative stress, increased release of excitatory amion acids, etc., which direct/indirect resulting in blood-brain barrier disruption and brain edema formation, as well as neurological deficit. Among those inflammatory responses is one of the most important mechanisms. Nowadays, beyond only a restricted number of hospitalized patients profiting from thrombolytic therapy, the vast majority of patients suffering from stroke resulted in disability even death owing to the limited therapies. Delaying the development of brain damage and protecting neuron are remained.
     There a large number of agents have been proved to display anti-inflammation, anti-oxidation and anti-apoptosis properties but never employed in treating cerebral ischemia. Telmisartan, an angiotensin type1receptor blocker (ARB), is a classical medicine controlling hypertention and protecting target organs. It's well established that telmisartan alleviated the infarct volume owing to anti-inflammation property by activating peroxisome prolixferator-activated receptors (PPARy). PPARy, activated by ligands, is a key transcription factor possessing multiple protective roles. So on the basis. telmisartan, the unique ARB as agonist of PPARy, attracted more attention for cerebral ischemia treatment.
     Male Sprague-Dawley rats were subjected to permanent middle cerebral artery occlusion (pMCAO). The time course expression of PEDF, PPARy, NF-κB, Egr-1, MMP-9, PEDF and Claudin-5were evaluated. Telmisartan (PPARγ agonist) and GW9662(PPARy antagonist) were systemic administrated to explore the effect on PPARγ, PEDF, NF-κB, Egr-1, MMP-9, PEDF and Claudin-5expression at24h after cerebral ischemia by immunohistochemistry, western blot, qRT-PCR. The neurological deficits, brain water content and infarct volume were measured.
     The study was divided into three part list as below.
     Part I The inflammatory responses after focal cerebral ischemia in rats and the anti-inflammatory role of telmisartan
     Object:This study is to evaluate the time course expression of PPARy, NF-κB, Egr-1and MMP-9, then explore the regulation mechanisms of telmisartan on them.
     Methods:Male Sprague-Dawley rats,230g~250g weight, were subjected to permanent middle cerebral artery occlusion (pMCAO). Experiment1:SD rats were randomly divided into normal,3h,6h,24h,48h and72h groups after operation. Experiment2:SD rats were randomly divided into4groups:sham-operated group, Vehicle group, Telmisartan group, and Telmisartan+GW9662group. Telmisartan group received telmisartan (Boehringer Ingelheim, Ingelheim, Germany) by feeding tubes at30mg/kg dissolved in PBS1hour before MCAO for avoiding emesis and then once daily thereafter. Vehicle groups received equal volume PBS in the same manner. Telmisartan+GW9662group received telmisartan before operation and GW9662(Sigma-Aldrich, USA) after MCAO immediately injected intraperitoneally at4mg/kg dissolved in3%DMSO diluted in PBS. Neurological deficit scores were evaluated by an examiner blinded to the experimental groups at respective time point. After that, rats were anesthetized with10%chloral hydrate and the brains were quickly removed. The time course expression of PPARy, NF-κB, Egr-1and MMP-9were evaluated by western blot and quantitative real-time PCR. The underling regulation mechanisms at24h were explored by administration of telmisartan and GW9662by immunohistochemistry, western blot and quantitative real-time PCR. Infarct volume was determined by2,3,5-triphenyltetrazolium chloride (TTC) and brain water content was measured using the standard wet-dry method at24h in the4groups.
     Results:Compared with normal group, the neurological deficits were obviously exacerbated from24h to72h after MCAO in our study (P<0.05). At the same time, PPARy expression was obviously decreased, but NF-κB, Egr-1and MMP-9expression were increased, relative to those in normal group(P<0.05). Compared with the vehicle group, the neurological deficits, brain water content and infarct volume were dramatically alleviated by telmisartan at24h after MCAO (P<0.05), and the decrease of PPARy was significantly up-regulated and the increase of Egr-1, NF-κB and MMP-9was down-regulated by telmisartan at24h (P<0.05). Telmisartan's profits were all reversed by GW9662co-administration(P<0.05). No significant differences were observed between Vehicle group and Telmisartan+GW9662group animals (P>0.05).
     Conclusions:At the early stage of focal cerebral ischemia, the inflammatory responses were triggered and responsible for the neurological deficits. PPARy was the key regular controlling Egr-1, NF-kB and MMP-9expression. Telmisartan down-regulated Egr-1, NF-κB and MMP-9by PPARy signaling, alleviated the neurological deficits, brain water content and infarct volume.
     Part Ⅱ The intrinsic PEDF expression after focal cerebral ischemia in rats and the regulation of telmisartan
     Object:This study is to evaluate the time course expression of PEDF and explore the underling regulation mechanisms of telmisartan.
     Methods:Male Sprague-Dawley rats,230g^250g weight, were subjected to permanent middle cerebral artery occlusion (pMCAO). Experiment1:SD rats were randomly divided into normal,3h,6h,24h,48h and72h groups after operation. Experiment2:SD rats were randomly divided into4groups:sham-operated group, Vehicle group, Telmisartan group, and Telmisartan+GW9662group. Telmisartan group received telmisartan (Boehringer Ingelheim, Ingelheim, Germany) by feeding tubes at30mg/kg dissolved in PBS1hour before MCAO for avoiding emesis and then once daily thereafter. Vehicle groups received equal volume PBS in the same manner. Telmisartan+GW9662group received telmisartan before operation and GW9662(Sigma-Aldrich, USA) after MCAO immediately injected intraperitoneally at4mg/kg dissolved in3%DMSO diluted in PBS. Neurological deficit scores were evaluated by an examiner blinded to the experimental groups at respective time point. After that, rats were anesthetized with10%chloral hydrate and the brains were quickly removed. The time course expression of PEDF was evaluated by western blot and quantitative real-time PCR. The underling regulation mechanisms at24h were explored by administration of telmisartan and GW9662by immunohistochemistry, western blot and quantitative real-time PCR. Infarct volume was determined by2,3,5-triphenyltetrazolium chloride (TTC) and brain water content was measured using the standard wet-dry method at24h in the4groups.
     Results:Compared with normal group, the neurological deficits were obviously exacerbated from24h to72h after MCAO in our study (P0.05). At the same time, the intrinsic PEDF expression was obviously decreased, relative to that in normal group (P<0.05). Compared with the vehicle group, the neurological deficits, brain water content and infarct volume were dramatically alleviated by telmisartan at24h after MCAO (P<0.05), and the decrease of the intrinsic PEDF was significantly up-regulated by telmisartan at24h (P<0.05). Telmisartan's profits were all reversed by GW9662co-administration (P<0.05). No significant differences were observed between Vehicle group and Telmisartan+GW9662group animals (P>0.05).
     Conclusions:It's the first report that the intrinsic PEDF were reduced at the early stage of focal cerebral ischemia, which was probably responsible for the neurological deficits and brain edema. PPARγ was the key regular controlling the intrinsic PEDF expression. Telmisartan normalizing the intrinsic PEDF by PPARy signal could contribute to the alleviated infarct volume, neurological deficits and brain edema in the study.
     Part Ⅲ The permeability of blood-brain barrier after focal cerebral ischemia in rats and the protection of telmisartan
     Object:This study is to evaluate the time course expression of Claudin-5and MMP-9and explore the underling regulation mechanisms of telmisartan.
     Methods:Male Sprague-Dawley rats,230g-250g weight, were subjected to permanent middle cerebral artery occlusion (pMCAO). Experiment1:SD rats were randomly divided into normal,3h,6h,24h,48h and72h groups after operation. Experiment2:SD rats were randomly divided into4groups:sham-operated group, Vehicle group, Telmisartan group, and Telmisartan+GW9662group. Telmisartan group received telmisartan (Boehringer Ingelheim, Ingelheim, Germany) by feeding tubes at30mg/kg dissolved in PBS1hour before MCAO for avoiding emesis and then once daily thereafter. Vehicle groups received equal volume PBS in the same manner. Telmisartan+GW9662group received telmisartan before operation and GW9662(Sigma-Aldrich, USA) after MCAO immediately injected intraperitoneally at4mg/kg dissolved in3%DMSO diluted in PBS. Neurological deficit scores were evaluated by an examiner blinded to the experimental groups at respective time point. After that, rats were anesthetized with10%chloral hydrate and the brains were quickly removed. The time course expression of Claudin-5and MMP-9were evaluated by western blot and quantitative real-time PCR. The underling regulation mechanisms at24h were explored by administration of telmisartan and GW9662by immunohistochemistry, western blot and quantitative real-time PCR. Infarct volume was determined by2,3,5-triphenyltetrazolium chloride (TTC) and brain water content was measured using the standard wet-dry method at24h in the4groups.
     Results:Compared with normal group, the neurological deficits were obviously exacerbated from24h to72h after MCAO in our study(P<0.05). At the same time, the Claudin-5and MMP-9expression was obviously decreased, relative to that in normal group (P<0.05). Compared with the vehicle group, the neurological deficits, brain water content and infarct volume were dramatically alleviated by telmisartan at24h after MCAO (P<0.05), and the decrease of the intrinsic PEDF was significantly up-regulated by telmisartan at24h(P<0.05). Telmisartan's profits were all reversed by GW9662co-administration (P<0.05). No significant differences were observed between Vehicle group and Telmisartan+GW9662group animals (P>0.05).
     Conclusions:The blood-brain barrier was dramatically disrupted at the early stage of focal cerebral ischemia, which was responsible for the brain edema. PPARy was the key factor regulating Claudin-5and protecting BBB integrity. Telmisartan normalizing Claudin-5by inhibiting MMP-9through PPARγ signal could contribute to the alleviated infarct volume, neurological deficits and brain edema in the study.
引文
1 Toni D, Chamorro A, Kaste M, et al. Acute treatment of ischaemic stroke. European Stroke Initiative. Cerebrovasc Dis,2004,17:30-46
    2 Kappelle LJ, Van Der, Worp HB. Treatment or prevention of complications of acute ischemic stroke. Curr Neurol Neurosci Rep.2004, 4:36-41
    3 Wu JS, Cheung WM, Tsai YS, et al. Ligand-activated peroxisome proliferator-activated receptor-gamma protects against ischemic cerebral infarction and neuronal apoptosis by 14-3-3 epsilon upregulation. Circulation,2009,119:1124-34
    4 Wang CX, Ding X, Noor R, et al. Rosiglitazone alone or in combination with tissue plasminogen activator improves ischemic brain injury in an embolic model in rats. J Cereb Blood Flow Metab,2009,29(10): 1683-1694
    5 Longa EZ, Weinstein PR, Carlson S, et al. Reversible middle cerebral artery occlusion without craniectomy in rats, Stroke,1989,20:84-91
    6 Liu Y, Zhang XJ, Yang CH, et al. Oxymatrine protects rat brains against permanent focal ischemia and down-regulates NF-kappaB expression. Brain Res,2009,1268:174-180
    7 Yang C, Zhang X, Fan H. et al. Curcumin up-regulates transcription factor Nrf2, HO-1 expression and protects rat brains against focal ischemia. Brain Res,2009,1282:133-141
    8 Tureyen K, Brooks N, Bowen K, et al. Transcription factor early growth response-1 induction mediates inflammatory gene expression and brain damage following transient focal ischemia. J Neurochem,2008,105: 1313-1324
    9 Ma J, Ren Z, Ma Y, Xu L, et al. Targeted knockdown of EGR-1 inhibits IL-8 production and IL-8-mediated invasion of prostate cancer cells through suppressing EGR-1/NF-kappaB synergy. J Biol Chem,2009,284: 34600-34606
    10 Yan S F, Fujita T, Lu J, et al. Egr-1, a master switch coordinating upregulation of divergent gene families underlying ischemic stress. Nature Med,2000,6:1355-1361
    11 Nurmi A, Lindsberg PJ, Koistinaho M, et al. Nuclear factor-kappaB contributes to infarction after permanent focal ischemia. Stroke,2004,35: 987-991
    12 Shin SY, Kim JH, Baker A, et al. Transcription factor Egr-1 is essential for maximal matrix metalloproteinase-9 transcription by tumor necrosis factor alpha. Mol Cancer Res,2010,8:507-519
    13 Wu KI, Schmid-Schonbein GW. Nuclear factor kappa B and matrix metalloproteinase induced receptor cleavage in the spontaneously hypertensive rat. Hypertension.2011,57:261-268
    14 Dong X, Song YN, Liu WG, et al. Mmp-9, a potential target for cerebral ischemic treatment. Curr Neuropharmacol,2009,7:269-275
    15 Hu Q, Chen C, Khatibi NH, et al. Lentivirus-mediated transfer of MMP-9 shRNA provides neuroprotection following focal ischemic brain injury in rats. Brain Res,2011,1367:347-359
    16 Haraguchi T, Iwasaki K, Takasaki K, et al. Telmisartan, a partial agonist of peroxisome proliferator-activated receptor gamma, improves impairment of spatial memory and hippocampal apoptosis in rats treated with repeated cerebral ischemia. Brain Res,2010,1353:125-132
    17 Kobayashi T, Kawamata T, Shibata N, et al. Angiotensin II type 1 receptor blocker telmisartan reduces cerebral infarct volume and peri-infarct cytosolic phospholipase A(2) level in experimental stroke. J Neurotrauma, 2009,26:2355-2364
    18 Thiel G, Rossler OG, Immediate-early transcriptional response to angiotensin Ⅱ in human adrenocortical cells. Endocrinology,2011,152: 4211-4223
    19 Kim S, Kawamura M, Wanibuchi H, et al. Angiotensin II type 1 receptor blockade inhibits the expression of immediate-early genes and fibronectin in rat injured artery. Circulation,1995,92:88-95
    20 Hoetzel A, Dolinay T, Vallbracht S, et al. Carbon monoxide protects against ventilator-induced lung injury via PPAR-gamma and inhibition of Egr-1. Am J Respir Crit Care Med,2008,177:1223-1232
    21 Zhang HL, Xu M, Wei C, et al. Neuroprotective effects of pioglitazone in a rat model of permanent focal cerebral ischemia are associated with peroxisome proliferator-activated receptor gamma-mediated suppression of nuclear factor-κB signaling pathway. Neuroscience,2011,176:381-395
    22 Nagai N, Izumi-Nagai K, Oike Y, et al. Suppression of diabetes-induced retinal inflammation by blocking the angiotensin Ⅱ type 1 receptor or its downstream nuclear factor-kappaB pathway. Invest Ophthalmol Vis Sci, 2007,48:4342-4350
    23 Lv B, Wang H, Tang Y, et al. High-mobility group box 1 protein induces tissue factor expression in vascular endothelial cells via activation of NF-kappaB and Egr-1. Thromb Haemost,2009,102:352-359
    24 Blessing E, Preusch M, Kranzhofer R, et al. Anti-atherosclerotic properties of telmisartan in advanced atherosclerotic lesions in apolipoprotein E deficient mice. Atherosclerosis,2008,199:295-303
    25 Gashler A, Sukhatme VP. Early growth response protein 1 (Egr-1): prototype of a zinc-finger family of transcription factors. Prog Nucleic Acid Res Mol Biol,1995,50:191-224
    26 Shin SY. Kim JH, Baker A. et al. Transcription factor Egr-1 is essential for maximal matrix metalloproteinase-9 transcription by tumor necrosis factor alpha. Mol Cancer Res,2010,8:507-519
    27 Okada M, Harada T, Kikuzuki R, et al. Effects of telmisartan on right ventricular remodeling induced by monocrotaline in rats. J Pharmacol Sci, 2009,111:193-200
    28 Malek R, Borowicz KK, Jargiello M, et al. Role of NF-κB in the central nervous system. Pharmacol,2007,59:25-33
    29 Fujita T, Asai T, Andrassy M, et al. PKC beta regulates ischemia/reperfusion injury in the lung. J Clin Invest,2004,113: 1615-1623
    30 Xu Y, Toure F, Qu W, et al. Advanced glycation end product (AGE)-receptor for AGE (RAGE) signaling and up-regulation of Egr-1 in hypoxic macrophages. J Biol Chem,2010,285:23233-23240
    31张祥建,李春岩,李海燕,等.实验性大鼠脑梗死后脑水肿规律及行为学改变.脑与神经疾病杂志,2005,13:26-28
    32 Longa EZ,Weinstein PR,Carlson S, et al. Reversible middle cerebral artery occlusion without craniectomy in rats. Stroke,1989,20:84-91
    33 Tatlisumak T, Carano RA, Takano K, et al. A novel endothelin antagonist, A-127722, attenuates ischemic lesion size in rats with temporary middle cerebral 24 artery occlusion:a diffusion and perfusion MRI study. Stroke, 1998,29:850-857
    34 Yi JH, Park SW, Kapadia R, et al. Role of transcription factors in mediating post-ischemic cerebral inflammation and braindamage. Neurochem Int,2007,50(7-8):1014-1027
    35 Liu M, Wu B, Wang WZ, et al. Stroke in China:epidemiology, prevention, and management strategies. Lancet Neurol,2007,6:456-464
    36 Lee SR, Kim HY, Hong JS, et al. PPARgamma agonist pioglitazone reduces matrix metalloproteinase-9 activity and neuronal damage after focal cerebral ischemia. Biochem Biophys Res Commun,2009,380: 17-21
    37 Sanagi T, Yabe T, Yamada H. Gene transfer of PEDF attenuates ischemic brain damage in the rat middle cerebral artery occlusion model. J Neurochem,2008,106:1841-1854
    38 Ho TC, Chen SL, Yang YC. PEDF induces p53-mediated apoptosis through PPAR gamma signaling in human umbilical vein endothelial cells. Cardiovasc Res,2007,76:213-223
    39 Ho TC, Yang YC, Chen SL, et al. Pigment epithelium-derived factor induces THP-1 macrophage apoptosis and necrosis by the induction of the peroxisome proliferator-activated receptor gamma. Mol Immunol,2007, 45:898-909
    40 Ho TC, Chen SL, Shih SC, et al. Pigment epithelium-derived factor (PEDF) promotes tumor cell death by inducing macrophage membrane tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). J Biol Chem,2011,286:35943-35954
    41 Ho TC, Chen SL, Shih SC, et al. Pigment epithelium-derived factor is an intrinsic antifibrosis factor targeting hepatic stellate cells. Am J Pathol, 2010,177:1798-1811
    42 Wang SH, Liang CJ, Wu JC, et al. Pigment epithelium-derived factor reduces the PDGF-induced migration and proliferation of human aortic smooth muscle cells through PPARy activation. Int J Biochem Cell Biol, 2012,44:280-289
    43 Lange J, Yafai Y, Reichenbach A, et al. Regulation of pigment epithelium-derived factor production and release by retinal glial (Muller) cells under hypoxia. Invest Ophthalmol Vis Sci,2008,49:5161-5167
    44 Notari L, Miller A, Martinez A, et al. Pigment epithelium-derived factor is a substrate for matrix metalloproteinase type 2 and type 9:implications for down-regulation in hypoxia. Invest. Ophthalmol Vis Sci,2005,46: 2736-2747
    45 Jiao H, Wang Z, Liu Y, et al. Specific role of tight junction proteins claudin-5, occludin, and ZO-1 of the blood-brain barrier in a focal cerebral ischemic insult. J Mol Neurosci,2011,44:130-139
    46 Martins T, Baptista S, Goncalves J, et al. Methamphetamine transiently increases the blood-brain barrier permeability in the hippocampus:role of tight junction proteins and matrix metalloproteinase-9. Brain Res,2011, 1411:28-40
    47 Zhao X, Strong R, Zhang J, et al. Neuronal PPARgamma deficiency increases susceptibility to brain damage after cerebral ischemia. J Neurosci,2009,29:6186-6195
    48 Schock SC, Xu J, Duquette PM, et al. Rescue of neurons from ischemic injury by peroxisome proliferatoractivated receptor-gamma requires a novel essential cofactor LMO4. J Neurosci,2008,28:12433-12444
    49 Barnes PJ, Adcock IM, Transcription factors and asthma. Eur Respir J, 1998,12:221-234 (review)
    50 Vemuganti R, Tureyen K, Brooks N, et al. Transcription factor NGFI-A/EGR1 induction mediates postischemic inflammatory gene expression and brain damage in rodents. Stroke,2007,38:556
    1 Auriel E, Bornstein NM. Neuroprotection in acute ischemic stroke-current status. J Cell Mol Med,2010,14(9):2200-2202
    2 Chavez JC, Hurko O, Barone FC, et al. Pharmacologic interventions for stroke:looking beyond the thrombolysis time window into the penumbra with biomarkers, not a stopwatch. Stroke,2009,40(10):e558-563
    3 Toth G,Albers GW. Use of MRI to estimate the therapeutic window in acute stroke:is perfusion-weighted imaging/diffusion-weighted imaging mismatch an EPITHET for salvageable ischemic brain tissue? Stroke, 2009,40(1):333-335
    4 Bilak MM, Corse AM, Bilak SR, et al. Pigment epithelium-derived factor (PEDF) protects motor neurons from chronic glutamate-mediated neurodegeneration. J Neuropathol Exp Neurol,1999,58(7):719-728
    5 Sanagi T, Yabe T, Yamada H. Gene transfer of PEDF attenuates ischemic brain damage in the rat middle cerebral artery occlusion model. J Neurochem,2008,106(4):1841-1854
    6 DeCoster MA, Schabelman E, Tombran-Tink J, et al. Neuroprotection by pigment epithelial-derived factor against glutamate toxicity in developing primary hippocampal neurons. J Neurosci Res,1999,56(6):604-610
    7 Falk T, Zhang S, Sherman SJ. Pigment epithelium derived factor (PEDF) is neuroprotective in two in vitro models of Parkinson's disease. Neurosci Lett,2009,458(2):49-52
    8 Taniwaki T, Becerra SP, Chader GJ, et al. Pigment epithelium-derived factor is a survival factor for cerebellar granule cells in culture. J Neurochem,1995,64(6):2509-2517
    9 Houenou LJ, D'Costa AP, Li L, et al. Pigment epithelium-derived factor promotes the survival and differentiation of developing spinal motor neurons. J Comp Neurol,1999,412:506-514
    10 Takita H, Yoneya S, Gehlbach PL, et al. Retinal neuroprotection against ischemic injury mediated by intraocular gene transfer of pigment epithelium-derived factor. Invest Ophthalmol Vis Sci,2003,44: 4497-4504
    11 Drevon CA. Fatty acids and expression of adipokines. Biochim Biophys Acta,2005,1740(2):287-292
    12 Sawant S, Aparicio S, Tink AR, et al. Regulation of factors controlling angiogenesis in liver development:a role for PEDF in the formation and maintenance of normal vasculature. Biochem Biophys Res Commun,2004, 325(2):408-413
    13 Quan GM, Ojaimi J, Li Y, et al. Localization of pigment epithelium-derived factor in growing mouse bone. Calcif Tissue Int,2005, 76(2):146-153
    14 Tombran-Tink J, Mazuruk K, Rodriguez IR, et al. Organization, evolutionary conservation, expression and unusual Alu density of the human gene for pigment epithelium-derived factor, a unique neurotrophic serpin. Mol Vis,1996,2:11
    15 Ho TC, Chen SL, Yang YC, et al. PEDF induces p53-mediated apoptosis through PPAR gamma signaling in human umbilical vein endothelial cells. Cardiovasc Res,2007,76:213-223
    16 Ho TC, Yang YC, Chen SL, et al. Pigment epithelium-derived factor induces THP-1 macrophage apoptosis and necrosis by the induction of the peroxisome proliferator-activated receptor gamma. Mol Immunol,2007, 45:898-909
    17 Ho TC, Chen SL, Shih SC, et al. Pigment epithelium-derived factor (PEDF) promotes tumor cell death by inducing macrophage membrane tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). J Biol Chem,2011,286:35943-35954
    18 Ho TC, Chen SL, Shih SC, et al. Pigment epithelium-derived factor is an intrinsic antifibrosis factor targeting hepatic stellate cells. Am J Pathol, 2010,177:1798-1811
    19 Wang SH, Liang CJ, Wu JC, et al. Pigment epithelium-derived factor reduces the PDGF-induced migration and proliferation of human aortic smooth muscle cells through PPARy activation. Int J Biochem Cell Biol, 2012,44:280-289
    20 Xu X, Zhang SS, Barnstable CJ, et al. Molecular phylogeny of the antiangiogenic and neurotrophic serpin, pigment epithelium derived factor in vertebrates. BMC Genomics,2006,7:248
    21 Alberdi E, Aymerich MS, Becerra SP. Binding of pigment epithelium-derived factor (PEDF) to retinoblastoma cells and cerebellar granule neurons. Evidence for a PEDF receptor. J Biol Chem,1999,274: 31605-31612
    22 Aymerich MS, Alberdi EM, Martinez A, et al. Evidence for pigment epithelium-derived factor receptors in the neural retina. Invest Ophthalmol Vis Sci,2001,42:3287-3293
    23 Ogata N, Wang L, Jo N, et al. Pigment epithelium derived factor as a neuroprotective agent against ischemic retinal injury. Curr Eye Res,2001; 22:245-252
    24 Takita H, Yoneya S, Gehlbach PL, et al. Invest Ophthalmol Vis Sci.2003 Oct;44(10):4497-504. Retinal neuroprotection against ischemic injury mediated by intraocular gene transfer of pigment epithelium-derived factor
    25 Notari L, Miller A, Martinez A, et al. Pigment epithelium-derived factor is a substrate for matrix metalloproteinase type 2 and type 9:implications for downregulation in hypoxia. Invest Ophthalmol Vis Sci,2005,46(8): 2736-2747
    26 Dong X, Song YN, Liu WG, et al. Mmp-9, a potential target for cerebral ischemic treatment. Curr Neuropharmacol,2009,7:269-275
    27 Hu Q, Chen C, Khatibi NH, et al. Lentivirus-mediated transfer of MMP-9 shRNA provides neuroprotection following focal ischemic brain injury in rats. Brain Res,2011,1367:347-359
    28 Yabe T, Wilson D, Schwartz JP. NF-κB activation is required for the neuroprotective effects of pigment epithelium-derived factor (PEDF) on cerebellar granule neurons. J Biol Chem,2001; 276:43313-43319
    29 Pang IH, Zeng H, Fleenor DL, et al. Pigment epithelium-derived factor protects retinal ganglion cells. BMC Neurosci.2007,8:11
    30 Yan S F, Fujita T, Lu J, et al. Egr-1, a master switch coordinating upregulation of divergent gene families underlying ischemic stress. Nature Med,2000,6:1355-1361
    31 Nurmi A, Lindsberg PJ, Koistinaho M, et al. Nuclear factor-kappaB contributes to infarction after permanent focal ischemia. Stroke,2004,35: 987-991
    32 Zhang HL, Xu M, Wei C, et al. Neuroprotective effects of pioglitazone in a rat model of permanent focal cerebral ischemia are associated with peroxisome proliferator-activated receptor gamma-mediated suppression of nuclear factor-KB signaling pathway. Neuroscience,2011,176:381-395
    33 Lv B, Wang H, Tang Y, et al. High-mobility group box 1 protein induces tissue factor expression in vascular endothelial cells via activation of NF-kappaB and Egr-1. Thromb Haemost,2009,102:352-359
    34 Blessing E, Preusch M, Kranzhofer R, et al. Anti-atherosclerotic properties of telmisartan in advanced atherosclerotic lesions in apolipoprotein E deficient mice. Atherosclerosis,2008,199:295-303
    35 Okada M, Harada T, Kikuzuki R, et al. Effects of telmisartan on right ventricular remodeling induced by monocrotaline in rats. J Pharmacol Sci, 2009,111:193-200
    36 Lange J, Yafai Y, Reichenbach A, et al. Regulation of pigment epithelium-derived factor production and release by retinal glial (Muller) cells under hypoxia. Invest Ophthalmol Vis Sci,2008,49:5161-5167
    37 Chung C, Shugrue C, Nagar A, et al. Ethanol exposure depletes hepatic pigment epithelium-derived factor, a novel lipid regulator. Gastroenterology,2009,136,331-340
    1 Nitta T, Hata M, Gotoh S, Seo Y, et al. Size-selective loosening of the blood-brain barrier in claudin-5-deficient mice. J Cell Biol,2003,161(3): 653-660
    2 Wang Z, Leng Y, Tsai LK, et al. Valproic acid attenuates blood-brain barrier disruption in a rat model of transient focal cerebral ischemia:the roles of HDAC and MMP-9 inhibition. J Cereb Blood Flow Metab,2011, 31(1):52-57
    3 Jiao H, Wang Z, Liu Y, et al. Specific role of tight junction proteins claudin-5, occludin, and ZO-1 of the blood-brain barrier in a focal cerebral ischemic insult. J Mol Neurosci,2011,44(2):130-139
    4 Martins T, Baptista S, Goncalves J, et al. Methamphetamine transiently increases the blood-brain barrier permeability in the hippocampus:role of tight junction proteins and matrix metal]oproteinase-9. Brain Res,2011, 1411:28-40
    5 Dong X, Song YN, Liu WG, et al. Mmp-9, a potential target for cerebral ischemic treatment. Curr Neuropharmacol,2009,7:269-275
    6 Wang CX, Ding X, Noor R, et al. Rosiglitazone alone or in combination with tissue plasminogen activator improves ischemic brain injury in an embolic model in rats. J Cereb Blood Flow Metab,2009,29(10): 1683-1694
    7 Lee SR, Kim HY, Hong JS, et al. PPARgamma agonist pioglitazone reduces matrix metalloproteinase-9 activity and neuronal damage after focal cerebral ischemia. Biochem Biophys Res Commun,2009,380: 17-21
    8 Blessing E, Preusch M, Kranzhofer R, et al. Anti-atherosclerotic properties of telmisartan in advanced atherosclerotic lesions in apolipoprotein E deficient mice. Atherosclerosis,2008,199:295-303
    9 Wu JS, Cheung WM, Tsai YS, et al. Ligand-activated peroxisome proliferator-activated receptor-gamma protects against ischemic cerebral infarction and neuronal apoptosis by 14-3-3 epsilon upregulation. Circulation,2009,119:1124-34
    10 Furuse M, Hirase T, Itoh M, et al. Occludin:a novel integral membrane protein localizing at tight junctions. J Cell Biol,1993,123:1777-1788
    11 Furuse M, Fujita K, Hiiragi T, et al. Claudin-1 and-2:novel integral membrane proteins localizing at tight junctions with no sequence similarity to occludin. J Cell Biol,1998a,141; 1539-1550
    12 Martin-Padura I, Lostaglio S, Schneemann M, et al. Junctional adhesion molecule, a novel member of the immunoglobulin superfamily that distributes at intercellular junctions and modulates monocyte transmigration. J Cell Biol,1998,142; 117-127
    13 Tsukita S, Furuse M, Itoh M. Multi-functional strands in tight junctions. Nat Rev Mol Cell Biol,2001,2:285-293
    14 Morita K, Sasaki H, Furuse M, et al. Endothelial claudin: claudin-5/TMVCF constitutes tight junction strands in endothelial cells. J Cell Biol,1999b,147; 185-194
    15 Maejima Y, Okada H, Haraguchi G, et al. Telmisartan, a unique ARB, improves left ventricular remodeling of infracted heart by activating PPAR gamma. Lab Invest,2011,91:932-944
    16 Ruhul Amin AR, Senea T. Oo ML, et al. Secretion of matrix metalloproteinase-9 by the proinflammatory cytokine, IL-1 beta:a role for the dual signalling pathways, Akt and Erk. Genes Cells,2003,8:515-523
    17 Magnoni S, Baker A, Thomson S, et al. Neuroprotective effect of adenoviral-mediated gene transfer of TIMP-1 and-2 in ischemic brain injury. Gene Ther,2007,14:621-625
    18 Pfefferkorn T, Rosenberg GA. Closure of the blood-brain barrier by matrix metalloproteinase inhibition reduces rtPAmediated mortality in cerebral ischemia with delayed reperfusion. Stroke,2003,34:2025-2030
    19 Rosenberg GA, Estrada EY, Dencoff JE. Matrix metalloproteinases and TIMPs are associated with blood-brain barrier opening after reperfusion in rat brain. Stroke,1998,29:2189-2195
    20 Wu JS, Cheung WM, Tsai YS, et al. Ligand-activated peroxisome proliferator-activated receptor-gamma protects against ischemic cerebral infarction and neuronal apoptosis by 14-3-3 epsilon upregulation. Circulation,2009,119:1124-34
    21 Wang CX, Ding X, Noor R, et al. Rosiglitazone alone or in combination with tissue plasminogen activator improves ischemic brain injury in an embolic model in rats. J Cereb Blood Flow Metab,2009,29(10): 1683-1694
    22 Haraguchi T, Iwasaki K, Takasaki K, et al. Telmisartan, a partial agonist of peroxisome proliferator-activated receptor gamma, improves impairment of spatial memory and hippocampal apoptosis in rats treated with repeated cerebral ischemia. Brain Res,2010,1353:125-132
    23 Kobayashi T, Kawamata T, Shibata N, et al. Angiotensin Ⅱ type 1 receptor blocker telmisartan reduces cerebral infarct volume and peri-infarct cytosolic phospholipase A(2) level in experimental stroke. J Neurotrauma, 2009,26:2355-2364
    24 Maejima Y, Okada H, Haraguchi G, et al. Telmisartan, a unique ARB, improves left ventricular remodeling of infracted heart by activating PPAR gamma. Lab Invest,2011,91:932-944
    25 Honda M, Nakagawa S, Hayashi K, et al. Adrenomedullin improves the blood-brain barrier function through the expression of claudin-5. Cell Mol Neurobiol,2006,26:109-118
    26 Gidday JM, Gasche YG, Copin JC, et al. Leukocyte-derived matrix metalloproteinase-9 mediates blood-brain barrier breakdown and is proinflammatory after transient focal cerebral ischemia. Am J Physiol Heart Circ Physiol,2005,289:H558-H568
    27 Chakraborti S, Mandal M, Das S, et al. Regulation of matrix metalloproteinases:An overview. Mol Cell Biochem,2003,253:269-285
    28 Ellerbroek SM, Halbleib JM, Benavidez M, et al. Phosphatidylinositol 3-kinase activity in epidermal growth factor-stimulated matrix metalloproteinase-9 production and cell surface association. Cancer Res, 2001,61:1855-1861
    29 Liu JF, Crepin M, Liu JM, et al. FGF-2 and TPA induce matrix metalloproteinase-9 secretion in MCF-7 cells through PKC activation of the Ras/ERK pathway. Biochem Bioph Res Commun,2002,293: 1174-1182
    30 Ruhul Amin AR, Senga T, Oo ML, et al. Secretion of matrix metalloproteinase-9 by the proinflammatory cytokine, IL-1 beta:a role for the dual signalling pathways, Akt and Erk. Genes Cells.2003,8:515-523.
    31 Suyama K, Shapiro I, Guttman M, et al. A signaling pathway leading to metastasis is controlled by N-cadherin and the FGF receptor. Cancer Cell, 2002,2:301-314
    1 Nuclear Receptors Nomenclature Committee. A unified nomenclature system for the nuclear receptor superfamily. Cell,1999,97:161-163
    2 Issemann I, Green S. Activation of a member of the steroid hormone receptor superfamily by peroxisome proliferators. Nature,1990,347: 645-650
    3 Laudet V, Hanni C, Coll J, et al. volution of the nuclear receptor gene superfamily. EMBO J,1992,11:1003-1013
    4 Collino M, Patel NS, Thiemermann C. PPARs as new therapeutic targets for the treatment of cerebral ischemia/reperfusion injury. Ther Adv Cardiovasc Dis,2008,2:179-197
    5 Dreyer C, Krey G, Keller H, et al. Control of the peroxisomal beta-oxidation pathway by a novel family of nuclear hormone receptors. Cell,1992,68:879-887
    6 Chandra V, Huang P, Hamuro Y, et al. Structure of the intact PPAR-y-RXR-a nuclear receptor complex on DNA. Nature,2008,456: 350-356
    7 Willson TM, Lambert MH, Kliewer SA. Peroxisome proliferator-activated receptor gamma and metabolic disease. Annu Rev Biochem,2001,70: 341-367
    8 Escher P, Wahli W. Peroxisome proliferator-activated receptors:insight into multiple cellular functions. Mutat Res,2000,448:121-138
    9 Chang TH, Szabo E. Induction of differentiation and apoptosis by ligands of peroxisome proliferator-activated receptor-g in non-small cell lung cancer. Cancer Res,2000,60:1129-1138
    10 Bradley MN, Zhou L, Smale ST. C/EBPb regulation in lipopolysaccharide-stimulated macrophages. Mol Cell Biol,2003,23: 4841-4858
    11 Blaschke F, Takata Y, Caglayan E, et al. Obesity, peroxisome proliferator-activated receptor, and atherosclerosis in type 2 diabetes. Arterioscler Thromb Vasc Biol,2006,26:28-40
    12 Berger JP, Akiyama TE, Meinke PT. PPARs:therapeutic targets for metabolic disease. Trends Pharmacol Sci,2005,26:244-251
    13 Verges B. Clinical interest of PPARs ligands. Diabetes,2004,30:7-12
    14 Kersten S, Desvergne B, Wahli W. Roles of PPARs in health and disease. Nature,2000,405:421-424
    15 Forman BM, Tontonoz P, Chen J, et al.15-Deoxy-delta 12, 14-prostaglandin J2 is a ligand for the adipocyte determination factor PPAR gamma. Cell,1995,83:803-812
    16 Kliewer SA, Lenhard JM, Willson TM, et al. prostaglandin J2 metabolite binds peroxisome proliferator-activated receptor gamma and promotes adipocyte differentiation. Cell,1995,83:813-819
    17 Gilroy DW, Colville-Nash PR, Willis D, et al. Inducible cyclooxygenase may have anti-inflammatory properties. Nature Med,1999,5:698-701
    18 Ricote M, Li AC, Willson TM, et al. The peroxisome proliferator-activated receptor-gamma is a negative regulator of macrophage activation. Nature, 1998,391:79-82
    19 Theocharis S, Margeli A, Kouraklis G. Peroxisome proliferators activated receptor-gamma ligands as potent antineoplastic agents. Curr Med Chem Anticancer Agents,2003,3:239-251
    20 Willson TM, Brown PJ, Strenbach DD, et al. The PPARs:from orphan receptors to drug discovery, J Med Chem,2000,43:527-550
    21 Margeli A, Kouraklis G, Theocharis S. Peroxisome proliferators activated receptor-gamma (PPAR-gamma) ligands and angiogenesis. Angiogenesis, 2003,6:165-169
    22 Theocharis S, Margeli A, Vielh P, et al. Peroxisome proliferatoractivated receptor-gamma ligands as cell-cycle modulators. Cancer Treat Rev,2004, 30:545-554
    23 Pershadsingh HA. Dual Peroxisome Proliferator-Activated Receptor-alpha/gamma Agonists:In the Treatment of Type 2 Diabetes Mellilus and the Metabolic Syndrome. Treat Endocrinol.2006,5:89-99
    24 Haraguchi T, Iwasaki K, Takasaki K, et al. Telmisartan, a partial agonist of peroxisome proliferator-activated receptor gamma, improves impairment of spatial memory and hippocampal apoptosis in rats treated with repeated cerebral ischemia. Brain Res,2010,1353:125-132
    25 Zhao X, Strong R, Zhang J, et al. Neuronal PPARgamma deficiency increases susceptibility to brain damage after cerebral ischemia. J Neurosci,2009,29:6186-6195
    26 Schock SC, Xu J, Duquette PM, et al. Rescue of neurons from ischemic injury by peroxisome proliferatoractivated receptor-gamma requires a novel essential cofactor LMO4. J Neurosci,2008,28:12433-12444
    27 Wu JS, Cheung WM, Tsai YS, et al. Ligand-activated peroxisome proliferatoractivated receptor-gamma protects against ischemic cerebral infarction and neuronal apoptosis by 14-3-3 epsilon upregulation. Circulation,2009,119:1124-1134
    28 Youle RJ, Strasser A. The Bcl-2 protein family:opposing activities that mediate cell death. Nat Rev Mol Cell Biol,2008,9:47-59
    29 Hengartner MO. The biochemistry of apoptosis. Nature,2000,407: 770-776
    30 Kerr JF, Wyllie AH, Currie AR. Apoptosis:a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer,1972,26: 239-257
    31 Fuenzalida K, Quintanilla R, Ramos P, et al. Peroxisome proliferator-activated receptor gamma up-regulates the Bcl-2 anti-apoptotic protein in neurons and induces mitochondrial stabilization and protection against oxidative stress and apoptosis. J Biol Chem,2007, 282:37006-37015
    32 Wu JS, Lin TN, Wu KK. Rosiglitazone and PPAR-gamma overexpression protect mitochondrial membrane potential and prevent apoptosis by upregulating anti-apoptotic Bcl-2 family proteins. J Cell Physiol,2009, 220:58-71
    33 Iadecola C, Alexander M. Cerebral ischemia and inflammation. Curr Opin Neurol,2001,14:89-94 (review)
    34 Iadecola C, Salkowski CA, Zhang F, et al. The transcription factor interferon regulatory factor 1 is expressed after cerebral ischemia and contributes to ischemic brain injury. J Exp Med,1999,189:719-727
    35 Graham SH, Hickey RW. Cyclooxygenases in central nervous system. A special role of cyclooxygenase 2 in neuronal cell death. Arch Neurol,2003, 60:628-630
    36 Mattson MP, Culmsee C, Yu ZF. Apoptotic and antiapoptotic mechanisms in stroke. Cell Tissue Res,2000,301:173-187
    37 Allahtavakoli M, Shabanzadeh A, Roohbakhsh A, et al. Combination therapy of rosiglitazone, a peroxisome proliferator-activated receptor-gamma ligand, and NMDA receptor antagonist (MK-801) on experimental embolic stroke in rats. Basic Clin Pharmacol Toxicol,2007, 101:309-314
    38 Allahtavakoli M, Shabanzadeh AP, Sadr SS, et al. Rosiglitazone, a peroxisome proliferator-activated receptor-gamma ligand, reduces infarction volume and neurological deficits in an embolic model of stroke, Clin Exp Pharmacol Physiol,2006,33:1052-1058
    39 Collino M, Aragno M, Mastrocola R, et al. Modulation of the oxidative stress and inflammatory response by PPAR-γ agonists in the hippocampus of rats exposed to cerebral ischemia/reperfusion. Eur J Pharmacol,2006b, 530:70-80
    40 Shimazu T, Inoue I, Araki N, et al. A peroxisome proliferator-activated receptor-γ agonist reduces infarct size in transient but not in permanent ischemia. Stroke,2005,36:353-359
    41 Sundararajan S, Gamboa JL, Victor NA, et al. Peroxisome proliferator-activated receptor-γ ligands reduce inflammation and infarction size in transient focal ischemia. Neuroscience,2005,130: 685-696
    42 Ou X, Zhao LA, Labiche R, et al. Neuronal expression of peroxisome proliferator-activated receptor-gamma (PPARgamma) and 15d-prostaglandin J2-mediated protection of brain after experimental cerebral ischemia in rat. Brain Res,2006,1096:196-203
    43 ZhaoY, Patzer A, Gohlke P, et al. The intracerebral application of the PPARy-ligand pioglitazone confers neuroprotection against focal ischemia in the rat brain. Eur J Neurosci,2005,22:278-282
    44 Zhao Y, Patzer A, Herdegen T, et al. Activation of cerebral peroxisome proliferator-activated receptors gamma promotes neuroprotection by attenuation of neuronal cyclooxygenase-2 overexpression after focal cerebral ischemia in rats. FASEB J,2006b,20:1162-1175
    45 Pascual G, Fong AL, Ogawa S, et al. A SUMOylationdependent pathway mediates transrepression of inflammatory response genes by PPAR-gamma. Nature,2005,437:759-763
    46 Lee J, Reding M. Effects of thiazolidinediones on stroke recovery:a case-matched controlled study. Neurochem Res,2006,32(4-5):635-638
    47 Forman BM, Tontonoz P, Chen J, et al.15-Deoxy-delta 12, 14-prostaglandin J2 is a ligand for the adipocyte determination factor PPAR gamma. Cell,1995,83:803-812
    48 Powell WS.15-deoxy-delta -PGJ2:endogenous PPARgamma ligand or minor eicosanoid degradation product? J Clin Invest,2003,112:828-830
    49 Heneka MT, Klockgether T, Feinstein DL. Peroxisome proliferators-activated receptor-gamma ligands reduce neuronal inducible nitric oxide synthase expression and cell death in vivo. J Neurosci,2000, 20:6862-6867
    50 Luo Y, Yin W, Signore AP, et al. Neuroprotection against focal ischemic brain injury by the peroxisome proliferator-activated receptor-gamma agonist rosiglitazone. J Neurochem,2006,97:435-448
    51 Tureyen K, Kapadia R, Bowen K, et al. Peroxisome proliferators-actiyated receptor-g agonists induce neuroprotection following transient focal ischemia in normotensive, normoglycemic as well as hypertensive and type-2 diabetic rodents. J Neurochem,2007,101:41-56
    52 Aoun P, Watson DG, Simpkins JW. Neuroprotective effects of PPARgamma agonists against oxidative insults in HT-22 cells. Eur J Pharmacol,2003,472:65-71
    53 Colca JR, McDonald WG, Waldon D J, et al. Identification of a novel mitochondrial protein ("mitoNEET") cross-linked specifically by a thiazolidinedione photoprobe. Am J Physiol Endocrinol Metab,2004,286: E252-E260
    54 Collino M, Aragno M, Mastrocola R, et al. Oxidative stress and inflammatory response evoked by transient cerebral ischemia/reperfusion: effects of the PPAR-alpha agonist WY14643. Free Radical Biol Metab, 2006a,41:579-589
    55 Lin TN, Cheung W M, Wu JS, et al.15d-prostaglandin J2 protects brain from ischemia-reperfusion injury. Arterioscler ThrombVasc Biol,2006,26: 481-487
    56 Pereira MP, Hurtado O, Cardenas A, et al. The nonthiazolidinedione PPARy agonist L-796,449 is neuroprotective in experimental stroke. J Neuropathol Exp Neurol,2005,64:797-805
    57 Romera C, Hurtado O, Mallolas J, et al. Ischemic preconditioning reveals that GLT1/EAAT2 glutamate transporter is a novel PPARgamma target gene involved in neurpprotection. J Cereb Blood Flow Metab,2007,27: 1327-1338
    58 Chu K, Lee ST. Koo JS, et al. Peroxisome proliferator-activated receptor-g agonist, rosiglitazone, promotes angiogenesis after focal cerebral ischemia. Brain Res,2006,1093:208-218
    59 Dormandy JA, Charbonnel B, Eckland DJ, et al. (2005) Secondary prevention of macrovascular events in patients with type 2 diabetes in the PROactive Study (PROspective pioglitAzone Clinical Trial In macro Vascular Events):a randomised controlled trial. Lancet,2005,366: 1279-1289
    60 Thiel G, Rossler OG, Immediate-early transcriptional response to angiotensin II in human adrenocortical cells. Endocrinology,2011,152: 4211-4223
    61 Kim S, Kawamura M, Wanibuchi H, et al. Angiotensin Ⅱ type 1 receptor blockade inhibits the expression of immediate-early genes and fibronectin in rat injured artery. Circulation,1995,92:88-95
    62 Hoetzel A, Dolinay T, Vallbracht S, et al. Carbon monoxide protects against ventilator-induced lung injury via PPAR-gamma and inhibition of Egr-1. Am. J. Respir. Crit Care Med,2008,177:1223-1232
    1 Tombran-Tink J, Johnson LV. Neuronal differentiation of retinoblastoma cells induced by medium conditioned by human RPE cells. Invest Ophthalmol Vis Sci,1989,30:1700-1707
    2 Becerra SP, Sagasti A, Spinella P, et al. Pigment epithelium-derived factor behaves like a noninhibitory serpin. Neurotrophic activity does not require the serpin reactive loop. J Biol Chem,1995; 270(43):25992-25999
    3 Xu X, Zhang SS, Barnstable CJ, et al. Molecular phylogeny of the antiangiogenic and neurotrophic serpin, pigment epithelium derived factor in vertebrates. BMC Genomics,2006,7:248
    4 Tombran-Tink J, Mazuruk K, Rodriguez IR, et al. Organization, evolutionary conservation, expression and unusual Alu density of the human gene for pigment epithelium-derived factor, a unique neurotrophic serpin. Mol Vis,1996,2:11
    5 Bilak MM, Corse AM, Bilak SR, et al. Pigment epithelium-derived factor (PEDF) protects motor neurons from chronic glutamate-mediaied neurodegeneration. J Neuropathol Exp Neurol,1999,58(7):719-728
    6 Drevon CA. Fatty acids and expression of adipokines. Biochim Biophys Acta,2005,1740(2):287-292
    7 Sawant S, Aparicio S, Tink AR, et al. Regulation of factors controlling angiogenesis in liver development:a role for PEDF in the formation and maintenance of normal vasculature. Biochem Biophys Res Commun,2004, 325(2):408-413
    8 Quan GM, Ojaimi J, Li Y, et al. Localization of pigment epithelium-derived factor in growing mouse bone. Calcif Tissue Int,2005, 76(2):146-153
    9 Sanagi T, Yabe T, Yamada H. Changes in pigment epithelium-derived factor expression following kainic acid induced cerebellar lesion in rat. Neurosci Lett,2007,424(1):66-71
    10 Sanagi T, Yabe T, Yamada H. Gene transfer of PEDF attenuates ischemic brain damage in the rat middle cerebral artery occlusion model. J Neurochem,2008,106(4):1841-1854
    11 DeCoster MA, Schabelman E, Tombran-Tink J, et al. Neuroprotection by pigment epithelial-derived factor against glutamate toxicity in developing primary hippocampal neurons. J Neurosci Res,1999,56(6):604-610.
    12 Falk T, Zhang S, Sherman SJ. Pigment epithelium derived factor (PEDF) is neuroprotective in two in vitro models of Parkinson's disease. Neurosci Lett,2009,458(2):49-52
    13 Taniwaki T, Becerra SP, Chader GJ, et al. Pigment epithelium-derived factor is a survival factor for cerebellar granule cells in culture. J Neurochem,1995,64(6):2509-2517
    14 Cao W, Tombran-Tink J, Elias R, et al. In vivo protection of photoreceptors from light damage by pigment epithelium-derived factor. Invest Ophthalmol Vis Sci,2001,42(7):1646-52
    15 Tombran-Tink J, Chader GG, Johnson LV. PEDF:a pigment epithelium-derived factor with potent neuronal differentiative activity. Exp Eye Res,1991,53(3):411-414
    16 Alberdi E, Aymerich MS, Becerra SP. Binding of pigment epithelium-derived factor (PEDF) to retinoblastoma cells and cerebellar granule neurons. Evidence for a PEDF receptor. J Biol Chem,1999,274: 31605-31612
    17 Aymerich MS, Alberdi EM, Martinez A, et al. Evidence for pigment epithelium-derived factor receptors in the neural retina. Invest Ophthalmol Vis Sci,2001,42:3287-3293
    18 Bernard A, Gao-Li J, Franco CA, et al. Laminin receptor involvement in the anti-angiogenic activity of pigment epithelium-derived factor. J Biol Chem,2009,284(16):10480-10490
    19 Notari L, Baladron V, Aroca-Aguilar JD, et al. Identification of a lipase-linked cell membrane receptor for pigment epithelium-derived factor. J Biol Chem,2006,281(49):38022-38037
    20 Yabe T, Wilson D, Schwartz JP. NF-κB activation is required for the neuroprotective effects of pigment epithelium-derived factor (PEDF) on cerebellar granule neurons. J Biol Chem,2001,276:43313-43319
    21 Volpert OV, Zaichuk T, Zhou W, et al. Inducer-stimulated Fas targets activated endothelium for destruction by anti-angiogenic thrombospondin-1 and pigment epithelium derived factor. Nature Med, 2002,8:349-357
    22 Ho TC, Chen SL, Yang YC, et al. PEDF induces p53-mediated apoptosis through PPAR gamma signaling in human umbilical vein endothelial cells. Cardiovasc Res,2007,76:213-223
    23 Ho TC, Yang YC, Chen SL, et al. Pigment epithelium-derived factor induces THP-1 macrophage apoptosis and necrosis by the induction of the peroxisome proliferator-activated receptor gamma. Mol Immunol,2007, 45:898-909
    24 Ho TC, Chen SL, Shih SC, et al. Pigment epithelium-derived factor (PEDF) promotes tumor cell death by inducing macrophage membrane tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). J Biol Chem,2011,286:35943-35954
    25 Ho TC, Chen SL, Shih SC, et al. Pigment epithelium-derived factor is an intrinsic antifibrosis factor targeting hepatic stellate cells. Am J Pathol, 2010,177:1798-1811
    26 Wang SH, Liang CJ, Wu JC, et al. Pigment epithelium-derived factor reduces the PDGF-induced migration and proliferation of human aortic smooth muscle cells through PPARy activation. Int J Biochem Cell Biol, 2012,44:280-289
    27 Araki T, Taniwaki T, Becerra SP, et al. Pigment epithelium-derived factor (PEDF) differentially protects immature but not mature cerebellar granule cells against apoptotic cell death. J Neurosci Res,1998,53:7-15
    28 Houenou LJ, D'Costa AP, Li L, et al. Pigment epithelium-derived factor promotes the survival and differentiation of developing spinal motor neurons. J Comp Neurol,1999,412:506-514
    29 Takita H, Yoneya S, Gehlbach PL, et al. Retinal neuroprotection against ischemic injury mediated by intraocular gene transfer of pigment epithelium-derived factor. Invest Ophthalmol Vis Sci, 2003,44: 4497.4504
    30 Li H, Tran VV, Hu Y, et al. A PEDF N-terminal peptide protects the retina from ischemic injury when delivered in PLGA nanospheres. Exp Eye Res, 2006,83(4):824-833
    31 Cayouette M, Smith SB, Becerra SP, et al. Pigment epithelium-derived factor delays the death of photoreceptors in mouse models of inherited retinal degenerations. Neurobiol Dis,1999,6:523-532
    32 Ogata N, Wang L, Jo N, et al. Pigment epithelium derived factor as a neuroprotective agent against ischemic retinal injury. Curr Eye Res,2001, 22:245-252
    33 Bilak MM, Becerra SP, Vincent AM, et al. Identification of the neuroprotective molecular region of pigment epithelium-derived factor and its binding sites on motor neurons. J Neurosci,2002,22:9378-9386
    34 Yabe T, Wilson D, Schwartz JP. NFkappaB activation is required for the neuroprotective effects of pigment epithelium-derived factor (PEDF) on cerebellar granule neurons. J Biol Chem,2001,276(46):43313-43319
    35 Pang IH, Zeng H, Fleenor DL, et al. Pigment epithelium-derived factor protects retinal ganglion cells. BMC Neurosci,2007,8:11
    36 Yasuda T, Fukuda-Tani M, Nihira T, et al. Correlation between levels of pigment epithelium-derived factor and vascular endothelial growth factor in the striatum of patients with Parkinson's disease. Exp Neurol,2007, 206(2):308-317
    37 Kuncl RW, Bilak MM, Bilak SR, et al. Pigment epithelium-derived factor is elevated in CSF of patients with amyotrophic lateral sclerosis. J Neurochem,2002,81(1):178-184
    38 Yamagishi S, Inagaki Y, Takeuchi M, et al. Is pigment epithelium-derived factor level in cerebrospinal fluid a promising biomarker for early diagnosis of Alzheimer's disease? Med Hypotheses,2004,63(1):115-117
    39 Castano EM, Roher AE, Esh CL, et al. Comparative proteomics of cerebrospinal fluid in neuropathologically-confirmed Alzheimer's disease and non-demented elderly subjects. Neurol Res,2006,28(2):155-163
    40 Davidsson P, Sjogren M, Andreasen N, et al. Studies of the pathophysiological mechanisms in frontotemporal dementia by proteome analysis of CSF proteins. Brain Res Mol Brain Res,2002,109(1-2): 128-133
    41 Hjelmeland LM, Cristofalo VJ, Funk W, et al. Senescence of the retinal pigment epithelium. Mol Vis,1999,5:33
    42 DiPaolo BR, Pignolo RJ, Cristofalo VJ. Identification of proteins differentially expressed in quiescent and proliferatively senescent fibroblast cultures. Exp Cell Res,1995,220:178-185
    43 Rychli K, Kaun C, Hohensinner P, et al. The antiangiogenic factor PEDF is present in the human heart and is regulated by anoxia in myocytes and fibroblasts. J Cell Mol Med,2010,14(1-2):198-205
    44 Lange J, Yafai Y, Reichenbach A, et al. Regulation of pigment epithelium-derived factor production and release by retinal glial (Muller) cells under hypoxia. Invest Ophthalmol Vis Sci,2008; 49(11):5161-5167
    45 Dawson DW, Volpert OV, Gillis P, et al. Pigment epithelium-derived factor: a potent inhibitor of angiogenesis. Science,1999,285(5425):245-248
    46 Notari L, Miller A, Martinez A, et al. Pigment epithelium-derived factor is a substrate for matrix metalloproteinase type 2 and type 9:implications for downregulation in hypoxia. Invest Ophthalmol Vis Sci,2005,46(8): 2736-47
    47 Ueda S, Yamagishi SI, Okuda S. Anti-vasopermeability effects of PEDF in retinal-renal disorders. Curr Mol Med,2010,10(3):279-283
    48 Yang XM, Yafai Y, Wiedemann P, et al. Hypoxia-induced upregulation of pigment epithelium-derived factor by retinal glial (Muller) cells. J Neurosci Res,2012,90(1):257-266
    49 Gao G, Li Y, Zhang D, et al. Unbalanced expression of VEGF and PEDF in ischemia-induced retinal neovascularization. FEBS Lett,2001,489(2-3): 270-276
    50 Takita H, Yoneya S, Gehlbach PL, et al. Retinal neuroprotection against ischemic injury mediated by intraocular gene transfer of pigment epithelium-derived factor. Invest Ophthalmol Vis Sci,2003,44(10): 4497.504
    51 Murakami Y, Ikeda Y, Yonemitsu Y, et al. Inhibition of nuclear translocation of apoptosis-inducing factor is an essential mechanism of the neuroprotective activity of pigment epithelium-derived factor in a rat model of retinal degeneration. Am J Pathol,2008,173(5):1326-1338

© 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