用户名: 密码: 验证码:
紫草素对局灶性脑缺血再灌注损伤小鼠的脑保护作用及其相关机制研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
缺血性脑血管病是临床常见病、多发病,具有致残率高、复发率高、恢复缓慢等特点。国际卫生组织统计在全球范围内平均每400个人中存在一个缺血性脑血管病患者;而在我国每年新发生的250万脑血管病患者中,大约有75%以上为缺血性脑血管病。缺血性脑血管病的死亡率仅次于心脏疾病,是人类主要的死亡原因之一,而在所有的缺血性脑血管病患者中约有30%患者由于溶栓治疗或栓子自发向远端推移出现血管再通,伴随着缺血再灌注损伤的发生。
     缺血再灌注损伤是导致患者病情加重的重要原因,近年来随着现代免疫学和分子生物学的迅速发展,对于脑缺血再灌注损伤病理生理机制的研究已取得了重大进步,认为其病理生理机制存在损伤级联反应,主要包括炎症损害、氧化应激、兴奋性氨基酸毒性、凋亡、细胞内钙超载、自由基损伤等几种因素相互作用形成了复杂的调控网络,造成一系列病理级联反应,可直接或者间接导致神经细胞凋亡/死亡。在脑缺血后复杂的病理损伤机制中,炎症反应、氧化应激是脑缺血再灌注损伤的主要原因,是目前神经科学研究的热点,抗氧化应激、减轻炎症损伤成为治疗缺血性脑血管病的主要途径之一。
     尽管已经有许多合成或天然物质的神经保护剂经过实验研究,证明具有抗炎、抗氧化、抗凋亡等作用,但是由于动物与人类生理机制及耐受性等方面的差异以及脑缺血后级联反应的复杂性,单一阻断某一病理环节的药物可能难以奏效,多数神经保护剂未能在临床得到广泛应用,出现临床和理论的严重脱节,所以在目前的临床治疗中,理想的神经保护剂应该具备针对缺血再灌注损伤多个损伤环节发挥保护作用的特性。
     紫草素(Shikonin)是存在于天然植物中的一种脂溶性萘醌类化合物,分子式为:C16H16O5,主要存在于紫草科植物紫草的根中,具有多种药理活性,研究表明紫草素具有抗氧化、抗肿瘤、抗炎、抗凋亡、抗病毒等生物学作用,并且毒副作用低,具有良好的临床应用潜力。
     本研究选用雄性CD-1小鼠为研究对象,采用线栓法建立脑缺血再灌注模型,在此模型基础上对脑缺血后炎症反应代表性因子TLR4,P38MAPK,NF-κB,TNF-α,氧化应激代表性因子HO-1,Nrf2,PI3K,AKT和MMP-9,claudin-5为代表的血脑屏障损伤程度进行观察,测定大脑皮层梗塞灶超氧化物歧化酶(Superoxide Dismutase,SOD)和丙二醛(Malondiadehyde,MDA)的含量。确定缺血后与炎症反应、氧化损伤的关系,通过选用萘醌类化合物紫草素进行干预,对干预前后梗死体积、脑含水量、神经功能缺损进行评估,观察紫草素对TLR4,P38MAPK,NF-κB,TNF-α,PI3K,AKT,HO-1,Nrf2,MMP-9及Claudin-5信号通路的作用,初步探讨其在缺血再灌注损伤后的脑保护作用机制,并应用PI3K特异性拮抗剂LY294002进行干预,观察上述各因子的变化,评价神经功能缺失、脑水肿和梗死体积情况。本研究分三部分,现将各部分内容叙述如下。
     第一部分紫草素对局灶性脑缺血再灌注损伤小鼠的抗氧化作用及其相关机制研究
     目的:观察脑缺血再灌注损伤后梗死体积、脑含水量、神经功能缺损评分、PI3K、AKT、HO-1、Nrf2的表达变化以及SOD活性和MDA含量的变化,研究紫草素对脑缺血再灌注损伤所致的氧化应激反应的作用,探讨紫草素抗氧化应激作用的相关机制。
     方法:选用成年雄性CD-1小鼠为研究对象,采用改良Longa线栓法制备大脑中动脉缺血再灌注模型。试验1:随机将CD-1小鼠分为2组:假手术组(Sham),手术组(MCAO):根据不同的时间点又分为3h、6h、24h、48h、72h五个亚组。试验2:随机将CD-1小鼠分为4组:假手术组(Sham)、手术组(Vehicle)、低剂量紫草素干预组(L-Shi)(10mg/Kg)、高剂量紫草素干预组(H-Shi)(25mg/Kg)。试验3:采用随机的方法分别将CD-1小鼠分为4组:假手术组(Sham)、溶剂对照组(Vehicle)、大剂量紫草素干预组(H-Shi)及紫草素+LY294002组(Shikonin+LY294002)。Sham组:假手术前以灌胃方式给予等量PBS+1%DMSO,每日一次,连续三天,Vehicle组:缺血再灌注前以灌胃方式给予等量PBS+1%DMSO,每日一次,连续三天,L-Shi组:缺血再灌注前以灌胃方式给予紫草素10mg/Kg,每日一次,连续三天,H-Shi组:缺血再灌注前以灌胃方式给予紫草素25mg/Kg,每日一次,连续三天,紫草素+LY294002组:缺血再灌注前以灌胃方式给予以灌胃方式给予紫草素25mg/Kg,每日一次,连续三天,并于术前15分钟脑室注射30μg LY-294002。各组取材前进行神经功能评分,干湿重法测定脑组织含水量,TTC染色评价脑梗死体积,检测超氧化物歧化酶(SOD)的活性,丙二醛(MDA)含量的变化情况,分别用免疫组化,Western-blotting和RT-qPCR的方法检测缺血脑组织中PI3K、AKT、HO-1、Nrf2蛋白和mRNA水平的变化。
     结果:Sham组中未见小鼠神经功能缺损;Vehicle组在24h和72h神经功能缺损评分均较严重,L-Shi组在24h和72h与Vehicle组相比可降低神经功能缺损,但差异无统计学意义(P>0.05),H-Shi组在24h和72h与Vehicle组相比可明显降低神经功能缺损,结果有统计学差异(P<0.05);与Vehicle组相比,L-Shi组在24h和72h可降低脑含水量,但是差异无统计学意义(P>0.05),而H-Shi组在24h和72h与Vehicle组相比可明显降低脑含水量,差异有统计学意义(P<0.05);L-Shi组在24h和72h可减小脑梗死体积,但是差异无统计学意义(P>0.05),H-Shi组在24h和72h与Vehicle组相比可明显降低脑梗死体积,差异有统计学意义(P<0.05),但是H-Shi对神经功能缺损、脑水肿、脑梗死体积这种改善作用可被PI3K抑制剂LY294002阻断,差异有统计学意义(P<0.05)。
     免疫组化结果显示,与Vehicle组相比,缺血后24h和72h,H-Shi组在24h和72h均可明显升高p-PI3K、p-AKT、HO-1、Nrf2的阳性细胞数,差异有统计学意义(P<0.05);L-Shi组在24h和72h可升高PI3K、AKT、HO-1、Nrf2的阳性细胞数,但是差异无统计学意义(P>0.05),H-Shi对PI3K、AKT、HO-1、Nrf2阳性细胞数的升高作用可被PI3K抑制剂LY294002阻断,差异有统计学意义(P<0.05)。Western blot结果显示,H-Shi在24h和72h可明显升高p-PI3K、p-AKT、HO-1、Nrf2的蛋白表达,差异有统计学意义(P<0.05);L-Shi组在24h和72h可升高PI3K、AKT、HO-1、Nrf2的蛋白表达,但差异无统计学意义(P>0.05),H-Shi对PI3K、AKT、HO-1、Nrf2蛋白的升高作用可被PI3K抑制剂LY294002阻断,差异有统计学意义(P<0.05)。与免疫组化和Western blot结果一致,H-Shi在24h和72h可升高HO-1、Nrf2的mRNA表达,差异有统计学意义(P<0.05);L-Shi组在24h和72h可升高HO-1、Nrf2的mRNA表达,但差异无统计学意义(P>0.05)。与Vehicle组相比,H-Shi可明显增加SOD,减低MDA的含量,差异有统计学意义(P<0.05);L-Shi组可增加SOD,减低MDA的含量,但差异无统计学意义(P<0.05)。
     结论:缺血小鼠脑组织中的HO-1水平出现动态表达的变化过程,紫草素可通过改善脑缺血再灌注损伤后的神经功能缺失,减轻脑水肿,减小脑梗死体积,诱导HO-1、Nrf2、p-PI3K及p-AKT的表达,使SOD含量明显增加,MDA含量明显减少,从而对局灶性缺血再灌注脑损伤小鼠发挥其神经保护作用,同时这种保护作用可被PI3K特异性拮抗剂LY294002阻断,因此我们推测紫草素的神经保护可能是通过PI3K/HO-1通路的激活实现其神经保护作用。
     第二部分紫草素对局灶性脑缺血再灌注损伤小鼠的抗炎作用及其相关机制研究
     目的:观察脑缺血再灌注损伤后TLR4/MAPKs/NF-κB/TNF-α的表达变化,研究紫草素对脑缺血再灌注损伤所致的炎症反应的作用,并探讨紫草素抗炎相关作用机制。
     方法:选用成年雄性CD-1小鼠为研究对象,应用改良线栓法制备大脑中动脉缺血再灌注模型。试验1:将CD-1小鼠随机分为2组:假手术组(Sham),手术组(MCAO),根据不同的时间点又分为3h、6h、24h、48h、72h五个亚组。试验2:将CD-1小鼠随机分为4组:假手术组(Sham)、手术组(Vehicle)、低剂量紫草素干预组(L-Shi)(10mg/Kg)、高剂量紫草素干预组(H-Shi)(25mg/Kg)。Sham组:假手术前以灌胃方式给予等量PBS+1%DMSO,每日一次,连续三天,Vehicle组:缺血再灌注前以灌胃方式给予等量PBS+1%DMSO,每日一次,连续三天,L-Shi组:缺血再灌注前以灌胃方式给予紫草素10mg/Kg,每日一次,连续三天,H-Shi组:缺血再灌注前以灌胃方式给予紫草素25mg/Kg,每日一次,连续三天。各组取材前进行神经功能评分,分别用免疫组化,Western-blotting和RT-qPCR的方法检测缺血脑组织中TLR4,P38MAPK,NF-κB,TNF-αmRNA和蛋白水平的变化。
     结果:在缺血再灌注早期TLR4、p-p38MAPK的表达呈逐渐增高的趋势,在24h达到高峰,可持续到48h,于72h逐渐开始下降。两种剂量紫草素干预后均可以使脑缺血再灌注后缺血脑组织炎症反应降低,TLR4/MAPKs/NF-κB/TNF-α mRNA表达下降。基因水平表达统计结果显示,H-Shi可明显减轻TLR4,p-p38MAPK,NF-κB,TNF-α表达,差异有统计学意义(P<0.05),而L-Shi组虽可减轻缺血后上调的TLR4,p-p38MAPK,NF-κB,TNF-α表达,但结果无统计学差异(P>0.05)。免疫组化和Western blot与基因水平表达统计结果一致。
     结论:紫草素对缺血再灌注小鼠有较好的神经保护作用,可改善脑缺血损伤后的神经功能缺失,减轻脑水肿,减小脑梗死体积,同时缺血再灌注损伤后的脑组织的TLR4,p-p38MAPK,NF-κB,TNF-α的表达变化与脑组织损伤程度一致,提示脑缺血再灌注后TLR4/MAPKs/NF-κB通路的参与了缺血后损伤;紫草素可下调TLR4,p-p38MAPK,TNF-α,抑制NF-κB的核转位,从而减轻缺血后的过度炎症反应,实现缺血后的神经保护作用。
     第三部分紫草素对局灶性脑缺血再灌注损伤小鼠的血脑屏障保护作用及其相关机制研究
     目的:观察脑缺血再灌注损伤后血脑屏障通透性变化,探讨紫草素对血脑屏障的保护作用及相关机制。
     方法:选用成年雄性CD-1小鼠为研究对象,采用改良线栓法制备大脑中动脉缺血再灌注模型。试验1:随机将CD-1小鼠分为2组:假手术组(Sham)(n=30),手术组(MCAO),每组又根据不同的时间点分为3h、6h、24h、48h、72h五个亚组(n=90)。试验2:随机将CD-1小鼠分为4组:假手术组(Sham)、手术组(Vehicle)、小剂量紫草素组(L-Shi)(10mg/Kg)、大剂量紫草素组(H-Shi)(25mg/Kg)。Sham组:假手术前以灌胃方式给予等量PBS+1%DMSO,每日一次,连续三天,Vehicle组:缺血再灌注前以灌胃方式给予等量PBS+1%DMSO,每日一次,连续三天,L-Shi组:缺血再灌注前以灌胃方式给予紫草素10mg/Kg,每日一次,连续三天,H-Shi组:缺血再灌注前以灌胃方式给予紫草素25mg/Kg,每日一次,连续三天。各组取材前进行神经功能评分,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。将各组小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-TPCR来观察脑缺血后Claudin-5以及MMP-9的mRNA和蛋白水平动态变化,通过Evans Blue的渗出情况评估血脑屏障的完整性。
     结果:在缺血再灌注早期MMP-9表达升高(P<0.05),至24h达到高峰,可持续至72h,与正常组比较,差异有统计学意义;两种剂量紫草素干预后均可以上调脑缺血再灌注后小鼠的Claudin-5表达,同时使MMP-9表达减少,H-Shi可明显诱导Claudin-5表达增加,减少MMP-9表达,差异有统计学意义,L-Shi组可上调诱导Claudin-5表达增加,减少MMP-9,但差异无统计学意义(P>0.05)。Claudin-5、MMP-9mRNA表达以CT值表示,Sham组、Vehicle组、L-Shi组、H-Shi组Claudin-5的CT值分别为:Claudin-51.75±0.03,0.19±0.10,0.63±0.28,1.63±0.38;MMP-90.25±0.06,1.87±0.23,1.62±0.10,1.16±0.15。Claudin-5、MMP-9蛋白的表达变化与基因水平改变类似。再灌注24h后,Evans Blue染色的结果显示,与Vehicle组相比,H-Shi能够明显减少Evans Blue的渗出,维持血脑屏障的完整性。
     结论: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。局灶性脑缺血再灌注后,缺血脑组织中Claudin-5、MMP-9表达的变化过程与脑组织损伤程度一致;紫草素通过抑制MMP-9、上调Claudin-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。
Ischemic cerebrovascular disease is the most common type of disease,which is the first leading cause of death and the most frequent cause ofpermanent disability in adults. Ischemic stroke is one of the main diseasesendangering people’s health in our state, have a high incidence, mortality,morbidity, high recurrence rate, and the characteristics of the slow recovery.The worldwide average of one in every400people suffer a stroke in anischemic cerebrovascular disease according to the World Health Organisation.There are250million patients with cerebrovascular disease each year in China,equating to more than75%with ischemic brain disease. Stroke is aslo aleading cause of mortality after heart disease, equating to9%of total deathseach year. Ischaemic stroke accounts for approximately80–85%of all cases,with approximately30%of the former undergoing haemorrhagictransformation.
     Brain tissue secondary injuries to ischemia often leads to an aggravatedillness after cerebral ischemia/reperfusion. Cerebral ischemia/reperfusioninjury is aslo a complex pathophysiologic process which is not been clarifiednow. With the rapid development of modern immunology and molecularbiology, recent studies found that the injuries caused by cerebral blood flowcessation and reperfusion are a rapid cascade reaction which includesinflammatory damage, oxidative stress, excitatory amino acid toxicity,apoptosis, intracellular calcium overload, free radical damage and so on.These pathophysiologic processes overlap and intercommunicate then form avicious cycle which direct/indirect resulting in blood-brain barrier disruptionand brain edema formation, neurological deficit, as well as cell apoptosis ornecrosis.
     Nowadays, beyond only a restricted number of hospitalized patients profiting from thrombolytic therapy, the vast majority of patients sufferingfrom stroke resulted in disability even death owing to the limited therapies.Delaying the development of brain damage and protecting neuron areremained. There a large number of agents have been proved to displayanti-inflammation, anti-oxidation properties but never employed in treatingcerebral ischemia. It is a focus in neuroscience that looking for the idealneuroprotective agents that can block ischemic cascade reaction.
     Shikonin is one of the major naphthoquinone pigment extracted from atraditional herbal medicine Lithospermum erythrorhizon. Evidence has shownthat shikonin possesses a wide range of biological effects, such asantimicrobial, anticancer, antithrombotic, and anti-inflammatory activities totreat thermal edema, arthritis, atherosclerosis. However, the molecular targetsand mechanisms underlying shikonin are not completely characterized, andthe effect of the shikonin in acute stroke is still unknown. In the present study,we demonstrated the unexplored potential of shikonin for the treatment ofcerebral ischemic damage and its potential mechanism.
     ICR mice were induced into focal cerebral ischemiaby transient middlecerebral artery occlusion (MCAO), and received shikonin treatmentimmediately after MCAO. The present study examined the ability of shikonin,the major constituents of Chinese herb Lithospermum erythrorhizon, to induceexpression of HO-1, p85(PI3-kinase) and AKT phospholation, NF-E2-relatedfactor-2(Nrf2) and analyzed its signaling mechanism in mice brain. Theneurological deficits, brain water content, infarct volume and the expression ofTLR4, p-p38MAPK, NF-κB, TNF-α, MMP-9and claudin-5were measured at24h and72h after cerebral ischemia/reperfusion. The activities of superoxidedismutase (SOD) and malondialdehyde (MDA) content in ischemic corticaltissue were aslo detected to examine the oxidative response at24h and72hafter ischemia.
     The study was divided into three part list as below.
     PartⅠAnti-oxidative effect of shikonin on focal cerebralischemia/reperfusion injury in mice and its mechanism of signal transduction pathway
     Objective: This study is to evaluate the time course expression regularityof HO-1, Nrf2, p85(PI3-kinase) and p-AKT, and estimate the anti-oxidativeeffects of shikonin on focal cerebral ischemia/reperfusion injury in mice andexplore its possible mechanisms and related signal transduction pathways.
     Methods: CD-1mice were subjected to transient focal cerebralischemia/reperfusion model was established by middle cerebral arteryocclusion using modified suture occlusion technique. Experiment1: The micewere randomly divided into Sham group(n=30), MCAO group(n=90), Thetime course expression of HO-1in the brain tissue after MCAO. Two groupswere studied, including Sham group, MCAO group. The last two groupincluded3h,6h,24h,48h,72h sub-groups. Experiment2: Shikonin’sneuroprotection against damage from cerebral ischemia/reperfusion. Shikoninwas injected intraperitoneally after MCAO. Mice were reanesthetized andkilled at24h and72h after MCAO. In this part, mice were individed into4groups randomly. Shikonin was injected intraperitoneally immediately afterMCAO. Group1: Sham-operated group (Sham): animals received shamoperation; group2: Vehicle controls (Vehicle): animals received transientMCAO and equal volume PBS including1%DMSO respectively once a dayfor three times before surgery; group3: Shikonin low dose group (L-Shi):animals received transient MCAO and10mg/kg of Shikonin respectively oncea day for three times before surgery; group4: MCAO-Shikonin high dosegroup (H-Shi): animals received transient MCAO and25mg/kg of Shikoninrespectively once a day for three times before surgery. Drug or solvent wasadministered before MCAO, then once daily thereafter. Experiment3:Shikonin was administered before MCAO. Sham-operated group (Sham):animals received sham operation; vehicle controls (Vehicle): animals receivedtransient MCAO and equal volume PBS including1%DMSO; Shikonin lowdose group (L-Shi): animals received transient MCAO and10mg/kg ofShikonin respectively once a day for three times before surgery; MCAO-Shikonin high dose group (H-Shi): animals received transient MCAO and25 mg/kg of Shikonin respectively once a day for three times before surgery;Shikonin+LY-294002group: animals received transient MCAO and25mg/kg of Shikonin respectively once a day for three times before surgery andLY-294002(10μL10mM dissolved in3%DMSO). Infarct volume wasmeasured by TTC staining and morphologic changes were observed by H.E.we measured superoxide dismutase (SOD) and malondialdehyde (MDA) ofcerebral tissue to evaluate antioxidation activitis of shikonin.Immunohistochemistry, RT-qPCR and Western blot were used to analyse theexpression of HO-1, p85(PI3-kinase) and p-AKT and Nrf2.
     Results: Compared with Sham group, HO-1were upregulated at geneand protein level in ischemia/reperfusion brain, beginning at6h and peakingat24h after MCAO (P<0.05). Treatment of shikonin for indicated timeperiods increased up-regulation of HO-1in ischemic brain after cerebralischemia. In addition, treatment of various concentrations of shikonin for24halso increased HO-1expression in a concentration-dependent manner.Treatment with shikonin for indicated time periods, or with variousconcentrations of shikonin also increased HO-1mRNA expression in atime-dependent or a concentration dependent manners. Taken together, thesefindings demonstrate that shikonin increases HO-1protein and mRNAexpression. Shikonin high dose (25mg/kg) upregulated Nrf2and HO-1inMCAO-affected brain tissue, The Nrf2CT of Vehicle group, MCAO group,L-Shi group, H-Shi group were (0.22±0.27,0.85±0.10,0.70±0.05,0.53±0.06). Shikonin reduced infarct volume (P<0.05), and behavioral deficitscaused by MCAO. And also, shikonin significantly increased the activities ofSOD and decreased the production of MDA at24h and72h after ischemia inH-Shi group. Phospho-PI3K and phospho-Akt were examined withimmunohistochemistry, Western blot and RT-qPCR. Few cells were stainedwith phospho-PI3K and phospho-Akt in the cortex in sham group byimmunohistochemistry. In vehicle group, the number of positive cells ofphospho-PI3K and phospho-Akt significantly decreased in the ischemic cortex.In H-Shi group, the number of positive cells of phospho-PI3K and phospho-Akt was significantly increased compared with vehicle group (P <0.05). However, there were no significant differences about the positive cellsbetween vehicle group and L-Shi group. In agreement with the results ofwestern blotting, the mRNA and protein expression of phospho-PI3K andphospho-Akt were down-regulated in Vehicle group compared with Shamgroup (P <0.05). The expression of those factors was significantly increasedin H-Shi group (P<0.05). Whereas L-Shi group did not display changes ofphospho-PI3K and phospho-Akt expression compared with Vehicle group. Tofurther investigate whether the PI3K/Akt pathway mediates theneuroprotection of shikonin in cerebral I/R injury, we treated the mice with aPI3K inhibitor LY-294002(i.c.v.,10μL10mM LY-294002dissolved in3%DMSO) at15min before ischemia. Western blot analysis showed that thePI3K inhibitor LY-294002inhibited the increase of p-Akt level induced byshikonin treatment and almost restored p-Akt level to basal level. In addition,shikonin-induced HO-1expression was antagonized by treatment with PI-3kinase inhibitors LY294002.
     Conclusions: Nrf2, HO-1were induced at the early stage after MCAO.Shikonin reduce cerebral infarct volume and improve neurologic impairmentand protected the brain from damage caused by MCAO, we found thatshikonin increased HO-1mRNA and protein expression time-dependently. Inaddition, shikonin-induced HO-1expression was attenuated by PI3-kinase(phosphatidylinositol3-kinase) inhibitors LY294002. Treatment of mice withshikonin also induced p85(PI3-kinase) and AKT phospholation. Shikoninalso increased NF-E2-related factor-2(Nrf2) accumulation in the nucleus.Moreover, shikonin-induced increase of Nrf2was reduced by PI3-kinaseinhibitors. Thus, shikonin may be useful as a therapeutic agent for thetreatment of cerebral ischemic-associated disorders. The neuroprotection ofshikonin was accomplished by antioxidation. These findings suggest thatshikonin-increased HO-1expression is mediated by Nrf2activation throughthe PI3-kinase/AKT pathway.
     PartⅡ Anti-inflammatory effect of shikonin on focal cerebral ischemia/reperfusion injury in mice and its mechanism of signaltransduction pathway
     Objective: This study is to evaluate the expression regularity of TLR4,MAPKs, NF-κB, TNF-α and estimate the anti-inflammatory effects ofshikonin on focal cerebral ischemia/reperfusion injury in mice and explore itspossible mechanisms and related signal transduction pathways.
     Methods: CD-1mice were subjected to transient focal cerebralischemia/reperfusion model in rats was established by middle cerebral arteryocclusion using modified suture occlusion technique. Experiment1: The micewere randomly divided into Sham group(n=30), MCAO group(n=90), Thetime course expression of TLR4, MAPKs, NF-κB, TNF-α in the brain tissueafter MCAO. Two groups were studied, including Sham group, MCAO group.The last two group included3h,6h,24h,48h and72h sub-groups.Experiment2: Shikonin’s neuroprotection against damage from cerebralischemia/reperfusion. In this part, mice were individed into4groups randomly.Shikonin (98%) was purchased from Santa Cruz Biotechnology; it wasdissolved in dimethyl sulfoxide (DMSO) as a10mmol/L stock solution andstored at–20℃. For all experiments the final concentration of the testedcompound was prepared by diluting the stock with PBS including1%DMSO.Shikonin was administered before MCAO. Mice were reanesthetized andkilled at24h and72h after MCAO. In this part, mice were individed into4groups randomly. Group1: Sham-operated group (Sham): animals receivedsham operation; group2: Vehicle controls (Vehicle): animals receivedtransient MCAO and equal volume PBS including1%DMSO respectivelyonce a day for three times before surgery; group3: Shikonin low dose group(L-Shi): animals received transient MCAO and10mg/kg of Shikoninrespectively once a day for three times before surgery; group4: MCAO-Shikonin high dose group (H-Shi): animals received transient MCAO and25mg/kg of Shikonin respectively once a day for three times before surgery.Drug or solvent was administered before MCAO, then once daily thereafter.Immunohistochemistry, RT-qPCR and Western blot were used to analyse the expression of TLR4, MAPKs, NF-κB, TNF-α.
     Results: TLR4, p-p38MAPK, NF-κB and TNF-α were examined withimmunohistochemistry, Western blot and RT-qPCR. Few cells were stainedwith TLR4, p-p38MAPK, NF-κB and TNF-α in the cortex in sham group byimmunohistochemistry. In vehicle group, the number of positive cells of TLR4,p-p38MAPK, NF-κB and TNF-α significantly increased in the ischemic cortex,among which the location of NF-κB is in nucleus. In H-Shi group, the numberof positive cells of TLR4, p-p38MAPK, NF-κB and TNF-α was significantlydecreased compared with vehicle group (P <0.05). Moreover, positive nucleiof NF-κB were also reduced and lots of cells labeled by NF-κB were stainedonly in cytoplasm (P <0.05). However, there were no significant differencesabout the positive cells between vehicle group and L-Shi group.We found the protein and mRNA levels of TLR4, p-p38MAPK, NF-κB andTNF-α in ischemic tissue were upregulated at24h and72h after MCAO. Wefirst analyzed the protein levels of nuclear NF-κB p65and total TLR4,p-p38MAPK, TNF-α and MMP-9by western blot. The NF-κB p65was rich incytosolic fractions but poor in nuclear extracts in brain tissue of Sham group.In contrast, the protein level of NF-κB in Vehicle group was significantlyenhanced in nuclear fraction and concurrently decreased in cytosol at24h and72h after ischemia, indicating the translocation of these NF-κB subunits fromthe cytosol to the nucleus. High dose of shikonin significantly decreased theexpressions TLR4, p-p38MAPK, NF-κB and TNF-α at24h and72h afterMCAO. However, there were no significant differences in the protein levels ofTLR4, p-p38MAPK, NF-κB and TNF-α between Vehicle group and L-Shigroup. Another parallel set of samples treated with L-Shi and H-Shi werealso evaluated for mRNA expression of TLR4, p-p38MAPK, NF-κB andTNF-α by RT-qPCR. In agreement with the results of western blotting, themRNA expression of TLR4, p-p38MAPK, NF-κB and TNF-α wereup-regulated in Vehicle group compared with Sham group (P <0.05). Theover-expression of those factors was significantly decreased in H-Shi group(P<0.05). Whereas L-Shi group did not display changes of TLR4, p-p38MAPK, NF-κB and TNF-α expression compared with Vehicle group.
     Conclusions: The expression of TLR4, p-p38MAPK, NF-κB andTNF-α were up-regulated after ischemia. Systemic administration of shikoninis effective which can decrease the expression of TLR4, p-p38MAPK, NF-κBand TNF-α. Therefore, the excessive inflammation of the brain ischemia wasalleviated.
     Part Ⅲ The permeability of blood-brain barrier after focal cerebralischemia in rats and the protection of shikonin
     Objective: This study is to evaluate the time course expression ofClaudin-5and MMP-9and explore the underling regulation mechanisms ofshikonin.
     Methods: CD-1mice were subjected to transient focal cerebralischemia/reperfusion model in rats was established by middle cerebral arteryocclusion using modified suture occlusion technique. Experiment1: The micewere randomly divided into Sham group(n=30), MCAO group(n=90), Thetime course expression of Claudin-5and MMP-9in the brain tissue afterMCAO. Two groups were studied, including Sham group, MCAO group. Thelast two group included3h,6h,24h,48h and72h sub-groups. Experiment2:shikonin’s neuroprotection against damage from cerebral ischemia/reperfusion.shikonin was administered before MCAO. Mice were reanesthetized andkilled at24h and72h after MCAO. In this part, mice were individed into4groups randomly. Group1: Sham-operated group (Sham): animals receivedsham operation; group2: Vehicle controls (Vehicle): animals receivedtransient MCAO and equal volume PBS including1%DMSO respectivelyonce a day for three times before surgery; group3: Shikonin low dose group(L-Shi): animals received transient MCAO and10mg/kg of Shikoninrespectively once a day for three times before surgery; group4: MCAO-Shikonin high dose group (H-Shi): animals received transient MCAO and25mg/kg of Shikonin respectively once a day for three times before surgery.Drug or solvent was administered before MCAO, then once daily thereafter.RT-qPCR and Western blot were used to analyse the expression of Claudin-5 and MMP-9. The loss of BBB integrity was assessed by leakage of Evans bluefrom microvessels after intravenous injection.
     Results: There was considerable leak of Evans blue into the ischemicbrain after MCAO, showing that the BBB had been disrupted in the ischemichemisphere. Compared with Vehicle group, Evans blue evasion of braintissues was significantly decreased in H-Shi group (P<0.05). Whileclaudin-5's expression was increased in both L-Shi and H-Shi groups (P<0.01) at gene and protein levels by western blotting and RT-qPCR at24h and72h, but low dose of shikonin did not display significant level. At the sametime, the MMP-9expression was obviously decreased in H-Shi group (P<0.05), relative to that in normal group.
     Conclusions: The blood-brain barrier was dramatically disrupted at theearly stage of focal cerebral ischemia, which was responsible for the brainedema. Shikonin normalizing Claudin-5by inhibiting MMP-9couldcontribute to the alleviated infarct volume, neurological deficits and brainedema in the study.
引文
1Thrift AG, Dewey HM, Macdonell RA, et al. Incidence of the major strokesubtypes: initial findings from the North East Melbourne stroke incidencestudy (NEMESIS). Stroke,2001,32:1732~1738
    2Crowder RJ, Freman RS. PhosPhatidylinositol3-Kinase and Akt Proteinkinase are necessary and sufficient for the survival of nerve gowthfactor-dependent sympathetic neurons. J Neurosci,1998,18:2933~2943
    3Shin WH, Park SJ, Kim EJ. Effect of anthocyanins in middle cerebralartery occlusion and reperfusion model of cerebral ischemia in rats. LifeSci,2006,79:130~137
    4Chan PH. Reactive oxygen radicals in signaling and damage in theischemic brain, J. Cereb. Blood Flow Metab,2001,21:2~14
    5Mori T, Tateishi N, Kagamiishi Y, et al. Attenuation of a delayed increasein the extracellular glutamate level in the peri-infarct area following focalcerebral ischemia by a novel agent ONO-250696. Neurochem. Int,2004,45:381~387
    6Nita DA, Nita V, Spulber S, et al. Oxidative damage following cerebralischemia depends on reperfusion-A biochemical study in rat. J. Cell. Mol.Med,2001,5:163~170
    7Mishina M, Sakimura K, Mori H, et al. A single amino acid residuedetermines the Ca2+permeability of AMPA-selective glutamate receptorchannels. Biochem. Biophys. Res.Commun,1991,180:813~821
    8Lewen A, Matz P, Chan PH. Free radical pathways in CNS injury. JNeurotrauma,2000,17:871~890
    9Budd SL. Mechanisms of neuronal damage in brain hypoxia/ischemia:focus on the role of mitochondrial calcium accumulation. Pharmacol Ther,1998,80:203~229
    10Lewen A, Matz P, Chan PH. Free radical pathways in CNS injury. JNeurotrauma,2001,17:871~890
    11Nurmi A, Lindsberg PJ, Koistinaho M, et al. Nuclear factor-kappaBcontributes to infarction after permanent focal ischemia. Stroke,2004,35:987~991
    12Dong X, Song YN, Liu WG, et al. Mmp-9, a potential target for cerebralischemic treatment. Curr Neuropharmacol,2009,7:269~275
    13Martins T, Baptista S, Gon alves J, et al. Methamphetamine transientlyincreases the blood-brain barrier permeability in the hippocampus: role oftight junction proteins and matrix metalloproteinase-9. Brain Res,2011,1411:28~40
    14Moi P, Chan K, Asunis I, et al. Isolation of NF-E2-related factor2(Nrf2),a NF-E2-like basic leucine zipper t ranscriptional activator that binds to the tandem NF-E2/AP1repeat of the beta-globin locus cont rol region.Proc Natl Acad Sci U SA,1994,91:9926~9930
    15Yoshimi N, Wang A, Morishita Y, et al. Modifying effects of fungal andherb metabolites on azoxymethaneinduced intestinal carcinogenesis in rats.Cancer Sci,1992,83:1273~1278
    16Singh F, Gao D, Lebwohl MG, et al. Shikonin modulates cell proliferationby inhibiting epiderm al growth factor receptor signaling in humanepidermoid carcinoma cells. Cancer Lett,2003,200:115~121
    17Hisa T, Kimura Y, Takada K, et al. Shikonin, an ingredient ofLithospermum erythrorhizon, inhibits angiogenesis in vivo and in vitro.Anticancer Res,1998,18:783~790
    18Chen X, Yang L, Oppenheim JJ, et al. Cellular pharmacology studies ofshikonin derivatives. Phytother Res,2002,16:199~209
    19Staniforth V, Wang SY, Shyur LF, et al. Shikonins, phytocompounds fromLithospermum erythrorhizon, inhibit the transcriptional activation ofhuman TNF-α promoter in vivo. J Bio Chem,2004,279:5877~5885
    20Collino M, Aragno M, Mastrocola R, et al. Oxidative stress andinflammatory response evoked by transient cerebral ischemia/reperfusion:effects of the PPAR-alpha agonist WY14643. Free Radic Biol Med,2006,41:579~589
    21Longa EZ, Weinstein PR, Carlson S, et al. Reversible middle cerebralartery occlusion without craniectomy in rats. Stroke,1989,20:84~91
    22Laing RJ, Jakubowski J, Laing RW, et al. Middle cerebral artery occlusionwithout eraniectomy in rats:which method works best? Stroke,1993,24:294~297
    23Tatlisumak T, Carano RA, Takano K, et al. A novel endothelin antagonist,A-127722, attenShites ischemic lesion size in rats with temporary middlecerebral24artery occlusion: a diffusion and perfusion MRI study. Stroke,1998,29:850~857
    24Toni D, Chamorro A, Kaste M, et al. Acute treatment of ischaemic stroke.European Stroke Initiative. Cerebrovasc Dis,2004,17:30~46
    25Kappelle LJ, Van Der, Worp HB. Treatment or prevention ofcomplications of acute ischemic stroke. Curr Neurol Neurosci Rep,2004,4:36~41
    26Rosen DR, Siddique T, Patterson D, et al. Mutations in Cu/Zn superoxidedismutase gene are associated with familial amyotrophic lateral sclerosis.Nature,1993,362:59~62
    27Schettler V, Methe H, Staschinsky D, et al. Review: theoxidant/antioxidant balance during regular low density lipoproteinapheresis.Ther Apher,1999,3:219~226
    28Moi P, Chan K, Asunis I, et al. Isolation of NF-E2-related factor2(Nrf2),a NF-E2-like basic leucine zipper t ranscriptional activator that binds to the tandem NF-E2/AP1repeat of the beta-globin locus cont rol region.Proc Natl Acad Sci U SA,1994,91:9926~9930
    29Jain AK, Bloom DA, Jaiswal AK, et al. Nuclear import and export signal sin cont rol of Nrf2. J Biol Chem,2005,280:29158~29168
    30Giudice A, Montella M. Activation of the Nrf2-ARE signaling pathway: apromising strategy in cancer prevention. Bioessays,2006,28:169~181
    31Chan PH. Reactive oxygen radicals in signaling and damage in theischemic brain. J. Cereb Blood Flow Metab,2001,21:2~14
    32Chen L, Wang L, Zhang X, et al.The protection by Octreotide againstexperimental ischemic stroke: Up-regulated transcription factor Nrf2,HO-1and down-regulated NF-κB expression. Brain Res,2012,26:80~87
    33Li M, Zhang X, Cui L, et al. The neuroprotection of oxymatrine incerebral ischemia/reperfusion is related to nuclear factor erythroid2-related factor2(nrf2)-mediated antioxidant response: role of nrf2andhemeoxygenase-1expression. Biol Pharm Bull,2011,34:595~601
    34Yang C, Zhang X, Fan H, et al. Curcumin upregulates transcription factorNrf2, HO-1expression and protects rat brains against focal ischemia.Brain Res,2009,1282:133~141
    35Li L, Zhang X, Cui L, et al. Ursolic acid promotes the neuroprotection byactivating Nrf2pathway after cerebral ischemia in mice. Brain Res,2013,1497:32~39
    1Toni D, Chamorro A, Kaste M, et al. Acute treatment of ischaemic stroke.European Stroke Initiative. Cerebrovasc Dis,2004,17:30~46
    2Kappelle LJ, Van Der, Worp HB. Treatment or prevention of complicationsof acute ischemic stroke. Curr Neurol Neurosci Rep,2004,4:36~41
    3Wu JS, Cheung WM, Tsai YS, et al. Ligand-activated peroxisomeproliferator-activated receptor-gamma protects against ischemic cerebralinfarction and neuronal apoptosis by14-3-3epsilon upregulation.Circulation,2009,119:1124~1134
    4Wang CX, Ding X, Noor R, et al. Rosiglitazone alone or in combinationwith tissue plasminogen activator improves ischemic brain injury in anembolic model in rats. J Cereb Blood Flow Metab,2009,29:1683~1694
    5Akira S. Toll-like recept or signaling. J Biol Chem,2003,278:38105~38108
    6Akira S, Takeda K. Toll-like receptor signalling. Nat Rev Immunol,2004,4:499~511
    7Abreu MT, Arditi M. Innate immunity and toll-like receptors: clinicalimplic ations of basic science research. J Pediatr,2004,144:421~429
    8Okun E, Griffioen KJ, Lathia JD, et al. Toll-like receptors inneurodegeneration. Brain Res Rev,2009,59:278~292
    9Wang L, Zhang X, Liu L, et al. Tanshinone II A down-regulates HMGB1,RAGE, TLR4, NF-kappaB expr ession, ameliorat es BBB permeab ilityand endothelial cell function, and protects rat brains against focal ischemia.Brain Res,2010,1321:143~151
    10Fan H, Li L, Zhang X, et al. Oxymatrine downregu lates TLR4, TLR2,MyD88, and NF-kappaB and protects rat brains against focal ischemia.Mediators Inflamm,2009,2009:704~706.
    11Liu Y, Zhang X, Yang C, et al. Oxymatrine protects rat brains againstpermanent focal ischemia and downregulates NF-κB expression. BrainRes,2009,1321:143~151
    12Wang L, Zhang X, Liu L, et al. Atorvastatin protects rat brains againstpermanent focal ischemia and downregulates HMGB1, HMGB1receptors(RAGE and TLR4), NF-kappaB expression. Neurosci Lett,2010,471:152~156
    13Fan H, Cook JA. Molecular mechanisms of endotoxin tolerance.JEndotoxin Res,2004,10:71~74
    14Allan SM, Rothwell NJ. Cytoki nes and acute neurodegenerati on. Nat RevNeurosci,2001,2:734~744
    15Hayashi F, Smith KD, Ozinsky A, et al. The innate im mune respons e tobacterial flagellin is mediated by Toll-like receptor5. Nature,2001,410:
    1099~1103
    16Yi JH, Park SW, Kapadia R, et al. Role of transcription factors inmediating post-ischemic cerebral inflammation and brain damage.Neurochem Int,2007,50:1014~1027
    17Zheng Z, Yenari MA. Post-ischemic inflammation: molecular mechanismsand therapeutic implications. Neurol Res,2004,26:884~892
    18Rosenberg GA. Matrix metalloproteinases in neuroinflammation. Glia,2002,39:279~291
    19Rubin LL, Staddon JM. The cell biology of the blood–brain barrier. AnnuRev Neurosci,1999,22:11~28
    20Ruhul AR, Senga T, Oo ML, et al. Secretion of matrix metalloproteinase-9by the proinflammatory cytokine, IL-1beta: a role for the dual signallingpathways, Akt and Erk. Genes Cells,2003,8:515~523
    21Yoshimi N, Wang A, Morishita Y, et al. Modifying effects of fungal andherb metabolites on azoxymethaneinduced intestinal carcinogenesis in rats.Cancer Sci,1992,83:1273~1278
    22Singh F, Gao D, Lebwohl MG, et al. Shikonin modulates cell proliferationby inhibiting epiderm al growth factor receptor signaling in humanepidermoid carcinoma cells. Cancer Lett,2003,200:115~121
    23Hisa T, Kimura Y, Takada K, et al. Shikonin, an ingredient ofLithospermum erythrorhizon, inhibits angiogenesis in vivo and in vitro.Anticancer Res,1998,18:783~790
    24Chen X, Yang L, Oppenheim JJ, et al. Cellular pharmacology studies ofshikonin derivatives. Phytother Res,2002,16:199~209
    25Staniforth V, Wang SY, Shyur LF, et al. Shikonins, phytocompounds fromLithospermum erythrorhizon, inhibit the transcriptional activation ofhuman TNF-α promoter in vivo. J Bio Chem,2004,279:5877~5885
    26Tanaka S, Tajima M, Tsukada M, et al. A comparative study onantiinflammatory activities of the enantiomers, shikonin and alkannin. JNat Prod,1986,49:466~469
    27An S, Park YD, Paik YK, et al. Human ACAT inhibitory effects ofshikonin derivatives from Lithospermum erythrorhizon. Bioorg MedChem Lett,2007,17:1112~1116
    28Kundakovic T, Fokialakis N, Dobric S, et al. Evaluation of theanti-inflammatory and cytotoxic activities of naphthazarine derivativesfrom Onosma leptantha. Phytomedicine,2006,13:290~294
    29Cui L, Zhang X, Yang R, et al. Baicalein is neuro protective in rat MCAOmodel: role of12/15-lipoxygena se, mitogen-activated protein kinase andcytosolic phospho lipase A2. Pharmacol Biochem Behav,2010,96:469~475
    30Qiao H, Zhang X, Zhu C, et al. Luteolin downregulates TLR4, TLR5,NF-κB and p-p38MAPK expression, upregulates the p-ERK expression,and protects rat brains against focal ischemia. Brain Res,2012,1448:71~81
    31Longa EZ, Weinstein PR, Carlson S, et al. Reversible middle cerebralartery occlusion without craniectomy in rats. Stroke,1989,20:84~91
    32Aing RJ, Jakubowski J, Laing RW, et al. Middle cerebral artery occlusionwithout eraniectomy in rats:which method works best? Stroke,1993,24:294~297
    33ThomPson AB, Bohiing T, Heires A, et al. Lower resPiraory tract ironburden is increased in assosciate with cigarette smoking. J Lab Clin Med,1991,117:494
    34Wesselius LJ, Nelson ME, Skikne BS. Increased released of ferritin andiron buiron loaded alveolar maeroPhages in cigarette smokers. Am JResPir Crit Care Med,1994,150:690~695
    35Lambeth, JD. NOX enzymes and the biology of reactive oxygen.Nat RevImrnunol,2004,181~189
    36Medzhitov R, Preston-Hurlburt P, Janeway CA. A human homologue ofthe Drosophila Toll protein signals activation of adaptive immunity.Nature,1997,388:394~397
    37Caso JR, Pradillo JM, Hurta do O, et al. Toll-like receptor4is involved inbrain damage and inflamma tion after expe rimental stroke. Circulation,2007,115:1599~1608
    38Tang SC, Arumugam TV, Xu X, et al. Pivotal role for neuronal Toll-likereceptors in ischemic brain injury and functional deficits. Proc Natl AcadSci USA,2007,104:13798~13803
    39Broad, Kirby JA, Jones DE, et al. Toll-like receptor interactions: toleranceof MyD88-dependent cytokines but enhancement of MyD88-independentinterferon-beta production. Immunology,2007,120:103~111
    40Yan SF, Fujita T, Lu J, et al. Egr-1, a master switch coordinatingupregulation of divergent gene families underlying ischemic stress. NatureMed,2000,6:1355~1361
    41Nurmi A, Lindsberg PJ, Koistinaho M, et al. Nuclear factor-kappaBcontributes to infarction after permanent focal ischemia. Stroke,2004,35:987~991
    42Sironi L, Banfi C, Brioschi M, et al. Activation of NF-κB and ERK1/2after permanent focal ischemia is abolished by simvastatin treatment.Neurobiol Dis,2006,22:445~451
    43Zhang HL, Gu ZL, Savitz SI, et al. Neuropro tective effects of prostaglandinA(1) in rat models of permanent focal cerebral ischemi a areassociated with nuclear factor-kappa B inhibition and peroxisomeproliferator-activ ated receptor-gamma up-r egulation. J Neurosci Res,2008,86:1132~1141
    44Ren JL, Pan JS, Lu YP. Inflammatory signaling and cellular senescence.Cell Signal,2008,21:378~383
    45De Keulenaer GW, Ushi o-Fukai M, Yin Q, et al. Convergence ofredox-sensitive and mitogen-activated protein kinase signaling pathwaysin tumor necrosis factor-alpha-mediated monocyt echemo attractantprotein-1induction in vascular smooth muscle cells. Arterioscler.Thromb Vasc Biol,2000,20:385~391
    46Thompson WL, Van Eldik LJ. Inflammatory cyto kines stimulate thechemo kines CCL2/M CP-1and CCL7/MCP-3through NF-kappaB andMAPK dependent pathwa ys in rat astrocytes. Brain Res,2009,1287:47~57
    47Hershko DD, Robb BW, Wray CJ, et al. Superinduction of IL-6bycycloheximide is associated with mRNA stabilization and sustainedactivation of p38map kinase and NF-kappaB in cultured caco-2cells. JCell Biochem,2004,5:951~961
    48Safirstein R, DiMari J, Megyesi J, et al. Mechanism s of renal repair andsurvival following acute injury. Semin Nephrol,1998,18:519~522
    49Ren JL, Pan JS, Lu YP. Inflammatory signaling and cellular senescence.Cell Signal,2008,21:378~383
    50Teeling JL, Perry VH. Systemic infection and inflammation in acute CNSinjury and chronic neurodegeneration: underlying mechanisms.Neuroscience,2008,158:1062~1073
    51Wang MJ, Lin SZ, Kuo JS, et al. Urocortin modulates inflammatoryresponse and neurotoxicity induced by microglial activation. J Immunol,2007,179:6204~6214
    52Kanaan NM, Kordower JH, Collier TJ. Age and region-specific responsesof microglia, but not astrocytes, suggest a role in selective vulnerability ofdopamine neurons after1-methyl-4-phenyl-1,2,3,6-tetrahydropyridineexposure in monkeys. GLIA,2008,56:1199~1214
    53Kanaan NM, Kordower JH, Collier TJ. Age-related changes in glial cellsof dopamine midbrain subregions in rhesus monkeys. Neurobiol Aging,2010,31:937~952
    54Itoh K, Mochizuki M, Ishii Y, et al. Transcription factor Nrf2regulatesinflammation by mediating the effect of15-deoxy-Delta(12,14)-prostaglandin. Mol Cell Biol,2004,24:36~45
    55Rahman I, Biswas SK, Kirkham PA. Regulation of inflammation andredox signaling by dietary polyphenols. Biochem Pharmacol,2006,72:1439~1452
    1Nitta T, Hata M, Gotoh S, Seo Y, et al. Size-selective loosening of theblood-brain barrier in claudin-5-deficient mice. J Cell Biol,2003,161:653~660
    2Wang Z, Leng Y, Tsai LK, et al. Valproic acid attenuates blood-brainbarrier disruption in a rat model of transient focal cerebral ischemia: theroles of HDAC and MMP-9inhibition. J Cereb Blood Flow Metab,2011,31:52~57
    3Martins T, Baptista S, Gon alves J, et al. Methamphetamine transientlyincreases the blood-brain barrier permeability in the hippocampus: role oftight junction proteins and matrix metalloproteinase-9. Brain Res,2011,1411:28~40
    4Dong X, Song YN, Liu WG, et al. Mmp-9, a potential target for cerebralischemic treatment. Curr Neuropharmacol,2009,7:269~275
    5Jiao H, Wang Z, Liu Y, et al. Specific role of tight junction proteinsclaudin-5, occludin, and ZO-1of the blood-brain barrier in a focal cerebralischemic insult. J Mol Neurosci,2011,44:130~139
    6Yi JH, Park SW, Kapadia R, et al. Role of transcription factors inmediating post-ischemic cerebral inflammation and brain damage.Neurochem Int,2007,50:1014~1027
    7Zheng Z, Yenari MA. Post-ischemic inflammation: molecular mechanismsand therapeutic implications. Neurol Res,2004,26:884~892
    8Rosenberg GA. Matrix metalloproteinases in neuroinflammation. Glia,2002,39:279~291
    9Rubin LL, Staddon JM. The cell biology of the blood–brain barrier. AnnuRev Neurosci,1999,22:11~28
    10Ruhul AR, Senga T, Oo ML, et al. Secretion of matrix metalloproteinase-9by the proinflammatory cytokine, IL-1beta: a role for the dual signallingpathways, Akt and Erk. Genes Cells,2003,8:515~523
    11Longa EZ, Weinstein PR, Carlson S, et al. Reversible middle cerebralartery occlusion without craniectomy in rats. Stroke,1989,20:84~91
    12Aing RJ, Jakubowski J, Laing RW, et al. Middle cerebral artery occlusionwithout eraniectomy in rats: which method works best? Stroke,1993,24:294~297
    13Furuse M, Hirase T, Itoh M, et al. Occludin: a novel integral membraneprotein localizing at tight junctions. J Cell Biol,1993,123:1777~1788
    14Furuse M, Fujita K, Hiiragi T, et al. Claudin-1and-2: novel integralmembrane proteins localizing at tight junctions with no sequencesimilarity to occludin. J Cell Biol,1998a,141:1539~1550
    15Martin-Padura I, Lostaglio S, Schneemann M, et al. Junctional adhesionmolecule, a novel member of the immunoglobulin superfamily thatdistributes at intercellular junctions and modulates monocytetransmigration. J Cell Biol,1998,142:117~127
    16Morita K, Sasaki H, Furuse M, et al. Endothelial claudin:claudin-5/TMVCF constitutes tight junction strands in endothelial cells. JCell Biol,1999b,147:185~194
    17Tsukita S, Furuse M, Itoh M. Multi-functional strands in tight junctions.Nat Rev Mol Cell Biol,2001,2:285~293
    18Honda M, Nakagawa S, Hayashi K, et al. Adrenomedullin improves theblood-brain barrier function through the expression of claudin-5. Cell MolNeurobiol,2006,26:109~118
    19Magnoni S, Baker A, Thomson S, et al. Neuroprotective effect ofadenoviral-mediated gene transfer of TIMP-1and-2in ischemic braininjury. Gene Ther,2007,14:621~625
    20Pfefferkorn T, Rosenberg GA. Closure of the blood-brain barrier by matrixmetalloproteinase inhibition reduces rtPAmediated mortality in cerebralischemia with delayed reperfusion. Stroke,2003,34:2025~2030
    21Rosenberg GA, Estrada EY, Dencoff JE. Matrix metalloproteinases andTIMPs are associated with blood-brain barrier opening after reperfusion inrat brain. Stroke,1998,29:2189~2195
    22Gidday JM, Gasche YG, Copin JC, et al. Leukocyte-derived matrixmetalloproteinase-9mediates blood-brain barrier breakdown and isproinflammatory after transient focal cerebral ischemia. Am J PhysiolHeart Circ Physiol,2005,289: H558~H568
    23Chakraborti S, Mandal M, Das S, et al. Regulation of matrixmetalloproteinases: An overview. Mol Cell Biochem,2003,253:269~285
    24Ellerbroek SM, Halbleib JM, Benavidez M, et al. Phosphatidylinositol3-kinase activity in epidermal growth factor-stimulated matrixmetalloproteinase-9production and cell surface association. Cancer Res,2001,61:1855~1861
    25Liu JF, Crépin M, Liu JM, et al. FGF-2and TPA induce matrixmetalloproteinase-9secretion in MCF-7cells through PKC activation ofthe Ras/ERK pathway. Biochem Bioph Res Commun,2002,293:1174~1182
    26Ruhul Amin AR, Senga T, Oo ML, et al. Secretion of matrixmetalloproteinase-9by the proinflammatory cytokine, IL-1beta: a role forthe dual signalling pathways, Akt and Erk. Genes Cells,2003,8:515~523
    27Suyama K, Shapiro I, Guttman M, et al. A signaling pathway leading tometastasis is controlled by N-cadherin and the FGF receptor. Cancer Cell2002,2:301~314
    1Rosenberg GA. Matrix metalloproteinases in neuroinflammation. Glia,2002,39:279~291
    2Fingleton B. Matrix metalloproteinases as valid clinical targets. Curr.Pharm. Des,2007,13:333~346
    3Ruhul Amin AR, Senga T, Oo ML, et al. Secretion of matrixmetalloproteinase-9by the proinflammatory cytokine, IL-1beta: a role forthe dual signalling pathways, Akt and Erk. Genes Cells,2003,8:515~523
    4Asahi M, Asahi K, Jung JC, et al. Role for matrix metalloproteinase9afterfocal cerebral ischemia: effects of gene knockout and enzyme inhibitionwith BB-94. J Cereb Blood Flow Metab,2000,20:1681~1689
    5Asahi M, Wang X, Mori T, et al. Effects of matrix metalloproteinase-9gene knock-out on the proteolysis of blood-brain barrier and white mattercomponents after cerebral ischemia. J Neurosci,2001,21:7724~7732
    6Magnoni S, Baker A, Thomson S, et al. Neuroprotective effect ofadenoviral-mediated gene transfer of TIMP-1and-2in ischemic braininjury. Gene Ther,2007,14:621~625
    7Pfefferkorn T, Rosenberg GA. Closure of the blood-brain barrier by matrixmetalloproteinase inhibition reduces rtPAmediated mortality in cerebralischemia with delayed reperfusion. Stroke,2003,34:2025~2030
    8Romanic A.M, White RF, Arleth AJ, et al. Matrix metalloproteinaseexpression increases after cerebral focal ischemia in rats-Inhibition ofmatrix metalloproteinase-9reduces infarct size. Stroke,1998,29:1020~1030
    9Rosenberg GA, Estrada EY, Dencoff JE. Matrix metalloproteinases andTIMPs are associated with blood-brain barrier opening after reperfusion inrat brain. Stroke,1998,29:2189~2195
    10Zhang XJ, Wang Y, Yamamoto G, et al. Expression of matrixmetalloproteinases MMP-2, MMP-9and their tissue inhibitors TIMP-1and TIMP-2in the epithelium and stroma of salivary gland pleomorphicadenomas. Histopathology,2009,55:250~260
    11Murphy G, Nagase H. Progress in matrix metalloproteinase research. MolAspects Med,2008,29:290~308
    12Chakraborti S, Mandal M, Das S, et al. Regulation of matrixmetalloproteinases: An overview. Mol Cell. Biochem,2003,253:269~285
    13Vanden Steen PE, Van Aelst I, Hvidberg V, et al. The hemopexin andO-glycosylated domains tune gelatinaseB/MMP-9bioavailability viainhibition and binding to cargo receptors. J Biol Chem,2006,281:18626~18637
    14Planas AM, Solé S, Justicia C. Expression and activation of matrixmetalloproteinase-2and-9in rat brain after transient focal cerebralischemia. Neurobiol Dis,2001,8:834~846
    15Rosell A, Ortega-Aznar A, Alvarez-Sabín J, et al. Increased brainexpression of matrix metalloproteinase-9after ischemic and hemorrhagichuman stroke. Stroke,2006,37:1399~1406
    16Rosell A, Cuadrado E, Ortega-Aznar A, et al. MMP-9-Positive neutrophilinfiltration is associated to blood-brain barrier breakdown and basal laminatype IV collagen degradation during hemorrhagic transformation afterhuman ischemic stroke. Stroke,2008,39:1121~1126
    17Rosenberg GA. Matrix metalloproteinases and their multiple roles inneurodegenerative diseases. Lancet Neurol,2009,8:205~216
    18Rosell A, Lo EH. Multiphasic roles for matrix metalloproteinases afterstroke. Curr Opin. Pharmacol,2008,8:82~89
    19Gurney KJ, Estrada EY, Rosenberg GA. Blood-brain barrier disruption bystromelysin-1facilitates neutrophil infiltration in neuroinflammation.Neurobiol Dis,2006,23:87~96
    20Solé S, Petegnief V, Gorina R, et al. Activation of matrixmetalloproteinase-3and agrin cleavage in cerebral ischemia/reperfusion. J.Neuropathol. Exp Neurol,2004,63:338~349
    21Gasche Y, Fujimura M, Morita-Fujimura Y, et al. Early appearance ofactivated matrix metalloproteinase-9after focal cerebral ischemia in mice:A possible role in blood-brain barrier dysfunction. J Cereb Blood FlowMetab,1999,19:1020~1028
    22Park KP, Rosell A, Foerch C, et al. Plasma and brain matrixmetalloproteinase-9after acute focal cerebral ischemia in Rats. Stroke,2009,40:2836~2842
    23Montaner J, Alvarez-Sabín J, Molina C, et al. Matrix metalloproteinaseexpression after human cardioembolic stroke-Temporal profile andrelation to neurological impairment. Stroke,2001,32:1759~1766
    24Montaner J, Rovira A, Molina CA, et al. Plasmatic level ofneuroinflammatory markers predict the extent of diffusionweighted imagelesions in hyperacute stroke. J Cereb Blood Flow Metab,2003,23:1403~1407
    25Horstmann S, Kalb P, Koziol J, et al. Profiles of mtrix metalloproteinases,their inhibitors, and laminin in stroke patients-Influence of differenttherapies. Stroke,2003,34:2165~2170
    26Zhao BQ, Ikeda Y, Ihara H, et al. Essential role of endogenous tissueplasminogen activator through matrix metalloproteinase9induction andexpression on heparinproduced cerebral hemorrhage after cerebralischemia in mice. Blood,2004,103:2610~2616
    27Copin JC, Merlani P, Sugawara T, et al. Delayed matrix metalloproteinaseinhibition reduces intracerebral hemorrhage after embolic stroke in rats.Exp. Neurol,2008,213:196~201
    28Montaner J, Alvarez-Sabín J, Molina CA, et al. Matrix metalloproteinaseexpression is related to hemorrhagic transformation after cardioembolicstroke. Stroke,2001,32:2762~2767
    29Castellanos M, Leira R, Serena J, et al. Plasma metalloproteinase-9concentrationpredicts hemorrhagic transformation in acute ischemic stroke.Stroke,2003,34:40~46
    30Montaner J, Molina CA, Monasterio J, et al. Matrix metalloproteinase-9pretreatment level predicts intracranial hemorrhagic complications afterthrombolysis in human stroke. Circulation,2003,107:598~603
    31Zhao BQ, Wang S, Kim HY, et al. Role of matrix metalloproteinases indelayed cortical responses after stroke. Nat. Med,2006,12:441~445
    32Lee SR, Kim HY, Rogowska J, et al. Involvement of matrixmetalloproteinase in neuroblast cell migration from the subventricularzone after stroke. J Neurosci,2006,26:3491~3495
    33Zhao BQ, Tejima E, Lo EH. Neurovascular proteases in brain injury,hemorrhage and remodeling after stroke.25th Princeton Conference onCerebrovascular Disease. Portland,2006,748~752
    34Ma Z, Shah RC, Chang MJ, et al. Coordination of cell signaling,chromatin remodeling, histone modifications, and regulator recruitment inhuman matrix metalloproteinase9gene transcription. Mol Cell Biol,2004,24:5496~5509
    35Ellerbroek SM, Halbleib JM, Benavidez M, et al. Phosphatidylinositol3-kinase activity in epidermal growth factor-stimulated matrixmetalloproteinase-9production and cell surface association. Cancer Res,2001,61:1855~1861
    36Liu JF, Crépin M, Liu JM, et al. FGF-2and TPA induce matrixmetalloproteinase-9secretion in MCF-7cells through PKC activation ofthe Ras/ERK pathway. Biochem Bioph Res Commun,2002,293:1174~1182
    37Suyama K, Shapiro I, Guttman M, et al. A signaling pathway leading tometastasis is controlled by N-cadherin and the FGF receptor. Cancer Cell,2002,2:301~314
    38Busti C, Falcinelli E, Momi S, et al. Matrix metalloproteinases andperipheral arterial disease. Intern. Emerg Med,2010,5:13~25
    39Coussens LM, Tinkle CL, Hanahan D, et al. MMP-9supplied by bonemarrow-derived cells contributes to skin carcinogenesis. Cell,2000,103:481~490
    40Ho TY, Bagnell CA. Relaxin induces matrix metalloproteinase-9throughactivation of nuclear factor kappa B in humanTHP-1cells. Ann N Y AcadSci,2005,104:314~316
    41Sun JT, Feng A., Zhang Y, et al. Fucoidan increases TNF-a-inducedMMP-9secretion in monocytic cell line U937. Inflamm Res,2010,59:271~276
    42Gidday JM, Gasche YG, Copin JC, et al. Leukocyte-derived matrixmetalloproteinase-9mediates blood-brain barrier breakdown and isproinflammatory after transient focal cerebral ischemia. Am J Physiol.Heart Circ Physiol,2005,289: H558~H568
    43Wang GM, Guo QM, Hossain M, et al. Bone marrowderived cells are themajor source of MMP-9contributing to blood–brain barrier dysfunctionand infarct formation after ischemic stroke in mice. Brain Res,2009,1294:183~192
    44Zhao X, Benveniste EN. Transcriptional Activation of human matrixmetalloproteinase-9gene expression by multiple Co-activators. J Mol Biol,2008,383:945~956
    45Candelario-Jalil E, Yang Y, Rosenberg GA. Diverse roles of matrixmetalloproteinases and tissue inhibitors of metalloproteinases inneuroinflammation and cerebral ischemia. Neuroscience,2009,158:983~994
    46Gu Z, Kaul M, Yan B, et al. S-nitrosylation of matrix metalloproteinases:Signaling pathway to neuronal cell death. Science,2002,297:1186~1190
    47Manabe S, Gu Z, Lipon SA. Activation of matrix metalloproteinase-9vianeuronal nitric oxide synthase contributes to NMDA-induced retinalganglion cell death. Invest. Ophthalmol. Vis Sci,2005,46:4747~4753
    48Dzwonek J, Rylski M, Kaczmarek L. Matrix metalloproteinases and theirendogenous inhibitors in neuronal physiology of the adult brain. FEBSLett,2004,567:129~135
    49Jiang X, Namura S, Nagata I. Matrix metalloproteinase inhibitorKB-R7785attenuates brain damage resulting from permanent focalcerebral ischemia in mice. Neurosci. Lett,2001,305:41~44
    50Machado LS, Kozak A, Ergul A, et al. Delayed minocycline inhibitsischemiaactivated matrix metalloprotein-ases2and9after experimentalstroke. BMC Neurosci,2006,7:56
    51Nagel S, Su Y, Horstmann S, et al. Minocycline and hypothermia forreperfusion injury after focal cerebral ischemia in the rat-Effects on BBBbreakdown and MMP expression in the acute and subacute phase. BrainRes,2008,1188:198~206
    52Lee H, Park JW, Kim SP, et al. Doxycycline inhibits matrixmetalloproteinase-9and laminin degradation after transient global cerebralischemia. Neurobiol Dis,2009,34:189~198
    53Burggraf D, Trinkl A, Dichgans M, et al. Doxycycline inhibits MMPs viamodulation of plasminogen activators in focal cerebral ischemia.Neurobiol Dis,2007,25:506~513
    54Guo RW, Yang LX, Wang H, et al. Angiotensin II induces matrixmetalloproteinase-9expression via a nuclear factor-kappaB-dependentpathway in vascular smooth muscle cells. Regul. Pept,2008,147:37~44
    55Hosomi N, Mizushige K, Kitadai M, et al. Induced hypertension treatmentto improve cerebral ischemic injury after transient forebrain ischemia.Brain Res,1999,835:188~196
    56Hosomi N, Nishiyama A, Ban CR, et al. Angiotensin type1receptorblockage improves ischemic injury following transient focal cerebralischemia. Neuroscience,2005,134:225~231
    57Liebetrau M, Burggraf D, Wunderlich N, et al. ACE inhibition reducesactivity of the plasminogen/plasmin and MMP systems in the brain ofspontaneous hypertensive stroke-prone rats. Neurosci. Lett,2005,376:205~209
    58Tanaka H, Takai S, Jin D, et al. Inhibition of matrix metalloproteinase-9activity by trandolapril after middle cerebral artery occlusion in rats.Hypertens. Res,2007,30:469~475
    59Candelario-Jalil E. Nimesulide as a promising neuroprotectant in brainischemia: New experimental evidences. Pharmacol. Res,2008,57:266~273
    60Candelario-Jalil E, Taheri S, Yang Y, et al. Cyclooxy genase inhibitionlimits blood-brain barrier disruption following intracerebralinjection oftumor necrosis factor-alpha in the rat. J. Pharmacol. Exp Ther,2007,323:488~498
    61Wang Y, Deng XL, Xiao XH, et al. A nonsteroidal anti-inflammatory agentprovides significant protection during focal ischemic stroke with decreasedexpression of matrix metalloproteinases. Curr Neurovasc Res,2007,4:176~183
    62Murono S, Yoshizaki T, Sato H, et al. Aspirin inhibits tumor cellinvasiveness induced by Epstein-Barr virus latent membrane protein1through suppression of matrix metalloproteinase-9expression. Cancer Res,2000,60:2555~2561
    63Ospina JA, Brevig HN, Krause DN, et al. Estrogen suppresses IL-1beta-mediated induction of COX-2pathway in rat cerebral blood vessels.Am. J. Physiol. Heart Circ. Physiol,2004,286: H2010~H2019
    64Liu R, Wen Y, Perez E, et al.17beta-Estradiol attenuates blood-brainbarrier disruption induced by cerebral ischemia-reperfusion injury infemale. Brain Res,2005,1060:55~61
    65Gao D, Zhang X, Jiang X, et al. Resveratrol reduces the elevated level ofMMP-9induced by cerebral ischemia-reperfusion in mice. Life Sci,2006,78:2564~2570
    66Woo JH, Lim JH, Kim YH, et al. Resveratrol inhibits phorbol myristateacetate-induced matrix metalloproteinase-9expression by inhibiting JNKand PKC delta signal transduction. Oncogene,2004,23:1845~1853
    67Zheng YQ, Liu JX, Wang JN, et al. Effects of crocin onreperfusion-induced oxidative/nitrative injury to cerebral microvesselsafter global cerebral ischemia. Brain Res,2007,1138:86~94
    68Cho JY, Kim IS, Jang Y, et al. Protective effect of quercetin, a naturalflavonoid against neuronal damage after transient global cerebral ischemia.Neurosci. Lett,2006,404:330~335
    69Tsubokawa T, Solaroglu I, Yatsushige H, et al. Cathepsin and calpaininhibitor E64d attenuates matrix metalloproteinase-9activity after focalcerebral ischemia in rats. Stroke,2006,37:1888~1894
    70Haddad M, Beray-Berthat V, Cogueran B, et al. Reduction of hemorrhagictransformation by PJ34, a poly(ADP-ribose)polymerase inhibitor, afterpermanent focal cerebral ischemia in mice. Eur J Pharmacol,2008,588:52~57
    71Gómez-Hernández A, Sánchez-Galán E, Ortego M, et al. Effect ofintensive Atorvastatin therapy on prostaglandin E2levels andmetalloproteinase-9activity in the plasma of patients with non-Stelevationacute coronary syndrome. Am J Cardiol,2008,102:12~18
    72Tziakas DN, Chalikias GK, Parissis JT, et al. Serum profiles of matrixmetalloproteinases and their tissue inhibitor in patients with acute coronarysyndromes. The effects of short-term atorvastatin administration. Int JCardiol,2004,94:269~277
    73Xu Z, Zhao S, Zhou H, et al. Atorvastatin lowers plasma matrixmetalloproteinase-9in patients with acute coronary syndrome. Clin Chem,2004,50:750~753
    74F erster C, Kahles T, Kietz S, et al. Dexamethasone induces theexpression of metalloproteinase inhibitor TIMP-1in the murine cerebralvascular endothelial cell line cEND. J Physiol,2007,580:937~949
    75Rush TS, Powers R. The application of X-ray, NMR, and molecularmodeling in the design of MMP inhibitors. Curr Top.Med Chem,2004,4:1311~1327
    76Lo EH. A new penumbra: transitioning from injury into repair after stroke.Nat Med,2008,14:497~500

© 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