莲房原花青素对老年认知障碍大鼠记忆功能的改善作用及其机制研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
正常老化是一个独立于病理条件下的正常而复杂的生物学过程。在衰老过程中,机体的许多系统都出现各种进行性的结构和功能的改变。在神经系统,与年龄增加相关的最主要的衰退之一是认知功能障碍。认知功能是人类与感知、思维、记忆、学习、想象和推理等有关的信息处理能力,是人类正常生存的必需能力和智能发展和维持的基础。人类和动物的老化都导致认知障碍。学习记忆能力作为认知功能的一部分,大量研究已经证实人和动物学习和记忆能力都存在增龄性衰退现象。这种年龄相关性的记忆损害在从正常认知到轻度认知障碍(MCI, mild cognitive impairment)再到老年痴呆(AD, Alzheimer's disease)的整个过程中都可能发生。虽然这种年龄相关性的记忆损害对生活质量的影响没有AD那样严重,但其在老年人群中的发病率非常高。研究表明有38%的年龄在60-78岁的老年人符合年龄相关性记忆障碍(AAMI, age associated memory impairment)的标准,并且9%的AAMI病人将发展为痴呆,而诊断为MCI的个体中55%的人在4.5年内发展为痴呆。因此年龄相关性记忆损害是影响老年生活质量的和生活乐趣的一个关键因素,有必要寻找安全而又切实可行的防治措施。为了解释年龄相关性记忆障碍的发生机制,人们提出了各种各样的假说,这其中,老化的胆碱能假说、老化的自由基假说、老化的一氧化氮假说是被广泛接受的解释。另外,老化过程中脑CREB介导的转录受损也越来越引起人们的注意。
     原花青素是一类多酚类物质,它们是由黄烷-3-醇单体组成的低聚物或多聚物,这些物质广泛分布于水果、蔬菜、种子、花以及树皮中。由于它们具有极强的抗氧化能力和极高的安全性,现在已经广受关注。原花青素清除自由基的能力明显优于维生素C、维生素E和β-胡萝卜素。除此之外,这些物质还具有抗菌、抗病毒、抗炎以及抗癌等多种功能。原花青素的代谢产物能够穿过血脑屏障而可被脑组织摄取。由于在脑中可以检测到这些代谢物的存在,因而其可能对脑功能发挥潜在的调节作用。原花青素在脑中的有益生物学作用一般认为是基于其优良的抗氧化能力。例如,以往的研究证实原花青素可抑制脑中的脂质过氧化和年龄相关性的DNA氧化损伤的蓄积。现在越来越多的证据表明原花青素所具有的其它一些调节机制可能也参与了对脑功能的调节作用。例如,蛋白组学研究表明原花青素能够系统地调节年轻大鼠脑中某些特定蛋白质的表达和活性;而(-)-表儿茶素则被发现可上调含有GluR2的AMPA受体,而上调此受体意味着对突触功能的潜在调节作用。
     为了探讨莲房原花青素(LSPC, procyanidins from the lotus seedpod)对AAMI的改善作用以及作用机制,我们首先确定LSPC对东莨菪碱所致记忆障碍模型小鼠和老年认知障碍大鼠记忆能力是否具有改善作用,然后从自由基假说、胆碱能假说、一氧化氮假说和CREB改变几个方面探讨LSPC改善老年认知障碍大鼠记忆能力的可能机制。
     第一部分行为学研究
     第一节LSPC对东莨菪碱所致记忆障碍小鼠模型学习记忆能力的影响
     目的确定LSPC对东莨菪碱所致记忆障碍小鼠学习记忆的改善作用。
     方法2月龄雄性昆明种小鼠50只(18-22 g)被随机分为5组(每组10只):对照组、低中高剂量组(L-、M-、-H-LSPC)和东莨菪碱组。L-、M-、H-LSPC组的小鼠分别给予50,100,150 mg/kg BW LSPC灌胃30 d,对照组和东莨菪碱组灌胃等量的蒸馏水。在进行行为学测试期间,L-、M-、H-LSPC组小鼠每天在第一次测试前1h灌胃LSPC, L-、M-、H-LSPC组和东莨菪碱组小鼠每天在第一次测试前10 min腹腔注射东莨菪碱(1mg/kg),对照组小鼠腹腔注射生理盐水。用Morris水迷宫和跳台试验评估小鼠学习记忆能力。在Morris水迷宫测试中,记录每次训练的潜伏期和游泳距离。跳台试验在Morris水迷宫结束7 d后进行,记录错误次数和潜伏期。跳台试验完成后,将小鼠处死并将脑匀浆进行AChE检测。
     结果三个剂量的LSPC处理组,除了L-LSPC组第1 d的潜伏期明显高于对照组外,其它时间和剂量的LSPC组的潜伏期与对照组相当。所有LSPC剂量组在第4和第5 d的潜伏期均明显少于东莨菪碱组;所有LSPC剂量组在第4和第5 d的游泳距离均明显少于东莨菪碱组并与对照组没有显著性差异。在跳台试验中,所有LSPC剂量组的错误次数均明显低于东莨菪碱组,同时所有LSPC剂量组的潜伏期均明显低于对照组,但是又都显著高于东莨菪碱组。LSPC剂量依赖性地抑制AChE活性,所有LSPC剂量组的AChE活性均明显低于东莨菪碱组并与对照组相比无显著性差异。
     结论通过Morris水迷宫和跳台试验评估,LSPC可改善东莨菪碱所致记忆障碍小鼠模型的学习记忆能力,其抑制AChE活性而增加胆碱能活性的作用可能参与了这一过程。
     第二节LSPC对老年认知障碍大鼠学习记忆能力的影响
     目的确定LSPC对老年认知障碍大鼠学习记忆能力的改善作用。
     方法以40只年轻雌性大鼠在Morris水迷宫中的潜伏期为基础,从200只18月龄雌性SD大鼠中筛选出老年认知正常(AU)大鼠和老年认知障碍(AI)大鼠。从年轻大鼠中随机挑选14只大鼠组成年轻组;从AU大鼠中随机挑选16只大鼠组成AU组;同时将AI大鼠随机分为3组(每组16只):AI组和低、高LSPC(L-、H-LSPC)剂量组。L-、H-LSPC组大鼠每日分别灌胃50和100 mg/kg BW LSPC,其它三组大鼠灌胃等量生理盐水。喂养周期为7 w。在其后进行的Morris水迷宫测试期间,所有动物在每日第一次训练前1 h继续进行相同处理。用与筛选时完全不同参照体系的Morris水迷宫试验评估各组大鼠的学习记忆能力。
     结果与年轻组和AU组相比,AI组大鼠在Morris水迷宫中的潜伏期明显延长。除H-LSPC组在训练的第1 d与AI组差异无显著性外,两个LSPC剂量组在测试期间的潜伏期均明显少于AI组。与年轻组和AU组相比,H-LSPC组的潜伏期在整个测试期间没有显著性差异。在训练的第3-5 d,H-LSPC组的游泳距离明显少于AI组;在训练的第2和第4d, L-LSPC组的游泳距离也明显少于AI组。在整个训练期间,H-LSPC组的游泳距离与年轻组和AU组相比均未见显著差异。
     结论LSPC可明显改善AI大鼠的空间学习记忆能力。
     第二部分机制研究
     第三节抗氧化和胆碱能系统的改变促进LSPC改善老年认知障碍大鼠学习记忆能力的机制研究
     目的探讨LSPC对AI大鼠海马和皮质抗氧化和胆碱能系统的作用。
     方法实验设计同第一部分第二节。将完成Morris水迷宫测试的动物处死后分离海马和皮质。每组随机挑选4只大鼠样本进行后续研究。对于剩下的样本,左侧海马和皮质匀浆后进行抗氧化和氧化损伤指标以及ChAT、AChE活性的检测。右侧海马和皮质用含有3 mg/ml毒扁豆碱的磷酸盐缓冲液匀浆后进行ACh检测。
     结果所有实验组大鼠海马和皮质的SOD活性均未见明显改变。两个老年组的海马和皮质CAT和GPx活性均显著低于年轻组,然而这两种酶在AU和AI组之间没有发现明显差异。与AI组相比,CAT、GPx活性在H-LSPC海马和皮质中明显增加,在L-LSPC组海马中也显著增加。在AU和AI组,海马和皮质的CAT/SOD和GPx/SOD比值均明显降低;与AI组相比,LSPC,特别是H-LSPC可逆转CAT/SOD和GPx/SOD比值的降低。AI和AU大鼠海马和皮质中GSH水平明显低于年轻大鼠,H-LSPC明显上调这两个部分的GSH水平。AU组和AI组海马和皮质的总抗氧化能力(T-AOC)均显著下降,而两个LSPC剂量组的T-AOC水平均明显升高并与年轻组无显著性差异。AU组和AI组海马和皮质MDA水平和蛋白羰基含量均明显高于年轻组。AU组和AI组之间的海马和皮质MDA水平未见显著性差异,然而AI组海马和皮质的蛋白羰基含量明显高于AU组。与AI组相比,两个LSPC剂量组海马和皮质中MDA水平和蛋白羰基含量均明显降低。各个实验组海马和皮质中ChAT活性均未见明显改变。AU和AI组大鼠海马AChE活性均显著低于年轻大鼠;AI组大鼠皮质AChE活性明显低于年轻大鼠,而AU组大鼠皮质AChE活性与年轻大鼠没有显著性差异。H-LSPC组海马AChE活性以及L-、H-LSPC组皮质AChE活性均明显高于AI组。与年轻组和AU组大鼠相比,AI组大鼠的海马和皮质ACh含量显著下降。LSPC可剂量依赖性地增加ACh含量,并且两个LSPC剂量组大鼠的海马和皮质ACh含量均与年轻组大鼠相当。
     结论LSPC改善AI大鼠海马和皮质抗氧化系统的功能。降低脑的氧化损伤和增加胆碱能系统功能参与了LSPC改善AI大鼠空间学习记忆能力的作用。
     第四节一氧化氮系统改变促进LSPC改善老年认知障碍大鼠学习记忆能力的机制研究
     目的探讨海马和皮质一氧化氮系统改变对LSPC改善AI大鼠学习记忆能力的影响。
     方法实验设计以及组织样品的准备同第二部分第三节。每组随机挑选4只大鼠进行实时定量PCR和western blot检测。其余样品的部分匀浆用于NO水平以及总NOS和分型NOS的检测。为了评估在蛋白酶酶抑制剂或磷酸酶抑制剂存在的情况下NOS活性,向匀浆中加入蛋白激酶抑制剂或磷酸酶抑制剂,然后用HEPES终止反应,再进行iNOS活性的检测。
     结果AI大鼠海马和皮质NO水平明显高于年轻大鼠。所有实验组的海马和皮质tNOS活性改变均与其NO水平的改变相平行。AI大鼠海马iNOS活性明显高于AU组和年轻组,同时AU大鼠海马iNOS活性与年轻大鼠没有明显差异。另外,虽然AU和AI大鼠海马:iNOS活性均明显降低,但是AI大鼠又比AU大鼠进一步明显降低。虽然AU和AI大鼠皮质iNOS活性均大幅显著增加同时nNOS活性明显降低,但是AU和AI大鼠皮质iNOS以及nNOS活性并没有明显差异。iNOS和nNOS的基因和蛋白表达也有相似的变化。用蛋白激酶抑制剂或磷酸酶抑制剂处理后,年轻组nNOS活性改变率变化最大,AU组也有相当大幅度的改变;然而这些抑制剂对AI大鼠海马nNOS活性改变率的影响则很小。LSPC明显降低AI大鼠海马和皮质iNOS活性并因此导致tNOS活性和NO水平降低。虽然LSPC并没有明显改变AI大鼠海马和皮质nNOS活性,但是却有增加AI大鼠海马nNOS基因和蛋白表达的趋势。因此,LSPC剂量组大鼠海马的单位nNOS蛋白活性降低。另外,虽然LSPC在蛋白激酶抑制剂存在的情况下并没有明显改变AI大鼠的海马nNOS活性,但却在磷酸酶抑制剂存在的情况下明显改变AI大鼠海马nNOS活性。结论AI大鼠认知功能的降低主要是海马iNOS和nNOS的表达和活性改变所致,而iNOS和nNOS在皮质中的改变则不是主要因素。LSPC不但降低AI大鼠海马iNOS的表达和活性,而且也可使海马nNOS磷酸化调节更加有效,从而抑制年龄相关性的NO过表达、增加海马NO-sGC-cGMP信号转导。表明LSPC可能通过海马NO系统的上述改变而改善AI大鼠的认知功能。
     第五节CREB磷酸化改变促进LSPC改善老年认知障碍大鼠学习记忆能力的机制研究
     目的探讨海马和皮质中CREB磷酸化改变对LSPC改善AI大鼠学习记忆能力的影响。方法实验设计以及组织样品的准备同第二部分第三节。每组随机挑选4只大鼠样本进行BDNFmRNA的实时定量PCR检测和CREB、pCREB、BDNF、ERK 42/44、pERK 42/44、CaMKIV、pCaMKIV和PKA C-a的western blot检测。
     结果AI大鼠海马CREB磷酸化程度明显低于年轻大鼠与AU大鼠;LSPC可剂量依赖性地明显增加AI大鼠海马CREB磷酸化程度。各个实验组的皮质CREB磷酸化程度没有明显差异。AI大鼠海马BDNF mRNA和蛋白表达水平明显低于年轻大鼠和AU大鼠;两个剂量的LSPC均明显增加AI大鼠海马BDNF mRNA和蛋白表达水平。各个实验组大鼠皮质BDNF mRNA和蛋白表达水平未见显著差异。AI大鼠海马ERK42、ERK44蛋白磷酸化程度显著低于年轻大鼠和AU大鼠。相对于AI组,L-LSPC组大鼠海马ERK42蛋白磷酸化程度有增加的趋势,H-LSPC组大鼠海马ERK42蛋白磷酸化程度在L-LSPC剂量组的基础上又明显增加,并与年轻组没有明显差异。两个LSPC剂量组海马ERK44蛋白磷酸化程度都明显高于AI组,并且与AU组相当。AI大鼠海马CaMKIV磷酸化状态显著低于年轻大鼠和AU大鼠;补充LSPC后没有改变海马的CaMKIV磷酸化状态。所有实验组动物海马PKA C-a水平均没有显著差异。
     结论相对于年轻大鼠和AU大鼠,AI大鼠海马CREB磷酸化程度明显降低,然而其皮质CREB磷酸化程度没有明显改变。AI大鼠海马ERK 42/44和CaMKIV磷酸化程度降低都促进其CREB活性的降低。海马CREB活性降低导致CREB依赖性的转录降低,从而损害长期记忆的形成。例如,AI大鼠海马BDNF mRNA和蛋白水平明显降低。这些结果表明海马CREB活性降低明显参与了年龄相关性老年记忆障碍的形成和发展过程。LSPC可通过ERK介导的通路活化海马CREB,从而易化长期记忆的形成过程,并因此改善AI大鼠的学习记忆能力。
Aging is a normal but complicated, not pathological, process, which characterized by slowly progressing impairments in structure and function of most systems. In central nervous system (CNS), age-related cognitive dysfunction is one of the most prominent features. Cognitive function refers to a person's ability to process information, such as perception, thinking, recognition, sensing, learning, memory, imagining, and reasoning, and which is necessary for survival as well as intellectual development and maintenance. As the natural process of aging progresses, humans and animals experience a progressive decline in overall cognitive function. As an integral part of cognitive function, learning and memory abilities have been demonstrated to marked decline in the course of normal aging. The age associated memory impairment (AAMI) may range from normal to a mild cognitive impairment, or to Alzheimer's disease (AD). Although its passive impact on quality of life is not as severe as AD, the age associated memory impairment is very prevalent in elderly people. It has been shown that thirty-eight percent of 60-78 years old individuals fulfill the criteria for AAMI, and in the follow-up nine percent of AAMI patients became demented. In addition, during a follow-up period of four and a half years fifty-five percent of individuals with the diagnosis of mild cognitive impairment developed dementia. Therefore, AAMI is one of crucial determinants of the quality and enjoyment of life of elderly people, and it is necessary to seek for some measures for prevention and treatment AAMI with little or not damage to body. Various hypothesizes have been proposed to try to explain the mechanisms of geriatric memory dysfunction. Among them, the cholinergic hypothesis of geriatric memory dysfunction, the free radical hypothesis of aging and the nitric oxide hypothesis of aging have been most widely accepted. In addition, the impaired CREB-mediated transcription in brain during aging has also been paid more and more attention.
     Procyanidins chemically belong to polyphenols, they are oligomeric and polymeric flavonoids comprised of flavan-3-ol monomeric subunits. These compounds are widely distributed in fruits, vegetables, seeds, flowers and bark. They have received considerable attention in recent years due to their excellent antioxidant activity and a broad spectrum of safety. Procyanidins posses significantly better free radical scavenging capability than vitamin C, E orβ-carotene. In addition, these compounds have also been reported to exhibit a wide range of biological effects including antibacterial, antiviral, anti-inflammatory, and anticarcinogen actions. The metabolites of procyanidins are able to cross the blood brain barrier and be uptaken and detected in brain and thus exert potential effects on brain functions. Beneficial bioactivities of procyanidins in brain are often claimed to be based on their antioxidant properties. For example, previous experiments demonstrated that procyanidins had an inhibiting effect on the lipid peroxidation and accumulation of age-related oxidative DNA damage in brain. But increasing evidence suggests that some other mechanisms may involve the modulation of brain functions. A proteomics study indicated that procyanidins were able to systematically change specific brain proteins in expression or modification in young rats. (-)-Epicatechin has been found to up-regulate GluR2-containing AMPA receptors, which means it has the potential to modulate synaptic function.
     In order to explore the effects of procyanidins extracted from the lotus seedpod (LSPC) on AAMI, the primary goal of this research is to find whether LSPC ameliorate memory impairment in scopolamine-induced mice and cognitively impaired aged rats. When the positive results were observed, we further explored the mechanisms which refer to cholinergic hypothesis, the free radical hypothesis, nitric oxide hypothesis and changes in CREB-mediated transcription.
     PartⅠBehavioral study
     Section one LSPC ameliorate scopolamine-induced memory impairment in mice
     Objective To determine the ameliorating effect of LSPC on the learning and memory impairments induced by scopolamine in mice.
     Methods Fifty two-month old male Kunming mice (18-22 g) were randomly divided into five groups with 10 mice in each group:the control group, the low, middle and high dose LSPC (L-, M-, and H-LSPC) groups and the scopolamine group. Mice in L-, M-, and H-LSPC groups were given 50,100,150 mg/kg BW LSPC orally by a gavage daily for 30 days respectively. The control group and scopolamine group were given an equivalent amount of distilled water daily by oral gavage. On each day of the behavioral tests period, mice in L-, M-, and H-LSPC groups were given LSPC 1 h before the first trial session. Animals in L-, M-, and H-LSPC groups and scopolamine group were treated with scopolamine (1 mg/kg, i.p) dissolved in normal saline at 10 min before the first trial session during the training phase of both behavioral tests. Mice in the control group received vehicle only. The capacities of memory and learning were evaluated by the Morris water maze and the step-down avoidance test. During each trial session in Morris water maze, escape latency and swimming distance were recorded. The step-down avoidance test was performed 7 days after the Morris water maze, and the numbers of errors and step-down latency were recorded. After completion of step-down avoidance test, the mice were killed and the brains were homogenized to determine the activity of AChE.
     Results No difference in escape latency was observed among the LSPC groups and the control group during all the training days only except that L-LSPC group had significantly longer escape latency relative to the control groups on day 1. Escape latency of all the three LSPC groups on day 4 and 5 were significantly shorter compared to that of scopolamine group. Mice in three LSPC groups on day 4 and 5 exhibited significantly shorter swimming distances compared with that in scopolamine group, but no differences were observed when compared with that in control group. In step-down avoidance test, reduced the number of errors in all the three LSPC groups were observed compared with scopolamine group. In addition, all the three LSPC groups had significant shorter latencies than the control group and significantly longer latencies than scopolamine group. LSPC inhibited AChE activity in a dose-dependent manner, and no significant difference was found among all the three LSPC groups and the control group.
     Conclusion LSPC has the ability to ameliorate the impairment of learning and memory induced by scopolamine in both Morris water maze test and step-down avoidance test, and increasing cholinergic activity by inhibition of AChE activity may involve in the ameliorative activities of LSPC on learning and memory dysfunction.
     Section two LSPC ameliorate memory impairment in cognitively impaired aged rats
     Objective To determine the ameliorating effect of LSPC on the learning and memory impairments in cognitively impaired aged rats.
     Methods Based on Morris water maze performance compared with forty young female rats, aged-unimpaired (AU) and aged-impaired (AI) rats were chosen from two hundred 18-month-old female Sprague-Dawley rats. The rats in young control group (n=14) and AU group (n=16) were randomly selected from the young rats and aged unimpaired rats respectively. The animals categorized as AI were randomly subdivided into three groups consisting of 16 animals each:AI group, low and high dose LSPC (L-and H-LSPC) groups. Rats in L-and H-LSPC groups were given 50 and 100 mg/kg BW LSPC orally by a gavage daily for 7 weeks, respectively. The other three groups were given orally an equivalent volume of distilled water daily. On each day of the behavioral tests period in Morris water maze, animals continued to be administered intragastrically 1 h before the first trial session. The learning and memory abilities of rats in different groups were assessed by retraining the animals in Morris water maze with entirely different cues and testing the animals as before.
     Results Rats in AI group exhibited a significant prolongation of escape latency in Morris water maze compared with those of young group and AU group. Both LSPC groups exhibited significantly shorter escape latencies than AI group during all sessions only except H-LSPC group on the first training day. Meanwhile, H-LSPC group did not exhibit significant difference in escape latency from young and AU groups. Significantly shorter swimming distance than that of AI group was observed in H-LSPC group on day 3-5 and L-LSPC group on day 2 and day 4. Compared with young and AU groups, H-LSPC group did not exhibit difference in swimming distance during all the test days.
     Conclusion LSPC has the ability to ameliorate the impairment of learning and memory in AI animals in Morris water maze test.
     Part II Mechanism study
     Section three Rejuvenation of antioxidant and cholinergic systems contributes to the effect of LSPC ameliorating memory impairment in cognitively impaired aged rats
     Objective To determine the effect of LSPC on the antioxidant and cholinergic systems in hippocampus and cerebral cortex in cognitively impaired aged rats.
     Methods Study design was the same as part I, section two. After completion of water maze test, animals were killed and the hippocampus and cerebral cortex were dissected. Four animal samples in each group were randomly selected for next study. For the remaining samples, hippocampus and cerebral cortex in left hemisphere were homogenized to determine the antioxidant capacity, oxidative damage markers as well as activities of ChAT and AChE. The hippocampus and cerebral cortex in right hemisphere were homogenized in phosphate buffer containing 3 mg/ml eserine for ACh assay.
     Results There were not significant changes in SOD activity in hippocampus and cerebral cortex in all the experimental groups. However, CAT and GPx activities of the both brain regions of old animals were significantly lower than those of young subjects, but no difference in activities was observed between AU and AI groups. Significant increase in CAT and GPx activities of both brain regions in H-LSPC group and in GPx activities of hippocampus in L-LSPC group were observed when compared with the AI group. The ratios of CAT/SOD and GPx/SOD were significantly lower in AU and AI groups. LSPC, especially H-LSPC, reversed the declines in these ratios when compared with AI. The levels of GSH in both brain regions of aged rats were significantly lower than those of young animals, and H-LSPC administration up-regulated significantly the GSH level in both regions. Hippocampal and cortical T-AOC in AU and AI groups markedly declined, and animals in both LSPC groups revealed higher T-AOC than that in AU and AI groups and were similar to their young counterparts. All AU and AI animals showed significant higher MDA levels and carbonyl content in hippocampus and cerebral cortex than young animals. There were not significant differences in MDA levels of both brain regions between AU and AI groups, whereas AI animals exhibited significantly higher carbonyl content in both brain regions than AU animals. Reduced MDA levels and carbonyl content in hippocampus and cerebral cortex in both LSPC groups were observed compared with AI group. There were not significant differences in ChAT activity in hippocampus and cerebral cortex in all the experimental groups. Animals in both AU and AI groups showed significantly lower AChE activities in hippocampus than their young counterparts. In cerebral cortex, animals in AI group also revealed significantly lower AChE activities than young animals, whereas AU animals were not significantly different from young animals. The AChE activities in hippocampus of H-LSPC group and in cerebral cortex of both LSPC groups were significantly higher than that in AI group. There was a substantial decrease in ACh content in both brain regions for AI group compared with young and AU groups. LSPC considerably increased ACh content in a dose-dependent manner, and no significant difference was found among both LSPC groups and the young group.
     Conclusion LSPC attenuated functional decline in antioxidant system in hippocampus and cerebral cortex of AI rats. The decreased oxidative damage to brain and increasing cholinergic activity may involve in the ameliorative effects of LSPC on learning and memory dysfunction in AI animals.
     Section four Changes in nitric oxide system contribute to the effect of LSPC ameliorating memory impairment in cognitively impaired aged rats
     Objective To determine the effects of LSPC on the nitric oxide system in hippocampus and cerebral cortex in cognitively impaired aged rats.
     Methods Study design and tissue preparation were the same as partⅡ, section three. Four animal samples in each group were randomly selected for real-time PCR and western blot analysis. For the remaining animal samples, part of the homogenate was used for NO level as well as total and NOS isoforms activities evaluation. In order to determination of the effect of phosphorylation/dephosphorylation on nNOS activity, homogenate were preincubated with protein kinase inhibitors cocktail or phosphatase inhibitor and terminated with HEPES, then nNOS activity were measured.
     Results The hippocampal and cortical NO levels in AI animals were markedly higher than that in young animals. The alterations in tNOS activity of both brain regions closely paralleled the changes in NO level of that in all the experimental groups. AI rats exhibited significantly higher hippocampal iNOS activities than AU and young animals, and no differences in hippocampal iNOS activity were observed between young and AU animals. In addition, although marked decrease in hippocampal nNOS activity occurred in all AU and AI rats, AI animals exhibited further significant lower hippocampal nNOS activities than AU animals. However, although cortical iNOS activities were increased by a large margin and at the same time nNOS activities were reduced in all AU and AI animals, there was no differences in both iNOS and nNOS activities between AI and AU animals. Similar alteration trends were also observed in terms of the gene and protein expressions in both iNOS and nNOS. When treated with protein kinase inhibitors or phosphatase inhibitor, although young group revealed the most alteration in the rate of nNOS activity change, AU group also had considerable changes in that. However, the effects of these inhibitors on the rates of nNOS activity change were almost abrogated in hippocampus of AI animals. Both doses of LSPC significantly decreased iNOS activities and consequently resulted in tNOS activities and NO levels decrease in hippocampus and cerebral cortex of AI animals. LSPC supplementation did not lead to marked alterations for hippocampal and cortical nNOS activity in AI animals, whereas AI animals revealed a trend to increase nNOS gene and protein expressions in hippocampus with LSPC supplementation. As a result, animals in LSPC supplementation groups should have a decline in nNOS activity per unit protein in hippocampus. Moreover, although LSPC supplementation had not marked effect on the rates of hippocampal nNOS activity change in the presence of protein kinase inhibitors in AI animals, these compounds considerably altered these rates in these animals in the presence of phosphatase inhibitor.
     Conclusion The alterations of expressions and activities in hippocampal but not cortical iNOS and nNOS accounted for the reduced cognitive behaviors in AI animals. LSPC supplementation not only decreased hippocampal iNOS activities and enzyme expression but also made hippocampal nNOS phosphorylation regulated effectively in AI animals, which led to the inhibition of age-related NO overproduction and increase of hippocampal NO-sGC-cGMP signal transduction in these animals. These changes in hippocampal NO system may involve, at least partly, in the ameliorative effects of LSPC on learning and memory dysfunction in AI animals.
     Section five Memory impairment in cognitively impaired aged rats associated with decreased hippocampal CREB phosphorylation: reversal by LSPC
     Objective To determine the effects of LSPC on CREB phosphorylation in hippocampus and cerebral cortex in cognitively impaired aged rats.
     Methods Study design and tissue preparation were the same as part II, section three. Four animal samples in each group were randomly selected for real-time PCR of BDNF mRNA and western blot analysis of CREB, pCREB, BDNF, ERK 42/44, pERK 42/44, CaMKIV, pCaMKIV and PKA C-α.
     Results The phosphorylation states of CREB in hippocampus of AI animals were markedly lower than that of young and AU animals. Supplementation of LSPC led to significant increase in hippocampal CREB phosphorylation with a dose-dependent manner. All groups were not significantly different from each other in cortical CREB phosphorylation. The BDNF mRNA and protein levels were markedly reduced in hippocampus of AI animals compared to that measured in young and AU animals. AI animals with LSPC supplementation uniformly exhibited significantly higher hippocampal BDNF mRNA and protein expression than AI animals. As for the BDNF mRNA and protein expression in cerebral cortex, no significant changes were detected in any of the groups. The significant lower states of phosphorylation in hippocampal ERK42 and ERK44 protein were observed in AI animals relative to young and AU animals. Animals in group with L-LSPC supplementation had a tendency to elevate the phosphorylation states of hippocampal ERK42 relative to AI rats, and rats in group with H-LSPC supplementation revealed further significant increase in that and were comparable with young animals. The phosphorylation states of hippocampal ERK44 in both LSPC groups were substantially increased than that in AI group and reach to the level of AU group. Substantial decrease in phosphorylation states of CaMKIV were detected in AI animals relative to young and AU counterparts. The phosphorylation states of this protein did not significantly increase in response to LSPC supplementation. No substantial alterations in the level of PKA C-a were detected among all the experimental groups.
     Conclusion The alterations of expressions and activities in hippocampal but not cortical iNOS and nNOS accounted for the reduced cognitive behaviors in AI animals. LSPC supplementation not only decreased hippocampal iNOS activities and enzyme expression but also made hippocampal nNOS phosphorylation regulated effectively in AI animals, which led to the inhibition of age-related NO overproduction and increase of hippocampal NO-sGC-cGMP signal transduction in these animals. These changes in hippocampal NO system may involve, at least partly, in the ameliorative effects of LSPC on learning and memory dysfunction in AI animals.
引文
1. Koivisto K, Reinikainen KJ, Hanninen T, et al. Prevalence of age-associated memory impairment in a randomly selected population from eastern Finland. Neurology,1995,45(4):741-747.
    2. Hanninen T, Hallikainen M, Koivisto K, et al. A follow-up study of age-associated memory impairment:neuropsychological predictors of dementia Journal of the American Geriatrics Society,1995,43(9):1007-1015.
    3. Halliwell B. Oxidants and the central nervous system:some fundamental questions. Is oxidant damage relevant to Parkinson's disease, Alzheimer's disease, traumatic injury or stroke? Acta Neurologica Scandinavica. Supplementum,1989,126:23-33.
    4. Francois C, Nguyen-Legros J, and Percheron G. Topographical and cytological localization of iron in rat and monkey brains. Brain Research,1981, 215(1-2):317-322.
    5. Kedar NP. Can we prevent Parkinson's and Alzheimer's disease? Journal of Postgraduate Medicine,2003,49(3):236-245.
    6. Gonenc S, Uysal N, Acikgoz 0, et al. Effects of melatonin on oxidative stress and spatial memory impairment induced by acute ethanol treatment in rats. Physiological Research,2005,54(3):341-348.
    7. Coyle JT, Price DL, and DeLong MR. Alzheimer's disease:a disorder of cortical cholinergic innervation. Science,1983,219(4589):1184-1190.
    8. Gibson GE, Peterson C, and Jenden DJ. Brain acetylcholine synthesis declines with senescence. Science,1981,213(4508):674-676.
    9. Wurtman RJ. Choline metabolism as a basis for the selective vulnerability of cholinergic neurons. Trends in Neurosciences,1992,15(4):117-122.
    10. Bartus RT, Dean RL,3rd, Beer B, et al. The cholinergic hypothesis of geriatric memory dysfunction. Science,1982,217(4558):408-414.
    11. Bartus RT. On neurodegenerative diseases, models, and treatment strategies:lessons learned and lessons forgotten a generation following the cholinergic hypothesis. Experimental Neurology,2000,163(2):495-529.
    12. Calabrese V, Mancuso C, Calvani M, et al. Nitric oxide in the central nervous system:neuroprotection versus neurotoxicity. Nature Reviews. Neuroscience,2007, 8(10):766-775.
    13. Guix FX, Uribesalgo I, Coma M, et al. The physiology and pathophysiology of nitric oxide in the brain. Progress in Neurobiology,2005,76(2):126-152.
    14. Dinerman JL, Dawson TM, Schell MJ, et al. Endothelial nitric oxide synthase localized to hippocampal pyramidal cells:implications for synaptic plasticity. Proceedings of the National Academy of Sciences of the United States of America, 1994,91(10):4214-4218.
    15. Contestabile A, Monti B, and Ciani E. Brain nitric oxide and its dual role in neurodegeneration/neuroprotection:understanding molecular mechanisms to devise drug approaches. Current Medicinal Chemistry,2003,10(20):2147-2174.
    16. Prast H and Philippu A. Nitric oxide as modulator of neuronal function. Progress In Neurobiology,2001,64(l):51-68.
    17. Edwards TM and Rickard NS. New perspectives on the mechanisms through which nitric oxide may affect learning and memory processes. Neuroscience and Biobehavioral Reviews,2007,31 (3):413-425.
    18. Susswein AJ, Katzoff A, Miller N, et al. Nitric oxide and memory. Neuroscientist, 2004,10(2):153-162.
    19. Moncada S and Bolanos JP. Nitric oxide, cell bioenergetics and neurodegeneration. Journal of Neurochemistry,2006,97(6):1676-1689.
    20. Zanelli SA, Trimmer PA, and Solenski NJ. Nitric oxide impairs mitochondrial movement in cortical neurons during hypoxia Journal of Neurochemistry,2006, 97(3):724-736.
    21. Silva AJ, Kogan JH, Frankland PW, et al. CREB and memory. Annual Review of Neuroscience,1998,21:127-148.
    22. Bourtchuladze R, Frenguelli B, Blendy J, et al. Deficient long-term memory in mice with a targeted mutation of the cAMP-responsive element-binding protein. Cell, 1994,79(1):59-68.
    23. Impey S, Mark M, Villacres EC, et al. Induction of CRE-Mediated Gene Expression by Stimuli That Generate Long-Lasting LTP in Area CA1 of the Hippocampus. Neuron,1996,16(5):973-982.
    24. Mizuno M, Yamada K, Maekawa N, et al. CREB phosphorylation as a molecular marker of memory processing in the hippocampus for spatial learning. Behavioural Brain Research,2002,133(2):135-141.
    25. Guzowski JF and McGaugh JL. Antisense oligodeoxynucleotide-mediated disruption of hippocampal cAMP response element binding protein levels impairs consolidation of memory for water maze training. Proceedings of the National Academy of Sciences of the United States of America,1997,94(6):2693-2698.
    26. Yin JC, Del Vecchio M, Zhou H, et al. CREB as a memory modulator:induced expression of a dCREB2 activator isoform enhances long-term memory in Drosophila Cell,1995,81(1):107-115.
    27. Carlezon WA, Jr., Duman RS, and Nestler EJ. The many faces of CREB. Trends in Neurosciences,2005,28(8):436-445.
    28. Barco A, Pittenger C, and Kandel ER. CREB, memory enhancement and the treatment of memory disorders:promises, pitfalls and prospects. Expert Opinion on Therapeutic Targets,2003,7(1):101-114.
    29. Foster TC, Sharrow KM, Masse JR, et al. Calcineurin links Ca2+ dysregulation with brain aging. Journal of Neuroscience,2001,21(11):4066-4073.
    30. Hattiangady B, Rao MS, Shetty GA, et al. Brain-derived neurotrophic factor, phosphorylated cyclic AMP response element binding protein and neuropeptide Y decline as early as middle age in the dentate gyrus and CA1 and CA3 subfields of the hippocampus. Experimental Neurology,2005,195(2):353-371.
    31. Asanuma M, Nishibayashi S, Iwata E, et al. Alterations of cAMP response element-binding activity in the aged rat brain in response to administration of rolipram, a cAMP-specific phosphodiesterase inhibitor. Brain Research. Molecular Brain Research,1996,41(1-2):210-215.
    32. Cos P, De Bruyne T, Hermans N, et al. Proanthocyanidins in health care:current and new trends. Current Medicinal Chemistry,2004,11 (10):1345-1359.
    33. Koleckar V, Kubikova K, Rehakova Z, et al. Condensed and hydrolysable tannins as antioxidants influencing the health. Mini Reviews in Medicinal Chemistry,2008, 8(5):436-447.
    34. Bagchi D, Garg A, Krohn RL, et al. Protective effects of grape seed proanthocyanidins and selected antioxidants against TPA-induced hepatic and brain lipid peroxidation and DNA fragmentation, and peritoneal macrophage activation in mice. General Pharmacology,1998,30(5):771-776.
    35. Abd El Mohsen MM, Kuhnle G, Rechner AR, et al. Uptake and metabolism of epicatechin and its access to the brain after oral ingestion. Free Radical Biology and Medicine,2002,33(12):1693-1702.
    36. Bagchi D, Garg A, Krohn RL, et al. Protective effects of grape seed proanthocyanidins and selected antioxidants against TPA-induced hepatic and brain lipid peroxidation and DNA fragmentation, and peritoneal macrophage activation in mice. General Pharmacology,1998,30(5):771-776.
    37. Devi A, Jolitha AB, and Ishii N. Grape seed proanthocyanidin extract (GSPE) and antioxidant defense in the brain of adult rats. Medical Science Monitor International Medical Journal of Experimental and Clinical Research,2006,12(4): BR124-129.
    38. Balu M, Sangeetha P, Murali G, et al. Modulatory role of grape seed extract on age-related oxidative DNA damage in central nervous system of rats. Brain Research Bulletin,2006,68(6):469-473.
    39. Gong Y, Liu L, Xie B, et al. Ameliorative effects of lotus seedpod proanthocyanidins on cognitive deficits and oxidative damage in senescence-accelerated mice. Behavioural Brain Research,2008,194(1):100-107.
    40. Williams RJ, Spencer JP, and Rice-Evans C. Flavonoids:antioxidants or signalling molecules? Free Radical Biology and Medicine,2004,36(7):838-849.
    41. Virgili F, Kobuchi H, and Packer L. Procyanidins extracted from Pinus maritima (Pycnogenol):scavengers of free radical species and modulators of nitrogen monoxide metabolism in activated murine RAW 264.7 macrophages. Free Radical Biology and Medicine,1998,24(7-8):1120-1129.
    42. Brixius K, Willms S, Napp A, et al. Crataegus special extract WS 1442 induces an endothelium-dependent, NO-mediated vasorelaxation via eNOS-phosphorylation at serine 1177. Cardiovascular Drugs and Therapy,2006,20(3):177-184.
    43. Stevens JF, Miranda CL, Wolthers KR, et al. Identification and in vitro biological activities of hop proanthocyanidins:inhibition of nNOS activity and scavenging of reactive nitrogen species. Journal of Agricultural and Food Chemistry,2002, 50(12):3435-3443.
    44. Maher P, Akaishi T, and Abe K. Flavonoid fisetin promotes ERK-dependent long-term potentiation and enhances memory. Proceedings of the National Academy of Sciences of the United States of America,2006,103(44):16568-16573.
    45. Williams CM, El Mohsen MA, Vauzour D, et al. Blueberry-induced changes in spatial working memory correlate with changes in hippocampal CREB phosphorylation and brain-derived neurotrophic factor (BDNF) levels. Free Radical Biology and Medicine,2008,45(3):295-305.
    46. Schroeter H, Bahia P, Spencer JP, et al. (-)Epicatechin stimulates ERK-dependent cyclic AMP response element activity and up-regulates GluR2 in cortical neurons. Journal of Neurochemistry,2007,101(6):1596-1606.
    47. Ling ZQ, Xie BJ, and Yang EL. Isolation, characterization, and determination of antioxidative activity of oligomeric procyanidins from the seedpod of Nelumbo nucifera Gaertn. Journal of Agricultural and Food Chemistry,2005, 53(7):2441-2445.
    48. Giovannini MG, Casamenti F, Bartolini L, et al. The brain cholinergic system as a target of cognition enhancers. Behavioural Brain Research,1997,83(1-2):1-5.
    49. Terry AV, Jr. and Buccafusco JJ. The cholinergic hypothesis of age and Alzheimer's disease-related cognitive deficits:recent challenges and their implications for novel drug development Journal of Pharmacology and Experimental Therapeutics,2003, 306(3):821-827.
    50. Morris R. Developments of a water-maze procedure for studying spatial learning in the rat Journal of Neuroscience Methods,1984, 11(1):47-60.
    51. Qiu H, Jin GQ, Jin RF, et al. Analysis of variance of repeated data measured by water maze with SPSS. Zhong Xi Yi Jie He Xue Bao,2007,5(1):101-105.
    52. Tomimoto H, Ohtani R, Shibata M, et al. Loss of cholinergic pathways in vascular dementia of the Binswanger type. Dementia and Geriatric Cognitive Disorders, 2005,19(5-6):282-288.
    53. Flood JF and Cherkin A. Scopolamine effects on memory retention in mice:a model
    of dementia? Behavioral and Neural Biology,1986,45(2):169-184.
    54. Buccafusco JJ, Terry AV, Jr., Webster SJ, et al. The scopolamine-reversal paradigm in rats and monkeys:the importance of computer-assisted operant-conditioning memory tasks for screening drug candidates. Psychopharmacology,2007.
    55. Jin SH, Park JK, Nam KY, et al. Korean red ginseng saponins with low ratios of protopanaxadiol and protopanaxatriol saponin improve scopolamine-induced learning disability and spatial working memory in mice. Journal of Ethnopharmacology,1999,66(2):123-129.
    56. Rubio J, Dang H, Gong M, et al. Aqueous and hydroalcoholic extracts of Black Maca (Lepidium meyenii) improve scopolamine-induced memory impairment in mice. Food and Chemical Toxicology,2007,45(10):1882-1890.
    57. Hermans RH, Hunter DE, McGivern RF, et al. Behavioral sequelae in young rats of acute intermittent antenatal hypoxia Neurotoxicology and Teratology,1992, 14(2):119-129.
    58. Gold PE. Acetylcholine modulation of neural systems involved in learning and memory. Neurobiology of Learning and Memory,2003,80(3):194-210.
    59. Chang Q and Gold PE. Switching memory systems during learning:changes in patterns of brain acetylcholine release in the hippocampus and striatum in rats. Journal of Neuroscience,2003,23(7):3001-3005.
    60. Fadda F, Cocco S, and Stancampiano R. Hippocampal acetylcholine release correlates with spatial learning performance in freely moving rats. NeuroReport, 2000, 11(10):2265-2269.
    61. Rowe WB, Blalock EM, Chen KC, et al. Hippocampal expression analyses reveal selective association of immediate-early, neuroenergetic, and myelinogenic pathways with cognitive impairment in aged rats. Journal of Neuroscience,2007, 27(12):3098-3110.
    62. D'Hooge R and De Deyn PP. Applications of the Morris water maze in the study of learning and memory. Brain Research. Brain Research Reviews,2001,36(1):60-90.
    63. Lindner MD. Reliability, distribution, and validity of age-related cognitive deficits in the Morris water maze. Neurobiology of Learning and Memory,1997, 68(3):203-220.
    64. Cassel JC, Lazaris A, Birthelmer A, et al. Spatial reference-(not working-or procedural-) memory performance of aged rats in the water maze predicts the magnitude of sulpiride-induced facilitation of acetylcholine release by striatal slices. Neurobiology of Aging,2007,28(8):1270-1285.
    65. Berry B, McMahan R, and Gallagher M. Spatial learning and memory at defined points of the estrous cycle:effects on performance of a hippocampal-dependent task. Behavioral Neuroscience,1997,111(2):267-274.
    66. Stackman RW, Blasberg ME, Langan CJ, et al. Stability of spatial working memory across the estrous cycle of Long-Evans rats. Neurobiology of Learning and Memory, 1997,67(2):167-171.
    67. Warren SG and Juraska JM. Spatial and nonspatial learning across the rat estrous cycle. Behavioral Neuroscience,1997, 111(2):259-266.
    68. Miller JC. Sex differences in dopaminergic and cholinergic activity and function in the nigro-striatal system of the rat. Psychoneuroendocrinology,1983,8(2):225-236.
    69. LeFevre J and McClintock MK. Reproductive senescence in female rats:a longitudinal study of individual differences in estrous cycles and behavior. Biology of Reproduction,1988,38(4):780-789.
    70. Barja G. Free radicals and aging. Trends In Neurosciences,2004,27(10):595-600.
    71. Poon HF, Calabrese V, Scapagnini G, et al. Free radicals and brain aging. Clinics In Geriatric Medicine,2004,20(2):329-359.
    72. Harman D. Free radical theory of aging. Mutation Research,1992,275(3-6): 257-266.
    73. Siqueira IR, Fochesatto C, de Andrade A, et al. Total antioxidant capacity is impaired in different structures from aged rat brain. International Journal of Developmental Neuroscience,2005,23(8):663-671.
    74. Rodrigues Siqueira I, Fochesatto C, da Silva Torres IL, et al. Aging affects oxidative state in hippocampus, hypothalamus and adrenal glands of Wistar rats. Life Sciences, 2005,78(3):271-278.
    75. Murali G and Panneerselvam C. Modulatory role of glutathione monoester in augmenting age-associated neuronal antioxidant system. Experimental Aging Research,2008,34(4):419-436.
    76. Navarro A and Boveris A. Rat brain and liver mitochondria develop oxidative stress and lose enzymatic activities on aging. American Journal of Physiology Regulatory Integrative and Comparative Physiology,2004,287(5):R1244-1249.
    77. Ravindranath V, Shivakumar BR, and Anandatheerthavarada HK. Low glutathione levels in brain regions of aged rats. Neuroscience Letters,1989,101(2):187-190.
    78. Calabrese V, Scapagnini G, Ravagna A, et al. Increased expression of heat shock proteins in rat brain during aging:relationship with mitochondrial function and glutathione redox state. Mechanisms of Ageing and Development,2004,125(4): 325-335.
    79. Fukui K, Onodera K, Shinkai T, et al. Impairment of learning and memory in rats caused by oxidative stress and aging, and changes in antioxidative defense systems. Annals of the New York Academy of Sciences,2001,928:168-175.
    80. Fukui K, Omoi NO, Hayasaka T, et al. Cognitive impairment of rats caused by oxidative stress and aging, and its prevention by vitamin E. Annals of the New York Academy of Sciences,2002,959:275-284.
    81. Dringen R. Metabolism and functions of glutathione in brain. Progress in Neurobiology,2000,62(6):649-671.
    82. Formigari A, Irato P, and Santon A. Zinc, antioxidant systems and metallothionein in metal mediated-apoptosis:biochemical and cytochemical aspects. Comparative Biochemistry and Physiology. Toxicology & Pharmacology:CBP,2007,146(4): 443-459.
    83. Serrano F and Klann E. Reactive oxygen species and synaptic plasticity in the aging hippocampus. Ageing Research Reviews,2004,3(4):431-443.
    84. Forster MJ, Dubey A, Dawson KM, et al. Age-related losses of cognitive function and motor skills in mice are associated with oxidative protein damage in the brain. Proceedings of the National Academy of Sciences of the United States of America, 1996,93(10):4765-4769.
    85. Mariani E, Polidori MC, Cherubini A, et al. Oxidative stress in brain aging, neurodegenerative and vascular diseases:an overview. Journal of Chromatography. B, Analytical Technologies in the Biomedical and Life Sciences,2005,827(1): 65-75.
    86. Arivazhagan P, Ayusawa D, and Panneerselvam C. Protective efficacy of alpha-lipoic acid on acetylcholinesterase activity in aged rat brain regions. Rejuvenation Research,2006,9(2):198-201.
    87. Carageorgiou H, Zarros A, and Tsakiris S. Selegiline long-term effects on brain acetylcholinesterase, (Na+,K+)-ATPase activities, antioxidant status and learning performance of aged rats. Pharmacological Research,2003,48(3):245-251.
    88. Yamaguchi T and Yamagata A. Serotonergic ligand binding in aging brain of experimental animals. Neurochemical Research,1991,16(4):469-473.
    89. Ikegami S. Behavioral impairment in radial-arm maze learning and acetylcholine content of the hippocampus and cerebral cortex in aged mice. Behavioural Brain Research,1994,65(1):103-111.
    90. Soreq H and Seidman S. Acetylcholinesterase-new roles for an old actor. Nature Reviews Neuroscience,2001,2(4):294-302.
    91. Weiner L, Kreimer D, Roth E, et al. Oxidative stress transforms acetylcholinesterase to a molten-globule-like state. Biochemical and Biophysical Research Communications,1994,198(3):915-922.
    92. Zou LB, Yamada K, Tanaka T, et al. Nitric oxide synthase inhibitors impair reference memory formation in a radial arm maze task in rats. Neuropharmacology, 1998,37(3):323-330.
    93. Holscher C, McGlinchey L, Anwyl R, et al.7-Nitro indazole, a selective neuronal nitric oxide synthase inhibitor in vivo, impairs spatial learning in the rat Learning and Memory,1996,2(6):267-278.
    94. Hara H, Waeber C, Huang PL, et al. Brain distribution of nitric oxide synthase in neuronal or endothelial nitric oxide synthase mutant mice using [3H]L-NG-nitro-arginine autoradiography. Neuroscience,1996,75(3):881-890.
    95. Son H, Hawkins RD, Martin K, et al. Long-term potentiation is reduced in mice that are doubly mutant in endothelial and neuronal nitric oxide synthase. Cell,1996, 87(6):1015-1023.
    96. Livak KJ and Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods,2001, 25(4):402-408.
    97. Bredt DS, Ferris CD, and Snyder SH. Nitric oxide synthase regulatory sites. Phosphorylation by cyclic AMP-dependent protein kinase, protein kinase C, and calcium/calmodulin protein kinase; identification of flavin and calmodulin binding sites. Journal of Biological Chemistry,1992,267(16):10976-10981.
    98. Song T, Hatano N, Horii M, et al. Calcium/calmodulin-dependent protein kinase Ⅰ inhibits neuronal nitric-oxide synthase activity through serine 741 phosphorylation. FEBS Letters,2004,570(1-3):133-137.
    99. Komeima K, Hayashi Y, Naito Y, et al. Inhibition of neuronal nitric-oxide synthase by calcium/ calmodulin-dependent protein kinase Ⅱalpha through Ser847 phosphorylation in NG108-15 neuronal cells. Journal of Biological Chemistry,2000, 275(36):28139-28143.
    100. Hayashi Y, Nishio M, Naito Y, et al. Regulation of neuronal nitric-oxide synthase by calmodulin kinases. Journal of Biological Chemistry,1999,274(29):20597-20602.
    101. Rameau GA, Chiu LY, and Ziff EB. NMD A receptor regulation of nNOS phosphorylation and induction of neuron death Neurobiology of Aging,2003, 24(8):1123-1133.
    102. Law A, O'Donnell J, Gauthier S, et al. Neuronal and inducible nitric oxide synthase expressions and activities in the hippocampi and cortices of young adult, aged cognitively unimpaired, and impaired Long-Evans rats. Neuroscience,2002, 112(2):267-275.
    103. Jesko H, Chalimoniuk M, and Strosznajder JB. Activation of constitutive nitric oxide synthase(s) and absence of inducible isoform in aged rat brain. Neurochemistry International,2003,42(4):315-322.
    104. Law A, Dore S, Blackshaw S, et al. Alteration of expression levels of neuronal nitric oxide synthase and haem oxygenase-2 messenger RNA in the hippocampi and cortices of young adult and aged cognitively unimpaired and impaired Long-Evans rats. Neuroscience,2000,100(4):769-775.
    105. Liu P, Smith PF, Appleton I, et al. Potential involvement of NOS and arginase in age-related behavioural impairments. Experimental Gerontology,2004, 39(8):1207-1222.
    106. La Porta CA and Comolli R. Age-dependent modulation of PKC isoforms and NOS
    activity and expression in rat cortex, striatum, and hippocampus. Experimental Gerontology,1999,34(7):863-874.
    107. Necchi D, Virgili M, Monti B, et al. Regional alterations of the NO/NOS system in the aging brain:a biochemical, histochemical and immunochemical study in the rat Brain Research,2002,933(1):31-41.
    108. Liu P, Smith PF, Appleton I, et al. Nitric oxide synthase and arginase in the rat hippocampus and the entorhinal, perirhinal, postrhinal, and temporal cortices: regional variations and age-related changes. Hippocampus,2003,13(7):859-867.
    109. Liu P, Smith PF, Appleton I, et al. Regional variations and age-related changes in nitric oxide synthase and arginase in the sub-regions of the hippocampus. Neuroscience,2003,119(3):679-687.
    110. Norris CM, Halpain S, and Foster TC. Alterations in the balance of protein kinase/phosphatase activities parallel reduced synaptic strength during aging. Journal of Neurophysiology,1998,80(3):1567-1570.
    111. Pascale A, Nogues X, Marighetto A, et al. Cytosolic hippocampal PKC and aging: correlation with discrimination performance. Neuroreport,1998,9(4):725-729.
    112. Karege F, Lambercy C, Schwald M, et al. Differential changes of cAMP-dependent protein kinase activity and 3H-cAMP binding sites in rat hippocampus during maturation and aging. Neuroscience Letters,2001,315(1-2):89-92.
    113. Chalimoniuk M and Strosznajder JB. Aging modulates nitric oxide synthesis and cGMP levels in hippocampus and cerebellum. Effects of amyloid beta peptide. Molecular and Chemical Neuropathology,1998,35(1-3):77-95.
    114. Law A, Gauthier S, and Quirion R. Say NO to Alzheimer's disease:the putative links between nitric oxide and dementia of the Alzheimer's type. Brain Research. Brain Research Reviews,2001,35(1):73-96.
    115. Terra X, Valls J, Vitrac X, et al. Grape-seed procyanidins act as antiinflammatory agents in endotoxin-stimulated RAW 264.7 macrophages by inhibiting NFkB signaling pathway. Journal of Agricultural and Food Chemistry,2007, 55(11):4357-4365.
    116. Josselyn SA, Shi C, Carlezon WA, Jr., et al. Long-term memory is facilitated by cAMP response element-binding protein overexpression in the amygdala Journal of
    Neuroscience,2001,21(7):2404-2412.
    117. Martin SJ, Grimwood PD, and Morris RG. Synaptic plasticity and memory:an evaluation of the hypothesis. Annual Review of Neuroscience,2000,23:649-711.
    118. Kandel ER. The molecular biology of memory storage:a dialogue between genes and synapses. Science,2001,294(5544):1030-1038.
    119. Si K, Giustetto M, Etkin A, et al. A neuronal isoform of CPEB regulates local protein synthesis and stabilizes synapse-specific long-term facilitation in aplysia Cell,2003,115(7):893-904.
    120. Impey S, McCorkle SR, Cha-Molstad H, et al. Defining the CREB regulon:a genome-wide analysis of transcription factor regulatory regions. Cell,2004, 119(7):1041-1054.
    121. Cha-Molstad H, Keller DM, Yochum GS, et al. Cell-type-specific binding of the transcription factor CREB to the cAMP-response element Proceedings of the National Academy of Sciences of the United States of America,2004, 101(37):13572-13577.
    122. Shaywitz AJ and Greenberg ME. CREB:a stimulus-induced transcription factor activated by a diverse array of extracellular signals. Annual Review of Biochemistry, 1999,68:821-861.
    123. Bekinschtein P, Cammarota M, Izquierdo I, et al. BDNF and memory formation and storage. Neuroscientist,2008,14(2):147-156.
    124. Jones MW, Errington ML, French PJ, et al. A requirement for the immediate early gene Zif268 in the expression of late LTP and long-term memories. Nature Neuroscience,2001,4(3):289-296.
    125. Tao X, Finkbeiner S, Arnold DB, et al. Ca2+ influx regulates BDNF transcription by a CREB family transcription factor-dependent mechanism. Neuron,1998, 20(4):709-726.
    126. Shieh PB, Hu SC, Bobb K, et al. Identification of a signaling pathway involved in calcium regulation of BDNF expression. Neuron,1998,20(4):727-740.
    127. Alonso M, Vianna MR, Depino AM, et al. BDNF-triggered events in the rat hippocampus are required for both short-and long-term memory formation. Hippocampus,2002,12(4):551-560.
    128. Cohen-Cory S and Fraser SE. Effects of brain-derived neurotrophic factor on optic axon branching and remodelling in vivo. Nature,1995,378(6553):192-196.
    129. Vicario-Abejon C. Collin C, McKay RD, et al. Neurotrophins induce formation of functional excitatory and inhibitory synapses between cultured hippocampal neurons. Journal of Neuroscience,1998,18(18):7256-7271.
    130. McAllister AK, Katz LC, and Lo DC. Neurotrophin regulation of cortical dendritic growth requires activity. Neuron,1996,17(6):1057-1064.
    131. Collin C, Vicario-Abejon C, Rubio ME, et al. Neurotrophins act at presynaptic terminals to activate synapses among cultured hippocampal neurons. European Journal of Neuroscience,2001,13(7):1273-1282.
    132. Porte Y, Buhot MC, and Mons N. Alteration of CREB phosphorylation and spatial memory deficits in aged 129T2/Sv mice. Neurobiology of Aging,2008, 29(10):1533-1546.
    133. Foster TC. Involvement of hippocampal synaptic plasticity in age-related memory decline. Brain Research. Brain Research Reviews,1999,30(3):236-249.
    134. van Praag H, Lucero MJ, Yeo GW, et al. Plant-derived flavanol (-)epicatechin enhances angiogenesis and retention of spatial memory in mice. Journal of Neuroscience,2007,27(22):5869-5878.
    1. Deprez S, Mila I, Huneau JF, et al. Transport of proanthocyanidin dimer, trimer, and polymer across monolayers of human intestinal epithelial Caco-2 cells. Antioxid Redox Signal,2001,3(6):957-967.
    2. Sano A, Yamakoshi J, Tokutake S, et al. Procyanidin B1 is detected in human serum after intake of proanthocyanidin-rich grape seed extract Bioscience, Biotechnology, and Biochemistry,2003,67(5):1140-1143.
    3. Shoji T, Masumoto S, Moriichi N, et al. Apple procyanidin oligomers absorption in rats after oral administration:analysis of procyanidins in plasma using the porter method and high-performance liquid chromatography/tandem mass spectrometry. Journal of Agricultural and Food Chemistry,2006,54(3):884-892.
    4. Spencer JP, Chaudry F, Pannala AS, et al. Decomposition of cocoa procyanidins in the gastric milieu Biochemical and Biophysical Research Communications,2000, 272(1):236-241.
    5. Rios LY, Bennett RN, Lazarus SA, et al. Cocoa procyanidins are stable during gastric transit in humans. American Journal of Clinical Nutrition,2002,76(5): 1106-1110.
    6. Deprez S, Brezillon C, Rabot S, et al. Polymeric proanthocyanidins are catabolized by human colonic microflora into low-molecular-weight phenolic acids. Journal of Nutrition,2000,130(11):2733-2738.
    7. Tsang C, Auger C, Mullen W, et al. The absorption, metabolism and excretion of flavan-3-ols and procyanidins following the ingestion of a grape seed extract by rats. British Journal of Nutrition,2005,94(2):170-181.
    8. Spencer JP, Schroeter H, Shenoy B, et al. Epicatechin is the primary bioavailable form of the procyanidin dimers B2 and B5 after transfer across the small intestine. Biochemical and Biophysical Research Communications,2001,285(3):588-593.
    9. Baba S, Osakabe N, Natsume M, et al. Absorption and urinary excretion of procyanidin B2 [epicatechin-(4beta-8)-epicatechin] in rats. Free Radical Biology and Medicine,2002,33(1):142-148.
    10. Steinberg FM, Holt RR, Schmitz HH, et al. Cocoa procyanidin chain length does not determine ability to protect LDL from oxidation when monomer units are controlled The Journal of nutritional biochemistry.2002,13(11):645-652.
    11. Stoupi S, Williamson G, Viton F, et al. In vivo bioavailability, absorption, excretion, and pharmacokinetics of [14C]procyanidin B2 in male rats. Drug Metabolism and Disposition:The Biological Fate of Chemicals,2010,38(2):287-291.
    12. van Praag H, Lucero MJ, Yeo GW, et al. Plant-derived flavanol (-)epicatechin enhances angiogenesis and retention of spatial memory in mice. Journal of Neuroscience,2007,27(22):5869-5878.
    13. Bagchi D, Garg A, Krohn RL, et al. Protective effects of grape seed proanthocyanidins and selected antioxidants against TPA-induced hepatic and brain lipid peroxidation and DNA fragmentation, and peritoneal macrophage activation in mice. General Pharmacology,1998,30(5):771-776.
    14. Cai YZ, Mei S, Jie X, et al. Structure-radical scavenging activity relationships of phenolic compounds from traditional Chinese medicinal plants. Life Sciences,2006, 78(25):2872-2888.
    15. Plumb GW, De Pascual-Teresa S, Santos-Buelga C, et al. Antioxidant properties of catechins and proanthocyanidins:effect of polymerisation, galloylation and glycosylation. Free Radical Research,1998,29(4):351-358.
    16. Lopes GK, Schulman HM, and Hermes-Lima M. Polyphenol tannic acid inhibits hydroxyl radical formation from Fenton reaction by complexing ferrous ions. Biochimica et BiophysicaActa,1999,1472(1-2):142-152.
    17. Puiggros F, Llopiz N, Ardevol A, et al. Grape seed procyanidins prevent oxidative injury by modulating the expression of antioxidant enzyme systems. Journal of Agricultural and Food Chemistry,2005,53(15):6080-6086.
    18. Devi A, Jolitha AB, and Ishii N. Grape seed proanthocyanidin extract (GSPE) and antioxidant defense in the brain of adult rats. Medical Science Monitor: International Medical Journal of Experimental and Clinical Research,2006, 12(4):BR124-129.
    19. Huynh HT and Teel RW. Selective induction of apoptosis in human mammary cancer cells (MCF-7) by pycnogenol. Anticancer Research,2000,20(4):2417-2420.
    20. Ito H, Kobayashi E, Takamatsu Y. et al. Polyphenols from Eriobotrya japonica and
    their cytotoxicity against human oral tumor cell lines. Chemical and Pharmaceutical Bulletin,2000,48(5):687-693.
    21. Ye X, Krohn RL, Liu W, et al. The cytotoxic effects of a novel IH636 grape seed proanthocyanidin extract on cultured human cancer cells. Molecular and Cellular Biochemistry,1999,196(1-2):99-108.
    22. Hertog MG, Feskens EJ, Hollman PC, et al. Dietary antioxidant flavonoids and risk of coronary heart disease:the Zutphen Elderly Study. Lancet,1993, 342(8878):1007-1011.
    23. Tebib K, Bitri L, Besancon P, et al. Polymeric grape seed tannins prevent plasma cholesterol changes in high-cholesterol-fed rats. Food Chemistry,1994, 49(4):403-406.
    24. Lacaille D, Franck U, and Wagner H. Search for potential angiotensin converting enzyme (ACE)-inhibitors from plants. Phytomedicine,2001,8(1):47-52.
    25. Leikert JF, Rathel TR, Wohlfart P, et al. Red wine polyphenols enhance endothelial nitric oxide synthase expression and subsequent nitric oxide release from endothelial cells. Circulation,2002,106(13):1614-1617.
    26. Cortes SF, Valadares YM, de Oliveira AB, et al. Mechanism of endothelium-dependent vasodilation induced by a proanthocyanidin-rich fraction from Ouratea semiserrata Planta Medica,2002,68(5):412-415.
    27. Fitzpatrick DF, Bing B, and Rohdewald P. Endothelium-dependent vascular effects of Pycnogenol. Journal of Cardiovascular Pharmacology,1998,32(4):509-515.
    28. Lotito SB, Actis-Goretta L, Renart ML, et al. Influence of oligomer chain length on the antioxidant activity of procyanidins. Biochemical and Biophysical Research Communications,2000,276(3):945-951.
    29. Pearson DA, Schmitz HH, Lazarus SA, et al. Inhibition of in vitro low-density lipoprotein oxidation by oligomeric procyanidins present in chocolate and cocoas. Methods in Enzymology,2001,335:350-360.
    30. Schewe T, Sadik C, Klotz LO, et al. Polyphenols of cocoa:inhibition of mammalian 15-lipoxygenase. Biological Chemistry,2001,382(12):1687-1696.
    31. Chang WC and Hsu FL. Inhibition of platelet aggregation and arachidonate metabolism in platelets by procyanidins. Prostaglandins Leukotrienes and Essential Fatty Acids,1989,38(3):181-188.
    32. Putter M, Grotemeyer KH, Wurthwein G, et al. Inhibition of smoking-induced platelet aggregation by aspirin and pycnogenol. Thrombosis Research,1999, 95(4):155-161.
    33. Yamakoshi J, Saito M, Kataoka S, et al. Safety evaluation of proanthocyanidin-rich extract from grape seeds. Food and Chemical Toxicology,2002,40(5):599-607.
    34. Wren AF, Cleary M, Frantz C, et al.90-day oral toxicity study of a grape seed extract (IH636) in rats. Journal of Agricultural and Food Chemistry,2002,50(7): 2180-2192.
    35. Bentivegna SS and Whitney KM. Subchronic 3-month oral toxicity study of grape seed and grape skin extracts. Food and Chemical Toxicology,2002,40(12): 1731-1743.
    36. Clifton PM. Effect of Grape Seed Extract and Quercetin on Cardiovascular and Endothelial Parameters in High-Risk Subjects. Journal of Biomedicine & Biotechnology,2004,2004(5):272-278.
    37. Koivisto K, Reinikainen KJ, Hanninen T, et al. Prevalence of age-associated memory impairment in a randomly selected population from eastern Finland Neurology,1995,45(4):741-747.
    38. Hanninen T, Hallikainen M, Koivisto K, et al. A follow-up study of age-associated memory impairment:neuropsychological predictors of dementia. Journal of the American Geriatrics Society,1995,43(9):1007-1015.
    39. Dringen R. Metabolism and functions of glutathione in brain. Progress In Neurobiology,2000,62(6):649-671.
    40. Polidori MC. Antioxidant micronutrients in the prevention of age-related diseases. Journal of Postgraduate Medicine,2003,49(3):229-235.
    41. Floyd RA. Antioxidants, oxidative stress, and degenerative neurological disorders. Proceedings of the Society for Experimental Biology and Medicine,1999,222(3): 236-245.
    42. Gonenc S, Uysal N, Acikgoz O, et al. Effects of melatonin on oxidative stress and spatial memory impairment induced by acute ethanol treatment in rats. Physiological Research,2005,54(3):341-348.
    43. Messaoudi M, Tricoire A, Lalonde R, et al. Effects of MPTP on lever-pressing for light extinction in rats. European Journal of Pharmacology,1996,299(1-3):17-20.
    44. Liu R, Liu IY, Bi X, et al. Reversal of age-related learning deficits and brain oxidative stress in mice with superoxide dismutase/catalase mimetics. Proceedings of the National Academy of Sciences of the United States of America,2003, 100(14):8526-8531.
    45. Sano M, Ernesto C, Thomas RG, et al. A controlled trial of selegiline, alpha-tocopherol, or both as treatment for Alzheimer's disease. The Alzheimer's Disease Cooperative Study. New England Journal of Medicine,1997, 336(17):1216-1222.
    46. Perrig WJ, Perrig P, and Stahelin HB. The relation between antioxidants and memory performance in the old and very old. Journal of The American Geriatrics Society,1997,45(6):718-724.
    47. Tomimoto H, Ohtani R, Shibata M, et al. Loss of cholinergic pathways in vascular dementia of the Binswanger type. Dementia and Geriatric Cognitive Disorders, 2005,19(5-6):282-288.
    48. Bartus RT, Dean RL,3rd, Beer B, et al. The cholinergic hypothesis of geriatric memory dysfunction. Science,1982,217(4558):408-414.
    49. Bartus RT. On neurodegenerative diseases, models, and treatment strategies:lessons learned and lessons forgotten a generation following the cholinergic hypothesis. Experimental Neurology,2000,163(2):495-529.
    50. Francis PT, Palmer AM, Sims NR, et al. Neurochemical studies of early-onset Alzheimer's disease. Possible influence on treatment New England Journal of Medicine,1985,313(1):7-11.
    51. Strong R, Hicks P, Hsu L, et al. Age-related alterations in the rodent brain cholinergic system and behavior. Neurobiology of Aging,1980, 1(1):59-63.
    52. Fong TG, Vogelsberg V, Neff NH, et al. GM1 and NGF synergism on choline acetyltransferase and choline uptake in aged brain. Neurobiology of Aging,1995, 16(6):917-923.
    53. Sherman KA and Friedman E. Pre-and post-synaptic cholinergic dysfunction in aged rodent brain regions:new findings and an interpretative review. International Journal of Developmental Neuroscience,1990,8(6):689-708.
    54. Decker MW. The effects of aging on hippocampal and cortical projections of the forebrain cholinergic system. Brain Research,1987,434(4):423-438.
    55. Gallagher M, Burwell RD, Kodsi MH, et al. Markers for biogenic amines in the aged rat brain:relationship to decline in spatial learning ability. Neurobiology of Aging,1990, 11(5):507-514.
    56. DeKosky ST, Ikonomovic MD, Styren SD, et al. Upregulation of choline acetyltransferase activity in hippocampus and frontal cortex of elderly subjects with mild cognitive impairment. Annals of Neurology,2002,51(2):145-155.
    57. Arivazhagan P, Ayusawa D, and Panneerselvam C. Protective efficacy of alpha-lipoic acid on acetylcholinesterase activity in aged rat brain regions. Rejuvenation Research,2006,9(2):198-201.
    58. Sharma D and Singh R. Centrophenoxine activates acetylcholinesterase activity in hippocampus of aged rats. Indian Journal of Experimental Biology,1995, 33(5):365-368.
    59. Das A, Dikshit M, and Nath C. Profile of acetylcholinesterase in brain areas of male and female rats of adult and old age. Life Sciences,2001,68(13):1545-1555.
    60. Buccafusco JJ and Terry AV, Jr. Multiple central nervous system targets for eliciting beneficial effects on memory and cognition. Journal of Pharmacology and Experimental Therapeutics,2000,295(2):438-446.
    61. Terry AV, Jr. and Buccafusco JJ. The cholinergic hypothesis of age and Alzheimer's disease-related cognitive deficits:recent challenges and their implications for novel drug development Journal of Pharmacology and Experimental Therapeutics,2003, 306(3):821-827.
    62. Gracon SI, Knapp MJ, Berghoff WG, et al. Safety of tacrine:clinical trials, treatment IND, and postmarketing experience. Alzheimer Disease and Associated Disorders,1998,12(2):93-101.
    63. Dinerman JL, Dawson TM, Schell MJ, et al. Endothelial nitric oxide synthase localized to hippocampal pyramidal cells:implications for synaptic plasticity. Proceedings of the National Academy of Sciences of the United States of America, 1994,91(10):4214-4218.
    64. Colasanti M, Persichini T, Fabrizi C, et al. Expression of a NOS-III-like protein in human astroglial cell culture. Biochemical and Biophysical Research Communications,1998,252(3):552-555.
    65. Rameau GA, Tukey DS, Garcin-Hosfield ED, et al. Biphasic coupling of neuronal nitric oxide synthase phosphorylation to the NMDA receptor regulates AMPA receptor trafficking and neuronal cell death. Journal of Neuroscience,2007,27(13): 3445-3455.
    66. Bredt DS, Ferris CD, and Snyder SH. Nitric oxide synthase regulatory sites. Phosphorylation by cyclic AMP-dependent protein kinase, protein kinase C, and calcium/calmodulin protein kinase; identification of flavin and calmodulin binding sites. Journal of Biological Chemistry,1992,267(16):10976-10981.
    67. Rameau GA, Chiu LY, and Ziff EB. NMDA receptor regulation of nNOS phosphorylation and induction of neuron death Neurobiology of Aging,2003, 24(8):1123-1133.
    68. Smolenski A, Burkhardt AM, Eigenthaler M, et al. Functional analysis of cGMP-dependent protein kinases I and II as mediators of NO/cGMP effects. Naunyn-Schmiedebergs Archives of Pharmacology,1998,358(1):134-139.
    69. Prast H and Philippu A. Nitric oxide as modulator of neuronal function. Progress in Neurobiology,2001,64(1):51-68.
    70. Prast H and Philippu A. Nitric oxide releases acetylcholine in the basal forebrain. European Journal of Pharmacology,1992,216(1):139-140.
    71. Calabrese V, Mancuso C, Calvani M, et al. Nitric oxide in the central nervous system:neuroprotection versus neurotoxicity. Nature Reviews. Neuroscience,2007, 8(10):766-775.
    72. Pannu R and Singh I. Pharmacological strategies for the regulation of inducible nitric oxide synthase:neurodegenerative versus neuroprotective mechanisms. Neurochemistry International,2006,49(2):170-182.
    73. Warner DS, Sheng H, and Batinic-Haberle I. Oxidants, antioxidants and the ischemic brain. Journal of Experimental Biology,2004,207(Pt 18):3221-3231.
    74. Dawson VL, Dawson TM, Bartley DA, et al. Mechanisms of nitric oxide-mediated neurotoxicity in primary brain cultures. Journal of Neuroscience,1993,13(6): 2651-2661.
    75. La Porta CA and Comolli R. Age-dependent modulation of PKC isoforms and NOS activity and expression in rat cortex, striatum, and hippocampus. Experimental Gerontology,1999,34(7):863-874.
    76. Mollace V, Rodino P, Massoud R, et al. Age-dependent changes of NO synthase activity in the rat brain. Biochemical and Biophysical Research Communications, 1995,215(3):822-827.
    77. Necchi D, Virgili M, Monti B, et al. Regional alterations of the NO/NOS system in the aging brain:a biochemical, histochemical and immunochemical study in the rat Brain Research,2002,933(1):31-41.
    78. Siles E, Martinez-Lara E, Canuelo A, et al. Age-related changes of the nitric oxide system in the rat brain. Brain Research,2002,956(2):385-392.
    79. Liu P, Smith PF, Appleton I, et al. Age-related changes in nitric oxide synthase and arginase in the rat prefrontal cortex. Neurobiology of Aging,2004,25(4):547-552.
    80. Chalimoniuk M and Strosznajder JB. Aging modulates nitric oxide synthesis and cGMP levels in hippocampus and cerebellum. Effects of amyloid beta peptide. Molecular and Chemical Neuropathology,1998,35(1-3):77-95.
    81. Liu P, Smith PF, Appleton I, et al. Nitric oxide synthase and arginase in the rat hippocampus and the entorhinal, perirhinal, postrhinal, and temporal cortices: regional variations and age-related changes. Hippocampus,2003,13(7):859-867.
    82. Liu P, Smith PF, Appleton I, et al. Regional variations and age-related changes in nitric oxide synthase and arginase in the sub-regions of the hippocampus. Neuroscience,2003,119(3):679-687.
    83. Liu P, Smith PF, Appleton I, et al. Hippocampal nitric oxide synthase and arginase and age-associated behavioral deficits. Hippocampus,2005,15(5):642-655.
    84. Jesko H, Chalimoniuk M, and Strosznajder JB. Activation of constitutive nitric oxide synthase(s) and absence of inducible isoform in aged rat brain. Neurochemistry International,2003,42(4):315-322.
    85. Law A, Dore S, Blackshaw S, et al. Alteration of expression levels of neuronal nitric oxide synthase and haem oxygenase-2 messenger RNA in the hippocampi and cortices of young adult and aged cognitively unimpaired and impaired Long-Evans rats. Neuroscience,2000,100(4):769-775.
    86. Law A, O'Donnell J, Gauthier S, et al. Neuronal and inducible nitric oxide synthase expressions and activities in the hippocampi and cortices of young adult, aged cognitively unimpaired, and impaired Long-Evans rats. Neuroscience,2002, 112(2):267-275.
    87. Carlezon WA, Jr., Duman RS, and Nestler EJ. The many faces of CREB. Trends in Neurosciences,2005,28(8):436-445.
    88. Herdegen T and Leah JD. Inducible and constitutive transcription factors in the mammalian nervous system:control of gene expression by Jun, Fos and Krox, and CREB/ATF proteins. Brain Research. Brain Research Reviews,1998, 28(3):370-490.
    89. Wu X and McMurray CT. Calmodulin kinase II attenuation of gene transcription by preventing cAMP response element-binding protein (CREB) dimerization and binding of the CREB-binding protein Journal of Biological Chemistry,2001, 276(3):1735-1741.
    90. Conkright MD, Guzman E, Flechner L, et al. Genome-wide analysis of CREB target genes reveals a core promoter requirement for cAMP responsiveness. Molecular Cell,2003, 11(4):1101-1108.
    91. Yin JC, Wallach JS, Del Vecchio M, et al. Induction of a dominant negative CREB transgene specifically blocks long-term memory in Drosophila Cell,1994,79(1): 49-58.
    92. Bourtchuladze R, Frenguelli B, Blendy J, et al. Deficient long-term memory in mice with a targeted mutation of the cAMP-responsive element-binding protein. Cell, 1994,79(1):59-68.
    93. Yin JC, Del Vecchio M, Zhou H, et al. CREB as a memory modulator:induced expression of a dCREB2 activator isoform enhances long-term memory in Drosophila Cell,1995,81(1):107-115.
    94. Josselyn SA, Shi C, Carlezon WA, Jr., et al. Long-term memory is facilitated by cAMP response element-binding protein overexpression in the amygdala Journal of Neuroscience,2001,21(7):2404-2412.
    95. Pittenger C, Huang YY, Paletzki RF, et al. Reversible inhibition of CREB/ATF transcription factors in region CA1 of the dorsal hippocampus disrupts hippocampus-dependent spatial memory. Neuron,2002,34(3):447-462.
    96. Yang SN, Huang LT, Wang CL, et al. Prenatal administration of morphine decreases CREBSerine-133 phosphorylation and synaptic plasticity range mediated by glutamatergic transmission in the hippocampal CA1 area of cognitive-deficient rat offspring. Hippocampus,2003,13(8):915-921.
    97. Florian C, Mons N, and Roullet P. CREB antisense oligodeoxynucleotide administration into the dorsal hippocampal CA3 region impairs long-but not short-term spatial memory in mice. Learning and Memory,2006,13(4):465-472.
    98. Guzowski JF and McGaugh JL. Antisense oligodeoxynucleotide-mediated disruption of hippocampal cAMP response element binding protein levels impairs consolidation of memory for water maze training. Proceedings of the National Academy of Sciences of the United States of America,1997,94(6):2693-2698.
    99. Taubenfeld SM, Wiig KA, Monti B, et al. Fornix-dependent induction of hippocampal CCAAT enhancer-binding protein [beta] and [delta] Co-localizes with phosphorylated cAMP response element-binding protein and accompanies long-term memory consolidation Journal of Neuroscience,2001,21(1):84-91.
    100. Mizuno M, Yamada K, Maekawa N, et al. CREB phosphorylation as a molecular marker of memory processing in the hippocampus for spatial learning. Behavioural Brain Research,2002,133(2):135-141.
    101. Silva AJ, Kogan JH, Frankland PW, et al. CREB and memory. Annual Review of Neuroscience,1998,21:127-148.
    102. Countryman RA and Gold PE. Rapid forgetting of social transmission of food preferences in aged rats:relationship to hippocampal CREB activation Learning & Memory,2007,14(5):350-358.
    103. Kudo K, Wati H, Qiao C, et al. Age-related disturbance of memory and CREB phosphorylation in CA1 area of hippocampus of rats. Brain Research,2005, 1054(1):30-37.
    104. Foster TC, Sharrow KM, Masse JR, et al. Calcineurin links Ca2+ dysregulation with brain aging. Journal of Neuroscience,2001,21(11):4066-4073.
    105. Brightwell JJ, Gallagher M, and Colombo PJ. Hippocampal CREB1 but not CREB2 is decreased in aged rats with spatial memory impairments. Neurobiology of Learning and Memory.2004.81(1):19-26.
    106. Porte Y, Buhot MC, and Mons N. Alteration of CREB phosphorylation and spatial memory deficits in aged 129T2/Sv mice. Neurobiology of Aging,2008, 29(10):1533-1546.
    107. Hattiangady B, Rao MS, Shetty GA, et al. Brain-derived neurotrophic factor, phosphorylated cyclic AMP response element binding protein and neuropeptide Y decline as early as middle age in the dentate gyrus and CA1 and CA3 subfields of the hippocampus. Experimental Neurology,2005,195(2):353-371.
    108. Monti B, Berteotti C, and Contestabile A. Dysregulation of memory-related proteins in the hippocampus of aged rats and their relation with cognitive impairment Hippocampus,2005,15(8):1041-1049.
    109. Mouravlev A, Dunning J, Young D, et al. Somatic gene transfer of cAMP response element-binding protein attenuates memory impairment in aging rats. Proceedings of the National Academy of Sciences of the United States of America,2006, 103(12):4705-4710.

© 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