用户名: 密码: 验证码:
重复经颅磁刺激改善老化相关的认知功能损伤的电生理机制及潜在代谢产物的变化
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
重复经颅磁刺激(rTMS)是一种无损伤、无侵入性的物理治疗方法,目前已逐渐得到人们关注。研究报道rTMS可以提高正常老化和阿尔兹海默病(AD)患者的认知功能损伤。rTMS对认知功能的改善是通过网络化,多通路调节脑组织结构和功能发挥作用的。研究报道,rTMS作用于神经系统可以提高神经元的兴奋性,并增加突触可塑性。rTMS改善突触可塑性的效应可以表现为长时程增强(LTP)效应。动物实验证明,在应用高频rTMS改善动物认知功能的同时,所记录到的海马脑片LTP显著增强了。在基因水平、蛋白质水平、代谢产物水平的研究表明,rTMS可以通过调节改善神经元可塑性相关基因、蛋白的表达和代谢产物平衡保护神经元。然而,高频rTMS改善认知功能时提高神经元兴奋性的具体机制及rTMS改善认知功能相关的物质基础目前还不是十分清楚。
     海马和额皮层是与认知功能密切相关的脑区,在老化过程和一些神经系统退行性疾病中也是最易受损伤的区域。研究表明海马神经元兴奋性在正常老化过程中是下降的,并表现为后超极化幅度的增大、静息电位的超极化。神经元的信息传递编码为动作电位进行传递,因此,如果动作电位的形成变慢,信息传递速率会受到影响,这样在神经网络信息传递中,效率就会降低。研究已经证明与青年个体相比,老年个体的神经元兴奋性显著下降低,而神经元兴奋性的下降在认知过程中起到了关键作用。神经元兴奋性降低的具体机制--物质基础,是如何变化的。这一变化在病理生理刺激,基因改变等作用方式下如何表现为动态的、多系统的变化。应用生物体液和组织对机体病理机制进行代谢物质的检测,结合非靶标多元统计分析方法,分析疾病过程中的物质变化基础,目前在许多领域得到了广泛应用。这种物质检测分析方法的优势是在疾病预防和治疗过程中,可以发现机体生化改变的生物标志物。在对AD疾病的研究中,已有应用非靶标多元统计方法对AD病人脑脊液进行了分析,发现AD病人机体的新陈代谢与正常对照人员相比,发生了明显变化。
     脑老化是脑组织随着年龄增长发生的组织结构和功能的变化。随着年龄的增长,个体在脑老化过程中可表现为学习记忆能力的降低。啮齿类动物作为实验模型研究脑老化及老化相关的认知功能障碍具有很多优点:啮齿类动物的海马和前额皮层在老化中与人类相似,是非常容易受损的,并伴有结构和功能的变化。昆明小鼠在老化过程中可表现出认知功能的损伤,是研究脑老化的理想动物模型。在对啮齿类动物进行认知功能评估的行为学实验中,被动逃避反应实验和新物体识别实验是经典的认知功能测试方法。
     本课题中我们观察了rTMS是如何改善昆明小鼠老化过程中认知功能的损伤,及其内在电生理机制和代谢物质基础。
     1rTMS改善老化引起的认知功能障碍
     观察rTMS是否可以改善昆明小鼠在年龄增长过程中出现的认知功能损害。为证明rTMS对认知功能的影响,应用被动逃避反应实验和新物体识别实验对小鼠的认知功能进行检测。
     方法:3-4月龄(青年)昆明小鼠,9-10月龄(成年)昆明小鼠,16-17月龄(老年)昆明小鼠。在实验室恒温20-25摄氏度适应饲养1天以适应实验室环境,然后用于实验。
     实验分组:
     (1)老年rTMS组:16-17月龄老年昆明小鼠,磁头于小鼠的颅顶部每天进行10组频率为25赫兹的磁刺激,总共刺激脉冲数为1000个,连续14天。
     (2)老年sham组:16-17月龄老年昆明小鼠,以磁头反面于小鼠颅顶部进行与rTMS组相似的无功能的磁刺激。
     (3)成年组:9-10月龄成年昆明小鼠,与老年sham组小鼠刺激方式相同。
     (4)青年组:3-4月龄青年昆明小鼠,与老年sham组小鼠刺激方式相同。
     各组动物在相同环境下饲养,然后通过被动逃避反应实验和新物体识别实验测试各组动物学习记忆能力的差异。
     结果:新物体识别实验成绩显示,在1h时间点,小鼠触碰两物体的总时间在青年组、成年组、老年组之间没有明显差异,在24h小鼠触碰两物体的总时间在三组之间同样也没有发现显著差异。在1h时间点,青年组和成年组之间的认知指数没有明显差异;与青年组和成年组相比,老年组的认知指数显著降低(P<0.05)。同样在24h时间点,与青年和成年组相比,老年组小鼠的认知指数显著降低(P<0.05);而青年组和成年组之间的认知指数没有显著差异。在1h时间点,小鼠触碰两物体的总时间在老年组和老年rTMS组没有明显差异,同样在24h也没有发现显著差异。在1h时间点,与老年组相比,老年rTMS组的认知指数显著提高(P<0.05)。同样在24h时间点,与老年组相比,老年rTMS组小鼠的认知指数显著提高(P<0.05)。
     被动逃避反应实验成绩显示,适应阶段的潜伏期在青年组、成年组和老年组没有显著差异。与青年组和成年组相比,学习阶段的电击次数在老年昆明鼠明显增多(P<0.05),而青年组和成年组之间无显著差异。与青年鼠和成年鼠相比,记忆阶段的被动逃避潜伏期在老年鼠出现了显著缩短(P<0.05),同样青年组和成年组之间没有显著差异。被动逃避反应实验适应阶段的潜伏期在老年组和老年rTMS组没有显著差异。与老年组相比,学习阶段的电击次数在老年rTMS昆明鼠明显减少(P<0.05)。同样与老年鼠相比,记忆阶段的被动逃避潜伏期在老年rTMS鼠出现了显著增长(P<0.05)。
     以上结果提示,昆明小鼠的认知功能在青年组和成年组没有出现明显差异,而在16月龄时开始出现下降,而应用rTMS可以改善老化引起的认知功能损伤。2rTMS改善老化相关的神经元电生理变化
     观察rTMS是否可以通过改善电压依赖性钙通道(VDCC)来调节神经元兴奋性,进而改善老年小鼠的认知功能障碍。
     方法:3-4月龄昆明小鼠,16-17月龄昆明小鼠。在相同实验室条件饲养,实验室恒温20-25摄氏度,然后用于实验。
     实验分组:
     (1)老年rTMS组:16-17月龄老年小鼠,磁头于小鼠的颅顶部每天进行10组频率为25赫兹的磁刺激,总共刺激脉冲数为1000个,连续14天。
     (2)老年sham组:16-17月龄老年小鼠,以磁头反面于小鼠颅顶部进行与rTMS组相似的无功能的磁刺激。
     (3)青年组:3-4月龄青年小鼠,与老年sham组小鼠刺激方式相同。
     各组动物在相同环境下饲养,然后通过行为学实验测试后,而后进行神经元兴奋性,VDCC等电生理指标的记录。
     结果:全细胞电流钳记录结果显示,rTMS可以改善老化引起的神经元兴奋性变化。与青年鼠相比,在老年鼠海马CA1区的神经元静息电位发生了显著超级化(P<0.05)。在应用rTMS后发现,与老年鼠相比,在老年磁刺激组小鼠海马CA1区的神经元静息电位显著去极化(P<0.05)。与青年组相比,老年组海马CA1区神经元的动作电位产生频率显著下降(P<0.05)。而高频经颅磁刺激可以显著提高老年神经元的动作电位的产生频率(P<0.05)。同样与青年组相比,在老年组海马CA1区神经元的后超极化幅度显著增大(P<0.05)。而应用rTMS后,可显著降低老年组的后超级化幅度(P<0.05)。而动作电位的阈值记录结果显示,在青年组、老年组和老年磁刺激组之间,没有发现显著差异。
     全细胞电流钳记录结果显示,rTMS可以显著降低老年海马CA1区神经元的VDCC。与老年对照组神经元相比,在测试电压分别为-20mV、-10mV和0mV时,在老年rTMS组神经元所记录的电流强度均显著降低(P<0.05)。
     以上结果表明,rTMS可以通过改善VDCC来调节神经元兴奋性,进而改善老年小鼠的认知功能障碍。3rTMS改善老化相关的脑组织代谢产物变化
     观察应用气质联用分析方法对小鼠脑组织代谢产物进行检测,应用偏最小二乘法判别分析(PLS-DA)和主成分分析(PCA)方法对结果进行分析,进一步应用one-wayANOVA统计方法深入分析差异代谢物。
     方法:3-4月龄昆明小鼠,9-10月龄昆明小鼠,16-17月龄昆明小鼠。在相同实验室条件饲养,实验室恒温20-25摄氏度,然后用于实验。
     实验分组:
     (1)老年rTMS组:16-17月龄老年小鼠,磁头于小鼠的颅顶部每天进行10组频率为25赫兹的磁刺激,总共刺激脉冲数为1000个,连续14天。
     (2)老年sham组:16-17月龄老年小鼠,以磁头反面于小鼠颅顶部进行与rTMS组相似的无功能的磁刺激。
     (3)青年组:3-4月龄青年小鼠,与老年sham组小鼠刺激方式相同。
     各组动物在相同环境下饲养,然后通过行为学实验测试后,应用气质联用方法进行脑组织代谢产物的测试。
     结果:脑组织代谢物轮廓在老化过程中和rTMS后的变化。PLS-DA得分图显示,三组样本被分布到了不同的区域。前两个主成分的累积R2Y是0.84,Q2是0.69。根据VIP>1,91种变量被选出来作为差异变量,其中一些变量被确定来自同一中代谢物,合并这些来自同一代谢产物的变量后,共得到23种差异代谢产物。
     进一步应用one-wayANOVA对这些差异代谢物进行进一步统计,发现这23种代谢物在青年组、老年组和老年磁刺激组具有显著性差异。与青年组相比,16种代谢产物在老年组发生了显著变化,其中丙氨酸、磷酸、丝氨酸、苏氨酸、苹果酸、乳酸、尿素和肌醇的含量显著下降,GABA、柠檬酸、油酸、5,8,11,14,17-二十碳五烯酸、单硬脂酸甘油脂、反式-9-十八碳烯酸、抗坏血酸和胆固醇含量显著增多(P<0.05)。与老年对照组小鼠相比,在老年rTMS组小鼠,21种代谢产物发生了变化,其中磷酸、富马酸、苏氨酸、苹果酸、柠檬酸、丙氨酸、尿素、GABA、丝氨酸、焦磷酸、乳酸、焦谷氨酸、天冬氨酸、肌酐、天冬氨酸和胆固醇含量显著减少(P<0.05),油酸、5,8,11,14,17-二十碳五烯酸、N乙酰天冬氨酸、甘油磷酸含量显著增多(P<0.05)。
     根据所筛选出的23种代谢差异物对青年组、老年组和老年rTMS组进行验证性主成分分析。得分图显示,根据前两个主成分,青年组、老年组和老年磁刺激组被显著区分开来,R2X和Q2分别是0.60和0.45。
     对所有差异代谢产物进行变化趋势分析,胆固醇、GABA、抗坏血酸和柠檬酸为所筛选出的rTMS逆转脑组织老化过程中的代谢产物。对被动逃避反应实验的成绩和胆固醇、GABA、抗坏血酸和柠檬酸的水平进行相关性分析。发现,胆固醇和被动逃避潜伏期的Pearson Correlation是-0.413(P<0.05),GABA、抗坏血酸和柠檬酸和被动逃避潜伏期的PearsonCorrelation是-0.25、-0.080和-0.273。
     上述结果表明,rTMS可以改善老化过程中认知功能相关的代谢产物紊乱,在这些差异代谢产物中,胆固醇可能是一个较为特殊、重要的认知功能相关的代谢标志物。
     结论
     (1)昆明小鼠的认知功能在青年组和成年组没有出现明显差异,而在16月龄时开始出现下降,应用rTMS可以改善老化引起的认知功能损伤。
     (2)rTMS可以通过改善VDCC来调节神经元兴奋性,进而改善老年小鼠的认知功能障碍。
     (3)rTMS可影响脑内代谢产物的变化,改善老化过程中认知功能相关的代谢产物紊乱,在这些差异代谢产物中,胆固醇可能是一个较为特殊、重要的认知功能相关的代谢标志物。
Repetitive transcranial magnetic stimulation (rTMS) is a noninvasivebrain stimulation technique that has recently received increasing interests as atherapeutic neurorehabilitative tool. Studies confirmed that chronic highfrequency rTMS ameliorated cognitive impairment in normally agingindividuals and patients with Alzheimer disease. The function of rTMS tocognition was related to multiple pathways and networks. rTMS can increasethe excitability of the neuron, and promote the neuronal synaptic plasticity.These aftereffects have been proposed to represent long-lasting changes ofsynaptic efficacy and were therefore termed ‘long-term potentiation (LTP)like’ phenomena. In parallel with the increased LTP recorded fromhippocampal slices, there was a significant enhancement of cognitiveperformance in behavioral test in animals exposed to high-frequency rTMS.Several studies have shown that rTMS promoted neuronal plasticity relatedgenes, proteins expression, and metabolites, remodeling of the neurons.However, the mechanisms underlying the improvement in the neuronalexcitability and cognitive performance after high-frequency rTMS have notbeen well understood.
     Brain aging is associated with structural and functional changes thatinvariably lead to impairment in cognitive functions even in healthyindividuals, as well as to changes that increase the brain’s susceptibility toneurodegenerative disorders. Rodents offer several benefits as models toinvestigate the mechanisms and to identify the potential treatment ofage-related cognitive decline. Such as, similar to humans, information thatrequires prefrontal cortex and hippocampus processing is particularlyvulnerable to ageing. Previous study reported that Kunming mice as an ideal animal model exhibited an age-related cognitive impairment during normalaging. Passive avoidance task and novel object recognition test were theclassic methods to appraise the cognitive ability in rodent.
     It is well accepted that the hippocampus and prefrontal cortex is a veryimportant region related to learning and memory and also known as the mostvulnerable region affected by the internal and external changes such as aging,stroke or other neurodegenerative diseases. Studies indicated that the neuronalexcitability in hippocampus decreased during normal aging, represented asenhanced after-hyperpolarization (AHP) and hyperpolarized resting membranepotential (RMP). Such neuronal hypoexcitability, when placed within thecontext of an active neuronal network, will likely result in less efficienttransmission of information encoded in the form of action potential (AP),which in turn will reduce the computational efficacy of the network. Severalstudies shown that compared with younger counterparts, the reduced intrinsicneuronal excitability in central nervous system structure in aged individualsplayed a crucial role in the impairment of cognitive processing. The materialbasis of specific mechanisms to reduce neuronal excitability changes in thepathophysiological stimuli with a mode of action of multi-system changes.Metabonomics combined with untargeted multivariate analysis, has beenextensively applied to many fields, such as understanding the diseases of thebiochemical basis in the process of diagnosis and treatment according to themetabolic profiles in biological fluids and tissues. In the study with untargetedmultivariate analysis to analyze cerebrospinal fluid of AD patients found thatcompared with normal individuals, metabolism in AD patients was obviouslychanged.
     In this study, we observed the application of rTMS to improve theage-related cognition impairment, and the internal mechanisms and materialbasis.
     1rTMS improved age-related cognitive dysfunction
     Application of passive avoidance response task and novel objectrecognition (NOR) task to test the cognition of mice,we observed that whether rTMS can improve cognitive impairment in aged Kunming mice.
     Methods:3-4month-old Kunming mice,9-10month-old Kunming mice,and16-17month-old Kunming mice were housed with conditions oftemperature (20-25°C) to adapt the laboratory environment, and then used forthe experiments.
     Groups:
     (1) Aged rTMS group: mice (16-17months old) exposed to highfrequency rTMS (25Hz) with the coil placed just above the head of the micefor14consecutive-days,10trains per day,100pulses per train.
     (2) Aged group: mice (16-17months old) were treated similar to agedrTMS mice by the reverse side of the coil without rTMS effect.
     (3) Adoult group: mice (9-10months old) were treated same as agedmice by the reverse side of the coil for a sham purpose.
     (4) Young group: mice (3-4months old) were treated same as aged miceby the reverse side of the coil for a sham purpose.
     Each group of animals housed in the same environment, and then testedwith passive avoidance response task and novel object recognition task.
     Result: the performance of NOR: The total time of the mice touching thetwo objects was no significant difference between young, matured and agedmice during the NOR test at1h or24h point. It was found that compared withyoung and matured mice, the cognitive index decreased significantly in agedmice in the NOR test at either1h or24h point (P<0.05). The total time of themice touching the two objects was not significantly different between rTMSand sham mice at1h or24h point. The aged mice in the sham group devotedmore time exploring familiar object than novel object, while the aged mice inrTMS group spent more time in exploring the novel object than familiar object.Thus, the cognitive index in rTMS group tested at either1h or24h increased,compared with those in the sham group (P<0.05).
     The latency of passive avoidance test was not significantly differentamong young mice, matured mice, aged mice and aged rTMS mice inadaptation trial indicated that aging and rTMS did not affect the native tendency of rodent step-down off a small, elevated platform to a corner. Timesof electric shock in aged mice were significantly increased, compared withyoung mice in the acquisition sessions (P<0.05), and no difference betweenyoung group and matured mice, and application of rTMS in aged mice coulddecrease the times of electric shock significantly (P<0.05). It was found thatcompared with the young mice, passive avoidance latency in matured micewas no significant difference, and passive avoidance latency in aged micesignificantly decreased (P<0.05). The latency in aged mice delivered14successive-days rTMS increased significantly (P<0.05), compared with agedmice without rTMS effect.
     These data showed that, cognition was no difference between younggroup and adult group, and age-dependent learning and memory deficits canbe detected at16months of age in Kunming mice. The deficits of cognition inthe aged mice were significantly improved by chronic high-frequency rTMS.
     2The electrophysiological mechanisms of rTMS to improve theage-related cognition impariment
     We observed that whether rTMS improved cognitive dysfunction in agedmice by modulating neuronal excitability by regulating Voltage-dependentCa2+current (VDCC).
     Methods:3-4month-old Kunming mice,9-10month-old Kunming mice,and16-17month-old Kunming mice were housed with conditions oftemperature (20-25°C), and then used for the experiments.
     Groups:
     (1) Aged rTMS group: mice (16-17months old) exposed to highfrequency rTMS (25Hz) with the coil placed just above the head of the micefor14consecutive-days,10trains per day,100pulses per train.
     (2) Aged group: mice (16-17months old) were treated similar to agedrTMS mice by the reverse side of the coil without rTMS effect.
     (3) Young group: mice (3-4months old) were treated same as aged miceby the reverse side of the coil for a sham purpose.
     Each group of animals housed in the same environment, and then recorded the electrophysiological indexes, after the behavioral task.Result: Whole-cell current clamp recordings revealed that the RMP was morehyperpolarized in CA1pyramidal neurons (CA1-PNs) in slices prepared fromaged mice compared with young mice (P<0.05). Application of rTMSsignificantly elevated the RMP in CA1-PNs prepared from aged micecompared with the aged mice without rTMS (P<0.05). The number of APs wassignificantly reduced in CA1-PNs of aged mice compared with those of youngmice (P<0.05). High-frequency rTMS significantly increased the number ofAPs elicited by the current injection in CA1-PNs of aged mice compared withthose without rTMS (P<0.05). The amplitude of AHP after a series of APs wassignificantly increased in aged mice compared with that in young mice(P<0.05). Application of rTMS significantly decreased the AHP amplitude inthe aged mice compared with the aged mice without rTMS (P<0.05). Thethreshold potential of the first AP fired in response to a300-pA currentinjection revealed no significant differences among matured, aged and agedrTMS mice.
     VDCC of the CA1-PNs was recorded with whole-cell patch clamp involtage-clamped at-50mV and depolarized from-40mV to20mVincremental10mV voltage steps with150-ms duration. The full currentdensity-voltage curves indicated that Ca2+currents recorded from aged rTMSneurons were decreased compared with those from aged sham neurons.Analysis indicated that the Ca2+current density was significantly smaller fromaged rTMS neurons when compared with aged neurons at test potential of-20mV,-10mV and0mV (P<0.05).
     These results suggest that, rTMS can modulate neuronal excitability byregulating VDCC, to improve age-related cognitive dysfunction.3rTMS impoved cognition impairment by regulating the age-relatedmetabolites disordersThe metabolites of prefrontal cortex in mice were tested with gaschromatography mass spectrometry (GC-MS), and analyzed with partial leastsquares analysis (PLS-DA) and principal component analysis (PCA),then analsized the different metabolites with one-way ANOVA
     Methods:3-4month-old Kunming mice,9-10month-old Kunming mice,and16-17month-old Kunming mice were housed with conditions oftemperature (20-25°C), and then tested for the experiments.
     Groups:
     (1) Aged rTMS group: mice (16-17months old) exposed to highfrequency rTMS (25Hz) with the coil placed just above the head of the micefor14consecutive-days,10trains per day, and100pulses per train.
     (2) Aged group: mice (16-17months old) were treated similar to agedrTMS mice by the reverse side of the coil without rTMS effect.
     (3) Young group: mice (3-4months old) were treated same as aged miceby the reverse side of the coil for a sham purpose.
     Each group of animals housed in the same environment, and thenrecorded the metabolites in prefrontal cortex, after the behavioral task.
     Result: The score plot of the PLS-DA model showed separation ofsamples in different groups. The model generated with two components had acumulative R2Y of0.84and a cumulative Q2of0.69. According to the valueof VIP>1and after merging the variables from the same metabolites,23identified variables were collected.To accurately evaluate the changes of metabolites level, one-way ANOVA andpost hoc analysis was employed to these metabolites ratios, and significantdifferences were found in the23variables from young group, aged group andaged rTMS group, which were considered as the potential different biomarkers.Plot data of above indicated that compared with young group,16metaboliteswere altered significantly in aged mice, metabolites of Alanine (Ala),Phosphoric acid (Pho), Serine (Ser), Threonine (Thr), Malic acid (Mal), Lacticacid (Lac), Urea and Myo-Inositol (M-In) decreased significantly (P<0.05).Metabolites of Gamma-Aminobutyric acid (GABA), Citric acid (Cit), Oleicacid (Ole),5,8,11,14,17-Eicosapentaenoic acid (Eic), Monostearin (M-Ste),Trans-9-Octadecenoic acid (Oct), Ascorbic acid (Asc) and Cholesterol (Cho)were significantly increased during aging (P<0.05). Compared with aged group,21metabolites were altered in aged rTMS mice. It could be found thatcompared with aged mice metabolites of Pho, Fumaric acid (Fum), Thr, Mal,Cit, Ala, Urea, GABA, Ser, Pyrophosphate (P-Pho), M-In, Lac, Pyroglutamicacid (P-Glu), Aspartic acid (Asp), Creatinine (Cre), Asc and Cho decreasedsignificantly (P<0.05), while metabolites of Ole, Eic, N Acetyl aspartic (NAA),Phosphaglyceride (P-Gly) were increased significantly (P<0.05).
     Based on selected23difference metabolites from young mice, aged mice,and aged rTMS mice, PCA analysis was used as the verify classificationmethod for modeling the discrimination. The score plots of the first twoprincipal components allowed visualization of the data and comparing thethree-group samples. The R2X and Q2were0.60and0.45. The PCA scoreplot showed the samples from different groups were scattered into threedifferent regions.
     Cholesterol, GABA, ascorbic acid and citric acid are as the screenedmetabolites of brain tissue by the way of rTMS reversed metabolic profile inthe aging process. The correlation between the performance of passiveavoidance task and the levels of metabolites of cholesterol, GABA, Asc, andCit in the tested mice were assessed. The Pearson Correlation of cholesterolwith passive avoidance latency is-0.413(P<0.05), and metabolites of GABA,Asc, and Cit with the avoidance latency is-0.25,-0.080, and-0.273withoutsignificant difference.
     The above results show that, rTMS can improve cognition-relatedmetabolites disorders during aging.
     Conclution
     (1) These data showed that under the conditions used here, cognitionindex was no significant difference between young group and adult group, andage-dependent learning and memory deficits can be detected at16months inKunming mice. The deficits of cognition in the aged mice were significantlyimproved by chronic high-frequency rTMS.
     (2) These results suggest that, rTMS can modulate neuronal excitabilityby regulating VDCC, to improve age-related cognitive dysfunction.
     (3) The above results show that, rTMS can improve cognition-relatedmetabolites disorders during aging, and cholesterol may be one of the mostimportant metabolites correlated with cognition.
引文
1Hallett M. Transcranial magnetic stimulation: a primer. Neuron,2007,55:187~199
    2Nardone R, Bergmann J, Christova M, et al. Effect of transcranial brainstimulation for the treatment of Alzheimer disease: a review. Int JAlzheimers Dis,2012,2012:687909
    3Guse B, Falkai P, Wobrock T. Cognitive effects of high~frequencyrepetitive transcranial magnetic stimulation: a systematic review. JNeural Transm,2010,117:105~122
    4Kim SH, Han HJ, Ahn HM, et al. Effects of five daily high~frequencyrTMS on Stroop task performance in aging individuals. Neurosci Res,2012,74:256~260
    5Foster TC. Dissecting the age~related decline on spatial learning andmemory tasks in rodent models: N~methyl~D~aspartate receptors andvoltage~dependent Ca2+channels in senescent synaptic plasticity. ProgNeurobiol,2012,96:283~303
    6Morrison JH, Baxter MG. The ageing cortical synapse: hallmarks andimplications for cognitive decline. Nat Rev Neurosci,2012,13:240~250
    7Chen GH, Wang YJ, Zhang LQ, et al. Age~and sex~related disturbancein a battery of sensorimotor and cognitive tasks in Kunming mice.Physiol Behav,2004,83:531~541
    8Giovannini MG, Pazzagli M, Malmberg~Aiello P, et al. Inhibition ofacetylcholine~induced activation of extracellular regulated proteinkinase prevents the encoding of an inhibitory avoidance response in therat. Neuroscience,2005,136:15~32
    9Romanova GA, Shakova FM, Gorbatov VY, et al. Effect of antibodiesto glutamate on retention of conditioned passive avoidance response inrats with ischemic injury of the prefrontal cortex. Bull Exp Biol Med,2010,149:289~292
    10Lee CH, Yoo KY, Choi JH, et al. Neuronal damage is much delayed andmicrogliosis is more severe in the aged hippocampus induced bytransient cerebral ischemia compared to the adult hippocampus. JNeurol Sci,2010,294:1~6
    11Vanguilder HD, Freeman WM. The hippocampal neuroproteome withaging and cognitive decline: past progress and future directions. FrontAging Neurosci,2011,3:8
    12West MJ, Coleman PD, Flood DG, et al. Differences in the pattern ofhippocampal neuronal loss in normal ageing and Alzheimer's disease.Lancet,1994,344:769~772
    13Thickbroom GW. Transcranial magnetic stimulation and synapticplasticity: experimental framework and human models. Exp Brain Res,2007,180:583~593
    14Ahmed Z, Wieraszko A. Modulation of learning and hippocampal,neuronal plasticity by repetitive transcranial magnetic stimulation(rTMS). Bioelectromagnetics,2006,27:288~294
    1Hallett M. Transcranial magnetic stimulation: a primer. Neuron,2007,55:187~199
    2Nardone R, Bergmann J, Christova M, et al. Effect of transcranial brainstimulation for the treatment of Alzheimer disease: a review. Int JAlzheimers Dis,2012,2012:687909
    3Ziemann U, Paulus W, Nitsche MA, et al. Consensus: Motor cortexplasticity protocols. Brain Stimul,2008,1:164~182
    4Thickbroom GW. Transcranial magnetic stimulation and synapticplasticity: experimental framework and human models. Exp Brain Res,2007,180:583~593
    5Ahmed Z, Wieraszko A. Modulation of learning and hippocampal,neuronal plasticity by repetitive transcranial magnetic stimulation(rTMS). Bioelectromagnetics,2006,27:288~294
    6Lee CH, Yoo KY, Choi JH, et al. Neuronal damage is much delayed andmicrogliosis is more severe in the aged hippocampus induced bytransient cerebral ischemia compared to the adult hippocampus. JNeurol Sci,2010,294:1~6
    7Vanguilder HD, Freeman WM. The hippocampal neuroproteome withaging and cognitive decline: past progress and future directions. FrontAging Neurosci,2011,3:8
    8West MJ, Coleman PD, Flood DG, et al. Differences in the pattern ofhippocampal neuronal loss in normal ageing and Alzheimer's disease.Lancet,1994,344:769~772
    9Disterhoft JF, Oh MM. Alterations in intrinsic neuronal excitabilityduring normal aging. Aging Cell,2007,6:327~336
    10Randall AD, Booth C, Brown JT. Age~related changes to Na+channelgating contribute to modified intrinsic neuronal excitability. NeurobiolAging,2012,33:2715~2720
    11Burgdorf J, Kroes RA, Weiss C, et al. Positive emotional learning isregulated in the medial prefrontal cortex by GluN2B~containingNMDA receptors. Neuroscience,2011,192:515~523
    12Moyer JR, Jr., Thompson LT, Black JP, et al. Nimodipine increasesexcitability of rabbit CA1pyramidal neurons in an age~andconcentration~dependent manner. J Neurophysiol,1992,68:2100~2109
    13Power JM, Wu WW, Sametsky E, et al. Age~related enhancement of theslow outward calcium~activated potassium current in hippocampal CA1pyramidal neurons in vitro. J Neurosci,2002,22:7234~7243
    14Landfield PW, Pitler TA. Prolonged Ca2+~dependentafterhyperpolarizations in hippocampal neurons of aged rats. Science,1984,226:1089~1092
    15Matthews EA, Linardakis JM, Disterhoft JF. The fast and slowafterhyperpolarizations are differentially modulated in hippocampalneurons by aging and learning. J Neurosci,2009,29:4750~4755
    16Oh MM, Oliveira FA, Disterhoft JF. Learning and aging related changesin intrinsic neuronal excitability. Front Aging Neurosci,2010,2:2
    17Foster TC. Dissecting the age~related decline on spatial learning andmemory tasks in rodent models: N~methyl~D~aspartate receptors andvoltage~dependent Ca2+channels in senescent synaptic plasticity. ProgNeurobiol,2012,96:283~303
    18Thibault O, Landfield PW. Increase in single L~type calcium channelsin hippocampal neurons during aging. Science,1996,272:1017~1020
    19Deyo RA, Straube KT, Disterhoft JF. Nimodipine facilitates associativelearning in aging rabbits. Science,1989,243:809~811
    20Kumar A, Foster TC.17beta~estradiol benzoate decreases the AHPamplitude in CA1pyramidal neurons. J Neurophysiol,2002,88:621~626
    21Norris CM, Halpain S, Foster TC. Reversal of age~related alterations insynaptic plasticity by blockade of L~type Ca2+channels. J Neurosci,1998,18:3171~3179
    22Rose GM, Ong VS, Woodruff~Pak DS. Efficacy of MEM1003, a novelcalcium channel blocker, in delay and trace eyeblink conditioning inolder rabbits. Neurobiol Aging,2007,28:766~773
    23Naundorf B, Wolf F, Volgushev M. Unique features of action potentialinitiation in cortical neurons. Nature,2006,440:1060~1063
    24Ahmed MA, Darwish ES, Khedr EM, et al. Effects of low versus highfrequencies of repetitive transcranial magnetic stimulation on cognitivefunction and cortical excitability in Alzheimer's dementia. J Neurol,2012,259:83~92
    25Kim SH, Han HJ, Ahn HM, et al. Effects of five daily high~frequencyrTMS on Stroop task performance in aging individuals. Neurosci Res,2012,74:256~260
    26Guse B, Falkai P, Wobrock T. Cognitive effects of high~frequencyrepetitive transcranial magnetic stimulation: a systematic review. JNeural Transm,2010,117:105~122
    27Rosenzweig ES, Barnes CA. Impact of aging on hippocampal function:plasticity, network dynamics, and cognition. Prog Neurobiol,2003,69:143~179
    28McKay BM, Matthews EA, Oliveira FA, et al. Intrinsic neuronalexcitability is reversibly altered by a single experience in fearconditioning. J Neurophysiol,2009,102:2763~2770
    29Wu WW, Oh MM, Disterhoft JF. Age~related biophysical alterations ofhippocampal pyramidal neurons: implications for learning and memory.Ageing Res Rev,2002,1:181~207
    30Disterhoft JF, Oh MM. Pharmacological and molecular enhancement oflearning in aging and Alzheimer's disease. J Physiol Paris,2006,99:180~192
    31Miranda PC, Hallett M, Basser PJ. The electric field induced in thebrain by magnetic stimulation: a3~D finite~element analysis of theeffect of tissue heterogeneity and anisotropy. IEEE Trans Biomed Eng,2003,50:1074~1085
    32Wassermann EM, Lisanby SH. Therapeutic application of repetitivetranscranial magnetic stimulation: a review. Clin Neurophysiol,2001,112:1367~1377
    33Huerta PT, Volpe BT. Transcranial magnetic stimulation, synapticplasticity and network oscillations. J Neuroeng Rehabil,2009,6:7
    34Gant JC, Thibault O. Action potential throughput in aged rathippocampal neurons: regulation by selective forms ofhyperpolarization. Neurobiol Aging,2009,30:2053~2064
    35Zhang L, McBain CJ. Potassium conductances underlyingrepolarization and after~hyperpolarization in rat CA1hippocampalinterneurones. J Physiol,1995,488(Pt3):661~672
    36Sun P, Wang F, Wang L, et al. Increase in cortical pyramidal cellexcitability accompanies depression~like behavior in mice: atranscranial magnetic stimulation study. J Neurosci,2011,31:16464~16472
    37Burke SN, Barnes CA. Senescent synapses and hippocampal circuitdynamics. Trends Neurosci,2010,33:153~161
    38Vlachos A, Muller~Dahlhaus F, Rosskopp J, et al. Repetitive magneticstimulation induces functional and structural plasticity of excitatorypostsynapses in mouse organotypic hippocampal slice cultures. JNeurosci,2012,32:17514~17523
    39Cheeran B, Talelli P, Mori F, et al. A common polymorphism in thebrain~derived neurotrophic factor gene (BDNF) modulates humancortical plasticity and the response to rTMS. J Physiol,2008,586:5717~5725
    40Wang F, Geng X, Tao HY, et al. The restoration after repetitivetranscranial magnetic stimulation treatment on cognitive ability ofvascular dementia rats and its impacts on synaptic plasticity inhippocampal CA1area. J Mol Neurosci,2010,41:145~155
    41Armstrong CM, Hille B. Voltage~gated ion channels and electricalexcitability. Neuron,1998,20:371~380
    42Catterall WA. Structure and regulation of voltage~gated Ca2+channels.Annu Rev Cell Dev Biol,2000,16:521~555
    43Yamakage M, Namiki A. Calcium channels~~basic aspects of theirstructure, function and gene encoding; anesthetic action on thechannels~~a review. Can J Anaesth,2002,49:151~164
    44Shkryl VM, Nikolaenko LM, Kostyuk PG, et al. High~thresholdcalcium channel activity in rat hippocampal neurones during hypoxia.Brain Res,1999,833:319~328
    45Lukyanetz EA, Shkryl VM, Kravchuk OV, et al. Action of hypoxia ondifferent types of calcium channels in hippocampal neurons. BiochimBiophys Acta,2003,1618:33~38
    1Morrison JH, Baxter MG. The ageing cortical synapse: hallmarks andimplications for cognitive decline. Nat Rev Neurosci,2012,13:240~250
    2Tisserand DJ, Jolles J. On the involvement of prefrontal networks incognitive ageing. Cortex,2003,39:1107~1128
    3Giovannini MG, Pazzagli M, Malmberg~Aiello P, et al. Inhibition ofacetylcholine~induced activation of extracellular regulated proteinkinase prevents the encoding of an inhibitory avoidance response in therat. Neuroscience,2005,136:15~32
    4Romanova GA, Shakova FM, Gorbatov VY, et al. Effect of antibodiesto glutamate on retention of conditioned passive avoidance response inrats with ischemic injury of the prefrontal cortex. Bull Exp Biol Med,2010,149:289~292
    5Hallett M. Transcranial magnetic stimulation: a primer. Neuron,2007,55:187~199
    6Guse B, Falkai P, Wobrock T. Cognitive effects of high~frequencyrepetitive transcranial magnetic stimulation: a systematic review. JNeural Transm,2010,117:105~122
    7Ahmed Z, Wieraszko A. Modulation of learning and hippocampal,neuronal plasticity by repetitive transcranial magnetic stimulation(rTMS). Bioelectromagnetics,2006,27:288~294
    8Hellmann J, Juttner R, Roth C, et al. Repetitive magnetic stimulation ofhuman~derived neuron~like cells activates cAMP~CREB pathway. EurArch Psychiatry Clin Neurosci,2012,262:87~91
    9Ma J, Zhang Z, Su Y, et al. Magnetic stimulation modulates structuralsynaptic plasticity and regulates BDNF~TrkB signal pathway incultured hippocampal neurons. Neurochem Int,2013,62:84~91
    10Keck ME, Sillaber I, Ebner K, et al. Acute transcranial magneticstimulation of frontal brain regions selectively modulates the release ofvasopressin, biogenic amines and amino acids in the rat brain. Eur JNeurosci,2000,12:3713~3720
    11Michael N, Gosling M, Reutemann M, et al. Metabolic changes afterrepetitive transcranial magnetic stimulation (rTMS) of the left prefrontalcortex: a sham~controlled proton magnetic resonance spectroscopy (1HMRS) study of healthy brain. Eur J Neurosci,2003,17:2462~2468
    12Ben~Shachar D, Belmaker RH, Grisaru N, et al. Transcranial magneticstimulation induces alterations in brain monoamines. J Neural Transm,1997,104:191~197
    13Nicholson JK, Connelly J, Lindon JC, et al. Metabonomics: a platformfor studying drug toxicity and gene function. Nat Rev Drug Discov,2002,1:153~161
    14Yin P, Mohemaiti P, Chen J, et al. Serum metabolic profiling ofabnormal savda by liquid chromatography/mass spectrometry. JChromatogr B Analyt Technol Biomed Life Sci,2008,871:322~327
    15Kellert M, Wagner S, Lutz U, et al. Biomarkers of furan exposure bymetabolic profiling of rat urine with liquid chromatography~tandemmass spectrometry and principal component analysis. Chem ResToxicol,2008,21:761~768
    16Jia L, Wang C, Kong H, et al. Effect of PA~MSHA vaccine on plasmaphospholipids metabolic profiling and the ratio of Th2/Th1cells withinimmune organ of mouse IgA nephropathy. J Pharm Biomed Anal,2007,43:646~654
    17Wang Y, Wang J, Yao M, et al. Metabonomics study on the effects of theginsenoside Rg3in a beta~cyclodextrin~based formulation ontumor~bearing rats by a fully automatic hydrophilicinteraction/reversed~phase column~switching HPLC~ESI~MSapproach. Anal Chem,2008,80:4680~4688
    18Czech C, Berndt P, Busch K, et al. Metabolite profiling of Alzheimer'sdisease cerebrospinal fluid. PLoS One,2012,7: e31501
    19Meng J, Zhang X, Wu H, et al. Morphine~induced conditioned placepreference in mice: metabolomic profiling of brain tissue to find"molecular switch" of drug abuse by gas chromatography/massspectrometry. Anal Chim Acta,2012,710:125~130
    20Yao H, Shi P, Zhang L, et al. Untargeted metabolic profiling revealspotential biomarkers in myocardial infarction and its application. MolBiosyst,2010,6:1061~1070
    21Wu H, Xue R, Lu C, et al. Metabolomic study for diagnostic model ofoesophageal cancer using gas chromatography/mass spectrometry. JChromatogr B Analyt Technol Biomed Life Sci,2009,877:3111~3117
    22Jonsson P, Bruce SJ, Moritz T, et al. Extraction, interpretation andvalidation of information for comparing samples in metabolic LC/MSdata sets. Analyst,2005,130:701~707
    23Paban V, Fauvelle F, Alescio~Lautier B. Age~related changes inmetabolic profiles of rat hippocampus and cortices. Eur J Neurosci,2010,31:1063~1073
    24Zhang X, Liu H, Wu J, et al. Metabonomic alterations in hippocampus,temporal and prefrontal cortex with age in rats. Neurochem Int,2009,54:481~487
    25Yoshiike Y, Kimura T, Yamashita S, et al. GABA(A) receptor~mediatedacceleration of aging~associated memory decline in APP/PS1mice andits pharmacological treatment by picrotoxin. PLoS One,2008,3: e3029
    26Lasarge CL, Banuelos C, Mayse JD, et al. Blockade of GABA(B)receptors completely reverses age~related learning impairment.Neuroscience,2009,164:941~947
    27Marjanska M, Lehericy S, Valabregue R, et al. Brain dynamicneurochemical changes in dystonic patients: A magnetic resonancespectroscopy study. Mov Disord,2013,28:201~209
    28Frei B, Stocker R, England L, et al. Ascorbate: the most effectiveantioxidant in human blood plasma. Adv Exp Med Biol,1990,264:155~163
    29Head E. Oxidative damage and cognitive dysfunction: antioxidanttreatments to promote healthy brain aging. Neurochem Res,2009,34:670~678
    30Harrison FE. A critical review of vitamin C for the prevention ofage~related cognitive decline and Alzheimer's disease. J Alzheimers Dis,2012,29:711~726
    31Bowman GL. Ascorbic acid, cognitive function, and Alzheimer'sdisease: a current review and future direction. Biofactors,2012,38:114~122
    32Dietschy JM, Turley SD. Cholesterol metabolism in the brain. CurrOpin Lipidol,2001,12:105~112
    33Tsui~Pierchala BA, Encinas M, Milbrandt J, et al. Lipid rafts inneuronal signaling and function. Trends Neurosci,2002,25:412~417
    34Toman RE, Spiegel S, Faden AI. Role of ceramide in neuronal celldeath and differentiation. J Neurotrauma,2000,17:891~898
    35Cutler RG, Kelly J, Storie K, et al. Involvement of oxidativestress~induced abnormalities in ceramide and cholesterol metabolism inbrain aging and Alzheimer's disease. Proc Natl Acad Sci U S A,2004,101:2070~2075
    36Yu ZF, Nikolova~Karakashian M, Zhou D, et al. Pivotal role for acidicsphingomyelinase in cerebral ischemia~induced ceramide and cytokineproduction, and neuronal apoptosis. J Mol Neurosci,2000,15:85~97
    37Hartfield PJ, Mayne GC, Murray AW. Ceramide induces apoptosis inPC12cells. FEBS Lett,1997,401:148~152
    38Kellner~Weibel G, Geng YJ, Rothblat GH. Cytotoxic cholesterol isgenerated by the hydrolysis of cytoplasmic cholesteryl ester andtransported to the plasma membrane. Atherosclerosis,1999,146:309~319
    39Genestier L, Prigent AF, Paillot R, et al. Caspase~dependent ceramideproduction in Fas~and HLA class I~mediated peripheral T cellapoptosis. J Biol Chem,1998,273:5060~5066
    40Kellner~Weibel G, Jerome WG, Small DM, et al. Effects of intracellularfree cholesterol accumulation on macrophage viability: a model forfoam cell death. Arterioscler Thromb Vasc Biol,1998,18:423~431
    41Wang L, Schuster GU, Hultenby K, et al. Liver X receptors in thecentral nervous system: from lipid homeostasis to neuronaldegeneration. Proc Natl Acad Sci U S A,2002,99:13878~13883
    42Eckert GP, Vardanian L, Rebeck GW, et al. Regulation of centralnervous system cholesterol homeostasis by the liver X receptor agonistTO~901317. Neurosci Lett,2007,423:47~52
    43Hooijmans CR, Kiliaan AJ. Fatty acids, lipid metabolism and Alzheimerpathology. Eur J Pharmacol,2008,585:176~196
    44Hirsch~Reinshagen V, Burgess BL, Wellington CL. Why lipids areimportant for Alzheimer disease? Mol Cell Biochem,2009,326:121~129
    45Kim J, Basak JM, Holtzman DM. The role of apolipoprotein E inAlzheimer's disease. Neuron,2009,63:287~303
    46Jansen D, Janssen CI, Vanmierlo T, et al. Cholesterol and synapticcompensatory mechanisms in Alzheimer's disease mice brain duringaging. J Alzheimers Dis,2012,31:813~826
    47Honjo M, Tanihara H, Nishijima K, et al. Statin inhibitsleukocyte~endothelial interaction and prevents neuronal death inducedby ischemia~reperfusion injury in the rat retina. Arch Ophthalmol,2002,120:1707~1713
    48Buxbaum JD, Geoghagen NS, Friedhoff LT. Cholesterol depletion withphysiological concentrations of a statin decreases the formation of theAlzheimer amyloid Abeta peptide. J Alzheimers Dis,2001,3:221~229
    1Henderson G, Tomlinson BE, Gibson PH. Cell counts in human cerebralcortex in normal adults throughout life using an image analysingcomputer. J Neurol Sci,1980,46:113~136
    2Devaney KO, Johnson HA. Neuron loss in the aging visual cortex ofman. J Gerontol,1980,35:836~841
    3West MJ, Slomianka L, Gundersen HJ. Unbiased stereologicalestimation of the total number of neurons in thesubdivisions of the rathippocampus using the optical fractionator. Anat Rec,1991,231:482~497
    4Rapp PR, Gallagher M. Preserved neuron number in the hippocampusof aged rats with spatial learning deficits. Proc Natl Acad Sci U S A,1996,93:9926~9930
    5Rasmussen T, Schliemann T, Sorensen JC, et al. Memory impaired agedrats: no loss of principal hippocampal and subicular neurons. NeurobiolAging,1996,17:143~147
    6Calhoun ME, Kurth D, Phinney AL, et al. Hippocampal neuron andsynaptophysin~positive bouton number in aging C57BL/6mice.Neurobiol Aging,1998,19:599~606
    7Burke SN, Barnes CA. Neural plasticity in the ageing brain. Nat RevNeurosci,2006,7:30~40
    8Martin SJ, Grimwood PD, Morris RG. Synaptic plasticity and memory:an evaluation of the hypothesis. Annu Rev Neurosci,2000,23:649~711
    9Sorra KE, Harris KM. Overview on the structure, composition, function,development, and plasticity of hippocampal dendritic spines.Hippocampus,2000,10:501~511
    10Harris KM. Structure, development, and plasticity of dendritic spines.Curr Opin Neurobiol,1999,9:343~348
    11Geinisman Y, deToledo~Morrell L, Morrell F, et al. Age~related loss ofaxospinous synapses formed by two afferent systems in the rat dentategyrus as revealed by the unbiased stereological dissector technique.Hippocampus,1992,2:437~444
    12Poe BH, Linville C, Riddle DR, et al. Effects of age and insulin~likegrowth factor~1on neuron and synapse numbers in area CA3ofhippocampus. Neuroscience,2001,107:231~238
    13Geinisman Y, Ganeshina O, Yoshida R, et al. Aging, spatial learning,and total synapse number in the rat CA1stratum radiatum. NeurobiolAging,2004,25:407~416
    14Geinisman Y. Structural synaptic modifications associated withhippocampal LTP and behavioral learning. Cereb Cortex,2000,10:952~962
    15Buchs PA, Muller D. Induction of long~term potentiation is associatedwith major ultrastructural changes of activated synapses. Proc NatlAcad Sci U S A,1996,93:8040~8045
    16Toni N, Buchs PA, Nikonenko I, et al. Remodeling of synapticmembranes after induction of long~term potentiation. J Neurosci,2001,21:6245~6251
    17Hering H, Sheng M. Dendritic spines: structure, dynamics andregulation. Nat Rev Neurosci,2001,2:880~888
    18Shi L, Linville MC, Tucker EW, et al. Differential effects of aging andinsulin~like growth factor~1on synapses in CA1of rat hippocampus.Cereb Cortex,2005,15:571~577
    19Nicholson DA, Yoshida R, Berry RW, et al. Reduction in size ofperforated postsynaptic densities in hippocampal axospinous synapsesand age~related spatial learning impairments. J Neurosci,2004,24:7648~7653
    20Benkovic SA, Connor JR. Ferritin, transferrin, and iron in selectedregions of the adult and aged rat brain. J Comp Neurol,1993,338:97~113
    21Connor JR, Pavlick G, Karli D, et al. A histochemical study ofiron~positive cells in the developing rat brain. J Comp Neurol,1995,355:111~123
    22Cotrina ML, Gao Q, Lin JH, et al. Expression and function of astrocyticgap junctions in aging. Brain Res,2001,901:55~61
    23Peters A, Moss MB, Sethares C. The effects of aging on layer1ofprimary visual cortex in the rhesus monkey. Cereb Cortex,2001,11:93~103
    24Harry GJ. Microglia during development and aging. Pharmacol Ther,2013:
    25Kohman RA. Aging microglia: relevance to cognition and neuralplasticity. Methods Mol Biol,2012,934:193~218
    26Peters A, Moss MB, Sethares C. Effects of aging on myelinated nervefibers in monkey primary visual cortex. J Comp Neurol,2000,419:364~376
    27Peters A, Sethares C. Aging and the myelinated fibers in prefrontalcortex and corpus callosum of the monkey. J Comp Neurol,2002,442:277~291
    28Malone MJ, Szoke MC. Neurochemical studies in aging brain. I.Structural changes in myelin lipids. J Gerontol,1982,37:262~267
    29Sloane JA, Hinman JD, Lubonia M, et al. Age~dependent myelindegeneration and proteolysis of oligodendrocyte proteins is associatedwith the activation of calpain~1in the rhesus monkey. J Neurochem,2003,84:157~168
    30Lichtenwalner RJ, Forbes ME, Bennett SA, et al.Intracerebroventricular infusion of insulin~like growth factor~Iameliorates the age~related decline in hippocampal neurogenesis.Neuroscience,2001,107:603~613
    31Nacher J, Alonso~Llosa G, Rosell DR, et al. NMDA receptor antagonisttreatment increases the production of new neurons in the aged rathippocampus. Neurobiol Aging,2003,24:273~284
    32Bondolfi L, Ermini F, Long JM, et al. Impact of age and caloricrestriction on neurogenesis in the dentate gyrus of C57BL/6mice.Neurobiol Aging,2004,25:333~340
    33Weinreb O, Drigues N, Sagi Y, et al. The application of proteomics andgenomics to the study of age~related neurodegeneration andneuroprotection. Antioxid Redox Signal,2007,9:169~179
    34Chen W, Ji J, Xu X, et al. Proteomic comparison between human youngand old brains by two~dimensional gel electrophoresis andidentification of proteins. Int J Dev Neurosci,2003,21:209~216
    35Feldmann RE, Jr., Maurer MH, Hunzinger C, et al. Reduction in ratphosphatidylethanolamine binding protein~1(PEBP1) after chroniccorticosterone treatment may be paralleled by cognitive impairment: afirst study. Stress,2008,11:134~147
    36Ramos JW, Townsend DA, Piarulli D, et al. Deletion of PEA~15inmice is associated with specific impairments of spatial learning abilities.BMC Neurosci,2009,10:134
    37Quintana C, Gutierrez L. Could a dysfunction of ferritin be adeterminant factor in the aetiology of some neurodegenerative diseases?Biochim Biophys Acta,2010,1800:770~782
    38Sato Y, Yamanaka H, Toda T, et al. Comparison of hippocampalsynaptosome proteins in young~adult and aged rats. Neurosci Lett,2005,382:22~26
    39VanGuilder HD, Yan H, Farley JA, et al. Aging alters the expression ofneurotransmission~regulating proteins in the hippocampalsynaptoproteome. J Neurochem,2010,113:1577~1588
    40Mendelsohn AR, Larrick JW. Epigenetic~mediated decline in synapticplasticity during aging. Rejuvenation Res,2012,15:98~101
    41Lister JP, Barnes CA. Neurobiological changes in the hippocampusduring normative aging. Arch Neurol,2009,66:829~833
    42Foster TC, Kumar A. Susceptibility to induction of long~termdepression is associated with impaired memory in aged Fischer344rats.Neurobiol Learn Mem,2007,87:522~535
    43Shankar S, Teyler TJ, Robbins N. Aging differentially alters forms oflong~term potentiation in rat hippocampal area CA1. J Neurophysiol,1998,79:334~341
    44Griffin R, Nally R, Nolan Y, et al. The age~related attenuation inlong~term potentiation is associated with microglial activation. JNeurochem,2006,99:1263~1272
    45Kelly L, Grehan B, Chiesa AD, et al. The polyunsaturated fatty acids,EPA and DPA exert a protective effect in the hippocampus of the agedrat. Neurobiol Aging,2011,32:2318e2311~2315
    46Rex CS, Kramar EA, Colgin LL, et al. Long~term potentiation isimpaired in middle~aged rats: regional specificity and reversal byadenosine receptor antagonists. J Neurosci,2005,25:5956~5966
    47Foster TC. Involvement of hippocampal synaptic plasticity inage~related memory decline. Brain Res Brain Res Rev,1999,30:236~249
    48Burke SN, Barnes CA. Senescent synapses and hippocampal circuitdynamics. Trends Neurosci,2010,33:153~161
    49Kumar A, Thinschmidt JS, Foster TC, et al. Aging effects on the limitsand stability of long~term synaptic potentiation and depression in rathippocampal area CA1. J Neurophysiol,2007,98:594~601
    50Randall AD, Booth C, Brown JT. Age~related changes to Na+channelgating contribute to modified intrinsic neuronal excitability. NeurobiolAging,2012,33:2715~2720
    51McKay BM, Matthews EA, Oliveira FA, et al. Intrinsic neuronalexcitability is reversibly altered by a single experience in fearconditioning. J Neurophysiol,2009,102:2763~2770
    52Toescu EC, Vreugdenhil M. Calcium and normal brain ageing. CellCalcium,2010,47:158~164
    53Thibault O, Landfield PW. Increase in single L~type calcium channelsin hippocampal neurons during aging. Science,1996,272:1017~1020
    54Disterhoft JF, Oh MM. Alterations in intrinsic neuronal excitabilityduring normal aging. Aging Cell,2007,6:327~336
    55LaFerla FM. Calcium dyshomeostasis and intracellular signalling inAlzheimer's disease. Nat Rev Neurosci,2002,3:862~872
    56Rose GM, Ong VS, Woodruff~Pak DS. Efficacy of MEM1003, a novelcalcium channel blocker, in delay and trace eyeblink conditioning inolder rabbits. Neurobiol Aging,2007,28:766~773
    57Kumar A, Foster TC.17beta~estradiol benzoate decreases the AHPamplitude in CA1pyramidal neurons. J Neurophysiol,2002,88:621~626
    58Hess EJ, Jen JC, Jinnah HA, et al. Neuronal voltage~gated calciumchannels: brief overview of their function and clinical implications inneurology. Neurology,2010,75:937; author reply937~938
    59Power JM, Wu WW, Sametsky E, et al. Age~related enhancement of theslow outward calcium~activated potassium current in hippocampal CA1pyramidal neurons in vitro. J Neurosci,2002,22:7234~7243
    60Disterhoft JF, Oh MM. Pharmacological and molecular enhancement oflearning in aging and Alzheimer's disease. J Physiol Paris,2006,99:180~192
    61Hell JW, Westenbroek RE, Warner C, et al. Identification anddifferential subcellular localization of the neuronal class C and class DL~type calcium channel alpha1subunits. J Cell Biol,1993,123:949~962
    62Foster TC. Dissecting the age~related decline on spatial learning andmemory tasks in rodent models: N~methyl~D~aspartate receptors andvoltage~dependent Ca2+channels in senescent synaptic plasticity. ProgNeurobiol,2012,96:283~303
    1Larson EB, Kukull WA, Katzman RL. Cognitive impairment: dementiaand Alzheimer's disease. Annu Rev Public Health,1992,13:431~449
    2Glenner GG, Wong CW. Alzheimer's disease: initial report of thepurification and characterization of a novel cerebrovascular amyloidprotein.1984. Biochem Biophys Res Commun,2012,425:534~539
    3Haass C, Selkoe DJ. Cellular processing of beta~amyloid precursorprotein and the genesis of amyloid beta~peptide. Cell,1993,75:1039~1042
    4De Strooper B, Annaert W. Proteolytic processing and cell biologicalfunctions of the amyloid precursor protein. J Cell Sci,2000,113(Pt11):1857~1870
    5De Strooper B. Aph~1, Pen~2, and Nicastrin with Presenilin generate anactive gamma~Secretase complex. Neuron,2003,38:9~12
    6Simons K, Ikonen E. Functional rafts in cell membranes. Nature,1997,387:569~572
    7Brown DA, London E. Functions of lipid rafts in biological membranes.Annu Rev Cell Dev Biol,1998,14:111~136
    8Korade Z, Kenworthy AK. Lipid rafts, cholesterol, and the brain.Neuropharmacology,2008,55:1265~1273
    9Reid PC, Urano Y, Kodama T, et al. Alzheimer's disease: cholesterol,membrane rafts, isoprenoids and statins. J Cell Mol Med,2007,11:383~392
    10Vetrivel KS, Thinakaran G. Membrane rafts in Alzheimer's diseasebeta~amyloid production. Biochim Biophys Acta,2010,1801:860~867
    11Guardia~Laguarta C, Coma M, Pera M, et al. Mild cholesterol depletionreduces amyloid~beta production by impairing APP trafficking to thecell surface. J Neurochem,2009,110:220~230
    12Hur JY, Welander H, Behbahani H, et al. Active gamma~secretase islocalized to detergent~resistant membranes in human brain. FEBS J,2008,275:1174~1187
    13Cordy JM, Hussain I, Dingwall C, et al. Exclusively targetingbeta~secretase to lipid rafts by GPI~anchor addition up~regulatesbeta~site processing of the amyloid precursor protein. Proc Natl AcadSci U S A,2003,100:11735~11740
    14Riddell DR, Christie G, Hussain I, et al. Compartmentalization ofbeta~secretase (Asp2) into low~buoyant density, noncaveolar lipid rafts.Curr Biol,2001,11:1288~1293
    15Ehehalt R, Keller P, Haass C, et al. Amyloidogenic processing of theAlzheimer beta~amyloid precursor protein depends on lipid rafts. J CellBiol,2003,160:113~123
    16Morell P, Jurevics H. Origin of cholesterol in myelin. Neurochem Res,1996,21:463~470
    17Poirier J, Baccichet A, Dea D, et al. Cholesterol synthesis andlipoprotein reuptake during synaptic remodelling in hippocampus inadult rats. Neuroscience,1993,55:81~90
    18Pfrieger FW. Outsourcing in the brain: do neurons depend oncholesterol delivery by astrocytes? Bioessays,2003,25:72~78
    19Mulder M. Sterols in the central nervous system. Curr Opin Clin NutrMetab Care,2009,12:152~158
    20Bjorkhem I, Meaney S. Brain cholesterol: long secret life behind abarrier. Arterioscler Thromb Vasc Biol,2004,24:806~815
    21Puglielli L, Konopka G, Pack~Chung E, et al. Acyl~coenzyme A:cholesterol acyltransferase modulates the generation of the amyloidbeta~peptide. Nat Cell Biol,2001,3:905~912
    22Hutter~Paier B, Huttunen HJ, Puglielli L, et al. The ACAT inhibitorCP~113,818markedly reduces amyloid pathology in a mouse model ofAlzheimer's disease. Neuron,2004,44:227~238
    23Simons M, Keller P, De Strooper B, et al. Cholesterol depletion inhibitsthe generation of beta~amyloid in hippocampal neurons. Proc Natl AcadSci U S A,1998,95:6460~6464
    24Fassbender K, Simons M, Bergmann C, et al. Simvastatin stronglyreduces levels of Alzheimer's disease beta~amyloid peptides Abeta42and Abeta40in vitro and in vivo. Proc Natl Acad Sci U S A,2001,98:5856~5861
    25Barrett PJ, Song Y, Van Horn WD, et al. The amyloid precursor proteinhas a flexible transmembrane domain and binds cholesterol. Science,2012,336:1168~1171
    26Nicholson AM, Ferreira A. Increased membrane cholesterol mightrender mature hippocampal neurons more susceptible tobeta~amyloid~induced calpain activation and tau toxicity. J Neurosci,2009,29:4640~4651
    27Usui K, Hulleman JD, Paulsson JF, et al. Site~specific modification ofAlzheimer's peptides by cholesterol oxidation products enhancesaggregation energetics and neurotoxicity. Proc Natl Acad Sci U S A,2009,106:18563~18568
    28Ferrera P, Mercado~Gomez O, Silva~Aguilar M, et al. Cholesterolpotentiates beta~amyloid~induced toxicity in human neuroblastomacells: involvement of oxidative stress. Neurochem Res,2008,33:1509~1517
    29Gamba P, Testa G, Sottero B, et al. The link between altered cholesterolmetabolism and Alzheimer's disease. Ann N Y Acad Sci,2012,1259:54~64
    30Bjorkhem I, Cedazo~Minguez A, Leoni V, et al. Oxysterols andneurodegenerative diseases. Mol Aspects Med,2009,30:171~179
    31Vaya J, Schipper HM. Oxysterols, cholesterol homeostasis, andAlzheimer disease. J Neurochem,2007,102:1727~1737
    32Buhaescu I, Izzedine H. Mevalonate pathway: a review of clinical andtherapeutical implications. Clin Biochem,2007,40:575~584
    33Takai Y, Sasaki T, Matozaki T. Small GTP~binding proteins. PhysiolRev,2001,81:153~208
    34Montaner S, Perona R, Saniger L, et al. Multiple signalling pathwayslead to the activation of the nuclear factor kappaB by the Rho family ofGTPases. J Biol Chem,1998,273:12779~12785
    35Perona R, Montaner S, Saniger L, et al. Activation of the nuclearfactor~kappaB by Rho, CDC42, and Rac~1proteins. Genes Dev,1997,11:463~475
    36Williams LM, Lali F, Willetts K, et al. Rac mediates TNF~inducedcytokine production via modulation of NF~kappaB. Mol Immunol,2008,45:2446~2454
    37Clarke RM, Lyons A, O'Connell F, et al. A pivotal role for interleukin~4in atorvastatin~associated neuroprotection in rat brain. J Biol Chem,2008,283:1808~1817
    38Lindberg C, Crisby M, Winblad B, et al. Effects of statins on microglia.J Neurosci Res,2005,82:10~19
    39Cordle A, Landreth G.3~Hydroxy~3~methylglutaryl~coenzyme Areductase inhibitors attenuate beta~amyloid~induced microglialinflammatory responses. J Neurosci,2005,25:299~307
    40Massaro M, Zampolli A, Scoditti E, et al. Statins inhibitcyclooxygenase~2and matrix metalloproteinase~9in humanendothelial cells: anti~angiogenic actions possibly contributing toplaque stability. Cardiovasc Res,2010,86:311~320
    41Brown MS, Goldstein JL. The SREBP pathway: regulation ofcholesterol metabolism by proteolysis of a membrane~boundtranscription factor. Cell,1997,89:331~340
    42Jurevics H, Morell P. Cholesterol for synthesis of myelin is made locally,not imported into brain. J Neurochem,1995,64:895~901
    43Koudinov AR, Koudinova NV. Essential role for cholesterol in synapticplasticity and neuronal degeneration. FASEB J,2001,15:1858~1860
    44Mauch DH, Nagler K, Schumacher S, et al. CNS synaptogenesispromoted by glia~derived cholesterol. Science,2001,294:1354~1357
    45Shie FS, Jin LW, Cook DG, et al. Diet~induced hypercholesterolemiaenhances brain A beta accumulation in transgenic mice. Neuroreport,2002,13:455~459
    46Bales KR, Verina T, Dodel RC, et al. Lack of apolipoprotein Edramatically reduces amyloid beta~peptide deposition. Nat Genet,1997,17:263~264
    47Strittmatter WJ, Weisgraber KH, Huang DY, et al. Binding of humanapolipoprotein E to synthetic amyloid beta peptide: isoform~specificeffects and implications for late~onset Alzheimer disease. Proc NatlAcad Sci U S A,1993,90:8098~8102
    48Strittmatter WJ, Saunders AM, Schmechel D, et al. Apolipoprotein E:high~avidity binding to beta~amyloid and increased frequency of type4allele in late~onset familial Alzheimer disease. Proc Natl Acad Sci U SA,1993,90:1977~1981
    49Holtzman DM, Bales KR, Wu S, et al. Expression of humanapolipoprotein E reduces amyloid~beta deposition in a mouse model ofAlzheimer's disease. J Clin Invest,1999,103: R15~R21
    50Deane R, Sagare A, Hamm K, et al. apoE isoform~specific disruption ofamyloid beta peptide clearance from mouse brain. J Clin Invest,2008,118:4002~4013
    51Poirier J. Apolipoprotein E and Alzheimer's disease. A role in amyloidcatabolism. Ann N Y Acad Sci,2000,924:81~90
    52Li L, Cao D, Garber DW, et al. Association of aortic atherosclerosiswith cerebral beta~amyloidosis and learning deficits in a mouse modelof Alzheimer's disease. Am J Pathol,2003,163:2155~2164
    53Greenwood CE, Winocur G. Learning and memory impairment in ratsfed a high saturated fat diet. Behav Neural Biol,1990,53:74~87
    54Winocur G, Greenwood CE. The effects of high fat diets andenvironmental influences on cognitive performance in rats. Behav BrainRes,1999,101:153~161
    55Winocur G, Greenwood CE. Studies of the effects of high fat diets oncognitive function in a rat model. Neurobiol Aging,2005,26Suppl1:46~49
    56Granholm AC, Bimonte~Nelson HA, Moore AB, et al. Effects of asaturated fat and high cholesterol diet on memory and hippocampalmorphology in the middle~aged rat. J Alzheimers Dis,2008,14:133~145
    57Ullrich C, Pirchl M, Humpel C. Hypercholesterolemia in rats impairsthe cholinergic system and leads to memory deficits. Mol Cell Neurosci,2010,45:408~417
    58Tong XK, Nicolakakis N, Fernandes P, et al. Simvastatin improvescerebrovascular function and counters soluble amyloid~beta,inflammation and oxidative stress in aged APP mice. Neurobiol Dis,2009,35:406~414
    59Kurata T, Miyazaki K, Kozuki M, et al. Atorvastatin and pitavastatinimprove cognitive function and reduce senile plaque andphosphorylated tau in aged APP mice. Brain Res,2011,1371:161~170
    60Tong XK, Lecrux C, Rosa~Neto P, et al. Age~dependent rescue bysimvastatin of Alzheimer's disease cerebrovascular and memory deficits.J Neurosci,2012,32:4705~4715
    61Cibickova L. Statins and their influence on brain cholesterol. J ClinLipidol,2011,5:373~379
    62Wollmer MA. Cholesterol~related genes in Alzheimer's disease.Biochim Biophys Acta,2010,1801:762~773
    63Moncaster JA, Pineda R, Moir RD, et al. Alzheimer's diseaseamyloid~beta links lens and brain pathology in Down syndrome. PLoSOne,2010,5: e10659
    64Kivipelto M, Helkala EL, Laakso MP, et al. Apolipoprotein E epsilon4allele, elevated midlife total cholesterol level, and high midlife systolicblood pressure are independent risk factors for late~life Alzheimerdisease. Ann Intern Med,2002,137:149~155
    65Whitmer RA, Sidney S, Selby J, et al. Midlife cardiovascular riskfactors and risk of dementia in late life. Neurology,2005,64:277~281
    66Solomon A, Kareholt I, Ngandu T, et al. Serum cholesterol changesafter midlife and late~life cognition: twenty~one~year follow~up study.Neurology,2007,68:751~756
    67Reitz C, Luchsinger J, Tang MX, et al. Impact of plasma lipids and timeon memory performance in healthy elderly without dementia.Neurology,2005,64:1378~1383
    68Reitz C, Tang MX, Luchsinger J, et al. Relation of plasma lipids toAlzheimer disease and vascular dementia. Arch Neurol,2004,61:705~714
    69Mielke MM, Zandi PP, Sjogren M, et al. High total cholesterol levels inlate life associated with a reduced risk of dementia. Neurology,2005,64:1689~1695
    70Glass CK, Saijo K, Winner B, et al. Mechanisms underlyinginflammation in neurodegeneration. Cell,2010,140:918~934
    71Heneka MT, O'Banion MK, Terwel D, et al. Neuroinflammatoryprocesses in Alzheimer's disease. J Neural Transm,2010,117:919~947
    72Barres BA. The mystery and magic of glia: a perspective on their rolesin health and disease. Neuron,2008,60:430~440
    73Infante~Duarte C, Waiczies S, Wuerfel J, et al. New developments inunderstanding and treating neuroinflammation. J Mol Med (Berl),2008,86:975~985
    74Pahan K, Sheikh FG, Namboodiri AM, et al. Lovastatin andphenylacetate inhibit the induction of nitric oxide synthase andcytokines in rat primary astrocytes, microglia, and macrophages. J ClinInvest,1997,100:2671~2679
    75Ostrowski SM, Wilkinson BL, Golde TE, et al. Statins reduceamyloid~beta production through inhibition of protein isoprenylation. JBiol Chem,2007,282:26832~26844
    76Jick H, Zornberg GL, Jick SS, et al. Statins and the risk of dementia.Lancet,2000,356:1627~1631
    77Wolozin B, Kellman W, Ruosseau P, et al. Decreased prevalence ofAlzheimer disease associated with3~hydroxy~3~methyglutarylcoenzyme A reductase inhibitors. Arch Neurol,2000,57:1439~1443
    78Haag MD, Hofman A, Koudstaal PJ, et al. Statins are associated with areduced risk of Alzheimer disease regardless of lipophilicity. TheRotterdam Study. J Neurol Neurosurg Psychiatry,2009,80:13~17
    79Yaffe K, Barrett~Connor E, Lin F, et al. Serum lipoprotein levels, statinuse, and cognitive function in older women. Arch Neurol,2002,59:378~384
    80Zandi PP, Sparks DL, Khachaturian AS, et al. Do statins reduce risk ofincident dementia and Alzheimer disease? The Cache County Study.Arch Gen Psychiatry,2005,62:217~224
    81Rea TD, Breitner JC, Psaty BM, et al. Statin use and the risk of incidentdementia: the Cardiovascular Health Study. Arch Neurol,2005,62:1047~1051
    82Arvanitakis Z, Schneider JA, Wilson RS, et al. Statins, incidentAlzheimer disease, change in cognitive function, and neuropathology.Neurology,2008,70:1795~1802
    83Shepherd J, Blauw GJ, Murphy MB, et al. Pravastatin in elderlyindividuals at risk of vascular disease (PROSPER): a randomisedcontrolled trial. Lancet,2002,360:1623~1630
    84MRC/BHF Heart Protection Study of cholesterol lowering withsimvastatin in20,536high~risk individuals: a randomisedplacebo~controlled trial. Lancet,2002,360:7~22
    85Sparks DL, Sabbagh MN, Connor DJ, et al. Atorvastatin for thetreatment of mild to moderate Alzheimer disease: preliminary results.Arch Neurol,2005,62:753~757
    86Feldman HH, Doody RS, Kivipelto M, et al. Randomized controlledtrial of atorvastatin in mild to moderate Alzheimer disease: LEADe.Neurology,2010,74:956~964
    87Wolozin B. Statins and therapy of Alzheimer's disease: questions ofefficacy versus trial design. Alzheimers Res Ther,2012,4:3

© 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