用户名: 密码: 验证码:
性腺切除对小鼠脑衰老和脑热休克蛋白表达的作用,及FLZ抗Aβ生成的作用
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
随着世界人口平均寿命的增加,老年人口逐年递增,伴随衰老而来的神经退行性疾病如阿尔茨海默病(Alzheimer's disease,AD)和帕金森氏病(Parkinson's disease,PD)的发病率亦相应增加。因而全世界开始面临老龄化社会带来的诸多问题。为了延长人类的预期寿命,提高老年人的生命质量,抗衰老和防治衰老相关疾病受到了越来越多的重视。其中,脑衰老和衰老相关的神经退行性疾病是老年医学中最急需解决的问题之一。
     关于衰老机制的假说甚多,其中包括生殖系统衰老导致的性激素缺乏对脑衰老性病理改变的发展具有促进作用。雌激素是一种具有神经调节和神经保护作用的激素。雌激素通过激活其受体调控多种基因的表达而发挥神经保护作用,另外,它还具有抗氧化作用。绝经后雌激素水平的迅速下降可以导致认知和记忆能力的减退,增加神经退行性疾病老年痴呆和帕金森病的发病率。雄激素同样具有神经调节和神经保护作用。男性的睾酮水平在中年以后开始减退,老年男性的睾酮低水平导致认知记忆功能的减退,睾酮缺乏是AD的重要危险因素。
     在衰老过程中,脑组织暴露于多种应激刺激中,损伤的蛋白质逐渐积累。错误折叠蛋白质的聚集,对细胞的存活造成严重的威胁,促进神经退行性病变的发生。热休克蛋白(heat shock protein,HSP)作为分子伴侣,与错误折叠的蛋白质结合,促进其正确折叠或清除。热休克蛋白可以抵抗多种应激刺激导致的组织损伤,因此被视为内源性细胞保护因子。在多种应激刺激作用下,两种重要的热休克蛋白HSP70和HSP27,在大脑的许多区域存在明显的诱导表达。在衰老脑组织中,应激刺激诱导的热休克蛋白表达减少,导致内源性防御机制减弱。
     阿尔茨海默病是老年人常见的,以进行性的记忆和认知功能损伤为特征的神经退行性病变。老年斑的形成是AD最重要的特征性病理学改变。老年斑的主要成分淀粉样蛋白(Amyloidβ,Aβ)Aβ_(1-40)和Aβ_(1-42),在脑组织中的进行性沉积可以引发一系列神经损害性反应,促进神经退行性病变的发展。Aβ是由淀粉样前体蛋白(Amyloid precursor protein,APP)经α-,β-和γ-内分泌酶酶切加工而生成的。APP的酶切加工存在两种相互排斥的不同途径:淀粉样蛋白生成途径系APP被β-内分泌酶和γ-内分泌酶切割生成Aβ;非淀粉样蛋白生成途径系APP被α-内分泌酶切割生成sAPPα。抑制β-内分泌酶以减少Aβ沉积是抗AD药物研究的靶点。研究表明,增加α-内分泌酶的表达可以减少AD转基因小鼠海马的Aβ沉积,改善学习记忆功能。因此,促进α-内分泌酶对APP的酶切加工成为治疗AD的新手段。
     FLZ是人工全合成的番荔枝酰胺新衍化物。既往的研究证实FLZ具有明显的神经保护作用:保护Aβ导致的SH-SY5Y细胞的损伤;改善脑室注射Aβ导致的小鼠学习记忆能力的损害并且减轻海马病理学损伤。因此,FLZ具有研发为抗AD新药的前景,已申请专利保护。
     为了解性激素与衰老和神经退行性疾病发病的关系以及寻找抗AD新药,本论文主要从以下三个方面进行研究:
     第一部分:性腺切除加速小鼠脑功能衰老模型的建立
     为建立性腺切除促进脑功能衰老的模型,在本研究中,将2月龄年轻雄性和雌性ICR小鼠的性腺切除,继续饲养10个月至12月龄。实验用小鼠分为4组:雌性假手术组(female-sham,12月龄)、雌性切除卵巢组(female-OVX,12月龄)、雄性假手术组(male-sham,12月龄)、雄性切除睾丸组(male-ORX,12月龄)。评价各项指标时,临时领取雌性年轻小鼠(female-young,2月龄)和雄性年轻小鼠(male-young,2月龄),与上述各组进行对比分析。小鼠脑衰老相关指标的检测包括学习记忆功能及脑组织相关部位的神经病理学指标的变化。在水迷津和Morris水迷宫实验中,结果显示,与2月龄年轻小鼠比较,性腺切除小鼠空间学习记忆能力下降;免疫组织化学染色和Western blot结果显示,性腺切除小鼠脑组织Aβ、BACE1和phospho-Tau表达增加;而NGF、BDNF、NT3和TrkA表达水平减低,p75NTR表达水平增高。尼氏染色结果显示,性腺切除小鼠海马和皮层神经元尼氏体数量减少。与同性别年轻小鼠相比较,卵巢切除小鼠脑组织的上述衰老相关指标的改变比睾丸切除小鼠更加明显。假手术组小鼠的上述指标则与同性别年轻小鼠相比没有明显差异。由此表明,性腺切除导致的性激素长期缺乏可以加速小鼠脑衰老。表现为学习记忆功能的减退,脑组织Aβ沉积增多,而神经营养因子减少。
     第二部分:雌激素对卵巢切除小鼠脑组织热休克蛋白表达的调节
     热休克蛋白是一种内源性细胞保护因子,老年人耐受酷暑天气能力差,可能与热休克蛋白表达调控的失调有关。而热休克蛋白与AD和PD的发展亦有关系,为了解卵巢切除对小鼠脑组织热休克蛋白表达的影响,以及雌激素对热休克蛋白表达的调控作用,研究了小鼠切除卵巢后雌激素对高热刺激诱导的脑组织热休克蛋白表达的调节作用和相关机制。2月龄雌性小鼠经双侧卵巢切除或假手术后,继续饲养10个月。小鼠分成4组:年轻组(young,2月龄)、切除卵巢组(old-OVX,12月龄)、假手术组(old-sham,12月龄)、雌激素补充组(OVX+E2,12月龄)。雌激素补充组小鼠隔日皮下注射雌二醇0.5mg/kg,持续两周,以恢复其血清雌激素生理水平。将小鼠置于44℃烤箱中接受1h高热刺激后,将小鼠取出,置于室温环境中,3h后将小鼠处死。Western blot和RT-PCR结果显示,切除卵巢组小鼠脑组织高热刺激后HSP70蛋白的表达和mRNA的转录均明显低于年轻组小鼠。凝胶迁移阻滞实验检测调控热休克蛋白表达的关键因子,结果显示,切除卵巢组小鼠脑组织热休克因子和热休克元件的结合能力明显减弱。并且发现切除卵巢组小鼠在高热刺激后脑组织出现明显的DNA片断化损伤和caspase-3活性增高,线粒体细胞色素C和凋亡诱导因子的释放亦均增加。与切除卵巢组小鼠相比,雌激素补充组小鼠脑组织中,高热刺激后HSP70蛋白的表达和mRNA的转录明显增高,热休克因子和热休克元件的结合能力明显恢复;高热刺激导致的DNA片断化损伤、caspase-3活性的增高和线粒体细胞色素C和凋亡诱导因子的释放均明显减轻。由此可见,切除卵巢小鼠脑组织在高热刺激后HSP70蛋白的诱导表达能力减低,其机制与热休克因子和热休克元件的结合能力减弱有关。而补充雌激素可明显恢复高热刺激诱导的切除卵巢小鼠脑组织HSP70蛋白表达,并且保护高热刺激导致的脑组织损伤。这些结果均属新发现。
     第三部分:番荔枝酰胺衍生物FLZ通过促进α-内分泌酶介导的APP酶切加工而减少Aβ生成
     如前所述,增加α-内分泌酶的活性可以减少APP切割为Aβ,从而可能是治疗AD的新策略。FLZ在多种模型中具有神经保护作用。本文从α-内分泌酶介导的APP酶切加工角度对FLZ的作用进行研究。在本研究中,我们发现FLZ可以减少N2a(APPswe/PS1Δ9)细胞的Aβ生成,同时增加培养基中sAPPα的分泌。Westernblot结果显示,FLZ可以增加细胞膜表面ADAM10和APP的表达。免疫细胞化学染色显示FLZ可以增加ADAM10和APP在细胞膜表面的共定位。预先给与细胞蛋白运输抑制剂brefeldin能阻断FLZ介导的ADAM10和APP向细胞膜的运输,逆转FLZ抑制Aβ生成和增加sAPPα分泌的作用。在TgAPP/PS1转基因小鼠中,FLZ能减少海马Aβ的生成,增加ADAM10的表达。上述结果表明,FLZ通过促进α-内分泌酶介导的APP酶切加工而减少Aβ的生成。这为FLZ用于治疗AD提供了新的理论依据。
     综上所述,建立衰老动物模型对研究衰老机理、研发延缓衰老和防治衰老相关神经退行性疾病的药物具有重要意义。衰老研究的最终目的之一是为防治神经退行性疾病提供新策略。本文发现:(1)性腺切除小鼠与同性别年轻小鼠相比,空间学习记忆能力减退;脑组织Aβ、BACE1和phospho-Tau表达增加;海马和皮层神经元尼氏体数量减少;脑组织NGF、BDNF、NT3和TrkA表达减低,p75NTR表达水平增高。这些新发现提示:性腺切除导致的长期性激素缺乏可以加速脑衰老。(2)切除卵巢小鼠脑组织在高热刺激后HSP70蛋白的诱导表达减少,其机制与热休克因子和热休克元件的结合能力减弱有关。由此可见,雌激素的长期缺乏可加重小鼠脑组织内源性防御功能的减退。补充雌激素可使切除卵巢小鼠脑组织在高热刺激后HSP70蛋白的诱导表达功能恢复,并且保护高热刺激所致的脑组织损伤。因此,雌激素对热休克蛋白诱导表达的调节作用为其神经保护作用提供了新的依据。(3) FLZ可以通过促进α-内分泌酶介导的APP酶切加工而减少Aβ的生成,这为FLZ的神经保护作用提供了新的依据。
     此外,为寻找治疗肝功能衰竭新药,对抗肝炎新药双环醇的衍化物WLP-S-10对扑热息痛所致小鼠急性肝衰竭的保护作用及机制进行了研究,结果如下:
     急性肝衰竭是一种以肝功能迅速恶化为特征的严重急症。扑热息痛过量是药物中毒性急性肝衰竭的最常见原因。双环醇是我国自主研发的新型药物,在临床上广泛用于治疗病毒性肝炎,药物和化学毒物导致的肝损伤。药理学研究证明,双环醇对多种动物肝损伤模型导致的肝损伤具有保护作用。但是,双环醇属脂溶性化合物,不溶于水。目前临床只能口服,无法注射用药。在本研究中,我们合成了一系列水溶性双环醇衍化物。使用CCl_4导致的小鼠肝功能损伤模型,扑热息痛和D-氨基半乳糖加脂多糖所致的急性肝衰竭模型,对其中10种化合物进行筛选,发现其中的一种化合物(WLP-S-10)作用最明显。我们进而研究了该化合物对扑热息痛所致急性肝衰竭的保护作用和机制。提前1小时给小鼠腹腔注射WLP-S-10 200mg/kg,然后腹腔注射扑热息痛450mg/kg。结果显示,WLP-S-10可以降低小鼠死亡率,降低血清ALT和AST;防止线粒体GSH及ATP的耗竭;减轻线粒体肿胀和膜电位崩溃;减少线粒体细胞色素C和AIF的释放。HPLC结果显示,WLP-S-10可以抑制肝微粒体对扑热息痛的代谢。WLP-S-10在体内代谢转化为双环醇和蛋氨酸。结果表明,双环醇衍化物WLP-S-10对扑热息痛导致的小鼠急性肝衰竭有明显的保护作用。其作用的主要机制是抑制扑热息痛在肝脏的毒性代谢和增加肝脏GSH水平,从而保护线粒体功能,减轻扑热息痛导致的肝细胞死亡。本文已发表于Liver International,2008 Nov;28(9):1226-35。
Over several decades,with the rise of average life expectancy,aging population gradually increased,which results in increased incidence of neurodegenerative disease, such as Alzheimer's disease(AD) and Parkinson's disease(PD).The public has to face the problems which aging society brings to the whole world.In order to extend the lifespan and improve the living quality of aging people,researches on aging and age related disease have raised more concerns in recent years.Brain aging and age related neurodegenerative disease is the focus of neuroscience gerontology.
     There are several hypotheses about mechanism of aging.Aging of reproductive system is considered to accelerate brain aging and increase the incidence of neurodegenerative diseases.Substantial evidence suggests that estrogen is a neuroprotective and neuromodulatory hormone.Besides its anti-oxidative effect, estrogen can regulate expression of a variety of genes by activation of estrogen receptors. The sharp decline of estrogen level after menopause may lead to impaired memory and cognition,and increases the incidence of AD and PD.Testosterone also has neuroprotective and neuromodulatory effect.The decline of testosterone level after middle age could lead to impaired memory and cognition and increased incidence of neurodegenerative diseases in men.
     During aging,brain is exposed to various stressors,which leads to accumulation of damaged proteins.The aggregation of misfolded proteins becomes a threat to cell survival,and promotes pathogenesis of neurodegenerative diseases.Heat shock proteins (HSP) serve as molecular chaperons,binding to misfolded proteins,assisting to refold misfolded proteins and eliminate irreversibly damaged proteins.Therefore,heat shock proteins are considered as endogenous cytoprotective factors,with function to protect cells against a multitude of stresses.Two of the most important heat shock proteins Hsp70 and Hsp27,can be significantly induced in many areas of brain after stresses. During aging,stress-induced expression of heat shock proteins declines in the brain, which leads to impairment of endogenous defense system.
     AD is a neurodegenerative disease characterized clinically by progressive impairment of memory and cognition.One of the hallmarks of AD is the presence of senile plaques in the hippocampus,which are composed primarily of aggregated extracellular deposition of Aβ_(1-40) and Aβ_(1-42).The progressive accumulation of Aβaggregates is widely believed to be fundamental to initiate neurodegenerative pathogenesis.Aβis generated by proteolysis of amyloid precursor protein(APP) by three specific enzymes,α-,β-,andγ-secretases.APP is enzymatically processed by two distinct and mutually exclusive pathways:in amyloidogenic pathway,cleavage of APP byβ-secretase produces Aβ.Alternatively,in nonamyloidogenic pathway,cleavage of APP byα-secretase generates sAPPαInhibition ofβ-secretase to decrease Aβproduction has been taken as target for AD drug development.In addition,substantial evidence indicates that AD transgenic mice with over-expression ofα-secretase display improved learning and memory,and less Aβdeposition in the hippocampus.Therefore,upregulation ofα-secretase mediated APP processing may be novel therapeutic applications in AD.
     FLZ is a synthetic cyclic analogue of natural squamosamide.Previous studies demonstrated that FLZ had significant neuroprotective effect.FLZ protected against Aβ-induced toxicity in SH-SY5Y cells.It also ameliorated the impairment of learning and memory and pathological damage to the hippocampus induced by ventricular injection of Aβin mice.Therefore,FLZ could be a promising candidate for AD treatment,and it has applied for patent.
     In order to elucidate the relationship between sex hormones and aging related neurodegenerative diseases,and develop novel drugs for AD treatment,three parts of study were performed and are reported in this dissertation as following:
     PartⅠ:Establishment of gonadectomy-aecelerated brain aging model in mice
     In this study,we aim to develop a gonadectomy-accelerated brain aging model. Male or female mice of 2-month old were orchiectomized(ORX) or ovariectomized (OVX) bilaterally,and were kept for 10 months.All the mice were divided into 4 groups: female-sham(12-month old),female-OVX(12-month old),male-sham(12-month old) and male-ORX(12-month old).2-month old female(female-young) and male(male-young) mice were obtained before determination of brain aging-related parameters.The results of water maze show that spatial learning and memory was impaired in gonadectomized mice as compared with young mice.The results of western blotting and immunostanning show increased expression of Aβ,BACE1 and phospho-Tau,decreased expression of NGF,BDNF,NT3 and TrkA as well as increased expression of p75NTR in gonadectomized mice,as compared with young mice.The result of Nissl staining indicates that the amount of Nissl bodies decreased in the brain of gonadectomized mice. In addition,as compared with young mice of the same gender,the changes of brain aging related parameters were more obvious in ovariectomized mice than that in orchiectomized ones.There are no significant changes of these parameters in the brain of sham-operated mice as compared to young mice.As a result,gonadectomy can accelerate brain aging which is displayed as impaired spatial learning and memory,increased Aβexpression as well as decreased neurotrophins level in the brain.
     PartⅡ:The role of estrogen in modulating expression of brain heat shock proteins in ovariectomized mice
     Heat shock proteins serve as endogenous cytoprotective factors.The impaired thermotolerance in old people is probably due to diminished expression of heat shock proteins,and heat shock proteins also play a role in the pathogenesis of AD and PD.In order to elucidate the effect of ovariectomy and the modulatory effect of estrogen on expression of brain heat shock proteins,in this study,we investigated the regulatory effect of estrogen on heat stress-induced brain heat shock protein expression in ovariectomized mice and its underlying mechanism.Female mice of 2 months old were ovariectomized bilaterally or sham operated,and were kept for 10 months.Some ovariectomized mice were supplemented with 17β-estradiol(0.5 mg/kg/day every other day for 14 days,S.C.).Mice were divided into four groups:young(2-month old),old-OVX (12-month old),old-sham(12-month old) and OVX+17β-estradiol(E2)(12-month old),and were subjected to heat stress at 44℃in an oven for 1 hour followed by 3 hours at room temperature.The results of western blotting and RT-PCR show that in ovariectomized mice,heat stress-induced brain HSP70 expression and mRNA transcription are significantly lower than that of young mice.And the result of gel mobility shift assay shows that the binding activity of heat shock factor with heat shock element was also significantly impaired in the brain of ovariectomized mice.In the brain of mice from OVX+E2 group,supplement of estrogen restored heat stress-induced brain HSP70 expression,and attenuated heat stress-induced brain DNA fragmentation, caspase-3 activation and mitochondrial cytochrorne C and AIF leakage.The results suggest that in ovariectomized mice,heat stress-induced brain Hsp70 expression decreased and the mechanism was related to impaired binding of heat shock factor with heat shock element.Estrogen supplement restored heat stress-induced heat shock protein expression and protected against heat stress-induced brain injury in ovariectomized mice.
     PartⅢ:A squamosamide derivative FLZ reduces amyloidβproduction by increasingα-secretase mediated non-amyloidogenic APP processing
     As mentioned above,upregulation ofα-secretase mediated APP processing leads to decreased Aβproduction,which may be novel therapeutic applications in AD.The neuroprotective effect of FLZ has been proved in several models.In this study,we investigate the effect of FLZ onα-secretase mediated APP processing.We found that Aβproduction was reduced by FLZ in Aβ-expressing N2a(APPswe/PS1Δ9) neuroblastoma cells,which was paralleled with an increase in sAPPαin the medium.Moreover,the result of western blotting show that the active form of ADAM10 and APP expression were elevated at the cell surface of FLZ-treated cells,consistent with immunostaining result showing an enhanced co-localization of ADAM10 and APP at the membrane of FLZ treated cells.Pretreatment with protein trafficking inhibitor brefeldin blocked FLZ-induced translocation of ADAM10 and APP to the cell surface;it also reversed FLZ-induced increase in.sAPPαrelease and decrease in Aβproduction.Furthermore,oral administration of FLZ to APPswe/PS1 transgenic mice significantly reduced Aβlevel and increased ADAM10 level in hippocampus.These findings suggest that decreasing effects of FLZ on Aβproduction may be mediated by its promotion ofα-secretase mediated APP non-amyloidogenic processing,which provides evidence for therapeutic applications of FLZ in AD.
     Establishing good animal models of aging is very important for the research on aging and neurodegenerative disease.The ultimate goal of research on aging and neurodegenerative disease is to find novel therapeutic applications.To summarize the study:(1) As compared with young mice,gonadectomized mice had impaired spatial learning and memory,increased expression of Aβ,BACE1 and phospho-Tau,decreased Nissl bodies and decreased expression of NGF,BDNF,NT3 and TrkA as well as increased expression of p75NTR.All of these results indicate that long-term sex hormone deficiency by gonadectomy can accelerate brain aging.(2) Heat stress-induced brain Hsp70 expression decreased in ovariectomized mice and the mechanism is related to impaired binding of heat shock factor with heat shock element.It suggests that deficiency of estrogen during aging can exacerbate impairment of endogenous defense system in brain.Estrogen supplement restored heat stress-induced heat shock protein expression and protected against heat stress-induced brain injury in ovariectomized mice.Therefore, the modulatory effect of estrogen on heat shock protein expression provides a new evidence for its neuroprotective effect.(3) FLZ decreased Aβproduction by promotingα- secretase mediated APP non-amyloidogenic processing.Our study provides new evidence for therapeutic potential of FLZ for AD.
     In addition,in order to develop novel drugs for the treatment of acute liver failure,we investigated the protective effect and mechanism of bicyclol derivative WLP-S-10 against acetaminophen-induced acute liver failure in mice.The results are reported as follow:
     Acute liver failure is a severe emergency with rapid deterioration of liver function. Overdose of acetaminophen is the most common cause of drug-induced liver failure. Bicyclol is a novel anti-hepatitis drug and has been widely used to treat chronic viral hepatitis and drug-induced liver injury in China.Pharmacological investigation demonstrated that bicyclol has protective action against liver injury in several models, however,bicyclol is not water soluble;it is difficult to make injectable preparation for treatment of acute liver failure patients at present.In this study,a number of water soluble bicyclol derivatives were synthesized and screened in mice using CCl_4, acetaminophen or D-galactosamine plus LPS induced liver injury models.Among ten compounds,a methionine derivative of bicyclol(WLP-S-10) was shown to be the most effective one in lowering serum ALT level and reducing the mortality of mice.WLP-S-10 was,therefore,further studied in acetaminophen induced acute liver failure in mice.A single dose of WLP-S-10 200mg/kg was intraperitoneally injected 1 hour before administration of acetaminophen 450mg/kg.Pretreatment of mice with WLP-S-10 200mg/kg significantly reduced mortality of mice due to acute liver failure caused by lethal dosage of acetaminophen.The liver necrosis,serum ALT and AST elevation,and GSH depletion induced by injection of acetaminophen were all reduced.WLP-S-10 200mg/kg inhibited mitochondrial swelling,breakdown of membrane potential,depletion of mitochondrial ATP,and release of cytochrome C and AIF from mitochondria.In addition,the result of HPLC shows that WLP-S-10 200mg/kg inhibited metabolism of acetaminophen in liver microsomes.WLP-S-10 is converted into bicyclol and methionine. All the results indicate that WLP-S-10 is a novel potential compound against acetaminophen induced acute liver failure in mice,and its active mechanism is related to a summation of inhibiting bioactivation of acetaminophen and increasing liver GSH level, and subsequently attenuating mitochondrial dysfunction and hepatocyte death.The paper has been published in Liver International,2008 Nov;28(9):1226-35.
引文
[1]Hof PR,Morrison JH.The aging brain:morphomolecular senescence of cortical circuits[J].Trends Neurosci,2004,27(10):607-13.
    [2]Salat DH,Tuch DS,Hevelone ND,et al.Age-related changes in prefrontal white matter measured by diffusion tensor imaging[J].Ann N Y Acad Sci,2005,1064:37-49.
    [3]Masoro EJ.Use of rodents as models for the study of“normal aging”:conceptual and practical issues[J].Neurobiol Aging,1991,12(6):639-43.
    [4]Pallas M,Camins A,Smith MA,et al.From aging to Alzheimer's disease:unveiling“the switch”with the senescence-accelerated mouse model (SAMP8)[J].J Alzheimers Dis,2008,15(4):615-24.
    [5]Zhang D,Liu G,Shi J,et al.Coeloglossum viride var.bracteatum extract attenuates D-galactose and NaN02 induced memory impairment in mice[J].J Ethnopharmacol,2006,104(l-2):250-6.
    [6]Spencer JL,Waters EM,Romeo RD,et al.Uncovering the mechanisms of estrogen effects on hippocampal function[J].Front Neuroendocrinol,2008,29(2):219-37.
    [7]Craig MC,Murphy DG.Estrogen:effects on normal brain function and neuropsychiatric disorders[J].Climacteric,2007,10 Suppl 2:97-104.
    [8]Thakur MK,Sharma PK.Binding of estrogen receptor alpha promoter to nuclear proteins of mouse cerebral cortex:effect of age,sex,and gonadal steroids[J].Biogerontology,2008,9(6):467-78.
    [9]Manthey D,Behl C.From structural biochemistry to expression profiling:neuroprotective activities of estrogen[J].Neuroscience,2006,138(3):845-50.
    [10]Rocca WA,Bower JH,Maraganore DM,et al.Increased risk of cognitive impairment or dementia in women who underwent oophorectomy before menopause[J].Neurology,2007,69(11):1074-83.
    [11]Shuster LT,Gostout BS,Grossardt BR,et al.Prophylactic oophorectomy in premenopausal women and long-term health[J].Menopause Int,2008,14(3):111-6.
    [12]Sherwin BB,Henry JF.Brain aging modulates the neuroprotective effects of estrogen on selective aspects of cognition in women:a critical review[J].Front Neuroendocrinol,2008,29(1):88-113.
    [13]Beyenburg S,Watzka M,Clusmann H,et al.Androgen receptor mRNA expression in the human hippocampus[J].Neurosci Lett,2000,294(l):25-8.
    [14]Roselli CE,Klosterman S,Resko JA.Anatomic relationships between aromatase and androgen receptor mRNA expression in the hypothalamus and amygdala of adult male cynomolgus monkeys[J].J Comp Neurol,2001,439(2):208-23.
    [15]Leranth C,Petnehazy O,MacLusky NJ.Gonadal hormones affect spine synaptic density in the CA1 hippocampal sub field of male rats[J].J Neurosci,2003,23(5):1588-92.
    [16]Barrett-Connor E,Goodman-Gruen D,Patay B.Endogenous sex hormones and cognitive function in older men[J].J Clin Endocrinol Metab,1999,84(10):3681-5.
    [17]Moffat SD,Zonderman AB,Metter EJ,et al.Free testosterone and risk for Alzheimer disease in older men[J].Neurology,2004,62(2):188-93.
    [18]Almeida OP,Flicker L.Testosterone and dementia:too much ado about too little data[J].J Br Menopause Soc,2003,9(3):107-10.
    [19]Janowsky JS.The role of androgens in cognition and brain aging in men[J].Neuroscience,2006,138(3):1015-20.
    [20]Howieson DB,Holm LA,Kaye JA,et al.Neurologic function in the optimally healthy oldest old.Neuropsychological evaluation[J].Neurology,1993,43(10):1882-6.
    [21]Frye CA,Park D,Tanaka M,et al.The testosterone metabolite and neurosteroid 3alpha-androstanediol may mediate the effects of testosterone on conditioned place preference[J].Psychoneuroendocrinology,2001,26(7):731-50.
    [22]Kritzer MF,McLaughlin PJ,Smirlis T,et al.Gonadectomy impairs T-maze acquisition in adult male rats[J].Horm Behav,2001,39(2):167-74.
    [23]Frye CA,Waif AA.Progesterone to ovariectomized mice enhances cognitive performance in the spontaneous alternation,object recognition,but not placement, water maze,and contextual and cued conditioned fear tasks[J].Neurobiol Learn Mem,2008,90(l):171-7.
    [24]Frick KM,Fernandez SM,Bulinski SC.Estrogen replacement improves spatial reference memory and increases hippocampal synaptophysin in aged female mice[J].Neuroscience,2002,115(2):547-58.
    [25]Markham JA,Pych JC,Juraska JM.Ovarian hormone replacement to aged ovariectomized female rats benefits acquisition of the morris water maze[J].Horm Behav,2002,42(3):284-93.
    [26]Aenlle KK,Kumar A,Cui L,et al.Estrogen effects on cognition and hippocampal transcription in middle-aged mice[J].Neurobiol Aging,2007,
    [27]Ramsden M,Nyborg AC,Murphy MP,et al.Androgens modulate beta-amyloid levels in male rat brain[J].J Neurochem,2003,87(4):1052-5.
    [28]Petanceska SS,Nagy V,Frail D,et al.Ovariectomy and 17beta-estradiol modulate the levels of Alzheimer's amyloid beta peptides in brain[J].Exp Gerontol,2000,35(9-10):1317-25.
    [29]Carroll JC,Rosario ER,Chang L,et al.Progesterone and estrogen regulate Alzheimer-like neuropathology in female 3xTg-AD mice[J].J Neurosci,2007,27(48):13357-65.
    [30]Levin-Allerhand JA,Lominska CE,Wang J,et al.17Alpha-estradiol and 17beta-estradiol treatments are effective in lowering cerebral amyloid-beta levels in AbetaPPSWE transgenic mice[J].J Alzheimers Dis,2002,4(6):449-57.
    [31]Sambamurti K,Kinsey R,Maloney B,et al.Gene structure and organization of the human beta-secretase (BACE) promoter[J].Faseb J,2004,18(9):1034-6.
    [32]Zhang QG,Wang R,Khan M,et al.Role of Dickkopf-1,an antagonist of the Wnt/beta-catenin signaling pathway,in estrogen-induced neuroprotection and attenuation of tau phosphorylation[J].J Neurosci,2008,28(34):8430-41.
    [33]Liu XA,Zhu LQ,Zhang Q,et al.Estradiol attenuates tau hyperphosphorylation induced by upregulation of protein kinase-A[J].Neurochem Res,2008,33(9):1811-20.
    [34]Shi HR,Zhu LQ,Wang SH,et al.17beta-estradiol attenuates glycogen synthase kinase-3beta activation and tau hyperphosphorylation in Akt-independent manner[J].J Neural Transm,2008,115:879-88.
    [35]Vanoye-Carlo A,Morales T,Ramos E,et al.Neuroprotective effects of lactation against kainic acid treatment in the dorsal hippocampus of the rat[J].Horm Behav,2008,53(1):112-23.
    [36]Takuma K,Hoshina Y,Arai S,et al.Ginkgo biloba extract EGb 761 attenuates hippocampal neuronal loss and cognitive dysfunction resulting from chronic restraint stress in ovariectomized rats[J].Neuroscience,2007,149(2):256-62.
    [37]Itahana K,Gampisi J,Dimri GR Methods to detect biomarkers of cellular senescence:the senescence-associated beta-galactosidase assay[J].Methods Mol Biol,2007,371:21-31.
    [38]Yegorov YE,Akimov SS,Hass R,et al.Endogenous beta-galactosidase activity in continuously nonproliferating cells[J].Exp Cell Res,1998,243(1):207-11.
    [39]Severino J,Allen RG,Balin S,et al.Is beta-galactosidase staining a marker of senescence in vitro and in vivo?[J].Exp Cell Res,2000,257(1):162-71.
    [40]Smith MA.Hippocampal vulnerability to stress and aging:possible role of neurotrophic factors[J].Behav Brain Res,1996,78(l):25-36.
    [41]Lindsay RM,Wiegand SJ,Altar CA,et al.Neurotrophic factors:from molecule to man[J].Trends Neurosci,1994,17(5):182-90.
    [42]Nawa H,Bessho Y,Carnahan J,et al.Regulation of neuropeptide expression in cultured cerebral cortical neurons by brain-derived neurotrophic factor[J].J Neurochem,1993,60(2):772-5.
    [43]Skaper SD.The biology of neurotrophins,signalling pathways,and functional peptide mimetics of neurotrophins and their receptors[J].CNS Neurol Disord Drug Targets,2008,7(l):46-62.
    [44]Huang EJ,Reichardt LF.Trk receptors:roles in neuronal signal transduction[J].Annu Rev Biochem,2003,72:609-42.
    [45]Sofroniew MV,Howe CL,Mobley WC.Nerve growth factor signaling,neuroprotection,and neural repair[J].Annu Rev Neurosci,2001,24:1217-81.
    [46]Cuello AC,Bruno MA,Bell KR NGF-cholinergic dependency in brain aging, MCI and Alzheimer's disease[J].Curr Alzheimer Res,2007,4(4):351-8.
    [47]Wu ZL,Ciallella JR,Flood DG,et al.Comparative analysis of cortical gene expression in mouse models of Alzheimer's disease[J].Neurobiol Aging,2006,27(3):377-86.
    [48]Durany N,Michel T,Kurt J,et al.Brain-derived neurotrophic factor and neurotrophin-3 levels in Alzheimer's disease brains[J].Int J Dev Neurosci,2000,18(8):807-13.
    [49]Singh M,Meyer EM,Simpkins JW.The effect of ovariectomy and estradiol replacement on brain-derived neurotrophic factor messenger ribonucleic acid expression in cortical and hippocampal brain regions of female Sprague-Dawley rats[J].Endocrinology,1995,136(5):2320-4.
    [50]Costantini C,Scrable H,Puglielli L.An aging pathway controls the TrkA to p75NTR receptor switch and amyloid beta-peptide generation[J].Embo J,2006,25(9):1997-2006.
    [1]Beere HM.Stressed to death:regulation of apoptotic signaling pathways by the heat shock proteins[J].Sci STKE,2001,2001:RE1.
    [2]Garrido C,Gurbuxani S,Ravagnan L,et al.Heat shock proteins:endogenous modulators of apoptotic cell death[J].Biochem Biophys Res Commun,2001, 286(3):433-42.
    [3]Kampinga HH.Chaperones in preventing protein denaturation in living cells and protecting against cellular stress[J].Handb Exp Pharmacol,2006,172:1-42.
    [4]Didelot C,Schmitt E,Brunet M,et al.Heat shock proteins:endogenous modulators of apoptotic cell death[J].Handb Exp Pharmacol,2006,172:171-98.
    [5]Franklin TB,Krueger-Naug AM,Clarke DB,et al.The role of heat shock proteins Hsp70 and Hsp27 in cellular protection of the central nervous system[J].Int J Hyperthermia,2005,21 (5):379-92.
    [6]Latchman DS.HSP27 and cell survival in neurones[J].Int J Hyperthermia,2005,21(5):393-402.
    [7]Ostberg JR,Kaplan KC,Repasky EA.Induction of stress proteins in a panel of mouse tissues by fever-range whole body hyperthermia[J].Int J Hyperthermia,2002,18(6):552-62.
    [8]Dillmann WH.Heat shock proteins and protection against ischemic injury[J].Infect Dis Obstet Gynecol,1999,7(l-2):55-7.
    [9]Currie RW,Ellison JA,White RF,et al.Benign focal ischemic preconditioning induces neuronal Hsp70 and prolonged astrogliosis with expression of Hsp27[J].Brain Res,2000,863(1-2):169-81.
    [10]Krueger-Naug AM,Emsley JG,Myers TL,et al.Administration of brain-derived neurotrophic factor suppresses the expression of heat shock protein 27 in rat retinal ganglion cells following axotomy[J].Neuroscience,2003,116(l):49-58.
    [11]Spencer JL,Waters EM,Romeo RD,et al.Uncovering the mechanisms of estrogen effects on hippocampal function[J].Front Neuroendocrinol,2008,29(2):219-37.
    [12]Craig MC,Murphy DG.Estrogen:effects on normal brain function and neuropsychiatric disorders[J].Climacteric,2007,10 Suppl 2:97-104.
    [13]Bjornstrom L,Sjoberg M.Mechanisms of estrogen receptor signaling:convergence of genomic and nongenomic actions on target genes[J].Mol Endocrinol,2005,19(4):833-42.
    [14]Hall JM,Couse JF,Korach KS.The multifaceted mechanisms of estradiol and estrogen receptor signaling[J].J Biol Chem,2001,276(40):36869-72.
    [15]Thakur MK,Sharma PK.Binding of estrogen receptor alpha promoter to nuclear proteins of mouse cerebral cortex:effect of age,sex,and gonadal steroids[J].Biogerontology,2008,9(6):467-78.
    [16]Kalesnykas G,Roschier U,Puolivali J,et al.The effect of aging on the subcellular distribution of estrogen receptor-alpha in the cholinergic neurons of transgenic and wild-type mice[J].Eur J Neurosci,2005,21(5):1437-42.
    [17]Rocca WA,Bower JH,Maraganore DM,et al.Increased risk of cognitive impairment or dementia in women who underwent oophorectomy before menopause[J].Neurology,2007,69(ll):1074-83.
    [18]Shuster LT,Gostout BS,Grossardt BR,et al.Prophylactic oophorectomy in premenopausal women and long-term health[J].Menopause Int,2008,14(3):111-6.
    [19]Sherwin BB,Henry JF.Brain aging modulates the neuroprotective effects of estrogen on selective aspects of cognition in women:a critical review[J].Front Neuroendocrinol,2008,29(1):88-113.
    [20]Benedetti MD,Maraganore DM,Bower JH,et al.Hysterectomy,menopause,and estrogen use preceding Parkinson's disease:an exploratory case-control study[J].Mov Disord,2001,16(5):830-7.
    [21]Chang CK,Chang CP,Liu SY,et al.Oxidative stress and ischemic injuries in heat stroke[J].Prog Brain Res,2007,162:525-46.
    [22]Lee WC,Wen HC,Chang CP,et al.Heat shock protein 72 overexpression protects against hyperthermia,circulatory shock,and cerebral ischemia during heatstroke[J].J Appl Physiol,2006,100(6):2073-82.
    [23]Min JN,Whaley RA,Sharpless NE,et al.CHIP deficiency decreases longevity,with accelerated aging phenotypes accompanied by altered protein quality control[J].Mol Cell Biol,2008,28(12):4018-25.
    [24]Rumora L,Lovric J,Sairam MR,et al.Impairments of heat shock protein expression and MAPK translocation in the central nervous system of follitropin receptor knockout mice[J].Exp Gerontol,2007,42(7):619-28.
    [25]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[J].Mech Ageing Dev,2004,125(4):325-35.
    [26]Pardue S,Groshan K,Raese JD,et al.Hsp70 mRNA induction is reduced in neurons of aged rat hippocampus after thermal stress[J].Neurobiol Aging,1992,13(6):661-72.
    [27]Manthey D,Behl C.From structural biochemistry to expression profiling:neuroprotective activities of estrogen[J].Neuroscience,2006,138(3):845-50.
    [28]Nikolic I,Liu D,Bell JA,et al.Treatment with an estrogen receptor-betaselective agonist is cardioprotective[J].J Mol Cell Cardiol,2007,42(4):769-80.
    [29]Chatterjee S,Premachandran S,Sharma D,et al.Therapeutic treatment with Larginine rescues mice from heat stroke-induced death:physiological and molecular mechanisms[J].Shock,2005,24(4):341-7.
    [30]Bao XQ,Liu GT.Induction of heat shock protein 27 and 70 overexpression by bicyclol attenuates concanavalin A-induced liver injury through suppression of NF-{kappa} B in mice[J].Mol Pharmacol,2009
    [31]Calabrese V,Cornelius C,Mancuso C,et al.Cellular stress response:a novel target for chemoprevention and nutritional neuroprotection in aging,neurodegenerative disorders and longevity[J].Neurochem Res,2008,33(12):2444-71.
    [32]Blake MJ,Fargnoli J,Gershon D,et al.Concomitant decline in heat-induced hyperthermia and HSP70 mRNA expression in aged rats[J].Am J Physiol,1991,260:R663-7.
    [33]Li S,Zheng J,Carmichael ST.Increased oxidative protein and DNA damage but decreased stress response in the aged brain following experimental stroke[J].Neurobiol Dis,2005,18(3):432-40.
    [34]Galli RL,Bielinski DF,Szprengiel A,et al.Blueberry supplemented diet reverses age-related decline in hippocampal HSP70 neuroprotection[J].Neurobiol Aging,2006,27(2):344-50.
    [35]Horowitz M,Robinson SD.Heat shock proteins and the heat shock response during hyperthermia and its modulation by altered physiological conditions[J]. Prog Brain Res,2007,162:433-46.
    [36]Shamovsky I,Gershon D.Novel regulatory factors of HSF-1 activation:facts and perspectives regarding their involvement in the age-associated attenuation of the heat shock response[J].Mech Ageing Dev,2004,125(10-11):767-75.
    [37]Marriott LK,McGann-Gramling KR,Hauss-Wegrzyniak B,et al.Estrogen replacement regimen and brain infusion of lipopolysaccharide differentially alter steroid receptor expression in the uterus and hypothalamus[J].Endocrine,2007,32(3):317-28.
    [38]Voss MR,Stallone JN,Li M,et al.Gender differences in the expression of heat shock proteins:the effect of estrogen[J].Am J Physiol Heart Circ Physiol,2003,285(2):H687-92.
    [39]Chen GZ,Luo BD,Wang HQ,et al.Effects of hyperthermia on rat hippocampal pyramidal cell apoptosis in vitro[J].Di Yi Jun Yi Da Xue Xue Bao,2003,23(3):233-5.
    [40]White MG,Emery M,Nonner D,et al.Caspase activation contributes to delayed death of heat-stressed striatal neurons[J].J Neurochem,2003,87(4):958-68.
    [41]White MG,Luca LE,Nonner D,et al.Cellular mechanisms of neuronal damage from hyperthermia[J].Prog Brain Res,2007,162:347-71.
    [42]Obrenovitch TP.Molecular physiology of preconditioning-induced brain tolerance to ischemia[J].Physiol Rev,2008,88(l):211-47.
    [43]Al-Khlaiwi T,Al-Drees A,Gursoy E,et al.Estrogen protects cardiac myogenic (H9c2) rat cells against lethal heat shock-induced cell injury:modulation of estrogen receptor alpha,glucocorticoid receptors,heat shock protein 70,and iNOS[J].J Cardiovasc Pharmacol,2005,45(3):217-24.
    [44]Suzuki T,Bramlett HM,Ruenes G,et al.The effects of early post-traumatic hyperthermia in female and ovariectomized rats[J].J Neurotrauma,2004,21(7):842-53.
    [45]Plahta WC,Clark DL,Colbourne F.17beta-estradiol pretreatment reduces CA1 sector cell death and the spontaneous hyperthermia that follows forebrain ischemia in the gerbil[J].Neuroscience,2004,129(1):187-93.
    [1]Selkoe DJ.Alzheimer's disease:genes,proteins,and therapy[J].Physiol Rev,2001,81(2):741-66.
    [2]Hardy J,Selkoe DJ.The amyloid hypothesis of Alzheimer's disease:progress and problems on the road to therapeutics[J].Science,2002,297(5595):353-6.
    [3]Cummings JL,Vinters HV,Cole GM,et al.Alzheimer's disease:etiologies,pathophysiology,cognitive reserve,and treatment opportunities[J].Neurology, 1998,51:S2-17;discussion S65-7.
    [4]Dudas B,Cornelli U,Lee JM,et al.Oral and subcutaneous administration of the glycosaminoglycan C3 attenuates Abeta (25-35)-induced abnormal tau protein imrnunoreactivity in rat brain[J].Neurobiol Aging,2002,23(1):97-104.
    [5]Yallampalli S,Micci MA,Taglialatela G.Ascorbic acid prevents beta-amyloidinduced intracellular calcium increase and cell death in PC 12 cells[J].Neurosci Lett,1998,251(2):105-8.
    [6]Monji A,Utsumi H,Ueda T,et al.Amyloid-beta-protein (A beta) (25-35)-associated free radical generation is strongly influenced by the aggregational state of the peptides[J].Life Sci,2002,70(7):833-41.
    [7]Rah JC,Kim HS,Kim SS,et al.Effects of carboxyl-terminal fragment of Alzheimer's amyloid precursor protein and amyloid beta-peptide on the production of cytokines and nitric oxide in glial cells[J].Faseb J,2001,15(8):1463-5.
    [8]Walsh DM,Klyubin I,Fadeeva JV,et al.Naturally secreted oligomers of amyloid beta protein potently inhibit hippocampal long-term potentiation in vivo[J].Nature,2002,416(6880):535-9.
    [9]Gong Y,Chang L,Viola KL,et al.Alzheimer's disease-affected brain:presence of oligomeric A beta ligands (ADDLs) suggests a molecular basis for reversible memory loss[J].Proc Natl Acad Sci USA,2003,100(18):10417-22.
    [10]Sinha S,Lieberburg I.Cellular mechanisms of beta-amyloid production and secretion[J].Proc Natl Acad Sci U S A,1999,96(20):11049-53.
    [11]Yan R,Bienkowski MJ,Shuck ME,et al.Membrane-anchored aspartyl protease with Alzheimer's disease beta-secretase activity[J].Nature,1999,402(6761):533-7.
    [12]De Strooper B,Saftig P,Craessaerts K,et al.Deficiency of presenilin-1 inhibits the normal cleavage of amyloid precursor protein[J].Nature,1998,391(6665):387-90.
    [13]Lammich S,Kojro E,Postina R,et al.Constitutive and regulated alpha-secretase cleavage of Alzheimer's amyloid precursor protein by a disintegrin metalloprotease[J].Proc Natl Acad Sci USA,1999,96(7):3 922-7.
    [14]Nunan J,Small DH.Regulation of APP cleavage by alpha-,beta-and gammasecretases[J].FEBS Lett,2000,483(1):6-10.
    [15]Peng Y,Jiang L,Lee DY,et al.Effects of huperzine A on amyloid precursor protein processing and beta-amyloid generation in human embryonic kidney 293 APP Swedish mutant cells[J].J Neurosci Res,2006,84(4):903-11.
    [16]Luo Y,Smith JV,Paramasivam V,et al.Inhibition of amyloid-beta aggregation and caspase-3 activation by the Ginkgo biloba extract EGb761[J].Proc Natl Acad Sci USA,2002,99(19):12197-202.
    [17]Yang F,Lim GP,Begum AN,et al.Curcumin inhibits formation of amyloid beta oligomers and fibrils,binds plaques,and reduces amyloid in vivo[J].J Biol Chem,2005,280(7):5892-901.
    [18]Ghosh AK,Bilcer G,Hong L,et al.Memapsin 2 (beta-secretase) inhibitor drug,between fantasy and reality[J].Curr Alzheimer Res,2007,4(4):418-22.
    [19]Wolfe MS.Inhibition and modulation of gamma-secretase for Alzheimer's disease[J].Neurotherapeutics,2008,5(3):391-8.
    [20]Bandyopadhyay S,Goldstein LE,Lahiri DK,et al.Role of the APP nonamyloidogenic signaling pathway and targeting alpha-secretase as an alternative drug target for treatment of Alzheimer's disease[J].Curr Med Chem,2007,14(27):2848-64.
    [21]Wallace WC,Akar CA,Lyons WE.Amyloid precursor protein potentiates the neurotrophic activity of NGF[J].Brain Res Mol Brain Res,1997,52(2):201-12.
    [22]Mattson MP.Cellular actions of beta-amyloid precursor protein and its soluble and fibrillogenic derivatives[J].Physiol Rev,1997,77(4):1081-132.
    [23]Postina R,Schroeder A,Dewachter I,et al.A disintegrin-metalloproteinase prevents amyloid plaque formation and hippocampal defects in an Alzheimer disease mouse model[J].J Clin Invest,2004,113(10):1456-64.
    [24]Obregon DF,Rezai-Zadeh K,Bai Y,et al.ADAM 10 activation is required for green tea (-)-epigallocatechin-3-gallate-induced alpha-secretase cleavage of amyloid precursor protein[J].J Biol Chem,2006,281(24):16419-27.
    [25]Fang F,Liu GT.Protective effects of compound FLZ on beta-amyloid peptide- (25-35)-induced mouse hippocampal injury and learning and memory impairment[J].Acta Pharmacol Sin,2006,27(6):651-8.
    [26]Fang F,Liu GT.Novel squamosamide derivative (compound FLZ) attenuates Abeta25-35-induced toxicity in SH-SY5Y cells[J].Acta Pharmacol Sin,2008,29(2):152-60.
    [27]Thinakaran G,Teplow DB,Siman R,et al.Metabolism of the“Swedish”amyloid precursor protein variant in neuro2a (N2a) cells.Evidence that cleavage at the“beta-secretase”site occurs in the golgi apparatus[J].J Biol Chem,1996,271(16):9390-7.
    [28]Waxman EA,Baconguis I,Lynch DR,et al.N-methyl-D-aspartate receptordependent regulation of the glutamate transporter excitatory amino acid carrier 1[J].J Biol Chem,2007,282(24):17594-607.
    [29]Borchelt DR,Thinakaran G,Eckman CB,et al.Familial Alzheimer's diseaselinked presenilin 1 variants elevate Abetal-42/1-40 ratio in vitro and in vivo[J].Neuron,1996,17(5):1005-13.
    [30]Fu Z,Lee SH,Simonetta A,et al.Differential roles of Rapl and Rap2 small GTPases in neurite retraction and synapse elimination in hippocampal spiny neurons[J].J Neurochem,2007,100(1):118-31.
    [31]Zimmermann M,Gardoni F,Marcello E,et al.Acetylcholinesterase inhibitors increase ADAM 10 activity by promoting its trafficking in neuroblastoma cell lines[J].J Neurochem,2004,90(6):1489-99.
    [32]Hwang EM,Kim SK,Sohn JH,et al.Furin is an endogenous regulator of alphasecretase associated APP processing[J].Biochem Biophys Res Commun,2006,349(2):654-9.
    [33]Skovronsky DM,Fath S,Lee VM,et al.Neuronal localization of the TNFalpha converting enzyme (TACE) in brain tissue and its correlation to amyloid plaques[J].J Neurobiol,2001,49(l):40-6.
    [34]Xu H,Sweeney D,Wang R,et al.Generation of Alzheimer beta-amyloid protein in the trans-Golgi network in the apparent absence of vesicle formation[J],Proc Natl Acad Sci USA,1997,94(8):3748-52.
    [35]Xu H,Shields D.Prohormone processing in the trans-Golgi network: endoproteolytic cleavage of prosomatostatin and formation of nascent secretory vesicles in permeabilized cells[J].J Cell Biol,1993,122(6):1169-84.
    [36]Selkoe DJ.Cell biology of the amyloid beta-protein precursor and the mechanism of Alzheimer's disease[J].Annu Rev Cell Biol,1994,10:373-403.
    [37]Klausner RD,Donaldson JG,Lippincott-Schwartz J.Brefeldin A:insights into the control of membrane traffic and organelle structure[J].J Cell Biol,1992,116(5):1071-80.
    [38]Colciaghi F,Borroni B,Pastorino L,et al.[alpha]-Secretase ADAM10 as well as[alpha]APPs is reduced in platelets and CSF of Alzheimer disease patients[J].Mol Med,2002,8(2):67-74.
    [39]Bernstein HG,Bukowska A,Krell D,et al.Comparative localization of ADAMs 10 and 15 in human cerebral cortex normal aging,Alzheimer disease and Down syndrome[J].J Neurocytol,2003,32(2):153-60.
    [40]Gatta LB,Albertini A,Ravid R,et al.Levels of beta-secretase BACE and alphasecretase ADAM 10 mRNAs in Alzheimer hippocampus[J].Neuroreport,2002,13(16):2031-3.
    [41]Thomas G.Furin at the cutting edge:from protein traffic to embryogenesis and disease[J].Nat Rev Mol Cell Biol,2002,3(10):753-66.
    [42]Anderson ED,VanSlyke JK,Thulin CD,et al.Activation of the furin endoprotease is a multiple-step process:requirements for acidification and internal propeptide cleavage[J].Embo J,1997,16(7):1508-18.
    [43]Lopez-Perez E,Zhang Y,Frank SJ,et al.Constitutive alpha-secretase cleavage of the beta-amyloid precursor protein in the furin-deficient LoVo cell line:involvement of the pro-hormone convertase 7 and the disintegrin metalloprotease ADAM10[J].J Neurochem,2001,76(5):1532-9.
    [44]Chen M,Fernandez HL.Stimulation of beta-amyloid precursor protein alphaprocessing by phorbol ester involves calcium and calpain activation[J].Biochem Biophys Res Commun,2004,316(2):332-40.
    [45]Xu H,Greengard P,Gandy S.Regulated formation of Golgi secretory vesicles containing Alzheimer beta-amyloid precursor protein[J].J Biol Chem,1995,270(40):23243-5.
    [46]Koo EH.Phorbol esters affect multiple steps in beta-amyloid precursor protein trafficking and amyloid beta-protein production[J].Mol Med,1997,3(3):204-11.
    [47]Skovronsky DM,Moore DB,Milla ME,et al.Protein kinase C-dependent alphasecretase competes with beta-secretase for cleavage of amyloid-beta precursor protein in the trans-golgi network[J].J Biol Chem,2000,275(4):2568-75.
    [48]Lee W,Boo JH,Jung MW,et al.Amyloid beta peptide directly inhibits PKC activation[J].Mol Cell Neurosci,2004,26(2):222-31.
    [49]Olariu A,Yamada K,Mamiya T,et al.Memory impairment induced by chronic intracerebroventricular infusion of beta-amyloid (1-40) involves downregulation of protein kinase C[J].Brain Res,2002,957(2):278-86.
    [50]Yogev-Falach M,Amit T,Bar-Am O,et al.Involvement of MAP kinase in the regulation of amyloid precursor protein processing by novel cholinesterase inhibitors derived from rasagiline[J].Faseb J,2002,16(12):1674-6.
    [51]Yang HQ,Pan J,Ba MW,et al.New protein kinase C activator regulates amyloid precursor protein processing in vitro by increasing alpha-secretase activity[J].Eur J Neurosci,2007,26(2):381-91.
    [52]Iwata N,Tsubuki S,Takaki Y,et al.Identification of the major Abetal-42-degrading catabolic pathway in brain parenchyma:suppression leads to biochemical and pathological deposition[J].Nat Med,2000,6(2):143-50.
    [53]Eckman EA,Reed DK,Eckman CB.Degradation of the Alzheimer's amyloid beta peptide by endothelin-converting enzyme[J].J Biol Chem,2001,276(27):24540-8.
    [54]Caccamo A,Oddo S,Sugarman MC,et al.Age-and region-dependent alterations in Abeta-degrading enzymes:implications for Abeta-induced disorders[J].Neurobiol Aging,2005,26(5):645-54.
    [1]Barnham KJ,Cappai R,Beyreuther K,et al.Delineating common molecular mechanisms in Alzheimer's and prion diseases[J].Trends Biochem Sci,2006,31:465-472.
    [2]Hinault MP,Ben-Zvi A,Goloubinoff P.Chaperones and proteases:cellular foldcontrolling factors of proteins in neurodegenerative diseases and aging[J].J Mol Neurosci,2006,30:249-265.
    [3]Calabrese V,Guagliano E,Sapienza M,et al.Redox regulation of cellular stress response in aging and neurodegenerative disorders:role of vitagenes[J].Neurochem Res,2007,32:757-773.
    [4]Poon F,Calabrese V,Scapagnini G,et al.Free radicals:key to brain aging and heme oxygenase as a cellular response to oxidative stress[J].J Gerontol A Biol Sci Med Sci,2004,59:478-493.
    [5]Mosser DD,Morimoto RI.Molecular chaperones and the stress of oncogenesis[J].Oncogene,2004,23:2907-2918.
    [6]Calabrese V,Butterfield DA,Scapagnini G,et al.Redox regulation of heat shock protein expression by signaling involving nitric oxide and carbon monoxide:relevance to brain aging,neurodegenerative disorders,and longevity[J].Antioxid Redox Signal,2006,8:444-477.
    [7]Sultana R,Ravagna A,Mohmmad-Abdul H,et al.Ferulic acid ethyl ester protects neurons against amyloid beta-peptide(l-42)-induced oxidative stress and neurotoxicity:relationship to antioxidant activity[J].J Neurochem,2005,92:749-758.
    [8]Calabrese V,Stella AM,Butterfield DA,et al.Redox regulation in neurodegeneration and longevity:role of the heme oxygenase and HSP70 systems in brain stress tolerance[J].Antioxid Redox Signal,2004,6:895-913.
    [9]Calabrese V,Ravagna A,Colombrita C,et al.Acetylcarnitine induces heme oxygenase in rat astrocytes and protects against oxidative stress:involvement of the transcription factor Nrf2[J].J Neurosci Res,2005,79:509-521.
    [10]Maines MD.The heme oxygenase system:a regulator of second messenger gases[J].Annu Rev Pharmacol Toxicol,1997,37:517-554.
    [11]Maines MD.Zinc-protoporphyrin is a selective inhibitor of heme oxygenase activity in the neonatal rat[J].Biochim Biophys Acta,1981,673:339-350.
    [12]Mancuso C.Heme oxygenase and its products in the nervous system[J].Antioxid Redox Signal,2004,6:878-887.
    [13]Sun J,Hoshino H,Takaku K,et al.Hemoprotein Bachl regulates enhancer availability of heme oxygenase-1 gene[J].EMBO J,2002,21:5216-5224.
    [14]Stewart D,Killeen E,Naquin R,et al.Degradation of transcription factor Nrf2 via the ubiquitin-proteasome pathway and stabilization by cadmium[J].J Biol Chem,2003,278:2396-2402.
    [15]Scapagnini G,D'Agata V,Calabrese V,et al.Gene expression profiles of heme oxygenase isoforms in the rat brain[J].Brain Res,2002,954:51-59.
    [16]Nakamura H.Thioredoxin and its related molecules:update 2005[J].Antioxid Redox Signal,2005,7:823-828.
    [17]Sadek CM,Jimenez A,Damdimopoulos AE,et al.Characterization of human thioredoxin-like 2.A novel microtubule-binding thioredoxin expressed predominantly in the cilia of lung airway epithelium and spermatid manchette and axoneme[J],J Biol Chem,2003,278:13133-13142.
    [18]Pekkari K,Holmgren A.Truncated thioredoxin:physiological functions and mechanism[J].Antioxid Redox Signal,2004,6:53-61.
    [19]Gromer S,Urig S,Becker K.The thioredoxin system—from science to clinic[J].Med Res Rev,2004,24:40-89.
    [20]Kaimul AM,Nakamura H,Masutani H,et al.Thioredoxin and thioredoxin-binding protein-2 in cancer and metabolic syndrome[J].Free Radic Biol Med,2007,43:861-868.
    [21]Yoshida T,Oka S,Masutani H,et al.The role of thioredoxin in the aging process:involvement of oxidative stress[J].Antioxid Redox Signal,2003,5:563-570.
    [22]Wiesel P,Foster LC,Pellacani A,et al.Thioredoxin facilitates the induction of heme oxygenase-1 in response to inflammatory mediators[J].J Biol Chem,2000,275:24840-24846,
    [23]Kabe Y,Ando K,Hirao S,et al.Redox regulation of NF-kappaB activation:distinct redox regulation between the cytoplasm and the nucleus[J].Antioxid Redox Signal,2005,7:395-403.
    [24]Hirota K,Nakamura H,Masutani H,et al.Thioredoxin superfamily and thioredoxininducing agents[J].Ann NY Acad Sci,2002,957:189-199.
    [25]Poon F,Calabrese V,Scapagnini G,et al.Free radicals:key to brain aging and heme oxygenase as a cellular response to oxidative stress[J].J Gerontol A Biol Sci Med Sci,2004,59:478-493.
    [26]Butterfield DA,Castegna A,Pocernich CB,et al.Nutritional approaches to combat oxidative stress in Alzheimer's disease[J].J Nutr Biochem,2002,13:444-461.
    [27]Sultana R,Boyd-Kimball D,Cai J,et al.Proteomics analysis of the Alzheimer's disease hippocampal proteome[J].J Alzheimers Dis,2007,11:153-164
    [28]Magrane J,Smith RC,Walsh K,et al.Heat shock protein 70 participates in the neuroprotective response to intracellularly expressed beta-amyloid in neurons[J].J Neurosci,2004,24:1700-1706.
    [29]Magrane J,Rosen KM,Smith RC,et al.Intraneuronal beta-amyloid expression downregulates the Akt survival pathway and blunts the stress response[J].J Neurosci,2005,25:10960-10969.
    [30]Elliott E,Tsvetkov P,Ginzburg I.BAG-1 associates with Hsc70.Tau complex and regulates the proteasomal degradation of Tau protein[J].J Biol Chem,2007,282:37276-37284.
    [31]Kumar P,Ambasta RK,Veereshwarayya V,et al.CHIP and HSPs interact with beta-APP in a proteasome-dependent manner and influence Abeta metabolism[J].Hum Mol Genet,2007,16:848-864.
    [32]Sahara N,Murayama M,Mizoroki T,et al.In vivo evidence of CHIP up-regulation attenuating tau aggregation[J].J Neurochem,2005,94:1254-1263.
    [33]Chiu PY,Leung HY,Poon MK,et al.Chronic schisandrin B treatment improves mitochondrial antioxidant status and tissue heat shock protein production in various tissues of young adult and middle-aged rats[J].Biogerontology,2006,7:199-210.
    [34]Unno K,Asakura H,Shibuya Y,et al.Stress response caused by chronic alcohol intake in aged rat brain[J].Alcohol Clin Exp Res,2002,26:1017-1023.
    [35]Rumora L,Lovric J,Sairam MR,et al.Impairments of heat shock protein expression and MAPK translocation in the central nervous system of follitropin receptor knockout mice[J].Exp Gerontol,2007,42:619-628.
    [36]Crivello NA,Rosenberg IH,Dallal GE,et al.Age-related changes in neutral sphingomyelin-specific phospholipase C activity in striatum,hippocampus,and frontal cortex:implication for sensitivity to stress and inflammation[J].Neurochem Int,2005,47:573-579.
    [37]Horowitz M,Robinson SD.Heat shock proteins and the heat shock response during hyperthermia and its modulation by altered physiological conditions[J].Prog Brain Res,2007,162:433-446.
    [38]Li S,Zheng J,Carmichael ST.Increased oxidative protein and DNA damage but decreased stress response in the aged brain following experimental stroke[J].Neurobiol Dis,2005,18:432-440.
    [39]Sharma S,Kaur G Neuroprotective potential of dietary restriction against kainate-induced excitotoxicity in adult male Wistar rats[J.].Brain Res Bull,2005,67:482-491.
    [40]Schipper HM.Heme oxygenase expression in human central nervous system disorders[J].Free Radic Biol Med,2004,37:1995-2011.
    [41]Calabrese V,Sultana R,Scapagnini G,et al.Nitrosative stress,cellular stress response,and thiol homeostasis in patients with Alzheimer's disease[J].Antioxid Redox Signal,2006,8:1975-1986.
    [42]Schipper HM,Bennett DA,Liberman A,et al.Glial heme oxygenase-1 expression in Alzheimer disease and mild cognitive impairment[J].Neurobiol Aging,2006,27:252-261.
    [43]Vaya J,Song W,Khatib S,et al.Effects of heme oxygenase-1 expression on sterol homeostasis in rat astroglia[J].Free Radic Biol Med,2007,42:864-871.
    [44]Takeda A,Perry G,Abraham NG,et al.Overexpression of heme oxygenase in neuronal cells,the possible interaction with Tau[J].J Biol Chem,2000,275:5395-5399.
    [45]Infante J,Rodriguez-Rodriguez E,Mateo I,et al.Gene-gene interaction between heme oxygenase-1 and liver X receptor-beta and Alzheimer's disease risk[J].Neurobiol Aging,2008,[Epub ahead of print]
    [46]Dwyer BE,Takeda A,Zhu X,et al.Ferric cycle activity and Alzheimer disease[J].Curr Neurovasc Res,2005,2:261-267.
    [47]Ghribi O,Golovko MY,Larsen B,et al.Deposition of iron and beta-amyloid plaques is associated with cortical cellular damage in rabbits fed with long-term cholesterol-enriched diets[J].J Neurochem,2006,99:438-449.
    [48]Hirose W,Ikematsu K,Tsuda R.Age-associated increases in heme oxygenase-1 and ferritin immunoreactivity in the autopsied brain[J].Leg Med,2003,1:S360-366.
    [49]Colombrita C,Calabrese V,Stella AM,et al.Regional rat brain distribution of heme oxygenase-1 and manganese superoxide dismutase mRNA:relevance of redox homeostasis in the aging processes[J].Exp Biol Med,2003,228:517-524.
    [50]Chernova T,Nicotera P,Smith AG.Heme deficiency is associated with senescence and causes suppression of N-mefhyl-D-aspartate receptor subunits expression in primary cortical neurons[J].Mol Pharmacol,2006,69:697-705.
    [51]Pertusa M,Garcia-Matas S,Rodriguez-Farre E,et al.Astrocytes aged in vitro show a decreased neuroprotective capacity[J].J Neurochem,2007,101:794-805.
    [52]Ewing JF,Maines MD.Regulation and expression of heme oxygenase enzymes in aged-rat brain:age related depression in HO-1 and HO-2 expression and altered stress-response[J].J Neural Transm,2006,113:439-454.
    [53]Hoekstra KA,Velleman SG.Brain microvascular and intracranial artery resistance to atherosclerosis is associated with heme oxygenase and ferritin in Japanese quail[J].Mol Cell Biochem,2008,307:1-12.
    [54]Lovell MA,Xie C,Gabbita SP,et al.Decreased thioredoxin and increased thioredoxin reductase levels in Alzheimer's disease brain[J].Free Radic Biol Med,2000,28:418-427.
    [55]Akterin S,Cowburn RF,Miranda-Vizuete A,et al.Involvement of glutaredoxin-1 and thioredoxin-1 in beta-amyloid toxicity and Alzheimer's disease[J].Cell Death Differ,2006,13:1454-1465.
    [56]Takagi Y,Hattori I,Nozaki K,et al.Excitotoxic hippocampal injury is attenuated in thioredoxin transgenic mice[J].J Cereb Blood Flow Metab,2000,20:829-833.
    [57]Masutani H,Bai J,Kim YC,et al.Thioredoxin as a neurotrophic cofactof and an important regulator of neuroprotection[J].Mol Neurobiol,2004,29:229-242.
    [58]Chiueh C,Lee S,Andoh T,et al.Induction of antioxidative and antiapoptotic thioredoxin supports neuroprotective hypothesis of estrogen[J].Endocrine,2003,21:27-31.
    [59]de Magalhaes JP,Cabral JA,Magalhaes D.The influence of genes on the aging process of mice:a statistical assessment of the genetics of aging[J].Genetics,2005,169:265-274.
    [60]Mitsui A,Hamuro J,Nakamura H,et al.Overexpression of human thioredoxm in transgenic mice controls oxidative stress and life span[J].Antioxid Redox Signal,2002,4:693-696.
    [61]Svensson MJ,Larsson J.Thioredoxin-2 affects lifespan and oxidative stress in Drosophila[J].Hereditas,2007,144:25-32.
    [62]Perez VI,Lew CM,Cortez LA,et al.Thioredoxin 2 haploinsufficiency in mice results in impaired mitochondrial function and increased oxidative stress[J].Free Radic Biol Med,2008,44:882-892.
    [63]Yoshida T,Kondo N,Oka S,et al.Thioredoxin-binding protein-2 (TBP-2):its potential roles in the aging process[J].Biofactors,2006,27:47-51.
    [64]Yoshida T,Nakamura H,Masutani H,et al.The involvement of thioredoxin and thioredoxin binding protein-2 on cellular proliferation and aging process[J].Ann N YAcadSci,2005,1055:1-12.
    [65]Osawa T,Sugiyama Y,Inayoshi M,et al.Anti-oxidative activity of tetrahydrocurcuminoids[J].Biosci Biotechnol Biochem,1995,59:1609-1612.
    [66]Balogun E,Foresti R,Green CJ,et al.Changes in temperature modulate heme oxygenase-1 induction by curcumin in renal epithelial cells[J].Biochem Biophys Res Commun,2003,308:950-955.
    [67]Kato K,Ito H,Kamei K,et al.Stimulation of the stress-induced expression of stress proteins by curcumin in cultured cells and in rat tissues in vivo[J].Cell Stress Chaperones,1998,3:152-160.
    [68]McDaniel MA,Maier SF,Einstein GO.“Brain-specific”nutrients:a memory cure[J]?Nutrition,2003,19:957-975.
    [69]Spagnoli A,Lucca U,Menasce G,et al.Long-term acetyl-L-carnitine treatment in Alzheimer's disease[J].Neurology,1991,41:1726-1732.
    [70]Abdul HM,Calabrese V,Calvani M,et al.Acetyl-L-carnitine-induced up-regulation of heat shock proteins protects cortical neurons against amyloid-beta peptide 1-42-mediated oxidative stress and neurotoxicity:implications for Alzheimer's disease[J].J Neurosci Res,2006,84:398-408.
    [71]Calabrese V,Ravagna A,Colombrita C,et al.Acetylcarnitine induces heme oxygenase in rat astrocytes and protects against oxidative stress:involvement of the transcription factor Nrf2[J].J Neurosci Res,2005,79:509-521.
    [72]Zhuang H,Kim YS,Koehler RC et al.Potential mechanism by which resveratrol,a red wine constituent,protects neurons[J].Ann N Y Acad Sci,2003,993:276-286.
    [73]Nakao A,Kaczorowski DJ,Zuckerbraun BS,et al.Galantamine and carbon monoxide protect brain microvascular endothelial cells by heme oxygenase-1 induction[J].Biochem Biophys Res Commun,2008,367:674-679.
    [74]Liu J,Yin F,Zheng X,et al.Geniposide,a novel agonist for GLP-1 receptor,prevents PC 12 cells from oxidative damage via MAP kinase pathway[J].Neurochem Int,2007,51:361-369.
    [75]Chow AM,Brown IR.Induction of heat shock proteins in differentiated human and rodent neurons by celastrol[J].Cell Stress Chaperones,2007,12:37-44.
    [76]Perluigi M,Joshi G,Sultana R,et al.In vivo protective effects of ferulic acid ethyl ester against amyloid-beta peptide 1-42-induced oxidative stress[J].J Neurosci Res,2006,84:418-426.
    [77]Wang X,Qin ZH,Leng Y,et al.Prostaglandin Al inhibits rotenone-induced apoptosis in SH-SY5Y cells[J].J Neurochem,2002,83:1094-1102.

© 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