Amyloid beta-Protein Precursor

淀粉样 β 蛋白前体
  • 文章类型: Journal Article
    目的:探讨槲皮素对APP/PS1双转基因小鼠阿尔茨海默病(AD)样病理的改善作用及其机制。有助于理解AD的发病机制。方法:将30只APP/PS1转基因小鼠随机分为模型组(APP/PS1),槲皮素组(APP/PS1+Q),盐酸多奈哌齐组(APP/PS1+DON)。同时,有10只相同年龄的C57小鼠作为对照组。治疗后三个月,使用Morris水迷宫(MWM)试验评估槲皮素对AD小鼠的影响,Y迷宫实验,免疫组织化学,免疫荧光,和西方印迹。结果:水迷宫和Y迷宫结果表明槲皮素可明显改善APP/PS1转基因AD小鼠的认知障碍。此外,血清酶联免疫吸附试验(ELISA)结果表明槲皮素升高MDA,超氧化物歧化酶(SOD),CAT,GSH,乙酰胆碱(ACh),和AD小鼠的乙酰胆碱酯酶(AChE)水平。苏木精-伊红(HE)染色,尼氏染色,海马组织硫黄染色显示槲皮素可降低AD小鼠的神经元损伤和Aβ蛋白积累。Westernblot验证了与氧化应激和凋亡相关的Kelch样ECH相关蛋白1(Keap1)/核因子红细胞2相关因子2(Nrf2)/HO-1通路中的蛋白表达,确认槲皮素增强AD小鼠认知的潜在分子机制。此外,蛋白质印迹结果表明槲皮素显著改变Keap1/Nrf2/HO-1途径中的蛋白表达。此外,分子对接分析表明Keap1/Nrf2/HO-1通路中的Keap1、NQO1、HO-1、caspase-3、Bcl-2和Bax蛋白可能是槲皮素的潜在调控靶点。这些发现将为槲皮素在AD治疗中的临床应用提供分子基础。结论:槲皮素可改善APP/PS1双转基因小鼠认知功能损害及AD样病理,可能与槲皮素激活Keap1/Nrf2/HO-1通路和减少细胞凋亡有关。
    Objective: The objective of the study is to investigate whether quercetin ameliorates Alzheimer\'s disease (AD)-like pathology in APP/PS1 double transgenic mice and its hypothesized mechanism, contributing to the comprehension of AD pathogenesis. Methods: A total of 30 APP/PS1 transgenic mice were randomized into model group (APP/PS1), quercetin group (APP/PS1+Q), and donepezil hydrochloride group (APP/PS1+DON). Simultaneously, there were 10 C57 mice of the same age served as a control group. Three months posttreatment, the effects of quercetin on AD mice were evaluated using the Morris water maze (MWM) test, Y maze experiment, immunohistochemistry, immunofluorescence, and western blotting. Results: Results from the water maze and Y maze indicated that quercetin significantly improved cognitive impairment in APP/PS1 transgenic AD mice. Additionally, serum enzyme-linked immunosorbent assay (ELISA) results demonstrated that quercetin elevated MDA, superoxide dismutase (SOD), CAT, GSH, acetylcholine (ACh), and acetylcholinesterase (AChE) levels in AD mice. Hematoxylin-eosin (HE) staining, Nissl staining, and hippocampal tissue thioflavine staining revealed that quercetin reduced neuronal damage and Aβ protein accumulation in AD mice. Western blot validated protein expression in the Kelch-like ECH-associated protein 1 (Keap1)/nuclear factor erythroid 2-related factor 2 (Nrf2)/HO-1 pathway associated with oxidative stress and apoptosis, confirming quercetin\'s potential molecular mechanism of enhancing AD mouse cognition. Furthermore, western blot findings indicate that quercetin significantly alters protein expression in the Keap1/Nrf2/HO-1 pathway. Moreover, molecular docking analysis suggests that Keap1, NQO1, HO-1, caspase-3, Bcl-2, and Bax proteins in the Keap1/Nrf2/HO-1 pathway may be potential regulatory targets of quercetin. These findings will provide a molecular basis for quercetin\'s clinical application in AD treatment. Conclusion: Quercetin can improve cognitive impairment and AD-like pathology in APP/PS1 double transgenic mice, potentially related to quercetin\'s activation of the Keap1/Nrf2/HO-1 pathway and reduction of cell apoptosis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • DOI:
    文章类型: English Abstract
    目的基于线粒体自噬和核苷酸结合寡聚化结构域样受体家族pyrin结构域3(NLRP3)炎症小体通路,探讨法舒地尔改善淀粉样前体蛋白/早老素-1(APP/PS1)转基因小鼠认知功能障碍的机制.方法将APP/PS1小鼠分为模型组和治疗组,以C57BL/6小鼠为对照组。治疗组给予法舒地尔(25mg/kg)腹腔注射,每日1次,连续2个月,对照组和模型组注射等量生理盐水。通过水迷宫和Y迷宫试验检测小鼠的行为;Nissl染色和神经元特异性核抗原(NeuN)免疫荧光组织化学染色用于评估神经元的数量和形态。末端脱氧核苷酸转移酶介导的dUTP-生物素缺口末端标记法(TUNEL)染色检测神经元凋亡;免疫荧光组织化学染色检测P62和NLRP3的表达;实时荧光定量PCR检测十号染色体上缺失的磷酸酶和张力蛋白同源物(PTEN)诱导的推定激酶1(PINK1)的mRNA表达水平,Parkin和NLRP3;Westernblot检测PINK1、Parkin、P62,微管相关蛋白1轻链3(LC3),NLRP3,衔接蛋白凋亡相关斑点样蛋白(ASC)和白介素18(IL-18)。结果水迷宫和Y迷宫实验结果显示,治疗组小鼠认知行为明显改善,其空间记忆和探索能力明显增强;Nissl染色和NeuN免疫荧光组织化学染色结果显示,模型组神经元数量和Nissl体数量均低于对照组,法舒地尔治疗后神经元形态和数量得到改善。TUNEL染色结果还显示,法舒地尔处理后,APP/PS1小鼠脑组织凋亡细胞数量减少;与对照组相比,模型组PINK1和Parkin的表达降低,而P62、LC3、NLRP3、ASC和IL-18的表达增加。法舒地尔治疗后,PINK1、Parkin、LC3增加了,而P62、NLRP3、ASC、IL-18下降。结论法舒地尔可改善APP/PS1小鼠的认知功能和神经元损伤,其机制可能与促进线粒体自噬和抑制NLRP3炎性体的活化有关。
    Objective To explore the mechanism of fasudil improving cognitive dysfunction in amyloid precursor protein/presenilin-1 (APP/PS1) transgenic mice based on mitophagy and nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome pathway. Methods APP/PS1 mice were divided into model group and treatment group, and C57BL/6 mice were used as control group. The treatment group was given intraperitoneal injection of Fasudil (25 mg/kg) once daily for 2 months, while the control group and the model group were injected with the same volume of normal saline. The behavior of mice was detected by water maze and Y maze test; Nissl staining and neuron-specific nuclear antigen (NeuN) immunofluorescence histochemical staining were used to evaluate the number and morphology of neurons. Terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling assay (TUNEL) staining was used to detect neuronal apoptosis; The expression of P62 and NLRP3 was detected by immunofluorescence histochemical staining; Real time fluorescence quantitative PCR was used to detect the mRNA expression levels of phosphatase and tensin homolog deleted on chromosome ten (PTEN) -induced putative kinase 1 (PINK1), Parkin and NLRP3; Western blot analysis was used to detect the expression of PINK1, Parkin, P62, microtubule-associated protein 1 light chain 3 (LC3), NLRP3, adapter protein apoptosis-associated speck-like protein (ASC) and interleukin-18 (IL-18). Results The results of the water maze and Y maze showed that the cognitive behavior of mice in treatment group was significantly improved, and their spatial memory and exploration abilities were significantly enhanced; The results of Nissl staining and NeuN immunofluorescence histochemical staining showed that the number of neurons and Nissl bodies were lower in the model group than that in the control group, while the morphology and number of neurons were improved after fasudil treatment. The results of TUNEL staining also showed that the number of apoptotic cells in the brain tissue of APP/PS1 mice were decreased after fasudil treatment; Compared with the control group, the expression of PINK1 and Parkin in the model group decreased, while the expression of P62, LC3, NLRP3, ASC and IL-18 increased. After treatment with fasudil, the expression of PINK1, Parkin, and LC3 increased, while the expression of P62, NLRP3, ASC, and IL-18 decreased. Conclusions Fasudil can improve the cognitive function and neuronal damage in APP/PS1 mice, and its mechanism may be related to promoting mitochondrial autophagy and inhibiting the activation of NLRP3 inflammasomes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    背景:中药固本健脑叶方(GBJNY)在中医治疗学习记忆障碍和老年失眠方面有着悠久的历史。该配方来源于孙思淼的五剂补药。此外,现代药理学研究揭示了其改善认知障碍和改善睡眠-觉醒昼夜节律紊乱的能力。然而,其疗效的确切机制仍然难以捉摸。
    目的:目前的研究通过转录组测序和实验验证,探讨了GBJNY在阿尔茨海默病昼夜节律睡眠-觉醒障碍和认知功能障碍中的调节作用和可能的机制。
    方法:利用LC-MS/MS串联技术定性地辨别存在于GBJNY中的活性组分。APP/PS1小鼠接受GBJNY或褪黑素连续治疗3个月。利用Morris水迷宫(MWM)测试对小鼠的学习和记忆能力进行评估,同时利用脑电图(EEG)和肌电图(EMG)研究睡眠变化。同时,研究了小鼠海马时钟基因的节律性。随后,我们用HE染色,高尔基染色,和免疫荧光观察GBJNY对突触损伤和神经元丢失的影响。我们进行了高通量测序以分析小鼠的mRNA表达谱,旨在鉴定差异表达基因(DEGs)。随后,我们进行了GO和KEGG富集分析,以探索相关的信号通路。此外,我们评估了小鼠海马中PI3K/AKT/mTOR通路和Aβ沉积相关蛋白的表达水平。通过这种全面的方法,我们试图阐明和验证GBJNY在APP/PS1小鼠中的潜在作用机制.
    结果:结果显示216个DEG。在此之后,我们进行了GO富集和KEGG通路分析,以更深入地研究mRNA靶基因的区别和基本功能。富集分析强调了PI3K/Akt/mTOR信号通路作为其中最关键的信号通路的重要性。通过体内实验,进一步证明,给予GBJNY可增强APP/PS1小鼠的记忆和学习能力.此外,GBJNY治疗导致睡眠-觉醒昼夜节律的改变,其特征在于觉醒减少和非快速眼动(NREM)睡眠增加。此外,Per1、Per2、Clock、在处理的小鼠的海马中注意到Cry1、Cry2和Bmal1mRNA。特别值得注意的是观察到海马内淀粉样β(Aβ)沉积的减少,神经元突触完整性的改善,和mTOR的上调,Akt,海马区PI3K蛋白表达。这些发现强调了PI3K/Akt/mTOR信号通路在减轻睡眠-觉醒昼夜节律紊乱中的关键参与。
    结论:GBJNY增强了APP/PS1小鼠的认知能力,改变了时钟基因表达模式,减轻睡眠-觉醒昼夜节律中断。基本机制似乎与PI3K/Akt/mTOR通路调节有关,为潜在的临床应用奠定了基础。
    BACKGROUND: The Chinese formula Guben-Jiannao Ye (GBJNY) formula has a long history of usage in traditional Chinese medicine (TCM) for the treatment of learning and memory disorders as well as senile insomnia. This formulation is derived from Sun Simiao\'s five tonic pills. Furthermore, modern pharmacological investigations have revealed its ability to improve cognitive impairment and ameliorate sleep-wake circadian rhythm disorders. However, the precise mechanism underlying its efficacy remains elusive.
    OBJECTIVE: The current research explored the modulatory effects and possible mechanisms of GBJNY in circadian rhythm sleep-wake disorders and cognitive dysfunction in Alzheimer\'s disease using transcriptome sequencing and experimental validation.
    METHODS: The LC-MS/MS tandem technology was utilized to qualitatively discern the active components present in GBJNY. The APP/PS1 mice received continuous treatment with GBJNY or Melatonin for 3 months. The learning and memory abilities of mice were assessed utilizing the Morris water maze (MWM) test, while sleep changes were studied utilizing the electroencephalogram (EEG) and electromyogram (EMG). Concurrently, mice\'s hippocampus clock gene rhythmicity was investigated. Subsequently, we employed HE staining, Golgi staining, and immunofluorescence to observe GBJNY\'s impact on synaptic damage and neuronal loss. We performed high-throughput sequencing to analyze the mRNA expression profiles of mice, aiming to identify differentially expressed genes (DEGs). Subsequently, we conducted GO and KEGG enrichment analyses to explore associated signaling pathways. Furthermore, we evaluated the expression levels of proteins involved in the PI3K/AKT/mTOR pathway and Aβ deposition in the hippocampus of mice. Through this comprehensive approach, we sought to elucidate and validate the potential mechanisms of action of GBJNY in APP/PS1 mice.
    RESULTS: Results showed 216 DEGs. Following this, we conducted GO enrichment and KEGG pathway analyses to delve deeper into the distinctions and fundamental functions of the mRNA target genes. The enrichment analysis underscored the prominence of the PI3K/Akt/mTOR signaling pathway as the most pivotal among them. Through in vivo experiments, it was further demonstrated that the administration of GBJNY enhanced memory and learning capacities in APP/PS1 mice. Additionally, GBJNY treatment resulted in alterations in the sleep-wake circadian rhythm, characterized by reduced wakefulness and an increase in non-rapid eye movement (NREM) sleep. Moreover, alterations in the peak expression of Per1, Per2, Clock, Cry1, Cry2, and Bmal1 mRNA were noted in the hippocampus of treated mice. Particularly noteworthy were the observed reductions in amyloid-beta (Aβ) deposition within the hippocampus, improvements in neuronal synaptic integrity, and upregulation of mTOR, Akt, and PI3K protein expression in the hippocampal region. These findings underscore the critical involvement of the PI3K/Akt/mTOR signaling pathway in mitigating disturbances in sleep-wake circadian rhythms.
    CONCLUSIONS: GBJNY enhanced the cognitive performance of APP/PS1 mice and altered clock gene expression patterns, alleviating sleep-wake circadian rhythm disruptions. The fundamental mechanism appears to be linked to the PI3K/Akt/mTOR pathway regulation, offering a foundation for potential clinical applications.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    阿尔茨海默病(AD)与β淀粉样蛋白(Aβ)的神经毒性作用密切相关,导致突触损伤,神经元丢失和认知功能障碍。先前的体外研究已经证明了corilagin抵抗Aβ诱导的氧化应激的潜力,炎性损伤,和Aβ生产中的β位点淀粉样前体蛋白裂解酶1(BACE1)活性。然而,Corilagin对阿尔茨海默病的体内保护作用仍未被研究。目的探讨corilagin对APP/PS1小鼠的保护作用及其机制。通过逐步被动回避和Morris水迷宫测试评估小鼠的认知功能。Nissl染色用于评估海马中的神经元损伤。使用ELISA和Western印迹分析来确定相关蛋白表达。透射电镜观察海马神经元的突触超微结构。高尔基染色用于评估海马锥体神经元的树突形态和树突棘密度。进行免疫组织化学和Western印迹以检查突触相关蛋白的表达。结果表明,corilagin可改善APP/PS1小鼠的学习记忆能力,减少海马神经元损伤,抑制BACE1并减少Aβ生成。它还改善突触可塑性和突触相关蛋白的表达。Corilagin通过抑制BACE1有效减少Aβ的产生,最终减少神经元的损失并增强突触可塑性以改善突触传递。本研究揭示了corilagin在阿尔茨海默病中的潜在治疗作用。
    Alzheimer\'s disease (AD) is closely associated with the neurotoxic effects of amyloid-β (Aβ), leading to synaptic damage, neuronal loss and cognitive dysfunction. Previous in vitro studies have demonstrated the potential of corilagin to counteract Aβ-induced oxidative stress, inflammatory injury, and β-site amyloid precursor protein cleaving enzyme-1 (BACE1) activity in Aβ production. However, the in vivo protective effects of corilagin on Alzheimer\'s disease remain unexplored. The purpose of this study was to investigate the protective effects of corilagin on APP/PS1 mice and the underlying mechanisms. The cognitive function of the mice was assessed by step-through passive avoidance and Morris water maze tests. Nissl staining was used to evaluate neuronal damage in the hippocampus. ELISA and Western blotting analyses were used to determine the associated protein expression. Transmission electron microscopy was utilized to observe the synaptic ultrastructure of hippocampal neurons. Golgi staining was applied to assess dendritic morphology and dendritic spine density in hippocampal pyramidal neurons. Immunohistochemistry and Western blotting were performed to examine the expression of synaptic-associated proteins. The results showed that corilagin improves learning and memory in APP/PS1 mice, reduces hippocampal neuron damage, inhibits BACE1 and reduces Aβ generation. It also improves synaptic plasticity and the expression of synaptic-associated proteins. Corilagin effectively reduces Aβ generation by inhibiting BACE1, ultimately reducing neuronal loss and enhancing synaptic plasticity to improve synaptic transmission. This study sheds light on the potential therapeutic role of corilagin in Alzheimer\'s disease.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    随着阿尔茨海默病(AD)发病机制的快速破译,人们普遍认为,大脑中错误折叠的淀粉样蛋白β(Aβ)的积累可能导致AD的神经变性。虽然很多证据证明了Aβ的神经毒性,Aβ在神经系统中的作用是复杂的。然而,需要更全面的研究来深入了解Aβ40单体的生理效应。探讨Aβ的生理机制,我们使用质谱法研究了由较低亚微摩尔浓度的Aβ诱导的改变的蛋白质组事件。将人神经母细胞瘤SH-SY5Y细胞暴露于五种不同浓度的Aβ1-40单体,并在四个时间点收集。蛋白质组学分析揭示了参与生物过程的蛋白质的时程行为,如RNA剪接,核运输和蛋白质定位。进一步的生物学研究表明,Aβ40单体可能激活PI3K/AKT信号来调节p-Tau,Ezrin和MAP2。这三种蛋白与树突形态发生有关,神经元极性,突触发生,轴突建立和轴突伸长。此外,Aβ40单体可以通过激活ERK1/2途径抑制BACE1和APP的表达来调节其生理形式。对Aβ病理生理机制的全面探索有利于探索新的治疗方法。
    With the rapid progress in deciphering the pathogenesis of Alzheimer\'s disease (AD), it has been widely accepted that the accumulation of misfolded amyloid β (Aβ) in the brain could cause the neurodegeneration in AD. Although much evidence demonstrates the neurotoxicity of Aβ, the role of Aβ in the nervous system are complex. However, more comprehensive studies are needed to understand the physiological effect of Aβ40 monomers in depth. To explore the physiological mechanism of Aβ, we employed mass spectrometry to investigate the altered proteomic events induced by a lower submicromolar concentration of Aβ. Human neuroblastoma SH-SY5Y cells were exposed to five different concentrations of Aβ1-40 monomers and collected at four time points. The proteomic analysis revealed the time-course behavior of proteins involved in biological processes, such as RNA splicing, nuclear transport and protein localization. Further biological studies indicated that Aβ40 monomers may activate PI3K/AKT signaling to regulate p-Tau, Ezrin and MAP2. These three proteins are associated with dendritic morphogenesis, neuronal polarity, synaptogenesis, axon establishment and axon elongation. Moreover, Aβ40 monomers may regulate their physiological forms by inhibiting the expression of BACE1 and APP via activation of the ERK1/2 pathway. A comprehensive exploration of pathological and physiological mechanisms of Aβ is beneficial for exploring novel treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    我们研究了黄芪灵(AST)治疗阿尔茨海默病(AD)的作用机制。采用网络药理学方法分析了AST与AST、AD,和神经炎症,在实验中使用患有AD的APP/PS1转基因小鼠;具体而言,通过Morris水迷宫和八臂放射状迷宫试验分析了AST对小鼠行为的影响,ELISA法检测组织炎症因子水平,H&E及免疫组化染色分析病理改变。网络药理学分析结果表明,AST对AD的神经炎症具有多靶点作用。通过分子对接和动力学分析,COX2可能是AST的靶标。此外,动物实验结果表明AST能改善AD小鼠的行为,增强了运动和记忆能力,同时,它抑制了组织中炎症因子的表达和小胶质细胞的活化。这项研究发现,AST可以通过COX2抑制小胶质细胞的活化,从而改善AD的神经炎症。
    We investigate the mechanism of action of astragalin (AST) in the treatment of Alzheimer\'s disease (AD). Network pharmacology was conducted to analyze the relationships among AST, AD, and neuroinflammation, The APP/PS1 transgenic mice with AD were used in the experiments; to be specific, the influence of AST on the behavior of mice was analyzed by Morris water maze and eight-arm radial maze tests, the tissue inflammatory factor levels were detected by ELISA, and pathological changes were analyzed by H&E and immunohistochemical staining. Analysis results of network pharmacology suggested that AST exerted the multi-target effect on neuroinflammation in AD. Through molecular docking and dynamics analyses, COX2 might be the target of AST. Moreover, animal experimental results demonstrated that AST improved the behavior of AD mice, and enhanced the motor and memory abilities, meanwhile, it suppressed the expression of inflammatory factors in tissues and the activation of microglial cells. this study discovers that AST can suppress microglial cell activation via COX2 to improve neuroinflammation in AD.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    阿尔茨海默病(AD)是一种退行性痴呆疾病,可导致大脑皮层的颞叶和额叶萎缩。灵贵竹干(LGZG),经典的中草药配方,最初被公认为是一种安全有效的治疗心血管疾病的方法。本研究旨在评估LGZG在AD进展中的作用及其分子机制。C57BL/6小鼠分为6组:正常小鼠,淀粉样前体蛋白/早老素1(APP/PS1)小鼠(模型组),阳性对照组(多奈哌齐治疗模型小鼠),高,中、低LGZG组(模型小鼠7g/kg/d,分别为3.5g/kg/d或1.75g/kg/dLGZG)。水迷宫结果显示,与模型小鼠相比,高、中LGZG组的逃避潜伏期和路径长度有所下降,下降程度呈剂量依赖性。采用Nissl染色对6组海马切片进行分析,Perls\'铁染色和免疫荧光测定。结果表明,LGZG可以恢复AD小鼠的形态学异常,减轻铁沉积。GXP4阳性细胞明显增加。MDA,通过生化试验测定Fe2+和GSH,其结果表明LGZG可以归一化MDA,与未处理的APP/PS1模型相比,AD模型中的Fe2+和GSH水平。小鼠接受的LGZG剂量越高,对这些分子水平的影响越强烈。Westernblot结果显示LGZG可以影响NeuN,AMPK,p53、SLC7A11和GPX4在AD模型海马中的水平,这是与AMPK途径相关的所有蛋白质。总之,LGZG通过AMPK通路缓解氧化应激和铁凋亡对AD具有神经保护作用。
    Alzheimer\'s disease (AD) is a degenerative dementia illness that causes atrophy of the temporal and frontal lobes of the cerebral cortex. Linggui Zhugan (LGZG), a classic Chinese herbal formula, was initially recognized as a safe and effective treatment of cardiovascular diseases for long history. This study intended to assess the effects and the molecular mechanism of LGZG on AD progress. C57BL/6 mice were divided into six groups: normal mice, amyloid precursor protein/presenilin 1 (APP/PS1) mice (model group), positive control group (model mice treated with donepezil), high, medium and low LGZG group (model mice treated with 7g/kg/d, 3.5g/kg/d or 1.75g/kg/d LGZG respectively). Water maze results showed that the escape latency and path length of high and medium LGZG groups declined compared to the model mice, the decline degree was dose-dependent. The hippocampal slices of six groups were analyzed by Nissl-staining, Perls\' iron staining and immunofluorescence assay. The results indicated LGZG could restore morphological anomalies and alleviate iron deposition of AD mice, and the GXP4 positive cells increased significantly. The MDA, Fe2+ and GSH were measured by biochemical testing, whose results illustrated that LGZG could normalize MDA, Fe2+ and GSH levels in AD model compared to un-treated APP/PS1 model. The higher dose of LGZG the mice received, the more intensive effects on those levels of molecules. Western blot results showed that LGZG could affect NeuN, AMPK, p53, SLC7A11 and GPX4 levels in the hippocampus of AD model, which was all proteins related to AMPK pathway. In conclusion, LGZG has a neuroprotective effect on AD through AMPK pathway by alleviating oxidative stress and ferroptosis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    背景:体育锻炼已被证明对阿尔茨海默病(AD)患者有益,尽管潜在的机制还没有完全理解。
    方法:将六个月大的淀粉样前体蛋白/早老素1(APP/PS1)转基因(Tg)小鼠和野生型(Wt)小鼠随机分为久坐组(Tg-Sed,Wt-Sed)或运动组(Tg-Ex,Wt-Ex)承担12周,中等强度跑步机运行程序。因此,测试了所有小鼠的记忆功能和淀粉样β(Aβ)水平,并检查了皮质和海马组织中tau和蛋白激酶B(Akt)/糖原合酶激酶3(GSK3)的磷酸化。
    结果:Tg-Sed小鼠记忆严重受损,Aβ水平较高,tau的磷酸化增加,GSK3α酪氨酸279和GSK3β酪氨酸216,但在两种组织中GSK3αserine21,GSK3βserine9和Aktserine473的磷酸化均低于各自组织中的Wt-Sed小鼠。Tg-Ex小鼠表现出记忆功能的显着改善以及较低水平的Aβ和较少的tau磷酸化(两种组织),GSK3α酪氨酸279(两种组织),和GSK3β酪氨酸216(仅海马),但是GSK3αserine21(两种组织)的磷酸化增加,GSK3βserine9(仅海马),和Aktserine473(两种组织)与相应组织中的Tg-Sed小鼠相比。
    结论:中等强度有氧运动对改善9月龄APP/PS1小鼠的记忆功能非常有效,最有可能是通过皮质和海马中GSK3α/β磷酸化的差异调节。
    BACKGROUND: Physical exercise has been shown to be beneficial for individuals with Alzheimer\'s disease (AD), although the underlying mechanisms are not fully understood.
    METHODS: Six-month-old Amyloid precursor protein/Presenilin 1 (APP/PS1) transgenic (Tg) mice and wild-type (Wt) mice were randomly assigned to either a sedentary group (Tg-Sed, Wt-Sed) or an exercise group (Tg-Ex, Wt-Ex) undertaking a 12-week, moderate-intensity treadmill running program. Consequently, all mice were tested for memory function and amyloid β (Aβ) levels and phosphorylation of tau and protein kinase B (Akt)/glycogen synthase kinase-3 (GSK3) were examined in tissues of both the cortex and hippocampus.
    RESULTS: Tg-Sed mice had severely impaired memory, higher levels of Aβ, and increased phosphorylation of tau, GSK3α tyrosine279, and GSK3β tyrosine216, but less phosphorylation of GSK3α serine21, GSK3β serine9, and Akt serine473 in both tissues than Wt-Sed mice in respective tissues. Tg-Ex mice showed significant improvement in memory function along with lower levels of Aβ and less phosphorylation of tau (both tissues), GSK3α tyrosine279 (both tissues), and GSK3β tyrosine216 (hippocampus only), but increased phosphorylation of GSK3α serine21 (both tissues), GSK3β serine9 (hippocampus only), and Akt serine473 (both tissues) compared with Tg-Sed mice in respective tissues.
    CONCLUSIONS: Moderate-intensity aerobic exercise is highly effective in improving memory function in 9-month-old APP/PS1 mice, most likely through differential modulation of GSK3α/β phosphorylation in the cortex and hippocampus.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    自噬是一种高度保守的分解代谢机制,通过它可以去除不必要或功能失调的细胞成分。自噬的失调与各种神经退行性疾病有关。包括阿尔茨海默病(AD)。了解影响自噬的分子机制/分子可能为开发针对AD和其他神经退行性疾病的治疗策略提供重要见解。吞并衔接子含有磷酸酪氨酸结合域的蛋白1(GULP1)是一种衔接子,可与淀粉样前体蛋白(APP)相互作用,以通过未确定的机制促进淀粉样β肽的产生。新的证据表明GULP1在自噬中起作用。这里,我们显示GULP1通过与自噬相关14(ATG14)的相互作用参与自噬,是自噬体形成的调节剂。GULP1通过调节III类磷脂酰肌醇3激酶复合物1(PI3KC3-C1)活性增强ATG14对自噬的刺激作用。GULP1的作用被破坏GULP1-ATG14相互作用的GULP1突变(GULP1m)减弱。相反,PI3KC3-C1活性在表达APP的细胞中增强,但在表达不结合GULP1的APP突变体的细胞中不增强,这表明GULP1-APP在调节PI3KC3-C1活性中的作用。值得注意的是,GULP1促进ATG14靶向内质网(ER)。此外,ATG14和APP的水平在表达GULP1的细胞的自噬液泡(AVs)中升高,但在表达GULP1m的细胞中不升高。APP加工在共表达GULP1和ATG14的细胞中显著增强。因此,GULP1通过促进APP进入AV来改变APP处理。总之,我们揭示了GULP1在增强ATG14靶向ER以刺激自噬和,因此,APP处理。
    Autophagy is a highly conserved catabolic mechanism by which unnecessary or dysfunctional cellular components are removed. The dysregulation of autophagy has been implicated in various neurodegenerative diseases, including Alzheimer\'s disease (AD). Understanding the molecular mechanism(s)/molecules that influence autophagy may provide important insights into developing therapeutic strategies against AD and other neurodegenerative disorders. Engulfment adaptor phosphotyrosine-binding domain-containing protein 1 (GULP1) is an adaptor that interacts with amyloid precursor protein (APP) to promote amyloid-β peptide production via an unidentified mechanism. Emerging evidence suggests that GULP1 has a role in autophagy. Here, we show that GULP1 is involved in autophagy through an interaction with autophagy-related 14 (ATG14), which is a regulator of autophagosome formation. GULP1 potentiated the stimulatory effect of ATG14 on autophagy by modulating class III phosphatidylinositol 3-kinase complex 1 (PI3KC3-C1) activity. The effect of GULP1 is attenuated by a GULP1 mutation (GULP1m) that disrupts the GULP1-ATG14 interaction. Conversely, PI3KC3-C1 activity is enhanced in cells expressing APP but not in those expressing an APP mutant that does not bind GULP1, which suggests a role of GULP1-APP in regulating PI3KC3-C1 activity. Notably, GULP1 facilitates the targeting of ATG14 to the endoplasmic reticulum (ER). Moreover, the levels of both ATG14 and APP are elevated in the autophagic vacuoles (AVs) of cells expressing GULP1, but not in those expressing GULP1m. APP processing is markedly enhanced in cells co-expressing GULP1 and ATG14. Hence, GULP1 alters APP processing by promoting the entry of APP into AVs. In summary, we unveil a novel role of GULP1 in enhancing the targeting of ATG14 to the ER to stimulate autophagy and, consequently, APP processing.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    目的:探讨阿尔茨海默病(AD)的发病机制,通过网络药理学研究人参皂苷Rg1抗AD的潜在作用靶点和信号通路。PharmMapper,和Uniprot数据库,而GeneCards数据库用于检查淀粉样前体蛋白(APP)和AD的各自靶标。然后,通过Venny工具探索了人参皂苷Rg1和APP之间的共同靶标,活动组件和目标之间的交互网络图是通过Cytoscape软件构建的,以及GO富集和KEGG途径注释分析。此外,GeneCards和FerrDb数据库发现了与铁死亡相关的基因。此外,人参皂苷Rg1、APP、铁性凋亡,并对AD进行了预测和分析。最后,通过免疫组织化学方法评价人参皂苷Rg1和liproxstrain-1对APP/PS1小鼠增殖和分化的影响。结果:人参皂苷Rg1、APP、铁性凋亡,AD有12个hub基因。GO富集和KEGG通路剖析注解,EGFR、SRC,蛋白质水解,蛋白质磷酸化,松弛素途径,FoxO信号通路通过调节APP相关信号通路,在人参皂苷Rg1调控铁凋亡抗AD的潜在机制中起重要作用。APP/PS1小鼠实验证实人参皂苷Rg1和liproxstrain-1可以促进细胞增殖和分化。结论:人参皂苷Rg1、APP和铁细胞凋亡可能作用于EGFR。SRC,松弛素和FoxO信号通路调节蛋白质代谢,蛋白磷酸化等途径改善AD症状。
    OBJECTIVE: To explore the pathogenesis of Alzheimer\'s disease (AD), the potential targets and signaling pathways of ginsenoside Rg1 against AD were investigated by network pharmacology.
    METHODS: Ginsenoside Rg1 targets were identified through PubChem, PharmMapper, and Uniprot databases, while the GeneCards database was used to examine the respective targets of amyloid precursor protein (APP) and AD. Then, the common targets between ginsenoside Rg1 and APP were explored by the Venny tool, the interaction network diagram between the active components and the targets was built via Cytoscape software, as well as GO enrichment and KEGG pathway annotation analysis were performed. Furthermore, genes associated with ferroptosis were found by the GeneCards and FerrDb databases. Besides, the connection among ginsenoside Rg1, APP, ferroptosis, and AD was predicted and analyzed. Finally, the effects of ginsenosides Rg1 and liproxstain-1 on the proliferation and differentiation of APP/PS1 mice were evaluated by immunohistochemistry.
    RESULTS: Ginsenoside Rg1, APP, ferroptosis, and AD had 12 hub genes. GO enrichment and KEGG pathway annotation analysis showed that EGFR, SRC, protein hydrolysis, protein phosphorylation, the Relaxin pathway, and the FoxO signaling pathway play an important role in the potential mechanism of ginsenoside Rg1\'s under regulation of ferroptosis anti-AD through the modulation of APP-related signaling pathways. The APP/PS1 mice experiment verified that ginsenosides Rg1 and liproxstain-1 can promote the proliferation and differentiation.
    CONCLUSIONS: Ginsenoside Rg1, APP and ferroptosis may act on EGFR, SRC, the Relaxin and FoxO signaling pathways to regulate protein metabolism, protein phosphorylation and other pathways to improve AD symptoms.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号