tauopathy

Tau 病
  • 文章类型: Journal Article
    背景:脱氧核糖核酸酶2(DNaseII)在清除细胞质双链DNA(dsDNA)中起关键作用。DNA酶II的缺乏导致DNA在细胞质中的积累。神经元中的持续dsDNA是衰老和神经退行性疾病(包括阿尔茨海默病(AD))的早期病理标志。然而,目前尚不清楚DNaseII和神经元细胞质dsDNA如何影响神经发病机制。Tau过度磷酸化是AD发病的关键因素。DNaseII和神经元细胞质dsDNA对神经元tau过度磷酸化的影响仍未阐明。
    方法:用免疫组织化学和免疫标记法检测不同年龄WT和Tau-P301S小鼠的神经元DNaseII和dsDNA水平,用ELISA法测定AD患者血浆中DNaseⅡ的水平。为了研究DNaseII对tau蛋白病变的影响,磷酸化tau的水平,磷酸激酶,磷酸酶突触蛋白,神经元DNaseII缺陷型WT小鼠脑中的神经胶质增生和促炎细胞因子,通过免疫标记评估神经元DNaseII缺陷型Tau-P301S小鼠和神经元DNaseII过表达的Tau-P301S小鼠,免疫印迹或ELISA。使用莫里斯水迷宫测试确定认知表现,Y-迷宫测试,新颖的物体识别测试和开放现场测试。
    结果:AD患者的大脑和血浆中DNaseII的水平显着降低。DNaseII还在WT和Tau-P301S小鼠的神经元中年龄依赖性地降低,随着dsDNA在细胞质中积累的增加。神经元DNA酶II缺乏诱导的DNA积累通过上调细胞周期蛋白依赖性样激酶5(CDK5)和钙/钙调蛋白激活的蛋白激酶II(CaMKII)和下调磷酸酶蛋白磷酸酶2A(PP2A)来驱动tau磷酸化。此外,DNaseII敲低诱导并显著加剧神经元丢失,WT和Tau-P301S小鼠的神经炎症和认知缺陷,分别,而神经元DNaseII的过表达显示出治疗益处。
    结论:DNaseII缺乏和细胞质dsDNA积累可以启动tau磷酸化,提示DNaseII是tau相关疾病的潜在治疗靶点。
    BACKGROUND: Deoxyribonuclease 2 (DNase II) plays a key role in clearing cytoplasmic double-stranded DNA (dsDNA). Deficiency of DNase II leads to DNA accumulation in the cytoplasm. Persistent dsDNA in neurons is an early pathological hallmark of senescence and neurodegenerative diseases including Alzheimer\'s disease (AD). However, it is not clear how DNase II and neuronal cytoplasmic dsDNA influence neuropathogenesis. Tau hyperphosphorylation is a key factor for the pathogenesis of AD. The effect of DNase II and neuronal cytoplasmic dsDNA on neuronal tau hyperphosphorylation remains unclarified.
    METHODS: The levels of neuronal DNase II and dsDNA in WT and Tau-P301S mice of different ages were measured by immunohistochemistry and immunolabeling, and the levels of DNase II in the plasma of AD patients were measured by ELISA. To investigate the impact of DNase II on tauopathy, the levels of phosphorylated tau, phosphokinase, phosphatase, synaptic proteins, gliosis and proinflammatory cytokines in the brains of neuronal DNase II-deficient WT mice, neuronal DNase II-deficient Tau-P301S mice and neuronal DNase II-overexpressing Tau-P301S mice were evaluated by immunolabeling, immunoblotting or ELISA. Cognitive performance was determined using the Morris water maze test, Y-maze test, novel object recognition test and open field test.
    RESULTS: The levels of DNase II were significantly decreased in the brains and the plasma of AD patients. DNase II also decreased age-dependently in the neurons of WT and Tau-P301S mice, along with increased dsDNA accumulation in the cytoplasm. The DNA accumulation induced by neuronal DNase II deficiency drove tau phosphorylation by upregulating cyclin-dependent-like kinase-5 (CDK5) and calcium/calmodulin activated protein kinase II (CaMKII) and downregulating phosphatase protein phosphatase 2A (PP2A). Moreover, DNase II knockdown induced and significantly exacerbated neuron loss, neuroinflammation and cognitive deficits in WT and Tau-P301S mice, respectively, while overexpression of neuronal DNase II exhibited therapeutic benefits.
    CONCLUSIONS: DNase II deficiency and cytoplasmic dsDNA accumulation can initiate tau phosphorylation, suggesting DNase II as a potential therapeutic target for tau-associated disorders.
    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
    过度磷酸化tau蛋白的异常积累在一系列称为tau蛋白病的神经退行性疾病中起着关键作用,包括阿尔茨海默病(AD)。我们最近概念化了异源双功能嵌合体的设计,用于选择性地促进tau和磷酸酶之间的接近,因此特别促进tau去磷酸化和去除。这里,我们试图优化tau去磷酸化靶向嵌合体(DEPTAC)的构建,并获得了一个新的嵌合体D14,该嵌合体在细胞和tau病小鼠模型中都能高效降低tau磷酸化,同时显示有限的细胞毒性。此外,D14改善了用毒性tau-K18片段处理的原代培养海马神经元的神经变性,并改善tau蛋白病小鼠的认知功能。这些结果表明D14是治疗tau蛋白病的经济有效的候选药物。
    Abnormal accumulation of hyperphosphorylated tau protein plays a pivotal role in a collection of neurodegenerative diseases named tauopathies, including Alzheimer\'s disease (AD). We have recently conceptualized the design of hetero-bifunctional chimeras for selectively promoting the proximity between tau and phosphatase, thus specifically facilitating tau dephosphorylation and removal. Here, we sought to optimize the construction of tau dephosphorylating-targeting chimera (DEPTAC) and obtained a new chimera D14, which had high efficiency in reducing tau phosphorylation both in cell and tauopathy mouse models, while showing limited cytotoxicity. Moreover, D14 ameliorated neurodegeneration in primary cultured hippocampal neurons treated with toxic tau-K18 fragments, and improved cognitive functions of tauopathy mice. These results suggested D14 as a cost-effective drug candidate for the treatment of tauopathies.
    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
    淀粉样蛋白-β(Aβ)容易错误折叠成神经毒性聚集体,产生高水平的活性氧(ROS),导致进行性氧化损伤并最终导致细胞死亡。因此,同时抑制Aβ聚集和清除ROS可能是缓解阿尔茨海默病病理的一种有希望的治疗策略。基于先前开发的靶向所有形式的Aβ42的抗体1F12,我们以可生物降解的介孔二氧化硅纳米粒子为载体,负载超小氧化铈纳米晶体(bMSNs@Ce-1F12),开发了Aβ42和ROS双靶向纳米复合材料。通过修饰脑靶向狂犬病病毒糖蛋白29(RVG29-bMSNs@Ce-1F12),这种智能纳米复合材料可以有效地瞄准大脑富含Aβ的区域。结合外周和中枢神经系统治疗,RVG29-bMSNs@Ce-1F12可通过抑制Aβ42的错误折叠而显著缓解AD症状,加速Aβ42清除,清除ROS。此外,这种Aβ42和ROS双靶向策略表现出的ROS清除和Aβ清除的协同作用也减少了过度磷酸化tau的负担,缓解神经胶质细胞活化,并最终改善APP/PS1小鼠的认知功能。我们的发现表明RVG29-bMSNs@Ce-1F12是一种有前途的纳米药物,可以促进AD的多靶点治疗。
    Amyloid-β (Aβ) readily misfolds into neurotoxic aggregates, generating high levels of reactive oxygen species (ROS), leading to progressive oxidative damage and ultimately cell death. Therefore, simultaneous inhibition of Aβ aggregation and scavenging of ROS may be a promising therapeutic strategy to alleviate Alzheimer\'s disease pathology. Based on the previously developed antibody 1F12 that targets all forms of Aβ42, we developed an Aβ42 and ROS dual-targeting nanocomposite using biodegradable mesoporous silica nanoparticles as carriers to load ultra-small cerium oxide nanocrystals (bMSNs@Ce-1F12). By modifying the brain-targeted rabies virus glycoprotein 29 (RVG29-bMSNs@Ce-1F12), this intelligent nanocomposite can efficiently target brain Aβ-rich regions. Combined with peripheral and central nervous system treatments, RVG29-bMSNs@Ce-1F12 can significantly alleviate AD symptoms by inhibiting Aβ42 misfolding, accelerating Aβ42 clearance, and scavenging ROS. Furthermore, this synergistic effect of ROS scavenging and Aβ clearance exhibited by this Aβ42 and ROS dual-targeted strategy also reduced the burden of hyperphosphorylated tau, alleviated glial cell activation, and ultimately improved cognitive function in APP/PS1 mice. Our findings indicate that RVG29-bMSNs@Ce-1F12 is a promising nanodrug that can facilitate multi-target treatment of 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
    蛋白质病,定义为最终导致细胞死亡的蛋白质的异常积累,是神经退行性疾病最显著的病理特点之一。Tau病,以阿尔茨海默病(AD)为代表,和突触核蛋白病,以帕金森病(PD)为代表,在多个方面表现出相似性。AD表现出锥体外系症状,而痴呆也是晚期PD的主要标志。我们和其他研究人员已经依次显示了α-突触核蛋白(α-syn)和tau的交叉播种现象,突触核蛋白病和tau蛋白病之间的强化病理。高度重叠的临床和病理特征暗示两组疾病之间共有的致病机制。似乎适合于一种不同的神经退行性疾病的诊断和治疗策略也可以应用于更广泛的范围。因此,清楚了解tau蛋白病和突触核蛋白病之间的重叠和差异对于揭示神经退行性疾病之间复杂关联的性质至关重要.在这次审查中,我们从遗传原因方面讨论Tau蛋白病和突触核蛋白病的共同和多样化特征,临床表现,病理进展和针对病理的潜在常见治疗方法,目的是为制定疾病分类方案提供及时的更新,并为神经退行性疾病的治疗发展提供新的见解。
    Proteinopathy, defined as the abnormal accumulation of proteins that eventually leads to cell death, is one of the most significant pathological features of neurodegenerative diseases. Tauopathies, represented by Alzheimer\'s disease (AD), and synucleinopathies, represented by Parkinson\'s disease (PD), show similarities in multiple aspects. AD manifests extrapyramidal symptoms while dementia is also a major sign of advanced PD. We and other researchers have sequentially shown the cross-seeding phenomenon of α-synuclein (α-syn) and tau, reinforcing pathologies between synucleinopathies and tauopathies. The highly overlapping clinical and pathological features imply shared pathogenic mechanisms between the two groups of disease. The diagnostic and therapeutic strategies seemingly appropriate for one distinct neurodegenerative disease may also apply to a broader spectrum. Therefore, a clear understanding of the overlaps and divergences between tauopathy and synucleinopathy is critical for unraveling the nature of the complicated associations among neurodegenerative diseases. In this review, we discuss the shared and diverse characteristics of tauopathies and synucleinopathies from aspects of genetic causes, clinical manifestations, pathological progression and potential common therapeutic approaches targeting the pathology, in the aim to provide a timely update for setting the scheme of disease classification and provide novel insights into the therapeutic development for neurodegenerative diseases.
    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
    神经元丢失是阿尔茨海默病(AD)的中心问题,然而,迄今为止尚未开发出可以阻止AD相关神经变性的治疗方法。这里,我们开发了针对217位点磷酸化的人tau(p-tau217)的单克隆抗体(mAb2A7),并观察到p-tau217水平与AD患者的脑萎缩和认知障碍呈正相关.鼻内给药有效地将mAb2A7递送到雄性PS19tau病鼠脑中,具有靶接合和减少tau病理/聚集,而对总可溶性tau几乎没有影响。Further,mAb2A7治疗阻断了凋亡相关的神经元丢失和脑萎缩,逆转的认知缺陷,并改善雄性牛磺酸病鼠的运动功能。蛋白质组学分析显示,mAb2A7治疗逆转了主要与鼠tau蛋白病和AD脑中观察到的突触功能相关的蛋白质的改变。靶向总tau的抗体(13G4)也减弱了tau相关的病理学和神经变性,但损害了雄性tau病鼠的运动功能。这些结果暗示p-tau217是AD相关神经变性的潜在治疗靶标。
    Neuronal loss is the central issue in Alzheimer\'s disease (AD), yet no treatment developed so far can halt AD-associated neurodegeneration. Here, we developed a monoclonal antibody (mAb2A7) against 217 site-phosphorylated human tau (p-tau217) and observed that p-tau217 levels positively correlated with brain atrophy and cognitive impairment in AD patients. Intranasal administration efficiently delivered mAb2A7 into male PS19 tauopathic mouse brain with target engagement and reduced tau pathology/aggregation with little effect on total soluble tau. Further, mAb2A7 treatment blocked apoptosis-associated neuronal loss and brain atrophy, reversed cognitive deficits, and improved motor function in male tauopathic mice. Proteomic analysis revealed that mAb2A7 treatment reversed alterations mainly in proteins associated with synaptic functions observed in murine tauopathy and AD brain. An antibody (13G4) targeting total tau also attenuated tau-associated pathology and neurodegeneration but impaired the motor function of male tauopathic mice. These results implicate p-tau217 as a potential therapeutic target for AD-associated neurodegeneration.
    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
    微管相关Tau蛋白的病理过度磷酸化和聚集导致阿尔茨海默病(AD)和其他相关tau蛋白病变。目前,阿尔茨海默病没有治愈方法。适体作为生物技术和神经障碍治疗的下一代治疗剂提供了巨大的潜力。Tau蛋白的传统适体选择方法侧重于结合亲和力,而不是干扰病理性Tau。在这项研究中,我们开发了一种新的选择策略来富集在通常会促进Tau聚集的条件下与存活的单体Tau蛋白结合的DNA适体。采用这种方法,我们确定了一组适体候选物。值得注意的是,BW1c表现出对Tau蛋白的高结合亲和力(Kd=6.6nM),并有效抑制花生四烯酸(AA)诱导的Tau蛋白寡聚化和聚集。此外,它抑制无细胞系统中GSK3β介导的Tau过度磷酸化和细胞环境中冈田酸介导的Tau过度磷酸化。最后,眼眶后注射BW1ctau适体显示了穿过血脑屏障并进入神经元细胞体的能力。通过进一步的完善和发展,这些Tau适体可能为一流的神经治疗方法铺平道路,以减轻tau蛋白病相关的神经退行性疾病.
    Pathological hyperphosphorylation and aggregation of microtubule-associated Tau protein contribute to Alzheimer\'s Disease (AD) and other related tauopathies. Currently, no cure exists for Alzheimer\'s Disease. Aptamers offer significant potential as next-generation therapeutics in biotechnology and the treatment of neurological disorders. Traditional aptamer selection methods for Tau protein focus on binding affinity rather than interference with pathological Tau. In this study, we developed a new selection strategy to enrich DNA aptamers that bind to surviving monomeric Tau protein under conditions that would typically promote Tau aggregation. Employing this approach, we identified a set of aptamer candidates. Notably, BW1c demonstrates a high binding affinity (Kd=6.6 nM) to Tau protein and effectively inhibits arachidonic acid (AA)-induced Tau protein oligomerization and aggregation. Additionally, it inhibits GSK3β-mediated Tau hyperphosphorylation in cell-free systems and okadaic acid-mediated Tau hyperphosphorylation in cellular milieu. Lastly, retro-orbital injection of BW1c tau aptamer shows the ability to cross the blood brain barrier and gain access to neuronal cell body. Through further refinement and development, these Tau aptamers may pave the way for a first-in-class neurotherapeutic to mitigate tauopathy-associated neurodegenerative disorders.
    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
    过度磷酸化tau蛋白聚集体的积累是阿尔茨海默病(AD)的关键致病事件,并诱导线粒体功能障碍和活性氧(ROS)过度产生。然而,由于血脑屏障(BBB)和AD的复杂病理造成的障碍,AD的治疗仍然具有挑战性。经鼻递送代表了规避BBB和将药物递送至脑的有效手段。在这项研究中,黑磷(BP)被用作药物载体和抗氧化剂,并负载有tau聚集抑制剂,亚甲蓝(MB),获得BP-MB。对于鼻内递送,我们通过交联羧甲基壳聚糖(CMCS)和醛PluronicF127(F127-CHO)胶束制备了热敏水凝胶。将BP-MB纳米复合材料掺入水凝胶中以获得BP-MB@凝胶。BP-MB@Gel可以鼻内注射,提供高的鼻粘膜保留和控制药物释放。鼻内给药后,BP-MB不断释放并传递到大脑,通过抑制tau神经病理学发挥协同治疗作用,恢复线粒体功能,缓解神经炎症,从而诱导AD小鼠模型的认知改善。这些发现突出了脑靶向药物递送在AD复杂病理管理中的潜在策略。本文受版权保护。保留所有权利。
    The accumulation of hyperphosphorylated tau protein aggregates is a key pathogenic event in Alzheimer\'s disease (AD) and induces mitochondrial dysfunction and reactive oxygen species overproduction. However, the treatment of AD remains challenging owning to the hindrance caused by the blood-brain barrier (BBB) and the complex pathology of AD. Nasal delivery represents an effective means of circumventing the BBB and delivering drugs to the brain. In this study, black phosphorus (BP) is used as a drug carrier, as well as an antioxidant, and loaded with a tau aggregation inhibitor, methylene blue (MB), to obtain BP-MB. For intranasal (IN) delivery, a thermosensitive hydrogel is fabricated by cross-linking carboxymethyl chitosan and aldehyde Pluronic F127 (F127-CHO) micelles. The BP-MB nanocomposite is incorporated into the hydrogel to obtain BP-MB@Gel. BP-MB@Gel could be injected intranasally, providing high nasal mucosal retention and controlled drug release. After IN administration, BP-MB is continuously released and delivered to the brain, exerting synergistic therapeutic effects by suppressing tau neuropathology, restoring mitochondrial function, and alleviating neuroinflammation, thus inducing cognitive improvements in mouse models of AD. These findings highlight a potential strategy for brain-targeted drug delivery in the management of the complex pathologies of AD.
    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
    tau蛋白的异常过度磷酸化和积累在阿尔茨海默病(AD)和许多其他tau蛋白病变的神经变性中起关键作用。选择性消除过度磷酸化的tau有望用于治疗这些疾病。我们已经概念化了一个战略,命名为去磷酸化靶向嵌合体(DEPTACs),专门劫持磷酸酶到tau以削弱其过度磷酸化。这里,我们对每个组成基序进行了逐步优化,以生成具有促进去磷酸化和随后清除病理性tau的合理有效性的DEPTACs.具体来说,对于其中一个选定的嵌合体,D16,我们证明了其在体外挽救由神经毒性K18-tau种子引起的神经变性的显着效率。此外,D16的静脉内给药还减轻了AD小鼠脑中的tau病理并改善了记忆缺陷。这些结果表明DEPTAC作为tau磷酸化的靶向调节剂,对AD和其他tau蛋白病具有治疗潜力。
    Abnormal hyperphosphorylation and accumulation of tau protein play a pivotal role in neurodegeneration in Alzheimer\'s disease (AD) and many other tauopathies. Selective elimination of hyperphosphorylated tau is promising for the therapy of these diseases. We have conceptualized a strategy, named dephosphorylation-targeting chimeras (DEPTACs), for specifically hijacking phosphatases to tau to debilitate its hyperphosphorylation. Here, we conducted the step-by-step optimization of each constituent motif to generate DEPTACs with reasonable effectiveness in facilitating the dephosphorylation and subsequent clearance of pathological tau. Specifically, for one of the selected chimeras, D16, we demonstrated its significant efficiency in rescuing the neurodegeneration caused by neurotoxic K18-tau seeds in vitro. Moreover, intravenous administration of D16 also alleviated tau pathologies in the brain and improved memory deficits in AD mice. These results suggested DEPTACs as targeted modulators of tau phosphorylation, which hold therapeutic potential for AD and other tauopathies.
    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:阿尔茨海默病(AD)tau蛋白病的“pr病毒样”特征及其与淀粉样蛋白β(Aβ)的关系从未在灵长类动物的系统发育上接近人类的实验研究。
    方法:我们在内嗅皮层中注射了17只猕猴,其中纳克是从AD大脑中纯化的有种子能力的tau聚集体或从老年匹配的健康大脑中提取的对照提取物,有或没有侧脑室共注射寡聚Aβ。
    结果:病理性tau注射液在18个月后增加脑脊液(CSF)p-tau181浓度。Tau病理从内嗅皮层扩散到海马三突触环和扣带皮层,恢复Braak阶段I至IV的实验进展。许多AD相关的分子网络受到tau种子注射的影响,而与蛋白质组学分析中的Aβ注射无关。然而,我们发现了成熟的神经原纤维缠结,CSF总tau浓度增加,以及仅在Aβ共同注射的猕猴中的突触前和突触后变性。
    结论:低聚-Aβ介导了tau病理的成熟及其在猕猴中的神经元毒性,但不是其最初的传播。
    结论:这项研究支持从AD大脑中提取的错误折叠tau的“朊病毒样”特性。这项研究从经验上验证了拟人化大脑中的Braak分期。这项研究强调了寡聚Aβ在驱动tau病理学成熟和毒性中的作用。这项工作建立了更接近人类病理学的早期散发性AD的新型动物模型。
    The \"prion-like\" features of Alzheimer\'s disease (AD) tauopathy and its relationship with amyloid-β (Aβ) have never been experimentally studied in primates phylogenetically close to humans.
    We injected 17 macaques in the entorhinal cortex with nanograms of seeding-competent tau aggregates purified from AD brains or control extracts from aged-matched healthy brains, with or without intracerebroventricular co-injections of oligomeric-Aβ.
    Pathological tau injection increased cerebrospinal fluid (CSF) p-tau181 concentration after 18 months. Tau pathology spreads from the entorhinal cortex to the hippocampal trisynaptic loop and the cingulate cortex, resuming the experimental progression of Braak stage I to IV. Many AD-related molecular networks were impacted by tau seeds injections regardless of Aβ injections in proteomic analyses. However, we found mature neurofibrillary tangles, increased CSF total-tau concentration, and pre- and postsynaptic degeneration only in Aβ co-injected macaques.
    Oligomeric-Aβ mediates the maturation of tau pathology and its neuronal toxicity in macaques but not its initial spreading.
    This study supports the \"prion-like\" properties of misfolded tau extracted from AD brains. This study empirically validates the Braak staging in an anthropomorphic brain. This study highlights the role of oligomeric Aβ in driving the maturation and toxicity of tau pathology. This work establishes a novel animal model of early sporadic AD that is closer to the human pathology.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Case Reports
    暂无摘要。
    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