Amyloid precursor protein

淀粉样前体蛋白
  • 文章类型: Journal Article
    跨膜蛋白β-淀粉样前体蛋白(APP)在阿尔茨海默病(AD)的病理生理学中起着重要作用。β-淀粉样蛋白假说认为APP的异常加工会形成神经毒性的β-淀粉样蛋白聚集体,这导致在AD中观察到的认知障碍。虽然许多其他因素有助于AD,有必要更好地了解APP的突触功能。我们发现果蝇APP样(APPL)在与Kismet(Kis)的突触中具有共享和非共享的角色,染色质解旋酶结合域(CHD)蛋白。Kis是CHD7和CHD8的同源物,两者都涉及神经发育障碍,包括CHARGE综合征和自闭症谱系障碍,分别。在其中枢神经系统中表达人APP和BACE的kis和动物中功能突变的丧失显示谷氨酸受体亚基的减少,GluRIIC,GTP酶Rab11和骨形态发生蛋白(BMP),pMad,在果蝇幼虫神经肌肉接头(NMJ)。同样,像内吞这样的过程,幼虫运动,这些动物的神经传递是有缺陷的。我们的药理学和上位性实验表明,Kis和APPL之间存在功能关系,但是Kis不调节幼虫NMJ的appl表达。相反,它可能影响APPL的突触定位,可能是通过促进rab11转录。这些数据确定了AD中染色质重塑蛋白与异常突触功能之间的潜在机制联系。
    The transmembrane protein β-amyloid precursor protein (APP) is central to the pathophysiology of Alzheimer\'s disease (AD). The β-amyloid hypothesis posits that aberrant processing of APP forms neurotoxic β-amyloid aggregates, which lead to the cognitive impairments observed in AD. Although numerous additional factors contribute to AD, there is a need to better understand the synaptic function of APP. We have found that Drosophila APP-like (APPL) has both shared and non-shared roles at the synapse with Kismet (Kis), a chromatin helicase binding domain (CHD) protein. Kis is the homolog of CHD7 and CHD8, both of which are implicated in neurodevelopmental disorders including CHARGE Syndrome and autism spectrum disorders, respectively. Loss of function mutations in kis and animals expressing human APP and BACE in their central nervous system show reductions in the glutamate receptor subunit, GluRIIC, the GTPase Rab11, and the bone morphogenetic protein (BMP), pMad, at the Drosophila larval neuromuscular junction (NMJ). Similarly, processes like endocytosis, larval locomotion, and neurotransmission are deficient in these animals. Our pharmacological and epistasis experiments indicate that there is a functional relationship between Kis and APPL, but Kis does not regulate appl expression at the larval NMJ. Instead, Kis likely influences the synaptic localization of APPL, possibly by promoting rab11 transcription. These data identify a potential mechanistic connection between chromatin remodeling proteins and aberrant synaptic function 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
    铅(Pb)的暴露是一个主要的公共卫生问题,可能通过污染的土壤发生,空气,食物,或者水,无论是在日常生活中,或在危险职业中工作。尽管铅长期以来被认为是儿童的神经发育毒物,最近越来越多的流行病学研究表明,低水平铅暴露可能导致成人年龄相关的神经功能障碍。成年期环境铅暴露与晚发性阿尔茨海默病(AD)和痴呆的风险有关。
    尽管这种联系背后的生物学机制尚不清楚,已经提出铅暴露可能通过改变AD相关基因的表达而增加AD的风险,可能,通过激活AD相关病理的分子途径。
    我们使用一系列转基因小鼠研究了铅暴露,这些小鼠在淀粉样蛋白前体蛋白和早老素1(APPΔNL/ΔNLxPS1P264L/P264L)中引起AD敲入突变,与Leprdb/db小鼠杂交以赋予血管病理学易感性。
    我们的数据表明,尽管成年小鼠的铅暴露会损害认知功能,这种作用与淀粉样蛋白病理学的增加或常见AD相关基因表达的变化无关.铅暴露也导致血压显著升高,众所周知的铅的作用。有趣的是,尽管血压升高与基因型无关,只有携带AD相关突变的小鼠出现认知功能障碍,尽管脑血管病理学没有明显变化。
    这些结果提出了一种可能性,即成人与铅暴露相关的痴呆风险增加可能与其随后与预先存在或发展中的AD相关神经病理学的相互作用有关。
    UNASSIGNED: Exposure to lead (Pb) is a major public health problem that could occur through contaminated soil, air, food, or water, either during the course of everyday life, or while working in hazardous occupations. Although Pb has long been known as a neurodevelopmental toxicant in children, a recent and growing body of epidemiological research indicates that cumulative, low-level Pb exposure likely drives age-related neurologic dysfunction in adults. Environmental Pb exposure in adulthood has been linked to risk of late-onset Alzheimer\'s disease (AD) and dementia.
    UNASSIGNED: Although the biological mechanism underlying this link is unknown, it has been proposed that Pb exposure may increase the risk of AD via altering the expression of AD-related genes and, possibly, by activating the molecular pathways underlying AD-related pathology.
    UNASSIGNED: We investigated Pb exposure using a line of genetically modified mice with AD-causing knock-in mutations in the amyloid precursor protein and presenilin 1 (APPΔNL/ΔNL x PS1P264L/P264L) that had been crossed with Leprdb/db mice to impart vulnerability to vascular pathology.
    UNASSIGNED: Our data show that although Pb exposure in adult mice impairs cognitive function, this effect is not related to either an increase in amyloid pathology or to changes in the expression of common AD-related genes. Pb exposure also caused a significant increase in blood pressure, a well known effect of Pb. Interestingly, although the increase in blood pressure was unrelated to genotype, only mice that carried AD-related mutations developed cognitive dysfunction, in spite of showing no significant change in cerebrovascular pathology.
    UNASSIGNED: These results raise the possibility that the increased risk of dementia associated with Pb exposure in adults may be tied to its subsequent interaction with either pre-existing or developing AD-related neuropathology.
    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

    求助全文

  • 文章类型: Journal Article
    背景:Nav1.6与阿尔茨海默病(AD)的病理密切相关,和星形胶质细胞最近被确定为β-淀粉样蛋白(Aβ)的重要来源。然而,关于Nav1.6和星形胶质细胞衍生的Aβ之间的联系知之甚少。
    目的:本研究探讨Nav1.6在介导星形胶质细胞源性Aβ在AD中的关键作用,敲低星形胶质细胞Nav1.6通过促进自噬和溶酶体-APP融合减轻AD进展。
    方法:构建星形细胞Nav1.6敲低的小鼠模型,以研究星形细胞Nav1.6对淀粉样变性的影响。利用透射电镜观察星形细胞Nav1.6在自噬和溶酶体-APP(淀粉样前体蛋白)融合中的作用,免疫染色,Westernblot和膜片钳。使用免疫染色检测胶质细胞活化。使用膜片钳评估神经可塑性和神经网络,高尔基染色和脑电图记录。进行行为实验以评估认知缺陷。
    结果:星形细胞Nav1.6基因敲除可减少淀粉样变性,减轻神经胶质细胞活化和形态复杂性,改善神经可塑性和异常神经网络,以及促进APP/PS1小鼠的学习和记忆能力。星形细胞Nav1.6敲低通过促进溶酶体-APP融合减少自身衍生的Aβ,这与减弱反向Na+-Ca2+交换电流从而减少细胞内Ca2+以促进通过AKT/mTOR/ULK途径的自噬有关。
    结论:我们的发现揭示了星形胶质细胞特异性Nav1.6在减少星形胶质细胞衍生的Aβ中的关键作用,强调其作为调节AD进展的细胞特异性靶标的潜力。
    BACKGROUND: Nav1.6 is closely related to the pathology of Alzheimer\'s Disease (AD), and astrocytes have recently been identified as a significant source of β-amyloid (Aβ). However, little is known about the connection between Nav1.6 and astrocyte-derived Aβ.
    OBJECTIVE: This study explored the crucial role of Nav1.6 in mediated astrocyte-derived Aβ in AD and knockdown astrocytic Nav1.6 alleviates AD progression by promoting autophagy and lysosome-APP fusion.
    METHODS: A mouse model for astrocytic Nav1.6 knockdown was constructed to study the effects of astrocytic Nav1.6 on amyloidosis. The role of astrocytic Nav1.6 on autophagy and lysosome-APP(amyloid precursor protein) fusion was used by transmission electron microscope, immunostaining, western blot and patch clamp. Glial cell activation was detected using immunostaining. Neuroplasticity and neural network were assessed using patch-clamp, Golgi stain and EEG recording. Behavioral experiments were performed to evaluate cognitive defects.
    RESULTS: Astrocytic Nav1.6 knockdown reduces amyloidosis, alleviates glial cell activation and morphological complexity, improves neuroplasticity and abnormal neural networks, as well as promotes learning and memory abilities in APP/PS1 mice. Astrocytic Nav1.6 knockdown reduces itself-derived Aβ by promoting lysosome- APP fusion, which is related to attenuating reverse Na+-Ca2+ exchange current thus reducing intracellular Ca2+ to facilitate autophagic through AKT/mTOR/ULK pathway.
    CONCLUSIONS: Our findings unveil the crucial role of astrocyte-specific Nav1.6 in reducing astrocyte-derived Aβ, highlighting its potential as a cell-specific target for modulating AD progression.
    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
    暂无摘要。
    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

  • 文章类型: Journal Article
    天然化合物石杉碱A(HupA)对与年龄相关的学习和记忆缺陷的有益作用促进了该化合物作为促智剂。阿尔茨海默病(AD)的病理生理特征是淀粉样β(Aβ)的积累。毒性Aβ寡聚体在脑中表现出病理损伤之前很早就解释了认知功能障碍。在本研究中,我们研究了HupA对SH-SY5Y神经母细胞瘤细胞中淀粉样前体蛋白(APP)蛋白水解的影响。HupA下调了β位点淀粉样前体蛋白裂解酶1(BACE1)和早老素1(PS1)水平的表达,但增加了A整合素和金属蛋白酶10(ADAM10)的水平,同时Aβ水平显着降低。我们在此首次报告了计算机分子对接分析,该分析揭示了HupA与BACE1的功能活性位点结合。我们进一步分析了HupA对糖原合酶激酶3β(GSK3β)和tau磷酸化状态的影响。在这种情况下,根据目前的观察,我们认为HupA是APP加工的有效调节剂,能够在生理条件下调节tau稳态,在预防和治疗AD样疾病方面具有巨大潜力。
    The beneficial actions of the natural compound Huperzine A (Hup A) against age-associated learning and memory deficits promote this compound as a nootropic agent. Alzheimer\'s disease (AD) pathophysiology is characterized by the accumulation of amyloid beta (Aβ). Toxic Aβ oligomers account for the cognitive dysfunctions much before the pathological lesions are manifested in the brain. In the present study, we investigated the effects of Hup A on amyloid precursor protein (APP) proteolysis in SH-SY5Y neuroblastoma cells. Hup A downregulated the expression of β-site amyloid precursor protein cleaving enzyme 1 (BACE1) and presenilin 1 (PS1) levels but augmented the levels of A disintegrin and metalloproteinase 10 (ADAM10) with significant decrement in the Aβ levels. We herein report for the first time an in silico molecular docking analysis that revealed that Hup A binds to the functionally active site of BACE1. We further analyzed the effect of Hup A on glycogen synthase kinase-3 β (GSK3β) and phosphorylation status of tau. In this scenario, based on the current observations, we propose that Hup A is a potent regulator of APP processing and capable of modulating tau homeostasis under physiological conditions holding immense potential in preventing and treating AD like 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
    淀粉样前体蛋白(APP),分泌酶,和淀粉样蛋白β(Aβ)在阿尔茨海默病(AD)的背景下已被广泛研究。尽管如此,这些蛋白质的功能及其代谢尚不清楚。APP,分泌酶,和APP加工产物(Aβ和C末端片段)定位于内体,线粒体,内质网(ER),和线粒体/ER接触位点。研究暗示APP之间存在显著的关系,分泌酶功能,APP代谢,和线粒体功能。线粒体功能障碍是AD的关键病理标志,并与蛋白质稳定密切相关。这里,我们回顾了研究APP潜在功能的研究,分泌酶,和APP代谢物在线粒体功能和生物能量学的背景下。我们讨论了研究的含义和局限性,并强调了该领域仍然存在的知识差距。
    Amyloid precursor protein (APP), secretase enzymes, and amyloid beta (Aβ) have been extensively studied in the context of Alzheimer\'s disease (AD). Despite this, the function of these proteins and their metabolism is not understood. APP, secretase enzymes, and APP processing products (Aβ and C-terminal fragments) localize to endosomes, mitochondria, endoplasmic reticulum (ER), and mitochondrial/ER contact sites. Studies implicate significant relationships between APP, secretase enzyme function, APP metabolism, and mitochondrial function. Mitochondrial dysfunction is a key pathological hallmark of AD and is intricately linked to proteostasis. Here, we review studies examining potential functions of APP, secretase enzymes, and APP metabolites in the context of mitochondrial function and bioenergetics. We discuss implications and limitations of studies and highlight knowledge gaps that remain in the field.
    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),痴呆症最常见的原因,其特点是内存中断,认知,和个性,显着影响老年人的发病率和死亡率。然而,AD的确切病理生理机制尚不清楚,目前仍缺乏有效的AD治疗方案。人类诱导多能干细胞(iPSC)正在成为疾病研究的有希望的平台。提供建模与各种条件相关的基因突变的能力。患者来源的iPSC可用于模拟神经变性和神经发育障碍。在这项研究中,我们从一名65岁的AD患者外周血单个核细胞中产生了ADiPSCs,该患者在编码淀粉样前体蛋白的基因中携带E682K突变.来自ADiPSCs的脑类器官概括了AD表型,表现出显著增加的tau蛋白水平。我们的分析表明,AD的iPSC疾病模型是病理生理学研究和药物筛选的有价值的评估工具。
    Alzheimer\'s disease (AD), the most common cause of dementia, is characterized by disruptions in memory, cognition, and personality, significantly impacting morbidity and mortality rates among older adults. However, the exact pathophysiological mechanism of AD remains unknown, and effective treatment options for AD are still lacking. Human induced pluripotent stem cells (iPSC) are emerging as promising platforms for disease research, offering the ability to model the genetic mutations associated with various conditions. Patient-derived iPSCs are useful for modeling neurodegenerative and neurodevelopmental disorders. In this study, we generated AD iPSCs from peripheral blood mononuclear cells obtained from a 65-year-old patient with AD carrying the E682K mutation in the gene encoding the amyloid precursor protein. Cerebral organoids derived from AD iPSCs recapitulated the AD phenotype, exhibiting significantly increased levels of tau protein. Our analysis revealed that an iPSC disease model of AD is a valuable assessment tool for pathophysiological research and drug screening.
    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
    基因编辑技术有望创造一种新的治疗方法,通过单一干预即可实现永久性矫正。除了消除家族性疾病中的突变等位基因,基因编辑还可用于有利地操纵上游病理生理事件,并在更广泛的患者群体中改变病程,但很少有这样可行的治疗途径被报道。在这里,我们使用CRISPR-Cas9来编辑淀粉样前体蛋白(App)的最后一个外显子,与阿尔茨海默病(AD)有关。我们的策略有效地消除了APPC末端的内吞(YENPTY)基序,同时保留N端和补偿性APP同源物。这种操作有利地改变了沿着淀粉样蛋白途径的事件-抑制毒性APP-β-裂解片段(包括Aβ)和上调神经保护性APP-α-裂解产物。AAV驱动的编辑改善神经病理学,电生理,和AD敲入小鼠模型中的行为缺陷。效果持续数月,并且即使在种系App编辑后,在WT小鼠中也没有观察到异常;强调总体功效和安全性。App-KI小鼠神经胶质转录组的病理学改变,如单核RNA测序(sNuc-Seq)所示,也通过AppC末端编辑进行归一化。我们的策略利用了先天的转录规则,使末端外显子对无义衰变不敏感,上游操作预计对所有形式的AD都有效。这些研究为AD的一次性疾病修饰治疗提供了途径。
    Gene-editing technologies promise to create a new class of therapeutics that can achieve permanent correction with a single intervention. Besides eliminating mutant alleles in familial disease, gene-editing can also be used to favorably manipulate upstream pathophysiologic events and alter disease-course in wider patient populations, but few such feasible therapeutic avenues have been reported. Here we use CRISPR-Cas9 to edit the last exon of amyloid precursor protein (App), relevant for Alzheimer\'s disease (AD). Our strategy effectively eliminates an endocytic (YENPTY) motif at APP C-terminus, while preserving the N-terminus and compensatory APP-homologues. This manipulation favorably alters events along the amyloid-pathway - inhibiting toxic APP-β-cleavage fragments (including Aβ) and upregulating neuroprotective APP-α-cleavage products. AAV-driven editing ameliorates neuropathologic, electrophysiologic, and behavioral deficits in an AD knockin mouse model. Effects persist for many months, and no abnormalities are seen in WT mice even after germline App-editing; underlining overall efficacy and safety. Pathologic alterations in the glial-transcriptome of App-KI mice, as seen by single nuclei RNA-sequencing (sNuc-Seq), are also normalized by App C-terminus editing. Our strategy takes advantage of innate transcriptional rules that render terminal exons insensitive to nonsense-decay, and the upstream manipulation is expected to be effective for all forms of AD. These studies offer a path for a one-time disease-modifying treatment for AD.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号