Proteinopathy

蛋白质病
  • 文章类型: Journal Article
    小胶质细胞,大脑的常备免疫细胞,在阿尔茨海默病(AD)中变得功能失调,它们的异常免疫反应导致病理蛋白的积累和神经元损伤。遗传研究提示小胶质细胞参与AD的发展,促使人们对开发免疫调节疗法以预防或改善疾病的兴趣。然而,小胶质细胞在疾病中具有不同的功能状态,在AD中既发挥保护作用,又发挥有害作用,在很大程度上重叠并可能在疾病过程中转移,复杂的识别有效的治疗目标。使用转基因小鼠模型收集的大量证据支持小胶质细胞在病理进展中的积极作用,尽管结果各不相同,并且在研究时不同类型的模型和病理程度之间可能是矛盾的。这里,我们回顾了小胶质细胞免疫信号和反应,这些信号和反应有助于病理蛋白的积累和传播或直接影响神经元健康。我们还探索了使用诱导多能干细胞(iPSC)衍生的模型来研究活的人类小胶质细胞,以及它们如何为我们的AD知识做出贡献,并可能开始填补小鼠模型留下的空白。最终,鼠标和iPSC衍生模型有其自身的局限性,只有通过跨模型的综合观点以及对其互补观点和局限性的理解,才能全面了解AD中的小胶质细胞功能障碍。
    Microglia, the resident immune-competent cells of the brain, become dysfunctional in Alzheimer\'s disease (AD), and their aberrant immune responses contribute to the accumulation of pathological proteins and neuronal injury. Genetic studies implicate microglia in the development of AD, prompting interest in developing immunomodulatory therapies to prevent or ameliorate disease. However, microglia take on diverse functional states in disease, playing both protective and detrimental roles in AD, which largely overlap and may shift over the disease course, complicating the identification of effective therapeutic targets. Extensive evidence gathered using transgenic mouse models supports an active role of microglia in pathology progression, though results vary and can be contradictory between different types of models and the degree of pathology at the time of study. Here, we review microglial immune signaling and responses that contribute to the accumulation and spread of pathological proteins or directly affect neuronal health. We additionally explore the use of induced pluripotent stem cell (iPSC)-derived models to study living human microglia and how they have contributed to our knowledge of AD and may begin to fill in the gaps left by mouse models. Ultimately, mouse and iPSC-derived models have their own limitations, and a comprehensive understanding of microglial dysfunction in AD will only be established by an integrated view across models and an appreciation for their complementary viewpoints and limitations.
    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
    神经变性是由于异常蛋白的积累导致特定神经元损失的进行性事件。这些病理形式的蛋白质进一步恶化并干扰正常的生理机制,这可能导致免疫细胞的异常增殖和随后的炎症级联反应,并最终导致神经元丢失。最近,靶向异常的免疫疗法,蛋白质的病理形式已显示出一种有希望的方法来改变神经变性的进展。免疫治疗的最新进展导致了针对蛋白质病的新型抗体的开发,该抗体可以根除蛋白质的聚集,这从临床前和临床研究中可以看出。尽管如此,只有少数成功获得临床批准,而其他人由于缺乏临床疗效终点而停药。当前的综述讨论了临床试验中研究抗体的现状,他们的治疗目标,以及失败或成功的证据。
    Neurodegeneration is a progressive event leading to specific neuronal loss due to the accumulation of aberrant proteins. These pathologic forms of proteins further worsen and interfere with normal physiologic mechanisms, which can lead to abnormal proliferation of immune cells and subsequent inflammatory cascades and ultimately neuronal loss. Recently, immunotherapies targeting abnormal, pathologic forms of protein have shown a promising approach to modify the progression of neurodegeneration. Recent advances in immunotherapy have led to the development of novel antibodies against the proteinopathies which can eradicate aggregations of protein as evident from preclinical and clinical studies. Nonetheless, only a few of them have successfully received clinical approval, while others have been discontinued due to a lack of clinical efficacy endpoints. The current review discusses the status of investigational antibodies under clinical trials, their targets for therapeutic action, and evidence for failure or success.
    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
    目的:突变亨廷顿(mHTT)聚集体的正电子发射断层扫描(PET)成像是监测疾病进展以及亨廷顿病(HD)候选治疗干预措施疗效的潜在工具。迄今为止,重点主要是调查11C放射性配体;然而,有利的18F放射性示踪剂将促进未来的临床翻译。这项工作旨在在HD小鼠模型中使用体内和体外方法的组合来表征新型[18F]CHDI-650PET放射性示踪剂。
    方法:使用体外放射自显影对[18F]CHDI-650进行表征后,我们通过动态PET成像评估了9月龄时HD和野生型(WT)同窝的杂合(HET)zQ175DN小鼠模型的体内血浆和脑部放射性示踪剂稳定性以及动力学.此外,我们在3个月时与先前发表的[11C]CHDI-180R放射性配体进行了正面对比研究.
    结果:血浆和脑放射性代谢物谱表明[18F]CHDI-650的体内成像的合适代谢谱。在9月龄时的体外放射自显影和体内[18F]CHDI-650PET成像均显示出显著的基因型效应(p<0.0001),尽管测试-重测可靠性差。与[11C]CHDI-180R相比,3月龄的[18F]CHDI-650PET成像在基因型之间显示出更高的分化。
    结论:总体而言,[18F]CHDI-650允许区分9和3月龄的HET和WTzQ175DN小鼠。[18F]CHDI-650代表第一个合适的18F放射性配体,用于在小鼠中对mHTT聚集体成像,其临床评估正在进行中。
    OBJECTIVE: Positron emission tomography (PET) imaging of mutant huntingtin (mHTT) aggregates is a potential tool to monitor disease progression as well as the efficacy of candidate therapeutic interventions for Huntington\'s disease (HD). To date, the focus has been mainly on the investigation of 11C radioligands; however, favourable 18F radiotracers will facilitate future clinical translation. This work aimed at characterising the novel [18F]CHDI-650 PET radiotracer using a combination of in vivo and in vitro approaches in a mouse model of HD.
    METHODS: After characterising [18F]CHDI-650 using in vitro autoradiography, we assessed in vivo plasma and brain radiotracer stability as well as kinetics through dynamic PET imaging in the heterozygous (HET) zQ175DN mouse model of HD and wild-type (WT) littermates at 9 months of age. Additionally, we performed a head-to-head comparison study at 3 months with the previously published [11C]CHDI-180R radioligand.
    RESULTS: Plasma and brain radiometabolite profiles indicated a suitable metabolic profile for in vivo imaging of [18F]CHDI-650. Both in vitro autoradiography and in vivo [18F]CHDI-650 PET imaging at 9 months of age demonstrated a significant genotype effect (p < 0.0001) despite the poor test-retest reliability. [18F]CHDI-650 PET imaging at 3 months of age displayed higher differentiation between genotypes when compared to [11C]CHDI-180R.
    CONCLUSIONS: Overall, [18F]CHDI-650 allows for discrimination between HET and WT zQ175DN mice at 9 and 3 months of age. [18F]CHDI-650 represents the first suitable 18F radioligand to image mHTT aggregates in mice and its clinical evaluation is underway.
    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
    突变蛋白在细胞中的积累可以诱导蛋白质病并引起器官的功能损伤。最近,Cingulin(CGN)蛋白已被证明可以维持内耳毛细胞表皮板的形态,CGN中的移码突变会导致常染色体显性遗传的非综合征性听力损失。这里,我们发现突变CGN蛋白形成不溶性聚集体,这些聚集体在细胞内积累并导致细胞死亡。突变CGN在内耳中的表达导致小鼠严重的毛细胞死亡和听力损失,类似于人类患者的听觉表型。有趣的是,由移码突变产生的新肽中的人特异性残基(V1112)对于突变型人CGN的聚集和细胞毒性至关重要。此外,热休克因子1(HSF1)的表达减少了不溶性突变体CGN聚集体的积累并挽救了细胞死亡。总之,这些发现将突变特异性毒性多肽鉴定为CGN中耳聋突变的致病机制,可以通过表达细胞伴侣反应调节因子HSF1来靶向。
    Accumulation of mutant proteins in cells can induce proteinopathies and cause functional damage to organs. Recently, the Cingulin (CGN) protein has been shown to maintain the morphology of cuticular plates of inner ear hair cells and a frameshift mutation in CGN causes autosomal dominant non-syndromic hearing loss. Here, we find that the mutant CGN proteins form insoluble aggregates which accumulate intracellularly and lead to cell death. Expression of the mutant CGN in the inner ear results in severe hair cell death and hearing loss in mice, resembling the auditory phenotype in human patients. Interestingly, a human-specific residue (V1112) in the neopeptide generated by the frameshift mutation is critical for the aggregation and cytotoxicity of the mutant human CGN. Moreover, the expression of heat shock factor 1 (HSF1) decreases the accumulation of insoluble mutant CGN aggregates and rescues cell death. In summary, these findings identify mutant-specific toxic polypeptides as a disease-causing mechanism of the deafness mutation in CGN, which can be targeted by the expression of the cell chaperone response regulator HSF1.
    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
    遗传性甲状腺素运载蛋白淀粉样变性(hATTR)伴多发性神经病(以前称为家族性淀粉样多发性神经病(FAP))是一种地方性淀粉样变性,涉及蛋白质的有害聚集,最常见的是转甲状腺素蛋白(TTR),但有时也有载脂蛋白A-1或凝溶胶蛋白。hATTR似乎作为常染色体显性性状传播。已经鉴定出超过100个点突变,Val30Met替代是最常见的。然而,hATTR的发病机制和总体起源尚不清楚.这里,我们认为hATTR可能与有害金属接触有关。HATTR发病率在全球分布不均,三个最大的定义集群存在于日本,葡萄牙,和瑞典。这三个疫区也是古老的矿区,当地环境受到金属污染。有害金属有两种主要机制,摄取到组织和体液后,可以诱导hATTR。首先,金属可以直接影响表达,函数,和/或参与hATTR病理学的蛋白质的聚集。这种金属-蛋白质相互作用可能构成抗hATTR药物设计的分子靶标。第二,金属暴露可诱导HATTR相关基因突变,这可能发生在几代人之前。这两种机制可以并行发生。总之,在地球化学定义的区域,hATTR可能与金属暴露有关的可能性值得进一步关注。
    Hereditary transthyretin amyloidosis (hATTR) with polyneuropathy (formerly known as Familial Amyloid Polyneuropathy (FAP)) is an endemic amyloidosis involving the harmful aggregation of proteins, most commonly transthyretin (TTR) but sometimes also apolipoprotein A-1 or gelsolin. hATTR appears to be transmitted as an autosomal dominant trait. Over 100 point mutations have been identified, with the Val30Met substitution being the most common. Yet, the mechanism of pathogenesis and the overall origin of hATTR remain unclear. Here, we argue that hATTR could be related to harmful metal exposure. hATTR incidence is unevenly distributed globally, and the three largest defined clusters exist in Japan, Portugal, and Sweden. All three disease regions are also ancient mining districts with associated metal contamination of the local environment. There are two main mechanisms for how harmful metals, after uptake into tissues and body fluids, could induce hATTR. First, the metals could directly influence the expression, function, and/or aggregation of the proteins involved in hATTR pathology. Such metal-protein interactions might constitute molecular targets for anti-hATTR drug design. Second, metal exposure could induce hATTR -associated genetic mutations, which may have happened several generations ago. These two mechanisms can occur in parallel. In conclusion, the possibility that hATTR could be related to metal exposure in geochemically defined regions deserves further attention.
    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
    溶酶体和跨膜蛋白106B(TMEM106B)的遗传变异改变了各种神经退行性疾病的风险,尤其是额颞叶变性(FTLD)伴有颗粒原蛋白(PGRN)单plo功能不全,尽管涉及的分子机制尚未被理解。通过低温电子显微镜(cryo-EM)的发展,TMEM106B(TMEMCT)的C末端结构域的同型聚集体被发现为FTLD大脑中先前未发现的细胞溶质蛋白病,老年痴呆症,进行性核上性麻痹(PSP),路易体痴呆(DLB)患者。虽然尚不清楚TMEMCT聚集在神经元丢失中起什么作用,它在一系列与衰老相关的痴呆疾病中的存在表明参与了多蛋白病驱动的神经变性。为了确定TMEMCT聚集倾向和神经退行性潜能,我们表征了一种新型的转基因秀丽隐杆线虫模型,该模型表达了人TMEMCT片段,该片段构成了FTLD病例中所见的纤维状核心。我们发现,秀丽隐杆线虫中人TMEMCT的泛神经元表达会导致神经元功能障碍,如行为分析所证明的。TMEMCT蛋白的胞浆聚集伴随行为功能障碍驱动神经变性,如GABA能神经元的损失所示。探讨TMEM106B蛋白病的分子机制,我们探讨了PGRN丢失对TMEMCT表达的神经退行性影响。为此,我们产生了表达秀丽隐杆线虫的TMEMCT,缺失pgrn-1,人类PGRN的C.elegans直系同源物。pgrn-1的全部或部分损失均未改变我们的TMEMCT模型的运动表型,表明TMEMCT聚集发生在PGRN功能丧失的下游。我们还测试了tau蛋白病的遗传抑制因子挽救TMEMCT病理学的能力。我们发现spop-1,sut-2和sut-6的基因敲除导致蛋白质病表型的弱至无挽救,表明TMEM106B蛋白病的机制驱动因素可能与tau蛋白病不同。一起来看,我们的数据表明,TMEMCT聚集可以杀死神经元。Further,TMEMCT在C.elegans神经元中的表达为神经退行性疾病中TMEM106B蛋白病的功能表征提供了有用的模型。
    Genetic variation in the lysosomal and transmembrane protein 106B (TMEM106B) modifies risk for a diverse range of neurodegenerative disorders, especially frontotemporal lobar degeneration (FTLD) with progranulin (PGRN) haplo-insufficiency, although the molecular mechanisms involved are not yet understood. Through advances in cryo-electron microscopy (cryo-EM), homotypic aggregates of the C-Terminal domain of TMEM106B (TMEM CT) were discovered as a previously unidentified cytosolic proteinopathy in the brains of FTLD, Alzheimer\'s disease, progressive supranuclear palsy (PSP), and dementia with Lewy bodies (DLB) patients. While it remains unknown what role TMEM CT aggregation plays in neuronal loss, its presence across a range of aging related dementia disorders indicates involvement in multi-proteinopathy driven neurodegeneration. To determine the TMEM CT aggregation propensity and neurodegenerative potential, we characterized a novel transgenic C. elegans model expressing the human TMEM CT fragment constituting the fibrillar core seen in FTLD cases. We found that pan-neuronal expression of human TMEM CT in C. elegans causes neuronal dysfunction as evidenced by behavioral analysis. Cytosolic aggregation of TMEM CT proteins accompanied the behavioral dysfunction driving neurodegeneration, as illustrated by loss of GABAergic neurons. To investigate the molecular mechanisms driving TMEM106B proteinopathy, we explored the impact of PGRN loss on the neurodegenerative effect of TMEM CT expression. To this end, we generated TMEM CT expressing C. elegans with loss of pgrn-1, the C. elegans ortholog of human PGRN. Neither full nor partial loss of pgrn-1 altered the motor phenotype of our TMEM CT model suggesting TMEM CT aggregation occurs downstream of PGRN loss of function. We also tested the ability of genetic suppressors of tauopathy to rescue TMEM CT pathology. We found that genetic knockout of spop-1, sut-2, and sut-6 resulted in weak to no rescue of proteinopathy phenotypes, indicating that the mechanistic drivers of TMEM106B proteinopathy may be distinct from tauopathy. Taken together, our data demonstrate that TMEM CT aggregation can kill neurons. Further, expression of TMEM CT in C. elegans neurons provides a useful model for the functional characterization of TMEM106B proteinopathy in neurodegenerative disease.
    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
    脊髓小脑性共济失调7型(SCA7)是一种进行性神经退行性疾病,由其疾病蛋白中不间断的聚谷氨酰胺(polyQ)重复序列异常扩张引起,ataxin-7(ATXN7)。ATXN7是Spt-Ada-Gcn5乙酰转移酶(SAGA)的一部分,在染色质重塑中具有关键作用的进化保守的转录共激活复合物,细胞信号,神经分化,线粒体健康和自噬。SCA7主要是遗传的,其特征是遗传预期和高重复长度不稳定性。SCA7患者经历进行性共济失调,萎缩,痉挛,和失明。目前没有治愈SCA7的方法,治疗旨在缓解症状以提高生活质量。这里,我们报道了在野生型和人类疾病患者范围内具有polyQ重复的SCA7果蝇新品系。我们发现ATXN7表达在果蝇存活和视网膜不稳定中具有年龄和polyQ重复长度依赖性减少,伴随着ATXN7蛋白聚集的增加。这些新的产品线将为疾病进展提供重要的见解,将来可用于确定SCA7患者的治疗靶标。
    Spinocerebellar ataxia type 7 (SCA7) is a progressive neurodegenerative disorder resulting from abnormal expansion of an uninterrupted polyglutamine (polyQ) repeat in its disease protein, ataxin-7 (ATXN7). ATXN7 is part of Spt-Ada-Gcn5 acetyltransferase (SAGA), an evolutionarily conserved transcriptional coactivation complex with critical roles in chromatin remodeling, cell signaling, neurodifferentiation, mitochondrial health and autophagy. SCA7 is dominantly inherited and characterized by genetic anticipation and high repeat-length instability. Patients with SCA7 experience progressive ataxia, atrophy, spasticity, and blindness. There is currently no cure for SCA7, and therapies are aimed at alleviating symptoms to increase quality of life. Here, we report novel Drosophila lines of SCA7 with polyQ repeats in wild-type and human disease patient range. We find that ATXN7 expression has age- and polyQ repeat length-dependent reduction in fruit fly survival and retinal instability, concomitant with increased ATXN7 protein aggregation. These new lines will provide important insight on disease progression that can be used in the future to identify therapeutic targets for SCA7 patients.
    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
    在TDP43蛋白病患者中,磷酸化的TDP43(p-TDP43)在神经元的细胞质中积累。在tau蛋白病和α-突触核蛋白病的患者中也报道了p-TDP43的积累。我们研究了过表达人突变体tau(P301L)并表现出过磷酸化tau(hp-tau)和磷酸化αSyn(p-αSyn)积累的rTg4510小鼠大脑中p-TDP43积累的时空变化。免疫组织化学,在神经元的细胞质中观察到p-TDP43聚集体,随着年龄的增长。在具有p-TDP43聚集体和hp-tau和p-αSyn聚集体的细胞数量之间观察到显著正相关。通过多西环素处理抑制人突变型tau(P301L)表达减少了p-TDP43、hp-tau、还有p-αSyn.在高hp-tau区域发现了蛋白酶K抗性p-TDP43聚集体,和p-αSyn积累。肌糖基不溶性部分的蛋白质印迹显示了单体TDP43和p-TDP43的条带。这些结果表明,小鼠p-TDP43的积累与人突变tau(P301L)在rTg4510小鼠脑中的积累有关。hp-tau和p-αSyn的积累可能会促进对蛋白酶K具有抗性的不溶性sarkosylp-TDP43聚集体。TDP43和αSyn可能与蛋白质病的病理学有关,导致多种异常蛋白质的积累。
    In patients with TDP43 proteinopathy, phosphorylated TDP43 (p-TDP43) accumulates in the cytoplasm of neurons. The accumulation of p-TDP43 has also been reported in patients with tauopathy and α-synucleinopathy. We investigated spatiotemporal changes in p-TDP43 accumulation in the brains of rTg4510 mice that overexpressed human mutant tau (P301L) and exhibited hyperphosphorylated tau (hp-tau) and phosphorylated αSyn (p-αSyn) accumulation. Immunohistochemically, p-TDP43 aggregates were observed in the cytoplasm of neurons, which increased with age. A significant positive correlation was observed between the number of cells with p-TDP43 aggregates and hp-tau and p-αSyn aggregates. Suppression of the human mutant tau (P301L) expression by doxycycline treatment reduces the accumulation of p-TDP43, hp-tau, and p-αSyn. Proteinase K-resistant p-TDP43 aggregates were found in regions with high hp-tau, and p-αSyn accumulation. Western blotting of the sarkosyl-insoluble fraction revealed bands of monomeric TDP43 and p-TDP43. These results indicate that the accumulation of mouse p-TDP43 is associated with the accumulation of human mutant tau (P301L) in rTg4510 mouse brains. The accumulation of hp-tau and p-αSyn may promote sarkosyl-insoluble p-TDP43 aggregates that are resistant to proteinase K. The synergistic effects of tau, TDP43, and αSyn may be involved in the pathology of proteinopathies, leading to the accumulation of multiple abnormal proteins.
    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
    中枢神经系统(CNS)被广泛认为是唯一没有淋巴毛细血管的器官系统,可以促进间质代谢副产物的去除。因此,新发现的在神经系统中提供假淋巴活动的淋巴系统一直是神经科学最新研究的焦点。此外,调查结果显示,睡眠会刺激淋巴系统的消除行为,并与正常的大脑稳态有关。中枢神经系统通过淋巴系统清除潜在危险的化合物,特别是在睡眠期间。各种神经退行性疾病的脑功能和病理生理学的任何年龄相关改变都表明脑淋巴系统的紊乱。在这种情况下,β-淀粉样蛋白以及tau通过淋巴系统离开中枢神经系统,根据许多发现,老年人大脑的功能和脑脊液放电明显改变。因此,淋巴功能衰竭可能具有潜在的机制,该机制可能在几种神经退行性和年龄相关的认知疾病中具有治疗针对性。因此,人们迫切需要对淋巴系统和几种潜在的脑相关疾病之间的联系进行更多的研究。这里,在我们当前的评论文件中,我们回顾了目前的研究,在一些普遍的神经退行性疾病和神经精神疾病的参与,我们还讨论了几种治疗方法,饮食和生活方式的改变,可用于获得更彻底的性能和目的,以破译新的前景,临床应用于这些疾病的管理。
    The central nervous system (CNS) is widely recognized as the only organ system without lymphatic capillaries to promote the removal of interstitial metabolic by-products. Thus, the newly identified glymphatic system which provides a pseudolymphatic activity in the nervous system has been focus of latest research in neurosciences. Also, findings reported that, sleep stimulates the elimination actions of glymphatic system and is linked to normal brain homeostatis. The CNS is cleared of potentially hazardous compounds via the glymphatic system, particularly during sleep. Any age-related alterations in brain functioning and pathophysiology of various neurodegenerative illnesses indicates the disturbance of the brain\'s glymphatic system. In this context, β-amyloid as well as tau leaves the CNS through the glymphatic system, it\'s functioning and CSF discharge markedly altered in elderly brains as per many findings. Thus, glymphatic failure may have a potential mechanism which may be therapeutically targetable in several neurodegenerative and age-associated cognitive diseases. Therefore, there is an urge to focus for more research into the connection among glymphatic system and several potential brain related diseases. Here, in our current review paper, we reviewed current research on the glymphatic system\'s involvement in a number of prevalent neurodegenerative and neuropsychiatric diseases and, we also discussed several therapeutic approaches, diet and life style modifications which might be used to acquire a more thorough performance and purpose of the glymphatic system to decipher novel prospects for clinical applicability for the management of these diseases.
    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
    肌萎缩侧索硬化症(ALS)是一种致命的疾病。由于其病理机制尚不清楚,目前还没有有效的治疗方法。虽然它在病因和临床上都是高度异质性的,它有一个共同的显著标志,即,异常蛋白质聚集(APA)。ALS中APA的上游发病机理和下游作用是复杂的,对这种病理的研究对于理解ALS具有重要意义。在本文中,讨论了ALS中APA的发病机制及其候选治疗策略。
    Amyotrophic lateral sclerosis (ALS) is a fatal disease. As its pathological mechanisms are not well understood, there are no efficient therapeutics for it at present. While it is highly heterogenous both etiologically and clinically, it has a common salient hallmark, i.e., aberrant protein aggregation (APA). The upstream pathogenesis and the downstream effects of APA in ALS are sophisticated and the investigation of this pathology would be of consequence for understanding ALS. In this paper, the pathomechanism of APA in ALS and the candidate treatment strategies for it are discussed.
    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