KIF5A

KIF5A
  • 文章类型: Journal Article
    肌萎缩侧索硬化症(ALS)是一种破坏性的神经退行性疾病,其特征是对上下运动神经元的进行性损害。众所周知,遗传因素在ALS中起着至关重要的作用,因为基因研究不仅提高了我们对疾病机制的理解,而且有助于解开患者表现出的复杂表型。为了进一步了解中国人群中ALS的遗传景观,并探索个体之间的基因型-表型相关性,我们进行了全基因组测序,以筛选缺乏最常见ALS相关基因的34位中国家族性ALS(FALS)先证者的基因.在这个群体中,我们在一个先证者的KIF5A的N端结构域中发现了一个罕见的杂合错义突变(c.86A>G)。这一发现具有重要意义,因为自2018年以来,KIF5A基因的突变与欧洲队列中的ALS有关,主要以C端突变为特征。对该家族谱系内的临床表型的分析揭示了症状的延迟发作,延长的生存时间,和两个上肢的初始表现。这些观察结果强调了在具有KIF5A突变的ALS患者中观察到的临床异质性。总之,我们的研究有助于越来越多的证据将KIF5A与ALS联系起来,并增强了我们对这种疾病复杂遗传格局的理解.
    Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disorder characterized by progressive damage to both upper and lower motor neurons. Genetic factors are known to play a crucial role in ALS, as genetic studies not only advance our comprehension of disease mechanisms but also help unravel the complex phenotypes exhibited by patients. To gain further insights into the genetic landscape of ALS in the Chinese population and explore genotype-phenotype correlations among individuals, we conducted whole-genome sequencing to screen genes in 34 Chinese familial ALS (FALS) probands lacking the most common ALS-associated genes. Within this cohort, we identified a rare heterozygous missense mutation in the N-terminal domain of KIF5A (c.86A>G) in one of the probands. This finding is significant as mutations in the KIF5A gene have been implicated in ALS in European cohorts since 2018, predominantly characterized by C-terminal mutations. Analysis of the clinical phenotype within this familial lineage revealed a delayed onset of symptoms, an extended survival duration, and initial manifestations in both upper limbs. These observations underscore the clinical heterogeneity observed in ALS patients harboring KIF5A mutations. In conclusion, our study contributes to the growing body of evidence linking KIF5A to ALS and enhances our understanding of the intricate genetic landscape of this 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
    预后不良的癌症相关死亡的主要原因是肺腺癌(LUAD)。KIF5A,驱动蛋白超家族的重要成员,与恶性肿瘤的耐药性有关。本研究旨在探讨KIF5A在LUAD细胞多西他赛(DTX)耐药中的作用机制。生物信息学分析的结果,qRT-PCR和westernblot分析表明,KIF5A,参与糖酵解途径,在LUAD中高表达,并与糖酵解相关基因呈正相关。我们进一步验证KIF5A的沉默抑制DTX抗性,糖酵解,通过细胞计数试剂盒-8(CCK-8)在LUAD细胞中产生乳酸,流式细胞术,海马XFe96,乳酸,和葡萄糖测定。机械上,KIF5A促进LUAD的DTX抗性,并且这种效应在添加LDHA抑制剂时减弱。染色质免疫沉淀和双荧光素酶报告基因测定显示FOXP3转录激活KIF5A。敲除FOXP3可减少LUAD的乳酸产生并增强DTX敏感性,在同时过表达KIF5A后恢复。我们的发现表明,FOXP3通过上调KIF5A水平来增强乳酸的产生,从而增加了LUAD细胞中的DTX抗性。总之,我们的研究为改善LUAD的化疗敏感性提供了一个新的治疗靶点.
    A prominent cause of cancer-related fatalities with a poor prognosis is lung adenocarcinoma (LUAD). KIF5A, a crucial member of the kinesin superfamily, is linked to drug resistance in malignancies. This work aims to investigate the mechanism of KIF5A in docetaxel (DTX) resistance in LUAD cells. The results of bioinformatics analysis, qRT-PCR and western blot analysis show that KIF5A, which is involved in the glycolysis pathway, is highly expressed in LUAD and is positively correlated with glycolysis-related genes. We further verify that silencing of KIF5A inhibits DTX resistance, glycolysis, and lactate production in LUAD cells via cell counting kit-8 (CCK-8), flow cytometry, Seahorse XFe 96, lactate, and glucose assays. Mechanistically, KIF5A promotes DTX resistance in LUAD, and this effect is attenuated upon the addition of an LDHA inhibitor. Chromatin immunoprecipitation and dual-luciferase reporter assays reveal that FOXP3 transcriptionally activates KIF5A. Knockdown of FOXP3 reduces lactate production and enhances DTX sensitivity in LUAD, which is restored upon simultaneous overexpression of KIF5A. Our findings reveal that FOXP3 increases DTX resistance in LUAD cells by enhancing lactate production through the upregulation of KIF5A level. In conclusion, our study provides a novel treatment target for improving chemosensitivity in LUAD.
    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
    百草枯(PQ)是一种广谱除草剂,对人类健康有害。累积证据加强了PQ暴露与帕金森病(PD)发展之间的关联。然而,PQ诱导的神经毒性的潜在机制和有效干预措施尚不清楚.在这项研究中,C57BL/6J小鼠用PQ治疗(i.p.,10mg/kg,每周两次)和褪黑激素(i.g.,20mg/kg,每周两次)持续8周。结果表明,褪黑素预处理可以保护PQ引起的C57BL/6J小鼠的运动缺陷和中脑多巴胺能神经元损伤。在分离的初级中脑神经元和SK-N-SH细胞中,细胞活力降低,ROS总水平的升高,褪黑素减轻了PQ引起的轴突线粒体转运缺陷和线粒体功能障碍。筛选后表达驱动轴突线粒体运输的主马达,数据显示,褪黑素拮抗了PQ降低的KIF5A在小鼠中脑和SK-N-SH细胞中的表达。使用体外KIF5A过表达模型,发现KIF5A过表达抑制了PQ引起的SK-N-SH细胞的神经毒性和线粒体功能障碍。此外,MTNR1B(MT2)受体拮抗剂的应用,4-P-PDOT,显著抵消褪黑素对PQ诱导的神经毒性的保护作用。Further,在C57BL/6J小鼠中,Kif5a敲低减少了褪黑素诱导的运动缺陷和针对PQ的神经元损伤的缓解。本研究建立了环境神经毒物暴露与PD病因之间的因果关系,并为PD的发病机理提供了有效的干预靶标。
    Paraquat (PQ) is a broad-spectrum herbicide used worldwide and is a hazardous chemical to human health. Cumulative evidence strengthens the association between PQ exposure and the development of Parkinson\'s disease (PD). However, the underlying mechanism and effective interventions against PQ-induced neurotoxicity remain unclear. In this study, C57BL/6 J mice were treated with PQ (i.p., 10 mg/kg, twice a week) and melatonin (i.g., 20 mg/kg, twice a week) for 8 weeks. Results showed that PQ-induced motor deficits and midbrain dopaminergic neuronal damage in C57BL/6 J mice were protected by melatonin pretreatment. In isolated primary midbrain neurons and SK-N-SH cells, reduction of cell viability, elevation of total ROS levels, axonal mitochondrial transport defects and mitochondrial dysfunction caused by PQ were attenuated by melatonin. After screening of expression of main motors driving axonal mitochondrial transport, data showed that PQ-decreased KIF5A expression in mice midbrain and in SK-N-SH cell was antagonized by melatonin. Using the in vitro KIF5A-overexpression model, it was found that KIF5A overexpression inhibited PQ-caused neurotoxicity and mitochondrial dysfunction in SK-N-SH cells. In addition, application of MTNR1B (MT2) receptor antagonist, 4-P-PDOT, significantly counteracted the protection of melatonin against PQ-induced neurotoxicity. Further, Kif5a-knockdown diminished melatonin-induced alleviation of motor deficits and neuronal damage against PQ in C57BL/6 J mice. The present study establishes a causal link between environmental neurotoxicants exposure and PD etiology and provides effective interventive targets in the pathogenesis of PD.
    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
    Kinesin-1是一种微管马达,可沿着微管运输细胞货物。KIF5A是人类三种驱动蛋白-1亚型之一,所有这些都被电机和C末端尾部近端区域的IAK基序之间的相互作用自动抑制。KIF5A的C末端尾部比其他两种驱动蛋白-1同工型(KIF5B和KIF5C)长80个残基,目前尚不清楚它是否有助于自身抑制。KIF5A的突变会引起神经元疾病,并可能影响自身抑制,如报道的,跳过外显子27的突变,改变其C端序列。这里,我们结合了负染色电子显微镜,交联质谱(XL-MS)和AlphaFold2结构预测,以确定全长自抑制KIF5A同二聚体的分子结构,在没有轻链的情况下。我们证明KIF5A形成了一个紧凑的,弯曲的构象,通过盘绕线圈2和3之间的弯曲,围绕P687。WTKIF5A的XL-MS揭示了电机中残基之间的广泛相互作用,在线圈1和电机之间,在卷曲线圈1和2之间,具有卷曲线圈3和4,以及C末端尾部的近端区域和处于自动禁止状态的电机,但不在远端C末端区域和分子的其余部分之间。虽然外显子-27KIF5A剪接突变体的阴性染色电子显微镜显示存在自抑制分子,XL-MS分析表明其自抑制状态更不稳定。我们的模型提供了一个概念框架,用于了解运动和茎域内的突变如何影响运动活动。
    Kinesin-1 is a microtubule motor that transports cellular cargo along microtubules. KIF5A is one of three kinesin-1 isoforms in humans, all of which are autoinhibited by an interaction between the motor and an IAK motif in the proximal region of the C-terminal tail. The C-terminal tail of KIF5A is ∼80 residues longer than the other two kinesin-1 isoforms (KIF5B and KIF5C) and it is unclear if it contributes to autoinhibition. Mutations in KIF5A cause neuronal diseases and could affect autoinhibition, as reported for a mutation that skips exon 27, altering its C-terminal sequence. Here, we combined negative-stain electron microscopy, crosslinking mass spectrometry (XL-MS) and AlphaFold2 structure prediction to determine the molecular architecture of the full-length autoinhibited KIF5A homodimer, in the absence of light chains. We show that KIF5A forms a compact, bent conformation, through a bend between coiled-coils 2 and 3, around P687. XL-MS of WT KIF5A revealed extensive interactions between residues in the motor, between coiled-coil 1 and the motor, between coiled-coils 1 and 2, with coiled-coils 3 and 4, and the proximal region of the C-terminal tail and the motor in the autoinhibited state, but not between the distal C-terminal region and the rest of the molecule. While negative-stain electron microscopy of exon-27 KIF5A splice mutant showed the presence of autoinhibited molecules, XL-MS analysis suggested that its autoinhibited state is more labile. Our model offers a conceptual framework for understanding how mutations within the motor and stalk domain may affect motor activity.
    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
    细胞骨架运动蛋白是水解ATP以产生沿着细胞骨架细丝的力和运动的必要分子机器。动力蛋白和驱动蛋白超家族的成员在运输生物有效载荷(如蛋白质,细胞器,和囊泡)沿着微管途径,导致鞭毛和纤毛的跳动,并在有丝分裂和减数分裂纺锤体内作用,将复制的染色体分离到子代细胞。了解运动蛋白的潜在机制和行为对于为运动蛋白相关疾病的治疗提供更好的策略至关重要。这里,我们提供了使用杆状病毒/昆虫细胞系统重组表达Kinesin-1马达KIF5C的详细方案,并提供了使用全内反射荧光显微镜和光学镊子进行单分子研究的更新方案,以研究纯化马达的运动性和力的产生.
    Cytoskeletal motor proteins are essential molecular machines that hydrolyze ATP to generate force and motion along cytoskeletal filaments. Members of the dynein and kinesin superfamilies play critical roles in transporting biological payloads (such as proteins, organelles, and vesicles) along microtubule pathways, cause the beating of flagella and cilia, and act within the mitotic and meiotic spindles to segregate replicated chromosomes to progeny cells. Understanding the underlying mechanisms and behaviors of motor proteins is critical to provide better strategies for the treatment of motor protein-related diseases. Here, we provide detailed protocols for the recombinant expression of the Kinesin-1 motor KIF5C using a baculovirus/insect cell system and provide updated protocols for performing single-molecule studies using total internal reflection fluorescence microscopy and optical tweezers to study the motility and force generation of the purified motor.
    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
    编码驱动蛋白家族成员5A(KIF5A)的基因中的单核苷酸变体(SNV),一种参与微管顺行运输的神经元运动蛋白,与肌萎缩侧索硬化症(ALS)有关。ALS是一种快速进展和致命的神经退行性疾病,主要影响运动神经元。许多ALS相关的KIF5ASNV聚集在倒数第二个外显子27的剪接位点连接处附近,并预测会改变KIF5A的羧基末端(C端)货物结合域。外显子27的错误剪接,导致外显子排斥,被认为是这些SNV引起ALS的机制。是否所有邻近外显子27的SNV导致外显子排除尚不清楚。为了解决这个问题,我们在HEK293细胞中设计了一种体外小基因剪接测定法,该测定法揭示了对剪接的异质位点特异性作用:只有5'剪接位点(5'ss)SNV导致外显子跳跃。我们还量化了分化为运动神经元的选择CRISPR编辑的人干细胞和5'ssSNV敲入小鼠的神经元组织中的剪接,显示出相同的结果。此外,代表性3\'剪接位点的存活,5\'ss,与野生型运动神经元相比,截短的C-term变体KIF5A(v-KIF5A)运动神经元严重减少,明显的形态变化。虽然KIF5AmRNA的总水平在细胞系中具有可比性,v-KIF5A系的总KIF5A蛋白水平降低,提示蛋白质合成或稳定性受损。因此,尽管对RNA剪接有异质作用,KIF5ASNV同样降低了KIF5A蛋白的可用性,导致轴突运输缺陷和运动神经元病理。
    Single-nucleotide variants (SNVs) in the gene encoding Kinesin Family Member 5A (KIF5A), a neuronal motor protein involved in anterograde transport along microtubules, have been associated with amyotrophic lateral sclerosis (ALS). ALS is a rapidly progressive and fatal neurodegenerative disease that primarily affects the motor neurons. Numerous ALS-associated KIF5A SNVs are clustered near the splice-site junctions of the penultimate exon 27 and are predicted to alter the carboxy-terminal (C-term) cargo-binding domain of KIF5A. Mis-splicing of exon 27, resulting in exon exclusion, is proposed to be the mechanism by which these SNVs cause ALS. Whether all SNVs proximal to exon 27 result in exon exclusion is unclear. To address this question, we designed an in vitro minigene splicing assay in human embryonic kidney 293 cells, which revealed heterogeneous site-specific effects on splicing: only 5\' splice-site (5\'ss) SNVs resulted in exon skipping. We also quantified splicing in select clustered, regularly interspaced, short palindromic repeats-edited human stem cells, differentiated to motor neurons, and in neuronal tissues from a 5\'ss SNV knock-in mouse, which showed the same result. Moreover, the survival of representative 3\' splice site, 5\'ss, and truncated C-term variant KIF5A (v-KIF5A) motor neurons was severely reduced compared with wild-type motor neurons, and overt morphological changes were apparent. While the total KIF5A mRNA levels were comparable across the cell lines, the total KIF5A protein levels were decreased for v-KIF5A lines, suggesting an impairment of protein synthesis or stability. Thus, despite the heterogeneous effect on ribonucleic acid splicing, KIF5A SNVs similarly reduce the availability of the KIF5A protein, leading to axonal transport defects and motor neuron pathology.
    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
    Kinesin家族成员5A(KIF5A)是必不可少的,神经元特异性微管相关运动蛋白,负责各种细胞货物的顺行轴突运输。N端功能变体的丢失,微管结合域与遗传性痉挛性截瘫和遗传性运动神经病有关。这些变体导致突变蛋白沿微管加工的能力丧失。相反,在C末端获得功能剪接位点变体,KIF5A的货物结合域与肌萎缩侧索硬化症(ALS)相关,一种涉及上下运动神经元死亡的神经退行性疾病,最终导致运动单位(MU;α运动神经元及其支配的所有肌纤维)退化和死亡。这些ALS相关变体导致自身抑制的丧失,沿着微管的突变蛋白的增加,和改变货物绑定。为了研究体内ALS相关变体的分子和细胞后果,我们使用CRISPR-Cas9基因编辑将ALS相关KIF5A变体的鼠同源物引入C57BL/6小鼠中,该基因编辑在杂合(Kif5a+/c.3005+1G>A;HET)和纯合(Kif5ac.3005+1G>A/c.3005+1G>A;HOM)小鼠的神经元组织中产生突变Kif5amRNA和蛋白。HET和HOM小鼠在1岁时的行为和电生理(复合肌肉动作电位[CMAP]和MU数量估计[MUNE])结果测量中表现正常。当受到坐骨神经损伤时,与野生型(WT)小鼠相比,HET和HOM小鼠的MUNE恢复延迟且不完全恢复,表明MU修复受损。此外,年龄突变的Kif5a小鼠(两岁)的MUNE降低,而与损伤无关,与老年WT小鼠相比,损伤后延迟和不完全恢复的恶化。这些数据表明,ALS相关变体可能导致MU受损,以应对诸如损伤和衰老等生物学挑战。导致MU维修和保养失败。在这份报告中,我们呈现行为,具有ALS相关Kif5a变体的小鼠的电生理和病理学表征,以了解KIF5AC末端变体在体内的功能后果。
    Kinesin family member 5A (KIF5A) is an essential, neuron-specific microtubule-associated motor protein responsible for the anterograde axonal transport of various cellular cargos. Loss of function variants in the N-terminal, microtubule-binding domain are associated with hereditary spastic paraplegia and hereditary motor neuropathy. These variants result in a loss of the ability of the mutant protein to process along microtubules. Contrastingly, gain of function splice-site variants in the C-terminal, cargo-binding domain of KIF5A are associated with amyotrophic lateral sclerosis (ALS), a neurodegenerative disease involving death of upper and lower motor neurons, ultimately leading to degradation of the motor unit (MU; an alpha motor neuron and all the myofibers it innervates) and death. These ALS-associated variants result in loss of autoinhibition, increased procession of the mutant protein along microtubules, and altered cargo binding. To study the molecular and cellular consequences of ALS-associated variants in vivo, we introduced the murine homolog of an ALS-associated KIF5A variant into C57BL/6 mice using CRISPR-Cas9 gene editing which produced mutant Kif5a mRNA and protein in neuronal tissues of heterozygous (Kif5a+/c.3005+1G>A; HET) and homozygous (Kif5ac.3005+1G>A/c.3005+1G>A; HOM) mice. HET and HOM mice appeared normal in behavioral and electrophysiological (compound muscle action potential [CMAP] and MU number estimation [MUNE]) outcome measures at one year of age. When subjected to sciatic nerve injury, HET and HOM mice have delayed and incomplete recovery of the MUNE compared to wildtype (WT) mice suggesting an impairment in MU repair. Moreover, aged mutant Kif5a mice (aged two years) had reduced MUNE independent of injury, and exacerbation of the delayed and incomplete recovery after injury compared to aged WT mice. These data suggest that ALS-associated variants may result in an impairment of the MU to respond to biological challenges such as injury and aging, leading to a failure of MU repair and maintenance. In this report, we present the behavioral, electrophysiological and pathological characterization of mice harboring an ALS-associated Kif5a variant to understand the functional consequences of KIF5A C-terminal variants in vivo.
    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
    肝细胞肝癌(LIHC)是最常见的肝脏恶性肿瘤之一,具有较高的死亡率和发病率。因此,确定能够准确预测LIHC预后和治疗反应的潜在生物标志物至关重要.驱动蛋白家族成员5A(KIF5A)是一种基于微管的运动蛋白,参与大分子如细胞器蛋白在细胞中的运输。最近的研究表明,KIF5A的高表达与实体瘤的不良预后有关。包括膀胱癌,前列腺癌,和乳腺癌。然而,目前对LIHC中KIF5A表达的临床意义知之甚少。在这里,通过采用多组学生物信息学分析,我们全面揭示了KIF5A在LIHC患者临床特征分层中的潜在功能和预测价值,高KIF5A水平预测不利的临床结果。KIF5A相关网络和富集分析的结果表明,KIF5A可能参与基于微管的过程,抗原加工和通过MHCII类呈递外源肽抗原。此外,免疫浸润和免疫功能分析表明,上调的KIF5A可以预测具有更多CD8+T细胞和更高水平抗肿瘤免疫应答的独特肿瘤微环境.免疫组织化学染色(IHC)提供的证据进一步验证了我们在蛋白质水平上的发现。一起来看,KIF5A可能作为预测免疫治疗反应的新型预后生物标志物,并且可能成为LIHC抗癌策略的潜在靶标。
    Liver hepatocellular carcinoma (LIHC) is one of the most common liver malignancies with high mortality and morbidity. Thus, it is crucial to identify potential biomarker that is capable of accurately predicting the prognosis and therapeutic response of LIHC. Kinesin family member 5A (KIF5A) is a microtubule-based motor protein involved in the transport of macromolecules such as organelle proteins in cells. Recent studies have illustrated that the high expression of KIF5A was related to poor prognosis of solid tumors, including bladder cancer, prostate cancer, and breast cancer. However, little is currently known concerning the clinical significance of KIF5A expression in LIHC. Herein, by adopting multi-omics bioinformatics analysis, we comprehensively uncovered the potential function and the predictive value of KIF5A in stratifying clinical features among patients with LIHC, for which a high KIF5A level predicted an unfavorable clinical outcome. Results from KIF5A-related network and enrichment analyses illustrated that KIF5A might involve in microtubule-based process, antigen processing and presentation of exogenous peptide antigen via MHC class II. Furthermore, immune infiltration and immune function analyses revealed upregulated KIF5A could predict a unique tumor microenvironment with more CD8+T cells and a higher level of anti-tumor immune response. Evidence provided by immunohistochemistry staining (IHC) further validated our findings at the protein level. Taken together, KIF5A might serve as a novel prognostic biomarker for predicting immunotherapy response and could be a potential target for anti-cancer strategies for LIHC.
    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
    人类驱动蛋白家族成员5A(KIF5A)基因的突变最近被确定为肌萎缩侧索硬化症(ALS)的遗传原因。几种KIF5AALS变体引起外显子27跳跃,并预测产生具有改变的C末端尾部的运动蛋白(称为ΔExon27)。然而,潜在的致病机制仍然未知。这里,我们证实了KIF5A突变蛋白在患者iPSC来源的运动神经元中的表达。我们对单分子进行了ΔExon27的综合分析,细胞,和有机体水平。我们的结果表明,ΔExon27易于形成细胞质聚集体,并且具有神经毒性。突变缓解了运动自抑制,增加了运动自联想,导致微管的持续合成能力大大增强。最后,黑腹果蝇中ΔExon27的异位表达会导致机翼缺损,运动障碍,瘫痪过早死亡。我们的结果表明,功能获得是KIF5A相关ALS的潜在疾病机制。
    Mutations in the human kinesin family member 5A (KIF5A) gene were recently identified as a genetic cause of amyotrophic lateral sclerosis (ALS). Several KIF5A ALS variants cause exon 27 skipping and are predicted to produce motor proteins with an altered C-terminal tail (referred to as ΔExon27). However, the underlying pathogenic mechanism is still unknown. Here, we confirm the expression of KIF5A mutant proteins in patient iPSC-derived motor neurons. We perform a comprehensive analysis of ΔExon27 at the single-molecule, cellular, and organism levels. Our results show that ΔExon27 is prone to form cytoplasmic aggregates and is neurotoxic. The mutation relieves motor autoinhibition and increases motor self-association, leading to drastically enhanced processivity on microtubules. Finally, ectopic expression of ΔExon27 in Drosophila melanogaster causes wing defects, motor impairment, paralysis, and premature death. Our results suggest gain-of-function as an underlying disease mechanism in KIF5A-associated ALS.
    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
    KIF5A是驱动蛋白超家族运动蛋白,可在神经元中运输各种货物。Kif5a突变导致家族性肌萎缩侧索硬化(ALS)。这些ALS突变在Kif5a的内含子中,并诱导KIF5AmRNA的错误剪接,导致剪接出外显子27,该外显子在人KIF5A中编码KIF5A的货物结合尾结构域。因此,已经提出ALS是由KIF5A的功能丧失引起的。然而,关于KIF5A突变如何导致ALS的确切机制尚不清楚.这里,我们显示KIF5A的ALS相关突变体,KIF5A(Δexon27),倾向于在培养的小鼠细胞系中形成寡聚体和聚集体。有趣的是,纯化的KIF5A(Δexon27)寡聚体在体外显示出比野生型KIF5A更活跃的微管运动。纯化的KIF5A(Δ外显子27)在体外容易形成聚集体。此外,表达KIF5A(Δexon27)的秀丽隐杆线虫神经元显示形态缺陷。这些数据共同表明,KIF5A的ALS相关突变是毒性功能获得突变,而不是简单的功能丧失突变。
    KIF5A is a kinesin superfamily motor protein that transports various cargos in neurons. Mutations in Kif5a cause familial amyotrophic lateral sclerosis (ALS). These ALS mutations are in the intron of Kif5a and induce mis-splicing of KIF5A mRNA, leading to splicing out of exon 27, which in human KIF5A encodes the cargo-binding tail domain of KIF5A. Therefore, it has been suggested that ALS is caused by loss of function of KIF5A. However, the precise mechanisms regarding how mutations in KIF5A cause ALS remain unclear. Here, we show that an ALS-associated mutant of KIF5A, KIF5A(Δexon27), is predisposed to form oligomers and aggregates in cultured mouse cell lines. Interestingly, purified KIF5A(Δexon27) oligomers showed more active movement on microtubules than wild-type KIF5A in vitro. Purified KIF5A(∆exon27) was prone to form aggregates in vitro. Moreover, KIF5A(Δexon27)-expressing Caenorhabditis elegans neurons showed morphological defects. These data collectively suggest that ALS-associated mutations of KIF5A are toxic gain-of-function mutations rather than simple loss-of-function mutations.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号