Kinesins

驱动蛋白
  • 文章类型: Journal Article
    微管相关运动蛋白KIF1A的突变导致严重的神经系统疾病,称为KIF1A相关神经系统疾病(KAND)。尽管深入了解其分子机制,KIF1A-微管复合物的高分辨率结构仍未定义。这里,我们展示了二聚体微管结合的KIF1A的2.7-3.5µ分辨率结构,包括致病性P305L突变体,跨越各种核苷酸状态。我们的结构显示KIF1A以单和双头结合的构型结合微管,两个头都表现出明显的构象,头间连接紧密。值得注意的是,KIF1A的类特异性环12(K环)与α-和β-微管蛋白的C末端尾巴形成静电相互作用。P305L突变不会破坏这些相互作用,但会改变loop-12的构象,损害强微管结合。结构功能分析揭示了K环和头-头协调是KIF1A超常运动的主要决定因素。我们的发现促进了对KIF1A分子机制的理解,并为开发针对KAND的结构指导疗法提供了基础。
    Mutations in the microtubule-associated motor protein KIF1A lead to severe neurological conditions known as KIF1A-associated neurological disorders (KAND). Despite insights into its molecular mechanism, high-resolution structures of KIF1A-microtubule complexes remain undefined. Here, we present 2.7-3.5 Å resolution structures of dimeric microtubule-bound KIF1A, including the pathogenic P305L mutant, across various nucleotide states. Our structures reveal that KIF1A binds microtubules in one- and two-heads-bound configurations, with both heads exhibiting distinct conformations with tight inter-head connection. Notably, KIF1A\'s class-specific loop 12 (K-loop) forms electrostatic interactions with the C-terminal tails of both α- and β-tubulin. The P305L mutation does not disrupt these interactions but alters loop-12\'s conformation, impairing strong microtubule-binding. Structure-function analysis reveals the K-loop and head-head coordination as major determinants of KIF1A\'s superprocessive motility. Our findings advance the understanding of KIF1A\'s molecular mechanism and provide a basis for developing structure-guided therapeutics against KAND.
    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
    微管蛋白翻译后修饰(PTM)调节微管的动态特性及其与其他蛋白质的相互作用。然而,微管蛋白PTM的作用通常通过修饰酶的缺失或微管蛋白突变体的过表达间接揭示。在这项研究中,我们直接编辑内源性微管蛋白基因座来安装PTM模拟或致残突变,并研究它们对微管稳定性的影响,神经突生长,轴突再生,货物运输,秀丽隐杆线虫触觉受体神经元的感觉功能。我们发现β-微管蛋白S172磷酸化和K252乙酰化状态强烈影响微管动力学,神经突生长,和再生,而α-微管蛋白K40乙酰化作用影响不大。微管蛋白C末端尾部的聚谷氨酸化和去酪氨酸可能通过调节与驱动蛋白13的相互作用而对微管稳定性产生更微妙的影响。总的来说,我们的研究系统地评估和比较了几种微管蛋白PTM对神经元分化和再生的影响,并建立了一个体内平台来测试微管蛋白PTM在神经元中的功能.
    Tubulin posttranslational modifications (PTMs) modulate the dynamic properties of microtubules and their interactions with other proteins. However, the effects of tubulin PTMs were often revealed indirectly through the deletion of modifying enzymes or the overexpression of tubulin mutants. In this study, we directly edited the endogenous tubulin loci to install PTM-mimicking or -disabling mutations and studied their effects on microtubule stability, neurite outgrowth, axonal regeneration, cargo transport, and sensory functions in the touch receptor neurons of Caenorhabditis elegans. We found that the status of β-tubulin S172 phosphorylation and K252 acetylation strongly affected microtubule dynamics, neurite growth, and regeneration, whereas α-tubulin K40 acetylation had little influence. Polyglutamylation and detyrosination in the tubulin C-terminal tail had more subtle effects on microtubule stability likely by modulating the interaction with kinesin-13. Overall, our study systematically assessed and compared several tubulin PTMs for their impacts on neuronal differentiation and regeneration and established an in vivo platform to test the function of tubulin PTMs in neurons.
    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
    目前,研究了许多长链非编码RNA(lncRNA)作为肿瘤抑制因子在宫颈癌(CC)形成和发展中的作用.然而,lncRNA前列腺癌基因表达标志物1(PCGEM1),高表达不仅可加重卵巢癌的发生,还可诱导肿瘤的发生和子宫内膜癌的进展,在CC中没有研究过。本研究的目的是研究PCGEM1在CC中的表达和潜在作用。通过实时PCR检测PCGEM1在CC细胞中的相对表达。shRNA抑制PCGEM1表达后,扩散的变化,迁移,并通过CCK-8测定检测侵袭能力,EdU分析,和集落形成试验伤口愈合试验。通过蛋白质印迹和免疫荧光测定Transwell测定和上皮-间质转化(EMT)标志物的表达变化。PCGEM1,miR-642a-5p,和驱动蛋白家族成员5B(KIF5B)通过生物信息学分析和荧光素酶报告基因测定得到证实。结果显示PCGEM1在CC细胞内表达上调。细胞活力,迁移,shRNA抑制Hela和SiHa细胞中PCGEM1的表达后,侵袭能力明显降低。N-钙黏着蛋白被沉默了,但sh-PCGEM1升高了E-cadherin的表达。此外,通过在CC中使用miR-642a-5p,PCGEM1被证实为调节KIF5B水平的竞争性内源性RNA(ceRNA)。MiR-642a-5p下调部分挽救了sh-PCGEM1对细胞增殖的抑制作用,迁移,入侵,和EMT流程。总之,PCGEM1/miR-642a-5p/KIF5B信号轴可能是CC的新治疗靶点.本研究为CC的靶向治疗提供了研究基础和新方向。
    At present, the role of many long non-coding RNAs (lncRNAs) as tumor suppressors in the formation and development of cervical cancer (CC) has been studied. However, lncRNA prostate cancer gene expression marker 1 (PCGEM1), whose high expression not only aggravates ovarian cancer but also can induce tumorigenesis and endometrial cancer progression, has not been studied in CC. The objective of this study was to investigate the expression and the underlying role of PCGEM1 in CC. The relative expression of PCGEM1 in CC cells was detected by real-time PCR. After the suppression of PCGEM1 expression by shRNA, the changes in the proliferation, migration, and invasion capacities were detected via CCK-8 assay, EdU assay, and colony formation assay wound healing assay. Transwell assay and the changes in expressions of epithelial-to-mesenchymal transition (EMT) markers were determined by western blot and immunofluorescence. The interplay among PCGEM1, miR-642a-5p, and kinesin family member 5B (KIF5B) was confirmed by bioinformatics analyses and luciferase reporter assay. Results showed that PCGEM1 expressions were up-regulated within CC cells. Cell viabilities, migration, and invasion were remarkably reduced after the suppression of PCGEM1 expression by shRNA in Hela and SiHa cells. N-cadherin was silenced, but E-cadherin expression was elevated by sh-PCGEM1. Moreover, by sponging miR-642a-5p in CC, PCGEM1 was verified as a competitive endogenous RNA (ceRNA) that modulates KIF5B levels. MiR-642a-5p down-regulation partially rescued sh-PCGEM1\'s inhibitory effects on cell proliferation, migration, invasion, and EMT process. In conclusion, the PCGEM1/miR-642a-5p/KIF5B signaling axis might be a novel therapeutic target in CC. This study provides a research basis and new direction for targeted therapy of CC.
    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
    开花植物依靠花粉管的极化生长将精子细胞(SC)输送到胚囊进行双重受精。在花粉中,营养核(VN)和两个SC形成雄性胚芽单位(MGU)。然而,MGU定向运输的潜在机制尚不清楚。在这项研究中,我们提供了微管之间动态相互作用的第一张全图,肌动蛋白丝,和MGU在花粉萌发和试管生长过程中。微管的解聚和驱动蛋白活性的抑制导致VN向前和向后运动的速度增加和幅度放大。药理学冲洗实验进一步表明,微管参与协调MGU的定向运动。相比之下,抑制肌动球蛋白系统会导致VN迁移率的速度降低,但不会改变运动模式。此外,详细观察表明,VN的运动方向和速度与肌动球蛋白驱动的VN周围细胞质流的运动方向和速度密切相关。因此,我们提出,虽然基于肌动球蛋白的细胞质流对MGU的振荡运动有影响,微管和驱动蛋白可以避免MGU随着细胞质流漂移,并充当调节MGU在花粉中的正确定位和定向迁移的主要调节剂。
    Flowering plants rely on the polarized growth of pollen tubes to deliver sperm cells (SCs) to the embryo sac for double fertilization. In pollen, the vegetative nucleus (VN) and two SCs form the male germ unit (MGU). However, the mechanism underlying directional transportation of MGU is not well understood. In this study, we provide the first full picture of the dynamic interplay among microtubules, actin filaments, and MGU during pollen germination and tube growth. Depolymerization of microtubules and inhibition of kinesin activity result in an increased velocity and magnified amplitude of VN\'s forward and backward movement. Pharmacological washout experiments further suggest that microtubules participate in coordinating the directional movement of MGU. In contrast, suppression of the actomyosin system leads to a reduced velocity of VN mobility but without a moving pattern change. Moreover, detailed observation shows that the direction and velocity of VN\'s movement are in close correlations with those of the actomyosin-driven cytoplasmic streaming surrounding VN. Therefore, we propose that while actomyosin-based cytoplasmic streaming influences on the oscillational movement of MGU, microtubules and kinesins avoid MGU drifting with the cytoplasmic streaming and act as the major regulator for fine-tuning the proper positioning and directional migration of MGU in pollen.
    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
    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
    背景:驱动蛋白家族成员26B(KIF26B)与多种肿瘤的发生和进展密切相关。然而,关于其在口腔鳞状细胞癌(OSCC)中的作用的研究有限。本文旨在研究KIF26B在OSCC中的表达水平及其机制。
    方法:进行实时定量聚合酶链反应(RT-qPCR)和Westernblot分析,以评估35例OSCC标本及其相应的非癌组织中KIF26B的表达水平。在HSC6和SCC25细胞中实现KIF26B的过表达和沉默,分别,导致KIF26B过表达和si-KIF26B细胞系的建立,指定为KIF26B组和si-KIF26B组。使用5-乙炔基-2'-脱氧尿苷(EdU)标记和克隆形成进行增殖测定,以评估这些组中细胞的增殖能力。使用Transwell测定法评估KIF26B和si-KIF26B组中的细胞的侵入和迁移能力。此外,通过Westernblot分析KIF26B对糖原合成酶激酶(GSK)-3β/β-catenin通路的影响。
    结果:根据RT-qPCR和Westernblot分析的结果,OSCC组织中KIF26B的表达明显高于正常组织(p<0.01)。KIF26B的过表达显著加速了细胞迁移,入侵,和增殖(p<0.01),而KIF26B的敲减显着抑制了这些过程(p<0.01)。此外,KIF26B过表达导致活性β-连环蛋白水平升高,p-GSK-3和c-myc(p<0.01),而KIF26B沉默降低了这些蛋白质的水平(p<0.01)。
    结论:我们的研究结果表明,KIF26B可能作为癌基因在OSCC的发病和进展中发挥作用。本研究为确定OSCC的潜在治疗靶点奠定了基础。
    BACKGROUND: Kinesin family member 26B (KIF26B) has been closely linked to the occurrence and progression of various tumors. However, there is limited research on its role in oral squamous cell carcinoma (OSCC). This article aims to investigate the expression levels and mechanisms of KIF26B in OSCC.
    METHODS: Real time quantity polymerase chain reaction (RT-qPCR) and Western blot analyses were conducted to assess the expression levels of KIF26B in 35 OSCC specimens and their corresponding non-cancerous tissues. Overexpression and silencing of KIF26B were achieved in HSC6 and SCC25 cells, respectively, resulting in the establishment of KIF26B-overexpressing and si-KIF26B cell lines, designated as the KIF26B group and si-KIF26B group. Proliferation assays using 5-Ethynyl-2\'-deoxyuridine (EdU) labeling and clone formation were performed to evaluate the proliferative capacity of cells in these groups. The invasive and migratory abilities of cells in the KIF26B and si-KIF26B groups were assessed using Transwell assay. Additionally, the influence of KIF26B on the glycogen synthase kinase (GSK)-3β/β-catenin pathway was investigated through Western blot analysis.
    RESULTS: According to the results of RT-qPCR and Western blot analyses, the expression of KIF26B was predominantly higher in OSCC tissues compared to normal tissues (p < 0.01). Overexpression of KIF26B notably accelerated cell migration, invasion, and proliferation (p < 0.01), whereas knockdown of KIF26B significantly inhibited these processes (p < 0.01). Additionally, KIF26B overexpression led to increased levels of active β-catenin, p-GSK-3, and c-myc (p < 0.01), while KIF26B silencing decreased the levels of these proteins (p < 0.01).
    CONCLUSIONS: Our findings suggest that KIF26B may play a role in the pathogenesis and progression of OSCC as an oncogene. This study establishes a foundation for the identification of potential therapeutic targets for OSCC.
    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
    内耳的感觉毛细胞利用专门的带状突触将感觉刺激传递到中枢神经系统。这种感觉传递需要快速和持续的神经递质释放,它依赖于毛细胞突触前的大量突触小泡。在神经元中的工作表明,驱动蛋白运动蛋白沿着微管将突触物质运送到突触前,但是新的突触物质如何到达毛细胞的突触前还不清楚。我们表明,驱动蛋白运动蛋白Kif1a和完整的微管网络对于富集毛细胞突触前的突触小泡是必需的。我们使用遗传学和药理学来破坏Kif1a功能并损害斑马鱼侧线系统毛细胞中的微管网络。我们发现这些操作减少了毛细胞突触前的突触-囊泡种群。使用电子显微镜,随着体内钙成像和电生理学,我们表明,突触小泡的供应减少会对带状突触功能产生不利影响。Kif1a突变体表现出自发囊泡释放和诱发的突触后钙反应的显着减少。此外,我们发现kif1a突变体表现出受损的流变倾向,一种依赖于侧线毛细胞对持续流动刺激的反应能力的行为。总的来说,我们的结果表明,基于Kif1a的微管运输是至关重要的,以丰富的突触小泡在毛细胞的活动区,一个对正常的带状突触功能至关重要的过程。
    Kif1amRNA存在于斑马鱼毛细胞中Kif1a的缺失会破坏带状突触中突触小泡的富集微管的破坏会耗尽带状突触中的突触小泡Kif1a突变体的带状突触和感觉系统功能受损。
    Sensory hair cells of the inner ear utilize specialized ribbon synapses to transmit sensory stimuli to the central nervous system. This sensory transmission necessitates rapid and sustained neurotransmitter release, which relies on a large pool of synaptic vesicles at the hair-cell presynapse. Work in neurons has shown that kinesin motor proteins traffic synaptic material along microtubules to the presynapse, but how new synaptic material reaches the presynapse in hair cells is not known. We show that the kinesin motor protein Kif1a and an intact microtubule network are necessary to enrich synaptic vesicles at the presynapse in hair cells. We use genetics and pharmacology to disrupt Kif1a function and impair microtubule networks in hair cells of the zebrafish lateral-line system. We find that these manipulations decrease synaptic-vesicle populations at the presynapse in hair cells. Using electron microscopy, along with in vivo calcium imaging and electrophysiology, we show that a diminished supply of synaptic vesicles adversely affects ribbon-synapse function. Kif1a mutants exhibit dramatic reductions in spontaneous vesicle release and evoked postsynaptic calcium responses. Additionally, we find that kif1a mutants exhibit impaired rheotaxis, a behavior reliant on the ability of hair cells in the lateral line to respond to sustained flow stimuli. Overall, our results demonstrate that Kif1a-based microtubule transport is critical to enrich synaptic vesicles at the active zone in hair cells, a process that is vital for proper ribbon-synapse function.
    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
    非洲裔美国人(AA)女性患三阴性乳腺癌(TNBC)的可能性是欧洲裔女性的两倍。此外,患有TNBC的AA女性比欧美(EA)女性的疾病病程更具侵略性。因此,在患有TNBC的AA患者中,鉴定种族特异性生物标志物和改善生存结局的临床需求尚未得到满足.负端定向微管运动蛋白驱动蛋白家族成员C1(KIFC1)促进中心体聚集和染色体不稳定性,并且通常在TNBC中过表达。先前的发现表明,KIFC1在来自AA的TNBC细胞中的细胞增殖和迁移中起作用,并且在患有TNBC的AA患者中,核KIFC1(nKIFC1)的水平特别高。KIFC1在间期的核定位可能是其先前未被识别的种族特异性关联的基础。在这项研究中,我们发现在来自AAs的TNBC细胞中,nKIFC1在EA细胞上与肿瘤抑制肌球蛋白重链9(MYH9)相互作用。用KIFC1和MYH9的商业抑制剂处理AATNBC细胞破坏了KIFC1和MYH9之间的相互作用。为了表征TNBC中KIFC1-MYH9-MYC轴的种族差异,我们建立了纯合KIFC1敲除(KO)TNBC细胞系。KIFC1KO显著抑制增殖,迁移,在AATNBC细胞中和侵袭,但在EATNBC细胞中没有。RNA测序分析表明,参与细胞迁移的基因显著下调,入侵,与来自EA的TNBC细胞系相比,来自AA的KIFC1KO的转移。这些数据表明,从机制上讲,AA患者中nKIFC1在驱动TNBC进展和转移中的作用强于EA患者,KIFC1可能是AA患者TNBC的关键治疗靶点。
    African American (AA) women are twice as likely to develop triple-negative breast cancer (TNBC) as women of European descent. Additionally, AA women with TNBC present a much more aggressive disease course than their European American (EA) counterparts. Thus, there is an unmet clinical need to identify race-specific biomarkers and improve survival outcomes in AA patients with TNBC. The minus-end directed microtubule motor protein kinesin family member C1 (KIFC1) promotes centrosome clustering and chromosomal instability and is often overexpressed in TNBC. Previous findings suggest that KIFC1 plays a role in cell proliferation and migration in TNBC cells from AAs and that the levels of nuclear KIFC1 (nKIFC1) are particularly high in AA patients with TNBC. The nuclear localization of KIFC1 in interphase may underlie its previously unrecognized race-specific association. In this study, we found that in TNBC cells derived from AAs, nKIFC1 interacted with the tumor suppressor myosin heavy chain 9 (MYH9) over EA cells. Treatment of AA TNBC cells with commercial inhibitors of KIFC1 and MYH9 disrupted the interaction between KIFC1 and MYH9. To characterize the racial differences in the KIFC1-MYH9-MYC axis in TNBC, we established homozygous KIFC1 knockout (KO) TNBC cell lines. KIFC1 KO significantly inhibited proliferation, migration, and invasion in AA TNBC cells but not in EA TNBC cells. RNA sequencing analysis showed significant downregulation of genes involved in cell migration, invasion, and metastasis upon KIFC1 KO in TNBC cell lines from AAs compared to those from EAs. These data indicate that mechanistically, the role of nKIFC1 in driving TNBC progression and metastasis is stronger in AA patients than in EA patients, and that KIFC1 may be a critical therapeutic target for AA patients with TNBC.
    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
    背景:囊泡运输(VT)与肿瘤进展和免疫力有复杂的关系。但是室性心动过速在透明细胞肾细胞癌(ccRCC)中的预后意义尚不清楚。因此,我们旨在根据VT建立预后模型来预测ccRCC患者的总生存期。
    方法:我们使用来自TCGA数据库的患者数据,并通过差异表达分析建立了具有13个VT相关基因(VTRGs)的预后模型,LASSO回归,和单变量/多变量Cox分析。该模型进行了内部和外部验证,生存分析和ROC曲线显示出优异的预测能力。此外,更高的模型风险评分对应于更晚期的肿瘤进展.为了进一步了解患者不同预后的潜在原因,我们对该模型在风险组中鉴定的差异表达基因进行了富集分析.通过qRT-PCR和细胞功能实验验证了SAA1和KIF18B在ccRCC中的表达水平和作用。
    结果:体液免疫反应和cAMP信号通路可能是导致预后不良的生物学过程和通路。对免疫微环境的进一步分析表明,预后不良的ccRCC患者具有高度的免疫浸润特征。我们比较了GDSC数据库中来自不同预后患者的样品的药物反应数据,并确定了与预后相关的药物敏感性差异。最后,我们证明了SAA1和KIF18B可以增加增殖,迁移,细胞实验和ccRCC细胞的侵袭能力。
    结论:总之,我们进一步揭示了VTRGs在ccRCC预后中的重要性。
    BACKGROUND: Vesicular transport (VT) has a complex relationship with tumor progression and immunity. But prognostic significance of VT in clear cell renal cell carcinoma (ccRCC) is unclear. Thus, we aimed to establish a prognostic model according to VT to predict overall survival of ccRCC patients.
    METHODS: We used patient data from TCGA database and built a prognostic model with 13 VT-related genes (VTRGs) by differential expression analysis, LASSO regression, and univariate/multivariate Cox analysis. The model was validated internally and externally, and survival analysis and ROC curves depicted excellent predictive ability. Furthermore, higher modeled riskscores corresponded to more advanced tumor progression. To further understand the potential reasons for different prognoses in patients, we did enrichment analysis on differentially expressed genes identified by the model in risk groups. The expression levels and roles of SAA1 and KIF18B in ccRCC were verified by qRT-PCR and cell function experiments.
    RESULTS: Humoral immune response and cAMP signaling pathway may be the biological processes and pathways leading to poor prognosis. Further analysis of immune microenvironment presented that ccRCC patients with poor prognoses had highly immune-infiltrated characteristics. We compared the drug response data of samples from different prognostic patients in the GDSC database and identified drug sensitivity differences associated with prognosis. Finally, we demonstrated that SAA1 and KIF18B could increase the proliferation, migration, and invasion ability of ccRCC cells using cellular experiments.
    CONCLUSIONS: In summary, we further revealed the importance of VTRGs in ccRCC prognosis.
    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
    产生血小板的巨核细胞(MK)主要存在于骨髓中,它们在每个细胞周期中复制其DNA含量,从而产生具有复杂分界膜系统的多倍体细胞。虽然已经确定了前血小板形成过程中细胞骨架重组的关键要素,什么启动血小板释放到血管正弦仍然很大程度上难以捉摸。使用细胞周期指示器,我们观察到一个独特的现象,在此期间,MK中扩增的中心体经历有丝分裂后的聚类,紧随其后的是前血小板形成,仅发生在相间的G1中。在短期饥饿小鼠后,G1期的强制细胞周期停滞不仅在体外而且在体内都增加了前血小板的形成。我们发现抑制中心体蛋白驱动蛋白家族成员C1(KIFC1)损害了聚集和随后的前血小板形成,而KIFC1缺陷小鼠表现出血小板计数减少。总之,我们确定KIFC1-和细胞周期介导的中心体聚集是MK形成前血小板的重要引发剂。
    Platelet-producing megakaryocytes (MKs) primarily reside in the bone marrow, where they duplicate their DNA content with each cell cycle resulting in polyploid cells with an intricate demarcation membrane system. While key elements of the cytoskeletal reorganizations during proplatelet formation have been identified, what initiates the release of platelets into vessel sinusoids remains largely elusive. Using a cell cycle indicator, we observed a unique phenomenon, during which amplified centrosomes in MKs underwent clustering following mitosis, closely followed by proplatelet formation, which exclusively occurred in G1 of interphase. Forced cell cycle arrest in G1 increased proplatelet formation not only in vitro but also in vivo following short-term starvation of mice. We identified that inhibition of the centrosomal protein kinesin family member C1 (KIFC1) impaired clustering and subsequent proplatelet formation, while KIFC1-deficient mice exhibited reduced platelet counts. In summary, we identified KIFC1- and cell cycle-mediated centrosome clustering as an important initiator of proplatelet formation from MKs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号