Active Transport, Cell Nucleus

主动转运, 细胞核
  • 文章类型: Journal Article
    蛋白质和信使RNA的核质转运分析一直是先进的微观方法的重点。最近,有可能使用电子显微镜和光学显微镜来识别和观察通过核孔复合体的单个前核糖体颗粒.在这次审查中,我们专注于核糖体前颗粒在细胞核中的运输,以及它们通过毛孔的方式。
    The analysis of nucleocytoplasmic transport of proteins and messenger RNA has been the focus of advanced microscopic approaches. Recently, it has been possible to identify and visualize individual pre-ribosomal particles on their way through the nuclear pore complex using both electron and light microscopy. In this review, we focused on the transport of pre-ribosomal particles in the nucleus on their way to and through the pores.
    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
    人类呼吸道病毒是人类最普遍的疾病原因,高传染性RSV是婴儿细支气管炎和病毒性肺炎的主要原因。对I型IFN的反应是针对病毒感染的主要防御。然而,RSV蛋白已被证明拮抗I型IFN介导的抗病毒先天免疫,特异性抑制细胞内IFN信号传导。呼吸道上皮细胞是RSV感染的主要目标。在这项研究中,我们发现RSV-NS1干扰了上皮细胞的IFN-αJAK/STAT信号通路。RSV-NS1表达显着增强IFN-α介导的STAT1磷酸化,但不增强pSTAT2;RSV-NS1对STAT1和STAT2总蛋白水平均无影响。然而,RSV-NS1的表达显著降低ISRE和GAS启动子活性和抗病毒IRG表达。进一步的机制研究表明RSV-NS1结合STAT1,蛋白质模型表明STAT1和RSV-NS1之间可能的相互作用位点。STAT1的核易位在RSV-NS1存在下减少。此外,STAT1与核运输衔接蛋白的相互作用,KPNA1,也减少了,提示RSV阻断STAT1核易位的机制。的确,减少STAT1进入细胞核可能解释RSV抑制IFNJAK/STAT启动子激活和抗病毒基因诱导。总之,这些结果描述了RSV控制抗病毒IFN-αJAK/STAT应答的新机制,这增强了我们对RSV呼吸道疾病进展的理解。
    Human respiratory viruses are the most prevalent cause of disease in humans, with the highly infectious RSV being the leading cause of infant bronchiolitis and viral pneumonia. Responses to type I IFNs are the primary defense against viral infection. However, RSV proteins have been shown to antagonize type I IFN-mediated antiviral innate immunity, specifically dampening intracellular IFN signaling. Respiratory epithelial cells are the main target for RSV infection. In this study, we found RSV-NS1 interfered with the IFN-α JAK/STAT signaling pathway of epithelial cells. RSV-NS1 expression significantly enhanced IFN-α-mediated phosphorylation of STAT1, but not pSTAT2; and neither STAT1 nor STAT2 total protein levels were affected by RSV-NS1. However, expression of RSV-NS1 significantly reduced ISRE and GAS promoter activity and anti-viral IRG expression. Further mechanistic studies demonstrated RSV-NS1 bound STAT1, with protein modeling indicating a possible interaction site between STAT1 and RSV-NS1. Nuclear translocation of STAT1 was reduced in the presence of RSV-NS1. Additionally, STAT1\'s interaction with the nuclear transport adapter protein, KPNA1, was also reduced, suggesting a mechanism by which RSV blocks STAT1 nuclear translocation. Indeed, reducing STAT1\'s access to the nucleus may explain RSV\'s suppression of IFN JAK/STAT promoter activation and antiviral gene induction. Taken together these results describe a novel mechanism by which RSV controls antiviral IFN-α JAK/STAT responses, which enhances our understanding of RSV\'s respiratory disease progression.
    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
    异常蛋白质的聚集是神经变性如聚谷氨酰胺(polyQ)和其他重复扩增疾病中的常见病理标志。通过在果蝇肠肠细胞中过表达ataxin3C末端polyQ扩增,我们建立了脊髓小脑性共济失调类型3(SCA3)的肠梗阻模型,并报道了核相关内体(NAEs)的新作用-将polyQ递送至核质.在这个模型中,伴随着显著增加的RAB5阳性NAEs是富含polyQ的大量核质网,异常的核包膜内陷,内质网显著减少,表明功能失调的核质运输和受损的内膜组织。始终如一,Rab5而不是Rab7RNAi进一步降低polyQ相关NAE,抑制内膜解体,缓解疾病模型。有趣的是,自噬蛋白在与polyQ相关的NAEs中富集,并发挥非经典自噬作用,因为自噬分子的遗传操作对NAEs和SCA3毒性表现出不同的影响。即,Atg1或Atg12的下调减轻,而Atg5RNAi加重果蝇肠和复眼的疾病表型。我们的发现,因此,提供新的机制见解,并强调以内体为中心的核质运输和稳态内膜分配在polyQ疾病发病机理中的基本作用。
    Aggregation of aberrant proteins is a common pathological hallmark in neurodegeneration such as polyglutamine (polyQ) and other repeat-expansion diseases. Here through overexpression of ataxin3 C-terminal polyQ expansion in Drosophila gut enterocytes, we generated an intestinal obstruction model of spinocerebellar ataxia type3 (SCA3) and reported a new role of nuclear-associated endosomes (NAEs)-the delivery of polyQ to the nucleoplasm. In this model, accompanied by the prominently increased RAB5-positive NAEs are abundant nucleoplasmic reticulum enriched with polyQ, abnormal nuclear envelope invagination, significantly reduced endoplasmic reticulum, indicating dysfunctional nucleocytoplasmic trafficking and impaired endomembrane organization. Consistently, Rab5 but not Rab7 RNAi further decreased polyQ-related NAEs, inhibited endomembrane disorganization, and alleviated disease model. Interestingly, autophagic proteins were enriched in polyQ-related NAEs and played non-canonical autophagic roles as genetic manipulation of autophagic molecules exhibited differential impacts on NAEs and SCA3 toxicity. Namely, the down-regulation of Atg1 or Atg12 mitigated while Atg5 RNAi aggravated the disease phenotypes both in Drosophila intestines and compound eyes. Our findings, therefore, provide new mechanistic insights and underscore the fundamental roles of endosome-centered nucleocytoplasmic trafficking and homeostatic endomembrane allocation in the pathogenesis of polyQ 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
    Wnt/无翼(Wg)信号在发育和疾病中至关重要,包括癌症.典型的Wnt信号是由β-catenin/Armadillo(果蝇中的手臂)介导的,用IFT-A/Kinesin2复合物促进β-catenin/Arm的核易位。这里,我们证明了保守的小N末端Arm34-87/β-catenin肽与IFT140结合,作为减弱体内Wg/Wnt信号传导的主要干扰工具。Arm34-87表达拮抗内源性Wnt/Wg信号,导致其靶表达的减少。Arm34-87通过干扰内源性Arm/β-catenin的核易位抑制Wg/Wnt信号,并且这可以通过野生型β-连环蛋白或IFT140的水平来调节,其中Arm34-87效应被增强或抑制。重要的是,这种机制在哺乳动物中保守,等价的β-catenin24-79肽阻断核易位和途径激活,包括癌细胞。我们的工作表明,Wnt信号传导可以由确定的N末端β-联蛋白肽调节,因此可能作为治疗应用的切入点以减弱Wnt/β-联蛋白信号传导。
    Wnt/Wingless (Wg) signaling is critical in development and disease, including cancer. Canonical Wnt signaling is mediated by β-catenin/Armadillo (Arm in Drosophila) transducing signals to the nucleus, with IFT-A/Kinesin 2 complexes promoting nuclear translocation of β-catenin/Arm. Here, we demonstrate that a conserved small N-terminal Arm34-87/β-catenin peptide binds to IFT140, acting as a dominant interference tool to attenuate Wg/Wnt signaling in vivo. Arm34-87 expression antagonizes endogenous Wnt/Wg signaling, resulting in the reduction of its target expression. Arm34-87 inhibits Wg/Wnt signaling by interfering with nuclear translocation of endogenous Arm/β-catenin, and this can be modulated by levels of wild-type β-catenin or IFT140, with the Arm34-87 effect being enhanced or suppressed. Importantly, this mechanism is conserved in mammals with the equivalent β-catenin24-79 peptide blocking nuclear translocation and pathway activation, including in cancer cells. Our work indicates that Wnt signaling can be regulated by a defined N-terminal β-catenin peptide and thus might serve as an entry point for therapeutic applications to attenuate Wnt/β-catenin signaling.
    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

    求助全文

  • 文章类型: Journal Article
    由于细胞生理学介导的GTP可用性波动,核质转运(NCT)速率如何变化尚不清楚。在这个问题上,斯科特等人。(https://doi.org/10.1083/jcb.202308152)证明细胞迁移,传播,和核细胞骨架偶联影响GTP水平,从而调节NCT,RNA导出,和蛋白质合成。
    How nucleocytoplasmic transport (NCT) rates change due to cellular physiology-mediated fluctuations in GTP availability remains unclear. In this issue, Scott et al. (https://doi.org/10.1083/jcb.202308152) demonstrate that cell migration, spreading, and nucleocytoskeletal coupling impact GTP levels, thereby regulating NCT, RNA export, and protein synthesis.
    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
    信号识别颗粒(SRP)对于调节细胞内蛋白质的运输和分泌至关重要。具有高SRP9表达的肿瘤患者倾向于具有较差的总体存活率。然而,据我们所知,尚无报道描述SRP9定位与胰腺癌预后之间的关系。因此,本研究旨在探讨这种关系。使用未术前化疗或放疗的胰腺癌手术病例的切除标本对SRP9进行免疫组织化学染色显示,在某些情况下,SRP9优先在癌区的细胞核中表达,在其他情况下几乎没有发现,表明在前者中SRP9被转运到细胞核。比较SRP9核易位患者的预后,患者分为两组:核移位率>50%的患者和核移位率≤50%的患者.>50%组核转位率显著优于≤50%组核转位率(P=0.037)。随后进行了体外实验;特别是,在氨基酸缺乏的条件下,SRP9的核易位率降低,这表明这一现象涉及多种因素。为了进一步研究SRP9核易位的功能,通过将SRP9剪接变体(v1和v2)及其缺失C末端区域的缺失突变体引入MiaPaCa胰腺癌细胞进行体外实验。结果表明,无论C端缺失如何,两个剪接变体都显示出核易位,建议N端区域的作用。鉴于SRP9是一种RNA结合蛋白,RNA免疫沉淀的研究表明,参与癌症进展和蛋白质翻译的信号通路在核转位的v1和v2中下调。毫无疑问,对SRP9核易位的进一步研究将为优化胰腺癌的精确评估和治疗控制开辟一条途径.
    Signal recognition particles (SRPs) are essential for regulating intracellular protein transport and secretion. Patients with tumors with high SRP9 expression tend to have a poorer overall survival. However, to the best of our knowledge, no reports have described the relationship between SRP9 localization and prognosis in pancreatic cancer. Thus, the present study aimed to investigate this relationship. Immunohistochemical staining for SRP9 using excised specimens from pancreatic cancer surgery cases without preoperative chemotherapy or radiotherapy showed that SRP9 was preferentially expressed in the nucleus of the cancerous regions in some cases, which was hardly detected in other cases, indicating that SRP9 was transported to the nucleus in the former cases. To compare the prognosis of patients with SRP9 nuclear translocation, patients were divided into two groups: Those with a nuclear translocation rate of >50% and those with a nuclear translocation rate of ≤50%. The nuclear translocation rate of >50% group had a significantly better recurrence‑free survival than the nuclear translocation rate of ≤50% group (P=0.037). Subsequent in vitro experiments were conducted; notably, the nuclear translocation rate of SRP9 was reduced under amino acid‑deficient conditions, suggesting that multiple factors are involved in this phenomenon. To further study the function of SRP9 nuclear translocation, in vitro experiments were performed by introducing SRP9 splicing variants (v1 and v2) and their deletion mutants lacking C‑terminal regions into MiaPaCa pancreatic cancer cells. The results demonstrated that both splicing variants showed nuclear translocation regardless of the C‑terminal deletions, suggesting the role of the N‑terminal regions. Given that SRP9 is an RNA‑binding protein, the study of RNA immunoprecipitation revealed that signaling pathways involved in cancer progression and protein translation were downregulated in nuclear‑translocated v1 and v2. Undoubtedly, further studies of the nuclear translocation of SRP9 will open an avenue to optimize the precise evaluation and therapeutic control of pancreatic cancer.
    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
    细胞分裂对真核细胞提出了挑战:染色体如何在膜核隔室的范围内有效地分离?不同的生物体通过调节核隔室的程度而进化出了不同的解决方案,从完整的核包络线破裂到通过核包络线扩展完全维持核分隔。在这些极端之间存在许多中间形式,表明细胞分裂过程中的核动力学是惊人的可塑性。在这次审查中,我们强调了核分裂的进化多样性,集中在两个定义特征:(1)染色体区隔和(2)核质运输。Further,我们强调了最近的证据,即分裂过程中的核行为可以在同一生物体的不同细胞环境中变化。在生物体内和生物体之间观察到的差异强调了根据特定环境和细胞要求量身定制的核分裂的动态进化。对不同核分裂的深入调查将增进我们对核子的认识,生理和病理状态。
    Cell division presents a challenge for eukaryotic cells: how can chromosomes effectively segregate within the confines of a membranous nuclear compartment? Different organisms have evolved diverse solutions by modulating the degree of nuclear compartmentalization, ranging from complete nuclear envelope breakdown to complete maintenance of nuclear compartmentalization via nuclear envelope expansion. Many intermediate forms exist between these extremes, suggesting that nuclear dynamics during cell division are surprisingly plastic. In this review, we highlight the evolutionary diversity of nuclear divisions, focusing on two defining characteristics: (1) chromosome compartmentalization and (2) nucleocytoplasmic transport. Further, we highlight recent evidence that nuclear behavior during division can vary within different cellular contexts in the same organism. The variation observed within and between organisms underscores the dynamic evolution of nuclear divisions tailored to specific contexts and cellular requirements. In-depth investigation of diverse nuclear divisions will enhance our understanding of the nucleus, both in physiological and pathological states.
    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
    RNA稳定性的模型已经经历了转化性转变,并揭示了细胞质加帽活性,这意味着转录物的子集被其核对应物自主地重新覆盖。本研究证明了mRNA加帽酶(CE,也称为RNA鸟苷酸转移酶和5'-磷酸酶;RNGTT),传统上公认的核本地化和功能,阐明其对细胞质加帽活性的贡献。CE中独特的核输出序列介导CE的XPO1依赖性核输出。值得注意的是,在亚砷酸钠诱导的氧化应激过程中,细胞质CE(cCE)聚集在应激颗粒(SGs)内。通过涉及分子对接和随后的免疫共沉淀的综合方法,我们识别eIF3b,SGs的组成部分,作为CE的互动伙伴,这意味着它在引导cCE到SGS方面具有潜在的作用。我们测量了来自非应激U2OS细胞的特定mRNA转录物的帽状态,压力和从压力中恢复,这表明cCE靶转录物在应激期间失去了帽,但在恢复阶段显着恢复了帽稳定性。因此,这项全面的研究揭示了细胞质CE的一个新方面,通过维持靶mRNA的帽稳态来促进细胞从应激中恢复。
    The model of RNA stability has undergone a transformative shift with the revelation of a cytoplasmic capping activity that means a subset of transcripts are recapped autonomously of their nuclear counterparts. The present study demonstrates nucleo-cytoplasmic shuttling of the mRNA-capping enzyme (CE, also known as RNA guanylyltransferase and 5\'-phosphatase; RNGTT), traditionally acknowledged for its nuclear localization and functions, elucidating its contribution to cytoplasmic capping activities. A unique nuclear export sequence in CE mediates XPO1-dependent nuclear export of CE. Notably, during sodium arsenite-induced oxidative stress, cytoplasmic CE (cCE) congregates within stress granules (SGs). Through an integrated approach involving molecular docking and subsequent co-immunoprecipitation, we identify eIF3b, a constituent of SGs, as an interactive associate of CE, implying that it has a potential role in guiding cCE to SGs. We measured the cap status of specific mRNA transcripts from U2OS cells that were non-stressed, stressed and recovered from stress, which indicated that cCE-target transcripts lost their caps during stress but remarkably regained cap stability during the recovery phase. This comprehensive study thus uncovers a novel facet of cytoplasmic CE, which facilitates cellular recovery from stress by maintaining cap homeostasis of target mRNAs.
    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
    环状RNA(circularRNAs)在神经发生期间上调。circRNAs在哪里以及如何定位,以及它们在此过程中扮演的角色仍然难以捉摸。比较H9细胞和H9衍生的前脑(FB)神经元之间的核和细胞质circRNAs,我们发现,富含腺苷(A)的circRNAs的一个子集被限制在H9细胞核中,但在分化后输出到细胞溶胶中。circRNAs的这种亚细胞重新定位由poly(A)结合蛋白PABPC1调节。在H9核中,新产生的富含(A)的circRNAs被PABPC1结合并被核篮蛋白TPR捕获以阻止它们的输出。调节circRNAs中富含(A)的基序改变了它们的亚细胞定位,并且在H9胞质中引入富含(A)的circRNAs导致mRNA翻译抑制。此外,神经元分化后减少的核PABPC1能够输出富含(A)的circRNAs,包括circRTN4(2,3),这是神经突生长所必需的。这些发现揭示了circRNAs的亚细胞定位特征,将它们在神经发生过程中的加工和功能联系起来。
    Circular RNAs (circRNAs) are upregulated during neurogenesis. Where and how circRNAs are localized and what roles they play during this process have remained elusive. Comparing the nuclear and cytoplasmic circRNAs between H9 cells and H9-derived forebrain (FB) neurons, we identify that a subset of adenosine (A)-rich circRNAs are restricted in H9 nuclei but exported to cytosols upon differentiation. Such a subcellular relocation of circRNAs is modulated by the poly(A)-binding protein PABPC1. In the H9 nucleus, newly produced (A)-rich circRNAs are bound by PABPC1 and trapped by the nuclear basket protein TPR to prevent their export. Modulating (A)-rich motifs in circRNAs alters their subcellular localization, and introducing (A)-rich circRNAs in H9 cytosols results in mRNA translation suppression. Moreover, decreased nuclear PABPC1 upon neuronal differentiation enables the export of (A)-rich circRNAs, including circRTN4(2,3), which is required for neurite outgrowth. These findings uncover subcellular localization features of circRNAs, linking their processing and function during neurogenesis.
    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