nuclear export

核出口
  • 文章类型: Journal Article
    MITF,一个基本的螺旋环螺旋拉链(bHLHZip)转录因子,在黑素细胞发育中起着至关重要的作用,并作为癌基因发挥作用。我们在小鼠中对Mitf相关色素沉着表型的抑制因子进行了遗传筛选,并鉴定了在K316SUMO化位点终止MITF的基因内Mitf突变,导致C端固有无序区(IDR)的丢失。所得的蛋白质比野生型MITF具有更大的核但稳定性较差,并保留了DNA结合能力。作为二聚体,它可以将野生型和突变型MITF伴侣转移到细胞核中,提高自身的稳定性,从而确保核MITF的供应。smFRET分析显示K316SUMO化和S409磷酸化位点在单体之间的相互作用;这些相互作用在很大程度上解释了观察到的效果。MITF中复发性黑色素瘤相关E318K突变,这影响了K316的磺酰化,还改变与S409一致的蛋白质调节。这表明MITF的残基K316和S409受到SUMO化和磷酸化的影响,分别,通过构象变化对核定位和稳定性的中介作用。我们的工作提供了一种新的遗传抑制机制,以及一个明显有害的突变如何导致正常表型的例子。
    MITF, a basic Helix-Loop-Helix Zipper (bHLHZip) transcription factor, plays vital roles in melanocyte development and functions as an oncogene. We perform a genetic screen for suppressors of the Mitf-associated pigmentation phenotype in mice and identify an intragenic Mitf mutation that terminates MITF at the K316 SUMOylation site, leading to loss of the C-end intrinsically disordered region (IDR). The resulting protein is more nuclear but less stable than wild-type MITF and retains DNA-binding ability. As a dimer, it can translocate wild-type and mutant MITF partners into the nucleus, improving its own stability thus ensuring nuclear MITF supply. smFRET analysis shows interactions between K316 SUMOylation and S409 phosphorylation sites across monomers; these interactions largely explain the observed effects. The recurrent melanoma-associated E318K mutation in MITF, which affects K316 SUMOylation, also alters protein regulation in concert with S409. This suggests that residues K316 and S409 of MITF are impacted by SUMOylation and phosphorylation, respectively, mediating effects on nuclear localization and stability through conformational changes. Our work provides a novel mechanism of genetic suppression, and an example of how apparently deleterious mutations lead to normal phenotypes.
    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
    染色体区域维持1(CRM1),也被称为出口1(XPO1),是一种蛋白质,对于蛋白质和RNA通过核孔复合物转运到细胞质至关重要。小分子抑制剂对CRM1的抑制作用目前正在许多癌症中进行研究。包括白血病,实体器官恶性肿瘤和脑肿瘤。我们回顾了CRM1的结构,它在核出口中的作用,目前CRM1抑制剂的可用性,以及CRM1在许多不同的细胞过程中的作用。对CRM1如何在核输出以及其他细胞过程中发挥作用的更深入的了解可能允许开发其他新型CRM1抑制剂。
    Chromosome Region Maintenance 1 (CRM1), also known as Exportin 1 (XPO1), is a protein that is critical for transport of proteins and RNA to the cytoplasm through the nuclear pore complex. CRM1 inhibition with small molecule inhibitors is currently being studied in many cancers, including leukemias, solid organ malignancies and brain tumors. We review the structure of CRM1, its role in nuclear export, the current availability of CRM1 inhibitors, and the role of CRM1 in a number of distinct cellular processes. A deeper understanding of how CRM1 functions in nuclear export as well as other cellular processes may allow for the development of additional novel CRM1 inhibitors.
    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:DEAD-boxRNA解旋酶19A(DDX19A)在宫颈鳞状细胞癌中过度表达。然而,其在胃癌中的作用尚不清楚。本研究旨在探讨DDX19A在胃癌发生发展中的作用及机制。
    方法:通过定量聚合酶链反应评估DDX19A在胃癌和癌旁组织中的表达,西方印迹,和免疫组织化学染色。用CCK8、平板集落形成、和Transwell迁移分析。用免疫印迹法研究了DDX19A在胃癌细胞中的具体作用机制,RNA结合蛋白免疫沉淀,mRNA半衰期检测,和核和细胞质RNA分离。
    结果:DDX19A在胃癌中高表达,与胃癌的恶性临床病理特征和不良预后呈正相关。此外,DDX19A促进胃癌细胞增殖,迁移,和上皮-间质转化表型。机械上,DDX19A通过上调磷脂酰肌醇-3-激酶(PIK3CA)表达激活PI3K/AKT途径。此外,DDX19A与PIK3CAmRNA相互作用,稳定了它,并促进了它从原子核的出口。
    结论:我们的研究揭示了一种新的机制,DDX19A通过增强PIK3CAmRNA的稳定性和核输出来促进胃癌细胞的增殖和迁移,从而激活PI3K/AKT途径。
    BACKGROUND: DEAD-box RNA helicase 19 A (DDX19A) is overexpressed in cervical squamous cell carcinoma. However, its role in gastric cancer remains unclear. The present study aimed to explore the role and underlying mechanism of DDX19A in the development of gastric cancer.
    METHODS: The expression of DDX19A in gastric cancer and paracancerous tissues was evaluated through quantitative polymerase chain reaction, western blotting, and immunohistochemical staining. The biological functions of DDX19A in gastric cancer were determined using CCK8, plate colony-forming, and Transwell migration assays. The specific mechanism of DDX19A in gastric cancer cells was studied using western blotting, RNA-binding protein immunoprecipitation, mRNA half-life detection, and nuclear and cytoplasmic RNA isolation.
    RESULTS: DDX19A was highly expressed in gastric cancer and positively associated with malignant clinicopathological features and poor prognosis. Additionally, DDX19A promoted gastric cancer cell proliferation, migration, and epithelial-mesenchymal transition phenotypes. Mechanistically, DDX19A activated the PI3K/AKT pathway by upregulating phosphatidylinositol-3-kinase (PIK3CA) expression. Furthermore, DDX19A interacted with PIK3CA mRNA, stabilized it, and facilitated its export from the nucleus.
    CONCLUSIONS: Our study reveals a novel mechanism whereby DDX19A promotes the proliferation and migration of gastric cancer cells by enhancing the stability and nuclear export of PIK3CA mRNA, thereby activating the PI3K/AKT pathway.
    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
    病毒核糖核蛋白(vRNP)的核输出是甲型流感病毒(IAV)生命周期中的关键步骤,可能是开发抗IAV药物的有效靶标。已知宿主因子ras相关核蛋白(RAN)参与几种病毒的生命周期,但其在流感病毒复制中的作用尚不清楚。在本研究中,我们的目的是使用不同的细胞系和亚型毒株确定RAN在流感病毒复制中的功能。我们发现RAN对于vRNP的核出口至关重要,因为它增强了XPO1对病毒核输出蛋白NS2的结合亲和力。RAN的耗尽限制了细胞核中的vRNP复合物并减弱了各种亚型流感病毒的复制。使用硅化合物筛选,我们确定bepotastine可以解离RAN-XPO1-vRNP三聚体复合物,并在体外和体内表现出对流感病毒的有效抗病毒活性。这项研究证明了RAN在IAV复制中的重要作用,并表明其作为抗病毒剂的潜在用途。
    Nuclear export of the viral ribonucleoprotein (vRNP) is a critical step in the influenza A virus (IAV) life cycle and may be an effective target for the development of anti-IAV drugs. The host factor ras-related nuclear protein (RAN) is known to participate in the life cycle of several viruses, but its role in influenza virus replication remains unknown. In the present study, we aimed to determine the function of RAN in influenza virus replication using different cell lines and subtype strains. We found that RAN is essential for the nuclear export of vRNP, as it enhances the binding affinity of XPO1 toward the viral nuclear export protein NS2. Depletion of RAN constrained the vRNP complex in the nucleus and attenuated the replication of various subtypes of influenza virus. Using in silico compound screening, we identified that bepotastine could dissociate the RAN-XPO1-vRNP trimeric complex and exhibit potent antiviral activity against influenza virus both in vitro and in vivo. This study demonstrates the important role of RAN in IAV replication and suggests its potential use as an antiviral target.
    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
    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
    剖析控制哺乳动物转录本从产生到降解的调控机制需要定量测量穿过细胞的mRNA流量。我们开发了亚细胞TimeLapse-seq来测量RNA从染色质中释放的速率,从原子核导出,加载到多染色体上,并在人和小鼠细胞的细胞核和细胞质内降解。这些比率变化很大,然而,来自具有相关功能或被相同转录因子和RNA结合蛋白靶向的基因的转录物以相似的动力学流过亚细胞区室。验证这些关联发现了DDX3X与核出口之间的联系。对于数百个RNA代谢基因,大多数保留有内含子的转录本被核外泌体降解,而其余分子则以稳定的细胞质寿命输出。在染色质上停留更长时间的转录本有延伸的poly(A)尾巴,而细胞质mRNA则相反。最后,机器学习确定了预测mRNA不同生命周期的分子特征。
    Dissecting the regulatory mechanisms controlling mammalian transcripts from production to degradation requires quantitative measurements of mRNA flow across the cell. We developed subcellular TimeLapse-seq to measure the rates at which RNAs are released from chromatin, exported from the nucleus, loaded onto polysomes, and degraded within the nucleus and cytoplasm in human and mouse cells. These rates varied substantially, yet transcripts from genes with related functions or targeted by the same transcription factors and RNA-binding proteins flowed across subcellular compartments with similar kinetics. Verifying these associations uncovered a link between DDX3X and nuclear export. For hundreds of RNA metabolism genes, most transcripts with retained introns were degraded by the nuclear exosome, while the remaining molecules were exported with stable cytoplasmic lifespans. Transcripts residing on chromatin for longer had extended poly(A) tails, whereas the reverse was observed for cytoplasmic mRNAs. Finally, machine learning identified molecular features that predicted the diverse life cycles of mRNAs.
    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
    早幼粒细胞白血病蛋白(PML),一种肿瘤抑制蛋白,在细胞周期调控中起关键作用,凋亡,衰老和细胞代谢。这里,我们报道PML促进细胞凋亡和铁凋亡。我们的数据显示PML过表达抑制细胞增殖和迁移。PML过表达增加凋亡细胞,核聚变和线粒体膜电位的丧失,伴随着Bcl-2家族蛋白和活性氧(ROS)水平的调节,提示PML细胞凋亡增强。同时,PML过表达不仅增加了脂质ROS积累和丙二醛(MDA)含量,而且下调了溶质载体家族7成员11(SLC7A11)和谷胱甘肽过氧化物酶4(GPX4)的表达,表明PML增强了铁凋亡。此外,p53的敲除减弱了PML对SLC7A11和GPX4的影响,并通过PML过表达抑制了脂质ROS和ROS的增加。此外,PML从细胞核到细胞质的易位不仅促进细胞凋亡和铁凋亡,而且还抑制细胞增殖。一起来看,PML促进细胞凋亡和铁凋亡,其中p53的介导和PML的核输出起着重要作用。
    Promyelocytic leukemia protein (PML), a tumor suppressor protein, plays a key role in cell cycle regulation, apoptosis, senescence and cellular metabolism. Here, we report that PML promotes apoptosis and ferroptosis. Our data showed that PML over-expression inhibited cell proliferation and migration. PML over-expression increased apoptotic cells, nuclear condensation and the loss of mitochondrial membrane potential, accompanied by regulation of Bcl-2 family proteins and reactive oxygen species (ROS) level, suggesting that PML enhanced apoptosis. Meanwhile, PML over-expression not only increased lipid ROS accumulation and Malondialdehyde (MDA) content but also downregulated solute carrier family 7 member 11 (SLC7A11) and glutathione peroxidase 4 (GPX4) expression, indicating that PML enhanced ferroptosis. Additionally, knockdown of p53 attenuated the effect of PML on SLC7A11 and GPX4, and inhibited the increase of lipid ROS and ROS by PML over-expression. Moreover, translocation of PML from nucleus to cytoplasm not only promoted apoptosis and ferroptosis, but also inhibited cell proliferation. Taken together, PML promotes apoptosis and ferroptosis, in which the mediation of p53 and the nuclear export of PML play important roles.
    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
    核孔复合物(NPC)调节分子从细胞核的进出。小分子通过扩散通过NPCs,而大分子通过核动力蛋白进入和离开细胞核,作为运输因素。Exportin-1(XPO1)是一种蛋白质,它是核蛋白家族的重要成员,并将大分子从细胞核携带到细胞质。XPO1负责蛋白质的核-细胞质运输,核糖体RNA和核糖体生物发生所需的某些mRNA。此外,XPO1介导的核输出与各种类型的疾病有关,比如癌症,炎症和病毒感染。先前的研究已经证明了XPO1在癌变中的关键作用及其作为治疗靶标的潜力。新型开发的一代特异性XPO1抑制剂的临床使用及其与其他药物的组合来阻断XPO1介导的核输出是一种有前途的新治疗策略。本研究的目的是解释XPO1和阻断XPO1介导的核输出的抑制剂的工作机制。
    Nuclear pore complexes (NPCs) regulate the entry and exit of molecules from the cell nucleus. Small molecules pass through NPCs by diffusion while large molecules enter and exit the nucleus by karyopherins, which serve as transport factors. Exportin-1 (XPO1) is a protein that is an important member of the karyopherin family and carries macromolecules from the nucleus to the cytoplasm. XPO1 is responsible for nuclear-cytoplasmic transport of protein, ribosomal RNA and certain required mRNAs for ribosomal biogenesis. Furthermore, XPO1-mediated nuclear export is associated with various types of disease, such as cancer, inflammation and viral infection. The key role of XPO1 in carcinogenesis and its potential as a therapeutic target has been demonstrated by previous studies. Clinical use of novel developed generation-specific XPO1 inhibitors and their combination with other agents to block XPO1-mediated nuclear export are a promising new treatment strategy. The aim of the present study was to explain the working mechanism of XPO1 and inhibitors that block XPO1-mediated nuclear export.
    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
    癌症转移是乳腺癌(BC)患者死亡的主要原因。肝脏是乳腺癌转移的常见部位,乳腺癌肝转移(BCLMs)患者的5年生存率仅为8.5%左右。CircRNAs参与多种癌症相关的病理行为,它们独特的结构和对RNA降解的抗性使它们成为理想的诊断生物标志物和治疗靶标。因此,研究circRNAs在肿瘤转移中的作用和分子机制具有重要意义。通过circRNA微阵列和qRT-PCR实验将CircLIFR-007鉴定为BC转移中的关键环状RNA。进行细胞功能测定以探索circLIFR-007在乳腺癌细胞中的作用。体内实验验证了circLIFR-007的功能。进行了几种分子测定以研究潜在的机制。我们发现circLIFR-007在乳腺癌肝转移中起负控制因子的作用。CircLIFR-007通过输出hnRNPA1来上调YAP的磷酸化水平,以促进细胞质中hnRNPA1和YAP之间的结合。circirLIFR-007的过表达抑制了肝转移相关蛋白的表达,SREBF1和SNAI1受转录因子YAP调控。功能上,circLIFR-007在体内和体外均抑制乳腺癌细胞的增殖和转移。
    Cancer metastasis is the major cause of death in patients with breast cancer (BC). The liver is a common site of breast cancer metastasis, and the 5-year survival rate of patients with breast cancer liver metastases (BCLMs) is only about 8.5 %. CircRNAs are involved in a variety of cancer-related pathological behaviors, and their unique structure and resistance to RNA degradation enable them to serve as ideal diagnostic biomarkers and therapeutic targets. Therefore, it is important to investigate the role and molecular mechanism of circRNAs in cancer metastasis. CircLIFR-007 was identified as a critical circular RNA in BC metastasis by circRNAs microarray and qRT-PCR experiment. Cell function assays were performed to explore the effect of circLIFR-007 in breast cancer cells. Experiments in vivo validated the function of circLIFR-007. Several molecular assays were performed to investigate the underlying mechanisms. We found that circLIFR-007 acted as a negative controller in breast cancer liver metastasis. CircLIFR-007 upregulates the phosphorylation level of YAP by exporting hnRNPA1 to promote the combination between hnRNPA1 and YAP in the cytoplasm. Overexpression of circLIFR-007 suppressed the expression of liver metastasis-related proteins, SREBF1 and SNAI1, which were regulated by transcription factor YAP. Functionally, circLIFR-007 inhibits the proliferation and metastasis of breast cancer cells both in vivo and in vitro.
    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