Karyopherins

核动力素
  • 文章类型: Journal Article
    氮(N)缺乏反应对于植物的生存和繁殖至关重要。这里,通过表达全基因组关联研究(eGWAS),我们揭示了一种调节拟南芥中N缺乏反应所必需的microRNA(miRNA)动力学的机制。拟南芥中参与叶片N缺乏反应的三个NAC转录因子(TF)基因的差异表达水平与HASTY(HST)中的多态性最显着相关。其编码负责生成成熟miRNA的输入蛋白/输出蛋白家族蛋白。HST是缺氮诱导的叶片衰老的负调节因子,HST的破坏和过表达不同地改变了响应N缺乏的miRNA动力学,改变miRNA靶向转录物的水平。有趣的是,N缺乏阻止HST与HST相互作用蛋白的相互作用,DCL1和RAN1,以及一些miRNA。这表明HST介导的miRNA动力学调控共同控制由多个N缺乏反应相关的NACTFs介导的调控,从而成为N缺乏响应网络的中心。
    Nitrogen (N) deficiency responses are essential for plant survival and reproduction. Here, via an expression genome-wide association study (eGWAS), we reveal a mechanism that regulates microRNA (miRNA) dynamics necessary for N deficiency responses in Arabidopsis. Differential expression levels of three NAC transcription factor (TF) genes involved in leaf N deficiency responses among Arabidopsis accessions are most significantly associated with polymorphisms in HASTY (HST), which encodes an importin/exportin family protein responsible for the generation of mature miRNAs. HST acts as a negative regulator of N deficiency-induced leaf senescence, and the disruption and overexpression of HST differently modifies miRNA dynamics in response to N deficiency, altering levels of miRNAs targeting transcripts. Interestingly, N deficiency prevents the interaction of HST with HST-interacting proteins, DCL1 and RAN1, and some miRNAs. This suggests that HST-mediated regulation of miRNA dynamics collectively controls regulations mediated by multiple N deficiency response-associated NAC TFs, thereby being central to the N deficiency response network.
    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
    Exportin-1 (XPO1) is a major transporter for hundreds of proteins. Selinexor is the first generation XPO1 inhibitor. At present, selinexor has gained more attention in the application of multiple myeloma (MM). Meanwhile, the latest clinical trials have confirmed that whether it is a single agent or combined with other chemotherapy regimens, selinexor can also achieve good therapeutic effects in patients with leukemia and lymphoma. This review summarizes the results of preclinical studies and clinical trials of selinexor in treatment of non-MM hematological malignancies, aiming to explore how to choose single agent or in combination with other regimens as induction chemotherapy.
    UNASSIGNED: 塞利尼索在非多发性骨髓瘤血液肿瘤中的应用及研究进展.
    UNASSIGNED: .核输出蛋白1(XPO1)是数百种蛋白质的主要转运蛋白。塞利尼索是第一代XPO1抑制剂,目前在多发性骨髓瘤的治疗中获得了较多的关注,同时最新临床试验也证实,无论是单药还是联合其他化疗方案,塞利尼索在白血病、淋巴瘤中同样能取得较好的治疗效果。本文总结了塞利尼索治疗非多发性骨髓瘤血液肿瘤的临床前研究和临床试验结果,旨在探讨未来如何选择塞利尼索单药或联合其他方案进行诱导化疗。.
    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: English Abstract
    OBJECTIVE: To investigate the effects of selinexor, a inhibitor of nuclear export protein 1 (XPO1) on the proliferation inhibition and apoptosis of Kasumi-1 cells in acute myeloid leukemia (AML).
    METHODS: MTS method was used to detect the inhibitory effect of different concentrations of selinexor on the proliferation of Kasumi-1 cells at different time points. The apoptosis rate and cell cycle changes after treatment with different concentration of selinexor were detected by flow cytometry.
    RESULTS: Selinexor inhibited the growth of Kasumi-1 cells at different time points in a concentration-dependent manner (r 24 h=0.7592, r 48 h=0.9456, and r 72 h=0.9425). Selinexor inhibited Kasumi-1 cells growth in a time-dependent manner (r =0.9057 in 2.5 μmol/L group, r =0.9897 in 5 μmol/L group and r =0.9994 in 10 μmol/L group). Selinexor could induce apoptosis of Kasumi-1 cells in a dose-dependent manner (r =0.9732), and the apoptosis of Kasumi-1 cells was more obvious with the increase of drug concentration. The proportion of G0/G1 phase was significantly increased and the proportion of S phase was significantly decreased after the treatment of Kasumi-1 cells by selinexor. With the increase of drug concentration, the proportion of Kasumi-1 cells cycle arrest in G0/G1 phase was increased and the cell synthesis was decreased.
    CONCLUSIONS: Selinexor can promote the death of tumor cells by inhibiting Kasumi-1 cells proliferation, inducing apoptosis and blocking cell cycle.
    UNASSIGNED: XPO1抑制剂塞利尼索对急性髓系白血病Kasumi-1细胞增殖和凋亡的影响.
    UNASSIGNED: 探讨核输出蛋白1(XPO1)抑制剂塞利尼索对急性髓系白血病(AML)Kasumi-1细胞增殖与凋亡的影响。.
    UNASSIGNED: 用MTS法检测不同浓度塞利尼索(0、2.5、5、10 μmol/L)作用于Kasumi-1细胞不同时间点(24、48、72 h)的增殖抑制率;流式细胞术检测不同浓度塞利尼索作用于Kasumi-1细胞48 h后的细胞凋亡率和细胞周期变化情况。.
    UNASSIGNED: 塞利尼索在不同时间点均可抑制Kasumi-1细胞生长,并呈浓度依赖(r 24 h=0.7592,r 48 h=0.9456,r 72 h=0.9425);不同浓度塞利尼索均可抑制Kasumi-1细胞生长,并呈时间依赖(2.5 μmol/L组r =0.9057,5 μmol/L组r =0.9897,10 μmol/L组r =0.9994)。塞利尼索可诱导Kasumi-1细胞凋亡,且呈浓度依赖(r =0.9732),随着药物浓度的增大,诱导Kasumi-1细胞凋亡的作用越明显。塞利尼索作用于Kasumi-1细胞48 h后,G0/G1期细胞比例明显上升,S期细胞比例明显下降,并且随着塞利尼索药物浓度的增大,Kasumi-1细胞周期阻滞于G0 /G1期的细胞比例增加,细胞合成减少。.
    UNASSIGNED: 塞利尼索可抑制Kasumi-1细胞增殖,诱导细胞凋亡,阻滞细胞周期于G0/G1期,导致肿瘤细胞的死亡。.
    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
    XPO1(Exportin-1/CRM1)是一种核输出蛋白,在癌症中经常过表达,并充当肿瘤发生的驱动因素。目前靶向XPO1的小分子正被用于临床作为抗癌剂。我们将XPO1鉴定为自然杀伤(NK)细胞的靶标。使用免疫肽,我们已经鉴定了一种源自XPO1的肽,该肽可以在人类白细胞抗原C的情况下被激活的NK细胞受体KIR2DS2识别。肽可以被内源性加工并呈递以通过该受体特异性地激活NK细胞。尽管XPO1在癌症中的高表达通常与不良预后相关,我们证明了特定癌症的结果,比如肝细胞癌,如果伴随有NK细胞浸润的证据,则可以大大改善。因此,我们将XPO1鉴定为NK细胞识别的真正的肿瘤抗原,为NK细胞治疗实体肿瘤提供了个性化方法的机会。
    XPO1 (Exportin-1/CRM1) is a nuclear export protein that is frequently overexpressed in cancer and functions as a driver of oncogenesis. Currently small molecules that target XPO1 are being used in the clinic as anticancer agents. We identify XPO1 as a target for natural killer (NK) cells. Using immunopeptidomics, we have identified a peptide derived from XPO1 that can be recognized by the activating NK cell receptor KIR2DS2 in the context of human leukocyte antigen-C. The peptide can be endogenously processed and presented to activate NK cells specifically through this receptor. Although high XPO1 expression in cancer is commonly associated with a poor prognosis, we show that the outcome of specific cancers, such as hepatocellular carcinoma, can be substantially improved if there is concomitant evidence of NK cell infiltration. We thus identify XPO1 as a bona fide tumor antigen recognized by NK cells that offers an opportunity for a personalized approach to NK cell therapy for solid tumors.
    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:Exportin1(XPO1)是一种核出口蛋白,可促进各种物质的运输。XPO1在多种肿瘤中作为不良预后因素促进肿瘤发展,并且是筛选抑制剂的治疗靶标。然而,XPO1在口腔鳞状细胞癌(OSCC)中的作用尚未确定。
    方法:使用生物信息学工具研究OSCC中XPO1mRNA的表达模式,免疫组织化学检测证实了OSCC标本中XPO1蛋白的表达水平。生存分析评估XPO1对预后的影响。利用GO和KEGG富集分析来揭示由XPO1介导的信号通路。此外,我们检查了XPO1和AKT/MAPK/TGFBR1与免疫浸润之间的关联。
    结果:XPO1mRNA和蛋白表达水平在OSCC中显著增强,并与OSCC严重程度相关。增强的XPO1表达指示较差的存活率。功能分析表明,XPO1介导的途径与OSCC的细胞周期和DNA复制相关,并减少了免疫浸润。此外,XPO1mRNA和蛋白表达水平与AKT/MAPK/TGFBR1呈显著正相关。
    结论:XPO1作为OSCC预后不良的标志物,可以通过AKT/MAPK/TGFBR1促进OSCC。
    BACKGROUND: Exportin 1 (XPO1) is a nuclear export protein that facilitates the transportation of various substances. XPO1 promotes tumor development as a poor prognostic factor in a variety of tumors and is a therapeutic target for screening inhibitors. However, the role of XPO1 in oral squamous cell carcinoma (OSCC) has yet to be determined.
    METHODS: The expression patterns of XPO1 mRNA in OSCC were investigated using bioinformatics tools, and the expression levels of XPO1 protein in OSCC specimens were confirmed by immunohistochemical assays. Survival analysis was conducted to evaluate the impact of XPO1 on prognosis. GO and KEGG enrichment analyses were utilized to uncover the signaling pathways mediated by XPO1. Additionally, we examined the association between XPO1 and AKT/MAPK/TGFBR1 and immune infiltration.
    RESULTS: XPO1 mRNA and protein expression levels were significantly enhanced in OSCC and associated with OSCC severity. Enhanced XPO1 expression was indicative of poor survival. Functional analysis showed that XPO1 mediated pathways associated with cell cycle and DNA replication and reduced immune infiltration in OSCC. Additionally, XPO1 mRNA and protein expression levels had significant positive relationships with AKT/MAPK/TGFBR1.
    CONCLUSIONS: XPO1, as a marker of poor prognosis in OSCC, can promote OSCC through AKT/MAPK/TGFBR1.
    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
    黄病毒科是一个正链RNA病毒家族,包括人类病原体,如日本脑炎病毒(JEV),登革热病毒(DENV),寨卡病毒(ZIKV),西尼罗河病毒(WNV)病毒核心蛋白的核定位在黄病毒科中保守,这个功能可能是开发广泛的抗黄病毒药物的目标。然而,核心蛋白易位到细胞核的机制以及核易位在病毒生命周期中的重要性仍然未知。我们旨在确定核心蛋白核易位的分子机制。我们确定了importin-7(IPO7),importin-β家族蛋白,作为黄病毒科核心蛋白的核载体。核输入测定显示,核心蛋白通过IPO7转运到细胞核,而CRISPR/Cas9的IPO7缺失损害了它们的核易位。为了了解核心蛋白核易位的重要性,我们评估了野生型或IPO7缺陷细胞中感染性病毒或单轮感染性颗粒的产生;这两个过程在IPO7缺陷细胞中显著受损,而野生型和IPO7缺陷细胞中的细胞内感染性病毒水平相当。这些结果表明,IPO7介导的核心蛋白核易位与黄病毒感染性病毒颗粒的释放有关。
    Flaviviridae is a family of positive-stranded RNA viruses, including human pathogens, such as Japanese encephalitis virus (JEV), dengue virus (DENV), Zika virus (ZIKV), and West Nile virus (WNV). Nuclear localization of the viral core protein is conserved among Flaviviridae, and this feature may be targeted for developing broad-ranging anti-flavivirus drugs. However, the mechanism of core protein translocation to the nucleus and the importance of nuclear translocation in the viral life cycle remain unknown. We aimed to identify the molecular mechanism underlying core protein nuclear translocation. We identified importin-7 (IPO7), an importin-β family protein, as a nuclear carrier for Flaviviridae core proteins. Nuclear import assays revealed that core protein was transported into the nucleus via IPO7, whereas IPO7 deletion by CRISPR/Cas9 impaired their nuclear translocation. To understand the importance of core protein nuclear translocation, we evaluated the production of infectious virus or single-round-infectious-particles in wild-type or IPO7-deficient cells; both processes were significantly impaired in IPO7-deficient cells, whereas intracellular infectious virus levels were equivalent in wild-type and IPO7-deficient cells. These results suggest that IPO7-mediated nuclear translocation of core proteins is involved in the release of infectious virus particles of flaviviruses.
    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
    尽管RNAi和RNA剪接机制参与调节严重急性呼吸综合征冠状病毒2(SARS-CoV-2)的复制,它们在2019年冠状病毒病(COVID-19)发病机制中的确切作用仍不清楚。在这里,我们显示RNAi成分(Dicer和XPO5)和剪接因子(SRSF3和hnRNPA3)表达减少与COVID-19严重程度增加相关。SARS-CoV-2N蛋白诱导Dicer自噬降解,XPO5、SRSF3和hnRNPA3抑制miRNA生物发生和RNA剪接并触发DNA损伤,蛋白毒性应激,和肺炎。Dicer,XPO5、SRSF3和hnRNPA3敲低增加,虽然它们的过度表达减少,N蛋白诱导的肺炎的严重程度。年龄较大的小鼠表现出较低的Dicer表达,XPO5,SRSF3和hnRNPA3在其肺组织中表现出比年轻小鼠更严重的N蛋白诱导的肺炎。PJ34,一种聚(ADP-核糖)聚合酶抑制剂,或者阿那曲唑,芳香化酶抑制剂,通过恢复Dicer改善N蛋白或SARS-CoV-2引起的肺炎,XPO5、SRSF3和hnRNPA3表达。这些发现将有助于开发SARS-CoV-2相关肺炎的改进治疗方法。
    Though RNAi and RNA-splicing machineries are involved in regulating severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) replication, their precise roles in coronavirus disease 2019 (COVID-19) pathogenesis remain unclear. Herein, we show that decreased RNAi component (Dicer and XPO5) and splicing factor (SRSF3 and hnRNPA3) expression correlate with increased COVID-19 severity. SARS-CoV-2 N protein induces the autophagic degradation of Dicer, XPO5, SRSF3, and hnRNPA3, inhibiting miRNA biogenesis and RNA splicing and triggering DNA damage, proteotoxic stress, and pneumonia. Dicer, XPO5, SRSF3, and hnRNPA3 knockdown increases, while their overexpression decreases, N protein-induced pneumonia\'s severity. Older mice show lower expression of Dicer, XPO5, SRSF3, and hnRNPA3 in their lung tissues and exhibit more severe N protein-induced pneumonia than younger mice. PJ34, a poly(ADP-ribose) polymerase inhibitor, or anastrozole, an aromatase inhibitor, ameliorates N protein- or SARS-CoV-2-induced pneumonia by restoring Dicer, XPO5, SRSF3, and hnRNPA3 expression. These findings will aid in developing improved treatments for SARS-CoV-2-associated pneumonia.
    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
    共价药物的发现经历了复兴,许多亲电子小分子最近获得FDA批准。许多结构不同的亲电子小分子靶向半胱氨酸528处的exportin-1(XPO1/CRM1),包括核输出(SINE)selinexor的选择性抑制剂,2019年被FDA批准为抗癌药物。新兴证据支持靶向Cys528的XPO1调节剂的其他药理学类别,包括选择性转录激活抑制剂(SITAs)和诱导XPO1快速降解的探针。这里,我们分析了XPO1Cys528靶向探针的多个结构系列的结构-活性关系。我们观察到Cys528靶向小分子的亲电子部分在观察到的细胞行为中起决定性作用,亲电结构的细微变化足以将XPO1靶向探针转化为不同的药理学类别。这项研究代表了一个独特的案例研究,其中用于靶向特定半胱氨酸的亲电功能决定了多种靶向XPO1的小分子的药理作用。
    Covalent drug discovery has experienced a renaissance, with numerous electrophilic small molecules recently gaining FDA approval. Many structurally diverse electrophilic small molecules target exportin-1 (XPO1/CRM1) at cysteine 528, including the selective inhibitor of nuclear export (SINE) selinexor, which was FDA-approved as an anticancer agent in 2019. Emerging evidence supports additional pharmacological classes of XPO1 modulators targeting Cys528, including the selective inhibitors of transcriptional activation (SITAs) and probes that induce rapid degradation of XPO1. Here, we analyzed structure-activity relationships across multiple structural series of XPO1 Cys528-targeting probes. We observe that the electrophilic moiety of Cys528-targeting small molecules plays a decisive role in the cellular behavior observed, with subtle changes in electrophile structure being sufficient to convert XPO1-targeting probes to different pharmacological classes. This investigation represents a unique case study in which the electrophile functionality used to target a specific cysteine determines the pharmacological effect among diverse XPO1-targeting small molecules.
    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
    SF3B1突变经常发生在癌症中,但缺乏靶向治疗。XPO1抑制剂的临床试验,selinexor和eltanexor,在高危骨髓增生异常肿瘤(MDS)中,发现应答者富含SF3B1突变.鉴于XPO1(Exportin-1)是负责蛋白质和多种RNA物种出口的核出口国,这导致了SF3B1突变细胞对XPO1抑制敏感的假设,可能是由于剪接改变。在SF3B1野生型和突变细胞中XPO1抑制后的后续RNA测序显示RNA转录物的核保留增加,并且在SF3B1突变细胞中的选择性剪接增加,特别是影响凋亡途径的基因。为了确定与XPO1抑制协同作用的新型药物组合,使用eltanexor治疗进行正向遗传筛选,涉及抗凋亡靶标BCL2和BCLXL,通过体外和体内功能测试进行了验证。使用Sf3b1K700E条件性敲入小鼠在体内测试了这些靶标,这表明eltanexor和venetoclax(BCL2抑制剂)的组合对SF3B1突变细胞具有优先敏感性,而没有过度毒性。在这项研究中,我们揭示了SF3B1突变型MDS对XPO1抑制致敏的潜在机制,并在临床前合理化了eltanexor和venetoclax联合治疗高危MDS.
    SF3B1 mutations frequently occur in cancer yet lack targeted therapies. Clinical trials of XPO1 inhibitors, selinexor and eltanexor, in high-risk myelodysplastic neoplasms (MDS) revealed responders were enriched with SF3B1 mutations. Given that XPO1 (Exportin-1) is a nuclear exporter responsible for the export of proteins and multiple RNA species, this led to the hypothesis that SF3B1-mutant cells are sensitive to XPO1 inhibition, potentially due to altered splicing. Subsequent RNA sequencing after XPO1 inhibition in SF3B1 wildtype and mutant cells showed increased nuclear retention of RNA transcripts and increased alternative splicing in the SF3B1 mutant cells particularly of genes that impact apoptotic pathways. To identify novel drug combinations that synergize with XPO1 inhibition, a forward genetic screen was performed with eltanexor treatment implicating anti-apoptotic targets BCL2 and BCLXL, which were validated by functional testing in vitro and in vivo. These targets were tested in vivo using Sf3b1K700E conditional knock-in mice, which showed that the combination of eltanexor and venetoclax (BCL2 inhibitor) had a preferential sensitivity for SF3B1 mutant cells without excessive toxicity. In this study, we unveil the mechanisms underlying sensitization to XPO1 inhibition in SF3B1-mutant MDS and preclinically rationalize the combination of eltanexor and venetoclax for high-risk MDS.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号