UPF1

UPF1
  • 文章类型: Journal Article
    最近对遗传稳健性的研究表明,转录适应(TA)是生物体可以通过同源基因的激活来补偿基因突变的一种机制。这里,我们发现基因突变,在淀粉样前体蛋白-b(appb)基因中引入过早终止密码子(PTC),激活了另外两个应用程序家庭成员的TA,斑马鱼中的appa和淀粉样前体样蛋白2(aplp2)。观察到的appa和aplp2的转录反应需要突变体mRNA的降解,并且不依赖于Appb蛋白水平。此外,在人神经元祖细胞(hNPC)中观察到淀粉样前体蛋白(APP)家族成员之间的TA,然而,补偿仅在早期神经元分化期间存在,并且在分化更高的神经元阶段或成年斑马鱼大脑中无法检测到。使用击倒和化学抑制,我们表明,无义介导的mRNA衰变(NMD)参与突变mRNA的降解,Upf1和Upf2,NMD途径中的关键蛋白,调节APA的内源性转录水平,appb,aplp1和aplp2总之,我们的研究结果表明,App家族成员的表达水平受NMD通路的调控,使app/APPmRNA失稳的突变可通过TA在斑马鱼和人类神经元祖细胞中诱导其他家族成员的遗传补偿.显著性陈述增加APP水平的遗传变异与阿尔茨海默病(AD)病理生理学相关。因此,理解调节APP表达水平的机制是关键的兴趣。这里,我们确定转录适应是APP家族成员可以调节同一家族中基因表达水平以补偿另一个家族基因缺失的一种机制。在引入PTC后,补偿是通过NMD途径中介导mRNA衰减的因素驱动的。有趣的是,我们的数据还表明,即使在生理条件下,NMD监测机制也是微调APP家族成员mRNA水平的重要方面。因此,我们的发现为APP成员之间的薪酬提供了见解,并揭示了可以监管APP的新目标。
    Studies on genetic robustness recently revealed transcriptional adaptation (TA) as a mechanism by which an organism can compensate for genetic mutations through activation of homologous genes. Here, we discovered that genetic mutations, introducing a premature termination codon (PTC) in the amyloid precursor protein-b (appb) gene, activated TA of two other app family members, appa and amyloid precursor-like protein-2 (aplp2), in zebrafish. The observed transcriptional response of appa and aplp2 required degradation of mutant mRNA and did not depend on Appb protein level. Furthermore, TA between amyloid precursor protein (APP) family members was observed in human neuronal progenitor cells; however, compensation was only present during early neuronal differentiation and could not be detected in a more differentiated neuronal stage or adult zebrafish brain. Using knockdown and chemical inhibition, we showed that nonsense-mediated mRNA decay (NMD) is involved in degradation of mutant mRNA and that Upf1 and Upf2, key proteins in the NMD pathway, regulate the endogenous transcript levels of appa, appb, aplp1, and aplp2 In conclusion, our results suggest that the expression level of App family members is regulated by the NMD pathway and that mutations destabilizing app/APP mRNA can induce genetic compensation by other family members through TA in both zebrafish and human neuronal progenitors.
    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
    肝细胞癌(HCC)是一种常见的恶性肿瘤,具有复杂的异质性和耐药性。对铁性凋亡的抵抗与HCC的进展密切相关。虽然HCC肿瘤对铁凋亡的敏感性不同,这种异质性背后的确切因素仍不清楚.在这项研究中,我们试图阐明导致HCC铁凋亡抵抗的机制。在HCC细胞系Huh7中使用亚毒性浓度(IC20)的铁凋亡诱导擦除素进行的全基因组CRISPR/Cas9筛选显示,TRIM34是HCC中铁凋亡抗性的关键驱动因素。进一步的研究表明,TRIM34抑制肝癌细胞的铁凋亡,促进它们的扩散,迁移,和体内外侵袭。此外,TRIM34在肝癌肿瘤组织中表达升高,与预后不良有关。机械上,TRIM34直接与上移码1(UPF1)交互,无义介导的mRNA衰减(NMD)途径的核心组成部分,促进其泛素化和降解。这种相互作用抑制GPX4转录物降解,从而促进HCC中这种关键的铁凋亡抑制因子的蛋白质水平。鉴于癌症中铁死亡与适应性免疫反应之间的紧密串扰,具有TRIM34靶向敲低的HCC细胞表现出抗PD-1治疗的改善反应。一起来看,TRIM34/UPF1/GPX4轴介导HCC的铁凋亡抗性,从而促进恶性表型。因此,靶向TRIM34可能代表了一种有前途的HCC治疗新策略。
    Hepatocellular carcinoma (HCC) is a prevalent malignancy characterized by complex heterogeneity and drug resistance. Resistance to ferroptosis is closely related to the progression of HCC. While HCC tumors vary in their sensitivity to ferroptosis, the precise factors underlying this heterogeneity remain unclear. In this study, we sought to elucidate the mechanisms that contribute to ferroptosis resistance in HCC. Whole-genome CRISPR/Cas9 screen using a subtoxic concentration (IC20) of ferroptosis inducer erastin in the HCC cell line Huh7 revealed TRIM34 as a critical driver of ferroptosis resistance in HCC. Further investigation revealed that TRIM34 suppresses ferroptosis in HCC cells, promoting their proliferation, migration, and invasion both in vitro and in vivo. Furthermore, TRIM34 expression is elevated in HCC tumor tissues, correlating with a poor prognosis. Mechanistically, TRIM34 directly interacts with Up-frameshift 1 (UPF1), a core component of the nonsense-mediated mRNA decay (NMD) pathway, to promote its ubiquitination and degradation. This interaction suppresses GPX4 transcript degradation, thus promoting the protein levels of this critical ferroptosis suppressor in HCC. In light of the close crosstalk between ferroptosis and the adaptive immune response in cancer, HCC cells with targeting knockdown of TRIM34 exhibited an improved response to anti-PD-1 treatment. Taken together, the TRIM34/UPF1/GPX4 axis mediates ferroptosis resistance in HCC, thereby promoting malignant phenotypes. Targeting TRIM34 may thus represent a promising new strategy for HCC treatment.
    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
    人类无义介导的mRNA衰变(NMD)因子的突变在神经发育障碍中富集。我们表明,小鼠胚胎神经祖细胞中关键NMD因子Upf2的缺失会导致围产期小头畸形,而未成熟神经元中的缺失则不会。表明NMD在祖细胞中的关键作用。Upf2敲除(KO)延长放射状神经胶质祖细胞的细胞周期,促进它们向中间祖细胞的过渡,并导致上层神经元减少。CRISPRi筛选确定了Trp53敲低挽救Upf2KO祖细胞而没有全局逆转NMD抑制,暗示大多数NMD靶标对细胞周期缺陷的边际贡献。整合的功能基因组学表明,NMD降解选择性TRP53下游靶标,包括Cdkn1a,which,如果没有国家导弹防御系统的压制,减缓细胞周期。Trp53KO恢复祖细胞池并挽救Upf2KO小鼠的小头畸形。因此,NMD在发育中的大脑中的一个生理作用是降解选择性TRP53靶标以控制祖细胞周期和大脑大小。
    Mutations in human nonsense-mediated mRNA decay (NMD) factors are enriched in neurodevelopmental disorders. We show that deletion of key NMD factor Upf2 in mouse embryonic neural progenitor cells causes perinatal microcephaly but deletion in immature neurons does not, indicating NMD\'s critical roles in progenitors. Upf2 knockout (KO) prolongs the cell cycle of radial glia progenitor cells, promotes their transition into intermediate progenitors, and leads to reduced upper-layer neurons. CRISPRi screening identified Trp53 knockdown rescuing Upf2KO progenitors without globally reversing NMD inhibition, implying marginal contributions of most NMD targets to the cell cycle defect. Integrated functional genomics shows that NMD degrades selective TRP53 downstream targets, including Cdkn1a, which, without NMD suppression, slow the cell cycle. Trp53KO restores the progenitor cell pool and rescues the microcephaly of Upf2KO mice. Therefore, one physiological role of NMD in the developing brain is to degrade selective TRP53 targets to control progenitor cell cycle and brain size.
    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
    转录后mRNA调控塑造基因表达,然而,顺式元件和mRNA翻译界面如何调节mRNA的稳定性却知之甚少。我们发现翻译启动的力量,上游开放阅读框架(uORF)内容,密码子最优性,AU丰富的元素,microRNA结合位点,和开放阅读框(ORF)长度组合功能以调节mRNA的稳定性。机器学习分析确定ORF长度是调节mRNA衰变的最重要的保守特征。我们发现Upf1绑定翻译不好和未翻译的ORF,它们与更高的衰变率有关,包括具有uORF的mRNA和终止密码子后具有暴露ORF的mRNA。我们的研究强调了Upf1在监控具有翻译不良的暴露ORF的mRNA中的融合作用,例如具有长ORF的mRNA,ORF-like3\'UTR,和含有uORF的mRNA。我们建议Upf1调节不良/未翻译的ORF提供了一种统一的监测机制,可以调节外显子连接复合物(EJC)独立的无义介导的衰变(NMD)途径中的mRNA稳定性和稳态,我们将其称为ORF介导的衰变(OMD)。
    Post-transcriptional mRNA regulation shapes gene expression, yet how cis-elements and mRNA translation interface to regulate mRNA stability is poorly understood. We find that the strength of translation initiation, upstream open reading frame (uORF) content, codon optimality, AU-rich elements, microRNA binding sites, and open reading frame (ORF) length function combinatorially to regulate mRNA stability. Machine-learning analysis identifies ORF length as the most important conserved feature regulating mRNA decay. We find that Upf1 binds poorly translated and untranslated ORFs, which are associated with a higher decay rate, including mRNAs with uORFs and those with exposed ORFs after stop codons. Our study emphasizes Upf1\'s converging role in surveilling mRNAs with exposed ORFs that are poorly translated, such as mRNAs with long ORFs, ORF-like 3\' UTRs, and mRNAs containing uORFs. We propose that Upf1 regulation of poorly/untranslated ORFs provides a unifying mechanism of surveillance in regulating mRNA stability and homeostasis in an exon-junction complex (EJC)-independent nonsense-mediated decay (NMD) pathway that we term ORF-mediated decay (OMD).
    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
    外显子-外显子连接上游的外显子连接复合物(EJC)的沉积有助于通过防止已经剪接的mRNA中的虚假重新剪接事件来维持转录组完整性。在这里,我们研究了EJC对于2.2兆碱基长的人DMD前mRNA的正确剪接的重要性,编码肌营养不良蛋白,参与细胞骨架组织和细胞信号传导的必需蛋白。使用靶向RNA-seq,我们显示,在人肌肉细胞系中,EJC的eIF4A3和Y14核心成分的敲除会导致聚集在DMD转录本(Dp427m)3'末端的错误剪接事件的积累。这种失调在成年骨骼肌中普遍表达的短Dp71同种型中是保守的,并且用野生型eIF4A3和Y14蛋白而不是用EJC组装缺陷突变体eIF4A3拯救。MLN51蛋白和EJC相关的ASAP/PSAP复合物独立地调节受调节外显子71和78的包含。我们的数据证实了EJC在保持剪接保真度方面的保护作用,在DMD基因中,这对于保留存在于所有组织特异性同工型中的肌营养不良蛋白的关键C端蛋白-蛋白相互作用域的功能是必需的。鉴于EJC在维持肌营养不良蛋白完整性方面的作用,我们询问EJC是否也可能参与骨骼肌分化等复杂机制的调节.我们发现eIF4A3敲低通过阻断肌管形成来损害肌源性分化。总的来说,我们的数据为EJC在人类骨骼肌中的功能作用提供了新的见解。
    Deposition of the exon junction complex (EJC) upstream of exon-exon junctions helps maintain transcriptome integrity by preventing spurious re-splicing events in already spliced mRNAs. Here we investigate the importance of EJC for the correct splicing of the 2.2-megabase-long human DMD pre-mRNA, which encodes dystrophin, an essential protein involved in cytoskeletal organization and cell signaling. Using targeted RNA-seq, we show that knock-down of the eIF4A3 and Y14 core components of EJC in a human muscle cell line causes an accumulation of mis-splicing events clustered towards the 3\' end of the DMD transcript (Dp427m). This deregulation is conserved in the short Dp71 isoform expressed ubiquitously except in adult skeletal muscle and is rescued with wild-type eIF4A3 and Y14 proteins but not with an EJC assembly-defective mutant eIF4A3. MLN51 protein and EJC-associated ASAP/PSAP complexes independently modulate the inclusion of the regulated exons 71 and 78. Our data confirm the protective role of EJC in maintaining splicing fidelity, which in the DMD gene is necessary to preserve the function of the critical C-terminal protein-protein interaction domain of dystrophin present in all tissue-specific isoforms. Given the role of the EJC in maintaining the integrity of dystrophin, we asked whether the EJC could also be involved in the regulation of a mechanism as complex as skeletal muscle differentiation. We found that eIF4A3 knockdown impairs myogenic differentiation by blocking myotube formation. Collectively, our data provide new insights into the functional roles of EJC in human skeletal muscle.
    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:报道新发现的具有家族性渗出性玻璃体视网膜病变(FEVR)独特形式的TSPAN12突变,并找出TSPAN12中重复的新型内含子变异导致FEVR的可能机制。
    结果:通过基于面板的NGS检测到9个具有独特形式的FEVR的TSPAN12突变。MINI-Gene分析显示mRNA的两种剪接模式,处理两个不同的条带A和B,和突变型显示与Exon11跳跃的剪接模式的替换。野生型和突变型TSPAN12载体的构建显示过早终止密码子(PTC)的出现。体外表达检测显示,与野生型组相比,用突变载体转染的细胞中TSPAN12mRNA和蛋白质的表达水平显着下调。相反,翻译抑制剂CHX和UPF1的小干扰RNA(si-UPF1)显着增加了用突变载体转染的细胞中TSPAN12的mRNA或蛋白质表达。
    结论:在9例FEVR患者中报道了TSPAN12基因的9个突变,这些患者具有一系列独特的眼部异常。三个新的TSPAN12突变触发NMD会导致参与微纤维生物合成和组装的TSPAN12蛋白的减少,这可能会导致FEVR,并表明内含子序列分析可能是遗传咨询和产前诊断的重要工具。
    BACKGROUND: To report newly found TSPAN12 mutations with a unique form of familial exudative vitreoretinopathy (FEVR) and find out the possible mechanism of a repeated novel intronic variant in TSPAN12 led to FEVR.
    RESULTS: Nine TSPAN12 mutations with a unique form of FEVR were detected by panel-based NGS. MINI-Gene assay showed two splicing modes of mRNA that process two different bands A and B, and mutant-type shows replacement with the splicing mode of Exon11 hopping. Construction of wild-type and mutant TSPAN12 vector showed the appearance of premature termination codons (PTC). In vitro expression detection showed significant down-regulated expression level of TSPAN12 mRNAs and proteins in cells transfected with mutant vectors compared with in wild-type group. On the contrary, translation inhibitor CHX and small interfering RNA of UPF1 (si-UPF1) significantly increased mRNA or protein expression of TSPAN12 in cells transfected with the mutant vectors.
    CONCLUSIONS: Nine mutations in TSPAN12 gene are reported in 9 FEVR patients with a unique series of ocular abnormalities. The three novel TSPAN12 mutations trigger NMD would cause the decrease of TSPAN12 proteins that participate in biosynthesis and assembly of microfibers, which might lead to FEVR, and suggest that intronic sequence analysis might be a vital tool for genetic counseling and prenatal diagnoses.
    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
    Immunotherapy harnesses neoantigens encoded within the human genome, but their therapeutic potential is hampered by low expression, which may be controlled by the nonsense-mediated mRNA decay (NMD) pathway. This study investigates the impact of UPF1-knockdown on the expression of non-canonical/mutant proteins, employing proteogenomic to explore UPF1 role within the NMD pathway. Additionally, we conducted a comprehensive pan-cancer analysis of UPF1 expression and evaluated UPF1 expression in Triple-Negative Breast Cancer (TNBC) tissue in-vivo. Our findings reveal that UPF1-knockdown leads to increased translation of non-canonical/mutant proteins, particularly those originating from retained-introns, pseudogenes, long non-coding RNAs, and unannotated transcript biotypes. Moreover, our analysis demonstrates elevated UPF1 expression in various cancer types, with notably heightened protein levels in patient-derived TNBC tumors compared to adjacent tissues. This study elucidates UPF1 role in mitigating transcriptional noise by degrading transcripts encoding non-canonical/mutant proteins. Targeting this mechanism may reveal a new spectrum of neoantigens accessible to the antigen presentation pathway. Our novel findings provide a strong foundation for the development of therapeutic strategies aimed at targeting UPF1 or modulating the NMD pathway.
    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
    据报道,右美托咪定(Dex)在阿尔茨海默病(AD)中起神经保护作用。然而,具体机制尚不清楚.在AD模型中阐明Dex调控神经细胞凋亡的分子机制。将SH-SY5Y细胞用Aβ1-42(10μM)处理24h后,建立了体外AD模型。通过RIP分析验证了UPF1,lncRNASNHG14和HSPB8之间的相互作用。细胞活力,凋亡,基因的水平,并通过CCK-8测定法检测蛋白质,流式细胞术,蛋白质印迹,和qRT-PCR,分别。Dex下调lncRNASNHG14水平并抑制神经细胞凋亡。LncRNASNHG14过表达逆转了Dex对AD模型神经细胞凋亡的抑制作用。LncRNASNHG14通过招募UPF1减弱HSPB8mRNA的稳定性。HSPB8过表达抑制AD模型神经细胞凋亡。此外,HSPB8敲低可逆转Dex对AD模型神经细胞凋亡的抑制作用。我们的研究表明,Dex通过抑制lncRNASNHG14/UPF1轴促进HSPB8的表达,从而抑制AD中神经细胞的凋亡。
    Dexmedetomidine (Dex) is reported to play a neuroprotective role in Alzheimer\'s disease (AD). However, the specific mechanism remains unclear. Figure out the underlying molecular mechanism of Dex regulating nerve cell apoptosis in the AD model. The AD model in vitro was established after SH-SY5Y cells were treated with Aβ1 - 42 at (10 μM) for 24 h. The interaction among UPF1, lncRNA SNHG14, and HSPB8 was verified by RIP assay. Cell viability, apoptosis, the level of genes, and proteins were detected by CCK-8 assay, flow cytometry, Western blot, and qRT-PCR, respectively. Dex downregulated lncRNA SNHG14 level and inhibited apoptosis of nerve cells. LncRNA SNHG14 overexpression reversed the inhibitory effect of Dex on nerve cell apoptosis in the AD model. LncRNA SNHG14 attenuated HSPB8 mRNA stability by recruiting UPF1. HSPB8 overexpression inhibited apoptosis of nerve cells in the AD model. Moreover, HSPB8 knockdown reversed the inhibitory effect of Dex on nerve cell apoptosis in the AD model. Our study demonstrated that Dex promoted HSPB8 expression via inhibiting the lncRNA SNHG14/UPF1 axis to inhibit nerve cell apoptosis in AD.
    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
    mRNA监测途径对于准确的基因表达和维持翻译稳态至关重要,确保生产功能齐全的蛋白质。对mRNA质量控制途径的未来见解将使我们能够了解如何控制细胞mRNA水平,如何消除有缺陷或不需要的mRNA,以及这些失调如何导致人类疾病。在这里,我们回顾翻译偶联mRNA质量控制机制,包括不停止和不停止的mRNA衰变途径,描述它们的机制,共享的交易因素,和差异。我们还描述了我们对无义介导的mRNA衰变(NMD)途径的理解的进展,强调最近的机械发现,新因素的发现,以及NMD在细胞生理学中的作用及其对人类疾病的影响。
    mRNA surveillance pathways are essential for accurate gene expression and to maintain translation homeostasis, ensuring the production of fully functional proteins. Future insights into mRNA quality control pathways will enable us to understand how cellular mRNA levels are controlled, how defective or unwanted mRNAs can be eliminated, and how dysregulation of these can contribute to human disease. Here we review translation-coupled mRNA quality control mechanisms, including the non-stop and no-go mRNA decay pathways, describing their mechanisms, shared trans-acting factors, and differences. We also describe advances in our understanding of the nonsense-mediated mRNA decay (NMD) pathway, highlighting recent mechanistic findings, the discovery of novel factors, as well as the role of NMD in cellular physiology and its impact on human 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
    RNA结合蛋白TRIM71/LIN-41是一种系统发育保守的发育调节因子,在哺乳动物干细胞重编程中起作用,大脑发育,和癌症。TRIM71通过发夹基序识别靶mRNA,并通过等待鉴定的分子机制沉默它们。这里,我们发现TRIM71通过RNA支持的与miRNA诱导的沉默复合物(miRISC)的核心成分TNRC6/GW182的相互作用抑制其靶标.我们证明了AGO2,TRIM71和UPF1各自招募TNRC6到特定的转录本组以使它们沉默。由于细胞TNRC6水平有限,竞争发生在沉默途径之间,这样AGO蛋白或AGO与TNRC6结合的丧失增强了其他途径的活性。我们得出结论,miRNA样沉默活性在不同的mRNA沉默途径中共享,并且使用TNRC6作为中心枢纽提供了整合其活性的手段。
    The RNA-binding protein TRIM71/LIN-41 is a phylogenetically conserved developmental regulator that functions in mammalian stem cell reprogramming, brain development, and cancer. TRIM71 recognizes target mRNAs through hairpin motifs and silences them through molecular mechanisms that await identification. Here, we uncover that TRIM71 represses its targets through RNA-supported interaction with TNRC6/GW182, a core component of the miRNA-induced silencing complex (miRISC). We demonstrate that AGO2, TRIM71, and UPF1 each recruit TNRC6 to specific sets of transcripts to silence them. As cellular TNRC6 levels are limiting, competition occurs among the silencing pathways, such that the loss of AGO proteins or of AGO binding to TNRC6 enhances the activities of the other pathways. We conclude that a miRNA-like silencing activity is shared among different mRNA silencing pathways and that the use of TNRC6 as a central hub provides a means to integrate their activities.
    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