RNA stability

RNA 稳定性
  • 文章类型: Journal Article
    背景:转录因子SIX2在几种肿瘤中的作用已被鉴定。然而,其在胃癌(GC)进展中的作用尚未揭示。我们的目标是探索SIX2对GC细胞干性的影响和潜在机制。
    方法:采用慢病毒感染在GC细胞中建立SIX2或PFN2的稳定表达。进行了功能增益和功能丧失实验,以检测干性标记的变化,流式细胞术配置文件,肿瘤球体形成,和肿瘤启动能力。ChIP,RNA测序,组织微阵列,并进行了生物信息学分析,以揭示SIX2和PFN2之间的相关性。通过组织微阵列分析阐明了SIX2/PFN2环介导作用的潜在机制,RNA稳定性分析,IP-MS,免疫共沉淀,和抑制JNK信号通路。
    结果:SIX2增强了GC细胞的干性。机械上,SIX2直接与PFN2的启动子结合并促进PFN2活性。PFN2,反过来,通过招募RNA结合蛋白YBX-1促进SIX2的mRNA稳定性,随后激活下游MAPK/JNK途径。
    结论:这项研究揭示了SIX2在控制GC细胞干细胞中的作用,定义负责这种调节的新型SIX2/PFN2调节环。这表明靶向SIX2/PFN2环用于GC处理的潜力(图形摘要)。
    BACKGROUND: The roles of the transcriptional factor SIX2 have been identified in several tumors. However, its roles in gastric cancer (GC) progression have not yet been revealed. Our objective is to explore the impact and underlying mechanisms of SIX2 on the stemness of GC cells.
    METHODS: Lentivirus infection was employed to establish stable expression SIX2 or PFN2 in GC cells. Gain- and loss-of-function experiments were conducted to detect changes of stemness markers, flow cytometry profiles, tumor spheroid formation, and tumor-initiating ability. ChIP, RNA-sequencing, tissue microarray, and bioinformatics analysis were performed to reveal the correlation between SIX2 and PFN2. The mechanisms underlying the SIX2/PFN2 loop-mediated effects were elucidated through tissue microarray analysis, RNA stability assay, IP-MS, Co-Immunoprecipitation, and inhibition of the JNK signaling pathway.
    RESULTS: The stemness of GC cells was enhanced by SIX2. Mechanistically, SIX2 directly bound to PFN2\'s promoter and promoted PFN2 activity. PFN2, in turn, promoted the mRNA stability of SIX2 by recruiting RNA binding protein YBX-1, subsequently activating the downstream MAPK/JNK pathway.
    CONCLUSIONS: This study unveils the roles of SIX2 in governing GC cell stemness, defining a novel SIX2/PFN2 regulatory loop responsible for this regulation. This suggests the potential of targeting the SIX2/PFN2 loop for GC treatment (Graphical Abstracts).
    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
    真核基因的表达在转录和转录后水平受到调控,监管的中断对人类疾病有重要贡献。5'm7GmRNA帽是转录后调控的中心节点,参与mRNA稳定和翻译效率。在哺乳动物中,DCP1a和DCP1b是mRNA帽水解酶DCP2的副同源辅因子蛋白。由于低等真核生物具有单一的DCP1辅因子,这种进化差异所获得的功能优势仍不清楚.我们报告了DCP1a和DCP1b的第一次功能解剖,证明它们是DCP2的非冗余辅因子,在脱盖复杂的完整性和特异性中具有独特的作用。DCP1a对于脱盖复合物组装以及脱盖复合物与mRNA帽结合蛋白之间的相互作用至关重要。DCP1b对于与蛋白质降解和翻译机制的复杂相互作用至关重要。DCP1a和DCP1b影响不同mRNA的周转。观察到DCP1a和DCP1b调控mRNA分子的不同本体论组,以及它们在取消复杂完整性方面的非冗余角色,提供了第一个证据,证明这些旁系同源物具有质量上不同的功能。
    Eukaryotic gene expression is regulated at the transcriptional and post-transcriptional levels, with disruption of regulation contributing significantly to human diseases. The 5\' m7G mRNA cap is a central node in post-transcriptional regulation, participating in both mRNA stabilization and translation efficiency. In mammals, DCP1a and DCP1b are paralogous cofactor proteins of the mRNA cap hydrolase DCP2. As lower eukaryotes have a single DCP1 cofactor, the functional advantages gained by this evolutionary divergence remain unclear. We report the first functional dissection of DCP1a and DCP1b, demonstrating that they are non-redundant cofactors of DCP2 with unique roles in decapping complex integrity and specificity. DCP1a is essential for decapping complex assembly and interactions between the decapping complex and mRNA cap-binding proteins. DCP1b is essential for decapping complex interactions with protein degradation and translational machinery. DCP1a and DCP1b impact the turnover of distinct mRNAs. The observation that different ontological groups of mRNA molecules are regulated by DCP1a and DCP1b, along with their non-redundant roles in decapping complex integrity, provides the first evidence that these paralogs have qualitatively distinct functions.
    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
    信使RNA剪接和降解是基因表达调控的关键,其异常导致疾病。以前的估计动能率的方法有局限性,假设跨细胞的速率均匀。DeepKINET是一种深度生成模型,可从scRNA-seq数据中估计单细胞分辨率下的剪接和降解率。DeepKINET在模拟和代谢标记数据集上优于现有方法。应用于前脑和乳腺癌数据,它鉴定了负责动力学速率多样性的RNA结合蛋白。DeepKINET还分析了剪接因子突变对红系谱系细胞中靶基因的影响。DeepKINET有效地揭示了转录后调控中的细胞异质性。
    Messenger RNA splicing and degradation are critical for gene expression regulation, the abnormality of which leads to diseases. Previous methods for estimating kinetic rates have limitations, assuming uniform rates across cells. DeepKINET is a deep generative model that estimates splicing and degradation rates at single-cell resolution from scRNA-seq data. DeepKINET outperforms existing methods on simulated and metabolic labeling datasets. Applied to forebrain and breast cancer data, it identifies RNA-binding proteins responsible for kinetic rate diversity. DeepKINET also analyzes the effects of splicing factor mutations on target genes in erythroid lineage cells. DeepKINET effectively reveals cellular heterogeneity in post-transcriptional regulation.
    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
    癌症是一种严重威胁人类健康的疾病,其发生和发展涉及复杂的分子机制。长链非编码RNA(lncRNAs)和RNA结合蛋白(RBP)是细胞内重要的调控分子。近年来在癌症研究中引起了广泛关注。lncRNAs和RBPs的结合在mRNA的转录后调控中起着至关重要的作用,通过调节mRNA的稳定性来影响与癌症相关的蛋白质的合成。这个,反过来,调节肿瘤细胞的恶性生物学行为,如增殖和转移,并在治疗抗性中起重要作用。本研究综述了lncRNA-RBP相互作用在多种恶性肿瘤mRNA稳定性调节中的作用。重点关注这种调控相互作用的分子机制。本综述旨在更深入地了解这些分子机制,为癌症的精准治疗提供新的策略和见解。
    Cancer is a disease that poses a serious threat to human health, the occurrence and development of which involves complex molecular mechanisms. Long non‑coding RNAs (lncRNAs) and RNA‑binding proteins (RBPs) are important regulatory molecules within cells, which have garnered extensive attention in cancer research in recent years. The binding of lncRNAs and RBPs plays a crucial role in the post‑transcriptional regulation of mRNA, affecting the synthesis of proteins related to cancer by regulating the stability of mRNA. This, in turn, regulates the malignant biological behaviors of tumor cells, such as proliferation and metastasis, and serves an important role in therapeutic resistance. The present study reviewed the role of lncRNA‑RBP interactions in the regulation of mRNA stability in various malignant tumors, with a focus on the molecular mechanisms underlying this regulatory interaction. The aim of the present review was to gain a deeper understanding of these molecular mechanisms to provide new strategies and insights for the precise treatment of 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
    目的:本研究旨在探讨IGF2BP2在食管鳞状细胞癌进展中的作用。
    方法:癌症基因组图谱(TCGA)数据集,转录组测序,和基因表达综合(GEO)数据集用于检测ESCC中m6A相关基因的表达。体外和体内试验用于探索IGF2BP2在ESCC中的作用。
    结果:IGF2BP2在人ESCC标本中显著过表达,通过分析GEO数据集证实了这一点。IGF2BP2过表达与ESCC患者的不良预后相关。改变IGF2BP2的表达影响细胞增殖,迁移,ESCC细胞的体外侵袭和体内致瘤性。IGF2BP2可以以m6A依赖性方式结合并稳定ESCC中的肝癌衍生生长因子(HDGF)转录本,并促进HDGF表达。
    结论:这些发现表明,新型IGF2BP2-HDGF轴对于ESCC癌症进展至关重要,可以作为开发治疗药物的靶标。
    OBJECTIVE: This study aimed to explore the role of IGF2BP2 in esophageal squamous cell carcinoma (ESCC) progression.
    METHODS: The Cancer Genome Atlas (TCGA) dataset, transcriptome sequencing, and the Gene Expression Omnibus (GEO) dataset were used to detect the expression of m6A-associated genes in ESCC. The in vitro and in vivo assays were used to explore the role of IGF2BP2 in ESCC.
    RESULTS: IGF2BP2 was significantly overexpressed in human ESCC specimens, which was confirmed by analyzing the GEO dataset. IGF2BP2 overexpression was correlated with poor prognosis in patients with ESCC. Altering the expression of IGF2BP2 influenced the proliferation, migration, and invasion of ESCC cells in vitro and tumorigenicity in vivo. IGF2BP2 could bind to and stabilize hepatoma-derived growth factor (HDGF) transcripts in ESCC in an m6A-dependent manner and promote HDGF expression.
    CONCLUSIONS: These findings indicate that the novel IGF2BP2-HDGF axis is pivotal for ESCC cancer progression and can serve as a target for developing therapeutics.
    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
    RNaseH1已被认为是一种内切核糖核酸酶,专门用于RNA-DNA杂交体中RNA部分的内部降解,其核糖核酸酶活性在核酸代谢的多方面是不可或缺的。然而,RNA酶H1介导的杂交裂解的分子机制仍未充分阐明.在这里,使用单分子方法,我们探讨了酿酒酵母RNaseH1的杂交裂解动力学。值得注意的是,单个RNaseH1酶显示3'至5'核糖核酸外切酶活性。RNA的定向降解顺序地进行,但不连续地进行,其中解链约6-bp杂种作为两个连续3-ntRNA切除的先决条件限制了每个催化循环内的总体速率。此外,复制蛋白A(RPA)可增强RNaseH1的3'至5'核解速率和持续合成能力,并刺激其5'至3'外切核糖核酸酶活性。这种刺激主要是通过杂交体的预分离来实现的,因此将RNaseH1转移到双向外切核糖核酸酶,进一步增强其裂解效率。这些发现揭示了RNase的前所未有的特征,并提供了RNaseH1增强的RPA持续杂种切割的动态视图。
    RNase H1 has been acknowledged as an endoribonuclease specializing in the internal degradation of the RNA moiety within RNA-DNA hybrids, and its ribonuclease activity is indispensable in multifaceted aspects of nucleic acid metabolism. However, the molecular mechanism underlying RNase H1-mediated hybrid cleavage remains inadequately elucidated. Herein, using single-molecule approaches, we probe the dynamics of the hybrid cleavage by Saccharomyces cerevisiae RNase H1. Remarkably, a single RNase H1 enzyme displays 3\'-to-5\' exoribonuclease activity. The directional RNA degradation proceeds processively and yet discretely, wherein unwinding approximately 6-bp hybrids as a prerequisite for two consecutive 3-nt RNA excisions limits the overall rate within each catalytic cycle. Moreover, Replication Protein A (RPA) reinforces RNase H1\'s 3\'-to-5\' nucleolytic rate and processivity and stimulates its 5\'-to-3\' exoribonuclease activity. This stimulation is primarily realized through the pre-separation of the hybrids and consequently transfers RNase H1 to a bidirectional exoribonuclease, further potentiating its cleavage efficiency. These findings unveil unprecedented characteristics of an RNase and provide a dynamic view of RPA-enhanced processive hybrid cleavage by RNase H1.
    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
    最近的研究表明,mRNA内部m7G及其作者蛋白METTL1与细胞代谢和癌症调节密切相关。这里,我们确定IGF2BP家族蛋白IGF2BP1-3可以优先结合内部mRNAm7G。这样的互动,尤其是IGF2BP3与M7G,可以促进癌细胞中m7G靶转录本的降解。IGF2BP3对m7G修饰的变化更敏感,而IGF2BP1更喜欢m6A来稳定结合的转录物。我们还证明了p53转录本,TP53在癌细胞中在其3'UTR处被m7G修饰。在胶质母细胞瘤中,可以通过调节IGF2BP3或通过dCas13b指导系统对m7G进行位点特异性靶向来调节修饰转录物的甲基化水平和半衰期,导致癌症进展和化学敏感性的调节。
    Recent studies have suggested that mRNA internal m7G and its writer protein METTL1 are closely related to cell metabolism and cancer regulation. Here, we identify that IGF2BP family proteins IGF2BP1-3 can preferentially bind internal mRNA m7G. Such interactions, especially IGF2BP3 with m7G, could promote the degradation of m7G target transcripts in cancer cells. IGF2BP3 is more responsive to changes of m7G modification, while IGF2BP1 prefers m6A to stabilize the bound transcripts. We also demonstrate that p53 transcript, TP53, is m7G-modified at its 3\'UTR in cancer cells. In glioblastoma, the methylation level and the half lifetime of the modified transcript could be modulated by tuning IGF2BP3, or by site-specific targeting of m7G through a dCas13b-guided system, resulting in modulation of cancer progression and chemosensitivity.
    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
    MYC上调是癌症的标志,其中MYC驱动致癌基因表达并提高整个癌细胞转录组的总RNA合成。尽管这种转录合成代谢促进了癌症的生长和生存,对过量细胞RNA诱导的后果和代谢应激知之甚少。在这里,我们发现RNA降解和下游核糖核苷酸分解代谢是MYC诱导癌细胞死亡的新机制。结合遗传学和代谢组学,我们发现MYC通过细胞质外泌体增加RNA衰变,导致细胞毒性RNA分解代谢产物和活性氧的积累。值得注意的是,肿瘤来源的外泌体突变消除了MYC诱导的细胞死亡,表明过量的RNA衰变可能对人类癌症有毒。在协议中,嘌呤补救作为一种补偿途径,减轻MYC诱导的核糖核苷酸分解代谢,嘌呤补救抑制剂损害MYC+肿瘤进展。一起,这些数据表明,MYC诱导的RNA衰变是一种可用于治疗的致癌应激.意义:MYC是预后不良癌症最常见的致癌驱动因素,但一直难以抑制治疗抑制。我们在MYC+癌症中发现了一个新的漏洞,其中MYC通过过量的RNA衰变诱导细胞死亡。加剧下游核糖核苷酸分解代谢的治疗剂为TNBC(三阴性乳腺癌)和其他MYC驱动的癌症提供了治疗上可处理的方法。
    Upregulation of MYC is a hallmark of cancer, wherein MYC drives oncogenic gene expression and elevates total RNA synthesis across cancer cell transcriptomes. Although this transcriptional anabolism fuels cancer growth and survival, the consequences and metabolic stresses induced by excess cellular RNA are poorly understood. Herein, we discover that RNA degradation and downstream ribonucleotide catabolism is a novel mechanism of MYC-induced cancer cell death. Combining genetics and metabolomics, we find that MYC increases RNA decay through the cytoplasmic exosome, resulting in the accumulation of cytotoxic RNA catabolites and reactive oxygen species. Notably, tumor-derived exosome mutations abrogate MYC-induced cell death, suggesting excess RNA decay may be toxic to human cancers. In agreement, purine salvage acts as a compensatory pathway that mitigates MYC-induced ribonucleotide catabolism, and inhibitors of purine salvage impair MYC+ tumor progression. Together, these data suggest that MYC-induced RNA decay is an oncogenic stress that can be exploited therapeutically. Significance: MYC is the most common oncogenic driver of poor-prognosis cancers but has been recalcitrant to therapeutic inhibition. We discovered a new vulnerability in MYC+ cancer where MYC induces cell death through excess RNA decay. Therapeutics that exacerbate downstream ribonucleotide catabolism provide a therapeutically tractable approach to TNBC (Triple-negative Breast Cancer) and other MYC-driven cancers.
    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
    2020年,辉瑞-BioNTech(BNT162b2)和Moderna(mRNA-1273)的信使核糖核酸(mRNA)疫苗的快速开发和授权标志着人类mRNA产品应用的重要里程碑。克服以前的障碍,如mRNA的不稳定性和免疫原性。本文回顾了纳入这些疫苗的策略修饰,以提高mRNA的稳定性和翻译效率。例如包含核苷修饰和优化的mRNA设计元件,包括5'帽和poly(A)尾。我们强调了有关这些mRNA疫苗的广泛系统性生物分布的新出现的问题,导致延长的炎症反应和其他安全问题。指导生物分布研究的监管框架对于评估当今使用的新mRNA制剂的安全性至关重要。mRNA疫苗的稳定性,它们的普遍分布,以及封装的mRNA的寿命以及破坏性和潜在致命的刺突(S)蛋白的无限产生需要减轻潜在不利影响的策略。这里,我们探索了小干扰RNA(siRNA)和核糖核酸酶靶向嵌合体(RIBOTACs)作为有前途的解决方案的潜力,失活,降解残留和持续的疫苗mRNA,从而潜在地防止不受控制的S蛋白产生并降低毒性。siRNA和RIBOTACs的靶向性质允许精确干预,提供了预防和减轻基于mRNA的治疗的不良事件的途径。这篇综述呼吁进一步研究siRNA和RIBOTAC作为mRNA疫苗技术的解毒剂和解毒产品的应用。
    The rapid development and authorization of messenger ribonucleic acid (mRNA) vaccines by Pfizer-BioNTech (BNT162b2) and Moderna (mRNA-1273) in 2020 marked a significant milestone in human mRNA product application, overcoming previous obstacles such as mRNA instability and immunogenicity. This paper reviews the strategic modifications incorporated into these vaccines to enhance mRNA stability and translation efficiency, such as the inclusion of nucleoside modifications and optimized mRNA design elements including the 5\' cap and poly(A) tail. We highlight emerging concerns regarding the wide systemic biodistribution of these mRNA vaccines leading to prolonged inflammatory responses and other safety concerns. The regulatory framework guiding the biodistribution studies is pivotal in assessing the safety profiles of new mRNA formulations in use today. The stability of mRNA vaccines, their pervasive distribution, and the longevity of the encapsulated mRNA along with unlimited production of the damaging and potentially lethal spike (S) protein call for strategies to mitigate potential adverse effects. Here, we explore the potential of small interfering RNA (siRNA) and ribonuclease targeting chimeras (RIBOTACs) as promising solutions to target, inactivate, and degrade residual and persistent vaccine mRNA, thereby potentially preventing uncontrolled S protein production and reducing toxicity. The targeted nature of siRNA and RIBOTACs allows for precise intervention, offering a path to prevent and mitigate adverse events of mRNA-based therapies. This review calls for further research into siRNA and RIBOTAC applications as antidotes and detoxication products for mRNA vaccine technology.
    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
    N6-甲基腺苷(m6A)修饰在RNA分子功能中起着重要作用,因此影响肝细胞癌(HCC)的发生和发展。在这里,应用多个数据集对HCC内的DEGs进行了全面分析,分析显示大量基因的显著失调.进行了功能和信号通路富集分析。Further,TP53RK结合蛋白(TPRKB)作为一个重要的因素出现,在HCC组织样本和细胞中表现出高表达水平,可以预测HCC患者的不良OS。体外敲除TPRKB的研究表明,TPRKB敲除通过抑制生存力减弱HCC细胞侵袭性的作用,菌落形成,侵入能力,和迁徙能力,诱导细胞周期停滞,促进肝癌细胞凋亡。体内研究表明,TPRKB敲低可显著抑制小鼠模型中的肿瘤生长。此外,该研究确定甲基转移酶5,N6-腺苷(METTL5)是通过m6A修饰TPRKB表达的潜在调节剂,通过增强TPRKBmRNA稳定性来正向调节TPRKB表达。METTL5和TPRKB对HCC细胞表型的动态影响进一步证实,TPRKB过表达部分消除了METTL5敲低对HCC细胞侵袭性的抗癌作用。最后,我们的发现揭示了TPRKB,在HCC中显著过表达,对促进肿瘤侵袭性产生关键影响,METTL5通过m6A甲基化对其表达进行正调控。这些见解加深了对HCC发病机制的理解,并为靶向治疗开辟了新的途径,强调METTL5-TPRKB轴是HCC管理中潜在的新治疗靶标。
    N6-methyladenosine (m6A) modification plays an important role in RNA molecular functions, therefore affecting the initiation and development of hepatocellular carcinoma (HCC). Herein, multiple datasets were applied to conduct a comprehensive analysis of DEGs within HCC and the analysis revealed significant dysregulation of numerous genes. Functional and signaling pathway enrichment analyses were performed. Further, TP53RK binding protein (TPRKB) emerged as a significant factor, exhibiting high expression level within HCC tissue samples and cells which could predict HCC patients\' poor OS. Knockdown investigations of TPRKB in vitro demonstrated the effect of TPRKB knockdown on attenuating the aggressiveness of HCC cells by suppressing the viability, colony formation, invasive ability, and migratory ability, inducing cell cycle arrest, and facilitating the apoptosis of HCC cells. Investigations in vivo revealed that TPRKB knockdown significantly suppressed tumor growth in mice model. Additionally, the study identified methyltransferase 5, N6-adenosine (METTL5) as a potential regulator of TPRKB expression via m6A modification, positively regulating TPRKB expression by enhancing TPRKB mRNA stability. The dynamic effects of METTL5 and TPRKB upon the phenotypes of HCC cells further confirmed that TPRKB overexpression partially abolished the anti-cancer effects of METTL5 knockdown upon the aggressiveness of HCC cells. Conclusively, our findings uncover that TPRKB, significantly overexpressed in HCC, exerts a critical effect on promoting tumor aggressiveness, and its expression shows to be positively regulated by METTL5 via m6A methylation. These insights deepen the understanding of HCC pathogenesis and open new avenues for targeted therapies, highlighting that METTL5-TPRKB axis is an underlying new therapeutic target in HCC management.
    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