pre-mRNA splicing

PRE - mRNA 拼接
  • 文章类型: Journal Article
    肿瘤抑制因子p53及其拮抗剂MDM2和MDM4整合应激信号。例如,核仁中核糖体的不平衡组装诱导p53。这里,我们显示核糖体蛋白L22(RPL22;eL22),在核糖体和核仁应激的条件下,促进MDM4外显子6的跳过。L22耗尽后,保持更多全长MDM4,导致p53活性降低和细胞增殖增强。L22与MDM4内含子6内的特定RNA元件结合,对应于茎环共有序列,导致6号外显子跳跃.这些内含子元件的靶向缺失在很大程度上消除了L22介导的外显子跳跃,并重新实现了细胞增殖。尽管有核仁应力。L22还控制L22L1(RPL22L1)和UBAP2LmRNA的可变剪接。因此,L22充当整合不同基因表达层的信号传导中间体。核糖体合成中的缺陷导致特定的可变剪接,最终触发p53介导的转录并阻止细胞增殖。
    The tumor suppressor p53 and its antagonists MDM2 and MDM4 integrate stress signaling. For instance, dysbalanced assembly of ribosomes in nucleoli induces p53. Here, we show that the ribosomal protein L22 (RPL22; eL22), under conditions of ribosomal and nucleolar stress, promotes the skipping of MDM4 exon 6. Upon L22 depletion, more full-length MDM4 is maintained, leading to diminished p53 activity and enhanced cellular proliferation. L22 binds to specific RNA elements within intron 6 of MDM4 that correspond to a stem-loop consensus, leading to exon 6 skipping. Targeted deletion of these intronic elements largely abolishes L22-mediated exon skipping and re-enables cell proliferation, despite nucleolar stress. L22 also governs alternative splicing of the L22L1 (RPL22L1) and UBAP2L mRNAs. Thus, L22 serves as a signaling intermediate that integrates different layers of gene expression. Defects in ribosome synthesis lead to specific alternative splicing, ultimately triggering p53-mediated transcription and arresting cell proliferation.
    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
    U1小核RNA(snRNA)形成核糖核蛋白颗粒(RNP),例如U1snRNP和U1-TAF15snRNP。U1snRNP是研究最多的RNPs之一,因为它在mRNA前剪接中起关键作用,通过与外显子/内含子连接处的序列直接相互作用来定义每个外显子的5'剪接位点(5'ss)。最近的报告支持U1snRNP在所有转录步骤中的作用,即启动,伸长率,和终止。U1-TAF15snRNP的功能了解较少,尽管它与转录机制相关,并可能通过与pre-mRNA内的5\'ss和/或5\'ss样序列相互作用来调节pre-mRNA剪接。隔离U1snRNA5'端的抗U1反义寡核苷酸(ASO)抑制U1snRNP的功能,包括转录和剪接。然而,尚不清楚U1snRNP的抑制是否通过深内含子序列影响pre-mRNA剪接的转录后调节。
    我们检查了隔离U1snRNA5'末端的抗U1ASO对脊髓性肌萎缩症(SMA)基因所有内部外显子转录和剪接的影响,SMN1和SMN2。我们的研究是通过采用多外显子跳跃检测测定法(MESDA)来实现的,该测定法可区分过早终止的转录本。我们使用SMN2超小基因来确定抗U1ASO在截短内含子的背景下是否对剪接有不同的影响。
    我们观察到由抗U1治疗引发的SMN1和SMN2的多个内部外显子的大量跳跃。提示U1snRNP在与深层内含子序列相互作用中的作用,具有截短内含子的SMN2超小基因的早期外显子对抗U1诱导的跳跃具有抗性。始终如一,靶向早期SMN1和SMN2外显子5'ss的工程化U1snRNA的过表达并不能防止由抗U1治疗引起的外显子跳跃。
    我们的结果揭示了U1snRNA相关的RNPs在通过深内含子序列执行的剪接调节中的独特作用。研究结果对于开发基于深内含子靶标的SMA新疗法具有重要意义。
    UNASSIGNED: The U1 small nuclear RNA (snRNA) forms ribonucleoprotein particles (RNPs) such as U1 snRNP and U1-TAF15 snRNP. U1 snRNP is one of the most studied RNPs due to its critical role in pre-mRNA splicing in defining the 5\' splice site (5\'ss) of every exon through direct interactions with sequences at exon/intron junctions. Recent reports support the role of U1 snRNP in all steps of transcription, namely initiation, elongation, and termination. Functions of U1-TAF15 snRNP are less understood, though it associates with the transcription machinery and may modulate pre-mRNA splicing by interacting with the 5\'ss and/or 5\'ss-like sequences within the pre-mRNA. An anti-U1 antisense oligonucleotide (ASO) that sequesters the 5\' end of U1 snRNA inhibits the functions of U1 snRNP, including transcription and splicing. However, it is not known if the inhibition of U1 snRNP influences post-transcriptional regulation of pre-mRNA splicing through deep intronic sequences.
    UNASSIGNED: We examined the effect of an anti-U1 ASO that sequesters the 5\' end of U1 snRNA on transcription and splicing of all internal exons of the spinal muscular atrophy (SMA) genes, SMN1 and SMN2. Our study was enabled by the employment of a multi-exon-skipping detection assay (MESDA) that discriminates against prematurely terminated transcripts. We employed an SMN2 super minigene to determine if anti-U1 ASO differently affects splicing in the context of truncated introns.
    UNASSIGNED: We observed substantial skipping of multiple internal exons of SMN1 and SMN2 triggered by anti-U1 treatment. Suggesting a role for U1 snRNP in interacting with deep intronic sequences, early exons of the SMN2 super minigene with truncated introns were resistant to anti-U1 induced skipping. Consistently, overexpression of engineered U1 snRNAs targeting the 5\'ss of early SMN1 and SMN2 exons did not prevent exon skipping caused by anti-U1 treatment.
    UNASSIGNED: Our results uncover a unique role of the U1 snRNA-associated RNPs in splicing regulation executed through deep intronic sequences. Findings are significant for developing novel therapies for SMA based on deep intronic targets.
    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
    转录后调控包括在DNA序列的初始拷贝转录成中间RNA分子(即,信使RNA),直到将此类分子用作生成蛋白质的模板。这些转录后调控机制的一个子集基本上注定要将未成熟的mRNA朝向其成熟形式进行处理,赋予足够的mRNA稳定性,提供相关内含子切除的手段,控制mRNA周转率和质量控制检查。在某些情况下增加了额外的复杂性层,由于成熟RNA分子中的离散核苷酸修饰是通过RNA编辑添加的,一个提供大量成熟mRNA多样性的过程。此外,许多转录后调控机制以细胞和组织特异性的方式发生,如选择性剪接和非编码RNA介导的调控。在这一章中,我们将简要总结目前的最新知识的一般转录后机制,而主要重点将致力于那些影响心脏发育和先天性心脏病的组织特异性转录后修饰。
    Posttranscriptional regulation comprises those mechanisms occurring after the initial copy of the DNA sequence is transcribed into an intermediate RNA molecule (i.e., messenger RNA) until such a molecule is used as a template to generate a protein. A subset of these posttranscriptional regulatory mechanisms essentially are destined to process the immature mRNA toward its mature form, conferring the adequate mRNA stability, providing the means for pertinent introns excision, and controlling mRNA turnover rate and quality control check. An additional layer of complexity is added in certain cases, since discrete nucleotide modifications in the mature RNA molecule are added by RNA editing, a process that provides large mature mRNA diversity. Moreover, a number of posttranscriptional regulatory mechanisms occur in a cell- and tissue-specific manner, such as alternative splicing and noncoding RNA-mediated regulation. In this chapter, we will briefly summarize current state-of-the-art knowledge of general posttranscriptional mechanisms, while major emphases will be devoted to those tissue-specific posttranscriptional modifications that impact on cardiac development and congenital heart disease.
    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
    选择性剪接(AS)是促进应激诱导的基因表达和蛋白质组多样性调节的重要机制。大规模测序技术允许鉴定通过应激反应性AS产生的转录本,对适应压力条件可能很重要。已经开发了几种生物信息学工具来从RNA测序结果中鉴定差异表达的可变剪接事件/转录物。本章介绍了使用rMATS工具进行差分选择性剪接分析的详细协议。此外,我们根据获得的输出文件提供了通过qRT-PCR验证检测到的剪接变体的指南.
    Alternative splicing (AS) is an important mechanism contributing to stress-induced regulation of gene expression and proteome diversity. Massive sequencing technologies allow the identification of transcripts generated via stress-responsive AS, potentially important for adaptation to stress conditions. Several bioinformatics tools have been developed to identify differentially expressed alternative splicing events/transcripts from RNA-sequencing results. This chapter describes a detailed protocol for differential alternative splicing analysis using the rMATS tool. In addition, we provide guidelines for validation of the detected splice variants by qRT-PCR based on the obtained output files.
    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
    RNA结合蛋白的转录后调节可以决定基因表达水平并驱动癌细胞蛋白质组的变化。鉴定蛋白质-RNA结合的机制,包括体内结合的优选序列基序,提供了对蛋白质-RNA网络以及它们如何影响mRNA结构的见解,函数,和稳定性。在这次审查中,我们将重点关注与新生或成熟mRNA中富含AU的元件(ARE)结合的蛋白质,它们在响应癌细胞遇到的压力中发挥作用。ARE结合蛋白(ARE-BPs)特异性影响可变剪接,稳定性,衰变和翻译,和形成富含RNA的生物分子缩合物,如胞质应激颗粒(SGs)。例如,最近的研究结果强调了ARE-BPs-如TIAR和HUR-在化疗耐药和编码促炎细胞因子的mRNA翻译调节中的作用.我们将讨论新出现的证据,即不同的ARE-BP活动模式影响白血病和淋巴瘤的发展,programming,适应微环境和化疗抗性。
    Post-transcriptional regulation by RNA binding proteins can determine gene expression levels and drive changes in cancer cell proteomes. Identifying mechanisms of protein-RNA binding, including preferred sequence motifs bound in vivo, provides insights into protein-RNA networks and how they impact mRNA structure, function, and stability. In this review, we will focus on proteins that bind to AU-rich elements (AREs) in nascent or mature mRNA where they play roles in response to stresses encountered by cancer cells. ARE-binding proteins (ARE-BPs) specifically impact alternative splicing, stability, decay and translation, and formation of RNA-rich biomolecular condensates like cytoplasmic stress granules (SGs). For example, recent findings highlight the role of ARE-BPs - like TIAR and HUR - in chemotherapy resistance and in translational regulation of mRNAs encoding pro-inflammatory cytokines. We will discuss emerging evidence that different modes of ARE-BP activity impact leukaemia and lymphoma development, progression, adaptation to microenvironment and chemotherapy resistance.
    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
    具有脑铁积累的神经变性(NBIA)是一种临床和遗传异质性疾病,其特征是在成年后基底神经节中铁沉积增加和神经系统进行性变性。然而,在儿童早期,没有进行早期诊断的特征性特征。在我们的研究中,一名女性儿童表现出全球发育迟缓,智力残疾,和高热惊厥,没有其他不同的临床表型。通过全外显子组测序(WES),从头无意义突变(c.726C>G,p。Tyr242Ter)在该孩子中鉴定出WDR45基因。她最终被诊断为β-螺旋桨蛋白相关神经变性(BPAN),最近发现的NBIA亚型之一。这种突变可能充当过早终止密码子(PSC),使突变的转录本被无义介导的mRNA衰变(NMD)降解,导致含PSC的mRNA水平降低。此外,通过微型基因剪接分析,这种突变可能导致前所未有的新转录本,WDR45的外显子9被无义相关剪接改变(NASA)排除.来自三重奏PBMC的总RNA的转录组测序(RNA-seq)揭示了患者中三种类型的选择性剪接事件。进一步的研究暗示铁转运基因的下调(TFRC,TFR2,SCARA5)可能是WDR45缺乏患者铁积累的潜在机制。这是关于NASA在WDR45中发生的第一份报告。这意味着接近剪接位点的无义突变可能通过一种以上的分子机制影响疾病的发病机理,在进行遗传咨询时应予以考虑。
    Neurodegeneration with brain iron accumulation (NBIA) is a clinically and genetically heterogeneous disease characterized by increased iron deposition in the basal ganglia and progressive degeneration of the nervous system in adulthood. However, in early childhood, there were no characteristic features to perform early diagnosis. In our study, a female child exhibited global developmental delay, intellectual disability, and febrile seizure without other distinct clinical phenotypes. Through whole exome sequencing (WES), a de novo nonsense mutation (c.726C > G, p. Tyr242Ter) of WDR45 gene was identified in this child. She was finally diagnosed as β-propeller protein-associated neurodegeneration (BPAN), one of the recently identified subtypes of NBIA. This mutation could act as a premature stop codon (PSC) which rendered the mutated transcripts to be degraded by nonsense-mediated mRNA decay (NMD), leading to decreased levels of PSC-containing mRNAs. Additionally, through mini-gene splicing assays, this mutation could result in an unprecedented novel transcript with the exon 9 of WDR45 excluded by nonsense-associated splicing alteration (NASA). Transcriptome sequencing (RNA-seq) on total RNAs from PBMCs of the trio revealed three types of alternative splicing events in the patient. Further research implied that downregulation of iron transport genes (TFRC, TFR2, SCARA5) might be the underlying mechanism for the iron accumulation in patients with deficient WDR45. This is the first report about NASA happening in WDR45. It implies that nonsense mutations approximal to splicing sites could affect the disease pathogenesis through more than one molecular mechanism and should be taken into consideration when conducting genetic counseling.
    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
    前mRNA剪接在两个步骤中催化:5'剪接位点(SS)切割和外显子连接。许多蛋白质与剪接体瞬时结合以特异性地影响这些步骤(第1和第2步骤因子)。我们最近确定Fyv6(人类中的FAM192A)是酿酒酵母中的第2步因子;然而,我们没有确定Fyv6对转录组的影响有多广泛。为了回答这个问题,我们已经使用RNA-Seq来分析剪接的变化。这些结果表明,Fyv6的丢失导致非共识的激活,分支点(BP)近端3'SS转录组范围。为了确定这些观察的分子基础,我们确定了含有Fyv6的酵母产品复合剪接体的高分辨率低温EM结构,其含量为2.3µ。该结构表明,Fyv6是与Prp22ATPase接触的唯一第2步因子,并且Fyv6的结合与第1步因子Yju2的结合是相互排斥的。然后,我们使用此结构来解剖Fyv6功能域,并解释fyv6Δ抑制子突变的遗传筛选结果。联合转录组,结构,遗传研究使我们能够提出一个模型,其中Yju2/Fyv6交换促进外显子连接,而Fyv6促进Prp22依赖性的使用,BP远端3'SS。
    Pre-mRNA splicing is catalyzed in two steps: 5\' splice site (SS) cleavage and exon ligation. A number of proteins transiently associate with spliceosomes to specifically impact these steps (1st and 2nd step factors). We recently identified Fyv6 (FAM192A in humans) as a 2nd step factor in S. cerevisiae; however, we did not determine how widespread Fyv6\'s impact is on the transcriptome. To answer this question, we have used RNA-seq to analyze changes in splicing. These results show that loss of Fyv6 results in activation of non-consensus, branch point (BP) proximal 3\' SS transcriptome-wide. To identify the molecular basis of these observations, we determined a high-resolution cryo-EM structure of a yeast product complex spliceosome containing Fyv6 at 2.3 Å. The structure reveals that Fyv6 is the only 2nd step factor that contacts the Prp22 ATPase and that Fyv6 binding is mutually exclusive with that of the 1st step factor Yju2. We then use this structure to dissect Fyv6 functional domains and interpret results of a genetic screen for fyv6Δ suppressor mutations. The combined transcriptomic, structural, and genetic studies allow us to propose a model in which Yju2/Fyv6 exchange facilitates exon ligation and Fyv6 promotes usage of consensus, BP distal 3\' SS.
    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
    SRSF1是SR蛋白家族的创始成员。它与其他SR蛋白可互换地用于体外pre-mRNA剪接,它调节各种可变剪接事件。SRSF1表达的失调有助于癌症和其他病理。这里,我们使用邻近标记和质谱法表征了SRSF1的相互作用组。这种方法产生了SRSF1样品中富集的190种蛋白质,独立于生物素标记结构域的N-或C-末端位置。检测到的蛋白质反映了SRSF1在前mRNA剪接中已建立的功能,并揭示了与剪接体蛋白质的其他连接,除了其他最近确定的功能。我们使用双分子荧光互补和体外结合测定验证了与剪接体RNA解旋酶DDX23/PRP28的强大相互作用。该相互作用由DDX23的N-末端RS样结构域以及RRM1和SRSF1的RS结构域两者介导。在前mRNA剪接期间,DDX23的ATP酶活性对于pre-B到B剪接体复合物转换以及从5'剪接位点释放U1snRNP至关重要。我们表明DDX23的N端结构域的RS样区域对于剪接体掺入很重要,而该结构域中较大的缺失会改变亚核定位。我们讨论了DDX23与SRSF1和其他SR蛋白的相互作用如何参与这些过程的调节。
    SRSF1 is the founding member of the SR protein family. It is required-interchangeably with other SR proteins-for pre-mRNA splicing in vitro, and it regulates various alternative splicing events. Dysregulation of SRSF1 expression contributes to cancer and other pathologies. Here, we characterized SRSF1\'s interactome using proximity labeling and mass spectrometry. This approach yielded 190 proteins enriched in the SRSF1 samples, independently of the N- or C-terminal location of the biotin-labeling domain. The detected proteins reflect established functions of SRSF1 in pre-mRNA splicing and reveal additional connections to spliceosome proteins, in addition to other recently identified functions. We validated a robust interaction with the spliceosomal RNA helicase DDX23/PRP28 using bimolecular fluorescence complementation and in vitro binding assays. The interaction is mediated by the N-terminal RS-like domain of DDX23 and both RRM1 and the RS domain of SRSF1. During pre-mRNA splicing, DDX23\'s ATPase activity is essential for the pre-B to B spliceosome complex transition and for release of U1 snRNP from the 5\' splice site. We show that the RS-like region of DDX23\'s N-terminal domain is important for spliceosome incorporation, while larger deletions in this domain alter subnuclear localization. We discuss how the identified interaction of DDX23 with SRSF1 and other SR proteins may be involved in the regulation of these processes.
    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
    前mRNA剪接在基因表达调控中起关键作用。最近的发现表明,前mRNA剪接的缺陷,由于某些剪接因子的功能障碍,可以影响对基因组监控机制至关重要的基因的表达,包括那些参与细胞对DNA损伤的反应。在这项研究中,我们分析了具有非功能性剪接体相关Gpl1-Gih35-Wdr83复合物的细胞对DNA损伤的反应.此外,我们研究了这种复合物在调节DNA损伤修复相关因子剪接中的作用。我们的发现表明,Gpl1-Gih35-Wdr83复合物中任何成分的缺失都会导致DNA修复因子未剪接的前mRNA的大量积累。因此,缺乏这种复合物的突变细胞对DNA损伤剂的敏感性增加。这些结果突出了Gpl1-Gih35-Wdr83复合物在调节DNA修复因子表达中的重要性,从而保护DNA损伤后基因组的稳定性。
    Pre-mRNA splicing plays a key role in the regulation of gene expression. Recent discoveries suggest that defects in pre-mRNA splicing, resulting from the dysfunction of certain splicing factors, can impact the expression of genes crucial for genome surveillance mechanisms, including those involved in cellular response to DNA damage. In this study, we analyzed how cells with a non-functional spliceosome-associated Gpl1-Gih35-Wdr83 complex respond to DNA damage. Additionally, we investigated the role of this complex in regulating the splicing of factors involved in DNA damage repair. Our findings reveal that the deletion of any component within the Gpl1-Gih35-Wdr83 complex leads to a significant accumulation of unspliced pre-mRNAs of DNA repair factors. Consequently, mutant cells lacking this complex exhibit increased sensitivity to DNA-damaging agents. These results highlight the importance of the Gpl1-Gih35-Wdr83 complex in regulating the expression of DNA repair factors, thereby protecting the stability of the genome following DNA damage.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Preprint
    剪接位点识别对于定义转录组至关重要。利沙普兰和兰纳普拉姆等药物改变了U1snRNP识别特定5'剪接位点(5'SS)的方式,并促进U1snRNP在这些位置的结合和剪接。尽管5种SS调节剂具有治疗潜力,它们相互作用和snRNP底物的复杂性排除了定义5'SS调制机制的可能性。我们已经结合了整体动力学测量和共定位单分子光谱学(CoSMoS),确定了通过branaplam调节-1A凸起的5'SS的顺序结合机制。我们的机制建立了U1-C蛋白与U1snRNP可逆结合,并且branaplam仅在与-1A凸出的5'SS接合后才与U1snRNP/U1-C复合物结合。结合和非结合的明确顺序解释了可逆的branaplam相互作用如何导致长寿命U1snRNP/5'SS复合物的形成。Branaplam是一种核糖核蛋白,不是单独的RNA双链体,靶向药物的作用取决于5'SS识别的基本性质。
    Splice site recognition is essential for defining the transcriptome. Drugs like risdiplam and branaplam change how U1 snRNP recognizes particular 5\' splice sites (5\'SS) and promote U1 snRNP binding and splicing at these locations. Despite the therapeutic potential of 5\'SS modulators, the complexity of their interactions and snRNP substrates have precluded defining a mechanism for 5\'SS modulation. We have determined a sequential binding mechanism for modulation of -1A bulged 5\'SS by branaplam using a combination of ensemble kinetic measurements and colocalization single molecule spectroscopy (CoSMoS). Our mechanism establishes that U1-C protein binds reversibly to U1 snRNP, and branaplam binds to the U1 snRNP/U1-C complex only after it has engaged a -1A bulged 5\'SS. Obligate orders of binding and unbinding explain how reversible branaplam interactions cause formation of long-lived U1 snRNP/5\'SS complexes. Branaplam is a ribonucleoprotein, not RNA duplex alone, targeting drug whose action depends on fundamental properties of 5\'SS recognition.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号