U1 snRNA

U1 snRNA
  • 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
    人UlsnRNA由多基因家族编码,所述多基因家族由转录的变体和缺陷的假基因组成。已经证明许多变体U1(vU1)snRNA不仅可以转录,而且可以通过添加三甲基化鸟苷帽进行处理,打包到snRNPs中,组装成剪接体,然而,它们促进前mRNA剪接的能力,到目前为止,没有经过测试。最近对人类snRNA基因的系统分析鉴定了178个U1snRNA基因,这些基因在基因组中以串联阵列或多个染色体上的单个基因存在。其中,发现15在人体组织和细胞系中表达,尽管其内源性基因座的水平明显较低,小于规范U1snRNA的0.001%。在这项研究中,我们发现,将变体置于RNA1-1基因调控元件的背景下,可将许多变体的表达提高到与标准U1snRNA相当的水平.应用先前建立的基于HeLa细胞的小基因报告基因测定来检查vU1snRNA支持pre-mRNA剪接的能力表明,即使外源表达的变体snRNA在细胞核中富集,只有少数对剪接有可测量的影响。
    The human U1 snRNA is encoded by a multigene family consisting of transcribed variants and defective pseudogenes. Many variant U1 (vU1) snRNAs have been demonstrated to not only be transcribed but also processed by the addition of a trimethylated guanosine cap, packaged into snRNPs, and assembled into spliceosomes; however, their capacity to facilitate pre-mRNA splicing has, so far, not been tested. A recent systematic analysis of the human snRNA genes identified 178 U1 snRNA genes that are present in the genome as either tandem arrays or single genes on multiple chromosomes. Of these, 15 were found to be expressed in human tissues and cell lines, although at significantly low levels from their endogenous loci, <0.001% of the canonical U1 snRNA. In this study, we found that placing the variants in the context of the regulatory elements of the RNU1-1 gene improves the expression of many variants to levels comparable to the canonical U1 snRNA. Application of a previously established HeLa cell-based minigene reporter assay to examine the capacity of the vU1 snRNAs to support pre-mRNA splicing revealed that even though the exogenously expressed variant snRNAs were enriched in the nucleus, only a few had a measurable effect on splicing.
    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
    CNOT1基因突变导致一种无法治愈的罕见神经系统疾病,主要表现为智力障碍的临床谱,发育迟缓,癫痫发作,和行为问题。在这项研究中,我们研究了与神经发育障碍相关的CNOT1的经典剪接位点变异(c.1343+1G>T),这是一个主调节器,协调基因表达,RNA去端化,和蛋白质泛素化。为了将CNOT1功能障碍与患者中观察到的神经发育表型联系起来,体外小基因试验用于验证CNOT1基因剪接位点变异c.1343+1G>T对mRNA剪接的影响。我们还探索了瞬时转染引入修饰的U1snRNA对校正剪接变体的影响。通过小基因在哺乳动物细胞中的表达,我们证明了该变体诱导了完整的外显子12跳跃,这解释了患者的临床状况,并为该变异的临床意义提供了额外的遗传诊断证据。此外,我们证实了异常剪接模式可以通过在mRNA水平上修饰的U1snRNA部分校正,这为修饰的U1snRNA在中和错误剪接模式的危险作用方面的治疗潜力提供了强有力的证据。
    Mutations in the CNOT1 gene lead to an incurable rare neurological disorder mainly manifested as a clinical spectrum of intellectual disability, developmental delay, seizures, and behavioral problems. In this study, we investigated a classical splice site variant of CNOT1 (c.1343+1G>T) associated with neurodevelopmental disorders, which was a master regulator, orchestrating gene expression, RNA deadenylation, and protein ubiquitination. To link CNOT1 dysfunction with the neurodevelopmental phenotype observed in a patient, in vitro minigene assay was used to verify the effect of CNOT1 gene splice site variant c.1343+1G>T on mRNA splicing. We also explored the impact of transient transfection introducing modified U1 snRNA on correcting the splicing variant. Through minigene expression in mammalian cells, we demonstrated that the variant induced complete exon 12 skipping, which explained the patient\'s clinical condition and provided additional genetic diagnosis evidence for the clinical significance of the variant. Moreover, we confirmed that the aberrant splice pattern could be partially corrected by the modified U1 snRNA at the mRNA level, which provided strong evidence for the therapeutic potential of modified U1 snRNA in neutralizing the hazardous effect of incorrect splicing patterns.
    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
    与编码转录物相比,不稳定的非编码RNA(ncRNA)的转录终止知之甚少。我们最近确定ZC3H4-WDR82(“限制器”)限制人类ncRNA转录,但它是如何做到这一点是未知的。这里,我们显示ZC3H4还与ARS2和核外泌体靶向复合物相关。接触ARS2和WDR82的ZC3H4结构域是ncRNA限制所必需的,表明它们存在于功能性复合体中。始终如一,ZC3H4、WDR82和ARS2共同转录控制重叠的ncRNAs群体。ZC3H4接近负延伸因子,PNUTS,我们显示它能够实现限制器功能,并且是终止所有主要RNA聚合酶II转录物类别的转录所必需的。与短ncRNAs相反,较长的蛋白质编码转录由U1snRNA支持,它在数百个基因上屏蔽了来自限制器和PNUTS的转录本。这些数据提供了对限制器和PNUTS转录的机制和控制的重要见解。
    The transcriptional termination of unstable non-coding RNAs (ncRNAs) is poorly understood compared to coding transcripts. We recently identified ZC3H4-WDR82 (\"restrictor\") as restricting human ncRNA transcription, but how it does this is unknown. Here, we show that ZC3H4 additionally associates with ARS2 and the nuclear exosome targeting complex. The domains of ZC3H4 that contact ARS2 and WDR82 are required for ncRNA restriction, suggesting their presence in a functional complex. Consistently, ZC3H4, WDR82, and ARS2 co-transcriptionally control an overlapping population of ncRNAs. ZC3H4 is proximal to the negative elongation factor, PNUTS, which we show enables restrictor function and is required to terminate the transcription of all major RNA polymerase II transcript classes. In contrast to short ncRNAs, longer protein-coding transcription is supported by U1 snRNA, which shields transcripts from restrictor and PNUTS at hundreds of genes. These data provide important insights into the mechanism and control of transcription by restrictor and PNUTS.
    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
    功效和安全性考虑因素构成了体内基因疗法开发过程中的必要步骤。在这里,我们评估了基于剪接因子的治疗方法在Opa1突变小鼠系中纠正突变诱导的剪接缺陷的疗效和安全性.我们将腺相关病毒应用于视网膜。病毒用设计用于纠正Opa1剪接缺陷的工程化U1snRNA剪接因子转导视网膜细胞。我们发现该治疗可有效增加野生型Opa1转录本。相应地,接受治疗的眼睛中Opa1蛋白水平显着增加。视网膜形态和功能的测量未显示与治疗相关的副作用,支持治疗的短期安全性。未检测到潜在脱靶基因的改变。我们的数据表明,应用工程化U1snRNA治疗剪接缺陷代表了一种有前途的体内治疗方法。该疗法增加了野生型Opa1转录本和蛋白质水平,没有可检测到的形态学,小鼠眼睛的功能或遗传副作用。U1基于snRNA的疗法可以针对特定的疾病基因突变进行定制,因此,提高了这种有前途的技术在治疗不同遗传性视网膜疾病方面的更广泛适用性的可能性。
    Efficacy and safety considerations constitute essential steps during development of in vivo gene therapies. Herein, we evaluated efficacy and safety of splice factor-based treatments to correct mutation-induced splice defects in an Opa1 mutant mouse line. We applied adeno-associated viruses to the retina. The viruses transduced retinal cells with an engineered U1 snRNA splice factor designed to correct the Opa1 splice defect. We found the treatment to be efficient in increasing wild-type Opa1 transcripts. Correspondingly, Opa1 protein levels increased significantly in treated eyes. Measurements of retinal morphology and function did not reveal therapy-related side-effects supporting the short-term safety of the treatment. Alterations of potential off-target genes were not detected. Our data suggest that treatments of splice defects applying engineered U1 snRNAs represent a promising in vivo therapeutic approach. The therapy increased wild-type Opa1 transcripts and protein levels without detectable morphological, functional or genetic side-effects in the mouse eye. The U1 snRNA-based therapy can be tailored to specific disease gene mutations, hence, raising the possibility of a wider applicability of this promising technology towards treatment of different inherited retinal diseases.
    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
    在衰老中,骨骼肌力量和再生能力下降,部分由于肌肉干细胞(MuSCs)的功能损害,然而,潜在的机制仍然难以捉摸。这里,我们利用大规模细胞计数来鉴定高CD47表达是功能失调的MuSCs(CD47hi)的一个标志,其再生能力受损,且以衰老为主.普遍的CD47hiMuSC子集通过旁分泌信号环抑制残留的功能性CD47loMuSC子集,导致扩散受损。我们发现,CD47水平升高在老年MuSCs的结果是增加U1snRNA表达,这破坏了替代性的聚腺苷酸化。通过吗啉代介导的CD47选择性多腺苷酸化阻断或血小板反应蛋白1/CD47信号传导的抗体阻断可以克服衰老MuSC再生功能的缺陷,导致老年小鼠的再生改善,具有治疗意义。我们的发现强调了MuSCs中CD47水平和功能的先前未被识别的年龄依赖性改变,这是在衰老过程中肌肉修复减少的基础。
    In aging, skeletal muscle strength and regenerative capacity decline, due in part to functional impairment of muscle stem cells (MuSCs), yet the underlying mechanisms remain elusive. Here, we capitalize on mass cytometry to identify high CD47 expression as a hallmark of dysfunctional MuSCs (CD47hi) with impaired regenerative capacity that predominate with aging. The prevalent CD47hi MuSC subset suppresses the residual functional CD47lo MuSC subset through a paracrine signaling loop, leading to impaired proliferation. We uncover that elevated CD47 levels on aged MuSCs result from increased U1 snRNA expression, which disrupts alternative polyadenylation. The deficit in aged MuSC function in regeneration can be overcome either by morpholino-mediated blockade of CD47 alternative polyadenylation or antibody blockade of thrombospondin-1/CD47 signaling, leading to improved regeneration in aged mice, with therapeutic implications. Our findings highlight a previously unrecognized age-dependent alteration in CD47 levels and function in MuSCs, which underlies reduced muscle repair in aging.
    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
    可变剪接占转录组多样性的很大一部分,因为大多数蛋白质编码基因被剪接成多个mRNA亚型。然而,剪接模式中的错误会导致错误剪接,并带来病理后果,如先天性疾病家族性自主神经障碍,杜氏肌营养不良症,和脊髓性肌萎缩症.已提出UsnRNP家族的小核RNA(snRNA)组分作为治疗错误剪接的治疗方式。U1snRNAs提供了巨大的希望,先前的研究证明了体内功效,提示额外的临床前发展是值得的。使能技术的改进,包括筛选方法,基因传递载体,基因编辑方法的相关考虑证明了U1snRNA作为治疗和研究工具的进一步发展。为了提供用户友好的协议,我们编译并展示了一个方法学工具包,用于用工程化U1snRNA对选择性剪接的pre-mRNA进行序列特异性靶向扰动。我们观察到在两个对比剪接环境中靶向的内源性pre-mRNA转录物的强大调节,SMN2外显子7和FAS外显子6,表现出工程化U1snRNA对包含和排除靶向外显子的效用和适用性。我们预计这些演示将有助于U1snRNA在研究真核细胞剪接调节中的可用性,增加更广泛的研究社区的可及性。
    Alternative splicing accounts for a considerable portion of transcriptomic diversity, as most protein-coding genes are spliced into multiple mRNA isoforms. However, errors in splicing patterns can give rise to mis-splicing with pathological consequences, such as the congenital diseases familial dysautonomia, Duchenne muscular dystrophy, and spinal muscular atrophy. Small nuclear RNA (snRNA) components of the U snRNP family have been proposed as a therapeutic modality for the treatment of mis-splicing. U1 snRNAs offer great promise, with prior studies demonstrating in vivo efficacy, suggesting additional preclinical development is merited. Improvements in enabling technologies, including screening methodologies, gene delivery vectors, and relevant considerations from gene editing approaches justify further advancement of U1 snRNA as a therapeutic and research tool. With the goal of providing a user-friendly protocol, we compile and demonstrate a methodological toolkit for sequence-specific targeted perturbation of alternatively spliced pre-mRNA with engineered U1 snRNAs. We observe robust modulation of endogenous pre-mRNA transcripts targeted in two contrasting splicing contexts, SMN2 exon 7 and FAS exon 6, exhibiting the utility and applicability of engineered U1 snRNA to both inclusion and exclusion of targeted exons. We anticipate that these demonstrations will contribute to the usability of U1 snRNA in investigating splicing modulation in eukaryotic cells, increasing accessibility to the broader research community.
    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
    遗传性视网膜营养不良是由超过250个基因的突变引起的,他们中的每一个携带几种类型的突变,可以导致不同的临床表型。色素性视网膜炎GTP酶调节因子(RPGR)的突变导致X连锁的色素性视网膜炎(RP)。RPGR内含子9中的核苷酸取代导致成熟mRNA的可变剪接同种型增加,轴承外显子9a(E9a)。这引入了一个终止密码子,导致蛋白质截短。旨在恢复受损的基因表达,我们开发了一种基于反义RNA的RPGRE9a跳跃治疗方法。我们设计了一组特定的U1反义snRNAs(U1_asRNAs),并在体外测试了它们的功效,在HEK-293T中与RPGR小基因报告系统瞬时共转染后,661W,和PC-12细胞系。因此,我们鉴定了三种有效介导E9a跳跃的嵌合U1_asRNA,纠正遗传缺陷。出乎意料的是,U1-5'反义构建体,在PC-12细胞中表现出最高的外显子跳跃效率,在HEK-293T和661W细胞中诱导E9a包含,表明在测试RNA剪接校正疗法时,临床前模型系统的选择要谨慎。我们的数据为基于U1_snRNA外显子跳跃的方法在RPGR中纠正剪接缺陷提供了原理证明。
    Inherited retinal dystrophies are caused by mutations in more than 250 genes, each of them carrying several types of mutations that can lead to different clinical phenotypes. Mutations in Retinitis Pigmentosa GTPase-Regulator (RPGR) cause X-linked Retinitis pigmentosa (RP). A nucleotide substitution in intron 9 of RPGR causes the increase of an alternatively spliced isoform of the mature mRNA, bearing exon 9a (E9a). This introduces a stop codon, leading to truncation of the protein. Aiming at restoring impaired gene expression, we developed an antisense RNA-based therapeutic approach for the skipping of RPGR E9a. We designed a set of specific U1 antisense snRNAs (U1_asRNAs) and tested their efficacy in vitro, upon transient cotransfection with RPGR minigene reporter systems in HEK-293T, 661W, and PC-12 cell lines. We thus identified three chimeric U1_asRNAs that efficiently mediate E9a skipping, correcting the genetic defect. Unexpectedly, the U1-5\'antisense construct, which exhibited the highest exon-skipping efficiency in PC-12 cells, induced E9a inclusion in HEK-293T and 661W cells, indicating caution in the choice of preclinical model systems when testing RNA splicing-correcting therapies. Our data provide a proof of principle for the application of U1_snRNA exon skipping-based approach to correct splicing defects in RPGR.
    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
    热休克(HS)反应涉及HS基因的快速诱导,而转录抑制在大多数其他基因中建立得更慢。以前的数据表明,这种抑制是由抑制RNA聚合酶II(RNAPII)暂停释放引起的,但在这里,我们表明HS强烈影响转录周期的其他阶段。有趣的是,而伸长率在HS时增加,加工能力显著下降,因此RNAPII经常无法到达基因的末端。的确,HS导致广泛的转录本在隐秘时提前终止,基因5'末端附近的内含子多腺苷酸化(IPA)位点,可能是通过抑制U1的电话脚本。这导致人类转录组的戏剧性重新配置,产生新的,以前没有注释,在细胞核中积累的短mRNA。一起,这些结果为高温下生长诱导的基本转录机制提供了新的思路,并表明在生理条件下可以发生全基因组向IPA位点使用的转变。
    The heat shock (HS) response involves rapid induction of HS genes, whereas transcriptional repression is established more slowly at most other genes. Previous data suggested that such repression results from inhibition of RNA polymerase II (RNAPII) pause release, but here, we show that HS strongly affects other phases of the transcription cycle. Intriguingly, while elongation rates increase upon HS, processivity markedly decreases, so that RNAPII frequently fails to reach the end of genes. Indeed, HS results in widespread premature transcript termination at cryptic, intronic polyadenylation (IPA) sites near gene 5\'-ends, likely via inhibition of U1 telescripting. This results in dramatic reconfiguration of the human transcriptome with production of new, previously unannotated, short mRNAs that accumulate in the nucleus. Together, these results shed new light on the basic transcription mechanisms induced by growth at elevated temperature and show that a genome-wide shift toward usage of IPA sites can occur under physiological conditions.
    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
    剪接改变已在肿瘤中广泛记录,其中癌细胞的增殖和播散由异常同种型变体的表达支持。剪接是由剪接体催化的,协调内含子去除和外显子连接的复杂过程的核糖核蛋白复合物。近年来,剪接体组件U1snRNA中的反复热点突变,SF3B1和U2AF1已在不同的肿瘤类型中被鉴定。原则上,这种突变对细胞非常有害,因为所有三个剪接体组件对于准确的剪接位点选择至关重要:U1snRNA对于3'剪接位点识别至关重要,SF3B1和U2AF1对于5'剪接位点选择很重要。尽管如此,它们似乎被选中来促进特定类型的癌症。这里,我们回顾了目前对癌症中这些突变的分子理解,重点关注它们如何影响剪接位点的选择和对癌症发展的影响。
    Splicing alterations have been widely documented in tumors where the proliferation and dissemination of cancer cells is supported by the expression of aberrant isoform variants. Splicing is catalyzed by the spliceosome, a ribonucleoprotein complex that orchestrates the complex process of intron removal and exon ligation. In recent years, recurrent hotspot mutations in the spliceosome components U1 snRNA, SF3B1, and U2AF1 have been identified across different tumor types. Such mutations in principle are highly detrimental for cells as all three spliceosome components are crucial for accurate splice site selection: the U1 snRNA is essential for 5′ splice site recognition, and SF3B1 and U2AF1 are important for 3′ splice site selection. Nonetheless, they appear to be selected to promote specific types of cancers. Here, we review the current molecular understanding of these mutations in cancer, focusing on how they influence splice site selection and impact on cancer development.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号