RNA, Small Nuclear

RNA,小型核
  • 文章类型: Journal Article
    剪接体通过四个连续阶段执行pre-mRNA剪接:组装,激活,催化作用,和拆卸。剪接体的激活,即将预催化剪接体(B复合物)重塑为活化剪接体(Bact复合物)和催化活化剪接体(B*复合物),涉及蛋白质成分和结构重排的主要流动。依靠剪接抑制剂,我们已经捕获了B和B*复合物之间的六个中间状态:pre-Bact,Bact-I,Bact-II,Bact-III,Bact-IV,和事后。他们的低温EM结构,以及催化步骤I剪接体(C络合物)的改进结构,揭示了催化中心如何在U6snRNA的内部茎环周围成熟,分支站点如何接近5'-拼接站点,RNA解旋酶PRP2如何重排以结合pre-mRNA,以及U2snRNP如何经历显著的运动以促进激活。我们确定了PRP2在剪接体激活中先前未被识别的关键作用。我们的研究概括了人类剪接体在催化活化过程中的分子编排。
    The spliceosome executes pre-mRNA splicing through four sequential stages: assembly, activation, catalysis, and disassembly. Activation of the spliceosome, namely remodeling of the pre-catalytic spliceosome (B complex) into the activated spliceosome (Bact complex) and the catalytically activated spliceosome (B* complex), involves major flux of protein components and structural rearrangements. Relying on a splicing inhibitor, we have captured six intermediate states between the B and B* complexes: pre-Bact, Bact-I, Bact-II, Bact-III, Bact-IV, and post-Bact. Their cryo-EM structures, together with an improved structure of the catalytic step I spliceosome (C complex), reveal how the catalytic center matures around the internal stem loop of U6 snRNA, how the branch site approaches 5\'-splice site, how the RNA helicase PRP2 rearranges to bind pre-mRNA, and how U2 snRNP undergoes remarkable movement to facilitate activation. We identify a previously unrecognized key role of PRP2 in spliceosome activation. Our study recapitulates a molecular choreography of the human spliceosome during its catalytic activation.
    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
    U6snRNA是富含尿苷的非编码RNA之一,在各种细胞中丰富而稳定,在内含子-套索剪接体(ILS)复合物中充当核心粒子。多倍体1-1D(ILP1)和NTC相关蛋白1(NTR1)水平升高,ILS复合体的两个保守分解因子,在完成内含子剪接后促进ILS复合物的分解。ILP1和NTR1的功能损害导致U6水平升高,而包含ILS复合物的其他snRNA未受影响。我们发现ILP1和NTR1对转录没有影响,U6snRNA的3'端磷酸结构或寡(U)尾。此外,我们发现ILP1和NTR1的突变导致ILS复合物的积累,阻碍U6与剪接因子的分离,导致U6的半衰期延长,并最终导致U6snRNA水平升高。我们的发现拓宽了对ILS拆卸因子ILP1和NTR1的功能的理解,并提供了对U6和ILS之间动态拆卸的见解。
    U6 snRNA is one of the uridine-rich non-coding RNAs, abundant and stable in various cells, function as core particles in the intron-lariat spliceosome (ILS) complex. The Increased Level of Polyploidy1-1D (ILP1) and NTC-related protein 1 (NTR1), two conserved disassembly factors of the ILS complex, facilitates the disintegration of the ILS complex after completing intron splicing. The functional impairment of ILP1 and NTR1 lead to increased U6 levels, while other snRNAs comprising the ILS complex remained unaffected. We revealed that ILP1 and NTR1 had no impact on the transcription, 3\' end phosphate structure or oligo(U) tail of U6 snRNA. Moreover, we uncovered that the mutation of ILP1 and NTR1 resulted in the accumulation of ILS complexes, impeding the dissociation of U6 from splicing factors, leading to an extended half-life of U6 and ultimately causing an elevation in U6 snRNA levels. Our findings broaden the understanding of the functions of ILS disassembly factors ILP1 and NTR1, and providing insights into the dynamic disassembly between U6 and ILS.
    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
    HP1α/CBX5是一种表观遗传调节因子,在多发性硬化症(MS)中具有可疑的作用。这里,在单核细胞上使用高深度RNA测序,我们确定了一个CBX5表达减少的MS患者子集,与疾病的进展阶段和广泛的转录组改变相关。对这些患者中罕见的非编码RNA物种的检查显示UsnRNA和增强子RNA的成熟/降解受损,表示积分器的活动减少,与MS风险增加有可疑联系的复合体。在蛋白质编码基因,整合子活性受损,表现为前mRNA剪接效率降低和RNA聚合酶II暂停释放调节的基因表达改变。小鼠中Cbx5的失活反映了大多数这些转录缺陷,并导致对实验性自身免疫性脑脊髓炎的超敏反应。总的来说,我们的观察表明,整合复合物在保护MS转录异常特征方面做出了重大贡献,HP1α/CBX5成为这种复合物活性的意想不到的调节剂。这些发现为MS的转录方面带来了新的见解,并为患者分层提供了潜在的新标准。
    HP1α/CBX5 is an epigenetic regulator with a suspected role in multiple sclerosis (MS). Here, using high-depth RNA sequencing on monocytes, we identified a subset of MS patients with reduced CBX5 expression, correlating with progressive stages of the disease and extensive transcriptomic alterations. Examination of rare non-coding RNA species in these patients revealed impaired maturation/degradation of U snRNAs and enhancer RNAs, indicative of reduced activity of the Integrator, a complex with suspected links to increased MS risk. At protein-coding genes, compromised Integrator activity manifested in reduced pre-mRNA splicing efficiency and altered expression of genes regulated by RNA polymerase II pause-release. Inactivation of Cbx5 in the mouse mirrored most of these transcriptional defects and resulted in hypersensitivity to experimental autoimmune encephalomyelitis. Collectively, our observations suggested a major contribution of the Integrator complex in safeguarding against transcriptional anomalies characteristic of MS, with HP1α/CBX5 emerging as an unexpected regulator of this complex\'s activity. These findings bring novel insights into the transcriptional aspects of MS and provide potential new criteria for patient stratification.
    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
    前信使RNA(pre-mRNA)剪接调节是研究由错误剪接引起的遗传疾病机制的有吸引力的方法。先前的报道已经表明,修饰的U7小核RNA(U7snRNA)是用于在体外和体内调节剪接的预期工具。迄今为止,很少有研究研究反义序列长度在修饰的U7snRNA中的作用。在这项研究中,我们设计了一系列不同长度的反义序列,并评估了它们诱导剪接调节的效率。为了表达修饰的U7snRNAs,我们构建了一系列编码巨细胞病毒(CMV)增强子的质粒DNA序列,人U1启动子,和具有不同长度的反义序列的修饰的小鼠U7snRNA。我们使用荧光素酶报告系统评估了体外剪接调节效率,以进行简单而精确的评估以及逆转录聚合酶链反应来监测剪接模式。我们的体外分析结果表明,修饰的小鼠U7snRNA的反义序列具有有效剪接调节的最佳长度,这取决于目标外显子。此外,太长或太短的反义序列降低剪接调节效率。为了确认再现性,我们使用两个靶基因进行了体外测定,小鼠Fas和小鼠Dmd。一起,我们的数据表明,反义序列长度应针对修饰的小鼠U7snRNA进行优化,以诱导有效的剪接调节.
    Pre-messenger RNA (pre-mRNA) splicing modulation is an attractive approach for investigating the mechanisms of genetic disorders caused by mis-splicing. Previous reports have indicated that a modified U7 small nuclear RNA (U7 snRNA) is a prospective tool for modulating splicing both in vitro and in vivo. To date, very few studies have investigated the role of antisense sequence length in modified U7 snRNA. In this study, we designed a series of antisense sequences with various lengths and evaluated their efficiency in inducing splicing modulation. To express modified U7 snRNAs, we constructed a series of plasmid DNA sequences which codes cytomegalovirus (CMV) enhancer, human U1 promoter, and modified mouse U7 snRNAs with antisense sequences of different lengths. We evaluated in vitro splicing modulation efficiency using a luciferase reporter system for simple and precise evaluation as well as reverse transcription-polymerase chain reaction to monitor splicing patterns. Our in vitro assay findings suggest that antisense sequences of modified mouse U7 snRNAs have an optimal length for efficient splicing modulation, which depends on the target exon. In addition, antisense sequences that were either too long or too short decreased splicing modulation efficiency. To confirm reproducibility, we performed an in vitro assay using two target genes, mouse Fas and mouse Dmd. Together, our data suggests that the antisense sequence length should be optimized for modified mouse U7 snRNAs to induce efficient splicing modulation.
    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剪接需要组装,重塑,以及称为剪接体1的多兆核糖核蛋白复合物的拆解。最近的研究为催化2-6的剪接体组装和重塑提供了启示,但拆解机制尚不清楚。这里,我们报告了线虫和人类末端内含子-套索剪接体的2.6至3.2µ分辨率的低温电子显微镜结构以及生化和遗传数据。我们的结果揭示了四个拆解因子和保守的RNA解旋酶DHX15如何启动剪接体拆解。拆卸因子探测大的内部和外部剪接体表面以检测连接的mRNA的释放。其中两个因素,TFIP11和C19L1,以及三个通用剪接体亚基,SYF1,SYF2和SDE2,然后在催化U6snRNA上对接并激活DHX15以启动拆卸。因此,U6控制前mRNA剪接的开始5和结束。一起来看,我们的结果解释了典型剪接体拆解的分子基础,并提供了一个框架来理解一般剪接体RNA解旋酶的控制和异常剪接体的丢弃。
    Precursor-mRNA (pre-mRNA) splicing requires the assembly, remodelling and disassembly of the multi-megadalton ribonucleoprotein complex called the spliceosome1. Recent studies have shed light on spliceosome assembly and remodelling for catalysis2-6, but the mechanism of disassembly remains unclear. Here we report cryo-electron microscopy structures of nematode and human terminal intron lariat spliceosomes along with biochemical and genetic data. Our results uncover how four disassembly factors and the conserved RNA helicase DHX15 initiate spliceosome disassembly. The disassembly factors probe large inner and outer spliceosome surfaces to detect the release of ligated mRNA. Two of these factors, TFIP11 and C19L1, and three general spliceosome subunits, SYF1, SYF2 and SDE2, then dock and activate DHX15 on the catalytic U6 snRNA to initiate disassembly. U6 therefore controls both the start5 and end of pre-mRNA splicing. Taken together, our results explain the molecular basis of the initiation of canonical spliceosome disassembly and provide a framework to understand general spliceosomal RNA helicase control and the discard of aberrant spliceosomes.
    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
    卵巢癌通常会对常规疗法产生耐药性,妨碍他们的有效性。这里,使用在化疗前后获得的体外配对卵巢癌腹水和体外治疗诱导的分泌体,我们表明,卵巢癌细胞在治疗后分泌的分子促进顺铂耐药并增强受体癌细胞的DNA损伤修复。即使化疗卵巢癌细胞与治疗诱导的分泌体的短期孵育也会诱导类似于长期治疗后在化学抗性患者来源的肿瘤细胞中观察到的变化。使用综合组学技术,我们发现离体和体外治疗诱导的分泌体富含剪接体成分,它从细胞核重新定位到细胞质,然后在治疗后进入细胞外囊泡。我们证明了这些分子对治疗诱导的分泌体的表型效应有实质性贡献。因此,SNU13和SYNCRIP剪接体蛋白促进治疗耐药,而外源性U12和U6atacsnRNAs刺激肿瘤生长。这些发现证明了治疗过程中剪接体网络扰动的重要性,并进一步强调了细胞外信号传导可能是导致卵巢癌治疗耐药性出现的关键因素。
    Ovarian cancer often develops resistance to conventional therapies, hampering their effectiveness. Here, using ex vivo paired ovarian cancer ascites obtained before and after chemotherapy and in vitro therapy-induced secretomes, we show that molecules secreted by ovarian cancer cells upon therapy promote cisplatin resistance and enhance DNA damage repair in recipient cancer cells. Even a short-term incubation of chemonaive ovarian cancer cells with therapy-induced secretomes induces changes resembling those that are observed in chemoresistant patient-derived tumor cells after long-term therapy. Using integrative omics techniques, we find that both ex vivo and in vitro therapy-induced secretomes are enriched with spliceosomal components, which relocalize from the nucleus to the cytoplasm and subsequently into the extracellular vesicles upon treatment. We demonstrate that these molecules substantially contribute to the phenotypic effects of therapy-induced secretomes. Thus, SNU13 and SYNCRIP spliceosomal proteins promote therapy resistance, while the exogenous U12 and U6atac snRNAs stimulate tumor growth. These findings demonstrate the significance of spliceosomal network perturbation during therapy and further highlight that extracellular signaling might be a key factor contributing to the emergence of ovarian cancer therapy 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
    剪接体使用一个催化中心进行两个连续的酯交换反应,因此需要在拼接的两个催化步骤之间进行重排。Prp16ATPase通过使一些对催化重要的相互作用不稳定而促进从催化中心的第一步构象中退出。为了更好地了解酿酒酵母催化中心内的重排,我们描述了调节Prp16:Cwc2,Prp8的N末端结构域和U6-41AACAAU46区域功能的因素。通过纠正prp16-302等位基因cs缺陷的突变体的遗传筛选来鉴定这些因子的等位基因。几个鉴定的U6,cwc2和prp8等位基因在剪接体催化构象的低温-EM结构中彼此紧密靠近。Cwc2和U6在第一步中与内含子序列相互作用,但它们似乎对第二步催化中心的稳定性没有贡献。另一方面,Prp8的N端片段不仅影响第一步的内含子定位,但是对于拼接的第一和第二步,它也在活性位点附近进行了重要的接触。通过确定对催化构象稳定性重要的相互作用,我们的遗传分析间接告知我们剪接体过渡态构象的特征。
    The spliceosome performs two consecutive transesterification reactions using one catalytic center, thus requiring its rearrangement between the two catalytic steps of splicing. The Prp16 ATPase facilitates exit from the first-step conformation of the catalytic center by destabilizing some interactions important for catalysis. To better understand rearrangements within the Saccharomyces cerevisiae catalytic center, we characterize factors that modulate the function of Prp16: Cwc2, N-terminal domain of Prp8, and U6-41AACAAU46 region. Alleles of these factors were identified through genetic screens for mutants that correct cs defects of prp16-302 alleles. Several of the identified U6, cwc2, and prp8 alleles are located in close proximity of each other in cryo-EM structures of the spliceosomal catalytic conformations. Cwc2 and U6 interact with the intron sequences in the first step, but they do not seem to contribute to the stability of the second-step catalytic center. On the other hand, the N-terminal segment of Prp8 not only affects intron positioning for the first step, but it also makes important contacts in the proximity of the active site for both the first and second steps of splicing. By identifying interactions important for the stability of catalytic conformations, our genetic analyses indirectly inform us about features of the transition-state conformation of the spliceosome.
    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
    单细胞RNA测序(scRNA-seq)是否捕获与单核RNA测序(snRNA-seq)相同的生物信息仍然不确定,并且可能与上下文相关。在这里,在匹配的正常腺癌人肺样本中进行了头对头比较,以评估scRNA-seq和snRNA-seq的生物学见解,并更好地了解从正常组织到肿瘤组织的细胞转变.这里,获得了160,621个细胞/细胞核的转录组。在非肿瘤肺,scRNA-seq和snRNA-seq之间的细胞类型比例差异很大,前者以免疫细胞为主(81.5%),后者以上皮细胞为主(69.9%)。在腺癌中观察到类似的结果,除了细胞类型异质性的总体增加和上皮起源细胞中拷贝数变异的更普遍之外,这表明恶性分配。无论检查细胞还是细胞核,从正常肺组织到腺癌发生的细胞类型转变并不总是一致的。不出所料,观察到全细胞和核转录组的大差异表达,但是配对的正常和肿瘤肺样本的细胞类型特异性变化揭示了一组参与癌症相关途径的细胞和细胞核中的共同基因。此外,我们发现,肺腺癌的配体-受体相互作用组景观在评估细胞或细胞核方面存在很大差异.新鲜标本中的免疫细胞消耗部分减轻了细胞和细胞核之间观察到的细胞类型组成的差异。然而,额外的操作影响了细胞活力,并放大了与应激反应相关的转录特征。总之,专注于绘制肺腺癌免疫景观的研究应用受益于新鲜样本中的scRNA-seq,而冷冻样本的snRNA-seq提供了一种低成本的替代方法来分析更多的上皮细胞和癌细胞,并产生与组织含量更紧密匹配的细胞类型比例。
    Whether single-cell RNA-sequencing (scRNA-seq) captures the same biological information as single-nucleus RNA-sequencing (snRNA-seq) remains uncertain and likely to be context-dependent. Herein, a head-to-head comparison was performed in matched normal-adenocarcinoma human lung samples to assess biological insights derived from scRNA-seq versus snRNA-seq and better understand the cellular transition that occurs from normal to tumoral tissue. Here, the transcriptome of 160,621 cells/nuclei was obtained. In non-tumor lung, cell type proportions varied widely between scRNA-seq and snRNA-seq with a predominance of immune cells in the former (81.5%) and epithelial cells (69.9%) in the later. Similar results were observed in adenocarcinomas, in addition to an overall increase in cell type heterogeneity and a greater prevalence of copy number variants in cells of epithelial origin, which suggests malignant assignment. The cell type transition that occurs from normal lung tissue to adenocarcinoma was not always concordant whether cells or nuclei were examined. As expected, large differential expression of the whole-cell and nuclear transcriptome was observed, but cell-type specific changes of paired normal and tumor lung samples revealed a set of common genes in the cells and nuclei involved in cancer-related pathways. In addition, we showed that the ligand-receptor interactome landscape of lung adenocarcinoma was largely different whether cells or nuclei were evaluated. Immune cell depletion in fresh specimens partly mitigated the difference in cell type composition observed between cells and nuclei. However, the extra manipulations affected cell viability and amplified the transcriptional signatures associated with stress responses. In conclusion, research applications focussing on mapping the immune landscape of lung adenocarcinoma benefit from scRNA-seq in fresh samples, whereas snRNA-seq of frozen samples provide a low-cost alternative to profile more epithelial and cancer cells, and yield cell type proportions that more closely match tissue content.
    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
    剪接体小核RNA(snRNAs)是真核细胞核质中丰富的一类基本的非编码小RNA,在剪接前体信使RNA(前mRNA)中起着至关重要的作用。它们通过DNA依赖性RNA聚合酶II(PolII)或III(PolIII)转录,并经历随后的加工和3'末端切割以成为成熟的snRNA。许多蛋白质因子参与转录起始,伸长率,终止,拼接,细胞定位,和snRNA的末端修饰过程。snRNAs的转录和加工通过各种机制在时空上进行调控。细胞内snRNAs的稳态平衡对生物体的生长发育具有重要意义。snRNA与特定的辅助蛋白组装以形成小的核核糖核蛋白颗粒(snRNP),其是负责前mRNA成熟的剪接体的基础组分。本文概述了生物功能,生物合成,终端结构,和snRNAs的组织特异性调控。
    Spliceosomal small nuclear RNAs (snRNAs) are a fundamental class of non-coding small RNAs abundant in the nucleoplasm of eukaryotic cells, playing a crucial role in splicing precursor messenger RNAs (pre-mRNAs). They are transcribed by DNA-dependent RNA polymerase II (Pol II) or III (Pol III), and undergo subsequent processing and 3\' end cleavage to become mature snRNAs. Numerous protein factors are involved in the transcription initiation, elongation, termination, splicing, cellular localization, and terminal modification processes of snRNAs. The transcription and processing of snRNAs are regulated spatiotemporally by various mechanisms, and the homeostatic balance of snRNAs within cells is of great significance for the growth and development of organisms. snRNAs assemble with specific accessory proteins to form small nuclear ribonucleoprotein particles (snRNPs) that are the basal components of spliceosomes responsible for pre-mRNA maturation. This article provides an overview of the biological functions, biosynthesis, terminal structure, and tissue-specific regulation of snRNAs.
    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
    烟酰胺腺嘌呤二核苷酸(NAD)是细胞代谢的关键成分,也是各种RNA的替代帽。然而,NADRNA帽的功能仍在研究中。我们研究了HIV-1感染细胞中RNA的NAD加帽,因为HIV-1负责NAD/NADH细胞池的消耗并引起细胞内糙皮病。通过将NADcaptureSeq协议应用于HIV-1感染和未感染的细胞,我们揭示了四个snRNAs(例如,U1)和四个snoRNA在感染HIV-1时失去了NAD上限。这里,我们提供的证据表明,NAD帽的存在会降低U1/HIV-1前mRNA双链体的稳定性。此外,我们证明了通过过表达NADRNA去盖酶DXO减少NAD加帽RNA的量导致HIV-1感染性增加。这表明NAD封端对HIV-1不利,并在其感染性中起作用。
    Nicotinamide adenine dinucleotide (NAD) is a critical component of the cellular metabolism and also serves as an alternative 5\' cap on various RNAs. However, the function of the NAD RNA cap is still under investigation. We studied NAD capping of RNAs in HIV-1-infected cells because HIV-1 is responsible for the depletion of the NAD/NADH cellular pool and causing intracellular pellagra. By applying the NAD captureSeq protocol to HIV-1-infected and uninfected cells, we revealed that four snRNAs (e.g., U1) and four snoRNAs lost their NAD cap when infected with HIV-1. Here, we provide evidence that the presence of the NAD cap decreases the stability of the U1/HIV-1 pre-mRNA duplex. Additionally, we demonstrate that reducing the quantity of NAD-capped RNA by overexpressing the NAD RNA decapping enzyme DXO results in an increase in HIV-1 infectivity. This suggests that NAD capping is unfavorable for HIV-1 and plays a role in its infectivity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号