RNA Splicing

RNA 剪接
  • 文章类型: Journal Article
    LncRNA在癌症进展和靶向中起着至关重要的作用,但很难确定参与结直肠癌(CRC)进展的关键lncRNAs.我们使用21对IV期CRC组织和邻近正常组织将FAM83H-AS1鉴定为肿瘤促进相关lncRNA。体外和体内实验表明,在CRC细胞中敲低FAM83H-AS1抑制肿瘤的增殖和转移,反之亦然。m6A修饰对于通过作者METTL3和读者IGF2BP2/IGFBP3的FAM83H-AS1RNA稳定性至关重要。PTBP1-一种RNA结合蛋白-负责CRC中的FAM83H-AS1功能。FAM83H-AS1的外显子4上的T4(1770-2440nt)和T5(2440-2743nt)提供了PTBP1RRM2相互作用的平台。我们的结果表明,m6A修饰通过磷酸化PTBP1对其RNA剪接作用失调FAM83H-AS1致癌作用。在患者来源的异种移植模型中,ASO-FAM83H-AS1显著抑制胃肠道(GI)肿瘤的生长,不仅是CRC,还有GC和ESCC。ASO-FAM83H-AS1和奥沙利铂/顺铂的组合与单独使用任一种药剂的治疗相比显著抑制肿瘤生长。值得注意的是,所有这三种胃肠道癌均有病理完全缓解。我们的研究结果表明,FAM83H-AS1靶向治疗将使主要接受铂类药物治疗的胃肠道肿瘤患者受益。
    LncRNA plays a crucial role in cancer progression and targeting, but it has been difficult to identify the critical lncRNAs involved in colorectal cancer (CRC) progression. We identified FAM83H-AS1 as a tumor-promoting associated lncRNA using 21 pairs of stage IV CRC tissues and adjacent normal tissues. In vitro and in vivo experiments revealed that knockdown of FAM83H-AS1 in CRC cells inhibited tumor proliferation and metastasis, and vice versa. m6A modification is critical for FAM83H-AS1 RNA stability through the writer METTL3 and the readers IGF2BP2/IGFBP3. PTBP1-an RNA binding protein-is responsible for the FAM83H-AS1 function in CRC. T4 (1770-2440nt) and T5 (2440-2743nt) on exon 4 of FAM83H-AS1 provide a platform for PTBP1 RRM2 interactions. Our results demonstrated that m6A modification dysregulated the FAM83H-AS1 oncogenic role by phosphorylated PTBP1 on its RNA splicing effect. In patient-derived xenograft models, ASO-FAM83H-AS1 significantly suppressed the growth of gastrointestinal (GI) tumors, not only CRC but also GC and ESCC. The combination of ASO-FAM83H-AS1 and oxaliplatin/cisplatin significantly suppressed tumor growth compared with treatment with either agent alone. Notably, there was pathological complete response in all these three GI cancers. Our findings suggest that FAM83H-AS1 targeted therapy would benefit patients primarily receiving platinum-based therapy in GI cancers.
    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
    这项研究分析了临床数据,全外显子组测序结果,以及发育迟缓和智力障碍儿童的体外小基因功能实验。男病人,4岁,出生后3个月开始出现癫痫发作,并表现出发育迟缓。在1至2岁之间进行康复训练。没有其他重要的家庭病史。通过全面的家族外显子组基因检测,在受影响的儿童中鉴定出OPHN1基因第11外显子的半合子变异:c.1025+1G>A.家庭隔离分析证实了患者母亲中存在这种变异,以前没有报道过。根据ACMG指南,该变异被归类为可能的致病变异.作为对这种变体的回应,设计并进行了体外小基因功能实验,证实突变影响基因mRNA的正常剪接,导致在Intron11的左侧保留56bp。已证实OPHN1:c.1025+1G>A是儿童X连锁智力障碍的致病原因,临床表型包括发育迟缓和癫痫发作。
    This research analyzes the clinical data, whole-exome sequencing results, and in vitro minigene functional experiments of a child with developmental delay and intellectual disability. The male patient, aged 4, began experiencing epileptic seizures at 3 months post-birth and has shown developmental delay. Rehabilitation training was administered between the ages of one and two. There were no other significant family medical histories. Through comprehensive family exome genetic testing, a hemizygous variant in the 11th exon of the OPHN1 gene was identified in the affected child: c.1025 + 1G > A. Family segregation analysis confirmed the presence of this variant in the patient\'s mother, which had not been previously reported. According to the ACMG guidelines, this variant was classified as a likely pathogenic variant. In response to this variant, an in vitro minigene functional experiment was designed and conducted, confirming that the mutation affects the normal splicing of the gene\'s mRNA, resulting in a 56 bp retention on the left side of Intron 11. It was confirmed that OPHN1: c.1025 + 1G > A is the pathogenic cause of X-linked intellectual disabilities in the child, with clinical phenotypes including developmental delay and seizures.
    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-seq带来了关于RNA加工中的畸变的重大发现,这些RNA变异与多种疾病有关。RNA中的异常剪接和单核苷酸变体(SNV)已被证明可以改变转录物的稳定性,本地化,和功能。特别是,ADAR的上调,一种介导腺苷到肌苷编辑的酶,先前已与肺腺癌细胞的侵袭性增加有关,并与剪接调节有关。尽管研究剪接和SNV的功能重要性,短读RNA-seq的使用限制了社区同时询问两种形式的RNA变异的能力。
    我们采用长读测序技术来获得全长转录物序列,在单分子水平上阐明变体对剪接变化的顺式效应。我们开发了一个计算工作流程来增强FLAIR,调用以长读数据表示的同工型模型的工具,将RNA变体调用与携带它们的相关同种型整合在一起。我们从具有和不具有ADAR敲低的H1975肺腺癌细胞产生具有高序列准确性的纳米孔数据。我们应用我们的工作流程来确定关键的肌苷同工型关联,以帮助阐明ADAR在肿瘤发生中的重要性。
    最终,我们发现长篇阅读方法为表征RNA变体和剪接模式之间的关系提供了有价值的见解。
    RNA-seq has brought forth significant discoveries regarding aberrations in RNA processing, implicating these RNA variants in a variety of diseases. Aberrant splicing and single nucleotide variants (SNVs) in RNA have been demonstrated to alter transcript stability, localization, and function. In particular, the upregulation of ADAR, an enzyme that mediates adenosine-to-inosine editing, has been previously linked to an increase in the invasiveness of lung adenocarcinoma cells and associated with splicing regulation. Despite the functional importance of studying splicing and SNVs, the use of short-read RNA-seq has limited the community\'s ability to interrogate both forms of RNA variation simultaneously.
    We employ long-read sequencing technology to obtain full-length transcript sequences, elucidating cis-effects of variants on splicing changes at a single molecule level. We develop a computational workflow that augments FLAIR, a tool that calls isoform models expressed in long-read data, to integrate RNA variant calls with the associated isoforms that bear them. We generate nanopore data with high sequence accuracy from H1975 lung adenocarcinoma cells with and without knockdown of ADAR. We apply our workflow to identify key inosine isoform associations to help clarify the prominence of ADAR in tumorigenesis.
    Ultimately, we find that a long-read approach provides valuable insight toward characterizing the relationship between RNA variants and splicing patterns.
    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
    白细胞介素-17(IL-17)家族细胞因子促进保护性炎症抵抗病原体,而且还促进自身免疫和肿瘤的发展。IL-17对调节性T细胞(Tregs)的直接信号尚未报道,可能有助于解释这些二分法反应。
    我们通过将Foxp3-YFP-Cre小鼠与Il17ra-flox小鼠(Il17raΔTreg小鼠)杂交,在Treg中产生了Il17ra的条件性敲除。随后,我们将Il17raΔTreg小鼠的骨髓细胞过继转移到散发性结直肠癌的小鼠模型(Cdx2-Cre/ApcF/),在结直肠癌中选择性消融IL-17对Tregs的直接信号传导。对来自小鼠结肠直肠肿瘤的纯化Treg进行单细胞RNA测序和批量RNA测序,并与人类肿瘤浸润的Treg细胞进行比较。
    IL-17受体A(IL-17RA)在存在于小鼠肠系膜淋巴结和结肠肿瘤中的Treg中表达。IL-17RA消融,特别是在Tregs中,导致Th17细胞增加,并加剧了肿瘤的发展。机械上,肿瘤浸润性Tregs表现出与其激活相关的独特基因特征,成熟,和抑制函数,并且该签名部分由IL-17向Tregs的直接信号传导支持。为了研究Treg编程的途径,我们发现肿瘤Tregs中IL-17RA的缺失导致RNA剪接减少,和下调几种RNA结合蛋白,已知这些蛋白可以调节可变剪接并促进Treg功能。
    IL-17直接向Tregs发出信号,并促进其成熟和功能。该信号传导途径构成了一个负反馈回路,可控制CRC中的促癌炎症。
    UNASSIGNED: Interleukin-17 (IL-17) family cytokines promote protective inflammation for pathogen resistance, but also facilitate autoimmunity and tumor development. A direct signal of IL-17 to regulatory T cells (Tregs) has not been reported and may help explain these dichotomous responses.
    UNASSIGNED: We generated a conditional knockout of Il17ra in Tregs by crossing Foxp3-YFP-Cre mice to Il17ra-flox mice (Il17ra ΔTreg mice). Subsequently, we adoptively transferred bone marrow cells from Il17ra ΔTreg mice to a mouse model of sporadic colorectal cancer (Cdx2-Cre +/Apc F/+), to selectively ablate IL-17 direct signaling on Tregs in colorectal cancer. Single cell RNA sequencing and bulk RNA sequencing were performed on purified Tregs from mouse colorectal tumors, and compared to those of human tumor infiltrating Treg cells.
    UNASSIGNED: IL-17 Receptor A (IL-17RA) is expressed in Tregs that reside in mouse mesenteric lymph nodes and colon tumors. Ablation of IL-17RA, specifically in Tregs, resulted in increased Th17 cells, and exacerbated tumor development. Mechanistically, tumor-infiltrating Tregs exhibit a unique gene signature that is linked to their activation, maturation, and suppression function, and this signature is in part supported by the direct signaling of IL-17 to Tregs. To study pathways of Treg programming, we found that loss of IL-17RA in tumor Tregs resulted in reduced RNA splicing, and downregulation of several RNA binding proteins that are known to regulate alternative splicing and promote Treg function.
    UNASSIGNED: IL-17 directly signals to Tregs and promotes their maturation and function. This signaling pathway constitutes a negative feedback loop that controls cancer-promoting inflammation in CRC.
    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:短读单细胞RNA测序(scRNA-seq)已用于研究细胞异质性,细胞命运,和转录动力学。对scRNA-seq数据中的剪接动力学建模具有挑战性,甚至在阐明从中提取测序片段的分子的剪接状态的看似简单的任务中也存在固有的困难。这个困难出现了,在某种程度上,从有限的读取长度和位置偏差,这大大降低了测序片段的特异性。因此,由于缺乏明确的证据,scRNA-seq中许多读段的剪接状态是不明确的.因此,我们需要可以恢复不明确读段的剪接状态的方法,反过来,可以提高下游分析的准确性和置信度。
    结果:我们开发了Forseti,一个预测模型,以概率方式将剪接状态分配给scRNA-seq读取。我们的模型有两个关键组成部分。首先,我们训练了一个结合亲和力模型,以指定在片段生成中使用给定转录组位点的概率。第二,我们拟合了一个强大的片段长度分布模型,该模型可以很好地推广来自不同物种和组织类型的数据集。Forseti组合这两个训练模型以通过对推定的片段进行评分来预测读段起源分子的剪接状态,该推定的片段将测序读段的每个比对与最接近的潜在引发位点相关联。利用模拟和实验数据,我们表明,我们的模型可以精确地预测许多读段的剪接状态,并确定多基因定位读段的真实基因起源。
    方法:Forseti和用于生成结果的代码可在https://github.com/COMBINE-lab/forseti上获得BSD3-clause许可证。
    BACKGROUND: Short-read single-cell RNA-sequencing (scRNA-seq) has been used to study cellular heterogeneity, cellular fate, and transcriptional dynamics. Modeling splicing dynamics in scRNA-seq data is challenging, with inherent difficulty in even the seemingly straightforward task of elucidating the splicing status of the molecules from which sequenced fragments are drawn. This difficulty arises, in part, from the limited read length and positional biases, which substantially reduce the specificity of the sequenced fragments. As a result, the splicing status of many reads in scRNA-seq is ambiguous because of a lack of definitive evidence. We are therefore in need of methods that can recover the splicing status of ambiguous reads which, in turn, can lead to more accuracy and confidence in downstream analyses.
    RESULTS: We develop Forseti, a predictive model to probabilistically assign a splicing status to scRNA-seq reads. Our model has two key components. First, we train a binding affinity model to assign a probability that a given transcriptomic site is used in fragment generation. Second, we fit a robust fragment length distribution model that generalizes well across datasets deriving from different species and tissue types. Forseti combines these two trained models to predict the splicing status of the molecule of origin of reads by scoring putative fragments that associate each alignment of sequenced reads with proximate potential priming sites. Using both simulated and experimental data, we show that our model can precisely predict the splicing status of many reads and identify the true gene origin of multi-gene mapped reads.
    METHODS: Forseti and the code used for producing the results are available at https://github.com/COMBINE-lab/forseti under a BSD 3-clause license.
    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
    I型干扰素(IFN-Is)通过引发IFN刺激基因(ISGs)的表达,在人类免疫缺陷病毒I(HIV-1)的先天免疫中至关重要。其中包括有效的宿主限制因子。虽然ISGs通过靶向病毒生命周期的各个阶段来限制宿主细胞内的病毒复制,鲜为人知的IFN抑制基因(IRepGs),包括RNA结合蛋白(RBPs),通过改变对有效的HIV-1基因表达至关重要的宿主依赖性因子的表达来影响病毒复制。宿主限制和依赖性因素决定了病毒复制效率;然而,目前,对与HIV-1感染有关的IRepGs的了解仍然非常有限.这篇综述提供了关于RNA结合蛋白家族影响的当前理解的全面概述。特别是剪接相关蛋白SRSF和hnRNP的两个家族,HIV-1基因表达和病毒复制。由于最近的发现特别表明SRSF1和hnRNPA0在各种细胞系和原代细胞中受到IFN-I的调节,包括肠固有层单核细胞(LPMC)和外周血单核细胞(PBMC),我们特别讨论了它们在先天免疫影响HIV-1复制的背景下的作用.
    Type I interferons (IFN-Is) are pivotal in innate immunity against human immunodeficiency virus I (HIV-1) by eliciting the expression of IFN-stimulated genes (ISGs), which encompass potent host restriction factors. While ISGs restrict the viral replication within the host cell by targeting various stages of the viral life cycle, the lesser-known IFN-repressed genes (IRepGs), including RNA-binding proteins (RBPs), affect the viral replication by altering the expression of the host dependency factors that are essential for efficient HIV-1 gene expression. Both the host restriction and dependency factors determine the viral replication efficiency; however, the understanding of the IRepGs implicated in HIV-1 infection remains greatly limited at present. This review provides a comprehensive overview of the current understanding regarding the impact of the RNA-binding protein families, specifically the two families of splicing-associated proteins SRSF and hnRNP, on HIV-1 gene expression and viral replication. Since the recent findings show specifically that SRSF1 and hnRNP A0 are regulated by IFN-I in various cell lines and primary cells, including intestinal lamina propria mononuclear cells (LPMCs) and peripheral blood mononuclear cells (PBMCs), we particularly discuss their role in the context of the innate immunity affecting HIV-1 replication.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Case Reports
    X连锁低磷酸盐血症(XLH)是一种罕见的遗传性肾脏磷酸盐消耗疾病,具有由PHEX基因功能丧失变体引起的高度可变的表型。具有轻度表型的个体的诊断可能是具有挑战性的并且经常延迟。这里,我们描述了一个XLH临床表现非常轻微的三代家族。该诊断是在一名39岁的妇女中意外发现的,该妇女由于儿童对肾小管病的诊断不清楚而被转诊进行基因检测。通过使用肾脏疾病基因小组的下一代测序进行的遗传测试鉴定了PHEX基因中的新型非规范剪接位点变体。隔离分析检测到咨询和\\的父亲,出现低磷酸盐血症和肾小管磷酸盐重吸收减少的患者,咨询公司的儿子也携带了这种变体。RNA研究表明,非常规剪接位点变异部分改变了PHEX基因的剪接,因为在只有一个PHEX基因拷贝的两个雄性成员中检测到野生型和异常剪接转录本。总之,这种情况有助于理解剪接变异与XLH疾病的可变表达之间的关系。该家族的轻度表型可以通过PHEX转录物与异常和野生型剪接的共存来解释。
    X-linked hypophosphatemia (XLH) is a rare inherited disorder of renal phosphate wasting with a highly variable phenotype caused by loss-of-function variants in the PHEX gene. The diagnosis of individuals with mild phenotypes can be challenging and often delayed. Here, we describe a three-generation family with a very mild clinical presentation of XLH. The diagnosis was unexpectedly found in a 39-year-old woman who was referred for genetic testing due to an unclear childhood diagnosis of a tubulopathy. Genetic testing performed by next-generation sequencing using a kidney disease gene panel identified a novel non-canonical splice site variant in the PHEX gene. Segregation analysis detected that the consultand\'s father, who presented with hypophosphatemia and decreased tubular phosphate reabsorption, and the consultand\'s son also carried this variant. RNA studies demonstrated that the non-canonical splice site variant partially altered the splicing of the PHEX gene, as both wild-type and aberrant splicing transcripts were detected in the two male members with only one copy of the PHEX gene. In conclusion, this case contributes to the understanding of the relationship between splicing variants and the variable expressivity of XLH disease. The mild phenotype of this family can be explained by the coexistence of PHEX transcripts with aberrant and wild-type 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
    目的:集落刺激因子1受体(CSF1R)相关性白质脑病是由CSF1R基因突变引起的一种进展迅速的神经退行性疾病。本研究旨在鉴定和研究CSF1R的一个新的内含子突变(c.1754-3C>G)对剪接的影响。
    方法:使用全外显子组测序鉴定了一种新的内含子突变。为了研究这种突变的影响,我们使用各种生物信息学工具来分析CSF1R基因的转录及其编码蛋白的三维结构。此外,进行逆转录聚合酶链反应(RT-PCR)以验证研究结果.
    结果:在CSF1R中发现了一个新的突变(c.1754-3C>G),由于3'剪接位点共有序列NYAG/G的破坏,导致外显子13跳跃。通过RT-PCR和Sanger测序在突变携带者的外周血中进一步验证了该外显子跳跃事件。蛋白质结构预测表明酪氨酸激酶结构域的破坏,截短的蛋白质显示出显著的结构改变。
    结论:我们的发现强调了内含子错误剪接突变在CSF1R相关白质脑病诊断和治疗中的重要性。
    OBJECTIVE: Colony stimulating factor 1 receptor (CSF1R)-related leukoencephalopathy is a rapidly progressing neurodegenerative disease caused by CSF1R gene mutations. This study aimed to identify and investigate the effect of a novel intronic mutation (c.1754-3C>G) of CSF1R on splicing.
    METHODS: A novel intronic mutation was identified using whole-exome sequencing. To investigate the impact of this mutation, we employed various bioinformatics tools to analyze the transcription of the CSF1R gene and the three-dimensional structure of its encoded protein. Furthermore, reverse transcription polymerase chain reaction (RT-PCR) was performed to validate the findings.
    RESULTS: A novel mutation (c.1754-3C>G) in CSF1R was identified, which results in exon 13 skipping due to the disruption of the 3\' splice site consensus sequence NYAG/G. This exon skipping event was further validated in the peripheral blood of the mutation carrier through RT-PCR and Sanger sequencing. Protein structure prediction indicated a disruption in the tyrosine kinase domain, with the truncated protein showing significant structural alterations.
    CONCLUSIONS: Our findings underscore the importance of intronic mis-splicing mutations in the diagnosis and management of CSF1R-related leukoencephalopathy.
    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
    核苷酸转换RNA测序技术询问细胞转录物中的化学RNA修饰,导致包含错配的读取。将所得读段映射到参考基因组的偏差仍然知之甚少。我们介绍splice_sim,剪接感知RNA-seq模拟和评估管道,以设定频率引入用户定义的核苷酸转换,创建转换和未转换读取的混合模型,并计算每个基因组注释的映射精度。通过在现实实验条件下模拟核苷酸转换RNA-seq数据集,包括代谢RNA标记和RNA亚硫酸氢盐测序,我们测量了小鼠和人类转录本的最先进的剪接阅读映射器的作图精度,并得出了防止数据解释偏差的策略。
    Nucleotide conversion RNA sequencing techniques interrogate chemical RNA modifications in cellular transcripts, resulting in mismatch-containing reads. Biases in mapping the resulting reads to reference genomes remain poorly understood. We present splice_sim, a splice-aware RNA-seq simulation and evaluation pipeline that introduces user-defined nucleotide conversions at set frequencies, creates mixture models of converted and unconverted reads, and calculates mapping accuracies per genomic annotation. By simulating nucleotide conversion RNA-seq datasets under realistic experimental conditions, including metabolic RNA labeling and RNA bisulfite sequencing, we measure mapping accuracies of state-of-the-art spliced-read mappers for mouse and human transcripts and derive strategies to prevent biases in the data interpretation.
    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
    U4snRNP在前信使RNA(mRNA)剪接中的重要作用已被充分确立。在这项研究中,我们利用特异性靶向U4snRNA的反义吗啉代寡核苷酸(AMO)在HeLa细胞中实现U4snRNP的功能性敲低。我们的结果表明,这种敲除导致全球内含子过早裂解和聚腺苷酸化(PCPA)事件,与U1AMO治疗观察到的效果相当,如mRNA3'-seq分析所示。此外,我们的研究表明,这可能是人类和小鼠细胞系中的常见现象。此外,我们表明U4AMO处理破坏了转录延伸,通过RNAPII的染色质免疫沉淀测序(ChIP-seq)分析证明。总的来说,我们的结果确定了U4snRNP在抑制PCPA中的独特作用,并表明了一个模型,其中剪接在共转录mRNA加工的背景下固有地抑制内含子切割和聚腺苷酸化.
    The essential role of U4 snRNP in pre-messenger RNA (mRNA) splicing has been well established. In this study, we utilized an antisense morpholino oligonucleotide (AMO) specifically targeting U4 snRNA to achieve functional knockdown of U4 snRNP in HeLa cells. Our results showed that this knockdown resulted in global intronic premature cleavage and polyadenylation (PCPA) events, comparable to the effects observed with U1 AMO treatment, as demonstrated by mRNA 3\'-seq analysis. Furthermore, our study suggested that this may be a common phenomenon in both human and mouse cell lines. Additionally, we showed that U4 AMO treatment disrupted transcription elongation, as evidenced by chromatin immunoprecipitation sequencing (ChIP-seq) analysis for RNAPII. Collectively, our results identified a unique role for U4 snRNP in the inhibition of PCPA and indicated a model wherein splicing intrinsically inhibits intronic cleavage and polyadenylation in the context of cotranscriptional mRNA processing.
    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