ADAR

ADAR
  • 文章类型: Journal Article
    腺苷-肌苷(A-to-I)RNA编辑是由腺苷脱氨酶作用于RNA(ADAR)家族酶介导的重要转录后修饰,通过将RNA分子中选定的核苷酸A改变为I来扩展转录组。最近,已经探索了使用治疗性指导寡核苷酸在特定位点指导ADAR编辑来纠正RNA中的致病突变。人类有两个活跃的ADAR,其偏好和特异性尚未得到很好的理解。为了研究它们的底物特异性,我们分别介绍了hADAR1和hADAR2,进入裂殖酵母(S.pombe),缺乏内源性ADAR,并评估了它们在体内的编辑活性。使用在最佳生长温度(30°C)下培养的S.pombe的转录组测序,与未编辑的野生型对照菌株相比,我们为hADAR1鉴定了483个A-to-I高置信度编辑位点,为hADAR2鉴定了404个位点.然而,这些位点在hADAR1和hADAR2表达菌株之间大多是不同的,共享33个常见位点,每个菌株的比例低于9%。它们对底物的差异特异性归因于它们对编辑位点的相邻序列的差异偏好。我们发现在相对于编辑站点的-3位置,HDAR1表现出一种向T的趋势,而hADAR2倾向于A。此外,当改变表达hADAR1-和hADAR2的菌株的生长温度时,我们观察到在20和35°C时它们的编辑位点增加,与它们在30°C下生长相比。然而,我们没有观察到hADAR1和hADAR2在三个温度下对相邻序列的偏好发生显著变化。在较低和较高的温度下,RNA编辑位点的巨大变化也被观察到之前在出芽酵母中的hADAR2,这可能是由于在这些不同温度下RNA折叠的影响,在许多其他因素中。我们注意到编辑位点周围较长长度的dsRNA的例子,这些例子在20或35°C诱导编辑,但在其他两个温度条件下不存在。我们发现基因的功能会受到转录本编辑的极大影响,其中50%以上的HADAR1和HADAR2的RNA编辑位点都在编码序列(CDS)中,其中60%以上导致蛋白质产品的氨基酸变化。这项研究揭示了两种活性人类ADARS之间底物选择性的广泛差异,即,ADAR1和ADAR2,并在使用RNA编辑方法利用两种不同的酶在体内治疗人类遗传疾病时提供了新的见解。
    Adenosine-to-inosine (A-to-I) RNA editing is an important post-transcriptional modification mediated by the adenosine deaminases acting on RNA (ADAR) family of enzymes, expanding the transcriptome by altering selected nucleotides A to I in RNA molecules. Recently, A-to-I editing has been explored for correcting disease-causing mutations in RNA using therapeutic guide oligonucleotides to direct ADAR editing at specific sites. Humans have two active ADARs whose preferences and specificities are not well understood. To investigate their substrate specificity, we introduced hADAR1 and hADAR2, respectively, into Schizosaccharomyces pombe (S. pombe), which lacks endogenous ADARs, and evaluated their editing activities in vivo. Using transcriptome sequencing of S. pombe cultured at optimal growth temperature (30 °C), we identified 483 A-to-I high-confident editing sites for hADAR1 and 404 for hADAR2, compared with the non-editing wild-type control strain. However, these sites were mostly divergent between hADAR1 and hADAR2-expressing strains, sharing 33 common sites that are less than 9% for each strain. Their differential specificity for substrates was attributed to their differential preference for neighboring sequences of editing sites. We found that at the -3-position relative to the editing site, hADAR1 exhibits a tendency toward T, whereas hADAR2 leans toward A. Additionally, when varying the growth temperature for hADAR1- and hADAR2-expressing strains, we observed increased editing sites for them at both 20 and 35 °C, compared with them growing at 30 °C. However, we did not observe a significant shift in hADAR1 and hADAR2\'s preference for neighboring sequences across three temperatures. The vast changes in RNA editing sites at lower and higher temperatures were also observed for hADAR2 previously in budding yeast, which was likely due to the influence of RNA folding at these different temperatures, among many other factors. We noticed examples of longer lengths of dsRNA around the editing sites that induced editing at 20 or 35 °C but were absent at the other two temperature conditions. We found genes\' functions can be greatly affected by editing of their transcripts, for which over 50% of RNA editing sites for both hADAR1 and hADAR2 in S. pombe were in coding sequences (CDS), with more than 60% of them resulting in amino acid changes in protein products. This study revealed the extensive differences in substrate selectivity between the two active human ADARS, i.e., ADAR1 and ADAR2, and provided novel insight when utilizing the two different enzymes for in vivo treatment of human genetic diseases using the RNA editing approach.
    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
    果蝇精子发生的动态过程涉及不对称分裂,有丝分裂,和减数分裂,最终导致成熟精子的产生。精子发生障碍可导致男性不育。果蝇中的ADAR(腺苷脱氨酶作用于RNA)突变会导致男性不育,然而,致病因素仍不清楚。在这项研究中,免疫荧光染色用于观察内源性ADAR蛋白,并通过荧光强度分析评估蛋白水平.此外,通过对转运放大区域长度的定量,检查了睾丸早期精子发生过程中的早期分化障碍和稳态改变,计数GSC(生殖干细胞)的数量,和生育实验。我们的发现表明,ADAR的缺失会导致睾丸尖端转运扩增细胞在老年雄性果蝇中积聚并变得不育。通过在早期生殖系细胞中过表达ADAR,男性不育可以部分救助。转录组分析表明,ADAR通过骨形态发生蛋白(BMP)信号通路维持早期精子发生稳态。一起来看,这些发现有可能有助于探索ADAR在早期精子发生中的作用。
    The dynamic process of Drosophila spermatogenesis involves asymmetric division, mitosis, and meiosis, which ultimately results in the production of mature spermatozoa. Disorders of spermatogenesis can lead to infertility in males. ADAR (adenosine deaminase acting on RNA) mutations in Drosophila cause male infertility, yet the causative factors remain unclear. In this study, immunofluorescence staining was employed to visualize endogenous ADAR proteins and assess protein levels via fluorescence-intensity analysis. In addition, the early differentiation disorders and homeostatic alterations during early spermatogenesis in the testes were examined through quantification of transit-amplifying region length, counting the number of GSCs (germline stem cells), and fertility experiments. Our findings suggest that deletion of ADAR causes testicular tip transit-amplifying cells to accumulate and become infertile in older male Drosophila. By overexpressing ADAR in early germline cells, male infertility can be partially rescued. Transcriptome analysis showed that ADAR maintained early spermatogenesis homeostasis through the bone-morphogenetic-protein (BMP) signaling pathway. Taken together, these findings have the potential to help explore the role of ADAR in early spermatogenesis.
    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
    尽管A到IRNA编辑导致与A到GDNA突变类似的效果,非同义RNA编辑(重新编码)被认为通过以时空方式调节蛋白质组多样性而赋予其适应性,避免基因组突变的多效性效应。昆虫Adar基因中Ser>Gly自动编辑位点的进化轨迹的最新发现表明,与不可编辑的密码子相比,具有可编辑的密码子具有选择性优势。然而,除了纯粹的观察,仍然缺乏证明单个RNA编辑位点适应性的定量方法。我们对113种双翅目进行了比较基因组分析,专注于果蝇的AdarSer>Gly自动重新编码位点。我们只在相应的地点发现了一个具有衍生Gly的物种,这种情况明显低于全基因组随机预期。这表明AdarSer>Gly位点在进化过程中不太可能被G基因组取代,从而表明可编辑状态优于硬连线基因组等位基因。对于Syt1基因中保守的Ile>Met重新编码观察到类似的趋势。从进化的角度来看,我们建立了一种比较基因组方法,用于定量证明单个编辑位点的适应性。在未来的功能研究中,应优先考虑此类自适应编辑站点。
    Although A-to-I RNA editing leads to similar effects to A-to-G DNA mutation, nonsynonymous RNA editing (recoding) is believed to confer its adaptiveness by \'epigenetically\' regulating proteomic diversity in a temporospatial manner, avoiding the pleiotropic effect of genomic mutations. Recent discoveries on the evolutionary trajectory of Ser>Gly auto-editing site in insect Adar gene demonstrated a selective advantage to having an editable codon compared to uneditable ones. However, apart from pure observations, quantitative approaches for justifying the adaptiveness of individual RNA editing sites are still lacking. We performed a comparative genomic analysis on 113 Diptera species, focusing on the Adar Ser>Gly auto-recoding site in Drosophila. We only found one species having a derived Gly at the corresponding site, and this occurrence was significantly lower than genome-wide random expectation. This suggests that the Adar Ser>Gly site is unlikely to be genomically replaced with G during evolution, and thus indicating the advantage of editable status over hardwired genomic alleles. Similar trends were observed for the conserved Ile>Met recoding in gene Syt1. In the light of evolution, we established a comparative genomic approach for quantitatively justifying the adaptiveness of individual editing sites. Priority should be given to such adaptive editing sites in future functional studies.
    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
    Adar介导的腺苷到肌苷(A到I)RNA编辑主要发生在细胞核中,并以灵活的方式使转录组多样化。从整个编辑事件的海洋中识别有益的编辑站点一直是一项具有挑战性的任务。昆虫Adar基因中的功能Ser>Gly自动重新编码位点在祖先节点中具有不可编辑的Ser密码子,指示具有可编辑状态的选择性优势。这里,我们将这个案例研究扩展到更多的后生动物物种,并寻找所有具有潜在不可编辑同义密码子的果蝇重新编码事件。有趣的是,在黑腹D中,丰富的非同义词编辑丰富了具有不可编辑对应物的密码子,但AdarSer>Gly案例表明,其他物种中的可编辑直系同源密码子不一定被编辑。使用可编辑密码子与祖先不可编辑密码子是推断RNA编辑的选择性优势的明智方法,由于构建不可编辑等位基因的可行性,因此可能优先考虑这些编辑位点进行功能研究。我们的研究提出了一个想法,以缩小有利的重新编码站点的候选范围。同时,我们强调,需要匹配的转录组来验证进化过程中编辑事件的保守性。
    Adar-mediated adenosine-to-inosine (A-to-I) RNA editing mainly occurs in nucleus and diversifies the transcriptome in a flexible manner. It has been a challenging task to identify beneficial editing sites from the sea of total editing events. The functional Ser>Gly auto-recoding site in insect Adar gene has uneditable Ser codons in ancestral nodes, indicating the selective advantage to having an editable status. Here, we extended this case study to more metazoan species, and also looked for all Drosophila recoding events with potential uneditable synonymous codons. Interestingly, in D. melanogaster, the abundant nonsynonymous editing is enriched in the codons that have uneditable counterparts, but the Adar Ser>Gly case suggests that the editable orthologous codons in other species are not necessarily edited. The use of editable versus ancestral uneditable codon is a smart way to infer the selective advantage of RNA editing, and priority might be given to these editing sites for functional studies due to the feasibility to construct an uneditable allele. Our study proposes an idea to narrow down the candidates of beneficial recoding sites. Meanwhile, we stress that the matched transcriptomes are needed to verify the conservation of editing events during evolution.
    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
    ADAR(作用于RNA的腺苷脱氨酶)蛋白是一组酶,通过将腺苷转化为RNA中的肌苷,在RNA编辑中起着至关重要的作用。该过程是在后生动物转录物中观察到的频繁的转录后事件。最近的研究表明,ADAR介导的RNA编辑在许多免疫相关疾病中普遍存在失调。比如人类癌症。我们全面回顾了ADAR作为模式识别者的功能及其介导免疫相关途径的能力。我们还强调了位点特异性RNA编辑在维持稳态及其与各种疾病的关系中的潜在作用。比如人类癌症。更重要的是,我们总结了用于预测和分析RNA编辑位点的最新尖端计算方法和数据资源。最后,我们介绍了网站定向ADAR编辑工具开发的最新进展。这篇综述介绍了ADAR介导的RNA编辑的最新概述,位点特异性RNA编辑如何潜在地影响疾病病理学,以及如何将它们用于治疗应用。
    ADAR (Adenosine Deaminases Acting on RNA) proteins are a group of enzymes that play a vital role in RNA editing by converting adenosine to inosine in RNAs. This process is a frequent post-transcriptional event observed in metazoan transcripts. Recent studies indicate widespread dysregulation of ADAR-mediated RNA editing across many immune-related diseases, such as human cancer. We comprehensively review ADARs\' function as pattern recognizers and their capability to contribute to mediating immune-related pathways. We also highlight the potential role of site-specific RNA editing in maintaining homeostasis and its relationship to various diseases, such as human cancers. More importantly, we summarize the latest cutting-edge computational approaches and data resources for predicting and analyzing RNA editing sites. Lastly, we cover the recent advancement in site-directed ADAR editing tool development. This review presents an up-to-date overview of ADAR-mediated RNA editing, how site-specific RNA editing could potentially impact disease pathology, and how they could be harnessed for therapeutic applications.
    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:由专门的细胞酶催化的腺苷(A)向肌苷(I)的转化或编辑代表了与疾病有关的最常见的转录后RNA修饰之一。A到I的转换可以发生在特定位点,并导致蛋白质组多样性的增加和RNA稳定性的变化。拼接,和监管。这样的位点可以通过下一代RNA测序检测为腺嘌呤到鸟嘌呤的序列变化,这导致来自多个全基因组调查的数百万个报告位点。尽管如此,在这些努力中缺乏广泛的独立验证,考虑到下一代测序的错误率相对较高,这一点至关重要,导致关于编辑网站当前简编的有效性以及基于它们的结论的挥之不去的问题。
    结果:引人注目的是,我们发现,目前的分析方法存在很高的假阳性率,公共数据库中很大一部分网站无法验证.在这项工作中,我们提出了这些问题的潜在解决方案,并提供了人类癌细胞系中A-to-I编辑位点的全面且广泛验证的列表。我们的发现表明,人类癌细胞系中大多数真正的A到I编辑位点都位于非编码转录本中,所谓的RNA“暗物质”。另一方面,许多ADAR编辑事件发生在人类蛋白质编码mRNA的外显子中,包括那些可以重新编码转录组的,代表假阳性,需要谨慎解释。尽管如此,尚未发现的增加人类蛋白质组多样性的真正的ADAR位点存在,需要进一步鉴定。
    结论:准确识别人类ADAR位点仍然是一个具有挑战性的问题,特别是蛋白质编码mRNA的外显子中的位点。因此,对ADAR编辑组的全基因组调查仍必须伴随着广泛的Sanger验证工作。然而,鉴于大量未知的人类ADAR站点,需要进一步发展分析技术,可能是基于深度学习解决方案的,以便快速可靠地鉴定任何样品中的editome。
    Conversion or editing of adenosine (A) into inosine (I) catalyzed by specialized cellular enzymes represents one of the most common post-transcriptional RNA modifications with emerging connection to disease. A-to-I conversions can happen at specific sites and lead to increase in proteome diversity and changes in RNA stability, splicing, and regulation. Such sites can be detected as adenine-to-guanine sequence changes by next-generation RNA sequencing which resulted in millions reported sites from multiple genome-wide surveys. Nonetheless, the lack of extensive independent validation in such endeavors, which is critical considering the relatively high error rate of next-generation sequencing, leads to lingering questions about the validity of the current compendiums of the editing sites and conclusions based on them.
    Strikingly, we found that the current analytical methods suffer from very high false positive rates and that a significant fraction of sites in the public databases cannot be validated. In this work, we present potential solutions to these problems and provide a comprehensive and extensively validated list of A-to-I editing sites in a human cancer cell line. Our findings demonstrate that most of true A-to-I editing sites in a human cancer cell line are located in the non-coding transcripts, the so-called RNA \'dark matter\'. On the other hand, many ADAR editing events occurring in exons of human protein-coding mRNAs, including those that can recode the transcriptome, represent false positives and need to be interpreted with caution. Nonetheless, yet undiscovered authentic ADAR sites that increase the diversity of human proteome exist and warrant further identification.
    Accurate identification of human ADAR sites remains a challenging problem, particularly for the sites in exons of protein-coding mRNAs. As a result, genome-wide surveys of ADAR editome must still be accompanied by extensive Sanger validation efforts. However, given the vast number of unknown human ADAR sites, there is a need for further developments of the analytical techniques, potentially those that are based on deep learning solutions, in order to provide a quick and reliable identification of the editome in any sample.
    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
    作为动物中最普遍的RNA修饰之一,腺苷到肌苷(A到I)RNA编辑通过以时空方式使蛋白质组多样化来促进生物体的环境适应。在苍蝇和蜜蜂中,编辑酶Adar独立获得了两个不同的自动重新编码位点,形成一个自动反馈回路,稳定整体编辑效率。这通过保持靶基因的正常功能来确保细胞内稳态。然而,在更广泛的昆虫中,目前尚不清楚这种Adar自动调节机制的进化动力学和意义。我们检索了涵盖五个主要昆虫目的377种节肢动物物种的基因组(半翅目,膜翅目,鞘翅目,双翅目,和鳞翅目),并在所有基因组中对齐Adar自动重新编码位点。我们发现,两个自动重新编码站点在两个排序物种(双翅目和膜翅目)的进化过程中经历了补偿性的得失,并且两个编辑站点之间是相互排斥的:一个可编辑站点与另一个不可编辑站点显着链接。Adar的这种自动重新编码机制可以灵活地使蛋白质组多样化并稳定全局编辑活动。许多昆虫独立选择不同的自动重新编码位点来实现反馈回路并调节全局RNA编辑组,在进化过程中揭示了一个有趣的现象。我们的研究揭示了进化力作用于昆虫RNA编辑活性的精确调控,从而加深了我们对RNA编辑在环境适应和进化中的功能重要性的理解。
    As one of the most prevalent RNA modifications in animals, adenosine-to-inosine (A-to-I) RNA editing facilitates the environmental adaptation of organisms by diversifying the proteome in a temporal-spatial manner. In flies and bees, the editing enzyme Adar has independently gained two different autorecoding sites that form an autofeedback loop, stabilizing the overall editing efficiency. This ensures cellular homeostasis by keeping the normal function of target genes. However, in a broader range of insects, the evolutionary dynamics and significance of this Adar autoregulatory mechanism are unclear. We retrieved the genomes of 377 arthropod species covering the five major insect orders (Hemiptera, Hymenoptera, Coleoptera, Diptera, and Lepidoptera) and aligned the Adar autorecoding sites across all genomes. We found that the two autorecoding sites underwent compensatory gains and losses during the evolution of two orders with the most sequenced species (Diptera and Hymenoptera), and that the two editing sites were mutually exclusive among them: One editable site is significantly linked to another uneditable site. This autorecoding mechanism of Adar could flexibly diversify the proteome and stabilize global editing activity. Many insects independently selected different autorecoding sites to achieve a feedback loop and regulate the global RNA editome, revealing an interesting phenomenon during evolution. Our study reveals the evolutionary force acting on accurate regulation of RNA editing activity in insects and thus deepens our understanding of the functional importance of RNA editing in environmental adaptation and evolution.
    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
    BaseEditor(BE)是通过将CRISPR/Cas系统与单独的脱氨酶相结合而开发的基因编辑工具,在DNA或RNA中实现精确的单碱基取代,而不产生DNA双链断裂(DSB)或需要活细胞中的供体DNA模板。Base编辑器提供比其他传统人工核酸酶系统更精确和安全的基因组编辑效果。如CRISPR/Cas9,因为Cas9诱导的DSB会对基因组造成严重的损伤。因此,基础编辑在生物医学领域有着重要的应用,包括基因功能调查,定向蛋白质进化,遗传谱系追踪,疾病建模,和基因治疗。自从两个主要的基础编辑器开发以来,胞嘧啶碱基编辑器(CBE)和腺嘌呤碱基编辑器(ABE),科学家开发了100多个优化的基础编辑器,提高了编辑效率,精度,特异性,瞄准范围,以及在体内传递的能力,大大提高了其在生物医学领域的应用潜力。这里,我们回顾了基础编辑的最新发展,总结它们在生物医学领域的应用,并讨论治疗应用的未来前景和挑战。
    Base editor (BE) is a gene-editing tool developed by combining the CRISPR/Cas system with an individual deaminase, enabling precise single-base substitution in DNA or RNA without generating a DNA double-strand break (DSB) or requiring donor DNA templates in living cells. Base editors offer more precise and secure genome-editing effects than other conventional artificial nuclease systems, such as CRISPR/Cas9, as the DSB induced by Cas9 will cause severe damage to the genome. Thus, base editors have important applications in the field of biomedicine, including gene function investigation, directed protein evolution, genetic lineage tracing, disease modeling, and gene therapy. Since the development of the two main base editors, cytosine base editors (CBEs) and adenine base editors (ABEs), scientists have developed more than 100 optimized base editors with improved editing efficiency, precision, specificity, targeting scope, and capacity to be delivered in vivo, greatly enhancing their application potential in biomedicine. Here, we review the recent development of base editors, summarize their applications in the biomedical field, and discuss future perspectives and challenges for therapeutic applications.
    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
    背景:ADAR是一种参与腺苷-肌苷RNA编辑的酶。然而,ADAR在肿瘤发生中的作用,programming,免疫疗法尚未完全阐明。
    方法:TCGA,GTEx和GEO数据库被广泛用于探索癌症中ADAR的表达水平。结合患者的临床信息,我们描述了ADAR在各种癌症中的风险特征.我们确定了富含ADAR及其相关基因的通路,并探索了ADAR表达与癌症免疫微环境评分和对免疫疗法的反应之间的关联。最后,我们特别探讨了ADAR在治疗膀胱癌免疫应答中的潜在价值,并通过实验验证了ADAR在膀胱癌发生发展中的关键作用.
    结果:ADAR在大多数癌症中以RNA和蛋白质水平高表达。ADAR与某些癌症的侵袭性有关,尤其是膀胱癌.此外,ADAR与免疫相关基因有关,尤其是免疫检查点基因,在肿瘤免疫微环境中。此外,ADAR的表达与多种肿瘤的突变负荷和微卫星不稳定性呈正相关,表明ADAR可用作免疫治疗的生物标志物。最后,我们证明ADAR是膀胱癌的关键致病因子。ADAR促进膀胱癌细胞的增殖和转移。
    结论:ADAR调节肿瘤免疫微环境,可作为肿瘤免疫治疗反应的生物标志物,为肿瘤的治疗提供了新的策略,尤其是膀胱癌.
    ADAR is an enzyme involved in adenosine-inosine RNA editing. However, the role of ADAR in tumorigenesis, progression, and immunotherapy has not been fully elucidated.
    The TCGA, GTEx and GEO databases were extensively utilized to explore the expression level of ADAR across cancers. Combined with the clinical information of patients, the risk profile of ADAR in various cancers was delineated. We identified pathways enriched in ADAR and their related genes and explored the association between ADAR expression and the cancer immune microenvironment score and response to immunotherapy. Finally, we specifically explored the potential value of ADAR in the treatment of the bladder cancer immune response and verified the critical role of ADAR in the development and progression of bladder cancer through experiments.
    ADAR is highly expressed in most cancers at both the RNA and protein level. ADAR is associated with the aggressiveness of some cancers, especially bladder cancer. In addition, ADAR is associated with immune-related genes, especially immune checkpoint genes, in the tumor immune microenvironment. Moreover, ADAR expression is positively correlated with tumor mutation burden and microsatellite instability in a variety of cancers, indicating that ADAR could be used as a biomarker of immunotherapy. Finally, we demonstrated that ADAR is a key pathogenic factor in bladder cancer. ADAR promoted proliferation and metastasis of bladder cancer cells.
    ADAR regulates the tumor immune microenvironment and can be used as a biomarker of the tumor immunotherapy response, providing a novel strategy for the treatment of tumors, especially bladder 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
    核转录mRNA的广泛的腺苷到肌苷(A到I)编辑是后生动物转录调控的标志。这里,通过分析涵盖Holozoa主要群体的22个物种的RNAeditome,我们提供了大量证据支持A-ImRNA编辑作为一种调控创新,起源于现存后生动物的最后一个共同祖先。这种古老的生物化学过程保存在大多数现存的后生动物门中,主要针对由进化上年轻的重复序列形成的内源性双链RNA(dsRNA)。我们还发现有义-反义转录物的分子间配对是在一些但不是所有谱系中形成用于A到I编辑的dsRNA底物的重要机制。同样,重新编码编辑很少在谱系之间共享,但优先针对双边神经和细胞骨架系统中涉及的基因。我们得出的结论是,后生动物A到I编辑可能首先成为抵抗重复序列来源的dsRNA的保障机制,后来由于其致突变性而被纳入多种生物过程。
    Extensive adenosine-to-inosine (A-to-I) editing of nuclear-transcribed mRNAs is the hallmark of metazoan transcriptional regulation. Here, by profiling the RNA editomes of 22 species that cover major groups of Holozoa, we provide substantial evidence supporting A-to-I mRNA editing as a regulatory innovation originating in the last common ancestor of extant metazoans. This ancient biochemistry process is preserved in most extant metazoan phyla and primarily targets endogenous double-stranded RNA (dsRNA) formed by evolutionarily young repeats. We also find intermolecular pairing of sense-antisense transcripts as an important mechanism for forming dsRNA substrates for A-to-I editing in some but not all lineages. Likewise, recoding editing is rarely shared across lineages but preferentially targets genes involved in neural and cytoskeleton systems in bilaterians. We conclude that metazoan A-to-I editing might first emerge as a safeguard mechanism against repeat-derived dsRNA and was later co-opted into diverse biological processes due to its mutagenic nature.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号