Inosine

肌苷
  • 文章类型: Journal Article
    新出现的证据表明,肠道微生物组通过产生多种代谢物对心血管疾病产生深远影响。使用心肌缺血再灌注(I/R)损伤的动物模型,我们发现预防性施用一种众所周知的益生菌,婴儿双歧杆菌(B.婴儿),在I/R后保留心脏收缩功能和预防不良心脏重塑方面表现出心脏保护作用,并且这些心脏保护作用由其代谢物肌苷概括。转录组学分析进一步揭示肌苷减轻I/R诱导的心脏炎症和细胞死亡。机制研究表明,肌苷抑制了促炎细胞因子的产生,并减少了树突状细胞和自然杀伤细胞的数量,通过激活腺苷A2A受体(A2AR)来实现,该受体在抑制时消除了肌苷的心脏保护作用。此外,使用C2C12成肌细胞的体外研究表明,当经历模拟心肌I/R损伤的氧-葡萄糖剥夺/复氧时,肌苷通过嘌呤补救途径作为三磷酸腺苷(ATP)生成的替代碳源来减轻细胞死亡。同样,肌苷逆转了I/R诱导的小鼠心脏ATP水平的降低。一起来看,我们的发现表明婴儿双歧杆菌或其代谢产物肌苷通过抑制心脏炎症和减轻心脏细胞死亡而对I/R发挥心脏保护作用。提示急性缺血性心脏损伤的预防性治疗选择。
    Emerging evidence has demonstrated the profound impact of the gut microbiome on cardiovascular diseases through the production of diverse metabolites. Using an animal model of myocardial ischemia-reperfusion (I/R) injury, we found that the prophylactic administration of a well-known probiotic, Bifidobacterium infantis (B. infantis), exhibited cardioprotective effects in terms of preserving cardiac contractile function and preventing adverse cardiac remodeling following I/R and that these cardioprotective effects were recapitulated by its metabolite inosine. Transcriptomic analysis further revealed that inosine mitigated I/R-induced cardiac inflammation and cell death. Mechanistic investigations elucidated that inosine suppressed the production of pro-inflammatory cytokines and reduced the numbers of dendritic cells and natural killer cells, achieved through the activation of the adenosine A2A receptor (A2AR) that when inhibited abrogated the cardioprotective effects of inosine. Additionally, in vitro studies using C2C12 myoblasts revealed that inosine attenuated cell death by serving as an alternative carbon source for adenosine triphosphate (ATP) generation through the purine salvage pathway when subjected to oxygen-glucose deprivation/reoxygenation that simulated myocardial I/R injury. Likewise, inosine reversed the I/R-induced decrease in ATP levels in mouse hearts. Taken together, our findings indicate that B. infantis or its metabolite inosine exerts cardioprotective effects against I/R by suppressing cardiac inflammation and attenuating cardiac cell death, suggesting prophylactic therapeutic options for acute ischemic cardiac injury.
    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
    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
    癌症是一种复杂的异质性疾病,其中许多遗传和表观遗传变化发生在肿瘤的发病和进展。最近的研究表明,RNA水平的变化也与肿瘤发生有关。例如腺苷到肌苷(A-to-I)RNA编辑。这里,我们系统地研究了大量非霍奇金淋巴瘤(NHL)样本中转录组范围的A-to-I编辑事件.使用计算管道确定NHL和正常样本在已知的A-to-I编辑位点的编辑水平的显著差异,我们在NHL亚型和正常样本之间鉴定了许多差异编辑的编辑位点.大多数差异编辑的位点位于非编码区,许多这样的位点在基因表达水平和编辑效率之间显示出很强的相关性,这表明在这些情况下,RNA编辑可能对癌细胞的异常基因调控状态产生直接影响。此外,我们通过证明仅基于全基因组RNA编辑谱就可以区分NHL和正常样品,甚至NHL亚型,从而在RNA编辑和NHL之间建立了牢固的联系。我们的研究建立了RNA编辑之间的紧密联系,NHL中的癌症和异常基因调控。
    Cancer is a complex and heterogeneous disease, in which a number of genetic and epigenetic changes occur in tumor onset and progression. Recent studies indicate that changes at the RNA level are also involved in tumorigenesis, such as adenosine-to-inosine (A-to-I) RNA editing. Here, we systematically investigate transcriptome-wide A-to-I editing events in a large number of samples from Non-Hodgkin lymphomas (NHLs). Using a computational pipeline that determines significant differences in editing level between NHL and normal samples at known A-to-I editing sites, we identify a number of differentially edited editing sites between NHL subtypes and normal samples. Most of the differentially edited sites are located in non-coding regions, and many such sites show a strong correlation between gene expression level and editing efficiency, indicating that RNA editing might have direct consequences for the cancer cell\'s aberrant gene regulation status in these cases. Moreover, we establish a strong link between RNA editing and NHL by demonstrating that NHL and normal samples and even NHL subtypes can be distinguished based on genome-wide RNA editing profiles alone. Our study establishes a strong link between RNA editing, cancer and aberrant gene regulation in NHL.
    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
    众所周知,七氟烷暴露会导致发育中的大脑中广泛的神经元细胞死亡。腺苷脱氨酶作用于RNA-1(ADAR1)依赖性腺苷-肌苷(A-I)RNA编辑在整个脑发育过程中动态调节。本研究旨在探讨ADAR1在七氟醚神经毒性中的作用。在这里,我们提供的证据表明,发育七氟醚启动触发神经元焦亡,细胞凋亡和坏死(PANoptosis),并引起炎症因子的释放,包括IL-1β,IL-18、TNF-α和IFN-γ。此外,ADAR1-P150,而不是ADAR1-P110,通过在七氟烷存在下与Z-DNA/RNA结合蛋白1(ZBP1)竞争与Z-RNA结合来抑制细胞PANoptosis和炎症反应。进一步的研究表明,ADAR1依赖性A-IRNA编辑减轻七氟醚诱导的神经元PANoptosis发育。要恢复RNA编辑,我们利用腺相关病毒(AAV)将工程化的环状ADAR募集指导RNA(cadRNAs)传递到细胞中,能够募集内源性腺苷脱氨酶以促进细胞A到IRNA编辑。如预期,AAV-cadRNAs减少七氟醚诱导的细胞Z-RNA产生和PANoptosis,ADAR1-P150shRNA转染可以废除。此外,AAV-cadRNAs递送改善七氟醚诱导的发育性空间和情绪认知缺陷而不影响运动活动。一起来看,这些结果说明,ADAR1-P150在七氟醚发育性神经毒性中通过A-to-IRNA编辑预防ZBP1依赖性PANoptosis中发挥了突出作用.应用工程化的cadRNA来纠正受损的ADAR1依赖性A到IRNA编辑为可能的临床预防和治疗提供了鼓舞人心的方向。
    It is well established that sevoflurane exposure leads to widespread neuronal cell death in the developing brain. Adenosine deaminase acting on RNA-1 (ADAR1) dependent adenosine-to-inosine (A-to-I) RNA editing is dynamically regulated throughout brain development. The current investigation is designed to interrogate the contributed role of ADAR1 in developmental sevoflurane neurotoxicity. Herein, we provide evidence to show that developmental sevoflurane priming triggers neuronal pyroptosis, apoptosis and necroptosis (PANoptosis), and elicits the release of inflammatory factors including IL-1β, IL-18, TNF-α and IFN-γ. Additionally, ADAR1-P150, but not ADAR1-P110, depresses cellular PANoptosis and inflammatory response by competing with Z-DNA/RNA binding protein 1 (ZBP1) for binding to Z-RNA in the presence of sevoflurane. Further investigation demonstrates that ADAR1-dependent A-to-I RNA editing mitigates developmental sevoflurane-induced neuronal PANoptosis. To restore RNA editing, we utilize adeno-associated virus (AAV) to deliver engineered circular ADAR-recruiting guide RNAs (cadRNAs) into cells, which is capable of recruiting endogenous adenosine deaminases to promote cellular A-to-I RNA editing. As anticipated, AAV-cadRNAs diminishes sevoflurane-induced cellular Z-RNA production and PANoptosis, which could be abolished by ADAR1-P150 shRNA transfection. Moreover, AAV-cadRNAs delivery ameliorates developmental sevoflurane-induced spatial and emotional cognitive deficits without influence on locomotor activity. Taken together, these results illustrate that ADAR1-P150 exhibits a prominent role in preventing ZBP1-dependent PANoptosis through A-to-I RNA editing in developmental sevoflurane neurotoxicity. Application of engineered cadRNAs to rectify the compromised ADAR1-dependent A-to-I RNA editing provides an inspiring direction for possible clinical preventions and therapeutics.
    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结合蛋白在协调所有生物体中的种系基因表达和发育中起着至关重要的作用。这里,我们报告了ADR-2的缺失,ADR-2是作用于RNA结合蛋白的RNA(ADAR)家族的腺苷脱氨酶的成员,也是秀丽隐杆线虫中唯一的腺苷到肌苷RNA编辑酶,可以在多种遗传背景下提高生育能力。首先,我们表明,ADR-2的RNA编辑缺失可恢复正常的胚胎生产,使转基因表达卵黄蛋白原(卵黄蛋白)与绿色荧光蛋白融合。使用这种表型,设计了高通量筛选来鉴定RNA结合蛋白,这些蛋白在耗尽时产生合成表型,同时损失adr-2.筛选揭示了ADR-2和SQD-1之间的遗传相互作用,SQD-1是RNA结合蛋白的异源核核糖核蛋白(hnRNP)家族的成员。显微镜,生殖测定,高通量测序表明,sqd-1对于幼年成年动物的卵子发生和卵子基因表达的发生至关重要,adr-2的缺失可以抵消sqd-1缺失对基因表达的影响,并挽救从精子发生到卵子发生的转换。一起,这些数据表明,ADR-2有助于抑制生育力,并提示了RNA编辑依赖性和非依赖性机制在调节胚胎发生中的新作用.
    RNA binding proteins play essential roles in coordinating germline gene expression and development in all organisms. Here, we report that loss of ADR-2, a member of the Adenosine DeAminase acting on RNA (ADAR) family of RNA binding proteins and the sole adenosine-to-inosine RNA editing enzyme in C. elegans, can improve fertility in multiple genetic backgrounds. First, we show that loss of RNA editing by ADR-2 restores normal embryo production to subfertile animals that transgenically express a vitellogenin (yolk protein) fusion to green fluorescent protein. Using this phenotype, a high-throughput screen was designed to identify RNA binding proteins that when depleted yield synthetic phenotypes with loss of adr-2. The screen uncovered a genetic interaction between ADR-2 and SQD-1, a member of the heterogenous nuclear ribonucleoprotein (hnRNP) family of RNA binding proteins. Microscopy, reproductive assays, and high-throughput sequencing reveal that sqd-1 is essential for the onset of oogenesis and oogenic gene expression in young adult animals, and that loss of adr-2 can counteract the effects of loss of sqd-1 on gene expression and rescue the switch from spermatogenesis to oogenesis. Together, these data demonstrate that ADR-2 can contribute to the suppression of fertility and suggest novel roles for both RNA editing-dependent and independent mechanisms in regulating embryogenesis.
    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分子含有在许多生物过程中起关键作用的多种修饰的核碱基。腺苷到肌苷(A到I)RNA编辑,哺乳动物细胞中最普遍的RNA修饰之一,与多种人类疾病有关。为了揭示A到IRNA编辑的功能,在特定部位准确定量肌苷是至关重要的。在这项研究中,我们开发了一种核酸内切酶介导的切割和实时荧光定量PCR方法,用于A到IRNA编辑(EM-qPCR),以定量分析单个位点的A到IRNA编辑。通过使用这种方法,我们成功地精确定量了哺乳动物细胞中各种转移RNA(tRNA)分子第34位(I34)的A-IRNA编辑水平.随后,这种方法被应用于睡眠不足小鼠的组织,揭示了睡眠剥夺小鼠和对照小鼠之间I34水平的显着变化。所提出的方法为在特定位点进行A到IRNA编辑的定量分析树立了先例,有助于更深入地了解A到IRNA编辑的生物学意义。
    RNA molecules contain diverse modified nucleobases that play pivotal roles in numerous biological processes. Adenosine-to-inosine (A-to-I) RNA editing, one of the most prevalent RNA modifications in mammalian cells, is linked to a multitude of human diseases. To unveil the functions of A-to-I RNA editing, accurate quantification of inosine at specific sites is essential. In this study, we developed an endonuclease-mediated cleavage and real-time fluorescence quantitative PCR method for A-to-I RNA editing (EM-qPCR) to quantitatively analyze A-to-I RNA editing at a single site. By employing this method, we successfully quantified the levels of A-to-I RNA editing on various transfer RNA (tRNA) molecules at position 34 (I34) in mammalian cells with precision. Subsequently, this method was applied to tissues from sleep-deprived mice, revealing a notable alteration in the levels of I34 between sleep-deprived and control mice. The proposed method sets a precedent for the quantitative analysis of A-to-I RNA editing at specific sites, facilitating a deeper understanding of the biological implications of A-to-I RNA editing.
    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
    腺苷到肌苷(A到I)RNA编辑重新编码遗传信息。除了使蛋白质组多样化,RNA重新编码的另一个诱人的优势是纠正有害的DNA突变并恢复祖先等位基因。有益的恢复性编辑的可靠证据在动物中非常罕见。通过在系统发育环境下搜索“收敛重新编码”,我们提出这个术语来判断特定编辑网站的潜在恢复功能。对于众所周知的哺乳动物Gln>Arg(Q>R)重编码位点,它在脊椎动物基因组中的祖先状态是编辑前的Gln,和所有470可用的哺乳动物基因组严格避免其他三种等效方式来实现蛋白质中的Arg。从他的>Arg中没有收敛的重新编码,或者Gln密码子的同义突变,可以归因于对编辑主题和结构的强大维护,但是没有直接的A到G突变是非常出乎意料的。有了类似的想法,我们在果蝇属中发现了会聚重新编码的病例,降低其恢复功能的可能性。总之,我们定义了一个有趣的收敛重新编码场景,其发生可以用作对重新编码网站是否具有唯一恢复作用的初步判断。我们的工作为RNA编辑的自然选择和进化提供了新的见解。
    Adenosine-to-inosine (A-to-I) RNA editing recodes the genetic information. Apart from diversifying the proteome, another tempting advantage of RNA recoding is to correct deleterious DNA mutation and restore ancestral allele. Solid evidences for beneficial restorative editing are very rare in animals. By searching for \"convergent recoding\" under a phylogenetic context, we proposed this term for judging the potential restorative functions of particular editing site. For the well-known mammalian Gln>Arg (Q>R) recoding site, its ancestral state in vertebrate genomes was the pre-editing Gln, and all 470 available mammalian genomes strictly avoid other three equivalent ways to achieve Arg in protein. The absence of convergent recoding from His>Arg, or synonymous mutations on Gln codons, could be attributed to the strong maintenance on editing motif and structure, but the absence of direct A-to-G mutation is extremely unexpected. With similar ideas, we found cases of convergent recoding in Drosophila genus, reducing the possibility of their restorative function. In summary, we defined an interesting scenario of convergent recoding, the occurrence of which could be used as preliminary judgements for whether a recoding site has a sole restorative role. Our work provides novel insights to the natural selection and evolution of RNA editing.
    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
    外写是一个研究转录后变化的领域。在这些修改中,腺苷转化为肌苷,作为鸟苷(A>I(G)),是已知的RNA编辑机制之一,由ADAR催化。这种类型的RNA编辑是哺乳动物中最常见的编辑类型,有助于生物多样性。A>I(G)RNA编辑平衡的破坏与疾病有关,包括几种癌症。癌症患者的耐药性是一个重要的公共卫生问题,导致治疗无反应性和疾病进展导致的死亡率增加,代表了这一领域研究人员的最大挑战。A>I(G)RNA编辑涉及免疫疗法和基因毒性药物反应和耐药性的几种机制。这篇综述研究了ADAR1与特定A>I(G)RNA编辑位点之间的关系,特别关注乳腺癌,以及这些位点对DNA损伤修复和抗癌治疗的免疫反应的影响。我们解决了潜在的机制,生物信息学,以及鉴定和验证A>I(G)RNA编辑位点的体外策略。我们收集了与A>I(G)RNA编辑和癌症相关的数据库,并讨论了理解A>I(G)RNA编辑模式的潜在临床和研究意义。了解ADAR1介导的A>I(G)RNA编辑在乳腺癌中的复杂作用,对于开发针对个体患者的个性化治疗方法具有重要意义。
    Epitranscriptomics is a field that delves into post-transcriptional changes. Among these modifications, the conversion of adenosine to inosine, traduced as guanosine (A>I(G)), is one of the known RNA-editing mechanisms, catalyzed by ADARs. This type of RNA editing is the most common type of editing in mammals and contributes to biological diversity. Disruption in the A>I(G) RNA-editing balance has been linked to diseases, including several types of cancer. Drug resistance in patients with cancer represents a significant public health concern, contributing to increased mortality rates resulting from therapy non-responsiveness and disease progression, representing the greatest challenge for researchers in this field. The A>I(G) RNA editing is involved in several mechanisms over the immunotherapy and genotoxic drug response and drug resistance. This review investigates the relationship between ADAR1 and specific A>I(G) RNA-edited sites, focusing particularly on breast cancer, and the impact of these sites on DNA damage repair and the immune response over anti-cancer therapy. We address the underlying mechanisms, bioinformatics, and in vitro strategies for the identification and validation of A>I(G) RNA-edited sites. We gathered databases related to A>I(G) RNA editing and cancer and discussed the potential clinical and research implications of understanding A>I(G) RNA-editing patterns. Understanding the intricate role of ADAR1-mediated A>I(G) RNA editing in breast cancer holds significant promise for the development of personalized treatment approaches tailored to individual patients\' A>I(G) RNA-editing profiles.
    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
    转化生长因子-β(TGF-β)信号通路在结直肠癌(CRC)的发病机制中起着关键作用。然而,TGF-β信号通路调控CRC的具体分子机制尚不完全清楚.在本研究中,代谢组学和转录组学用于筛选与CRC中TGF-β信号通路调控最相关的关键代谢物和调控基因.此外,逆转录-定量PCR,进行蛋白质印迹和Transwell测定以评估上皮-间质转化(EMT)的过程。代谢组学分析表明TGF-β1对嘌呤代谢有影响,导致嘌呤代谢物肌苷增加。肌苷的增加对于促进CRC细胞中的EMT和细胞迁移至关重要。此外,代谢组学和转录组学数据的综合分析显示,TGF-β1诱导含有漆酶结构域1(LACC1)的表达,一种参与肌苷调节的酶。敲除LACC1导致TGF-β1诱导的肌苷水平改变减少,CRC细胞中的EMT和细胞迁移。本研究的结果表明,TGF-β信号通路通过调节LACC1的表达参与了CRC嘌呤代谢的调节。此外,LACC1似乎通过提高嘌呤代谢物肌苷的水平来影响EMT和细胞迁移。
    Transforming growth factor-β (TGF-β) signaling pathway serves a pivotal role in the pathogenesis of colorectal cancer (CRC). However, the specific molecular mechanisms by which the TGF-β signaling pathway regulates CRC are still not fully understood. In the present study, metabolomics and transcriptomics were used to screen for key metabolites and regulatory genes most related to the regulation of the TGF-β signaling pathway in CRC. Additionally, reverse transcription-quantitative PCR, western blotting and Transwell assays were performed to assess the process of epithelial-mesenchymal transition (EMT). Metabolomics analysis indicated that TGF-β1 has an impact on purine metabolism, leading to an increase in the purine metabolite inosine. The increase of inosine is essential for facilitating EMT and cell migration in CRC cells. Furthermore, the integrated analysis of metabolomics and transcriptomics data revealed that TGF-β1 induces the expression of laccase domain-containing 1 (LACC1), an enzyme involved in the regulation of inosine. Knockdown of LACC1 resulted in a reduction of TGF-β1-induced alterations in inosine levels, EMT and cell migration in CRC cells. The results of the present study suggest that the TGF-β signaling pathway is involved in the regulation of purine metabolism in CRC through the modulation of LACC1 expression. Furthermore, LACC1 appears to influence EMT and cell migration by elevating the levels of the purine metabolite inosine.
    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
    作用于RNA的腺苷脱氨酶(ADAR)是已知将双链RNA中的腺苷转化为肌苷并参与宿主-病毒相互作用的进化上保守的酶。对可用的转录组数据进行荟萃分析,我们鉴定并鉴定了Chlamysfarreri中的八个ADAR转录本,养殖的海洋扇贝易受急性病毒性坏死病毒(AVNV)感染和死亡暴发的影响。因此,我们在知孔扇贝基因组中鉴定出6个ADAR基因,修改了以前的基因注释,并追踪可变剪接变体。详细来说,每个ADAR基因编码一个独特的功能域组合,总是包括腺苷脱氨酶结构域,RNA结合域和,在一个案例中,Z-DNA结合结构域的两个拷贝。经过系统发育分析,在ADAR1进化枝中聚集了5个FarreriADAR,以及来自不同动物门的序列。基因表达分析显示CF051320是表达最多的ADAR,尤其是眼睛和男性性腺.另外4个ADAR1基因和1个ADAR2基因表现出不同的表达水平,CF105370和CF051320在扇贝发育早期显著增加。ADAR介导的单碱基编辑,在成虫C.Farreri组织和发育阶段进行评估,主要在基因间区域检测到(83%和85%,分别)。总的来说,六个ADAR基因的表达模式以及在扇贝RNA-seq样本上计算的编辑和超编辑值支持ADAR1介导的编辑的适应性值,特别是在沉降前的幼虫阶段。
    Adenosine Deaminases Acting on RNA (ADARs) are evolutionarily conserved enzymes known to convert adenosine to inosine in double-stranded RNAs and participate in host-virus interactions. Conducting a meta-analysis of available transcriptome data, we identified and characterised eight ADAR transcripts in Chlamys farreri, a farmed marine scallop susceptible to Acute viral necrosis virus (AVNV) infections and mortality outbreaks. Accordingly, we identified six ADAR genes in the Zhikong scallop genome, revised previous gene annotations, and traced alternative splicing variants. In detail, each ADAR gene encodes a unique combination of functional domains, always including the Adenosine deaminase domain, RNA binding domains and, in one case, two copies of a Z-DNA binding domain. After phylogenetic analysis, five C. farreri ADARs clustered in the ADAR1 clade along with sequences from diverse animal phyla. Gene expression analysis indicated CF051320 as the most expressed ADAR, especially in the eye and male gonad. The other four ADAR1 genes and one ADAR2 gene exhibited variable expression levels, with CF105370 and CF051320 significantly increasing during early scallop development. ADAR-mediated single-base editing, evaluated across adult C. farreri tissues and developmental stages, was mainly detectable in intergenic regions (83 % and 85 %, respectively). Overall, the expression patterns of the six ADAR genes together with the editing and hyper-editing values computed on scallops RNA-seq samples support the adaptive value of ADAR1-mediated editing, particularly in the pre-settling larval stages.
    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