snoRNA

snoRNA
  • 文章类型: Journal Article
    这篇综述全面研究了小的非编码RNA(sncRNA)和胰腺导管腺癌(PDAC)之间的复杂相互作用。治疗选择有限的破坏性恶性肿瘤。我们的分析揭示了sncRNAs在PDAC生物学各个方面的关键作用,跨越诊断,发病机制,耐药性,和治疗策略。sncRNAs已经成为PDAC的有希望的生物标志物,在患病组织中表现出不同的表达谱。sncRNA差异表达模式,通常可在体液中检测到,保持早期和微创诊断方法的潜力。此外,sncRNAs在PDAC发病机制中表现出复杂的参与,调节关键的细胞过程,如增殖,凋亡,和转移。此外,对sncRNA介导的致病途径的机制见解阐明了新的治疗靶标和干预措施。这篇综述的一个重要焦点是致力于揭示PDAC中潜在的耐药性sncRNA机制。在分子水平上了解这些机制对于设计克服耐药性的策略至关重要。探索治疗前景,我们讨论了sncRNAs本身作为治疗剂的潜力,因为它们以高特异性调节基因表达的能力使它们成为靶向治疗的有吸引力的候选者.总之,这篇综述整合了PDAC中sncRNAs的最新知识,对他们的诊断提供一个整体的视角,致病性,和治疗相关性。通过阐明sncRNAs在PDAC生物学中的作用,这篇综述为开发新的诊断工具和靶向治疗方法提供了有价值的见解,对于改善PDAC患者的预后至关重要。
    This review comprehensively investigates the intricate interplay between small non-coding RNAs (sncRNAs) and pancreatic ductal adenocarcinoma (PDAC), a devastating malignancy with limited therapeutic options. Our analysis reveals the pivotal roles of sncRNAs in various facets of PDAC biology, spanning diagnosis, pathogenesis, drug resistance, and therapeutic strategies. sncRNAs have emerged as promising biomarkers for PDAC, demonstrating distinct expression profiles in diseased tissues. sncRNA differential expression patterns, often detectable in bodily fluids, hold potential for early and minimally invasive diagnostic approaches. Furthermore, sncRNAs exhibit intricate involvement in PDAC pathogenesis, regulating critical cellular processes such as proliferation, apoptosis, and metastasis. Additionally, mechanistic insights into sncRNA-mediated pathogenic pathways illuminate novel therapeutic targets and interventions. A significant focus of this review is dedicated to unraveling sncRNA mechanisms underlying drug resistance in PDAC. Understanding these mechanisms at the molecular level is imperative for devising strategies to overcome drug resistance. Exploring the therapeutic landscape, we discuss the potential of sncRNAs as therapeutic agents themselves as their ability to modulate gene expression with high specificity renders them attractive candidates for targeted therapy. In summary, this review integrates current knowledge on sncRNAs in PDAC, offering a holistic perspective on their diagnostic, pathogenic, and therapeutic relevance. By elucidating the roles of sncRNAs in PDAC biology, this review provides valuable insights for the development of novel diagnostic tools and targeted therapeutic approaches, crucial for improving the prognosis of PDAC patients.
    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
    细胞衰老是由各种应激引起的细胞周期停滞的不可逆状态。包括异常的癌基因激活,端粒缩短,和DNA损伤。通过一个全基因组的屏幕,我们发现了一种保守的小核仁RNA(snoRNA),SNORA13,是人类细胞和小鼠多种形式衰老所必需的。尽管SNORA13指导了核糖体解码中心保守核苷酸的假尿苷化,这种snoRNA的丢失对翻译的影响最小。相反,我们发现SNORA13负调节核糖体生物发生。诱导衰老的应激干扰核糖体生物发生,导致触发p53激活的游离核糖体蛋白(RP)的积累。SNORA13与RPL23直接相互作用,减少其掺入成熟的60S亚基,因此,增加免费RPs池,从而促进p53介导的衰老。因此,SNORA13通过与其在引导RNA修饰中的作用不同的非规范机制调节核糖体生物发生和p53途径。这些发现扩展了我们对snoRNA功能及其在细胞信号传导中的作用的理解。
    Cellular senescence is an irreversible state of cell-cycle arrest induced by various stresses, including aberrant oncogene activation, telomere shortening, and DNA damage. Through a genome-wide screen, we discovered a conserved small nucleolar RNA (snoRNA), SNORA13, that is required for multiple forms of senescence in human cells and mice. Although SNORA13 guides the pseudouridylation of a conserved nucleotide in the ribosomal decoding center, loss of this snoRNA minimally impacts translation. Instead, we found that SNORA13 negatively regulates ribosome biogenesis. Senescence-inducing stress perturbs ribosome biogenesis, resulting in the accumulation of free ribosomal proteins (RPs) that trigger p53 activation. SNORA13 interacts directly with RPL23, decreasing its incorporation into maturing 60S subunits and, consequently, increasing the pool of free RPs, thereby promoting p53-mediated senescence. Thus, SNORA13 regulates ribosome biogenesis and the p53 pathway through a non-canonical mechanism distinct from its role in guiding RNA modification. These findings expand our understanding of snoRNA functions and their roles in cellular signaling.
    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
    急性肾损伤(AKI)表示以肾小管细胞死亡和间质炎症为特征的肾功能的突然和长期下降。小核仁RNA(snoRNAs)在氧化应激和炎症中起关键作用,并可能在AKI过程中发挥重要作用,这仍然难以捉摸。在对AKI的反应中,肾小管中Snord3a的表达升高,这表明Snord3a缺乏减轻了AKI小鼠模型中的肾损伤。值得注意的是,Snord3a的缺乏对干扰素基因(STING)相关的铁死亡表型的刺激因子和肾小管损伤的进展具有缓解作用。机械上,Snord3a显示通过促进STING基因转录来调节STING信号轴;Snord3a反义寡核苷酸的施用在AKI小鼠模型中建立了显著的治疗优势。一起,这些发现阐明了STING的转录调控机制以及Snord3a-STING轴在AKI过程中铁死亡的关键作用,强调Snord3a是AKI的潜在预后和治疗靶点。
    Acute kidney injury (AKI) signifies a sudden and prolonged decline in kidney function characterized by tubular cell death and interstitial inflammation. Small nucleolar RNAs (snoRNAs) play pivotal roles in oxidative stress and inflammation, and may play an important role in the AKI process, which remains elusive. an elevated expression of Snord3a is revealed in renal tubules in response to AKI and demonstrates that Snord3a deficiency alleviates renal injury in AKI mouse models. Notably, the deficiency of Snord3a exhibits a mitigating effect on the stimulator of interferon genes (STING)-associated ferroptosis phenotypes and the progression of tubular injury. Mechanistically, Snord3a is shown to regulate the STING signaling axis via promoting STING gene transcription; administration of Snord3a antisense oligonucleotides establishes a significant therapeutic advantage in AKI mouse models. Together, the findings elucidate the transcription regulation mechanism of STING and the crucial roles of the Snord3a-STING axis in ferroptosis during AKI, underscoring Snord3a as a potential prognostic and therapeutic target for AKI.
    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
    近年来,关于人类RNA中A-I编辑的程度以及ADAR1在细胞编辑机制中的关键作用,已经积累了许多证据。已经表明,A-to-I编辑的发生和频率是组织特异性的,并且对于某些组织发育是必不可少的,比如肝脏。研究ADAR1对肝细胞功能的影响,我们已经创建了Huh7.5ADAR1KO细胞系。IFN治疗后,Huh7.5ADAR1KO细胞显示生长和翻译的快速停滞,他们无法从中恢复过来。我们通过采用基于对单独的多体谱RNA部分进行测序的方法分析了翻译体的变化。我们发现Huh7.5ADAR1KO细胞的转录组和翻译组发生显著变化。最突出的变化包括RNA聚合酶III对转录的负面影响以及snoRNA和YRNA水平的失调。此外,我们观察到ADAR1KO多聚体富含编码蛋白质的mRNA,这些蛋白质在广泛的生物过程中至关重要,例如RNA定位和RNA加工,而未结合的部分主要富集在编码核糖体蛋白和翻译因子的mRNA中。这表明ADAR1在小RNA代谢和核糖体生物发生中起着更重要的作用。
    In recent years, numerous evidence has been accumulated about the extent of A-to-I editing in human RNAs and the key role ADAR1 plays in the cellular editing machinery. It has been shown that A-to-I editing occurrence and frequency are tissue specific and essential for some tissue development, such as liver. To study the effect of ADAR1 function in hepatocytes, we have created Huh7.5 ADAR1 KO cell lines. Upon IFN treatment, the Huh7.5 ADAR1 KO cells show rapid arrest of growth and translation, from which they do not recover. We analyzed translatome changes by employing a method based on sequencing of separate polysome profile RNA fractions. We found significant changes in transcriptome and translatome of the Huh7.5 ADAR1 KO cells. The most prominent changes include negatively affected transcription by RNA polymerase III and the deregulation of snoRNA and Y RNA levels. Furthermore, we observed that ADAR1 KO polysomes are enriched in mRNAs coding for proteins pivotal in a wide range of biological processes such as RNA localization and RNA processing, whereas the unbound fraction is enriched mainly in mRNAs coding for ribosomal proteins and translational factors. This indicates that ADAR1 plays more relevant role in small RNA metabolism and ribosome biogenesis.
    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
    驱动神经胶质瘤进展的因素仍然知之甚少。这里,表观遗传调节因子TRIM24被确定为神经胶质瘤进展的驱动因素,其中TRIM24过表达促进HRasV12间变性星形细胞瘤(AA)进展为上皮样GBM(Ep-GBM)样肿瘤。TRIM24与HRasV12的共转染还诱导具有肿瘤蛋白p53基因(TP53)敲低的人神经干细胞(hNSC)的Ep-GBM样转化。此外,TRIM24在临床Ep-GBM标本中高表达。使用单细胞RNA测序(scRNA-Seq),作者表明TRIM24过表达影响肿瘤内异质性和肿瘤微环境.机械上,HRasV12激活RNA输出(PHAX)的磷酸化衔接子,并上调U3小核仁RNA(U3snoRNA)以招募Ku依赖性DNA依赖性蛋白激酶催化亚基(DNA-PKcs)。过表达的TRIM24也被PHAX招募到U3snoRNA,从而促进TRIM24在S767/768残基处的DNA-PKcs磷酸化。磷酸化TRIM24诱导表观基因组和转录因子网络重编程并促进Ep-GBM样转化。用小分子抑制剂NU7441靶向DNA-PKcs与替莫唑胺协同作用以降低Ep-GBM致瘤性并延长动物存活。这些发现为Ep-GBM样转化的表观遗传调控提供了新的见解,并为Ep-GBM患者提出了潜在的治疗策略。
    The factors driving glioma progression remain poorly understood. Here, the epigenetic regulator TRIM24 is identified as a driver of glioma progression, where TRIM24 overexpression promotes HRasV12 anaplastic astrocytoma (AA) progression into epithelioid GBM (Ep-GBM)-like tumors. Co-transfection of TRIM24 with HRasV12 also induces Ep-GBM-like transformation of human neural stem cells (hNSCs) with tumor protein p53 gene (TP53) knockdown. Furthermore, TRIM24 is highly expressed in clinical Ep-GBM specimens. Using single-cell RNA-sequencing (scRNA-Seq), the authors show that TRIM24 overexpression impacts both intratumoral heterogeneity and the tumor microenvironment. Mechanically, HRasV12 activates phosphorylated adaptor for RNA export (PHAX) and upregulates U3 small nucleolar RNAs (U3 snoRNAs) to recruit Ku-dependent DNA-dependent protein kinase catalytic subunit (DNA-PKcs). Overexpressed TRIM24 is also recruited by PHAX to U3 snoRNAs, thereby facilitating DNA-PKcs phosphorylation of TRIM24 at S767/768 residues. Phosphorylated TRIM24 induces epigenome and transcription factor network reprogramming and promotes Ep-GBM-like transformation. Targeting DNA-PKcs with the small molecule inhibitor NU7441 synergizes with temozolomide to reduce Ep-GBM tumorigenicity and prolong animal survival. These findings provide new insights into the epigenetic regulation of Ep-GBM-like transformation and suggest a potential therapeutic strategy for patients with Ep-GBM.
    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:微血管和大血管并发症在2型糖尿病患者中很常见。最近,人们对研究循环的非编码小RNA(sncRNA)作为糖尿病并发症发展的贡献者的潜力越来越感兴趣。在这项研究中,我们调查了2型糖尿病患者中循环sncRNAs水平与糖尿病肾病(DKD)的相关性。
    方法:使用小RNA-seq测定血浆sncRNAs水平,允许检测miRNAs,snoRNAs,piRNAs,tRNA片段,和各种其他sncRNA类。我们测试了2型糖尿病患者中差异表达的sncRNAs,有DKD(n=69)或无DKD(n=405)。在二级分析中,我们还测试了与eGFR的关联,白蛋白尿(UACR),和血浆蛋白质组。
    结果:总共7个sncRNAs与普遍DKD呈负相关(所有PFDR≤0.05)。包括一个microRNA(miR-143-5p),五个snoRNA(U8,SNORD118,SNORD24,SNORD107,SNORD87)和一个piRNA(piR-019825|DQ597218)。蛋白质组学分析表明,七个sncRNAs,尤其是piRNApiR-019825,与24种蛋白质的血浆水平相关,其中一些已知与肾功能相关,包括TNFsR-I(TNFRFS1A),DAN(NBL1)和胱抑素C(CST3)。
    结论:我们已经鉴定了新的非编码小RNA,主要来自microRNA以外的类,与糖尿病肾病有关。我们的结果表明,小的非编码RNA在DKD中的参与超出了已知的microRNA,并且还涉及其他类型的sncRNA,特别是snoRNA和piR-019825,以前从未与肾功能相关的研究。
    BACKGROUND: Micro- and macrovascular complications are common among persons with type 2 diabetes. Recently there has been growing interest to investigate the potential of circulating small non-coding RNAs (sncRNAs) as contributors to the development of diabetic complications. In this study we investigate to what extent circulating sncRNAs levels associate with prevalent diabetic kidney disease (DKD) in persons with type 2 diabetes.
    METHODS: Plasma sncRNAs levels were determined using small RNA-seq, allowing detection of miRNAs, snoRNAs, piRNAs, tRNA fragments, and various other sncRNA classes. We tested for differentially expressed sncRNAs in persons with type 2 diabetes, with DKD (n = 69) or without DKD (n = 405). In secondary analyses, we also tested the association with eGFR, albuminuria (UACR), and the plasma proteome.
    RESULTS: In total seven sncRNAs were negatively associated with prevalent DKD (all PFDR ≤ 0.05). Including one microRNA (miR-143-5p), five snoRNAs (U8, SNORD118, SNORD24, SNORD107, SNORD87) and a piRNA (piR-019825 | DQ597218). Proteomic analyses showed that the seven sncRNAs, and especially the piRNA piR-019825, were associated with plasma levels of 24 proteins of which several have known associations with kidney function including TNF sR-I (TNFRFS1A), DAN (NBL1) and cystatin C (CST3).
    CONCLUSIONS: We have identified novel small non-coding RNAs, primarily from classes other than microRNAs, that are associated with diabetic kidney disease. Our results show that the involvement of small non-coding RNAs in DKD goes beyond the already known microRNAs and also involves other classes of sncRNA, in particular snoRNAs and the piRNA piR-019825, that have never been studied before in relation to kidney function.
    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
    核糖体RNA(rRNA)在转录和随后的成熟过程中被广泛修饰。三种类型的修改,核糖部分的2'-O-甲基化,假吡啶化,和基础修改,通过snoRNA驱动的机制或独立的酶引入。修饰的核苷酸聚集在功能上重要的位点,包括肽基转移酶中心(PTC)。因此,据推测,修饰的核苷酸在确保核糖体的功能性中起着重要作用。在这项研究中,我们证明了七个25SrRNA修饰,包括四个进化保守的修改,在PTC附近可以同时耗尽而不损失细胞活力。构建了缺乏三个snoRNA基因(snR34,snR52和snR65)和/或表达spb1(D52A/E679K)和nop2(C424A/C478A)的无酶活性变体的酵母突变体。结果表明,PTC中的rRNA修饰共同有助于真核细胞中的有效翻译。25SrRNA中七个修饰核苷酸的缺乏导致细胞生长减少,冷灵敏度,翻译水平下降,和超精确的翻译,正如减少的误解和无稽之谈所表明的那样。修饰m5C2870在不存在其他六个修饰的核苷酸时至关重要。因此,PTC周围rRNA修饰核苷酸的模式对于最佳核糖体翻译活性和翻译保真度至关重要。
    Ribosomal RNAs (rRNAs) are extensively modified during the transcription and subsequent maturation. Three types of modifications, 2\'-O-methylation of ribose moiety, pseudouridylation, and base modifications, are introduced either by a snoRNA-driven mechanism or by stand-alone enzymes. Modified nucleotides are clustered at the functionally important sites, including peptidyl transferase center (PTC). Therefore, it has been hypothesised that the modified nucleotides play an important role in ensuring the functionality of the ribosome. In this study, we demonstrate that seven 25S rRNA modifications, including four evolutionarily conserved modifications, in the proximity of PTC can be simultaneously depleted without loss of cell viability. Yeast mutants lacking three snoRNA genes (snR34, snR52, and snR65) and/or expressing enzymatically inactive variants of spb1(D52A/E679K) and nop2(C424A/C478A) were constructed. The results show that rRNA modifications in PTC contribute collectively to efficient translation in eukaryotic cells. The deficiency of seven modified nucleotides in 25S rRNA resulted in reduced cell growth, cold sensitivity, decreased translation levels, and hyperaccurate translation, as indicated by the reduced missense and nonsense suppression. The modification m5C2870 is crucial in the absence of the other six modified nucleotides. Thus, the pattern of rRNA-modified nucleotides around the PTC is essential for optimal ribosomal translational activity and translational fidelity.
    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
    在酿酒酵母中,聚腺苷酸化形式的成熟(而不是前体)小的非编码RNA(sncRNAs)那些未能经历适当的3'-末端成熟的受到Rrp6p和Rrp47p的活性降解,这不需要核心外泌体和TRAMP组件的参与。与这一发现一致,Rrp6p/Rrp47p被证明作为外泌体独立的复合物存在,优先与这些sncRNA的成熟聚腺苷酸化形式结合。与这一观察一致,与核心核外泌体缺乏物理关联的Rrp6p(Rrp6p-ΔC2)的C末端截断版本支持其衰变,就像其全长版本一样。聚腺苷酸化由规范和非规范聚(A)聚合酶催化,Pap1p和Trf4p.对WT和rrp6-Δ菌株中的聚腺苷酸化概况的分析表明,大多数聚腺苷酸化位点对应于其成熟末端上游或下游的一到三个核苷酸,并且它们的聚(A)尾的范围为10-15个腺苷酸残基。最有趣的是,积累的聚腺苷酸化snRNA在rrp6-Δ菌株中具有功能,并组装成剪接体。因此,Rrp6p-Rrp47p定义了面包师酵母中不依赖核心核外泌体的新型RNA周转系统,该系统靶向不完全加工的聚腺苷酸化sncRNAs,该sncRNAs在不存在Rrp6p的情况下积累。
    In Saccharomyces cerevisiae, polyadenylated forms of mature (and not precursor) small non-coding RNAs (sncRNAs) those fail to undergo proper 3\'-end maturation are subject to an active degradation by Rrp6p and Rrp47p, which does not require the involvement of core exosome and TRAMP components. In agreement with this finding, Rrp6p/Rrp47p is demonstrated to exist as an exosome-independent complex, which preferentially associates with mature polyadenylated forms of these sncRNAs. Consistent with this observation, a C-terminally truncated version of Rrp6p (Rrp6p-ΔC2) lacking physical association with the core nuclear exosome supports their decay just like its full-length version. Polyadenylation is catalyzed by both the canonical and non-canonical poly(A) polymerases, Pap1p and Trf4p. Analysis of the polyadenylation profiles in WT and rrp6-Δ strains revealed that the majority of the polyadenylation sites correspond to either one to three nucleotides upstream or downstream of their mature ends and their poly(A) tails ranges from 10-15 adenylate residues. Most interestingly, the accumulated polyadenylated snRNAs are functional in the rrp6-Δ strain and are assembled into spliceosomes. Thus, Rrp6p-Rrp47p defines a core nuclear exosome-independent novel RNA turnover system in baker\'s yeast targeting imperfectly processed polyadenylated sncRNAs that accumulate in the absence of Rrp6p.
    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:透明细胞肾细胞癌(ccRCC)是最常见的肾细胞癌亚型,转移率高。酪氨酸激酶和检查点抑制剂等靶向治疗提高了治疗成功率。但治疗相关的副作用和肿瘤复发仍然是一个挑战。因此,ccRCC的死亡率仍然很高。转移前早期检测具有改善预后的巨大潜力,但目前尚无ccRCC特异性合适的生物标志物。因此,在过去的十年中,已经研究了来自体液的分子生物标志物。其中,来自尿液来源的细胞外囊泡(EV)的RNA非常有前途。
    方法:从78名受试者(54名ccRCC患者,24个尿石症对照)。RNA-seq在发现队列中进行,整个队列的一个子集(47ccRCC,16尿石症)。然后将读数映射到基因组,并且基于100nt长的连续基因组区域定量表达。进行聚类分析和差异区域表达分析,并根据年龄和性别进行调整。通过qPCR在整个群组中验证候选生物标志物。接收机工作特性,曲线下面积和比值比用于评估模型的诊断潜力.
    结果:RNA-seq表达数据的初始聚类分析显示受试者的性别分离,但不是肿瘤状态。因此,进行了以下分析,调整性别和年龄。ccRCC和尿石症患者之间差异表达的区域主要与小核仁RNA(snoRNA)重叠。通过定量PCR验证了四种snoRNAs(SNORD99、SNORD22、SNORD26、SNORA50C)的差异表达。然后使用校正回归模型将验证队列分类为ccRCC和无肿瘤受试者。相应的准确度范围从0.654到0.744。结合多个基因和肥胖和高血压危险因素的模型显示,SNORD99和SNORA50C的诊断性能提高,准确率高达0.811(p=0.0091)。
    结论:我们的研究发现了来自尿液来源的电动汽车的四种以前未被识别的snoRNA生物标志物,推进寻找一个强大的,易于使用的ccRCC筛选方法。
    BACKGROUND: Clear cell renal cell carcinoma (ccRCC) is the most common subtype of RCC with high rates of metastasis. Targeted therapies such as tyrosine kinase and checkpoint inhibitors have improved treatment success, but therapy-related side effects and tumor recurrence remain a challenge. As a result, ccRCC still have a high mortality rate. Early detection before metastasis has great potential to improve outcomes, but no suitable biomarker specific for ccRCC is available so far. Therefore, molecular biomarkers derived from body fluids have been investigated over the past decade. Among them, RNAs from urine-derived extracellular vesicles (EVs) are very promising.
    METHODS: RNA was extracted from urine-derived EVs from a cohort of 78 subjects (54 ccRCC patients, 24 urolithiasis controls). RNA-seq was performed on the discovery cohort, a subset of the whole cohort (47 ccRCC, 16 urolithiasis). Reads were then mapped to the genome, and expression was quantified based on 100 nt long contiguous genomic regions. Cluster analysis and differential region expression analysis were performed with adjustment for age and gender. The candidate biomarkers were validated by qPCR in the entire cohort. Receiver operating characteristic, area under the curve and odds ratios were used to evaluate the diagnostic potential of the models.
    RESULTS: An initial cluster analysis of RNA-seq expression data showed separation by the subjects\' gender, but not by tumor status. Therefore, the following analyses were done, adjusting for gender and age. The regions differentially expressed between ccRCC and urolithiasis patients mainly overlapped with small nucleolar RNAs (snoRNAs). The differential expression of four snoRNAs (SNORD99, SNORD22, SNORD26, SNORA50C) was validated by quantitative PCR. Confounder-adjusted regression models were then used to classify the validation cohort into ccRCC and tumor-free subjects. Corresponding accuracies ranged from 0.654 to 0.744. Models combining multiple genes and the risk factors obesity and hypertension showed improved diagnostic performance with an accuracy of up to 0.811 for SNORD99 and SNORA50C (p = 0.0091).
    CONCLUSIONS: Our study uncovered four previously unrecognized snoRNA biomarkers from urine-derived EVs, advancing the search for a robust, easy-to-use ccRCC screening method.
    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
    利什曼原虫是影响全世界数百万人的皮肤和内脏疾病的病原体。伪尿苷(Φ),rRNA上最丰富的修饰,寄生虫生命周期中的变化。螺旋69(H69)中特定的Φ水平的变化会影响核糖体功能。为了破译这种表型的分子机制,我们使用cryo-EM在〜2.4-3µ分辨率下确定了缺乏单个Φ及其亲本菌株的核糖体的结构。我们的发现证明了H69上的单个Φ对其结构的重要性以及其与螺旋44和特定tRNA相互作用的重要性。我们的研究表明,由于核糖体对tRNA的选择性调节,rRNA修饰会影响携带密码子偏倚的mRNA的翻译。基于高分辨率结构,我们提出了一种解释核糖体如何选择特定tRNA的机制。
    Leishmania is the causative agent of cutaneous and visceral diseases affecting millions of individuals worldwide. Pseudouridine (Ψ), the most abundant modification on rRNA, changes during the parasite life cycle. Alterations in the level of a specific Ψ in helix 69 (H69) affected ribosome function. To decipher the molecular mechanism of this phenotype, we determine the structure of ribosomes lacking the single Ψ and its parental strain at ∼2.4-3 Å resolution using cryo-EM. Our findings demonstrate the significance of a single Ψ on H69 to its structure and the importance for its interactions with helix 44 and specific tRNAs. Our study suggests that rRNA modification affects translation of mRNAs carrying codon bias due to selective accommodation of tRNAs by the ribosome. Based on the high-resolution structures, we propose a mechanism explaining how the ribosome selects specific tRNAs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号