PUS1

PUS1
  • 文章类型: Journal Article
    骨转移是前列腺癌预后不良的重要原因。最近的证据强调了假尿苷合成酶在实体瘤进展中的关键作用,然而,驱动前列腺癌转移的特定酶仍未被识别。本研究揭示了FOXA1/PUS1/EIF3b信号轴在前列腺癌骨转移中的新调控机制。我们发现前列腺癌组织中PUS1表达升高,与较高的临床分级和较差的预后有关。敲除PUS1抑制转移,与其酶活性无关,EIF3b作为下游效应物,免受泛素介导的PUS1降解。EIF3b的过表达抵消了由于PUS1敲低引起的转移抑制。此外,显示FOXA1通过与其启动子结合而增强PUS1表达。MogrosideIV-E,一种特定的PUS1抑制剂,通过降低PUS1表达证明了有效的抗转移作用。我们的发现强调了FOXA1/PUS1/EIF3b轴作为前列腺癌骨转移的关键介质,并表明靶向该途径可能是一种有前途的治疗策略。
    Bone metastasis is a significant contributor to the poor prognosis in prostate cancer. Recent evidence highlights the pivotal role of pseudouridine synthases in solid tumor progression, yet the specific enzyme driving prostate cancer metastasis remains unidentified. This study uncovers a novel regulatory mechanism of the FOXA1/PUS1/EIF3b signaling axis in prostate cancer bone metastasis. We identified elevated PUS1 expression in prostate cancer tissues, correlating with higher clinical grade and worse prognosis. Knockdown of PUS1 inhibited metastasis independently of its enzymatic activity, with EIF3b acting as a downstream effector, protected from ubiquitin-mediated degradation by PUS1. Overexpression of EIF3b countered the metastasis suppression due to PUS1 knockdown. Additionally, FOXA1 was shown to enhance PUS1 expression by binding to its promoter. Mogroside IV-E, a specific PUS1 inhibitor, demonstrated potent anti-metastatic effects by reducing PUS1 expression. Our findings highlight the FOXA1/PUS1/EIF3b axis as a critical mediator of prostate cancer bone metastasis and suggest that targeting this pathway could be a promising therapeutic strategy.
    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
    伪尿苷(Φ),尿苷的异构体,普遍存在于RNA中,包括tRNA,rRNA,和mRNA。人假尿苷合酶3(PUS3)催化tRNA中位置38/39的假尿苷化。然而,它识别其RNA靶标并实现位点特异性的分子机制仍然难以捉摸。这里,我们确定了apo形式并与三个tRNA结合的PUS3的单颗粒冷冻EM结构,显示对称PUS3同二聚体如何识别tRNA并将靶尿苷定位在其活性位点附近。结构指导和患者来源的突变验证了我们在互补生化测定中的结构发现。此外,我们在HEK293细胞中删除了PUS1和PUS3,并通过Pseudo-seq定位了转录组范围的Φ位点。尽管在tRNA和mRNA中可以检测到PUS1依赖性位点,我们没有发现人类PUS3修饰mRNA的证据.我们的工作为人类PUS3介导的tRNA修饰提供了分子基础,并解释了其tRNA修饰活性如何与智力障碍有关。
    Pseudouridine (Ψ), the isomer of uridine, is ubiquitously found in RNA, including tRNA, rRNA, and mRNA. Human pseudouridine synthase 3 (PUS3) catalyzes pseudouridylation of position 38/39 in tRNAs. However, the molecular mechanisms by which it recognizes its RNA targets and achieves site specificity remain elusive. Here, we determine single-particle cryo-EM structures of PUS3 in its apo form and bound to three tRNAs, showing how the symmetric PUS3 homodimer recognizes tRNAs and positions the target uridine next to its active site. Structure-guided and patient-derived mutations validate our structural findings in complementary biochemical assays. Furthermore, we deleted PUS1 and PUS3 in HEK293 cells and mapped transcriptome-wide Ψ sites by Pseudo-seq. Although PUS1-dependent sites were detectable in tRNA and mRNA, we found no evidence that human PUS3 modifies mRNAs. Our work provides the molecular basis for PUS3-mediated tRNA modification in humans and explains how its tRNA modification activity is linked to intellectual disabilities.
    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mRNAs上核糖体占据的全基因组测量已经实现了翻译区的经验鉴定,但是与注释编码区域重叠的编码区域的高置信度检测仍然具有挑战性。这里,我们报告了一种灵敏而强大的算法,该算法揭示了出芽酵母中388个N末端截短的蛋白质的翻译,比以前已知的多30倍。我们对它们进行了广泛的实验验证,并定义了两个类。第一类缺乏大部分的注释蛋白质,并且倾向于由截短的转录物产生。我们证明了两个这样的情况,Yap5截断和Pus1截断,具有条件特异性调节和与其各自注释的同工型不同的功能。第二类截短的蛋白质同种型仅缺少注释蛋白质的小区域,并且不太可能从替代转录物同种型产生。许多显示不同的亚细胞定位比他们的注释对应物,代表双重定位功能相同的蛋白质的共同策略。补充信息中包含了本文透明的同行评审过程的记录。
    Genome-wide measurement of ribosome occupancy on mRNAs has enabled empirical identification of translated regions, but high-confidence detection of coding regions that overlap annotated coding regions has remained challenging. Here, we report a sensitive and robust algorithm that revealed the translation of 388 N-terminally truncated proteins in budding yeast-more than 30-fold more than previously known. We extensively experimentally validated them and defined two classes. The first class lacks large portions of the annotated protein and tends to be produced from a truncated transcript. We show that two such cases, Yap5truncation and Pus1truncation, have condition-specific regulation and distinct functions from their respective annotated isoforms. The second class of truncated protein isoforms lacks only a small region of the annotated protein and is less likely to be produced from an alternative transcript isoform. Many display different subcellular localizations than their annotated counterpart, representing a common strategy for dual localization of otherwise functionally identical proteins. A record of this paper\'s transparent peer review process is included in the supplemental information.
    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
    非小细胞肺癌(NSCLC)是肺癌的主要病理类型。在这项研究中,多组学分析显示,在NSCLC中,假尿苷合酶1(PUS1)显著增加,PUS1的高表达与较短的OS(总生存期)相关。PFS(无进展生存),和NSCLC患者的PPS(进展后生存)。临床亚组分析显示,PUS1可能参与了NSCLC的发生发展。此外,TIMER,估计,IPS分析表明,PUS1的表达与免疫细胞浸润有关,PUS1的表达与DC细胞浸润呈显著负相关。GESA分析还表明PUS1可能参与DNA_REPAIR,E2F_目标,MYC_TARGETS_V2、G2M_CHECKPOINT和MYC_TARGETS_V1通路并通过MCM5或XPO1触发NSCLC恶性肿瘤。此外,PUS1可能是NSCLC治疗的潜在靶点。
    Non-small cell lung cancer (NSCLC) is the main pathological type of lung cancer. In this study, multi-omics analysis revealed a significant increase of pseudouridine synthase 1 (PUS1) in NSCLC and the high expression of PUS1 was associated with shorter OS (Overall Survival), PFS (Progression Free Survival), and PPS (Post Progression Survival) of NSCLC patients. Clinical subgroup analysis showed that PUS1 may be involved in the occurrence and development of NSCLC. Besides, TIMER, ESTIMATE, and IPS analysis suggested that PUS1 expression was associated with immune cell infiltration, and the expression of PUS1 was significantly negatively correlated with DC cell infiltration. GESA analysis also indicated PUS1 may involve in DNA_REPAIR, E2F_TARGETS, MYC_TARGETS_V2, G2M_CHECKPOINT and MYC_TARGETS_V1 pathways and triggered NSCLC malignancy through MCM5 or XPO1. Furthermore, PUS1 may be a potential target for NSCLC therapy.
    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
    肾细胞癌是最常见的恶性肿瘤之一。尽管肿瘤学研究和外科治疗迅速发展,RCC预后无明显改善。因此,探索RCC的病理分子机制和开发新的治疗靶点具有重要意义。在这里,通过生物信息学分析和体外细胞实验,我们报告说,假尿苷合酶1(PUS1)的表达,属于参与RNA修饰的PUS酶家族,与RCC进展密切相关。此外,上调的PUS1表达导致RCC癌细胞活力升高,迁移,入侵和群体形成能力,而降低的PUS1表达对RCC细胞产生相反的作用。因此,我们的发现显示了PUS1在RCC细胞中的潜在作用,提供PUS1参与RCC进展的证据,这可能有助于RCC的诊断和临床干预。
    Renal cell carcinoma (RCC) is one of the most common malignancies. Despite the rapid development of the oncology research and surgical treatment, the prognosis of RCC has not significantly improved. Thus, exploration of the pathological molecular mechanism and development of new therapeutic targets of RCC are of great importance. Herein, by bioinformatic analysis and in vitro cell experiments, we report that, the expression of pseudouridine synthase 1 (PUS1), belonging to the family of PUS enzymes that participate in RNA modifications, is closely associated with RCC progression. In addition, the upregulated PUS1 expression results in the elevated RCC cancer cell viability, migration, invasion and colony formation ability, whereas the decreased PUS1 expression exerts the opposite effects on RCC cells. Thus, our findings show the potential role of PUS1 in RCC cells, providing with evidence that PUS1 is involved in RCC progression, which may help contribute to RCC diagnosis and intervention in clinic.
    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
    假尿苷合酶(PUS)在肝细胞癌(HCC)中的作用机制尚不明确,本研究的目的是探讨PUS基因在HCC中的作用。
    根据癌症基因组图谱(TCGA)鉴定了PUS酶的差异表达和预后基因,国际癌症基因组联盟(ICGC)和基因表达谱交互式分析(GEPIA)数据库。对于确定的基因,假尿苷合酶1(PUS1),用于进一步研究。采用t检验分析PUS1的临床病理特征。通过Kaplan-Meier(KM)分析和Cox比例风险回归模型探索预后意义。应用受试者工作特征(ROC)曲线评价诊断和预后价值。注释数据库,可视化,综合发现(DAVID)和基因集富集分析(GSEA),以探索PUS1的机制。应用广西队列验证差异表达。进行了体外细胞实验以研究对增殖的影响,活性氧(ROS)水平,迁移,敲低PUS1后肝癌细胞的侵袭。
    PUS1在HCC组织中显著过表达,高PUS1患者的临床病理特征不理想。生存分析显示,高PUS1表达与低总生存率(OS)和1年无复发生存率(RFS)相关。是一个独立的危险因素。同时,ROC曲线显示PUS1对HCC具有诊断和预后意义。功能富集分析提示PUS1可能参与代谢途径,线粒体功能,非酒精性脂肪性肝病(NAFLD),和一些重要的致癌途径。细胞实验显示,敲低PUS1显著限制了迁移,扩散,侵袭能力和提高肝癌细胞的ROS水平。
    PUS1可能是HCC的预后生物标志物和潜在治疗靶标。
    UNASSIGNED: The mechanisms of pseudouridine synthase (PUS) are not definite in hepatocellular carcinoma (HCC), the objective of this study is to investigate the effect of PUS genes in HCC.
    UNASSIGNED: Differentially expressed and prognostic gene of PUS enzymes was identified based on The Cancer Genome Atlas (TCGA), International Cancer Genome Consortium (ICGC) and Gene Expression Profiling Interactive Analysis (GEPIA) databases. For the identified gene, pseudouridine synthase 1 (PUS1), was used for further research. The clinicopathological feature of PUS1 was analyzed by Student\'s t-test. Prognostic significance was explored by Kaplan-Meier (KM) analysis and Cox proportional hazards regression model. Receiver operating characteristic (ROC) curve was applied to appraise diagnostic and prognostic value. The Database for Annotation, Visualization, and Integrated Discovery (DAVID) and Gene Set Enrichment Analysis (GSEA) were implemented to explore mechanism of PUS1. A Guangxi cohort was applied to verify differential expression. In vitro cell experiments were implemented to investigate the influence for proliferation, reactive oxygen species (ROS) level, migration, and invasion of HCC cells after a knockdown of PUS1.
    UNASSIGNED: PUS1 was significantly overexpressed in HCC tissues, and patients with high PUS1 were related to unpromising clinicopathological features. Survival analysis revealed high PUS1 expression was associated with a poor overall survival (OS) and 1 year-recurrence free survival (RFS), was an independent risk factor. Meanwhile, ROC curve showed that PUS1 had a diagnostic and prognostic significance to HCC. Functional enrichment analysis implied that PUS1 may be involved in metabolic pathways, mitochondrial function, non-alcoholic fatty liver disease (NAFLD), and some important carcinogenic pathways. Cell assays revealed that knockdown of PUS1 significantly constrained the migration, proliferation, invasion and improved the ROS level of HCC cells.
    UNASSIGNED: PUS1 may be a prognostic biomarker and a underlying treatment target for HCC.
    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
    近年来,乳腺癌患者的预后有了显著改善,但由于其侵袭性和异质性,复发和预后差仍然很常见。仍然需要开发可靠的生物标志物来预测预后和治疗有效性。最近,越来越多的研究表明,假尿苷合成酶有助于许多癌症的发展,但是它们对乳腺癌的影响在很大程度上仍然未知。使用综合分析,我们选择假尿苷合成酶1(PUS1)作为候选生物标志物.然后使用131名乳腺癌患者的组织微阵列来确定PUS1的临床意义和预后价值。进行RNA测序分析以鉴定在对照和PUS1敲低细胞之间不同的下游基因。使用细胞增殖评估PUS1对细胞表型的影响,菌落形成,和transwell入侵测定。我们发现,与配对的正常组织相比,乳腺肿瘤过度表达PUS1。PUS1表达与三阴性乳腺癌(TNBC)状态(P=0.020)和肿瘤分级(P<0.0001)呈正相关,但与年龄无关(P=0.736),肿瘤大小(P=0.608),淋巴结(P=0.742),雌激素受体(ER)(P=0.162),孕激素受体(PR)(P=0.901),人表皮生长因子受体2(HER2)(P=0.608)或肿瘤分期(P=0.411)。相对而言,PUS1水平高的患者总生存时间较短(P=0.0001),无复发生存时间较短(P=0.0093).单因素和多因素生存分析显示,患者的总体生存受PUS1评分的独立影响(单因素CoxP<0.0001,HR=5.176,95%CI=2.420-11.07;多因素CoxP=0.001,HR=5.291,95%CI=1.893-14.78)。RNA测序数据显示,PUS1敲低显著影响一系列与癌症相关的生物学过程,如细胞增殖和细胞迁移的调控,以及KEGG途径,包括线粒体自噬和PI3K-Akt信号传导。体外,PUS1的敲低能显著抑制MDA-MB-231细胞和BT-549细胞的增殖和集落形成能力。此外,与相应的对照组相比,低PUS1表达组的肿瘤细胞侵袭能力显著减弱.我们的结果表明,PUS1是一种新的生物标志物,可预测乳腺癌患者的不良预后,并可能被证明是一个有希望的治疗靶标。
    Breast cancer patients\' outcomes have improved dramatically in recent years, but relapses and poor prognosis remain common due to its aggressiveness and heterogeneity. The development of reliable biomarkers is still needed for predicting prognosis and treatment effectiveness. Recently, a growing body of research suggests that pseudouridine synthases contribute to the development of many cancers, but their contribution to breast cancer remains largely unknown. Using an integrative analysis, we selected pseudouridine synthase1(PUS1) as the candidate biomarker. A tissue microarray of 131 breast cancer patients was then utilized to determine the clinical significance and prognostic value of PUS1. RNA sequencing analysis was conducted to identify downstream genes that differ between control and PUS1 knockdown cells. The effect of PUS1 on phenotypes of cells was assessed using cell proliferation, colony formation, and transwell invasion assays. We found that breast tumors overexpressed PUS1 compared with paired normal tissues. PUS1 expression was positively correlated with triple-negative breast cancer (TNBC) status (P= 0.020) and tumor grade (P <0.0001), but not with age (P= 0.736), tumor size (P= 0.608), lymph node (P= 0.742), oestrogen receptor (ER) (P= 0.162), progesterone receptor (PR) (P= 0.901), human epidermal growth factor receptor 2 (HER2) (P= 0.608) or tumor stage (P= 0.411). Comparatively, patients with high PUS1 levels had shorter overall survival time (P=0.0001) and relapse-free survival time (P = 0.0093). A univariate and multivariate survival analysis suggested that the overall survival of patients was independently influenced by the PUS1 score (Univariate Cox P <0.0001, HR=5.176, 95% CI =2.420-11.07; Multivariate Cox P = 0.001, HR = 5.291, 95% CI =1.893-14.78). RNA sequencing data revealed the PUS1 knockdown significantly affects a series of cancer related biological process such as regulation of cell proliferation and cell migration, as well as KEGG pathways including Mitophagy and PI3K-Akt signaling. In vitro, knockdown of PUS1 significantly suppressed the proliferation and colony formation abilities of MDA-MB-231 cells and BT-549 cells. Additionally, the ability of tumor cells to invade was remarkably attenuated in low PUS1 expression groups compared with the corresponding control groups. Our results suggested that PUS1 is a novel biomarker that predicts poor outcomes in patients with breast cancer and may prove to be a promising treatment target.
    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
    Pus1依赖性假尿苷化发生在许多tRNA和多个位置,然而,这种修改的功能影响还没有完全理解。我们分析了PUS1缺失对酵母中tRNAGlnCUG对CAG(Gln)密码子的基本解码的影响。在将修饰缺陷与tRNAGlnCUG的不稳定变体组合时观察到合成致死性,指向低修饰的tRNA的严重CAG解码缺陷。此外,我们证明了tRNAGlnCUG变体对UAG终止密码子的误读受Pus1的积极影响。遗传方法进一步表明,轻度升高的温度通过不稳定的tRNAGlnCAG变体降低了CAG和UAG的解码效率。我们还确定了tRNAHisGUG对CGC(Arg)密码子的误读,其中CGC解码器tRNAArgICG包含依赖Pus1的伪尿嘧啶,但不是误译的mRNA。我们发现Pus1的缺失增加了tRNAHis对CGC的误读,证明了修饰在与非同义近同源tRNA的竞争中的积极作用。pus1突变体和携带不稳定tRNAGlnCAG的菌株中的部分体内解码缺陷和表型可被快速tRNA衰变(RTD)相关的MET22的额外缺失所抑制,这表明RTD介导的tRNA不稳定参与。
    Pus1-dependent pseudouridylation occurs in many tRNAs and at multiple positions, yet the functional impact of this modification is incompletely understood. We analyzed the consequences of PUS1 deletion on the essential decoding of CAG (Gln) codons by tRNAGlnCUG in yeast. Synthetic lethality was observed upon combining the modification defect with destabilized variants of tRNAGlnCUG, pointing to a severe CAG-decoding defect of the hypomodified tRNA. In addition, we demonstrated that misreading of UAG stop codons by a tRNAGlnCUG variant is positively affected by Pus1. Genetic approaches further indicated that mildly elevated temperature decreases the decoding efficiency of CAG and UAG via destabilized tRNAGlnCAG variants. We also determined the misreading of CGC (Arg) codons by tRNAHisGUG, where the CGC decoder tRNAArgICG contains Pus1-dependent pseudouridine, but not the mistranslating tRNAHis. We found that the absence of Pus1 increased CGC misreading by tRNAHis, demonstrating a positive role of the modification in the competition against non-synonymous near-cognate tRNA. Part of the in vivo decoding defects and phenotypes in pus1 mutants and strains carrying destabilized tRNAGlnCAG were suppressible by additional deletion of the rapid tRNA decay (RTD)-relevant MET22, suggesting the involvement of RTD-mediated tRNA destabilization.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Mitochondrial myopathy, lactic acidosis and sideroblastic anemia 1 (MLASA1) is a rare disease caused by biallelic pathogenic variants in the PUS1 gene. There are eleven MLASA1 patients reported worldwide with the majority of the patients originating from the Shiraz region of Iran. The rarity of this disease poses challenges to counseling patients due to a lack of natural history data. This report reviews what is known regarding MLASA1 and describes two brothers with MLASA1 who were cared for over the course of 10 years at the University of California Los Angeles. The brothers suffered from chronic anemia, transfusion dependency and muscle wasting that lead to respiratory insufficiency and death in one of the brothers.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Case Reports
    肌病-乳酸性酸中毒-铁粒母细胞性贫血(MLASA)综合征是一种罕见的常染色体隐性遗传疾病。我们研究了一名43岁的女性,从小就患有轻度认知障碍和铁粒幼细胞性贫血。她后来患上了肝病,心肌病,和胰岛素依赖型糖尿病。肌肉无力出现在青春期,43岁时,她无法行走。在PUS1基因中鉴定出两个新的不同突变:c.487delA(p。I163Lfs*4)和c.884G>A(p。R295Q).来自骨骼肌活检的DNA的定量分析显示,与对照组相比,患者的线粒体DNA(mtDNA)含量显着增加。该患者的临床和分子发现扩大了MLASA综合征的基因型-表型谱。
    Myopathy-lactic acidosis-sideroblastic anemia (MLASA) syndrome is a rare autosomal recessive disease. We studied a 43-year-old female presenting since childhood with mild cognitive impairment and sideroblastic anemia. She later developed hepatopathy, cardiomyopathy, and insulin-dependent diabetes. Muscle weakness appeared in adolescence and, at age 43, she was unable to walk. Two novel different mutations in the PUS1 gene were identified: c.487delA (p.I163Lfs*4) and c.884 G>A (p.R295Q). Quantitative analysis of DNA from skeletal muscle biopsies showed a significant increase in mitochondrial DNA (mtDNA) content in the patient compared to controls. Clinical and molecular findings of this patient widen the genotype-phenotype spectrum in MLASA syndrome.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号