UPF1

UPF1
  • 文章类型: Journal Article
    最近对遗传稳健性的研究表明,转录适应(TA)是生物体可以通过同源基因的激活来补偿基因突变的一种机制。这里,我们发现基因突变,在淀粉样前体蛋白-b(appb)基因中引入过早终止密码子(PTC),激活了另外两个应用程序家庭成员的TA,斑马鱼中的appa和淀粉样前体样蛋白2(aplp2)。观察到的appa和aplp2的转录反应需要突变体mRNA的降解,并且不依赖于Appb蛋白水平。此外,在人神经元祖细胞(hNPC)中观察到淀粉样前体蛋白(APP)家族成员之间的TA,然而,补偿仅在早期神经元分化期间存在,并且在分化更高的神经元阶段或成年斑马鱼大脑中无法检测到。使用击倒和化学抑制,我们表明,无义介导的mRNA衰变(NMD)参与突变mRNA的降解,Upf1和Upf2,NMD途径中的关键蛋白,调节APA的内源性转录水平,appb,aplp1和aplp2总之,我们的研究结果表明,App家族成员的表达水平受NMD通路的调控,使app/APPmRNA失稳的突变可通过TA在斑马鱼和人类神经元祖细胞中诱导其他家族成员的遗传补偿.显著性陈述增加APP水平的遗传变异与阿尔茨海默病(AD)病理生理学相关。因此,理解调节APP表达水平的机制是关键的兴趣。这里,我们确定转录适应是APP家族成员可以调节同一家族中基因表达水平以补偿另一个家族基因缺失的一种机制。在引入PTC后,补偿是通过NMD途径中介导mRNA衰减的因素驱动的。有趣的是,我们的数据还表明,即使在生理条件下,NMD监测机制也是微调APP家族成员mRNA水平的重要方面。因此,我们的发现为APP成员之间的薪酬提供了见解,并揭示了可以监管APP的新目标。
    Studies on genetic robustness recently revealed transcriptional adaptation (TA) as a mechanism by which an organism can compensate for genetic mutations through activation of homologous genes. Here, we discovered that genetic mutations, introducing a premature termination codon (PTC) in the amyloid precursor protein-b (appb) gene, activated TA of two other app family members, appa and amyloid precursor-like protein-2 (aplp2), in zebrafish. The observed transcriptional response of appa and aplp2 required degradation of mutant mRNA and did not depend on Appb protein level. Furthermore, TA between amyloid precursor protein (APP) family members was observed in human neuronal progenitor cells; however, compensation was only present during early neuronal differentiation and could not be detected in a more differentiated neuronal stage or adult zebrafish brain. Using knockdown and chemical inhibition, we showed that nonsense-mediated mRNA decay (NMD) is involved in degradation of mutant mRNA and that Upf1 and Upf2, key proteins in the NMD pathway, regulate the endogenous transcript levels of appa, appb, aplp1, and aplp2 In conclusion, our results suggest that the expression level of App family members is regulated by the NMD pathway and that mutations destabilizing app/APP mRNA can induce genetic compensation by other family members through TA in both zebrafish and human neuronal progenitors.
    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
    肝细胞癌(HCC)是一种常见的恶性肿瘤,具有复杂的异质性和耐药性。对铁性凋亡的抵抗与HCC的进展密切相关。虽然HCC肿瘤对铁凋亡的敏感性不同,这种异质性背后的确切因素仍不清楚.在这项研究中,我们试图阐明导致HCC铁凋亡抵抗的机制。在HCC细胞系Huh7中使用亚毒性浓度(IC20)的铁凋亡诱导擦除素进行的全基因组CRISPR/Cas9筛选显示,TRIM34是HCC中铁凋亡抗性的关键驱动因素。进一步的研究表明,TRIM34抑制肝癌细胞的铁凋亡,促进它们的扩散,迁移,和体内外侵袭。此外,TRIM34在肝癌肿瘤组织中表达升高,与预后不良有关。机械上,TRIM34直接与上移码1(UPF1)交互,无义介导的mRNA衰减(NMD)途径的核心组成部分,促进其泛素化和降解。这种相互作用抑制GPX4转录物降解,从而促进HCC中这种关键的铁凋亡抑制因子的蛋白质水平。鉴于癌症中铁死亡与适应性免疫反应之间的紧密串扰,具有TRIM34靶向敲低的HCC细胞表现出抗PD-1治疗的改善反应。一起来看,TRIM34/UPF1/GPX4轴介导HCC的铁凋亡抗性,从而促进恶性表型。因此,靶向TRIM34可能代表了一种有前途的HCC治疗新策略。
    Hepatocellular carcinoma (HCC) is a prevalent malignancy characterized by complex heterogeneity and drug resistance. Resistance to ferroptosis is closely related to the progression of HCC. While HCC tumors vary in their sensitivity to ferroptosis, the precise factors underlying this heterogeneity remain unclear. In this study, we sought to elucidate the mechanisms that contribute to ferroptosis resistance in HCC. Whole-genome CRISPR/Cas9 screen using a subtoxic concentration (IC20) of ferroptosis inducer erastin in the HCC cell line Huh7 revealed TRIM34 as a critical driver of ferroptosis resistance in HCC. Further investigation revealed that TRIM34 suppresses ferroptosis in HCC cells, promoting their proliferation, migration, and invasion both in vitro and in vivo. Furthermore, TRIM34 expression is elevated in HCC tumor tissues, correlating with a poor prognosis. Mechanistically, TRIM34 directly interacts with Up-frameshift 1 (UPF1), a core component of the nonsense-mediated mRNA decay (NMD) pathway, to promote its ubiquitination and degradation. This interaction suppresses GPX4 transcript degradation, thus promoting the protein levels of this critical ferroptosis suppressor in HCC. In light of the close crosstalk between ferroptosis and the adaptive immune response in cancer, HCC cells with targeting knockdown of TRIM34 exhibited an improved response to anti-PD-1 treatment. Taken together, the TRIM34/UPF1/GPX4 axis mediates ferroptosis resistance in HCC, thereby promoting malignant phenotypes. Targeting TRIM34 may thus represent a promising new strategy for HCC treatment.
    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:报道新发现的具有家族性渗出性玻璃体视网膜病变(FEVR)独特形式的TSPAN12突变,并找出TSPAN12中重复的新型内含子变异导致FEVR的可能机制。
    结果:通过基于面板的NGS检测到9个具有独特形式的FEVR的TSPAN12突变。MINI-Gene分析显示mRNA的两种剪接模式,处理两个不同的条带A和B,和突变型显示与Exon11跳跃的剪接模式的替换。野生型和突变型TSPAN12载体的构建显示过早终止密码子(PTC)的出现。体外表达检测显示,与野生型组相比,用突变载体转染的细胞中TSPAN12mRNA和蛋白质的表达水平显着下调。相反,翻译抑制剂CHX和UPF1的小干扰RNA(si-UPF1)显着增加了用突变载体转染的细胞中TSPAN12的mRNA或蛋白质表达。
    结论:在9例FEVR患者中报道了TSPAN12基因的9个突变,这些患者具有一系列独特的眼部异常。三个新的TSPAN12突变触发NMD会导致参与微纤维生物合成和组装的TSPAN12蛋白的减少,这可能会导致FEVR,并表明内含子序列分析可能是遗传咨询和产前诊断的重要工具。
    BACKGROUND: To report newly found TSPAN12 mutations with a unique form of familial exudative vitreoretinopathy (FEVR) and find out the possible mechanism of a repeated novel intronic variant in TSPAN12 led to FEVR.
    RESULTS: Nine TSPAN12 mutations with a unique form of FEVR were detected by panel-based NGS. MINI-Gene assay showed two splicing modes of mRNA that process two different bands A and B, and mutant-type shows replacement with the splicing mode of Exon11 hopping. Construction of wild-type and mutant TSPAN12 vector showed the appearance of premature termination codons (PTC). In vitro expression detection showed significant down-regulated expression level of TSPAN12 mRNAs and proteins in cells transfected with mutant vectors compared with in wild-type group. On the contrary, translation inhibitor CHX and small interfering RNA of UPF1 (si-UPF1) significantly increased mRNA or protein expression of TSPAN12 in cells transfected with the mutant vectors.
    CONCLUSIONS: Nine mutations in TSPAN12 gene are reported in 9 FEVR patients with a unique series of ocular abnormalities. The three novel TSPAN12 mutations trigger NMD would cause the decrease of TSPAN12 proteins that participate in biosynthesis and assembly of microfibers, which might lead to FEVR, and suggest that intronic sequence analysis might be a vital tool for genetic counseling and prenatal diagnoses.
    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
    据报道,右美托咪定(Dex)在阿尔茨海默病(AD)中起神经保护作用。然而,具体机制尚不清楚.在AD模型中阐明Dex调控神经细胞凋亡的分子机制。将SH-SY5Y细胞用Aβ1-42(10μM)处理24h后,建立了体外AD模型。通过RIP分析验证了UPF1,lncRNASNHG14和HSPB8之间的相互作用。细胞活力,凋亡,基因的水平,并通过CCK-8测定法检测蛋白质,流式细胞术,蛋白质印迹,和qRT-PCR,分别。Dex下调lncRNASNHG14水平并抑制神经细胞凋亡。LncRNASNHG14过表达逆转了Dex对AD模型神经细胞凋亡的抑制作用。LncRNASNHG14通过招募UPF1减弱HSPB8mRNA的稳定性。HSPB8过表达抑制AD模型神经细胞凋亡。此外,HSPB8敲低可逆转Dex对AD模型神经细胞凋亡的抑制作用。我们的研究表明,Dex通过抑制lncRNASNHG14/UPF1轴促进HSPB8的表达,从而抑制AD中神经细胞的凋亡。
    Dexmedetomidine (Dex) is reported to play a neuroprotective role in Alzheimer\'s disease (AD). However, the specific mechanism remains unclear. Figure out the underlying molecular mechanism of Dex regulating nerve cell apoptosis in the AD model. The AD model in vitro was established after SH-SY5Y cells were treated with Aβ1 - 42 at (10 μM) for 24 h. The interaction among UPF1, lncRNA SNHG14, and HSPB8 was verified by RIP assay. Cell viability, apoptosis, the level of genes, and proteins were detected by CCK-8 assay, flow cytometry, Western blot, and qRT-PCR, respectively. Dex downregulated lncRNA SNHG14 level and inhibited apoptosis of nerve cells. LncRNA SNHG14 overexpression reversed the inhibitory effect of Dex on nerve cell apoptosis in the AD model. LncRNA SNHG14 attenuated HSPB8 mRNA stability by recruiting UPF1. HSPB8 overexpression inhibited apoptosis of nerve cells in the AD model. Moreover, HSPB8 knockdown reversed the inhibitory effect of Dex on nerve cell apoptosis in the AD model. Our study demonstrated that Dex promoted HSPB8 expression via inhibiting the lncRNA SNHG14/UPF1 axis to inhibit nerve cell apoptosis in AD.
    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
    上移码蛋白1(UPF1)对于无义介导的信使RNA衰变(NMD)至关重要。它以其在降解异常和特异性RNA中的细胞保护作用而闻名。UPF1在多种肿瘤中失调,这与不良预后和低总生存率有关。然而,UPF1在肺癌中的作用尚不清楚.目前的研究表明,UPF1可能是肿瘤治疗的潜在靶点。结果还证明了UPF1在调节肺癌增殖和转移方面的潜在功效。我们的发现表明,这些功能可归因于对FOXO1蛋白稳定性的抑制。此外,PBK参与了UPF1对FOXO1的监管。这一结果为肺癌患者提供了一种新的治疗策略。
    Up-frameshift protein 1 (UPF1) is essential for nonsense-mediated messenger RNA decay (NMD). It is best known for its cytoprotective role in degrading aberrant and specific RNAs. UPF1 is dysregulated in multiple tumors, which correlates with poor prognosis and low overall survival.However,the role of UPF1 in lung cancer remains unclear.Current study shows that UPF1 could be a potential target for oncology therapies. The results also demonstrated the potential efficiency of UPF1 in regulating the proliferation and metastasis of lung cancer. Our findings suggest that those functions can be attributed to the inhibition of the stability of FOXO1 protein. In addition, PBK participates in the regulation of FOXO1 by UPF1.This result provides a new therapeutic strategy for lung cancer 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
    通过体液免疫起作用的有效疫苗寻求产生高亲和力抗体。我们先前的研究确定了CXCR5的3UTR中单核苷酸多态性rs3922G与乙型肝炎疫苗无反应相关。CXCR5在暗区(DZ)和亮区(LZ)之间的差异表达对于组织生发中心(GC)的功能结构至关重要。在这项研究中,我们报道了RNA结合蛋白IGF2BP3可以通过无义介导的mRNA衰变途径与含有rs3922变体的CXCR5mRNA结合,促进其降解.IGF2BP3的缺乏导致CXCR5表达增加,这导致DZ和LZ之间CXCR5差异表达的消失,杂乱无章的GCs,异常体细胞超突变,和减少高亲和力抗体的产生。此外,IGF2BP3对含有rs3922G的序列的亲和力低于对rs3922A的亲和力,这可以解释对乙肝疫苗接种的无反应。一起,我们的研究结果表明,IGF2BP3通过与含rs3922的序列结合以调节CXCR5的表达,在GC中产生高亲和力抗体中起着至关重要的作用.本文受版权保护。保留所有权利。
    Effective vaccines that function through humoral immunity seek to produce high-affinity antibodies. Our previous research identified the single-nucleotide polymorphism rs3922G in the 3\'UTR of CXCR5 as being associated with nonresponsiveness to the hepatitis B vaccine. The differential expression of CXCR5 between the dark zone (DZ) and light zone (LZ) is critical for organizing the functional structure of the germinal center (GC). In this study, we report that the RNA-binding protein IGF2BP3 can bind to CXCR5 mRNA containing the rs3922 variant to promote its degradation via the nonsense-mediated mRNA decay pathway. Deficiency of IGF2BP3 leads to increased CXCR5 expression, which results in the disappearance of CXCR5 differential expression between DZ and LZ, disorganized GCs, aberrant somatic hypermutations, and reduced production of high-affinity antibodies. Furthermore, the affinity of IGF2BP3 for the rs3922G-containing sequence is lower than that for the rs3922A counterpart, which may explain the nonresponsiveness to the hepatitis B vaccination. Together, our findings suggest that IGF2BP3 plays a crucial role in the production of high-affinity antibodies in the GC by binding to the rs3922-containing sequence to regulate CXCR5 expression.
    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
    长非编码RNA(lncRNA)生长停滞特异性转录物5(GAS5)已被证明是许多癌症的调节因子,包括非小细胞肺癌(NSCLC)。因此,其在NSCLC发展过程中的作用及机制有待进一步揭示。实时定量PCR检测GAS5、脂肪量和肥胖相关蛋白(FTO)和含溴结构域蛋白4(BRD4)的表达水平。蛋白质印迹分析用于检测FTO的蛋白表达,BRD4、上移码蛋白1(UPF1)和自噬相关标志物。甲基化RNA免疫沉淀用于评估由FTO调节的GAS5的m6A水平。用MTT法测定细胞增殖和凋亡,EdU测定和流式细胞术。通过免疫荧光染色和透射电子显微镜评估自噬能力。构建异种移植肿瘤模型以探讨FTO和GAS5对体内NSCLC肿瘤生长的影响。通过下拉试验证实了UPF1和GAS5或BRD4之间的相互作用,RIP测定,双荧光素酶报告分析,和染色质免疫沉淀。荧光原位杂交用于分析GAS5和UPF1的共定位。放线菌素D处理用于评价BRD4mRNA稳定性。GAS5在NSCLC组织中下调,并与NSCLC患者的不良预后相关。FTO在NSCLC中高表达,它通过降低GAS5m6A甲基化水平来抑制GAS5的表达。FTO抑制GAS5在体外促进NSCLC细胞自噬性死亡,在体内抑制NSCLC肿瘤生长。此外,GAS5能够与UPF1相互作用以降低BRD4的mRNA稳定性。敲除BRD4逆转了GAS5或UPF1沉默对NSCLC自噬性细胞死亡的抑制作用。研究结果表明,由FTO介导的lncRNAGAS5可能通过与UPF1相互作用以降低BRD4mRNA的稳定性,从而促进NSCLC的自噬性细胞死亡。提示GAS5可能是NSCLC进展的重要治疗靶点。
    Long non-coding RNA (lncRNA) growth arrest-specific transcript 5 (GAS5) has been shown to be a regulator for many cancers, including non-small cell lung cancer (NSCLC). Therefore, its role and mechanism in the process of NSCLC deserve to be further revealed. The expression levels of GAS5, fat mass and obesity-associated protein (FTO) and bromodomain-containing protein 4 (BRD4) were detected by quantitative real-time PCR. Western blot analysis was used to examine the protein expression of FTO, BRD4, up-frameshift protein 1 (UPF1) and autophagy-related markers. Methylated RNA immunoprecipitation was used to assess the m6A level of GAS5 regulated by FTO. Cell proliferation and apoptosis were determined using MTT assay, EdU assay and flow cytometry. Autophagy ability was assessed by immunofluorescence staining and transmission electron microscope. Xenograft tumor model was constructed to explore the effects of FTO and GAS5 on NSCLC tumor growth in vivo. The interaction between UPF1 and GAS5 or BRD4 was confirmed by pull-down assay, RIP assay, dual-luciferase reporter assay, and chromatin immunoprecipitation. Fluorescent in situ hybridization was used to analyze the co-localization of GAS5 and UPF1. Actinomycin D treatment was employed to evaluate BRD4 mRNA stability. GAS5 was downregulated in NSCLC tissues and was associated with poor prognosis in NSCLC patients. FTO was highly expressed in NSCLC, and it inhibited GAS5 expression by reducing GAS5 m6A methylation level. GAS5 suppressed by FTO could promote the autophagic death of NSCLC cells in vitro and inhibit NSCLC tumor growth in vivo. In addition, GAS5 was able to interact with UPF1 to reduce the mRNA stability of BRD4. Knockdown of BRD4 reversed the inhibition of GAS5 or UPF1 silencing on the autophagic cell death of NSCLC. The findings of the study showed that lncRNA GAS5 mediated by FTO could contribute to the autophagic cell death of NSCLC by interacting with UPF1 to reduce BRD4 mRNA stability, suggesting that GAS5 might be a vital therapy target for NSCLC progression.
    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
    简介:环境压力促进影响基因表达的表观遗传改变,随后参与疾病的病理过程。在表观遗传法规中,十11易位(Tet)酶将DNA和RNA中的5-甲基胞嘧啶(5mC)氧化为5-羟甲基胞嘧啶(5hmC),并在疾病的发病机理中起关键作用。我们先前的结果表明,慢性应激增加了暴露于慢性轻度应激(CMS)的小鼠海马中细胞质Tet2的表达。在慢性应激相关过程中,细胞质Tet2是否会改变RNA5hmC修饰仍然是未知的。方法:探讨细胞质Tet2在CMS条件下的作用,建立CMS小鼠模型,通过斑点印迹法检测5hmCRNA的表达。我们通过免疫共沉淀结合质谱验证了Tet2及其相互作用蛋白的相互作用,并通过Tet2和上游移码1(Upf1)相互作用RNA的聚类分析筛选了下游靶基因。Westernblot检测蛋白的表达,qRT-PCR检测筛选的靶基因的表达。结果:在这项研究中,我们发现CMS条件下胞质Tet2表达增加导致5hmC总RNA修饰增加。Tet2与关键的无义介导的mRNA衰变(NMD)因子Upf1相互作用,调节应激相关基因如Unc5bmRNA的稳定性,从而可能影响神经发育。讨论:总之,这项研究表明,Tet2介导的RNA5hmC修饰参与应激相关的mRNA稳定性调节,并可能作为慢性应激相关疾病如抑郁症的潜在治疗靶点。
    Introduction: Environmental stress promotes epigenetic alterations that impact gene expression and subsequently participate in the pathological processes of the disorder. Among epigenetic regulations, ten-eleven Translocation (Tet) enzymes oxidize 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) in DNA and RNA and function as critical players in the pathogenesis of diseases. Our previous results showed that chronic stress increases the expression of cytoplasmic Tet2 in the hippocampus of mice exposed to chronic mild stress (CMS). Whether the cytoplasmic Tet2 alters RNA 5hmC modification in chronic stress-related processes remains largely unknown. Methods: To explore the role of cytoplasmic Tet2 under CMS conditions, we established CMS mice model and detected the expression of RNA 5hmC by dot blot. We verified the interaction of Tet2 and its interacting protein by co-immunoprecipitation combined with mass spectrometry and screened downstream target genes by cluster analysis of Tet2 and upstream frameshift 1 (Upf1) interacting RNA. The expression of protein was detected by Western blot and the expression of the screened target genes was detected by qRT-PCR. Results: In this study, we found that increased cytoplasmic Tet2 expression under CMS conditions leads to increase in total RNA 5hmC modification. Tet2 interacted with the key non-sense-mediated mRNA decay (NMD) factor Upf1, regulated the stability of stress-related genes such as Unc5b mRNA, and might thereby affect neurodevelopment. Discussion: In summary, this study revealed that Tet2-mediated RNA 5hmC modification is involved in stress-related mRNA stability regulation and may serve as a potential therapeutic target for chronic stress-related diseases such as depression.
    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:上移码蛋白1(UPF1)是含有过早终止密码子(PTCs)的mRNA的无义介导的mRNA衰减(NMD)的关键成分。在各种癌症中已经报道了UPF1的失调。然而,UPF1在肾透明细胞癌(ccRCC)中的表达及其临床意义尚不清楚。方法:为了检测UPF1在ccRCC中的表达及其与ccRCC临床特征的关系,从癌症基因组图谱(TCGA)分析大量RNA测序数据,基因表达综合(GEO)和ArrayExpress数据库。通过多种免疫评分算法评估UPF1对ccRCC的免疫微环境的影响,以使用ccRCC单细胞测序(scRNA)数据鉴定通常表达UPF1的细胞组。此外,通过加权基因相关网络分析(WGCNA)鉴定与UPF1共表达的基因,其次是KEGG和Reactome富集分析。进行了一系列功能实验以评估UPF1在肾癌细胞中的作用。最后,还进行了UPF1的泛癌症分析。结果:与正常组织相比,ccRCC患者肿瘤组织中UPF1mRNA和蛋白的表达水平明显下降。此外,UPF1低表达的患者预后较差.免疫微环境分析表明UPF1免疫细胞浸润密切相关,ccRCCscRNA-seq数据确定UPF1主要在巨噬细胞中表达。WGCNA分析表明,共表达基因的功能主要集中在细胞增殖和细胞过程中。实验表明,敲除UPF1可以促进入侵,ccRCC细胞的迁移和增殖。最后,泛癌症分析显示,UPF1疾病与各种癌症结局密切相关.结论:UPF1可能在ccRCC中起肿瘤抑制作用,并调节免疫微环境。UPF1的丢失可以预测ccRCC的预后,使其成为有前途的生物标志物,并为预防和治疗提供新的参考。
    Background: Up frameshift protein 1 (UPF1) is a key component of nonsense-mediated mRNA decay (NMD) of mRNA containing premature termination codons (PTCs). The dysregulation of UPF1 has been reported in various cancers. However, the expression profile of UPF1 and its clinical significance in clear cell renal cell carcinoma (ccRCC) remains unclear. Methods: In order to detect UPF1 expression in ccRCC and its relationship with the clinical features of ccRCC, bulk RNA sequencing data were analyzed from The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO) and ArrayExpress databases. The impact of UPF1 on the immune microenvironment of ccRCC was evaluated by multiple immune scoring algorithms to identify the cell groups that typically express UPF1 using ccRCC single cell sequencing (scRNA) data. In addition, genes co-expressed with UPF1 were identified by the weighted gene correlation network analysis (WGCNA), followed by KEGG and Reactome enrichment analysis. A series of functional experiments were performed to assess the roles of UPF1 in renal cancer cells. Finally, pan-cancer analysis of UPF1 was also performed. Results: Compared with normal tissues, the expression levels of UPF1 mRNA and protein in tumor tissues of ccRCC patients decreased significantly. In addition, patients with low expression of UPF1 had a worse prognosis. Analysis of the immune microenvironment indicated that UPF1 immune cell infiltration was closely related and the ccRCC scRNA-seq data identified that UPF1 was mainly expressed in macrophages. WGCNA analysis suggested that the functions of co-expressed genes are mainly enriched in cell proliferation and cellular processes. Experimental tests showed that knockdown of UPF1 can promote the invasion, migration and proliferation of ccRCC cells. Lastly, pan-cancer analysis revealed that UPF1 disorders were closely associated with various cancer outcomes. Conclusions: UPF1 may play a tumor suppressive role in ccRCC and modulate the immune microenvironment. The loss of UPF1 can predict the prognosis of ccRCC, making it a promising biomarker and providing a new reference for prevention and treatment.
    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:化疗耐药是导致胰腺癌患者预后不良的主要因素,而癌症的干性是与化疗耐药相关的最关键因素之一,也是癌症治疗的一个非常有前途的方向。然而,癌症干性的确切分子机制尚未完全阐明。
    方法:m6A-RNA免疫沉淀和测序用于筛选m6A相关的mRNA和lncRNA。qRT-PCR和FISH用于分析DDIT4-AS1表达。球样形成,菌落形成,进行蛋白质印迹和流式细胞术测定以分析PDAC细胞的癌症干细胞性和化学敏感性。进行异种移植实验以分析体内肿瘤形成率和生长。RNA测序,使用蛋白质印迹和生物信息学分析来鉴定DDIT4-AS1的下游途径。IP,进行RIP和RNA下拉测定以测试DDIT4-AS1、DDIT4和UPF1之间的相互作用。产生患者来源的异种移植(PDX)小鼠模型以评估对GEM的化学敏感性。
    结果:DDIT4-AS1被确定为ALKBH5的下游靶标之一,将HuR募集到m6A修饰的位点对于DDIT4-AS1的稳定至关重要。DDIT4-AS1在PDAC中上调,与不良预后呈正相关。DDIT4-AS1沉默抑制干性并增强对GEM(吉西他滨)的化学敏感性。机械上,DDIT4-AS1通过阻止SMG5和PP2A与UPF1的结合来促进UPF1的磷酸化,从而降低DDIT4mRNA的稳定性并激活mTOR通路。此外,在PDX衍生模型中抑制DDIT4-AS1增强了GEM对PDAC的抗肿瘤作用。
    结论:ALKBH5介导的m6A修饰导致PDAC中DDIT4-AS1过表达,和DDIT-AS1通过使DDIT4不稳定和激活mTOR通路增加癌症干性并抑制对GEM的化学敏感性。针对DDIT4-AS1及其通路的方法可能是治疗PDAC化学耐药的有效策略。
    Chemoresistance is a major factor contributing to the poor prognosis of patients with pancreatic cancer, and cancer stemness is one of the most crucial factors associated with chemoresistance and a very promising direction for cancer treatment. However, the exact molecular mechanisms of cancer stemness have not been completely elucidated.
    m6A-RNA immunoprecipitation and sequencing were used to screen m6A-related mRNAs and lncRNAs. qRT-PCR and FISH were utilized to analyse DDIT4-AS1 expression. Spheroid formation, colony formation, Western blot and flow cytometry assays were performed to analyse the cancer stemness and chemosensitivity of PDAC cells. Xenograft experiments were conducted to analyse the tumour formation ratio and growth in vivo. RNA sequencing, Western blot and bioinformatics analyses were used to identify the downstream pathway of DDIT4-AS1. IP, RIP and RNA pulldown assays were performed to test the interaction between DDIT4-AS1, DDIT4 and UPF1. Patient-derived xenograft (PDX) mouse models were generated to evaluate chemosensitivities to GEM.
    DDIT4-AS1 was identified as one of the downstream targets of ALKBH5, and recruitment of HuR onto m6A-modified sites is essential for DDIT4-AS1 stabilization. DDIT4-AS1 was upregulated in PDAC and positively correlated with a poor prognosis. DDIT4-AS1 silencing inhibited stemness and enhanced chemosensitivity to GEM (Gemcitabine). Mechanistically, DDIT4-AS1 promoted the phosphorylation of UPF1 by preventing the binding of SMG5 and PP2A to UPF1, which decreased the stability of the DDIT4 mRNA and activated the mTOR pathway. Furthermore, suppression of DDIT4-AS1 in a PDX-derived model enhanced the antitumour effects of GEM on PDAC.
    The ALKBH5-mediated m6A modification led to DDIT4-AS1 overexpression in PDAC, and DDIT-AS1 increased cancer stemness and suppressed chemosensitivity to GEM by destabilizing DDIT4 and activating the mTOR pathway. Approaches targeting DDIT4-AS1 and its pathway may be an effective strategy for the treatment of chemoresistance in PDAC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号