YTHDF2

YTHDF2
  • 文章类型: Journal Article
    背景:KIAA1429,N6-甲基腺苷(m6A)甲基转移酶复合物的调节亚基,与各种癌症的进展有关。然而,KIAA1429在胃癌(GC)中的作用及其潜在机制仍然难以捉摸。本研究旨在探讨KIAA1429在GC中的作用并阐明其潜在机制。
    方法:使用定量实时PCR(qRT-PCR)评估KIAA1429在GC中的表达模式和临床相关性,西方印迹,免疫组织化学(IHC),和生物信息学分析。体外和体内功能损失和增益测定,M6A斑点印迹分析,甲基化RNA免疫沉淀测序(MeRIP-seq),RNA-seq,MeRIP-qPCR,双荧光素酶报告分析,RNA稳定性分析,RNA免疫沉淀(RIP)测定,并进行了RNA下拉测定,以研究KIAA1429在GC中的生物学功能和潜在的分子机制。
    结果:GC组织中KIAA1429的mRNA和蛋白表达均高于正常胃组织。KIAA1429高表达与GC患者预后不良呈正相关。KIAA1429不只增进GC细胞增殖,菌落形成,G2/M细胞周期过渡,迁移,和体外侵袭,但也增强了体内GC肿瘤的生长和转移。机械上,KIAA1429以m6A-YTHDF2依赖性方式增加RASD1mRNA的m6A水平并增强其稳定性,从而提高其表达。RASD1敲低部分挽救了KIAA1429敲低诱导的GC细胞原癌能力受损。GC组织中KIAA1429和RASD1的表达水平呈负相关。
    结论:KIAA1429通过以m6A-YTHDF2依赖性方式使RASD1mRNA不稳定而下调RASD1表达,从而在GC中发挥原癌作用。KIAA1429可作为GC的预后生物标志物和治疗靶标。
    BACKGROUND: KIAA1429, a regulatory subunit of the N6-methyladenosine (m6A) methyltransferase complex, has been implicated in the progression of various cancers. However, the role of KIAA1429 in gastric cancer (GC) and its underlying mechanisms remain elusive. This study aimed to investigate the role of KIAA1429 in GC and to elucidate the underlying mechanisms.
    METHODS: The expression patterns and clinical relevance of KIAA1429 in GC were assessed using quantitative real-time PCR (qRT-PCR), Western blotting, immunohistochemistry (IHC), and bioinformatic analysis. In vitro and in vivo loss- and gain-of-function assays, m6A dot blot assays, methylated RNA immunoprecipitation sequencing (MeRIP-seq), RNA-seq, MeRIP-qPCR, dual luciferase reporter assays, RNA stability assays, RNA immunoprecipitation (RIP) assays, and RNA pull-down assays were performed to investigate the biological functions and underlying molecular mechanisms of KIAA1429 in GC.
    RESULTS: Both the mRNA and protein expression of KIAA1429 were greater in GC tissues than in normal gastric tissues. High KIAA1429 expression correlated positively with poor prognosis in GC patients. KIAA1429 not only promoted GC cell proliferation, colony formation, G2/M cell cycle transition, migration, and invasion in vitro but also enhanced GC tumor growth and metastasis in vivo. Mechanistically, KIAA1429 increased the m6A level of RASD1 mRNA and enhanced its stability in an m6A-YTHDF2-dependent manner, thereby upregulating its expression. RASD1 knockdown partially rescued the KIAA1429 knockdown-induced impairment of pro‑oncogenic ability in GC cells. The expression levels of KIAA1429 and RASD1 were negatively correlated in GC tissues.
    CONCLUSIONS: KIAA1429 plays a pro‑oncogenic role in GC by downregulating RASD1 expression through destabilizing RASD1 mRNA in an m6A-YTHDF2-dependent manner. KIAA1429 may serve as a prognostic biomarker and therapeutic target for GC.
    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
    就发病率和死亡率而言,宫颈癌在女性中排名第四。RNA结合蛋白YTHN6-甲基腺苷RNA结合蛋白F2(YTHDF2)通过不完全了解的机制促进癌症进展。我们展示了YTHDF2如何控制宫颈癌细胞的命运,以及YTHDF2是否可以成为宫颈癌治疗的有效靶标。进行球体形成和碱性磷酸酶染色测定以评估YTHDF2敲低后宫颈癌细胞的肿瘤干性。通过流式细胞术和TUNEL测定检测细胞凋亡。化合物4PBA和SP600125用于研究JNK,内质网应激,肿瘤干性,和凋亡。来自癌症基因组图谱(TCGA)数据库和基因表达综合(GEO)的数据显示GLI家族锌指2(GLI2)可能是YTHDF2的靶标。采用转录抑制剂放线菌素D和双荧光素酶报告基因测定来研究GLI2mRNA与YTHDF2之间的关联。产生裸鼠异种移植物以评估YTHDF2敲低对体内宫颈癌生长的影响。敲低YTHDF2上调GLI2的表达,导致JNK磷酸化和内质网应激。这些过程抑制了宫颈癌细胞的增殖及其肿瘤细胞的干性和促进凋亡。总之,YTHDF2的敲除显著影响宫颈癌细胞的进展,使其成为治疗宫颈癌的潜在靶点。
    Cervical cancer ranks fourth in women in terms of incidence and mortality. The RNA-binding protein YTH N6-methyladenosine RNA-binding protein F2 (YTHDF2) contributes to cancer progression by incompletely understood mechanisms. We show how YTHDF2 controls the fate of cervical cancer cells and whether YTHDF2 could be a valid target for the therapy of cervical cancer. Sphere formation and alkaline phosphatase staining assays were performed to evaluate tumor stemness of cervical cancer cells following YTHDF2 knockdown. Apoptosis was detected by flow cytometry and TUNEL assay. The compounds 4PBA and SP600125 were used to investigate the correlation between JNK, endoplasmic reticulum stress, tumor stemness, and apoptosis. Data from The Cancer Genome Atlas (TCGA) databases and Gene Expression Omnibus (GEO) revealed that GLI family zinc finger 2 (GLI2) might be the target of YTHDF2. The transcription inhibitor actinomycin D and dual-luciferase reporter gene assays were employed to investigate the association between the GLI2 mRNA and YTHDF2. Nude mouse xenografts were generated to assess the effects of YTHDF2 knockdown on cervical cancer growth in vivo. Knockdown of YTHDF2 up-regulated the expression of GLI2, leading to JNK phosphorylation and endoplasmic reticulum stress. These processes inhibited the proliferation of cervical cancer cells and their tumor cell stemness and promotion of apoptosis. In conclusion, the knockdown of YTHDF2 significantly affects the progression of cervical cancer cells, making it a potential target for treating cervical 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
    背景:蛋白质精氨酸甲基转移酶6(PRMT6)在各种病理生理过程和疾病中起着至关重要的作用。胶质母细胞瘤(GBM;WHO4级胶质瘤)是成人中最常见和致命的原发性脑肿瘤,预后极差,尽管不如其他系统性恶性肿瘤常见。我们目前的研究发现PRMT6在GBM中上调,增强肿瘤恶性程度。然而,PRMT6在GBM中的调控过程和潜在分子机制的细节仍未被探索。
    方法:使用神经胶质瘤公共数据库评估GBM中PRMT6的表达和预后意义,免疫组织化学(IHC),和免疫印迹。Scratch和Transwell测定法检查了GBM细胞迁移和侵袭。免疫印迹法评估了上皮间质转化(EMT)和Wnt-β-catenin通路相关蛋白的表达。双荧光素酶报告基因测定和ChIP-qPCR评估了PRMT6和YTHDF2之间的调控关系。裸鼠中的原位肿瘤模型评估了体内条件。
    结果:生物信息学分析表明PRMT6和YTHDF2在GBM中高表达,与预后不良有关。功能实验表明PRMT6和YTHDF2促进GBM迁移,入侵,EMT。机制实验揭示了PRMT6和CDK9共同调节YTHDF2表达。YTHDF2结合并促进Wnt-β-catenin通路的负调节因子APC和GSK3βmRNA的降解,激活它,从而增强GBM恶性肿瘤。
    结论:我们的结果表明PRMT6-YTHDF2-Wnt-β-Catenin轴促进GBM迁移,入侵,和体外和体内EMT,可能作为GBM的治疗靶标。
    BACKGROUND: Protein arginine methyltransferase 6 (PRMT6) plays a crucial role in various pathophysiological processes and diseases. Glioblastoma (GBM; WHO Grade 4 glioma) is the most common and lethal primary brain tumor in adults, with a prognosis that is extremely poor, despite being less common than other systemic malignancies. Our current research finds PRMT6 upregulated in GBM, enhancing tumor malignancy. Yet, the specifics of PRMT6\'s regulatory processes and potential molecular mechanisms in GBM remain largely unexplored.
    METHODS: PRMT6\'s expression and prognostic significance in GBM were assessed using glioma public databases, immunohistochemistry (IHC), and immunoblotting. Scratch and Transwell assays examined GBM cell migration and invasion. Immunoblotting evaluated the expression of epithelial-mesenchymal transition (EMT) and Wnt-β-catenin pathway-related proteins. Dual-luciferase reporter assays and ChIP-qPCR assessed the regulatory relationship between PRMT6 and YTHDF2. An in situ tumor model in nude mice evaluated in vivo conditions.
    RESULTS: Bioinformatics analysis indicates high expression of PRMT6 and YTHDF2 in GBM, correlating with poor prognosis. Functional experiments show PRMT6 and YTHDF2 promote GBM migration, invasion, and EMT. Mechanistic experiments reveal PRMT6 and CDK9 co-regulate YTHDF2 expression. YTHDF2 binds and promotes the degradation of negative regulators APC and GSK3β mRNA of the Wnt-β-catenin pathway, activating it and consequently enhancing GBM malignancy.
    CONCLUSIONS: Our results demonstrate the PRMT6-YTHDF2-Wnt-β-Catenin axis promotes GBM migration, invasion, and EMT in vitro and in vivo, potentially serving as a therapeutic target for GBM.
    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
    N6-甲基腺苷(m6A)是哺乳动物细胞中最丰富的mRNA修饰,在mRNA稳定性的调节中具有重要意义,翻译和拼接。此外,它主要通过募集YTHDF2阅读器蛋白在调节RNA降解中起关键作用。然而,通过YTHDF2结合对m6A甲基化mRNA的mRNA降解的选择性调节知之甚少。为了提高我们的认识,我们开发了m6A-BERT-Deg,适用于预测YTHDF2介导的m6A甲基化mRNA降解的BERT模型。我们通过整合HeLa细胞系的多个数据源,精心组装了高质量的训练数据集。为了克服小训练样本的局限性,我们采用了预训练微调策略,首先对427.760未标记的m6A位点序列进行模型的自监督预训练.测试结果证明了这种预训练策略在使m6A-BERT-Deg优于其他基准模型方面的重要性。我们进一步进行了全面的模型解释,并揭示了一个令人惊讶的发现,即m6A位点附近的辅因子的存在可能会破坏YTHDF2介导的mRNA降解,随后增强mRNA的稳定性。我们还将分析扩展到HEK293细胞系,对环境依赖性YTHDF2介导的mRNA降解的启示。
    N6-methyladenosine (m6A) is the most abundant mRNA modification within mammalian cells, holding pivotal significance in the regulation of mRNA stability, translation and splicing. Furthermore, it plays a critical role in the regulation of RNA degradation by primarily recruiting the YTHDF2 reader protein. However, the selective regulation of mRNA decay of the m6A-methylated mRNA through YTHDF2 binding is poorly understood. To improve our understanding, we developed m6A-BERT-Deg, a BERT model adapted for predicting YTHDF2-mediated degradation of m6A-methylated mRNAs. We meticulously assembled a high-quality training dataset by integrating multiple data sources for the HeLa cell line. To overcome the limitation of small training samples, we employed a pre-training-fine-tuning strategy by first performing a self-supervised pre-training of the model on 427 760 unlabeled m6A site sequences. The test results demonstrated the importance of this pre-training strategy in enabling m6A-BERT-Deg to outperform other benchmark models. We further conducted a comprehensive model interpretation and revealed a surprising finding that the presence of co-factors in proximity to m6A sites may disrupt YTHDF2-mediated mRNA degradation, subsequently enhancing mRNA stability. We also extended our analyses to the HEK293 cell line, shedding light on the context-dependent YTHDF2-mediated mRNA degradation.
    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
    许多病毒,包括口蹄疫病毒(FMDV),可以通过巨自噬/自噬促进宿主蛋白的降解,从而促进病毒复制。然而,在FMDV感染过程中,自噬与先天性免疫反应之间的调节机制尚不完全清楚.这里,我们发现宿主GTPBP4/NOG1(GTP结合蛋白4)是先天免疫应答的负调节因子。GTPBP4缺乏促进抗病毒先天性免疫反应,导致GTPBP4促进FMDV复制的能力。同时,GTPBP4缺陷型小鼠对FMDV感染的抗性更强。对抗宿主的抗病毒免疫,FMDV结构蛋白VP1促进GTPBP4的表达,VP1的第209个位点负责这种作用。机械上,FMDVVP1在病毒感染期间促进自噬,并在AKT-MTOR依赖性自噬途径中与YTHDF2(YTHN6-甲基腺苷RNA结合蛋白F2)相互作用并降解,导致GTPBP4mRNA和蛋白质水平增加。增加的GTPBP4抑制IRF3与Ifnb/Ifn-β启动子的结合,抑制FMDV诱导的I型干扰素的产生。总之,我们的研究揭示了VP1如何通过自噬途径负调节先天免疫的潜在机制,这将有助于了解FMDV感染期间宿主先天免疫反应的负调控以及GTPBP4和YTHDF2的功能。缩写:3-MA:3-甲基腺嘌呤;ACTB:肌动蛋白β;ATG:自噬相关;ChIP:染色质免疫沉淀;CQ:氯喹;DAPI:4',6-二氨基-2-苯基吲哚;dpi:感染后天数;EV71:肠道病毒71;FMDV:口蹄疫病毒;GTPP4/NOG1:GTP结合蛋白4;HIF1A:缺氧诱导因子1亚基α;hpt:转染后小时数;IFNB/IFN-β:干扰素β;IRF3:TAF1TTBtuoH蛋白3TMTN相关基因1基因1
    Many viruses, including foot-and-mouth disease virus (FMDV), can promote the degradation of host proteins through macroautophagy/autophagy, thereby promoting viral replication. However, the regulatory mechanism between autophagy and innate immune responses is not fully understood during FMDV infection. Here, we found that the host GTPBP4/NOG1 (GTP binding protein 4) is a negative regulator of innate immune responses. GTPBP4 deficiency promotes the antiviral innate immune response, resulting in the ability of GTPBP4 to promote FMDV replication. Meanwhile, GTPBP4-deficient mice are more resistant to FMDV infection. To antagonize the host\'s antiviral immunity, FMDV structural protein VP1 promotes the expression of GTPBP4, and the 209th site of VP1 is responsible for this effect. Mechanically, FMDV VP1 promotes autophagy during virus infection and interacts with and degrades YTHDF2 (YTH N6-methyladenosine RNA binding protein F2) in an AKT-MTOR-dependent autophagy pathway, resulting in an increase in GTPBP4 mRNA and protein levels. Increased GTPBP4 inhibits IRF3 binding to the Ifnb/Ifn-β promoter, suppressing FMDV-induced type I interferon production. In conclusion, our study revealed an underlying mechanism of how VP1 negatively regulates innate immunity through the autophagy pathway, which would contribute to understanding the negative regulation of host innate immune responses and the function of GTPBP4 and YTHDF2 during FMDV infection.Abbreviation: 3-MA:3-methyladenine; ACTB: actin beta; ATG: autophagy related; ChIP:chromatin immunoprecipitation; CQ: chloroquine; DAPI:4\',6-diamidino-2-phenylindole; dpi: days post-infection; EV71:enterovirus 71; FMDV: foot-and-mouth disease virus; GTPBP4/NOG1: GTPbinding protein 4; HIF1A: hypoxia inducible factor 1 subunit alpha;hpt:hours post-transfection; IFNB/IFN-β:interferon beta; IRF3: interferon regulatory factor 3; MAP1LC3/LC3:microtubule associated protein 1 light chain 3; MAVS: mitochondriaantiviral signaling protein; MOI: multiplicity of infection; MTOR:mechanistic target of rapamycin kinase; m6A: N(6)-methyladenosine;qPCR:quantitativePCR; SIRT3:sirtuin 3; SQSTM1/p62: sequestosome 1; STING1: stimulator ofinterferon response cGAMP interactor 1; siRNA: small interfering RNA;TBK1: TANK binding kinase 1; TCID50:50% tissue culture infectious doses; ULK1: unc-51 like autophagyactivating kinase 1; UTR: untranslated region; WT: wild type; YTHDF2:YTH N6-methyladenosine RNA binding protein F2.
    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
    前列腺癌是男性癌症相关死亡率的主要原因。RNA的失调腺嘌呤N-6甲基化(m6A)有助于癌症的恶性程度。mRNA上的m6A可能影响mRNA的剪接,营业额,运输和翻译。M6A发挥这些作用,至少部分地,通过专用的m6A阅读器蛋白,包括YTHDF2。YTHDF2是发育所必需的,而其失调在包括前列腺癌的各种癌症中可见。然而,YTHDF2在癌症中失调和功能的潜在机制仍然难以捉摸。这里,我们发现去泛素酶OTUB1通过抑制YTHDF2蛋白的泛素化增加其稳定性。通过体内和体外泛素化试验,OTUB1显示阻断泛素向YTHDF2的转移,而与其去泛素酶活性无关。此外,功能转录组数据和m6A测序数据的分析确定PRSS8为潜在的抑癌基因。OTUB1和YTHDF2降低PRSS8的mRNA和蛋白质水平,PRSS8是胰蛋白酶样丝氨酸蛋白酶。机械上,YTHDF2结合PRSS8mRNA并以m6A依赖性方式促进其降解。对细胞和小鼠模型的进一步功能研究表明,PRSS8是前列腺癌中OTUB1-YTHDF2轴的关键下游效应物。我们在前列腺癌细胞中发现,PRSS8通过E-cadherin降低核β-catenin水平,这与它的蛋白酶活性无关。总的来说,我们的研究揭示了YTHDF2蛋白稳定性的关键调节因子,并在前列腺癌中建立了功能性OTUB1-YTHDF2-PRSS8轴.
    Prostate cancer is a leading cause of cancer-related mortality in males. Dysregulation of RNA adenine N-6 methylation (m6A) contributes to cancer malignancy. m6A on mRNA may affect mRNA splicing, turnover, transportation, and translation. m6A exerts these effects, at least partly, through dedicated m6A reader proteins, including YTH domain-containing family protein 2 (YTHDF2). YTHDF2 is necessary for development while its dysregulation is seen in various cancers, including prostate cancer. However, the mechanism underlying the dysregulation and function of YTHDF2 in cancer remains elusive. Here, we find that the deubiquitinase OUT domain-containing ubiquitin aldehyde-binding protein 1 (OTUB1) increases YTHDF2 protein stability by inhibiting its ubiquitination. With in vivo and in vitro ubiquitination assays, OTUB1 is shown to block ubiquitin transfer to YTHDF2 independent of its deubiquitinase activity. Furthermore, analysis of functional transcriptomic data and m6A-sequencing data identifies PRSS8 as a potential tumor suppressor gene. OTUB1 and YTHDF2 decrease mRNA and protein levels of PRSS8, which is a trypsin-like serine protease. Mechanistically, YTHDF2 binds PRSS8 mRNA and promotes its degradation in an m6A-dependent manner. Further functional study on cellular and mouse models reveals PRSS8 is a critical downstream effector of the OTUB1-YTHDF2 axis in prostate cancer. We find in prostate cancer cells, PRSS8 decreases nuclear β-catenin level through E-cadherin, which is independent of its protease activity. Collectively, our study uncovers a key regulator of YTHDF2 protein stability and establishes a functional OTUB1-YTHDF2-PRSS8 axis in prostate 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
    非编码RNA(ncRNAs),包括环状RNA(circRNAs)和N6-甲基腺苷(m6A),已被证明在各种疾病的发展中起关键作用,包括肥胖和代谢紊乱相关的脂肪性肝病(MAFLD)。肥胖是一种由体内脂肪过度堆积引起的慢性疾病,最近变得更加普遍,是MAFLD的首要危险因素。肥胖的原因可能涉及遗传的相互作用,行为,和社会因素。m6ARNA甲基化可能为理解肥胖和MAFLD的发展以及基因表达的转录后调控提供了新的启示。特别是,circRNAs,microRNAs(miRNAs),m6A可能与MAFLD的进展有关。有趣的是,M6A修改可以调节翻译,降解,和ncRNAs的其他功能。miRNA/circRNAs也可以通过影响作者来调节m6A修饰,橡皮擦,和读者。反过来,ncRNAs可以以不同的方式调节m6A调节因子的表达。然而,关于这些ncRNAs和m6A如何相互作用以影响肝脏疾病的促进的证据有限。似乎m6A可以发生在DNA中,RNA,和可能与几种生物学特性相关的蛋白质。这项研究提供了对m6A修饰和ncRNAs与肝脏疾病关联的机制理解。尤其是MAFLD。理解m6A修饰和ncRNAs之间的关联可能有助于MAFLD治疗策略的发展。
    Noncoding RNAs (ncRNAs), including circular RNAs (circRNAs) and N6-methyladenosine (m6A), have been shown to play a critical role in the development of various diseases including obesity and metabolic disorder-associated fatty liver disease (MAFLD). Obesity is a chronic disease caused by excessive fat accumulation in the body, which has recently become more prevalent and is the foremost risk factor for MAFLD. Causes of obesity may involve the interaction of genetic, behavioral, and social factors. m6A RNA methylation might add a novel inspiration for understanding the development of obesity and MAFLD with post-transcriptional regulation of gene expression. In particular, circRNAs, microRNAs (miRNAs), and m6A might be implicated in the progression of MAFLD. Interestingly, m6A modification can modulate the translation, degradation, and other functions of ncRNAs. miRNAs/circRNAs can also modulate m6A modifications by affecting writers, erasers, and readers. In turn, ncRNAs could modulate the expression of m6A regulators in different ways. However, there is limited evidence on how these ncRNAs and m6A interact to affect the promotion of liver diseases. It seems that m6A can occur in DNA, RNA, and proteins that may be associated with several biological properties. This study provides a mechanistic understanding of the association of m6A modification and ncRNAs with liver diseases, especially for MAFLD. Comprehension of the association between m6A modification and ncRNAs may contribute to the development of treatment tactics for MAFLD.
    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
    N6-甲基腺苷(m6A)修饰通过影响肿瘤微环境(TME)来协调癌症形成和进展。对于肝细胞癌(HCC),免疫逃避和血管生成是其TME的特征。YTHN6-甲基腺苷RNA结合蛋白2(YTHDF2)的作用,作为一名m6读者,在调节HCCTME方面还没有完全理解。在这里,发现YTHDF2启动子区的三甲基化组蛋白H3赖氨酸4和H3赖氨酸27乙酰化修饰增强了其在HCC中的表达,并在HCC中上调YTHDF2预测预后较差。动物实验表明,Ythdf2耗竭抑制自发性肝癌形成,而其过度表达促进异种移植HCC进展。机械上,YTHDF2识别ETS变异转录因子5(ETV5)mRNA的5'非翻译区中的m6A修饰,并招募真核翻译起始因子3亚基B以促进其翻译。升高的ETV5表达诱导程序性死亡配体-1和血管内皮生长因子A的转录,从而促进HCC免疫逃避和血管生成。通过含有小干扰RNA的适体/脂质体靶向YTHDF2成功地抑制HCC免疫逃避和血管生成。一起,这一发现揭示了YTHDF2在HCC预后和靶向治疗中的潜在应用。
    N6-methyladenosine (m6A) modification orchestrates cancer formation and progression by affecting the tumor microenvironment (TME). For hepatocellular carcinoma (HCC), immune evasion and angiogenesis are characteristic features of its TME. The role of YTH N6-methyladenosine RNA binding protein 2 (YTHDF2), as an m6A reader, in regulating HCC TME are not fully understood. Herein, it is discovered that trimethylated histone H3 lysine 4 and H3 lysine 27 acetylation modification in the promoter region of YTHDF2 enhanced its expression in HCC, and upregulated YTHDF2 in HCC predicted a worse prognosis. Animal experiments demonstrated that Ythdf2 depletion inhibited spontaneous HCC formation, while its overexpression promoted xenografted HCC progression. Mechanistically, YTHDF2 recognized the m6A modification in the 5\'-untranslational region of ETS variant transcription factor 5 (ETV5) mRNA and recruited eukaryotic translation initiation factor 3 subunit B to facilitate its translation. Elevated ETV5 expression induced the transcription of programmed death ligand-1 and vascular endothelial growth factor A, thereby promoting HCC immune evasion and angiogenesis. Targeting YTHDF2 via small interference RNA-containing aptamer/liposomes successfully both inhibited HCC immune evasion and angiogenesis. Together, this findings reveal the potential application of YTHDF2 in HCC prognosis and targeted treatment.
    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)是第五最常诊断的癌症,在癌症相关死亡人数中排名第三。公认的长链非编码RNA(lncRNA)在几种癌症类型中的参与,包括HCC,启发本研究探索一种新的lncRNA在HCC进展中的功能重要性。为了实现这一点,在三组不同的HCC组织上进行了lncRNA微阵列分析,显示LINC00707是最显著上调的lncRNA。对其生物学功能的进一步研究表明,LINC00707充当癌基因,通过增强增殖来驱动HCC进展,肝癌细胞的迁移和侵袭。提供了机械见解,证明LINC00707与YTHN6-甲基腺苷RNA结合蛋白2(YTHDF2)相互作用,从而促进YTHDF2蛋白的泛素化依赖性降解。此外,发现LINC00707通过其与YTHDF2的相互作用影响NK-92MI细胞对HCC细胞的细胞毒性。这些发现有助于更深入地了解LINC00707在HCC进展中所起的作用。
    Hepatocellular carcinoma (HCC) is the fifth most frequently diagnosed cancer and ranks third in cancer-related fatalities. The recognized involvement of long noncoding RNAs (lncRNAs) in several cancer types, including HCC, inspired this study to explore a novel lncRNA\'s functional importance in the progression of HCC. To achieve this, lncRNA microarray analysis was conducted on three distinct sets of HCC tissues, revealing LINC00707 as the most significantly upregulated lncRNA. Further research into its biological functions has revealed that LINC00707 acts as an oncogene, driving HCC progression by enhancing the proliferation, migration and invasion of HCC cells. Mechanistic insights were provided, demonstrating that LINC00707 interacts with YTH N6-methyladenosine RNA-binding protein 2 (YTHDF2), thus facilitating the ubiquitination-dependent degradation of the YTHDF2 protein. Furthermore, LINC00707 was found to influence the cytotoxicity of NK-92MI cells against HCC cells through its interactions with YTHDF2. These findings significantly contribute to a deeper understanding of the role played by LINC00707 in the progression of 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
    目的:m6ARNA修饰通路在多种细胞过程和病毒生命周期中发挥重要作用。这里,我们研究了PIAS1与m6A阅读蛋白YTHDF2之间的关系,该蛋白通过与m6A修饰的RNA结合而参与调节RNA稳定性。我们发现YTHDF2的N端和C端区域都与PIAS1相互作用。我们显示PIAS1促进YTHDF2在三个特定赖氨酸残基处的SUMOylation。我们还证明PIAS1增强YTHDF2的抗EBV活性。我们进一步揭示了PIAS1介导其他YTHDF家族成员的SUMOylation,即,YTHDF1和YTHDF3,以限制EBV的复制。这些发现共同阐明了YTHDF蛋白在通过PIAS1介导的SUMO化控制病毒RNA衰变和EBV复制中的重要调节机制。
    YTH N6-methyladenosine RNA-binding protein F2 (YTHDF2) is a member of the YTH protein family that binds to N6-methyladenosine (m6A)-modified RNA, regulating RNA stability and restricting viral replication, including Epstein-Barr virus (EBV). PIAS1 is an E3 small ubiquitin-like modifier (SUMO) ligase known as an EBV restriction factor, but its role in YTHDF2 SUMOylation remains unclear. In this study, we investigated the functional regulation of YTHDF2 by PIAS1. We found that PIAS1 promotes the SUMOylation of YTHDF2 at three specific lysine residues (K281, K571, and K572). Importantly, PIAS1 synergizes with wild-type YTHDF2, but not a SUMOylation-deficient mutant, to limit EBV lytic replication. Mechanistically, YTHDF2 lacking SUMOylation exhibits reduced binding to EBV transcripts, leading to increased viral mRNA stability. Furthermore, PIAS1 mediates SUMOylation of YTHDF2\'s paralogs, YTHDF1 and YTHDF3, to restrict EBV replication. These results collectively uncover a unique mechanism whereby YTHDF family proteins control EBV replication through PIAS1-mediated SUMOylation, highlighting the significance of SUMOylation in regulating viral mRNA stability and EBV replication.IMPORTANCEm6A RNA modification pathway plays important roles in diverse cellular processes and viral life cycle. Here, we investigated the relationship between PIAS1 and the m6A reader protein YTHDF2, which is involved in regulating RNA stability by binding to m6A-modified RNA. We found that both the N-terminal and C-terminal regions of YTHDF2 interact with PIAS1. We showed that PIAS1 promotes the SUMOylation of YTHDF2 at three specific lysine residues. We also demonstrated that PIAS1 enhances the anti-EBV activity of YTHDF2. We further revealed that PIAS1 mediates the SUMOylation of other YTHDF family members, namely, YTHDF1 and YTHDF3, to limit EBV replication. These findings together illuminate an important regulatory mechanism of YTHDF proteins in controlling viral RNA decay and EBV replication through PIAS1-mediated SUMOylation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号