Mettl14

METTL14
  • 文章类型: Journal Article
    细胞分裂周期5-样(CDC5L)蛋白与各种癌症的发展有关。然而,其在肺腺癌(LUAD)进展中的作用仍不确定.我们的发现揭示了LUAD中CDC5L的频繁上调,这与较差的总体生存率和晚期临床分期相关。体外实验证明CDC5L过表达刺激细胞增殖,迁移,以及LUAD细胞的入侵,而CDC5L敲低对这些细胞过程具有抑制作用。此外,在异种移植小鼠模型中,沉默CDC5L可显着抑制肿瘤生长和转移。机械上,CDC5L通过转录调控WNT7B激活Wnt/β-catenin信号通路,从而促进LUAD进展。此外,METTL14介导的m6A修饰有助于以IGF2BP2依赖性方式上调CDC5L。总的来说,我们的研究揭示了一种新的分子机制,通过这种机制,m6A诱导的CDC5L通过WNT7B的转录调节激活Wnt/β-catenin通路,在LUAD中起癌基因的作用,提示CDC5L可能作为LUAD的一个有希望的预后标志物和治疗靶点。
    Cell division cycle 5-like (CDC5L) protein is implicated in the development of various cancers. However, its role in the progression of lung adenocarcinoma (LUAD) remains uncertain. Our findings revealed frequent upregulation of CDC5L in LUAD, which correlated with poorer overall survival rates and advanced clinical stages. In vitro experiments demonstrated that CDC5L overexpression stimulated the proliferation, migration, and invasion of LUAD cells, whereas CDC5L knockdown exerted suppressive effects on these cellular processes. Furthermore, silencing CDC5L significantly inhibited tumor growth and metastasis in a xenograft mouse model. Mechanistically, CDC5L activates the Wnt/β-catenin signaling pathway by transcriptionally regulating WNT7B, thereby promoting LUAD progression. Besides, METTL14-mediated m6A modification contributed to CDC5L upregulation in an IGF2BP2-dependent manner. Collectively, our study uncovers a novel molecular mechanism by which the m6A-induced CDC5L functions as an oncogene in LUAD by activating the Wnt/β-catenin pathway through transcriptional regulation of WNT7B, suggesting that CDC5L may serve as a promising prognostic marker and therapeutic target for LUAD.
    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:高级别浆液性卵巢癌(HGSOC)是一种常见的妇科恶性肿瘤,预后不良。中药配方二枝草灵汤(EZMLD)具有抗癌潜力。本研究旨在阐明EZMLD对HGSOC的体外和体内抗癌作用。
    方法:从Sprague-Dawley大鼠制备含EZMLD的血清,用于以不同浓度治疗SKOV3卵巢癌细胞24小时和48小时,以测定IC50。浓度为0%,5%,选择24小时的10%用于随后的体外实验。通过过表达这些基因并将EZMLD与METTL3/14敲低相结合,探索了METTL3和METTL14在SKOV3细胞中的作用。研究集中在细胞活力和凋亡,凋亡相关蛋白表达,和KRT8mRNAm6A修饰。对于体内研究,将36只BALB/c裸鼠分为6组,涉及EZMLD(6.75、13.5和27g/kg)和METTL3或METTL14敲除,每天EZMLD灌胃两周。
    结果:体外,在SKOV3细胞中,含EZMLD的血清在24小时时具有8.29%和在48小时时具有5.95%的IC50值。含EZMLD的血清降低SKOV3细胞活力并增加细胞凋亡。EZMLD上调METTL3/14和FAS介导的凋亡蛋白,而下调角蛋白8(KRT8)。EZMLD增加KRT8mRNAm6A甲基化。METTL3/14过表达降低SKOV3细胞活力,增加细胞凋亡,而METTL3/14敲除减轻了EZMLD的影响。在体内,EZMLD抑制SKOV3异种移植物生长,引起显著的细胞凋亡和调节蛋白表达。
    结论:EZMLD对卵巢癌具有治疗潜力,可考虑用于其他癌症类型。未来的研究可能会探索其超越细胞凋亡的更广泛的影响。
    BACKGROUND: High-grade serous ovarian cancer (HGSOC) is a common gynecologic malignancy with a poor prognosis. The traditional Chinese medicine formula Erzhimaoling decoction (EZMLD) has anticancer potential. This study aims to elucidate the anticancer effects of EZMLD on HGSOC in vitro and in vivo.
    METHODS: EZMLD-containing serum was prepared from Sprague-Dawley rats for treating SKOV3 ovarian cancer cells at varying concentrations for 24 h and 48 h to determine the IC50. Concentrations of 0%, 5%, and 10% for 24 h were chosen for subsequent in vitro experiments. The roles of METTL3 and METTL14 in SKOV3 cells were explored by overexpressing these genes and combining EZMLD with METTL3/14 knockdown. Investigations focused on cell viability and apoptosis, apoptosis-related protein expression, and KRT8 mRNA m6A modification. For in vivo studies, 36 BALB/c nude mice were divided into six groups involving EZMLD (6.75, 13.5, and 27 g/kg) and METTL3 or METTL14 knockdowns, with daily EZMLD gavage for two weeks.
    RESULTS: In vitro, EZMLD-containing serum had IC50 values of 8.29% at 24 h and 5.95% at 48 h in SKOV3 cells. EZMLD-containing serum decreased SKOV3 cell viability and increased apoptosis. EZMLD upregulated METTL3/14 and FAS-mediated apoptosis proteins, while downregulating Keratin 8 (KRT8). EZMLD increased KRT8 mRNA m6A methylation. METTL3/14 overexpression reduced SKOV3 cell viability and increased apoptosis, while METTL3/14 knockdown mitigated EZMLD\'s effects. In vivo, EZMLD suppressed SKOV3 xenografts growth, causing significant apoptosis and modulating protein expression.
    CONCLUSIONS: EZMLD has therapeutic potential for ovarian cancer and may be considered for other cancer types. Future research may explore its broader effects beyond cell apoptosis.
    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)甲基化是与药物成瘾相关的重要表观遗传机制。然而,m6A修饰与羟考酮奖励之间的关系研究较少。基于开放的现场测试,本研究使用染色质免疫沉淀PCR评估羟考酮奖励,免疫荧光,和RNA测序。以剂量和时间依赖性方式观察到纹状体神经元中羟考酮丰度引起的METTL14蛋白的显着增加和PP1α蛋白的减少。羟考酮显著增加LSD1表达,纹状体中H3K4me1表达降低。在野外测试中,纹状体内注射METTL14siRNA,HOTAIRsiRNA,或LSD1shRNA阻断羟考酮诱导的运动活性增加。用METTL14/HOTAIRsiRNA和LSD1shRNA处理后,PP1α的下调也受到抑制。LSD1shRNA也逆转了由羟考酮诱导的LSD1与CoRest的增强结合以及CoRest与PP1α基因的增强结合。此外,H3K4me1去甲基化也被处理阻断。总之,研究证实,METTL14介导的HOTAIR上调导致PP1α抑制,这反过来促进了LSD1的募集,从而催化H3K4me1去甲基化并促进羟考酮成瘾。
    N6-methyladenosine (m6A) methylation is a vital epigenetic mechanism associated with drug addiction. However, the relationship between m6A modification and oxycodone rewarding is less well explored. Based on an open field test, the present study evaluated oxycodone rewarding using chromatin immunoprecipitation PCR, immunofluorescence, and RNA sequencing. A marked increase in METTL14 protein and a decrease in PP1α protein due to oxycodone abundance in the striatal neurons were observed in a dose- and time-dependent manner. Oxycodone markedly increased LSD1 expression, and decreased H3K4me1 expression in the striatum. In the open field test, intra-striatal injection of METTL14 siRNA, HOTAIR siRNA, or LSD1 shRNA blocked oxycodone-induced increase in locomotor activity. The downregulation of PP1α was also inhibited after treatment with METTL14/HOTAIR siRNA and LSD1 shRNA. Enhanced binding of LSD1 with CoRest and of CoRest with the PP1α gene induced by oxycodone was also reversed by LSD1 shRNA. In addition, H3K4me1 demethylation was also blocked by the treatment. In summary, the investigation confirmed that METTL14-mediated upregulation of HOTAIR resulted in the repression of PP1α, which in turn facilitated the recruitment of LSD1, thus catalyzing H3K4me1 demethylation and promoting oxycodone addiction.
    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:N6-甲基腺苷(m6A)修饰对于调节RNA加工和表达至关重要,特别是在恶性肿瘤进展的情况下。然而,鼻咽癌(NPC)中m6A修饰的探索仍然非常有限。
    方法:使用m6A斑点印迹测定法分析NPC中的RNAm6A水平。从公共数据库以及RT-qPCR和免疫组织化学分析NPC组织中甲基转移酶样14(METTL14)的表达水平。通过体外和体内功能测定探讨了METTL14表达对NPC增殖和转移的影响。使用m6A和基因表达谱微阵列数据筛选METTL14的靶向基因。使用放线菌素D处理和多聚体分析来检测ANKRD22的半衰期和翻译效率。流式细胞术,免疫荧光和免疫沉淀用于验证ANKRD22对NPC细胞脂质代谢的作用。METTL14、GINS3、POLE2、PLEK2和FERMT1基因启动子附近的H3K27AC信号的ChIP-qPCR分析。
    结果:我们在NPC中发现了METTL14,与不良患者预后相关。体外和体内测定表明METTL14积极促进NPC细胞增殖和转移。METTL14在ANKRD22信使核糖核酸(mRNA)上催化m6A修饰,由读者IGF2BP2识别,导致增加的mRNA稳定性和更高的翻译效率。此外,ANKRD22,线粒体上的代谢相关蛋白,与SLC25A1相互作用以增强柠檬酸盐的运输,提高细胞内乙酰辅酶A含量。ANKRD22的这种双重影响促进了脂质代谢重编程和细胞脂质合成,同时通过提高细胞核中的表观遗传组蛋白乙酰化水平来上调与细胞周期(GINS3和POLE2)和细胞骨架(PLEK2和FERMT1)相关的基因的表达。有趣的是,我们的发现强调了在METTL14启动子附近ANKRD22介导的组蛋白H3赖氨酸27乙酰化(H3K27AC)信号升高,这有助于一个正反馈循环延续NPC的恶性进展。
    结论:确定的METTL14-ANKRD22-SLC25A1轴是NPC的有希望的治疗靶点,这些分子也可以作为新的诊断生物标志物。
    BACKGROUND: N6-methyladenosine (m6A) modification is essential for modulating RNA processing as well as expression, particularly in the context of malignant tumour progression. However, the exploration of m6A modification in nasopharyngeal carcinoma (NPC) remains very limited.
    METHODS: RNA m6A levels were analysed in NPC using m6A dot blot assay. The expression level of methyltransferase-like 14 (METTL14) within NPC tissues was analysed from public databases as well as RT-qPCR and immunohistochemistry. The influences on METTL14 expression on NPC proliferation and metastasis were explored via in vitro as well as in vivo functional assays. Targeted genes of METTL14 were screened using the m6A and gene expression profiling microarray data. Actinomycin D treatment and polysome analysis were used to detect the half-life and translational efficiency of ANKRD22. Flow cytometry, immunofluorescence and immunoprecipitation were used to validate the role of ANKRD22 on lipid metabolism in NPC cells. ChIP-qPCR analysis of H3K27AC signalling near the promoters of METTL14, GINS3, POLE2, PLEK2 and FERMT1 genes.
    RESULTS: We revealed METTL14, in NPC, correlating with poor patient prognosis. In vitro and in vivo assays indicated METTL14 actively promoted NPC cells proliferation and metastasis. METTL14 catalysed m6A modification on ANKRD22 messenger ribonucleic acid (mRNA), recognized by the reader IGF2BP2, leading to increased mRNA stability and higher translational efficiency. Moreover, ANKRD22, a metabolism-related protein on mitochondria, interacted with SLC25A1 to enhance citrate transport, elevating intracellular acetyl-CoA content. This dual impact of ANKRD22 promoted lipid metabolism reprogramming and cellular lipid synthesis while upregulating the expression of genes associated with the cell cycle (GINS3 and POLE2) and the cytoskeleton (PLEK2 and FERMT1) through heightened epigenetic histone acetylation levels in the nucleus. Intriguingly, our findings highlighted elevated ANKRD22-mediated histone H3 lysine 27 acetylation (H3K27AC) signals near the METTL14 promoter, which contributes to a positive feedback loop perpetuating malignant progression in NPC.
    CONCLUSIONS: The identified METTL14-ANKRD22-SLC25A1 axis emerges as a promising therapeutic target for NPC, and also these molecules may serve as novel diagnostic biomarkers.
    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
    METTL3和METTL14传统上被设置为以化学计量1:1的比例组装m6A甲基转移酶复合物,通过m6A修饰调节mRNA命运。然而,最近的研究表明,METTL3和METTL14在不同肿瘤类型中的表达水平和预后意义不一致,挑战他们在肿瘤环境中一贯的功能参与。利用癌症基因组图谱(TCGA)数据的泛癌症分析已经确定了表达模式的明显差异,功能角色,以及METTL3和METTL14与肿瘤负荷的相关性,特别是在食管鳞状细胞癌(ESCC)中。METTL3在EC109细胞中的敲除实验在体外和体内都能显著抑制细胞增殖,而METTL14敲低对增殖的影响相对减弱,并且不会显着改变METTL3蛋白水平。mRNA测序表明,METTL3单独控制1615个基因的表达,只有776个基因与METTL14共调控。此外,免疫荧光共定位研究提示METTL3和METTL14细胞定位存在差异.高效液相色谱-质谱(HPLC-MS)分析表明,METTL3与Nop56p连接的前rRNA复合物和mRNA剪接机制独特地相关。独立于METTL14。初步的生物信息学和多组学研究表明,METTL3在调节肿瘤细胞增殖中的自主作用及其参与mRNA剪接是潜在的关键分子机制。我们的研究为更深入地了解m6A甲基转移酶复合物以及针对METTL3的靶向肿瘤疗法的开发奠定了实验和理论基础。
    METTL3 and METTL14 are traditionally posited to assemble the m6A methyltransferase complex in a stoichiometric 1:1 ratio, modulating mRNA fate via m6A modifications. Nevertheless, recent investigations reveal inconsistent expression levels and prognostic significance of METTL3 and METTL14 across various tumor types, challenging their consistent functional engagement in neoplastic contexts. A pan-cancer analysis leveraging The Cancer Genome Atlas (TCGA) data has identified pronounced disparities in the expression patterns, functional roles, and correlations with tumor burden between METTL3 and METTL14, particularly in esophageal squamous cell carcinoma (ESCC). Knockdown experiments of METTL3 in EC109 cells markedly suppress cell proliferation both in vitro and in vivo, whereas METTL14 knockdown shows a comparatively muted effect on proliferation and does not significantly alter METTL3 protein levels. mRNA sequencing indicates that METTL3 singularly governs the expression of 1615 genes, with only 776 genes co-regulated with METTL14. Additionally, immunofluorescence co-localization studies suggest discrepancies in cellular localization between METTL3 and METTL14. High-performance liquid chromatography-mass spectrometry (HPLC-MS) analyses demonstrate that METTL3 uniquely associates with the Nop56p-linked pre-rRNA complex and mRNA splicing machinery, independent of METTL14. Preliminary bioinformatics and multi-omics investigations reveal that METTL3\'s autonomous role in modulating tumor cell proliferation and its involvement in mRNA splicing are potentially pivotal molecular mechanisms. Our study lays both experimental and theoretical groundwork for a deeper understanding of the m6A methyltransferase complex and the development of targeted tumor therapies focusing on METTL3.
    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:鼻咽癌(NPC)是鼻咽粘膜的恶性上皮性肿瘤,在世界范围内发病率很高。甲基转移酶样14(METTL14)是一种主要的RNAN6-腺苷甲基转移酶,通过调节RNA功能参与肿瘤进展。本研究旨在探讨METTL14在鼻咽癌中的生物学功能和作用机制。
    方法:采用实时定量聚合酶链反应(RT-qPCR)方法检测METTL14和含胺氧化酶铜1(AOC1)的表达。METTL14,AOC1,细胞周期蛋白D1,B细胞淋巴瘤-2(Bcl-2)的蛋白水平,和N-cadherin使用蛋白质印迹测量。细胞增殖,周期进展,凋亡,迁移,使用5-乙炔基-2'-脱氧尿苷(EdU)评估侵袭,殖民地的形成,流式细胞术,伤口划伤,和transwell分析。使用RNA免疫沉淀(RIP)验证了METTL14和AOC1之间的相互作用,甲基化RNA免疫沉淀(MeRIP),和双荧光素酶报告基因测定。通过体内异种移植肿瘤模型检查了METTL14对NPC肿瘤生长的生物学作用。
    结果:METTL14和AOC1在NPC组织和细胞中高表达。此外,METTL14敲低可能阻断NPC细胞增殖,迁移,入侵,体外诱导细胞凋亡。在机制上,METTL14可能通过m6A甲基化增强AOC1mRNA的稳定性。METTL14沉默可能在体内抑制NPC肿瘤生长。
    结论:METTL14可能通过调节AOC1mRNA的稳定性促进NPC细胞的发育,这为NPC治疗提供了一个有希望的治疗靶点。
    BACKGROUND: Nasopharyngeal carcinoma (NPC) is a malignant epithelial tumor of the nasopharyngeal mucosa with a high incidence rate all over the world. Methyltransferase-like 14 (METTL14) is a major RNA N6-adenosine methyltransferase implicated in tumor progression by regulating RNA function. This study is designed to explore the biological function and mechanism of METTL14 in NPC.
    METHODS: METTL14 and Amine oxidase copper containing 1 (AOC1) expression were detected by real-time quantitative polymerase chain reaction (RT-qPCR). The protein levels of METTL14, AOC1, Cyclin D1, B-cell lymphoma-2 (Bcl-2), and N-cadherin were measured using western blot. Cell proliferation, cycle progression, apoptosis, migration, and invasion were assessed using 5-ethynyl-2\'-deoxyuridine (EdU), Colony formation, flow cytometry, wound scratch, and transwell assays. The interaction between METTL14 and AOC1 was verified using RNA immunoprecipitation (RIP), methylated RNA immunoprecipitation (MeRIP), and dual-luciferase reporter assays. The biological role of METTL14 on NPC tumor growth was examined by the xenograft tumor model in vivo.
    RESULTS: METTL14 and AOC1 were highly expressed in NPC tissues and cells. Moreover, METTL14 knockdown might block NPC cell proliferation, migration, invasion, and induce cell apoptosis in vitro. In mechanism, METTL14 might enhance the stability of AOC1 mRNA via m6A methylation. METTL14 silencing might repress NPC tumor growth in vivo.
    CONCLUSIONS: METTL14 might boosted the development of NPC cells partly by regulating the stability of AOC1 mRNA, which provided a promising therapeutic target for NPC 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
    N6-甲基腺苷(m6A)甲基化通过调节细胞增殖和转移介导癌症发展。本研究旨在确定甲基转移酶14(METTL14)是否影响胃癌(GC)细胞功能及其潜在机制。使用定量实时PCR检测METTL14和TATA-box结合蛋白相关因子10(TAF10)水平,免疫组织化学测定,和Westernblot。使用细胞计数试剂盒-8评估生物学功能,集落形成,和transwell分析。使用RNA免疫沉淀分析了METTL14和TAF10之间的相互作用,甲基化RNA免疫沉淀,和荧光素酶报告基因测定。建立异种移植肿瘤小鼠模型以评估METTL14在体内的作用。结果表明,在GC组织和细胞中,METTL14低表达,TAF10高表达。METTL14过表达抑制GC细胞活力,殖民地,迁移,和入侵。TAF10被预测并证实与METTL14呈负相关。METTL14促进TAF10的m6A甲基化并抑制TAF10稳定性。此外,TAF10抵消了METTL14调节的细胞行为。METTL14的过表达抑制肿瘤生长和组织病理学。总之,METTL14通过减弱GC细胞增殖来抑制GC进展,迁移,和入侵。机械上,METTL14促进TAF10的m6A甲基化,抑制TAF10的稳定性,从而下调TAF10水平,这些结果为GC治疗提供了新的见解。
    N6-methyladenosine (m6A) methylation mediates cancer development by regulating cell proliferation and metastasis. This study aimed to identify whether methyltransferase 14 (METTL14) affects gastric cancer (GC) cellular functions and its underlying mechanism. METTL14 and TATA-box binding protein associated factor 10 (TAF10) levels were examined using quantitative real-time PCR, immunohistochemical assay, and Western blot. Biological functions were assessed using cell counting kit-8, colony formation, and transwell assays. The interaction between METTL14 and TAF10 was analyzed using RNA immunoprecipitation, methylated RNA immunoprecipitation, and luciferase reporter assay. A xenograft tumor mouse model was established to assess the role of METTL14 in vivo. The results suggested that METTL14 was low expressed and TAF10 was highly expressed in GC tissues and cells. METTL14 overexpression inhibited GC cell viability, colony, migration, and invasion. TAF10 was predicted and confirmed to be negatively related to METTL14. METTL14 promoted m6A methylation of TAF10 and inhibited TAF10 stability. Moreover, TAF10 counteracted the cellular behaviors regulated by METTL14. Overexpression of METTL14 inhibited tumor growth and histopathology. In conclusion, METTL14 inhibits GC progression by attenuating GC cell proliferation, migration, and invasion. Mechanistically, METTL14 promoted m6A methylation of TAF10, suppressed the stability of TAF10, and thus downregulated the TAF10 levels, These results provide a new insight into GC 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
    宫颈癌(CC)是全球女性中普遍存在的恶性肿瘤。这项研究旨在研究METTL14在索拉非尼诱导的CC铁性凋亡中的作用。在CC组织中测定METTL14表达和m6A甲基化,然后分析这些因素与临床特征的相关性。随后,METTL14在CC细胞系中被敲低,以及对细胞增殖的影响,使用CCK-8,显微镜评估线粒体形态和铁死亡,以及与铁死亡相关的标记,分别。使用qRT-PCR和荧光素酶报告基因测定验证METTL14和FTH1之间的调控关系。通过在索拉非尼处理后将细胞与靶向METTL14和FTH1的过表达载体或shRNA共转染,在体外和体内进一步研究了这种相互作用的功能意义。在CC组织中METTL14表达和m6A甲基化显著降低,较低的METTL14表达水平与CC患者预后较差相关。值得注意的是,在索拉非尼诱导的铁凋亡过程中,METTL14表达增加,和METTL14敲低减弱了索拉非尼在CC细胞中诱导的铁反应。FTH1被确定为METTL14的直接靶标,METTL14过表达导致FTH1mRNA的m6A甲基化增加,导致CC中FTH1的稳定性和表达降低。此外,FTH1过表达或用LY294002处理部分抵消了通过METTL14促进索拉非尼诱导的铁凋亡。体内异种移植实验表明,抑制METTL14可降低索拉非尼的抗癌作用,而抑制FTH1显著增强索拉非尼诱导的铁细胞凋亡并提高其抗癌功效。METTL14通过m6A甲基化降低FTH1mRNA的稳定性,从而增强索拉非尼诱导的铁细胞凋亡,这有助于通过PI3K/Akt信号通路抑制CC进展。
    Cervical cancer (CC) is a prevalent malignancy among women worldwide. This study was designed to investigate the role of METTL14 in sorafenib-induced ferroptosis in CC. METTL14 expression and m6A methylation were determined in CC tissues, followed by analyzes correlating these factors with clinical features. Subsequently, METTL14 was knocked down in CC cell lines, and the effects on cell proliferation, mitochondrial morphology and ferroptosis were assessed using CCK-8, microscopy, and markers associated with ferroptosis, respectively. The regulatory relationship between METTL14 and FTH1 was verified using qRT-PCR and luciferase reporter assays. The functional significance of this interaction was further investigated both in vitro and in vivo by co-transfecting cells with overexpression vectors or shRNAs targeting METTL14 and FTH1 after sorafenib treatment. METTL14 expression and m6A methylation were significantly reduced in CC tissues, and lower METTL14 expression levels were associated with a poorer CC patients\' prognosis. Notably, METTL14 expression increased during sorafenib-induced ferroptosis, and METTL14 knockdown attenuated the ferroptotic response induced by sorafenib in CC cells. FTH1 was identified as a direct target of METTL14, with METTL14 overexpression leading to increased m6A methylation of FTH1 mRNA, resulting in reduced stability and expression of FTH1 in CC. Furthermore, FTH1 overexpression or treatment with LY294002 partially counteracted the promotion of sorafenib-induced ferroptosis by METTL14. In vivo xenograft experiments demonstrated that inhibiting METTL14 reduced the anticancer effects of sorafenib, whereas suppression of FTH1 significantly enhanced sorafenib-induced ferroptosis and increased its anticancer efficacy. METTL14 reduces FTH1 mRNA stability through m6A methylation, thereby enhancing sorafenib-induced ferroptosis, which contributes to suppressing CC progression via the PI3K/Akt signaling pathway.
    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:N6-甲基腺苷(m6A)修饰在肺癌中起重要作用。然而,甲基转移酶样14(METTL14),作为M6A复合物的主要成分,报道较少涉及肺癌的免疫微环境。本研究旨在分析肺癌中METTL14与免疫检查点抑制剂程序性死亡受体1(PD-1)的关系。
    方法:CCK-8,集落形成,transwell,伤口愈合,和流式细胞术检测来探讨METTL14在肺癌进展中的作用。此外,用sh-METTL14和抗PD-1抗体治疗同基因模型小鼠,观察METTL14对免疫治疗的影响。流式细胞术和免疫组织化学(IHC)染色检测CD8的表达。进行RIP和MeRIP以评估METTL14和HSD17B6之间的关系。将LLC细胞和活化的小鼠PBMC体外共培养以模拟肿瘤微环境中的免疫细胞浸润。ELISA法检测IFN-γ和TNF-α水平。
    结果:在线数据库GEPIA显示,METTL14高表达表明肺癌患者预后不良。体外实验表明METTL14敲低抑制了肺癌的进展。体内测定显示METTL14敲低抑制肿瘤生长并增强对PD-1免疫疗法的反应。此外,METTL14敲低增强CD8+T细胞活化和浸润。更重要的是,METTL14敲低通过减少其m6A甲基化增加HSD17B6mRNA的稳定性。此外,HSD17B6过表达促进CD8+T细胞的活化。
    结论:METTL14的破坏有助于CD8+T细胞活化和通过HSD17B6的m6A修饰对PD-1的免疫治疗反应,从而抑制肺癌进展。
    BACKGROUND: N6-methyladenosine (m6A) modification plays an important role in lung cancer. However, methyltransferase-like 14 (METTL14), which serves as the main component of the m6A complex, has been less reported to be involved in the immune microenvironment of lung cancer. This study aimed to analyze the relationship between METTL14 and the immune checkpoint inhibitor programmed death receptor 1 (PD-1) in lung cancer.
    METHODS: CCK-8, colony formation, transwell, wound healing, and flow cytometry assays were performed to explore the role of METTL14 in lung cancer progression in vitro. Furthermore, syngeneic model mice were treated with sh-METTL14 andan anti-PD-1 antibody to observe the effect of METTL14 on immunotherapy. Flow cytometry and immunohistochemical (IHC) staining were used to detect CD8 expression. RIP and MeRIP were performed to assess the relationship between METTL14 and HSD17B6. LLC cells and activated mouse PBMCs were cocultured in vitro to mimic immune cell infiltration in the tumor microenvironment. ELISA was used to detect IFN-γ and TNF-α levels.
    RESULTS: The online database GEPIA showed that high METTL14 expression indicated a poor prognosis in patients with lung cancer. In vitro assays suggested that METTL14 knockdown suppressed lung cancer progression. In vivo assays revealed that METTL14 knockdown inhibited tumor growth and enhanced the response to PD-1 immunotherapy. Furthermore, METTL14 knockdown enhanced CD8+T-cell activation and infiltration. More importantly, METTL14 knockdown increased the stability of HSD17B6 mRNA by reducing its m6A methylation. In addition, HSD17B6 overexpression promoted the activation of CD8+ T cells.
    CONCLUSIONS: The disruption of METTL14 contributed to CD8+T-cell activation and the immunotherapy response to PD-1 via m6A modification of HSD17B6, thereby suppressing lung cancer progression.
    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:缺氧/复氧(H/R)诱导心肌细胞铁性凋亡,心肌缺血/再灌注损伤中的核心重塑事件。甲基转移酶样14(METTL14)是N6-甲基腺苷(m6A)修饰的作者。进行这项研究以破译METTL14在H/R诱导的心肌细胞铁性凋亡中的作用。
    方法:培养小鼠心肌细胞HL-1,并进行H/R处理。H/R处理后的铁死亡程度通过细胞计数试剂盒-8测定来评价,测定试剂盒(ROS/GSH/Fe2+),和蛋白质印迹(GPX4/ACSL4)。METTL14,pri-miR-146a-5p的细胞内表达,miR-146a-5p,或衔接蛋白磷酸酪氨酸与PH结构域和亮氨酸拉链1(APPL1)的相互作用通过实时定量聚合酶链反应或蛋白质印迹进行检查,通过m6A定量分析和RNA免疫沉淀来确定与DiGeorge临界区8(DGCR8)结合的pri-miR-146a-5p和m6A修饰的pri-miR-146a-5p的总m6A水平和表达。miR-146a-5p与APPL1的结合通过双荧光素酶测定来证明。
    结果:H/R处理诱导心肌细胞铁性凋亡(ROS增加,Fe2+,和ACSL4并降低GSH和GPX4)并上调METTL14表达。METTL14敲低减弱H/R诱导的心肌细胞铁凋亡。METTL14通过增加pri-miR-146a-5p上的m6A修饰来诱导DGCR8对pri-miR-146a-5p的识别,这促进pri-miR-146a-5p转化为miR-146a-5p并进一步抑制APPL1转录。miR-146a-5p上调或APPL1下调限制了METTL14下调对H/R诱导的心肌细胞铁性凋亡的抑制作用。
    结论:METTL14通过DGCR8识别和加工pri-miR-146a-5p促进miR-146a-5p表达,抑制APPL1转录并触发H/R诱导的心肌细胞铁凋亡。
    BACKGROUND: Hypoxia/reoxygenation (H/R) induces cardiomyocyte ferroptosis, a core remodeling event in myocardial ischemia/reperfusion injury. Methyltransferase-like 14 (METTL14) emerges as a writer of N6-methyladenosine (m6A) modification. This study was conducted to decipher the role of METTL14 in H/R-induced cardiomyocyte ferroptosis.
    METHODS: Mouse cardiomyocytes HL-1 were cultured and underwent H/R treatment. The degree of ferroptosis after H/R treatment was appraised by the cell counting kit-8 assay, assay kits (ROS/GSH/Fe2+), and Western blotting (GPX4/ACSL4). The intracellular expressions of METTL14, pri-miR-146a-5p, miR-146a-5p, or adaptor protein phosphotyrosine interacting with PH domain and leucine zipper 1 (APPL1) were examined by real-time quantitative polymerase chain reaction or Western blotting, with m6A quantification analysis and RNA immunoprecipitation to determine the total m6A level and the expression of pri-miR-146a-5p bound to DiGeorge critical region 8 (DGCR8) and m6A-modified pri-miR-146a-5p. The binding of miR-146a-5p to APPL1 was testified by the dual-luciferase assay.
    RESULTS: H/R treatment induced cardiomyocyte ferroptosis (increased ROS, Fe2+, and ACSL4 and decreased GSH and GPX4) and upregulated METTL14 expression. METTL14 knockdown attenuated H/R-induced cardiomyocyte ferroptosis. METTL14 induced the recognition of pri-miR-146a-5p by DGCR8 by increasing m6A modification on pri-miR-146a-5p, which promoted the conversion of pri-miR-146a-5p into miR-146a-5p and further repressed APPL1 transcription. miR-146a-5p upregulation or APPL1 downregulation limited the inhibitory effect of METTL14 downregulation on H/R-induced cardiomyocyte ferroptosis.
    CONCLUSIONS: METTL14 promoted miR-146a-5p expression through the recognition and processing of pri-miR-146a-5p by DGCR8, which repressed APPL1 transcription and triggered H/R-induced cardiomyocyte ferroptosis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号