N6-methyladenosine methylation

  • 文章类型: Journal Article
    背景:在肿瘤微环境中,生存压力普遍存在,肿瘤进展的有效驱动因素,血管生成,和治疗抗性。N6-甲基腺苷(m6A)甲基化已被认为是调节mRNA代谢各个方面的关键转录后机制。了解生存压力和m6A修饰之间的复杂相互作用,为肝细胞癌(HCC)进展的分子机制提供了新的见解,并突出了在HCC诊断和治疗中靶向生存压力-m6A轴的潜力。
    方法:在PubMed,MEDLINE,和WebofScience提供截至2024年4月发表的相关文章。用于搜索的关键词包括肝细胞癌,细胞生存,生存压力,N6-甲基腺苷,肿瘤微环境,应激反应,和缺氧。
    结果:这篇综述探讨了肝癌中生存压力和m6ARNA甲基化的多方面作用。强调生存压力如何调节m6A景观,m6A修饰对生存压力响应基因表达的影响,以及随之而来的对肝癌细胞存活的影响,扩散,转移,和对治疗的抵抗力。此外,我们探索了针对这种串扰的治疗潜力,提出利用对生存压力和m6ARNA甲基化机制的理解来开发新的策略,和更有效的治疗肝癌。
    结论:生存压力和m6ARNA甲基化之间的相互作用是影响HCC发病机制和进展的复杂调控网络。
    BACKGROUND: Within the tumor microenvironment, survival pressures are prevalent with potent drivers of tumor progression, angiogenesis, and therapeutic resistance. N6-methyladenosine (m6A) methylation has been recognized as a critical post-transcriptional mechanism regulating various aspects of mRNA metabolism. Understanding the intricate interplay between survival pressures and m6A modification provides new insights into the molecular mechanisms underlying hepatocellular carcinoma (HCC) progression and highlights the potential for targeting the survival pressures-m6A axis in HCC diagnosis and treatment.
    METHODS: A literature search was conducted in PubMed, MEDLINE, and Web of Science for relevant articles published up to April 2024. The keywords used for the search included hepatocellular carcinoma, cellular survival, survival pressure, N6-methyladenosine, tumor microenvironment, stress response, and hypoxia.
    RESULTS: This review delves into the multifaceted roles of survival pressures and m6A RNA methylation in HCC, highlighting how survival pressures modulate the m6A landscape, the impact of m6A modification on survival pressure-responsive gene expression, and the consequent effects on HCC cell survival, proliferation, metastasis, and resistance to treatment. Furthermore, we explored the therapeutic potential of targeting this crosstalk, proposing strategies that leverage the understanding of survival pressures and m6A RNA methylation mechanisms to develop novel, and more effective treatments for HCC.
    CONCLUSIONS: The interplay between survival pressures and m6A RNA methylation emerges as a complex regulatory network that influences HCC pathogenesis and 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
    非酒精性脂肪性肝病(NAFLD),包括其更严重的表现非酒精性脂肪性肝炎(NASH),是全球公共卫生挑战。这里,我们探讨去泛素化酶RPN11在NAFLD和NASH中的作用。肝细胞特异性RPN11敲除小鼠免受饮食诱导的肝脏脂肪变性,胰岛素抵抗,还有脂肪性肝炎.机械上,RPN11去泛素化并稳定METTL3,以增强酰基辅酶A(CoA)合成酶短链家族成员3(ACSS3)的m6A修饰和表达,产生丙酰辅酶A通过组蛋白丙酰化上调脂质代谢基因。RPN11-METTL3-ACSS3-组蛋白丙酸化途径在NAFLD患者的肝脏中被激活。Capzimin改善NAFLD对RPN11的药理抑制作用,NASH,和相关的代谢紊乱的小鼠和降低的脂质含量培养的人肝细胞在2D和3D。这些结果表明,RPN11是NAFLD/NASH的新型调节因子,抑制RPN11具有治疗潜力。
    Nonalcoholic fatty liver disease (NAFLD), including its more severe manifestation nonalcoholic steatohepatitis (NASH), is a global public health challenge. Here, we explore the role of deubiquitinating enzyme RPN11 in NAFLD and NASH. Hepatocyte-specific RPN11 knockout mice are protected from diet-induced liver steatosis, insulin resistance, and steatohepatitis. Mechanistically, RPN11 deubiquitinates and stabilizes METTL3 to enhance the m6A modification and expression of acyl-coenzyme A (CoA) synthetase short-chain family member 3 (ACSS3), which generates propionyl-CoA to upregulate lipid metabolism genes via histone propionylation. The RPN11-METTL3-ACSS3-histone propionylation pathway is activated in the livers of patients with NAFLD. Pharmacological inhibition of RPN11 by Capzimin ameliorated NAFLD, NASH, and related metabolic disorders in mice and reduced lipid contents in human hepatocytes cultured in 2D and 3D. These results demonstrate that RPN11 is a novel regulator of NAFLD/NASH and that suppressing RPN11 has therapeutic potential for the 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
    在RNA的所有化学修饰中,N6-甲基腺苷(m6A)修饰是最普遍和充分表征的RNA修饰,其在功能上涉及广泛的生物过程。m6A修饰发生在乙型肝炎病毒(HBV)RNA中,这种修饰以多种方式调节HBV生命周期。因此,了解HBVRNAm6A修饰的潜在机制对于理解HBV感染过程和相关发病机制至关重要。这里,我们描述了目前在病毒感染期间m6A-甲基化RNA的检测和表征中使用的方法。
    Of all the chemical modifications of RNAs, the N6-methyladenosine (m6A) modification is the most prevalent and well-characterized RNA modification that is functionally implicated in a wide range of biological processes. The m6A modification occurs in hepatitis B virus (HBV) RNAs and this modification regulates the HBV life cycle in several ways. Thus, understanding the mechanisms underlying m6A modification of HBV RNAs is crucial in understanding HBV infectious process and associated pathogenesis. Here, we describe the currently utilized method in the detection and characterization of m6A-methylated RNAs during viral infection.
    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:EB病毒(EBV)原发感染中存在N6-甲基腺苷(m6A)甲基化修饰,延迟,和裂解剂的再激活。它还修饰EBV潜伏基因和裂解基因。EBV相关胃癌(EBVaGC)是一种独特的GC分子亚型。我们假设EBV和m6A甲基化调节因子在EBVaGC中相互作用,以将其与其他类型的GC区分开。
    目的:研究m6A甲基化调节因子在EBVaGC中的作用机制,以确定与其他类型GC的区别因素。
    方法:首先,癌症基因图谱和基因表达综合数据库用于分析EBVaGC和EBV阴性GC(EBVnGC)之间m6A甲基化调节因子的表达模式。第二,我们确定了基因本体论(GO)和京都基因和基因组百科全书(KEGG)m6A相关差异表达基因的功能富集。我们定量了肿瘤微环境(TME)中免疫细胞和炎症因子的相对丰度。最后,细胞计数试剂盒-8细胞增殖试验,Transwell测试,并采用流式细胞术验证胰岛素样生长因子结合蛋白1(IGFBP1)在EBVaGC细胞系中的作用。
    结果:m6A甲基化调节因子参与了EBVaGC的发生和发展。与EBVnGC相比,m6A甲基化调节因子Wilms肿瘤1相关蛋白的表达水平,RNA结合基序蛋白15B,CBL原癌基因样1,富含亮氨酸的五肽重复序列,异质核核糖核蛋白A2B1、IGFBP1和胰岛素样生长因子2结合蛋白1在EBVaGC中显著下调(P<0.05)。IGFBP1表达水平较低的EBVaGC患者的总生存率明显较高(P=0.046)。GO和KEGG功能富集分析表明,EBVaGC中免疫途径被显著激活并富含免疫细胞浸润。与EBVnGC相比,活化的CD4+T细胞的浸润,激活的CD8+T细胞,单核细胞,激活的树突状细胞,和浆细胞样树突状细胞在EBVaGC中显著上调(P<0.001)。在EBVaGC,促炎因子白细胞介素(IL)-17、IL-21、干扰素-γ和免疫抑制因子IL-10的表达水平显著升高(P<0.05)。体外实验表明,IGFBP1在EBVaGC细胞系(SNU719)中的表达水平显着低于EBVnGC细胞系(AGS)(P<0.05)。IGFBP1过表达显著减弱SNU719细胞增殖和迁移,促进细胞凋亡。干扰IGFBP1显著促进AGS细胞的增殖和迁移,降低细胞凋亡水平。
    结论:m6A调节剂可以重塑EBVaGC的TME,它被归类为一种免疫发炎的表型,被称为“热”肿瘤。在这些监管机构中,我们证明IGFBP1影响增殖,迁移,和凋亡。
    BACKGROUND: N6-methyladenosine (m6A) methylation modification exists in Epstein-Barr virus (EBV) primary infection, latency, and lytic reactivation. It also modifies EBV latent genes and lytic genes. EBV-associated gastric cancer (EBVaGC) is a distinctive molecular subtype of GC. We hypothesized EBV and m6A methylation regulators interact with each other in EBVaGC to differentiate it from other types of GC.
    OBJECTIVE: To investigate the mechanisms of m6A methylation regulators in EBVaGC to determine the differentiating factors from other types of GC.
    METHODS: First, The Cancer Gene Atlas and Gene Expression Omnibus databases were used to analyze the expression pattern of m6A methylation regulators between EBVaGC and EBV-negative GC (EBVnGC). Second, we identified Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional enrichment of m6A-related differentially expressed genes. We quantified the relative abundance of immune cells and inflammatory factors in the tumor microenvironment (TME). Finally, cell counting kit-8 cell proliferation test, transwell test, and flow cytometry were used to verify the effect of insulin-like growth factor binding protein 1 (IGFBP1) in EBVaGC cell lines.
    RESULTS: m6A methylation regulators were involved in the occurrence and development of EBVaGC. Compared with EBVnGC, the expression levels of m6A methylation regulators Wilms tumor 1-associated protein, RNA binding motif protein 15B, CBL proto-oncogene like 1, leucine rich pentatricopeptide repeat containing, heterogeneous nuclear ribonucleoprotein A2B1, IGFBP1, and insulin-like growth factor 2 binding protein 1 were significantly downregulated in EBVaGC (P < 0.05). The overall survival rate of EBVaGC patients with a lower expression level of IGFBP1 was significantly higher (P = 0.046). GO and KEGG functional enrichment analyses showed that the immunity pathways were significantly activated and rich in immune cell infiltration in EBVaGC. Compared with EBVnGC, the infiltration of activated CD4+ T cells, activated CD8+ T cells, monocytes, activated dendritic cells, and plasmacytoid dendritic cells were significantly upregulated in EBVaGC (P < 0.001). In EBVaGC, the expression level of proinflammatory factors interleukin (IL)-17, IL-21, and interferon-γ and immunosuppressive factor IL-10 were significantly increased (P < 0.05). In vitro experiments demonstrated that the expression level of IGFBP1 was significantly lower in an EBVaGC cell line (SNU719) than in an EBVnGC cell line (AGS) (P < 0.05). IGFBP1 overexpression significantly attenuated proliferation and migration and promoted the apoptosis levels in SNU719. Interfering IGFBP1 significantly promoted proliferation and migration and attenuated the apoptosis levels in AGS.
    CONCLUSIONS: m6A regulators could remodel the TME of EBVaGC, which is classified as an immune-inflamed phenotype and referred to as a \"hot\" tumor. Among these regulators, we demonstrated that IGFBP1 affected proliferation, migration, and 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
    长期缺氧诱导心肌细胞凋亡是心肌缺血(MI)损伤的重要机制。有趣的是,长链非编码RNA心肌梗死相关转录物(LncMIAT)参与MI损伤的调控;LncMIAT影响缺氧诱导的心肌细胞凋亡进展的潜在机制尚不清楚.在本研究中,研究发现低氧可通过增加体外LncMIAT的表达促进心肌细胞凋亡。生物学研究和双荧光素酶基因报告基因测定进一步揭示LncMIAT能够与miR-708-5p结合以上调心肌细胞的p53介导的细胞死亡。LncMIAT的沉默或miR-708-5p的过表达导致p53介导的心肌细胞凋亡的显著减少。甲基化RNA免疫沉淀(MeRIP)-qPCR结果表明,缺氧通过AKLBH5-N6-甲基腺苷(m6A)甲基化对LncMIAT产生影响,因此缺氧通过m6A甲基化介导的LncMIAT/miR-708-5p/p53轴触发HL-1心肌细胞凋亡。沉默AKLBH5可显着减轻m6A甲基化介导的LncMIAT上调和p53介导的心肌细胞凋亡,同时促进miR-708-5p表达。一起来看,本研究强调LncMIAT在缺氧诱导的心肌细胞凋亡过程中可能是一个关键的生物学靶点.此外,研究表明,缺氧可通过调节m6A甲基化介导的LncMIAT/miR-708-5p/p53信号轴促进心肌细胞凋亡。
    Long-time hypoxia induced cardiomyocyte apoptosis is an important mechanism of myocardial ischemia (MI) injury. Interestingly, long noncoding RNA myocardial infarction-associated transcript (LncMIAT) has been involved in the regulation of MI injury; however, the underlying mechanism by which LncMIAT affects the progression of hypoxia-induced cardiomyocyte apoptosis remains unclear. In the present study, hypoxia was found to promote cardiomyocyte apoptosis through an increased expression of LncMIAT in vitro. Biological investigations and dual-luciferase gene reporter assay further revealed that LncMIAT was able to bind with miR-708-5p to upregulate the p53-mediated cell death of the cardiomyocytes. Silencing of LncMIAT or overexpression of miR-708-5p led to a significant reduction in p53-mediated cardiomyocyte apoptosis. The methylated RNA immunoprecipitation (MeRIP)-qPCR results showed that hypoxia exerted its effects on LncMIAT through AKLBH5-N6-methyladenosine (m6A) methylation and therefore hypoxia was shown to trigger HL-1 cardiomyocyte apoptosis via the m6A methylation-mediated LncMIAT/miR-708-5p/p53 axis. Silencing of AKLBH5 significantly alleviated the m6A methylation-mediated LncMIAT upregulation and p53-mediated cardiomyocyte apoptosis, while promoted miR-708-5p expression. Taken together, the present study highlighted that LncMIAT could act as a key biological target during hypoxia-induced cardiomyocyte apoptosis. In addition, it was shown that hypoxia could promote cardiomyocyte apoptosis through regulation of the m6A methylation-mediated LncMIAT/miR-708-5p/p53 signaling axis.
    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
    哮喘是一种以气道炎症为特征的慢性呼吸道疾病,过多的粘液产生,和气道重塑。哮喘的预防和治疗是临床研究中迫切需要解决的问题。近年来,N6-甲基腺苷甲基化(m6A)已成为涉及多种疾病的有希望的调节方法。ALKBH5(alkB同源物5)是在疾病病理学中广泛研究的去甲基酶。这项工作旨在探讨ALKBH5调节哮喘的潜在调节机制。我们建立了白细胞介素-13(IL-13)刺激的细胞模型来模拟哮喘的体外炎症环境。使用siRNA进行支气管上皮细胞中的ALKBH5敲低,并通过定量PCR(qPCR)分析敲低功效。通过细胞计数试剂盒8(CCK-8)和集落形成测定来测量细胞活力和增殖。通过测量总铁来评估铁沉积,Fe2+,脂质活性氧(ROS),丙二醛(MDA),和超氧化物歧化酶(SOD)水平。通过MeRIP测定检测N6-甲基腺苷甲基化(m6A)修饰的富集。在IL-13诱导模型中,ALKBH5的敲除显着提高了支气管上皮细胞的存活和集落形成能力。总铁的水平,Fe2+,脂质ROS,MDA显著升高,IL-13诱导的支气管上皮细胞中SOD水平降低,ALKBH5的消耗逆转了这些影响。敲除ALKBH5提高了m6A修饰的富集和谷胱甘肽过氧化物酶4(GPX4)的表达。敲除GPX4消除了siALKBH5的促增殖和抗铁凋亡作用。敲除ALKBH5可促进支气管上皮细胞的增殖,减轻细胞铁凋亡。
    Asthma is a chronic respiratory disease that is characterized by airway inflammation, excessive mucus production, and airway remodeling. Prevention and treatment for asthma is an urgent issue in clinical studies. In recent years, N6-methyladenosine methylation (m6A) has emerged as a promising regulatory approach involved in multiple diseases. ALKBH5 (alkB homolog 5) is a demethylase widely studied in disease pathologies. This work aimed to explore the regulatory mechanisms underlying the ALKBH5-regulated asthma. We established an interleukin-13 (IL-13)-stimulated cell model to mimic the in vitro inflammatory environment of asthma. ALKBH5 knockdown in bronchial epithelial cells was performed using siRNAs, and the knockdown efficacy was analyzed by quantitative PCR (qPCR). Cell viability and proliferation were measured by cell counting kit 8 (CCK-8) and colony formation assay. The ferroptosis was assessed by measuring the total iron, Fe2+, lipid reactive oxygen species (ROS), malondialdehyde (MDA), and superoxide dismutase (SOD) levels. The enrichment of N6-methyladenosine methylation (m6A) modification was detected by the MeRIP assay. Knockdown of ALKBH5 significantly elevated the survival and colony formation ability of bronchial epithelial cells in the IL-13 induction model. The levels of total iron, Fe2+, lipid ROS, and MDA were remarkedly elevated, and the SOD level was reduced in IL-13-induced bronchial epithelial cells, and depletion of ALKBH5 reversed these effects. Knockdown of ALKBH5 elevated the enrichment of m6A modification and expression of glutathione peroxidase 4 (GPX4). Knockdown of GPX4 abolished the pro-proliferation and anti-ferroptosis effects of siALKBH5. Knockdown of ALKBH5 improved the proliferation of bronchial epithelial cells and alleviated cell ferroptosis.
    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:N6-甲基腺苷(m6A)是最常见和最丰富的mRNA修饰,在生物过程和肿瘤的发展中起着至关重要的作用。然而,m6A甲基化在皮肤皮肤黑色素瘤(SKCM)中的作用尚不清楚。本研究分析了m6A相关功能基因在SKCM中的表达,旨在探讨去甲基酶ALKBH5介导的m6A修饰及其在人类SKCM中的潜在机制。方法:基于公共数据库,对SKCM中m6A相关基因表达情况进行了描述.MeRIP-Seq和RNA-Seq用于识别ALKBH5的下游靶标。进行了体内和体外功能表型和挽救功能实验,以探索SKCM中ALKBH5-m6A-ABCA1轴的机制。结果:我们发现SKCM中ALKBH5上调,与预后不良有关。ALKBH5可以促进黑色素瘤细胞增殖,菌落形成,迁移,和体外侵袭和抑制自噬,促进体内肿瘤生长和转移。我们鉴定了ABCA1,一种协助胆固醇流出的膜蛋白,作为ALKBH5介导的m6A去甲基化的下游靶标。最后,我们的数据表明,ALKBH5通过介导ABCA1的下调,以m6A依赖性方式降低ABCA1mRNA的稳定性,从而促进SKCM.结论:我们的发现显示了关键脱甲基酶ALKBH5介导的m6A修饰在SKCM进展中的功能价值。提示ALKBH5-m6A-ABCA1轴作为SKCM的潜在治疗靶点。
    Background: N6-methyladenosine (m6A) is the most common and abundant mRNA modification, playing an essential role in biological processes and tumor development. However, the role of m6A methylation in skin cutaneous melanoma (SKCM) is not yet clear. This study analyzed the expression of m6A-related functional genes in SKCM and aimed to explore the key demethylase ALKBH5 mediated m6A modification and its potential mechanism in human SKCM. Methods: Based on public databases, the m6A-related gene expression landscape in SKCM was portrayed. MeRIP-Seq and RNA-Seq were used to recognize the downstream target of ALKBH5. In vivo and in vitro functional phenotype and rescue functional experiments were performed to explore the mechanism of the ALKBH5-m6A-ABCA1 axis in SKCM. Results: We found ALKBH5 upregulated in SKCM, associated with poor prognosis. ALKBH5 can promote melanoma cell proliferation, colony formation, migration, and invasion and inhibit autophagy in vitro, facilitating tumor growth and metastasis in vivo. We identified ABCA1, a membrane protein that assists cholesterol efflux, as a downstream target of ALKBH5-mediated m6A demethylation. Finally, our data demonstrated that ALKBH5 promoted SKCM via mediating ABCA1 downregulation by reducing ABCA1 mRNA stability in an m6A-dependent manner. Conclusion: Our findings exhibited the functional value of the key demethylase ALKBH5 mediated m6A modification in the progression of SKCM, suggesting the ALKBH5-m6A-ABCA1 axis as a potential therapeutic target in SKCM.
    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:中枢神经系统缺血再灌注损伤通常会引起严重的并发症。内源性神经干细胞(NSC)的激活被认为是神经修复的有希望的治疗策略。然而,NSC激活的具体生物学过程和分子机制尚不清楚,N6-甲基腺苷(m6A)甲基化修饰在此过程中的作用尚未被探索。
    方法:对NSC进行缺氧/复氧(H/R)以模拟体内缺血再灌注。m6ARNA甲基化定量试剂盒用于测量m6A总RNA甲基化水平。定量实时PCR用于检测甲基转移酶和去甲基酶mRNA表达水平。对对照组和H/R组的NSCs进行甲基化RNA免疫沉淀测序(MeRIP-seq)和RNA测序(RNA-seq),并对测序结果进行生物信息学分析。最后,通过伤口愈合试验鉴定了神经干细胞的迁移能力,并且使用细胞计数试剂盒-8、EdU测定和细胞球化测定来评估NSCs的增殖能力。
    结果:在H/R处理后,在NSC中m6A修饰水平和Mettl14mRNA表达的总体增加。首次描述了H/R后NSC中m6A甲基化和mRNA的表达谱。通过对MeRIP-seq和RNA-seq结果的联合分析,我们验证了H/R后NSCs的增殖,受m6A甲基化修饰调控。确定了七个hub基因在调节过程中起关键作用。Mettl14的敲除显著抑制了NSCs的增殖。此外,对MeRIP-seq结果的单独分析表明,m6A甲基化调节细胞迁移和分化的方式不是影响mRNA表达.随后的实验证实了NSC的迁移能力被Mettl14的敲低所抑制。
    结论:MSCs在H/R后的生物学行为与m6A基因甲基化密切相关。Mettl14被证实参与细胞增殖和迁移。
    BACKGROUND: Ischemia-reperfusion injury to the central nervous system often causes severe complications. The activation of endogenous neural stem cells (NSCs) is considered a promising therapeutic strategy for nerve repair. However, the specific biological processes and molecular mechanisms of NSC activation remain unclear, and the role of N6-methyladenosine (m6A) methylation modification in this process has not been explored.
    METHODS: NSCs were subjected to hypoxia/reoxygenation (H/R) to simulate ischemia-reperfusion in vivo. m6A RNA methylation quantitative kit was used to measure the total RNA m6A methylation level. Quantitative real-time PCR was used to detect methyltransferase and demethylase mRNA expression levels. Methylated RNA immunoprecipitation sequencing (MeRIP-seq) and RNA sequencing (RNA-seq) were conducted for NSCs in control and H/R groups, and the sequencing results were analyzed using bioinformatics. Finally, the migration ability of NSCs was identified by wound healing assays, and the proliferative capacity of NSCs was assessed using the cell counting kit-8, EdU assays and cell spheroidization assays.
    RESULTS: Overall of m6A modification level and Mettl14 mRNA expression increased in NSCs after H/R treatment. The m6A methylation and expression profiles of mRNAs in NSCs after H/R are described for the first time. Through the joint analysis of MeRIP-seq and RNA-seq results, we verified the proliferation of NSCs after H/R, which was regulated by m6A methylation modification. Seven hub genes were identified to play key roles in the regulatory process. Knockdown of Mettl14 significantly inhibited the proliferation of NSCs. In addition, separate analysis of the MeRIP-seq results suggested that m6A methylation regulates cell migration and differentiation in ways other than affecting mRNA expression. Subsequent experiments confirmed the migration ability of NSCs was suppressed by knockdown of Mettl14.
    CONCLUSIONS: The biological behaviors of NSCs after H/R are closely related to m6A methylation of mRNAs, and Mettl14 was confirmed to be involved in cell proliferation and migration.
    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
    糖尿病肾病(DKD)是糖尿病的主要微血管并发症,与糖尿病相关的高血糖记忆携带疾病发生的风险,甚至在血糖损伤终止后。高血糖记忆的存在支持了涉及n6-甲基腺苷(m6A)修饰的表观遗传机制的概念。多项研究表明,m6A在DKD的发病机制中起关键作用。本文综述了m6ARNA修饰在DKD进展中的作用和机制。包括M6A修饰在病理过程中的调节作用,比如炎症,氧化应激,纤维化,和非编码(nc)RNA。这揭示了m6A在DKD的发生和发展中的重要性,提示m6A可能在高血糖记忆现象中起作用。这篇综述还讨论了一些灰色地带,如M6A修饰的多种酶,影响DKD的发展并提出对策。总之,这篇综述从m6A修饰的角度增强了我们对DKD的理解,并为未来的治疗策略提供了新的靶点.此外,这里讨论的见解支持DKD中存在高血糖记忆效应,这可能对新型治疗方法的发展产生深远的影响。我们假设m6ARNA修饰,作为调节DKD发展的关键因素,为DKD的深入探索提供了新的视角,为DKD患者的临床管理提供了新的选择。
    Diabetic kidney disease (DKD) is a major microvascular complication of diabetes, and hyperglycemic memory associated with diabetes carries the risk of disease occurrence, even after the termination of blood glucose injury. The existence of hyperglycemic memory supports the concept of an epigenetic mechanism involving n6-methyladenosine (m6A) modification. Several studies have shown that m6A plays a key role in the pathogenesis of DKD. This review addresses the role and mechanism of m6A RNA modification in the progression of DKD, including the regulatory role of m6A modification in pathological processes, such as inflammation, oxidative stress, fibrosis, and non-coding (nc) RNA. This reveals the importance of m6A in the occurrence and development of DKD, suggesting that m6A may play a role in hyperglycemic memory phenomenon. This review also discusses how some gray areas, such as m6A modified multiple enzymes, interact to affect the development of DKD and provides countermeasures. In conclusion, this review enhances our understanding of DKD from the perspective of m6A modifications and provides new targets for future therapeutic strategies. In addition, the insights discussed here support the existence of hyperglycemic memory effects in DKD, which may have far-reaching implications for the development of novel treatments. We hypothesize that m6A RNA modification, as a key factor regulating the development of DKD, provides a new perspective for the in-depth exploration of DKD and provides a novel option for the clinical management of patients with DKD.
    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:低级别神经胶质瘤(LGG)的生物学行为受到N6-甲基腺苷(m6A)甲基化的显着影响,一种重要的表观遗传改变。因此,利用调节m6A甲基化的基因建立LGG的预后模型至关重要.
    方法:使用TCGA和GTEx数据库。我们检查了LGG和正常组织中的m6A调节剂水平,并研究了PD-L1和PD-1的表达,免疫评分,免疫细胞浸润,不同LGG簇中的肿瘤免疫微环境(TIME)和潜在的潜在机制。我们还进行了免疫组织化学和RT-qPCR以鉴定必需的m6A调节因子。
    结果:结果显示,LGG组织中m6A调控元件表达明显增加,并与TMIE显著相关。观察到LGG组织和高风险队列中PD-L1和PD-1水平的大幅增加。PD-L1表达与FTO呈正相关,ZCCHC4和HNRNPD,而PD-1表达与FTO呈负相关,ZC3H7B,和HNRNPD。使用m6ARNA甲基化调节因子创建的预后特征显示与LGG患者的总体生存率密切相关。FTO和ZCCHC4被临床样本证实为独立的预后标志物。此外,结果显示两组患者的时间特征不同,指示与LGG相关的信号通路被破坏。
    结论:我们的结果表明,m6A调节因子在调节PD-L1/PD-1表达和免疫细胞浸润中起着至关重要的作用,从而对LGG的时间产生了相当大的影响。因此,m6A调节因子对LGG的预后具有准确的预测价值。
    BACKGROUND: The biological behavior of low-grade glioma (LGG) is significantly affected by N6-methyladenosine (m6A) methylation, an essential epigenetic alteration. Therefore, it is crucial to create a prognostic model for LGG by utilizing genes that regulate m6A methylation.
    METHODS: Using TCGA and GTEx databases. We examined m6A modulator levels in LGG and normal tissues, and investigated PD-L1 and PD-1 expression, immune scores, immune cell infiltration, tumor immune microenvironment (TIME) and potential underlying mechanisms in different LGG clusters. We also performed immunohistochemistry and RT-qPCR to identify essential m6A adjustment factor.
    RESULTS: The results showed that m6A regulatory element expression was significantly increased in LGG tissues and was significantly associated with TMIE. A substantial increase in PD-L1 and PD-1 levels in LGG tissues and high-risk cohorts was observed. PD-L1 expression was positively correlated with FTO, ZCCHC4, and HNRNPD, whereas PD-1 expression was negatively correlated with FTO, ZC3H7B, and HNRNPD. The prognostic signature created using regulators of m6A RNA methylation was shown to be strongly associated with the overall survival of LGG patients, and FTO and ZCCHC4 were confirmed as independent prognostic markers by clinical samples. Furthermore, the results revealed different TIME characteristics between the two groups of patients, indicating disrupted signaling pathways associated with LGG.
    CONCLUSIONS: Our results present that the m6A regulators play vital role in regulating PD-L1/PD-1 expression and the infiltration of immune cells, thereby exerting a sizable impact on the TIME of LGG. Therefore, m6A regulators have precise predictive value in the prognosis of LGG.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号