Meg3

MEG3
  • 文章类型: Journal Article
    长链非编码RNA(lncRNA)的失调,如母本表达基因3(MEG3)和长基因间非编码RNA重编程调节因子(linc-ROR),在结直肠癌的进展中起着至关重要的作用。我们旨在同时评估linc-ROR沉默和MEG3激活对结直肠癌细胞增殖的影响;并探索TP53相关通路的潜在机制。在双向CEA启动子(UM1)下克隆MEG3和linc-RORshRNA。随后,构建另外的载体以表达linc-RORshRNA(UM2)和MEG3(UM3)。用这些重组载体转染结直肠癌细胞系后,细胞活力实验,凋亡,进行细胞周期分析。此外,使用定量实时聚合酶链反应(qRT-PCR)评估TP53的转录活性和相关基因。有趣的是,UM1比UM2和UM3显著抑制两种细胞系的增殖。响应于UM1,在HCT116细胞中TP53转录物(10.46)比SW480细胞(6.16)显着增加;这导致TP53INP1,TP53I3,GDF15,CCKN1A和BAX的上调,和G1细胞周期蛋白(D1,E1)的下调。HCT116(36.35%)和SW480(16.64%)细胞的凋亡率比对照组增加。此外,UM1转染的HCT116细胞在G0/G1期表现出明显的停滞,伴随着G2/M细胞群体的减少。与单向向量相比,同时靶向方法在转录水平上增强了TP53的激活.细胞对UM1的反应导致TP53的快速上调,导致细胞增殖的抑制,细胞凋亡增加,和细胞周期停滞。这些发现表明,靶向MEG3和linc-ROR的协同作用可以作为TP53相关结肠癌的有希望的治疗策略。
    Dysregulation of long noncoding RNAs (lncRNAs), such as maternally expressed gene 3 (MEG3) and long intergenic noncoding RNA regulator of reprogramming (linc-ROR), plays a crucial role in colorectal cancer progression. We aimed to assess linc-ROR silencing and MEG3 activation on the colorectal cancer cell proliferation simultaneously; and explore the underlying mechanisms in the TP53-associated Pathway. The MEG3 and linc-ROR shRNA were cloned under the bidirectional CEA promoter (UM1). Subsequently, additional vectors were constructed to express linc-ROR shRNA (UM2) and MEG3 (UM3). After transfecting colorectal cancer cell lines with these recombinant vectors, experiments on cell viability, apoptosis, and cell cycle analysis were conducted. Furthermore, TP53\'s transcriptional activity and associated genes were assessed using quantitative real-time polymerase chain reaction (qRT-PCR). Interestingly, UM1 significantly inhibited the proliferation of both cell lines than UM2 and UM3. In response to UM1, TP53 transcript remarkably increased in HCT116 cells (10.46) than SW480 cells (6.16); which resulted in up-regulation of TP53INP1, TP53I3, GDF15, CCKN1A and BAX, and down-regulation of G1 cyclins (D1, E1). The rate of apoptosis increased in HCT116 (36.35 %) and SW480 (16.64 %) cells than control. Moreover, UM1-transfected HCT116 cells exhibited a notable arrest in the G0/G1 phase, accompanied by a reduction in the G2/M cell population. Compared to unidirectional vectors, the concurrent targeting approach enhanced TP53 activation at the transcription level. The cell response to UM1 resulted in rapid upregulation of TP53, leading to inhibition of cell proliferation, increased apoptosis, and cell cycle arrest. These findings suggest that the synergistic effect of targeting both MEG3 and linc-ROR could serve as a promising therapeutic strategy for TP53-associated colon 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
    目的:最近的研究表明,HOTTIP和MEG3与各种类型肿瘤的发生和进展有关,包括鼻咽癌(NPC)。本研究旨在阐明HOTTIP和MEG3多态性对鼻咽癌易感性和临床病理特征的影响。
    方法:本研究采用下一代测序和多重PCR分别在200例鼻咽癌和200例健康个体中评估HOTTIPrs1859168和MEG3rs7158663的多态性。通过qRT-PCR评估评估HOTTIP和MEG3表达。此外,在健康个体和NPC个体之间比较了rs1859168和rs7158663的基因型和等位基因频率,以阐明它们对NPC易感性的影响以及与临床病理特征的关系。
    结果:与健康队列相比,HOTTIPrs1859168CC基因型和C等位基因的存在与NPC发病率增加显著相关(p<0.05)。此外,MEG3rs7158663AA基因型和A等位基因也表明NPC的风险增加(p<0.05)。年龄的亚组分析,EBV感染,性别,国籍,吸烟,饮酒状态显示rs1859168和rs7158663基因型与这些潜在的混杂因素之间没有显着关联。此外,观察到rs1859168CC和rs7158663AA基因型与局部肿瘤浸润和淋巴结转移有关。此外,HOTTIP标出了标高,而NPC样本中的MEG3比正常鼻咽生物标本中的MEG3大幅减少。携带CC或CA基因型而不是HOTTIPrs1859168AA基因型的患者,有更高的HOTTIP水平,而rs7158663AA或GA基因型患者的MEG3表达明显低于GG基因型携带者。
    结论:具有HOTTIPrs1859168和MEG3rs7158663遗传变异的个体可能增加NPC易感性和相关临床病理特征的风险,可能通过影响HOTTIP和MEG3的表达。
    OBJECTIVE: Recent studies have indicated that HOTTIP and MEG3 are associated with the initiation and progression of various types of tumors, including nasopharyngeal carcinoma (NPC). This investigation aimed to elucidate the impact of HOTTIP and MEG3 polymorphisms on the susceptibility and clinicopathologic characteristics of NPC.
    METHODS: This research employed next-generation sequencing and multiplex PCR to assess the polymorphisms of HOTTIP rs1859168 and MEG3 rs7158663 in 200 NPC and 200 healthy individuals respectively. HOTTIP and MEG3 expression were assessed via qRT-PCR assessment. Furthermore, the genotypes and alleles frequency of rs1859168 and rs7158663 were compared between healthy and NPC individuals to elucidate their influence on NPC susceptibility and relation with clinicopathologic characteristics.
    RESULTS: In comparison with the healthy cohort, the presence of HOTTIP rs1859168 CC genotype and the C allele were markedly linked with increased NPC incidence (p < 0.05). Furthermore, the MEG3 rs7158663 AA genotype and the A allele also indicated an increased risk of NPC (p < 0.05). The subgroup analysis of age, EBV infection, gender, nationality, smoking, and drinking status revealed no marked association between rs1859168 and rs7158663 genotypes and these potential confounding factors. Moreover, it was observed that rs1859168 CC and rs7158663 AA genotypes were related to local tumor invasion and lymph node metastasis. Additionally, HOTTIP indicated a marked elevation, while MEG3 substantially reduced in NPC samples than the normal nasopharyngeal biospecimens. Patients who carried CC or CA genotypes rather than the HOTTIP rs1859168 AA genotype, had substantially higher HOTTIP levels, while patients with rs7158663 AA or GA genotypes indicated notably lower expression of MEG3 than GG genotype carriers.
    CONCLUSIONS: Individuals with genetic variants of HOTTIP rs1859168 and MEG3 rs7158663 might have an increased risk of NPC susceptibility and related clinicopathologic characteristics, potentially by affecting the expression of HOTTIP and MEG3.
    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
    前几代条件引起的代际和跨代表观遗传效应可有助于环境适应以及疾病易感性。先前在啮齿动物和人体模型中的研究表明,甲状腺激素的异常发育暴露会影响后代的内分泌功能和甲状腺激素敏感性。由于印迹3型脱碘酶基因(Dio3)调节甲状腺激素的敏感性,我们假设其表观遗传调控在甲状腺激素过度暴露个体的后代中发生了改变.使用DIO3缺陷小鼠作为发育性甲状腺毒症模型,我们调查了Dio3在后代中的总和等位基因表达以及生长和内分泌表型。我们观察到,男性和女性发育过度暴露于甲状腺激素以组织特异性方式改变了遗传完整后代中总的和等位基因的Dio3表达。这与甲状腺激素和瘦素的异常生长和新生儿水平有关。后代小鼠在Dlk1-Dio3印迹域中也表现出分子异常,包括Meg3甲基化增加和Dlk1-Dio3印迹域其他基因的胎儿脑表达改变。在最初在子宫内过度暴露于甲状腺激素的DIO3缺陷祖先的组织和种系中也观察到了这些分子异常。我们的结果提供了一种新的表观遗传自我记忆范例,通过该范例,Dio3基因剂量在给定个体中,以及它对甲状腺激素的依赖性发育暴露,影响自己在后代中的表达。这种在每一代中Dio3表达的表观遗传自校正机制可能对后代的发育生长和成人内分泌功能的适应性编程有帮助。
    Intergenerational and transgenerational epigenetic effects resulting from conditions in previous generations can contribute to environmental adaptation as well as disease susceptibility. Previous studies in rodent and human models have shown that abnormal developmental exposure to thyroid hormone affects endocrine function and thyroid hormone sensitivity in later generations. Since the imprinted type 3 deiodinase gene (Dio3) regulates sensitivity to thyroid hormones, we hypothesize its epigenetic regulation is altered in descendants of thyroid hormone overexposed individuals. Using DIO3-deficient mice as a model of developmental thyrotoxicosis, we investigated Dio3 total and allelic expression and growth and endocrine phenotypes in descendants. We observed that male and female developmental overexposure to thyroid hormone altered total and allelic Dio3 expression in genetically intact descendants in a tissue-specific manner. This was associated with abnormal growth and neonatal levels of thyroid hormone and leptin. Descendant mice also exhibited molecular abnormalities in the Dlk1-Dio3 imprinted domain, including increased methylation in Meg3 and altered foetal brain expression of other genes of the Dlk1-Dio3 imprinted domain. These molecular abnormalities were also observed in the tissues and germ line of DIO3-deficient ancestors originally overexposed to thyroid hormone in utero. Our results provide a novel paradigm of epigenetic self-memory by which Dio3 gene dosage in a given individual, and its dependent developmental exposure to thyroid hormone, influences its own expression in future generations. This mechanism of epigenetic self-correction of Dio3 expression in each generation may be instrumental in descendants for their adaptive programming of developmental growth and adult endocrine function.
    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
    大约30%-40%的分泌生长激素的垂体腺瘤(GHPAs)在GNAS(刺激性G蛋白的α亚基)中具有体细胞激活突变。GNAS中的突变与较小且侵入性较小的肿瘤的临床特征相关。然而,GNAS突变在GHPAs侵袭中的作用尚不清楚.使用标准聚合酶链反应(PCR)测序程序在GHPA中检测GNAS突变。用RT-qPCR评估突变相关的母体表达基因3(MEG3)的表达。使用慢病毒表达系统在GH3细胞中操作MEG3。使用Transwell测定法测量细胞侵袭能力,和上皮间质转化(EMT)相关蛋白通过免疫荧光和蛋白质印迹进行定量。最后,使用肿瘤细胞异种移植小鼠模型来验证MEG3对肿瘤生长和侵袭性的影响。与具有野生型GNAS的小鼠相比,具有突变GNAS的小鼠的GHPAs的侵袭力明显降低。始终如一,突变表达GNAS的GH3细胞的侵袭力降低。MEG3在携带突变GNAS的GHPAs中以高水平独特表达。因此,MEG3上调抑制肿瘤细胞侵袭,反过来,MEG3下调增加肿瘤细胞侵袭。机械上,GNAS突变抑制GHPAs中的EMT。突变的GNAS细胞中的MEG3通过Wnt/β-catenin信号通路的失活阻止细胞侵袭,进一步在体内验证。我们的数据表明,GNAS突变可能通过MEG3/Wnt/β-catenin信号通路的激活来调节EMT,从而抑制GHPAs中的细胞侵袭。
    Approximately 30%-40% of growth hormone-secreting pituitary adenomas (GHPAs) harbor somatic activating mutations in GNAS (α subunit of stimulatory G protein). Mutations in GNAS are associated with clinical features of smaller and less invasive tumors. However, the role of GNAS mutations in the invasiveness of GHPAs is unclear. GNAS mutations were detected in GHPAs using a standard polymerase chain reaction (PCR) sequencing procedure. The expression of mutation-associated maternally expressed gene 3 (MEG3) was evaluated with RT-qPCR. MEG3 was manipulated in GH3 cells using a lentiviral expression system. Cell invasion ability was measured using a Transwell assay, and epithelial-mesenchymal transition (EMT)-associated proteins were quantified by immunofluorescence and western blotting. Finally, a tumor cell xenograft mouse model was used to verify the effect of MEG3 on tumor growth and invasiveness. The invasiveness of GHPAs was significantly decreased in mice with mutated GNAS compared with that in mice with wild-type GNAS. Consistently, the invasiveness of mutant GNAS-expressing GH3 cells decreased. MEG3 is uniquely expressed at high levels in GHPAs harboring mutated GNAS. Accordingly, MEG3 upregulation inhibited tumor cell invasion, and conversely, MEG3 downregulation increased tumor cell invasion. Mechanistically, GNAS mutations inhibit EMT in GHPAs. MEG3 in mutated GNAS cells prevented cell invasion through the inactivation of the Wnt/β-catenin signaling pathway, which was further validated in vivo. Our data suggest that GNAS mutations may suppress cell invasion in GHPAs by regulating EMT through the activation of the MEG3/Wnt/β-catenin 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
    长非编码RNA(lncRNA)是长度为200个核苷酸或更多的RNA分子,其不被翻译成蛋白质。它们的表达是组织特异性的,绝大多数参与细胞过程和功能的调节。许多人类疾病,包括癌症,已被证明与失调的lncRNAs相关,为鉴别诊断提供潜在的治疗靶点和生物标志物。lncRNAs在神经系统中的表达在不同的细胞类型中有所不同,与神经元和神经胶质的机制有关,影响大脑的发育和功能。报告还显示了lncRNA分子的变化与脑肿瘤的病因之间的联系,包括多形性胶质母细胞瘤(GBM)。GBM是脑癌的侵袭性变体,预后不良,中位生存期为14-16个月。它被认为是一种脑特异性疾病,高度侵袭性的恶性细胞遍布神经组织,阻碍了完整的切除,导致术后复发,这是死亡的主要原因。GBM的早期诊断可以提高治疗效果,延长生存期。生物体液的lncRNA分析有望在其初始阶段检测肿瘤变化和更有效的治疗干预措施。这篇综述提供了GBM相关的lncRNAs失调的系统概述,重点关注患者血液中的lncRNA指纹。
    Long noncoding RNAs (lncRNAs) are RNA molecules of 200 nucleotides or more in length that are not translated into proteins. Their expression is tissue-specific, with the vast majority involved in the regulation of cellular processes and functions. Many human diseases, including cancer, have been shown to be associated with deregulated lncRNAs, rendering them potential therapeutic targets and biomarkers for differential diagnosis. The expression of lncRNAs in the nervous system varies in different cell types, implicated in mechanisms of neurons and glia, with effects on the development and functioning of the brain. Reports have also shown a link between changes in lncRNA molecules and the etiopathogenesis of brain neoplasia, including glioblastoma multiforme (GBM). GBM is an aggressive variant of brain cancer with an unfavourable prognosis and a median survival of 14-16 months. It is considered a brain-specific disease with the highly invasive malignant cells spreading throughout the neural tissue, impeding the complete resection, and leading to post-surgery recurrences, which are the prime cause of mortality. The early diagnosis of GBM could improve the treatment and extend survival, with the lncRNA profiling of biological fluids promising the detection of neoplastic changes at their initial stages and more effective therapeutic interventions. This review presents a systematic overview of GBM-associated deregulation of lncRNAs with a focus on lncRNA fingerprints in patients\' blood.
    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
    涉及长链非编码RNA的表观遗传过程调节内皮基因表达。然而,导致内皮功能障碍的潜在调节机制仍有待阐明。zeste同源物2(EZH2)的增强子是组蛋白H3K27三甲基化(H3K27me3)的重要变阻器,但EZH2RNA结合能力和EZH2:RNA功能相互作用尚未在缺血后血管生成中进行研究。我们使用甲醛/UV辅助交联连接和杂交测序,并确定了母本表达基因3(MEG3)的新作用。MEG3在内皮细胞中形成主要的RNA:RNA杂交结构。此外,MEG3:EZH2协助募集到染色质上。通过EZH2-染色质免疫沉淀,在MEG3耗尽之后,我们证明MEG3控制EZH2/H3K27me3募集到整合素亚基α4(ITGA4)启动子上。MEG3敲低或EZH2抑制(A-395)均促进ITGA4表达,并在体外改善内皮细胞迁移和与纤连蛋白的粘附。A-395抑制剂重新引导MEG3辅助染色质重塑,通过增加内皮功能和弹性提供直接的治疗益处。这种方法随后增加了小鼠缺血性损伤后小动脉中ITGA4的表达,从而促进动脉生成。我们的研究结果表明,MEG3在指导EZH2抑制ITGA4方面具有特定的作用。新的治疗策略可以拮抗MEG3:EZH2相互作用,用于临床前研究。
    Epigenetic processes involving long non-coding RNAs regulate endothelial gene expression. However, the underlying regulatory mechanisms causing endothelial dysfunction remain to be elucidated. Enhancer of zeste homolog 2 (EZH2) is an important rheostat of histone H3K27 trimethylation (H3K27me3) that represses endothelial targets, but EZH2 RNA binding capacity and EZH2:RNA functional interactions have not been explored in post-ischemic angiogenesis. We used formaldehyde/UV-assisted crosslinking ligation and sequencing of hybrids and identified a new role for maternally expressed gene 3 (MEG3). MEG3 formed the predominant RNA:RNA hybrid structures in endothelial cells. Moreover, MEG3:EZH2 assists recruitment onto chromatin. By EZH2-chromatin immunoprecipitation, following MEG3 depletion, we demonstrated that MEG3 controls recruitment of EZH2/H3K27me3 onto integrin subunit alpha4 (ITGA4) promoter. Both MEG3 knockdown or EZH2 inhibition (A-395) promoted ITGA4 expression and improved endothelial cell migration and adhesion to fibronectin in vitro. The A-395 inhibitor re-directed MEG3-assisted chromatin remodeling, offering a direct therapeutic benefit by increasing endothelial function and resilience. This approach subsequently increased the expression of ITGA4 in arterioles following ischemic injury in mice, thus promoting arteriogenesis. Our findings show a context-specific role for MEG3 in guiding EZH2 to repress ITGA4. Novel therapeutic strategies could antagonize MEG3:EZH2 interaction for pre-clinical studies.
    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:长链非编码RNA参与动脉粥样硬化的发病机制。由于长非编码RNA母体表达基因3(Meg3)可防止肝血管内皮细胞衰老和肥胖诱导的胰岛素抵抗,我们决定研究其在细胞衰老和动脉粥样硬化中的作用.
    结果:通过分析我们的数据以及来自基因表达综合数据库的人类和小鼠数据,我们发现Meg3在患有心血管疾病的人和小鼠中的表达降低,表明其在动脉粥样硬化中的潜在作用。在喂食西方饮食12周的Ldlr-/-小鼠中,通过化学修饰的反义寡核苷酸沉默Meg3使雄性和雌性小鼠动脉粥样硬化病变的形成减少了34.9%和20.1%,分别,通过正面油红O染色显示,这与血浆脂质谱的变化无关。细胞衰老标志物p21和p16的实时定量PCR分析显示,Meg3缺乏会加重肝细胞衰老,但不会加重主动脉根部的细胞衰老。产生人Meg3转基因小鼠以检查Meg3功能获得在PCSK9过表达诱导的动脉粥样硬化发展中的作用。Meg3过表达在Meg3敲入小鼠中促进动脉粥样硬化病变形成29.2%,而与其对血脂谱的影响无关。Meg3过表达抑制肝细胞衰老,而它可能通过损害线粒体功能和延迟细胞周期进程来促进主动脉细胞衰老。
    结论:我们的数据表明Meg3促进动脉粥样硬化病变的形成,而不依赖于其对血浆脂质分布的影响。此外,Meg3在动脉粥样硬化期间以组织特异性方式调节细胞衰老。因此,我们证明Meg3在细胞衰老和动脉粥样硬化中具有多方面的作用.
    OBJECTIVE: Long noncoding RNAs are involved in the pathogenesis of atherosclerosis. As long noncoding RNAs maternally expressed gene 3 (Meg3) prevents cellular senescence of hepatic vascular endothelium and obesity-induced insulin resistance, we decided to examine its role in cellular senescence and atherosclerosis.
    RESULTS: By analyzing our data and human and mouse data from the Gene Expression Omnibus database, we found that Meg3 expression was reduced in humans and mice with cardiovascular disease, indicating its potential role in atherosclerosis. In Ldlr-/- mice fed a Western diet for 12 weeks, Meg3 silencing by chemically modified antisense oligonucleotides attenuated the formation of atherosclerotic lesions by 34.9% and 20.1% in male and female mice, respectively, revealed by en-face Oil Red O staining, which did not correlate with changes in plasma lipid profiles. Real-time quantitative PCR analysis of cellular senescence markers p21 and p16 revealed that Meg3 deficiency aggravates hepatic cellular senescence but not cellular senescence at aortic roots. Human Meg3 transgenic mice were generated to examine the role of Meg3 gain-of-function in the development of atherosclerosis induced by PCSK9 overexpression. Meg3 overexpression promotes atherosclerotic lesion formation by 29.2% in Meg3 knock-in mice independent of its effects on lipid profiles. Meg3 overexpression inhibits hepatic cellular senescence, while it promotes aortic cellular senescence likely by impairing mitochondrial function and delaying cell cycle progression.
    CONCLUSIONS: Our data demonstrate that Meg3 promotes the formation of atherosclerotic lesions independent of its effects on plasma lipid profiles. In addition, Meg3 regulates cellular senescence in a tissue-specific manner during atherosclerosis. Thus, we demonstrated that Meg3 has multifaceted roles in cellular senescence and atherosclerosis.
    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:卵巢癌对女性健康构成严重威胁。由于难以早期发现,大多数患者被诊断为晚期疾病或腹膜转移。我们发现LncRNAMEG3是一种新型的肿瘤抑制因子,但其在肿瘤发生发展中的作用尚不清楚。
    方法:我们通过生物信息学分析研究了MEG3在泛癌症中的表达水平,尤其是妇科肿瘤。功能测定用于检测MEG3对卵巢癌恶性表型的影响。RIP,RNA下拉,MeRIP-qPCR,通过放线菌素D试验探讨m6A甲基化对MEG3的调控作用。荧光素酶报告基因测定,实施PCR和Western印迹以揭示MEG3的潜在机制。我们通过原位异种移植模型和IHC测定进一步证实了MEG3对体内肿瘤生长的影响。
    结果:在这项研究中,我们发现MEG3在各种癌症中下调,在卵巢癌中最明显的下调。MEG3抑制增殖,迁移,和卵巢癌细胞的侵袭。MEG3的过表达通过负调控miR-885-5p抑制VASH1的降解,抑制卵巢癌恶性表型。此外,我们证明MEG3在转录后水平受到调控。YTHDF2通过识别METTL3介导的m6A修饰促进MEG3衰变。与注射载体对照细胞相比,注射MEG3敲低细胞的小鼠显示出更大的肿瘤体积和更快的生长速率。
    结论:我们证明MEG3受METTL3/YTHDF2甲基化的影响,并通过结合miR-885-5p增加VASH1表达来抑制卵巢癌的增殖和转移。MEG3有望成为卵巢癌的治疗靶标。
    Ovarian cancer poses a serious threat to women\'s health. Due to the difficulty of early detection, most patients are diagnosed with advanced-stage disease or peritoneal metastasis. We found that LncRNA MEG3 is a novel tumor suppressor, but its role in tumor occurrence and development is still unclear.
    We investigated the expression level of MEG3 in pan-cancer through bioinformatics analysis, especially in gynecological tumors. Function assays were used to detect the effect of MEG3 on the malignant phenotype of ovarian cancer. RIP, RNA pull-down, MeRIP-qPCR, actinomycin D test were carried out to explore the m6A methylation-mediated regulation on MEG3. Luciferase reporter gene assay, PCR and Western blot were implemented to reveal the potential mechanism of MEG3. We further confirmed the influence of MEG3 on tumor growth in vivo by orthotopic xenograft models and IHC assay.
    In this study, we discovered that MEG3 was downregulated in various cancers, with the most apparent downregulation in ovarian cancer. MEG3 inhibited the proliferation, migration, and invasion of ovarian cancer cells. Overexpression of MEG3 suppressed the degradation of VASH1 by negatively regulating miR-885-5p, inhibiting the ovarian cancer malignant phenotype. Furthermore, we demonstrated that MEG3 was regulated at the posttranscriptional level. YTHDF2 facilitated MEG3 decay by recognizing METTL3‑mediated m6A modification. Compared with those injected with vector control cells, mice injected with MEG3 knockdown cells showed larger tumor volumes and faster growth rates.
    We demonstrated that MEG3 is influenced by METTL3/YTHDF2 methylation and restrains ovarian cancer proliferation and metastasis by binding miR-885-5p to increase VASH1 expression. MEG3 is expected to become a therapeutic target for ovarian 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:乳腺癌是全球最常见的癌症,是女性癌症死亡的主要原因。新的生物标志物对于这种疾病是非常必要的。
    目的:评估乳腺癌的新型长链非编码RNA生物标志物。
    方法:该研究包括通过筛选分析71个候选lncRNAs的表达,其中六个(四个不足,两个过表达)通过qPCR在与NST乳腺癌相关的肿瘤组织中进行验证和分析,与良性样本及其临床病理特征进行比较。
    结果:结果表明PTENP1、GNG12-AS1、MEG3和MAGI2-AS3的抑癌作用。PTENP1和GNG12-AS1的低水平与无进展和总生存率恶化相关。GNG12-AS1的表达降低与晚期阶段有关。较高的等级与PTENP1、GNG12-AS1和MAGI2-AS3的较低表达相关。MEG3和PTENP1水平降低与Ki-67阳性相关。NRSN2-AS1和UCA1lncRNAs过表达;较高水平的UCA1与多灶性相关。
    结论:结果表明,所研究的lncRNAs可能在乳腺癌中起重要作用,并且包含一个潜在的因素,应该在临床研究中进一步评估。
    UNASSIGNED: Breast cancer is the most commonly occurring cancer worldwide and is the main cause of death from cancer in women. Novel biomarkers are highly warranted for this disease.
    UNASSIGNED: Evaluation of novel long non-coding RNAs biomarkers for breast cancer.
    UNASSIGNED: The study comprised the analysis of the expression of 71 candidate lncRNAs via screening, six of which (four underexpressed, two overexpressed) were validated and analyzed by qPCR in tumor tissues associated with NST breast carcinomas, compared with the benign samples and with respect to their clinicopathological characteristics.
    UNASSIGNED: The results indicated the tumor suppressor roles of PTENP1, GNG12-AS1, MEG3 and MAGI2-AS3. Low levels of both PTENP1 and GNG12-AS1 were associated with worsened progression-free and overall survival rates. The reduced expression of GNG12-AS1 was linked to the advanced stage. A higher grade was associated with the lower expression of PTENP1, GNG12-AS1 and MAGI2-AS3. Reduced levels of both MEG3 and PTENP1 were linked to Ki-67 positivity. The NRSN2-AS1 and UCA1 lncRNAs were overexpressed; higher levels of UCA1 were associated with multifocality.
    UNASSIGNED: The results suggest that the investigated lncRNAs may play important roles in breast cancer and comprise a potential factor that should be further evaluated in clinical studies.
    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:本研究旨在探讨长链非编码RNA(lncRNA)MEG3在乳腺癌发生发展中的分子机制。
    方法:通过对癌症基因组图谱数据库中乳腺癌样本的生物信息学分析,预测了lncRNAMEG3、miRNA-330和CNN1之间的调控关系。lncRNAMEG3、miRNA-330和CNN1的差异表达首次在乳腺癌组织和细胞中得到验证。通过操纵其在MCF-7和BT-474细胞中的表达来评估lncRNAMEG3对乳腺癌恶性特性的影响。救援实验,双荧光素酶测定,和RNA免疫沉淀(RIP)实验进一步用于验证lncRNAMEG3,miRNA-330和CNN1之间的关系。
    结果:生物信息学分析显示,lncRNAMEGs和CNN1在乳腺癌组织中显著下调,而miR-330上调。在我们的乳腺癌样本队列中进一步验证了这些差异表达。lncRNAMEG3和CNN1的高表达水平以及miR-330的低表达与有利的总体存活显著相关。lncRNAMEG3的过表达显著抑制细胞活力,移民和入侵,S期细胞减少,促进细胞凋亡。双荧光素酶报告基因测定和RIP实验表明,lncRNAMEG3可以直接结合miR-330。此外,基于lncRNAMEG3过表达的miR-330模拟物通过降低CNN1表达改善了lncRNAMEG3在乳腺癌恶性特性中的肿瘤抑制作用。
    结论:我们的研究表明lncRNAMEG3是一种通过调节miR-330/CNN1轴的乳腺癌抑制因子。
    The current study aimed to investigate the molecular mechanism of long non-coding RNA (lncRNA) MEG3 in the development of breast cancer.
    The regulating relationships among lncRNA MEG3, miRNA-330 and CNN1 were predicted by bioinformatics analysis of breast cancer samples in the Cancer Genome Atlas database. The differential expression of lncRNA MEG3, miRNA-330 and CNN1 was first validated in breast cancer tissues and cells. The effects of lncRNA MEG3 on breast cancer malignant properties were evaluated by manipulating its expression in MCF-7 and BT-474 cells. Rescue experiments, dual-luciferase assays, and RNA immunoprecipitation (RIP) experiments were further used to validate the relationships among lncRNA MEG3, miRNA-330 and CNN1.
    Bioinformatics analysis showed that lncRNA MEGs and CNN1 were significantly downregulated in breast cancer tissues, while miR-330 was upregulated. These differential expressions were further validated in our cohort of breast cancer samples. High expression levels of lncRNA MEG3 and CNN1 as well as low expression of miR-330 were significantly associated with favorable overall survival. Overexpression of lncRNA MEG3 significantly inhibited cell viability, migration and invasion, decreased cells in S stage and promoted cell apoptosis. Dual-luciferase reporter gene assay and RIP experiments showed that lncRNA MEG3 could directly bind to miR-330. Moreover, miR-330 mimics on the basis of lncRNA MEG3 overexpression ameliorated the tumor-suppressing effects of lncRNA MEG3 in breast cancer malignant properties by decreasing CNN1 expression.
    Our study indicated lncRNA MEG3 is a breast cancer suppressor by regulating miR-330/CNN1 axis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号