ASCL2

  • 文章类型: Journal Article
    胰腺腺癌(PAAD)是一种危及生命的癌症。探索新的诊断和治疗靶点有助于改善其预后。tRNA衍生的非编码小RNA(tsRNA)是一种新型的基因表达调控因子,其调控异常与许多人类癌症密切相关。然而,PAAD中tsRNA的表达和功能尚不清楚。我们的研究使用RNA测序来鉴定在无葡萄糖或高葡萄糖培养基中培养的PAAD细胞中的tsRNA表达谱,并发现tRF-18-8R6546D2是一种未表征的tsRNA。在PAAD细胞和组织中具有显著的高表达。临床上,tRF-18-8R6546D2与PAAD患者的不良预后相关,可用于将其与健康人群区分开。功能上,在体外和体内,tRF-18-8R6546D2过表达促进PAAD细胞增殖,移民和入侵,抑制细胞凋亡,而tRF-18-8R6546D2敲低显示出相反的效果。机械上,tRF-18-8R6546D2部分通过直接沉默ASCL2并进一步调节其下游基因如MYC和CASP3来促进PAAD恶性肿瘤。这些发现表明tRF-18-8R6546D2是一种新的致癌因子,并且可以成为PAAD的有希望的诊断或预后生物标志物和治疗靶标。
    Pancreatic adenocarcinoma (PAAD) is a life-threatening cancer. Exploring new diagnosis and treatment targets helps improve its prognosis. tRNA-derived small non-coding RNAs (tsRNAs) are a novel type of gene expression regulators and their dysregulation is closely related to many human cancers. Yet the expression and functions of tsRNAs in PAAD are not well understood. Our study used RNA sequencing to identify tsRNA expression profiles in PAAD cells cultured in no or high glucose media and found tRF-18-8R6546D2 was an uncharacterized tsRNA, which has significantly high expression in PAAD cells and tissues. Clinically, tRF-18-8R6546D2 is linked to poor prognosis in PAAD patients and can be used to distinguish them from healthy populations. Functionally, in vitro and vivo, tRF-18-8R6546D2 over-expression promoted PAAD cell proliferation, migration and invasion, inhibited apoptosis, whereas tRF-18-8R6546D2 knock-down showed opposite effects. Mechanistically, tRF-18-8R6546D2 promoted PAAD malignancy partly by directly silencing ASCL2 and further regulating its downstream genes such as MYC and CASP3. These findings show that tRF-18-8R6546D2 is a novel oncogenic factor and can be a promising diagnostic or prognostic biomarker and therapeutic target for PAAD.
    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
    食道由分层上皮保护免受恶劣环境的影响,迅速更新。该上皮的稳态由基底层中的罕见干细胞群确保:角蛋白15+(Krt15+)细胞。然而,对调节其独特特征的分子机制知之甚少,即自我更新,效力和上皮再生。Achaete-scute家族BHLH转录因子2(ASCL2)在Krt15干细胞中强烈上调,并且已知有助于其他组织中的干细胞维持。在这里,我们研究了ASCL2在维持食管上皮正常和应激条件下的稳态中的作用.ASCL2过表达严重失调的细胞分化和细胞命运。由于细胞周期G1期的阻断或静止的诱导,增殖也降低。质谱分析证实了与分化和细胞周期相关的几种蛋白质的改变。此外,ASCL2的过表达增强了对放射和化疗药物的抗性。总的来说,这些结果表明ASCL2作为食管上皮增殖-分化平衡的关键调节因子的作用.
    The esophagus is protected from the hostile environment by a stratified epithelium, which renews rapidly. Homeostasis of this epithelium is ensured by a rare population of stem cells in the basal layer: Keratin 15+ (Krt15+) cells. However, little is known about the molecular mechanisms regulating their distinct features, namely self-renewal, potency and epithelial regeneration. Achaete-scute family BHLH transcription factor 2 (ASCL2) is strongly upregulated in Krt15+ stem cells and is known to contribute to stem cell maintenance in other tissues. Herein, we investigated the role of ASCL2 in maintaining homeostasis under normal and stress conditions in the esophageal epithelium. ASCL2 overexpression severely dysregulated cell differentiation and cell fate. Proliferation was also reduced due potentially to a blockage in the G1 phase of the cell cycle or an induction of quiescence. Mass spectrometry analysis confirmed alterations in several proteins associated with differentiation and the cell cycle. In addition, overexpression of ASCL2 enhanced resistance to radiation and chemotherapeutic drugs. Overall, these results denote the role of ASCL2 as a key regulator of the proliferation-differentiation equilibrium in the esophageal epithelium.
    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
    从腺癌到小细胞神经内分泌状态的跨分化与多种癌症类型的治疗抗性相关。为了深入了解转分化的潜在分子事件,我们对泛小细胞神经内分泌癌模型(称为PARCB)进行了多组学时程分析,使用人类前列腺基底细胞进行正向遗传转化,并确定共同的发育,弧形,和所有转换模型复制中的高熵轨迹。使用单细胞分辨率的进一步映射揭示了由ASCL1或ASCL2的互斥表达定义的两个不同的谱系。跨发育阶段的转录因子组的时间调节表明,细胞重编程先于神经元程序的诱导。TFAP4和ASCL1/2反馈被认为是ASCL1和ASCL2表达的潜在调节因子。我们的研究提供了时间转录模式,并揭示了前列腺癌和肺癌之间的泛组织相似性,以及与正常神经内分泌细胞状态的联系。
    Trans-differentiation from an adenocarcinoma to a small cell neuroendocrine state is associated with therapy resistance in multiple cancer types. To gain insight into the underlying molecular events of the trans-differentiation, we perform a multi-omics time course analysis of a pan-small cell neuroendocrine cancer model (termed PARCB), a forward genetic transformation using human prostate basal cells and identify a shared developmental, arc-like, and entropy-high trajectory among all transformation model replicates. Further mapping with single cell resolution reveals two distinct lineages defined by mutually exclusive expression of ASCL1 or ASCL2. Temporal regulation by groups of transcription factors across developmental stages reveals that cellular reprogramming precedes the induction of neuronal programs. TFAP4 and ASCL1/2 feedback are identified as potential regulators of ASCL1 and ASCL2 expression. Our study provides temporal transcriptional patterns and uncovers pan-tissue parallels between prostate and lung cancers, as well as connections to normal neuroendocrine cell states.
    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
    p57Kip2是细胞周期蛋白/CDK抑制剂和细胞增殖的负调节因子。这里,我们报道p57在肠道发育过程中以不依赖CDK的方式调节肠道干细胞(ISC)命运和增殖。在没有p57的情况下,肠道隐窝表现出增加的增殖和转运扩增细胞和Hopx+ISC的扩增,不再是静止的,而Lgr5+ISC不受影响。Hopx+ISC的RNA测序(RNA-seq)分析显示在不存在p57的情况下主要的基因表达变化。我们发现p57结合并抑制Ascl2的活性,Ascl2是一种对ISC规范和维持至关重要的转录因子,通过参与Ascl2靶基因启动子的辅抑制复合物的募集。因此,我们的数据表明,在肠道发育过程中,p57通过以不依赖CDK的方式抑制转录因子Ascl2,在维持HopxISC静止和抑制隐窝底部外部的ISC表型中起关键作用。
    p57Kip2 is a cyclin/CDK inhibitor and a negative regulator of cell proliferation. Here, we report that p57 regulates intestinal stem cell (ISC) fate and proliferation in a CDK-independent manner during intestinal development. In the absence of p57, intestinal crypts exhibit an increased proliferation and an amplification of transit-amplifying cells and of Hopx+ ISCs, which are no longer quiescent, while Lgr5+ ISCs are unaffected. RNA sequencing (RNA-seq) analyses of Hopx+ ISCs show major gene expression changes in the absence of p57. We found that p57 binds to and inhibits the activity of Ascl2, a transcription factor critical for ISC specification and maintenance, by participating in the recruitment of a corepressor complex to Ascl2 target gene promoters. Thus, our data suggest that, during intestinal development, p57 plays a key role in maintaining Hopx+ ISC quiescence and repressing the ISC phenotype outside of the crypt bottom by inhibiting the transcription factor Ascl2 in a CDK-independent manner.
    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
    在来自乳腺癌患者和五种乳腺癌细胞系的肿瘤样品中发现了ARHGAP25的下调。然而,其在乳腺癌中的确切作用和分子机制仍完全未知。在这里,我们发现敲低ARHGAP25在乳腺癌细胞中促进增殖,乳腺癌细胞的迁移和侵袭。机械上,ARHGAP25沉默促进了Wnt/β-catenin通路的激活及其下游分子(包括c-Myc,细胞周期蛋白D1,PCNA,MMP2,MMP9,蜗牛,和ASCL2)通过直接调节乳腺癌细胞中的Rac1/PAK1。体内异种移植实验表明ARHGAP25沉默促进肿瘤生长并激活Wnt/β-catenin途径。相比之下,ARHGAP25在体外和体内的过表达阻碍了上述所有癌症特性。有趣的是,ASCL2,Wnt/β-catenin通路的下游靶标,转录抑制了ARHGAP25的表达,因此构成了负反馈回路。此外,生物信息学分析表明,ARHGAP25与肿瘤免疫细胞浸润和不同免疫细胞亚群患者的生存密切相关。总的来说,我们的研究显示,ARHGAP25抑制了乳腺癌的肿瘤进展.它为乳腺癌的治疗提供了新的见解。
    Downregulation of ARHGAP25 was found in the tumor samples from breast cancer patients and five breast cancer cell lines. However, its precise role and molecular mechanisms in breast cancer remain completely unknown. Herein, we found that knockdown of ARHGAP25 in breast cancer cells promoted proliferation, migration and invasion of breast cancer cells. Mechanistically, ARHGAP25 silence facilitated the activation of the Wnt/β-catenin pathway and the upregulation of its downstream molecules (including c-Myc, Cyclin D1, PCNA, MMP2, MMP9, Snail and ASCL2) by directly regulating Rac1/PAK1 in breast cancer cells. In vivo xenograft experiments indicated ARHGAP25 silence promoted tumor growth and activated the Wnt/β-catenin pathway. In contrast, overexpression of ARHGAP25 in vitro and in vivo impeded all of the above cancer properties. Intriguingly, ASCL2, a downstream target of the Wnt/β-catenin pathway, transcriptionally repressed the expression of ARHGAP25 and therefore constituted a negative feedback loop. Moreover, bioinformatics analysis indicated that ARHGAP25 was significantly correlated with tumor immune cell infiltration and the survival of patients with different immune cell subgroups in breast cancer. Collectively, our work revealed that ARHGAP25 suppressed tumor progression of breast cancer. It provides a novel insight for the treatment of breast cancer.
    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:为了研究achaete-scute复合物样2(ASCL2)在胃腺癌(STAD)中的作用,我们分析了ASCL2抑制是否可以延迟癌症的发展,并进一步观察ASCL2与STAD中Toll样受体4(TLR4)激活的炎症之间的相关性,在体外和体内。
    方法:增殖,发展,炎症,使用sh-ASCL2慢病毒在体外和体内通过TLR4激活观察到STAD中的凋亡。分析ASCL2与炎症的关系。用免疫相关细胞的靶蛋白进行ASCL2的蛋白质印迹。分析STAD和相关ASCL2突变的预后。
    结果:STAD肿瘤组织中ASCL2水平升高,与正常组织相比,并带来更糟糕的预后。ASCL2与炎症呈负相关,TLR4与胃癌呈正相关。ASCL2在MGC803细胞中表达较高。Sh-ASCL2可以通过减少增殖来减少STAD的发展,肿瘤体积,和生物标志物水平和增加细胞凋亡的体外和体内。ASCL2的炎症作用是通过TLR4激活来调节的。ASCL2水平可能与CNTNAP3、CLIP1、C9orf84、ARIH2和IL1R2突变有关;与M2巨噬细胞和滤泡辅助性T细胞水平呈正相关;与中性粒细胞呈负相关,树突状细胞,单核细胞,CD8T细胞,和M1巨噬细胞水平;并参与STAD预后。
    结论:ASCL2可能通过激活TLR4调节STAD的炎症反应,并可能与相关免疫细胞有关。ASCL2可能是TLR4信号通路的上游靶因子。
    BACKGROUND: To investigate the role of achaete-scute complex-like 2 (ASCL2) in stomach adenocarcinoma (STAD), we analyze whether ASCL2 suppression could retard cancer development and further observe the relevance between ASCL2 and inflammation via Toll-like receptor 4 (TLR4) activation in STAD, both in vitro and in vivo.
    METHODS: Proliferation, development, inflammation, and apoptosis in STAD are observed using sh-ASCL2 lentivirus via TLR4 activation in vitro and in vivo. The relationship between ASCL2 and inflammation is analyzed. Western blotting of ASCL2 with the target protein of immune-associated cells is performed. The prognosis of STAD and associated ASCL2 mutation are analyzed.
    RESULTS: The ASCL2 level in STAD tumor tissues is increased, compared to normal tissues, and brings a worse prognosis. The ASCL2 shows a negative correlation with inflammation, and TLR4 reveals a positive correlation with gastric cancer. ASCL2 expression is high in MGC803 cells. Sh-ASCL2 could reduce STAD development by decreasing proliferation, tumor volume, and biomarker levels and increasing apoptosis in vitro and in vivo. The inflammatory role of ASCL2 is regulated through TLR4 activation. ASCL2 levels may be related to CNTNAP3, CLIP1, C9orf84, ARIH2, and IL1R2 mutations; positively correlated with M2 macrophage and T follicular helper cell levels; negatively correlated with neutrophil, dendritic cell, monocyte, CD8 T cell, and M1 macrophage levels; and involved in STAD prognosis.
    CONCLUSIONS: The ASCL2 may adjust inflammation in STAD through TLR4 activation and may be associated with related immune cells. ASCL2 is possibly an upstream target factor of the TLR4 signaling pathway.
    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
    自噬是一个高度保守的过程,对肿瘤进展和治疗反应至关重要。尽管自噬被认为可以维持成人弥漫性神经胶质瘤的干性表型,自噬和干性之间联系的分子基础知之甚少,这使得无法有效筛选将受益于自噬靶向治疗的人群。这里,ATG9B对于自我更新能力和肿瘤传播潜力是必需的。值得注意的是,ASCL2转录调节ATG9B的表达以维持干性特性。ASCL2-ATG9B轴是自噬活性的独立预后生物标志物和指标。此外,研究了高效的血脑屏障(BBB)通透性自噬抑制剂ROC-325,该抑制剂可以显着抑制ASCL2-ATG9B轴高胶质瘤的进展。这些数据表明,新的ASCL2-ATG9B信号轴对于维持干细胞表型和肿瘤进展至关重要。揭示了成人弥漫性神经胶质瘤潜在的自噬抑制策略。
    Autophagy is a highly conserved process that is vital for tumor progression and treatment response. Although autophagy is proposed to maintain the stemness phenotype in adult diffuse glioma, the molecular basis of the link between autophagy and stemness is poorly understood, which makes it impossible to effectively screen for the population that will benefit from autophagy-targeted treatment. Here, ATG9B as essential for self-renewal capacity and tumor-propagation potential is identified. Notably, ASCL2 transcriptionally regulates the expression of ATG9B to maintain stemness properties. The ASCL2-ATG9B axis is an independent prognostic biomarker and indicator of autophagic activity. Furthermore, the highly effective blood-brain barrier (BBB)-permeable autophagy inhibitor ROC-325, which can significantly inhibit the progression of ASCL2-ATG9B axisHigh gliomas as a single agent is investigated. These data demonstrate that a new ASCL2-ATG9B signaling axis is crucial for maintaining the stemness phenotype and tumor progression, revealing a potential autophagy inhibition strategy for adult diffuse gliomas.
    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:耐药性是影响结肠癌患者化疗疗效的重要因素。然而,诊断肿瘤细胞耐药性的临床标志物不仅数量少,但特异性也很低,肿瘤细胞耐药的作用机制尚不清楚。
    方法:使用癌症基因组图谱(TCGA)和基因型-组织表达泛癌症数据分析二肽酶1(DPEP1)表达。使用R软件中的生存包进行生存分析以评估DPEP1表达在结肠癌中的预后价值。采用相关性和维恩分析来鉴定关键基因。免疫组织化学,westernblot,qRT-PCR,免疫共沉淀,并进行了双荧光素酶报告基因实验,以探索DPEP1和Achaetescute-like2(ASCL2)之间的潜在关联。MTT法评价DPEP1和ASCL2在结肠癌耐药中的作用。
    结果:DPEP1在结肠癌组织中高表达。DPEP1表达与疾病特异性生存率呈负相关,但与总体生存率无关。生物信息学分析和实验表明,DPEP1和ASCL2在结肠癌组织中的表达呈显著正相关。机制研究表明,DPEP1通过抑制ASCL2蛋白泛素化介导的降解增强其稳定性。反过来,ASCL2充当转录因子以激活DPEP1基因的转录活性并增强其表达。此外,DPEP1还可以增强结肠癌干细胞标志物(LGR5,CD133和CD44)的表达,这增强了结肠癌细胞对化疗药物的耐受性。
    结论:我们的发现表明,DPEP1通过与ASCL2形成正反馈回路来增强肿瘤细胞的干性,从而改善对化疗药物的耐药性。
    Drug resistance is an important factor affecting the efficacy of chemotherapy in patients with colon cancer. However, clinical markers for diagnosing drug resistance of tumor cells are not only a few in number, but also low in specificity, and the mechanism of action of tumor cell drug resistance remains unclear.
    Dipeptidase 1 (DPEP1) expression was analyzed using the cancer genome atlas (TCGA) and genotype-Tissue Expression pan-cancer data. Survival analysis was performed using the survival package in R software to assess the prognostic value of DPEP1 expression in colon cancer. Correlation and Venn analyses were adopted to identify key genes. Immunohistochemistry, western blot, qRT-PCR, Co-immunoprecipitation, and dual-luciferase reporter experiments were carried out to explore the underlying associations between DPEP1 and Achaete scute-like 2 (ASCL2). MTT assays were used to evaluate the role of DPEP1 and ASCL2 in colon cancer drug resistance.
    DPEP1 was highly expressed in colon cancer tissues. DPEP1 expression correlated negatively with disease-specific survival but not with overall survival. Bioinformatics analysis and experiments showed that the expressions of DPEP1 and ASCL2 in colon cancer tissues were markedly positively correlated. Mechanistic research indicated that DPEP1 enhanced the stability of protein ASCL2 by inhibiting its ubiquitination-mediated degradation. In turn, ASCL2 functioned as a transcription factor to activate the transcriptional activity of the DPEP1 gene and boost its expression. Furthermore, DPEP1 also could enhance the expression of colon cancer stem cell markers (LGR5, CD133, and CD44), which strengthened the tolerance of colon cancer cells to chemotherapy drugs.
    Our findings reveal that the DPEP1 enhances the stemness of tumor cells by forming a positive feedback loop with ASCL2 to improve resistance to chemotherapy drugs.
    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
    Ascl2已被证明参与结直肠癌(CRC)的肿瘤发生,尽管其表观遗传调控机制在很大程度上是未知的。这里,我们发现,在CRC细胞中,与Ascl2位点的其他区域相比,Ascl2启动子(bp-1670~-1139)的甲基化显著增加,并且与升高的Ascl2mRNA表达相关。此外,在分析DNA甲基化数据后,我们发现启动子甲基化可预测CRC患者的生存,RNA-Seq数据,和来自MethHC数据库的410例CRC患者样本的临床数据,MEXPRESS数据库,和Cbioportal网站。利用建立的TET甲基胞嘧啶双加氧酶2(TET2)敲低和异位TET2催化结构域表达细胞模型,我们进行了糖基化羟甲基敏感定量PCR(qPCR),实时PCR,和蛋白质印迹分析,以进一步证实Ascl2启动子的超甲基化,CRC细胞中Ascl2表达升高的部分原因是TET2表达降低。此外,通过LC-MS/MS将BCLAF1鉴定为CRC细胞中的TET2相互作用剂,共免疫沉淀,免疫荧光共定位,和邻近连接测定。随后,我们发现TET2-BCLAF1复合物在Ascl2启动子处与CCGG位点周围的多个元件结合,并通过使用染色质免疫沉淀-qPCR和糖基化羟甲基-qPCR分析诱导其羟甲基化进一步抑制其超甲基化.最后,我们证明了在CRC细胞中TET2调节的Ascl2靶向茎基因表达独立于Wnt信号传导。一起来看,我们的数据表明,通过TET2-BCLAF1介导的启动子甲基化抑制CRC细胞中Ascl2表达的另一种选择,CRC祖细胞的Ascl2依赖性自我更新,和TET2-BCLAF1相关的CRC进展。
    Ascl2 has been shown to be involved in tumorigenesis in colorectal cancer (CRC), although its epigenetic regulatory mechanism is largely unknown. Here, we found that methylation of the Ascl2 promoter (bp -1670 ∼ -1139) was significantly increased compared to the other regions of the Ascl2 locus in CRC cells and was associated with elevated Ascl2 mRNA expression. Furthermore, we found that promoter methylation was predictive of CRC patient survival after analyzing DNA methylation data, RNA-Seq data, and clinical data of 410 CRC patient samples from the MethHC database, the MEXPRESS database, and the Cbioportal website. Using the established TET methylcytosine dioxygenase 2 (TET2) knockdown and ectopic TET2 catalytic domain-expression cell models, we performed glucosylated hydroxymethyl-sensitive quatitative PCR (qPCR), real-time PCR, and Western blot assays to further confirm that hypermethylation of the Ascl2 promoter, and elevated Ascl2 expression in CRC cells was partly due to the decreased expression of TET2. Furthermore, BCLAF1 was identified as a TET2 interactor in CRC cells by LC-MS/MS, coimmunoprecipitation, immunofluorescence colocalization, and proximity ligation assays. Subsequently, we found the TET2-BCLAF1 complex bound to multiple elements around CCGG sites at the Ascl2 promoter and further restrained its hypermethylation by inducing its hydroxymethylation using chromatin immunoprecipitation-qPCR and glucosylated hydroxymethyl-qPCR assays. Finally, we demonstrate that TET2-modulated Ascl2-targeted stem gene expression in CRC cells was independent of Wnt signaling. Taken together, our data suggest an additional option for inhibiting Ascl2 expression in CRC cells through TET2-BCLAF1-mediated promoter methylation, Ascl2-dependent self-renewal of CRC progenitor cells, and TET2-BCLAF1-related CRC progression.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • DOI:
    文章类型: Journal Article
    神经干细胞(NSC)可能提供有益和有希望的辅助治疗神经系统疾病,如阿尔茨海默病(AD),帕金森病(PD)和脊髓损伤。先前的研究表明,LncRNAFER1L4在许多生物学程序中起着至关重要的作用,例如入侵,新陈代谢,凋亡,和干细胞分化。然而,FER1L4在神经干细胞分化和生长中的作用尚不清楚.在目前的研究中,我们注意到FER1L4在TNFα诱导的神经干细胞中上调。FER1L4的异位表达抑制NSC增殖并诱导NSC分化成神经元和星形胶质细胞。使用Starbase在线软件,我们确定FER1L4是miR-874-3p的一个潜在靶基因。FER1L4的异位表达降低了NSC中miR-874-3p的表达。我们鉴定了Ascl2是miR-874-3p的一个靶基因。FER1L4的过表达增强了NSC中的Ascl2表达。此外,我们证明FER1L4通过调节Ascl2调节神经干细胞的增殖和分化。
    Neural stem cells (NSCs) may offer beneficeial and promising adjuncts for treatment of neurological diseases such as Alzheimer\'s disease (AD), Parkinson\'s disease (PD) and spinal cord injuries. Previous studies showed that LncRNA FER1L4 plays crucial roles in many biological procedures such as invasion, metabolism, apoptosis, and stem cell differentiation. However, the role of FER1L4 in differentiation and growth of NSCs remains unknown. In the present research, we noted that FER1L4 is upregulated in NSCs induced with TNFα. Ectopic expression of FER1L4 suppresses NSCs proliferation and induces NSCs differentiated into neurons and astrocytes. Using Starbase online software, we identified that FER1L4 is one potential target gene of miR-874-3p. Ectopic expression of FER1L4 decreases miR-874-3p expression in NSCs. We identified Ascl2 is one target gene for miR-874-3p. Overexpression of FER1L4 enhances Ascl2 expression in NSCs. Furthermore, we proved that FER1L4 modulates the proliferation and differentiation of NSCs via regulating Ascl2.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号