SNAI2

SNAI2
  • 文章类型: Journal Article
    RNAN6-甲基腺苷(m6A)读取器介导癌症进展。然而,m6A阅读器在前列腺癌致瘤性中的功能作用和潜在机制仍有待阐明。在这项研究中,我们证明,YTHDF3表达在去势抵抗性前列腺癌(CRPC)中升高,并且与高级别呈正相关,骨转移和低生存率。YTHDF3表达促进CRPC细胞增殖,上皮间质转化(EMT)和肿瘤进展。机械上,YTHDF3促进SPOP和NXK3.1的RNA降解,但稳定了依赖于m6A的TWIST1和SNAI2的RNA表达,以促进细胞增殖和EMT。此外,YTHDF3表达以m6A依赖性方式通过降解SPOP增强AKT活性。重要的是,我们发现褪黑激素可以与m6A竞争占据YTHDF3的m6A结合笼,导致抑制YTHFD3及其靶表达以及CRPC肿瘤生长。我们的发现揭示了YTHDF3在CRPC进展中的重要作用,并强调了褪黑激素在抗CRPC活性中的作用。
    RNA N6-methyladenosine (m6A) readers mediate cancer progression. However, the functional role and potential mechanisms of the m6A readers in prostate cancer tumorigenicity remain to be elucidated. In this study, we demonstrate that YTHDF3 expression is elevated in castration-resistant prostate cancer (CRPC) and positively correlated to high grade, bone metastasis and poor survival. YTHDF3 expression promoted CRPC cell proliferation, epithelial to mesenchymal transition (EMT) and tumour progression. Mechanistically, YTHDF3 promoted the RNA degradation of SPOP and NXK3.1 but stabilized RNA expressions of TWIST1 and SNAI2 dependent on m6A to facilitate cell proliferation and EMT. Additionally, YTHDF3 expression enhanced AKT activity via degrading SPOP in an m6A-dependent manner. Importantly, we found that melatonin can compete with m6A to occupy the m6A-binding cage of YTHDF3, leading to inhibition of YTHFD3 and its target expressions as well as CRPC tumour growth. Our findings uncover an essential role of YTHDF3 in the progression of CRPC and highlight the role of melatonin in anti-CRPC activity.
    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:肺癌是一种常见的恶性肿瘤,具有很高的发病率和死亡率。氨基葡萄糖6-磷酸N-乙酰转移酶(GNPNAT1),它是己糖胺生物合成途径(HBP)中的关键酶,已被鉴定为转移相关基因,并在肺腺癌(LUAD)中上调。然而,GNPNAT1在LUAD转移中的作用及相关机制尚不清楚。
    方法:我们分析了GNPNAT1在公共数据库中的表达,并通过免疫组织化学(IHC)证实了结果。基于癌症基因组图谱(TCGA)研究了GNPNAT1在LUAD中的生物学功能。通过使用表达数据(ESTIMATE)估计恶性肿瘤组织中的基质细胞和免疫细胞和通过估计RNA转录物的相对子集(CIBERSORT)R包来进行细胞类型鉴定,分析GNPNAT1与癌症免疫特征之间的相关性。GNPNAT1表达改变对LUAD细胞肿瘤发生的潜在机制,扩散,迁移,入侵,和转移进行了体外和体内探索。
    结果:我们证明GNPNAT1表达在LUAD中显著增加,并且与患者的总生存期(OS)呈负相关。hsa-miR-1-3p和hsa-miR-26a-5p被鉴定为GNPNAT1的上游miRNA靶标。GNPNAT1与CD8T细胞的浸润水平有关,记忆激活的CD4T细胞,NK细胞静息,巨噬细胞M0,巨噬细胞M1,中性粒细胞,γδT细胞,和嗜酸性粒细胞,虽然它与记忆静息CD4T细胞呈负相关,调节性T细胞(Tregs),静息NK细胞,单核细胞,静息树突状细胞,和静止的肥大细胞。GNPNAT1敲低显著抑制增殖,迁移,入侵,上皮-间质转化(EMT)过程,和LUAD细胞的转移,而GNPNAT1的过表达显示出相反的作用。拯救实验显示Snai2敲低可逆转GNPNAT1诱导的LUAD细胞迁移,入侵,EMT。机械上,GNPNAT1通过抑制LUAD中Snai2的泛素化降解促进癌细胞转移。
    结论:综合来看,这些数据表明GNPNAT1是LUAD患者的预后生物标志物.此外,GNPNAT1对于促进LUAD细胞的肿瘤发生和转移至关重要,并且可能是预防LUAD转移的潜在治疗靶标。
    BACKGROUND: Lung cancer is a common malignant tumor with high morbidity and mortality rate. Glucosamine 6-phosphate N-acetyltransferase (GNPNAT1), which serves as a critical enzyme in hexosamine biosynthetic pathway (HBP), has been identified as a metastasis-associated gene and is upregulated in lung adenocarcinoma (LUAD). However, the exact role and related mechanism of GNPNAT1 in LUAD metastasis remain unknown.
    METHODS: We analyzed the expression of GNPNAT1 in the public databases and confirmed the results by immunohistochemistry (IHC). The biological functions of GNPNAT1 in LUAD were investigated based on The Cancer Genome Atlas (TCGA). Correlations between GNPNAT1 and cancer immune characteristics were analyzed via the Estimation of Stromal and Immune cells in Malignant Tumor tissues using Expression data (ESTIMATE) and Cell-type Identification by Estimating Relative Subsets of RNA Transcript (CIBERSORT) R package. The underlying mechanisms of altered GNPNAT1 expression on LUAD cell tumorigenesis, proliferation, migration, invasion, and metastasis were explored in vitro and in vivo.
    RESULTS: We demonstrated that GNPNAT1 expression was significantly increased in LUAD and negatively associated with the overall survival (OS) of patients. hsa-miR-1-3p and hsa-miR-26a-5p were identified as upstream miRNA targets of GNPNAT1. GNPNAT1 was associated with the infiltration levels of CD8 T cells, memory-activated CD4 T cells, NK cells resting, macrophages M0, macrophages M1, neutrophils, gamma delta T cells, and eosinophils, while it was negatively correlated with memory-resting CD4 T cells, regulatory T cells (Tregs), resting NK cells, monocytes, resting dendritic cells, and resting mast cells. GNPNAT1 knockdown significantly inhibited proliferation, migration, invasion, epithelial-mesenchymal transition (EMT) process, and metastasis of LUAD cells, while overexpression of GNPNAT1 revealed the opposite effects. Rescue assay showed that Snai2 knockdown reversed GNPNAT1-induced LUAD cells migration, invasion, and EMT. Mechanistically, GNPNAT1 promoted cancer cell metastasis via repressing ubiquitination degradation of Snai2 in LUAD.
    CONCLUSIONS: Taken together, these data indicate that GNPNAT1 serves as a prognostic biomarker for LUAD patient. Additionally, GNPNAT1 is critical for promoting tumorigenesis and metastasis of LUAD cells and may be a potential therapeutic target for preventing LUAD metastasis.
    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:从DNA甲基化角度探讨miR-181a-5p在急性肾损伤(AKI)进展为肾间质纤维化(RIF)中的作用。材料与方法:通过收集临床样本证实miR-181a-5p的作用,将miR-181a-5p阿戈米尔注射到尾静脉,并体外转染miR-181a-5p模拟物。通过甲基化特异性PCR研究miR-181a-5p对AKI诱导的RIF的影响机制。生物信息学分析,转录组测序等。结果:MiR-181a-5p在AKI诱导的RIF中起重要作用。DNMT3b介导的miR-181a-5p启动子甲基化是miR-181a-5p下调的主要原因。HDAC9和SNAI2是miR-181a-5p的直接靶标。结论:DNMT3b介导的miR-181a-5p启动子超甲基化通过靶向HDAC9和SNAI2促进AKI诱导的RIF。
    [方框:见正文]。
    Aim: To explore the role of miR-181a-5p in the progression of acute kidney injury (AKI) to renal interstitial fibrosis (RIF) from the perspective of DNA methylation. Materials & methods: The role of miR-181a-5p was confirmed by collecting clinical samples, injecting miR-181a-5p agomir into tail vein, and transfecting miR-181a-5p mimic in vitro. The mechanism of miR-181a-5p\'s influence on AKI induced RIF was investigated by methylation-specific PCR, bioinformatic analysis, transcriptome sequencing and so on. Results: MiR-181a-5p plays an important role in AKI induced RIF. DNMT3b-mediated miR-181a-5p promoter hypermethylation is the main reason for the downregulation of miR-181a-5p. HDAC9 and SNAI2 are direct targets of miR-181a-5p. Conclusion: Hypermethylation of miR-181a-5p promoter mediated by DNMT3b promotes AKI induced RIF by targeting HDAC9 and SNAI2.
    [Box: see text].
    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
    盆腔器官脱垂(POP)是一组由盆腔支持组织中的细胞外基质(ECM)降解引起的疾病。富含半胱氨酸和丝氨酸的核蛋白1(CSRNP1)参与细胞增殖和存活调控,据报道促进人软骨细胞中的胶原蛋白分解。本研究旨在探讨CSRNP1对人源阴道成纤维细胞胶原代谢的影响。与正常来源的阴道成纤维细胞相比,在POP患者来源的阴道成纤维细胞中发现CSRNP1的高表达。功能实验表明,CSRNP1过表达导致增殖抑制,正常阴道成纤维细胞的凋亡和胶原降解。与此相符,CSRNP1沉默抑制过氧化氢(H2O2)引发的细胞凋亡,正常阴道成纤维细胞中的ROS产生和胶原损失。CSRNP1的沉默也降低了细胞衰老标记p21和γ-H2Ax(在Ser139磷酸化的组蛋白H2Ax)的表达,以及抑制由DNA损伤剂依托泊苷引起的正常阴道成纤维细胞的胶原蛋白分解。对阴道成纤维细胞的转录组学分析表明,受CSRNP1过表达影响的差异表达基因主要富集在Wnt信号通路中。用Wnt途径抑制剂DKK1治疗阻断了CSRNP1敲低引起的胶原沉积。机械上,CSRNP1被鉴定为Snail家族转录抑制因子2(SNAI2)的靶标。CSRNP1的强制表达逆转了抗凋亡,SNAI2在暴露于H2O2或依托泊苷的正常阴道成纤维细胞中的抗衰老和抗胶原蛋白损失作用。我们的研究表明,SNAI2/CSRNP1轴可能是POP进展的关键驱动因素,这为POP提供了潜在的治疗策略。
    Pelvic organ prolapse (POP) is a group of diseases caused by extracellular matrix (ECM) degradation in pelvic supportive tissues. Cysteine and serine rich nuclear protein 1 (CSRNP1) is involved in cell proliferation and survival regulation, and reportedly facilitates collagen breakdown in human chondrocytes. The present study aimed to probe the effect of CSRNP1 on collagen metabolism in human-derived vaginal fibroblasts. High expression of CSRNP1 was found in POP patient-derived vaginal fibroblasts in comparison to normal-derived vaginal fibroblasts. Following functional experiments revealed that CSRNP1 overexpression led to proliferation inhibition, apoptosis and collagen degradation in normal vaginal fibroblasts. In line with this, silencing of CSRNP1 inhibited hydrogen peroxide (H2O2)-triggered apoptosis, ROS generation and collagen loss in normal vaginal fibroblasts. Silencing of CSRNP1 also reduced the expression of cell senescence markers p21 and γ-H2Ax (the histone H2Ax phosphorylated at Ser139), as well as curbed collagen breakdown in normal vaginal fibroblasts caused by a DNA damage agent etoposide. Transcriptomic analysis of vaginal fibroblasts showed that differentially expressed genes affected by CSRNP1 overexpression were mainly enriched in the Wnt signaling pathway. Treatment with a Wnt pathway inhibitor DKK1 blocked CSRNP1 knockdown-caused collagen deposition. Mechanistically, CSRNP1 was identified to be a target of Snail family transcriptional repressor 2 (SNAI2). Forced expression of CSRNP1 reversed the anti-apoptotic, anti-senescent and anti-collagen loss effects of SNAI2 in normal vaginal fibroblasts exposed to H2O2 or etoposide. Our study indicates that the SNAI2/CSRNP1 axis may be a key driver in POP progression, which provides a potential therapeutic strategy for POP.
    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:横纹肌肉瘤(RMS)是一种罕见的恶性肿瘤,是儿童中最常见的软组织肉瘤。血管生成拟态(VM)是一种不同于传统肿瘤血管生成的新型肿瘤微循环模型,它不依赖内皮细胞为肿瘤生长提供足够的血液供应。近年来,VM已被证实与肿瘤进展密切相关。然而,RMS形成VM的能力尚未报告。
    方法:免疫组织化学,RT-qPCR和Westernblot检测SNAI2的表达水平及其临床意义。在体外和体内检查了调节SNAI2的血管生成拟态和恶性进展的生物学功能。质谱,联合免疫组织化学,免疫荧光染色,并进行泛素检测以探讨SNAI2的调控机制。
    结果:我们的研究表明SNAI2在RMS患者和RMS细胞系中异常表达,并促进RMS的增殖和转移。通过细胞小管形成实验,裸鼠基质胶塞实验,和免疫组织化学(IHC),我们证实RMS可以形成VM,SNAI2促进VM的形成。由于SNAI2是一种转录因子,不容易被下药,我们使用Co-IP结合质谱来筛选SNAI2结合蛋白USP7和TRIM21.USP7耗尽抑制RMSVM形成,通过促进SNAI2降解进行增殖和转移。我们进一步证明了TRIM21在人RMS组织中以低水平表达并抑制RMS细胞中的VM。TRIM21通过RMS中的泛素化促进SNAI2蛋白降解。去泛素化酶USP7和E3连接酶TRIM21以拮抗而非竞争模式起作用,并在控制SNAI2的稳定性以确定RMS的VM形成和进展中起关键作用。
    结论:我们的发现揭示了一种以前未知的机制,USP7和TRIM21平衡SNAI2泛素化水平,确定RMS血管生成拟态,扩散,和移民。这种新机制可能通过恢复TRIM21表达或抑制具有高SNAI2蛋白水平的RMS患者的USP7表达来提供新的靶向治疗以抑制RMS的发展。
    BACKGROUND: Rhabdomyosarcoma (RMS) is a rare malignancy and the most common soft tissue sarcoma in children. Vasculogenic mimicry (VM) is a novel tumor microcirculation model different from traditional tumor angiogenesis, which does not rely on endothelial cells to provide sufficient blood supply for tumor growth. In recent years, VM has been confirmed to be closely associated with tumor progression. However, the ability of RMS to form VM has not yet been reported.
    METHODS: Immunohistochemistry, RT-qPCR and western blot were used to test the expression level of SNAI2 and its clinical significance. The biological function in regulating vasculogenic mimicry and malignant progression of SNAI2 was examined both in vitro and in vivo. Mass spectrometry, co-immunohistochemistry, immunofluorescence staining, and ubiquitin assays were performed to explore the regulatory mechanism of SNAI2.
    RESULTS: Our study indicated that SNAI2 was abnormally expressed in patients with RMS and RMS cell lines and promoted the proliferation and metastasis of RMS. Through cell tubule formation experiments, nude mice Matrigel plug experiments, and immunohistochemistry (IHC), we confirmed that RMS can form VM and that SNAI2 promotes the formation of VM. Due to SNAI2 is a transcription factor that is not easily drugged, we used Co-IP combined with mass spectrometry to screen for the SNAI2-binding protein USP7 and TRIM21. USP7 depletion inhibited RMS VM formation, proliferation and metastasis by promoting SNAI2 degradation. We further demonstrated that TRIM21 is expressed at low levels in human RMS tissues and inhibits VM in RMS cells. TRIM21 promotes SNAI2 protein degradation through ubiquitination in the RMS. The deubiquitinase USP7 and E3 ligase TRIM21 function in an antagonistic rather than competitive mode and play a key role in controlling the stability of SNAI2 to determine the VM formation and progression of RMS.
    CONCLUSIONS: Our findings reveal a previously unknown mechanism by which USP7 and TRIM21 balance the level of SNAI2 ubiquitination, determining RMS vasculogenic mimicry, proliferation, and migration. This new mechanism may provide new targeted therapies to inhibit the development of RMS by restoring TRIM21 expression or inhibiting USP7 expression in RMS patients with high SNAI2 protein levels.
    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
    肌动蛋白是对许多细胞功能至关重要的蛋白质。其定位和活性受到与大量肌动蛋白结合蛋白的相互作用的调节。在酵母双杂交(Y2H)筛选系统中,蜗牛家族转录抑制因子2(SNAI2或slug)被鉴定为未知的潜在肌动蛋白结合蛋白。我们使用免疫沉淀验证了这种相互作用,并分析了slug和肌动蛋白之间的功能关系。由于两种蛋白质都被报道参与DNA双链断裂(DSB)修复,在使用阿霉素或紫外线照射治疗后,我们集中研究了它们在此过程中的相互作用。共聚焦显微镜显示,与NLS融合的肌动蛋白的过表达稳定了slug和γH2AX的复合物,DNA损伤修复的早期标记。
    Actin is a protein of central importance to many cellular functions. Its localization and activity are regulated by interactions with a high number of actin-binding proteins. In a yeast two-hybrid (Y2H) screening system, snail family transcriptional repressor 2 (SNAI2 or slug) was identified as a yet unknown potential actin-binding protein. We validated this interaction using immunoprecipitation and analyzed the functional relation between slug and actin. Since both proteins have been reported to be involved in DNA double-strand break (DSB) repair, we focused on their interaction during this process after treatment with doxorubicin or UV irradiation. Confocal microscopy elicits that the overexpression of actin fused to an NLS stabilizes complexes of slug and γH2AX, an early marker of DNA damage repair.
    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
    肝损伤刺激肝细胞复制和肝星状细胞(HSC)激活,从而驱动肝脏再生。异常HSC活化诱导肝纤维化。然而,肝脏再生和纤维化的潜在机制仍然知之甚少。这里,我们确定肝Snai1和Snai2是肝再生和纤维化的重要转录调节因子。部分肝切除术或CCl4治疗会增加Snai1和Snai2在肝脏中细胞周期蛋白A2和D1启动子上的发生率。Snai1和Snai2依次增加启动子H3K27乙酰化和细胞周期蛋白A2/D1表达。Snai1和Snai2的肝细胞特异性缺失,但不是单独的,抑制肝切除术或CCl4治疗后的细胞周期蛋白A2/D1表达和再生肝细胞增殖,但增加CCl4刺激的HSC激活和肝纤维化。相反,在CCl4治疗后,肝脏中的Snai2过表达增强肝细胞复制并抑制肝纤维化。这些结果表明,肝Snai1和Snai2直接促进,通过组蛋白修饰,修复肝细胞复制和间接抑制肝纤维化。
    Liver injury stimulates hepatocyte replication and hepatic stellate cell (HSC) activation, thereby driving liver regeneration. Aberrant HSC activation induces liver fibrosis. However, mechanisms underlying liver regeneration and fibrosis remain poorly understood. Here, we identify hepatic Snai1 and Snai2 as important transcriptional regulators for liver regeneration and fibrosis. Partial hepatectomy or CCl4 treatment increases occupancies of Snai1 and Snai2 on cyclin A2 and D1 promoters in the liver. Snai1 and Snai2 in turn increase promoter H3K27 acetylation and cyclin A2/D1 expressions. Hepatocyte-specific deletion of both Snai1 and Snai2, but not one alone, suppresses liver cyclin A2/D1 expression and regenerative hepatocyte proliferation after hepatectomy or CCl4 treatments but augments CCl4-stimulated HSC activation and liver fibrosis. Conversely, Snai2 overexpression in the liver enhances hepatocyte replication and suppresses liver fibrosis after CCl4 treatment. These results suggest that hepatic Snai1 and Snai2 directly promote, via histone modifications, reparative hepatocyte replication and indirectly inhibit liver fibrosis.
    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
    叉头盒M1(FOXM1)的异常表达与帕金森病(PD)的病理过程有关,但上游和下游监管机构仍然知之甚少。本研究旨在研究FOXM1在PD多巴胺能神经元损伤中的潜在机制。进行生物信息学分析以查明FOXM1的差异表达,该差异表达在鱼藤酮损伤小鼠的黑色组织和多巴胺能神经元MN9D细胞中得到了验证。分析了SP1、FOXM1、SNAI2和CXCL12之间的相互作用。为了评估它们对多巴胺能神经元损伤的影响,将慢病毒载体介导的FOXM1,SP1和CXCL12操作导入鱼藤酮损伤小鼠和MN9D细胞.SP1、FOXM1、SNAI2和CXCL12在鱼藤酮损伤小鼠和MN9D细胞中大量表达。沉默FOXM1可延缓鱼藤酮诱导的多巴胺能神经元损伤。机械上,SP1是FOXM1的上游转录因子,上调FOXM1表达,导致SNAI2和CXCL12表达增加。在体内,数据证实SP1通过激活FOXM1/SNAI2/CXCL12轴促进多巴胺能神经元损伤.我们的数据表明,SP1沉默对多巴胺能神经元有神经保护作用,这取决于失活的FOXM1/SNAI2/CXCL12轴。
    The aberrant expression of Forkhead box M1 (FOXM1) has been associated with the pathological processes of Parkinson\'s disease (PD), but the upstream and downstream regulators remain poorly understood. This study sought to examine the underlying mechanism of FOXM1 in dopaminergic neuron injury in PD. Bioinformatics analysis was conducted to pinpoint the differential expression of FOXM1, which was verified in the nigral tissues of rotenone-lesioned mice and dopaminergic neuron MN9D cells. Interactions among SP1, FOXM1, SNAI2, and CXCL12 were analyzed. To evaluate their effects on dopaminergic neuron injury, the lentiviral vector-mediated manipulation of FOXM1, SP1, and CXCL12 was introduced in rotenone-lesioned mice and MN9D cells. SP1, FOXM1, SNAI2, and CXCL12 abundant expression occurred in rotenone-lesioned mice and MN9D cells. Silencing of FOXM1 delayed the rotenone-induced dopaminergic neuron injury in vitro. Mechanistically, SP1 was an upstream transcription factor of FOXM1 and upregulated FOXM1 expression, leading to increased SNAI2 and CXCL12 expression. In vivo, data confirmed that SP1 promoted dopaminergic neuron injury by activating the FOXM1/SNAI2/CXCL12 axis. Our data indicate that SP1 silencing has neuroprotective effects on dopaminergic neurons, which is dependent upon the inactivated FOXM1/SNAI2/CXCL12 axis.
    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
    结直肠癌(CRC)是一种肠癌,每年导致600,000多人死亡。克服转移问题需要详细的研究来揭示潜在的分子机制。本研究旨在揭示非编码RNA调控结直肠癌转移的分子机制。基于TCGA-COAD患者队列的数据分析FTH1P3的表达谱。进行Q-PCR分析以验证FTH1P3在结直肠癌细胞中的表达。使用JASPR筛选FTH1P3转录因子。q-ChIP分析用于验证FTH1P3和转录因子之间的靶标。采用划痕试验和transwell试验评价结直肠癌细胞的迁移和侵袭能力。FTH1P3在CRC患者队列中高表达。FTH1P3通过调控上皮间质转化(EMT)诱导SW480细胞迁移和侵袭。此外,FTH1P3是SNAI2的直接靶标。SNAI2促进FTH1P3的表达。FTH1P3和SNAI2都被miR-218-5p直接靶向和抑制。有趣的是,异位FTH1P3导致miR-218-5p水平降低和核酸SNAI2蛋白表达水平升高。值得注意的是,只有异位SNAI2蛋白导致抑制的miR-218-5p和增加的FTH1P3,而SNAI23'UTR未能影响miR-218-5p和FTH1P3的表达。SNAI2转录激活FTH1P3表达。SNAI2和FTH1P3都是miR-218-5p的靶标。SNAI2/FTH1P3/miR-218-5p在调控CRC转移中形成正反馈回路。
    Colorectal cancer (CRC) is a type of intestinal cancer that causes more than 600,000 deaths every year. Overcoming the problems of metastasis requires detailed studies to reveal the potential molecular mechanisms. This study aims to reveal the molecular mechanism of CRC metastasis involving non-coding RNA regulation. The expression profile of FTH1P3 was analyzed based on the data of TCGA-COAD patient cohorts. Q-PCR analysis was performed to validate the expression of FTH1P3 in colorectal cancer cells. JASPR was used to screen transcription factors of FTH1P3. q-ChIP analysis was used to validate the target between FTH1P3 and transcription factor. Scratch assay and transwell assay were used to evaluate the migration and invasion ability of colorectal cancer cells. FTH1P3 is highly expressed in CRC patient cohort. FTH1P3 induced migration and invasion of SW480 cell through regulating epithelial-mesenchymal transition (EMT). In addition, FTH1P3 is a direct target of SNAI2. SNAI2 promotes the expression of FTH1P3. Both FTH1P3 and SNAI2 were directly targeted and repressed by miR-218-5p. Interestingly, ectopic FTH1P3 caused a decreased miR-218-5p level and an elevated nucleic SNAI2 protein expression level. Of note, only ectopic SNAI2 protein resulted in a repressed miR-218-5p and an increased FTH1P3, whereas SNAI2 3\'UTR failed to affect the expression of miR-218-5p and FTH1P3. SNAI2 transcriptionally activates FTH1P3 expression. Both SNAI2 and FTH1P3 are targets of miR-218-5p. SNAI2/FTH1P3/miR-218-5p form a positive feedback loop in the regulation of CRC metastasis.
    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
    横纹肌肉瘤(RMS)是一种儿科肌肉恶性肿瘤,具有分化受阻的细胞特征。NOTCH1是促进自我更新并阻断融合阴性RMS亚型分化的癌基因。然而,NOTCH1表达如何在肿瘤中转录维持尚不清楚.SNAI2和CTCF染色质结合的分析以及HiC分析显示,NOTCH1基因座下游的保守SNAI2/CTCF重叠峰标记了亚拓扑关联域(TAD)边界。SNAI2-CTCF峰的缺失表明它对于FN-RMS细胞的NOTCH1表达和活力是必需的。在SNAI2-CTCF峰缺失的细胞中重新引入组成型活化的NOTCH1-ΔE可恢复细胞活力。使用CRISPR/Cas9试剂的SNAI2的消融导致大部分RD和SMS-CTRFN-RMS细胞的损失。然而,少数存活的重新繁殖培养物的克隆已经恢复了NOTCH1。SNAI2消融后重新建立NOTCH1表达的细胞不能像SNAI2shRNA敲低细胞那样强烈分化;然而,SNAI2消融的细胞继续对电离辐射非常敏感。因此,我们发现了SNAI2和CTCF维持亚TAD边界促进而不是抑制NOTCH1表达的新机制。Further,我们证明SNAI2对放射后细胞凋亡的抑制独立于SNAI2/NOTCH1对自我更新和分化的影响。
    Rhabdomyosarcoma (RMS) is a pediatric malignancy of the muscle with characteristics of cells blocked in differentiation. NOTCH1 is an oncogene that promotes self-renewal and blocks differentiation in the fusion negative-RMS sub-type. However, how NOTCH1 expression is transcriptionally maintained in tumors is unknown. Analyses of SNAI2 and CTCF chromatin binding and HiC analyses revealed a conserved SNAI2/CTCF overlapping peak downstream of the NOTCH1 locus marking a sub-topologically associating domain (TAD) boundary. Deletion of the SNAI2-CTCF peak showed that it is essential for NOTCH1 expression and viability of FN-RMS cells. Reintroducing constitutively activated NOTCH1-ΔE in cells with the SNAI2-CTCF peak deleted restored cell-viability. Ablation of SNAI2 using CRISPR/Cas9 reagents resulted in the loss of majority of RD and SMS-CTR FN-RMS cells. However, the few surviving clones that repopulate cultures have recovered NOTCH1. Cells that re-establish NOTCH1 expression after SNAI2 ablation are unable to differentiate robustly as SNAI2 shRNA knockdown cells; yet, SNAI2-ablated cells continued to be exquisitely sensitive to ionizing radiation. Thus, we have uncovered a novel mechanism by which SNAI2 and CTCF maintenance of a sub-TAD boundary promotes rather than represses NOTCH1 expression. Further, we demonstrate that SNAI2 suppression of apoptosis post-radiation is independent of SNAI2/NOTCH1 effects on self-renewal and differentiation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号