E2F1 Transcription Factor

E2F1 转录因子
  • 文章类型: Journal Article
    RNA结合蛋白(RBP)是一类主要通过与不同类型的RNA相互作用而起作用的蛋白质,并在调节癌症相关基因的转录和翻译中起关键作用。然而,它们在肝细胞癌(HCC)进展中的作用尚不清楚。在这项研究中,我们分析了肝癌患者的RNA测序数据和相应的临床信息,以筛选预后RBPs.胰岛素样生长因子2mRNA结合蛋白3(IGF2BP3)被确定为肝癌的独立预后因素。它在HCC中上调,并与不良预后相关。通过使用HCC患者的组织微阵列的免疫组织化学分析来验证IGF2BP3表达的升高。IGF2BP3敲低抑制Hep3B和HepG2细胞增殖,而IGF2BP3过表达促进HuH-7和MHCC97H细胞的扩增。机械上,IGF2BP3通过调节E2F1表达来调节细胞增殖。IGF2BP3基因的DNA低甲基化可能会增加IGF2BP3的表达,从而增强HCC的细胞增殖。因此,IGF2BP3可能作为一个新的预后生物标志物和肝癌的潜在治疗靶点。
    RNA-binding proteins (RBPs) are a class of proteins that primarily function by interacting with different types of RNAs and play a critical role in regulating the transcription and translation of cancer-related genes. However, their role in the progression of hepatocellular carcinoma (HCC) remains unclear. In this study, we analyzed RNA sequencing data and the corresponding clinical information of patients with HCC to screen for prognostic RBPs. Insulin-like growth factor 2 mRNA-binding protein 3 (IGF2BP3) was identified as an independent prognostic factor for liver cancer. It is upregulated in HCC and is associated with a poor prognosis. Elevated IGF2BP3 expression was validated via immunohistochemical analysis using a tissue microarray of patients with HCC. IGF2BP3 knockdown inhibited the proliferation of Hep3B and HepG2 cells, whereas IGF2BP3 overexpression promoted the expansion of HuH-7 and MHCC97H cells. Mechanistically, IGF2BP3 modulates cell proliferation by regulating E2F1 expression. DNA hypomethylation of the IGF2BP3 gene may increase the expression of IGF2BP3, thereby enhancing cell proliferation in HCC. Therefore, IGF2BP3 may act as a novel prognostic biomarker and a potential therapeutic target for HCC.
    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:乳腺癌是女性最常见的恶性肿瘤之一。细胞分裂周期相关5(CDCA5),姐妹染色单体内聚力的主要调节剂,据报道在几种类型的癌症中上调。这里,探讨CDCA5在乳腺癌中的作用及调控机制。
    方法:通过免疫组织化学染色在乳腺癌标本中鉴定CDCA5的表达。采用组织芯片技术分析CDCA5表达与乳腺癌患者临床病理特征及预后的相关性。在CDCA5过表达/敲低的细胞和小鼠模型中探索了CDCA5在乳腺癌中的功能。共同IP,进行ChIP和双荧光素酶报告基因测定以阐明潜在的分子机制。
    结果:我们发现CDCA5在乳腺癌组织和细胞系中的表达水平更高,CDCA5的过度表达与乳腺癌患者的不良预后显著相关。此外,CDCA5敲低显著抑制增殖和迁移,同时在体外促进细胞凋亡。机械上,我们发现CDCA5在促进E2F转录因子1(E2F1)与叉头框M1(FOXM1)启动子的结合中起重要作用。此外,体外和体内数据显示FOXM1的消耗减轻了CDCA5过表达对乳腺癌的影响。此外,我们发现Wnt/β-catenin信号通路是CDCA5诱导乳腺癌进展所必需的.
    结论:我们建议CDCA5通过CDCA5/FOXM1/Wnt轴促进乳腺癌的进展,CDCA5可能成为乳腺癌治疗的新靶点。
    BACKGROUND: Breast cancer is one of the most common malignant tumors in women. Cell division cycle associated 5 (CDCA5), a master regulator of sister chromatid cohesion, was reported to be upregulated in several types of cancer. Here, the function and regulation mechanism of CDCA5 in breast cancer were explored.
    METHODS: CDCA5 expression was identified through immunohistochemistry staining in breast cancer specimens. The correlation between CDCA5 expression with clinicopathological features and prognosis of breast cancer patients was analyzed using a tissue microarray. CDCA5 function in breast cancer was explored in CDCA5-overexpressed/knockdown cells and mice models. Co-IP, ChIP and dual-luciferase reporter assay assays were performed to clarify underlying molecular mechanisms.
    RESULTS: We found that CDCA5 was expressed at a higher level in breast cancer tissues and cell lines, and overexpression of CDCA5 was significantly associated with poor prognosis of patients with breast cancer. Moreover, CDCA5 knockdown significantly suppressed the proliferation and migration, while promoted apoptosis in vitro. Mechanistically, we revealed that CDCA5 played an important role in promoting the binding of E2F transcription factor 1 (E2F1) to the forkhead box M1 (FOXM1) promoter. Furthermore, the data of in vitro and in vivo revealed that depletion of FOXM1 alleviated the effect of CDCA5 overexpression on breast cancer. Additionally, we revealed that the Wnt/β-catenin signaling pathway was required for CDCA5 induced progression of breast cancer.
    CONCLUSIONS: We suggested that CDCA5 promoted progression of breast cancer via CDCA5/FOXM1/Wnt axis, CDCA5 might serve as a novel therapeutic target for breast cancer 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
    作为泌尿系统第二常见的恶性肿瘤,肾细胞癌(RCC)的早期诊断标志物和治疗靶点的探索势在必行。大量研究表明,AURKB通过磷酸化下游底物促进肿瘤发展。然而,AURKB对肾透明细胞癌(ccRCC)进展的功能作用和调节机制尚不清楚.在目前的研究中,基于生物信息学分析,我们将AURKB鉴定为ccRCC进展的新关键基因。同时,我们观察到AURKB在ccRCC组织和细胞系中高表达,敲低ccRCC细胞中的AURKB在体外和体内抑制细胞增殖和迁移。确定CDC37为AURKB的激酶分子伴侣,ccRCC中的表型AURKB。AURKB/CDC37复合物通过在S67和S373处直接磷酸化MYC来介导MYC蛋白的稳定以促进ccRCC的发展。同时,我们证明了AURKB/CDC37复合物激活MYC转录CCND1,增强Rb磷酸化,并促进E2F1的发布,进而激活AURKB转录并在ccRCC中形成正前馈环。总的来说,我们的研究将AURKB确定为ccRCC的新标记,揭示了AURKB/CDC37复合物通过直接磷酸化MYC以增强其稳定性来促进ccRCC的新机制,首先提出了AURKB/E2F1正前馈环,突出显示AURKB可能是ccRCC的有希望的治疗靶标。
    As the second most common malignant tumor in the urinary system, renal cell carcinoma (RCC) is imperative to explore its early diagnostic markers and therapeutic targets. Numerous studies have shown that AURKB promotes tumor development by phosphorylating downstream substrates. However, the functional effects and regulatory mechanisms of AURKB on clear cell renal cell carcinoma (ccRCC) progression remain largely unknown. In the current study, we identified AURKB as a novel key gene in ccRCC progression based on bioinformatics analysis. Meanwhile, we observed that AURKB was highly expressed in ccRCC tissue and cell lines and knockdown AURKB in ccRCC cells inhibit cell proliferation and migration in vitro and in vivo. Identified CDC37 as a kinase molecular chaperone for AURKB, which phenocopy AURKB in ccRCC. AURKB/CDC37 complex mediate the stabilization of MYC protein by directly phosphorylating MYC at S67 and S373 to promote ccRCC development. At the same time, we demonstrated that the AURKB/CDC37 complex activates MYC to transcribe CCND1, enhances Rb phosphorylation, and promotes E2F1 release, which in turn activates AURKB transcription and forms a positive feedforward loop in ccRCC. Collectively, our study identified AURKB as a novel marker of ccRCC, revealed a new mechanism by which the AURKB/CDC37 complex promotes ccRCC by directly phosphorylating MYC to enhance its stability, and first proposed AURKB/E2F1-positive feedforward loop, highlighting AURKB may be a promising therapeutic target for ccRCC.
    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
    环状RNA(circularRNAs)已被认为是肿瘤发生中的关键调节因子,然而,大多数circRNAs在肝细胞癌(HCC)中的生物学功能和分子机制仍然难以捉摸。我们试图揭示circMYBL2在HCC中的表达谱和生物学作用。进行初始微阵列分析以探测HCC细胞中circMYBL2的表达谱,然后在HCC细胞系和组织中进行qRT-PCR分析,揭示了大约MYBL2的显著上调。随后进行实验以评估circMYBL2在HCC进展中的生物学功能。此外,生物信息学分析,qRT-PCR分析,荧光素酶报告基因测定,和蛋白质印迹分析用于研究circMYBL2、miR-1205和E2F1之间的相互作用。发现CircMYBL2在肿瘤组织和HCC细胞系中表现出明显的上调。circMYBL2的高表达增加了肝癌细胞的增殖和迁移,而大约MYBL2敲除会产生相反的效果。机械上,我们的结果表明,circMYBL2促进E2F1表达,并通过生成miR-1205促进HCC进展.我们的发现显示,circMYBL2通过circMYBL2/miR-1205/E2F1轴促进HCC进展,提示circMYBL2作为HCC治疗的新靶点或HCC的预后生物标志物的潜力。
    Circular RNAs (circRNAs) have been recognized as pivotal regulators in tumorigenesis, yet the biological functions as well as molecular mechanisms of the majority of circRNAs in hepatocellular carcinoma (HCC) remain elusive. We sought to unveil the expression profile and biological role of circMYBL2 in HCC. Initial microarray analyses were conducted to probe the expression profile of circMYBL2 in HCC cells, and qRT‒PCR analysis was then performed in HCC cell lines and tissues, revealing significant upregulation of circMYBL2. Subsequent experiments were conducted to evaluate the biological function of circMYBL2 in HCC progression. Furthermore, bioinformatics analysis, qRT‒PCR analysis, luciferase reporter assays, and western blot analysis were employed to investigate the interplay among circMYBL2, miR-1205, and E2F1. CircMYBL2 was found to exhibit marked upregulation in tumor tissues as well as HCC cell lines. Elevated expression of circMYBL2 increased the proliferation and migration of HCC cells, whereas circMYBL2 knockdown elicited contrasting effects. Mechanistically, our results indicated that circMYBL2 promoted E2F1 expression and facilitated HCC progression by sponging miR-1205. Our findings revealed that circMYBL2 contributed to HCC progression through the circMYBL2/miR-1205/E2F1 axis, suggesting the potential of circMYBL2 as a novel target for HCC treatment or a prognostic biomarker for HCC.
    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:肺腺癌(LUAD)是非小细胞癌。核糖核酸酶/血管生成素抑制剂1(RNH1)在有生命的癌症中发挥多种作用。E2F1是参与LUAD发育的关键转录因子。这里,我们分析了LUAD患者中RNH1的表达,研究RNH1在LUAD中的生物学功能,并通过E2F1在LUAD中展示其潜在机制。
    方法:在本研究中,我们基于数据库提供了RNH1在LUAD中的表达,并通过蛋白质印迹检测人LUAD组织中的RNH1来证实。构建慢病毒感染以沉默或过表达NCI-H1395和NCI-H1437细胞中的RNH1。我们通过MTT法评估RNH1对LUAD细胞增殖的作用,集落形成试验,和细胞周期检测。用Hoechst染色和流式细胞术评价RNH1对LUAD细胞凋亡的影响。通过Transwell实验研究了RNH1在侵袭和迁移中的功能。双荧光素酶测定,ChIP检测,并进行下拉实验以探讨E2F1在维持RNH1表达和功能中的相关性。探讨了E2F1对LUAD细胞RNH1功能的调控。进行小鼠实验以证实RNHl在LUAD中的体内作用。mRNA测序表明RNH1过表达改变了LUAD细胞的表达谱。
    结果:这项工作介绍了患者LUAD组织中RNH1的表达。重要的是,RNH1敲除增进了增殖,细胞的迁移和侵袭能力以及RNH1过表达产生了相反的作用。双荧光素酶实验证明,E2F1与RNH1启动子(-1064~-1054,-1514~-1504)结合,降低了RNH1的转录活性。ChIP分析表明,E2F1DNA富集在RNH1启动子(-1148~-943,-1628~-1423)。下拉法也显示了E2F1和RNH1启动子之间的关联(-1148~-943)。E2F1过表达有助于LUAD细胞的恶性行为,而RNH1过表达则逆转了它。高通量测序显示,RNH1过表达可诱导多基因表达改变,从而调节LUAD相关过程。
    结论:我们的研究表明,E2F1与RNH1启动子的结合可能导致RNH1表达的抑制,从而促进LUAD的发展。
    BACKGROUND: Lung adenocarcinoma (LUAD) is a non-small cell carcinoma. Ribonuclease/angiogenin inhibitor 1 (RNH1) exerts multiple roles in virous cancers. E2F1 is a critical transcription factor involved in the LUAD development. Here, we analyze the expression of RNH1 in LUAD patients, investigate the biological function of RNH1 in LUAD, and demonstrate its potential mechanisms through E2F1 in LUAD.
    METHODS: In the present study, we presented the expression of RNH1 in LUAD based on the database and confirmed it by western blot detection of RNH1 in human LUAD tissues. Lentiviral infection was constructed to silence or overexpress RNH1 in NCI-H1395 and NCI-H1437 cells. We assess the role of RNH1 on proliferation in LUAD cells by MTT assay, colony formation assays, and cell cycle detection. Hoechst staining and flow cytometry were used to evaluate the effects of RNH1 on apoptosis of LUAD cells. The function of RNH1 in invasion and migration was investigated by Transwell assay. Dual luciferase assay, ChIP detection, and pull-down assay were conducted to explore the association of E2F1 in the maintenance of RNH1 expression and function. The regulation of E2F1 on the functions of RNH1 in LUAD cells was explored. Mouse experiments were performed to confirm the in-vivo role of RNH1 in LUAD. mRNA sequencing indicated that RNH1 overexpression altered the expression profile of LUAD cells.
    RESULTS: RNH1 expression in LUAD tissues of patients was presented in this work. Importantly, RNH1 knockdown improved the proliferation, migration and invasion abilities of cells and RNH1 overexpression produced the opposite effects. Dual luciferase assay proved that E2F1 bound to the RNH1 promoter (-1064 ∼ -1054, -1514 ∼ -1504) to reduce the transcriptional activity of RNH1. ChIP assay indicated that E2F1 DNA was enriched at the RNH1 promoter (-1148 ∼ -943, -1628 ∼ -1423). Pull-down assays also showed the association between E2F1 and RNH1 promoter (-1148 ∼ -943). E2F1 overexpression contributed to the malignant behavior of LUAD cells, while RNH1 overexpression reversed it. High-throughput sequencing showed that RNH1 overexpression induced multiple genes expression changes, thereby modulating LUAD-related processes.
    CONCLUSIONS: Our study demonstrates that binding of E2F1 to the RNH1 promoter may lead to inhibition of RNH1 expression and thus promoting the development of 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
    高水平的H2A。Z促进黑色素瘤细胞增殖并与不良预后相关。然而,两种截然不同的H2A的作用。黑色素瘤中的Z组蛋白伴侣复合物SRCAP和P400-TIP60仍不清楚。这里,我们表明SRCAP的单个亚基耗尽,P400和VPS72(YL1)不仅导致H2A的损失。Z沉积到染色质中,但也减少了黑素瘤细胞中H4的乙酰化。H4乙酰化的这种丧失尤其在与H2A直接结合的细胞周期基因的启动子处发现。Z和它的监护人,提示H2A之间的协调调节。Z沉积和H4乙酰化促进它们的表达。三个亚基的击倒下调E2F1及其靶标,导致类似于H2A的细胞周期停滞。Z损耗。然而,不像H2A。Z缺乏,共享H2A的损失。Z伴侣亚基YL1诱导细胞凋亡。此外,YL1在黑素瘤组织中过度表达,其上调与患者预后不良有关。一起,这些发现为未来靶向H2A提供了理论基础.Z伴侣作为黑色素瘤治疗的表观遗传策略。
    High levels of H2A.Z promote melanoma cell proliferation and correlate with poor prognosis. However, the role of the two distinct H2A.Z histone chaperone complexes SRCAP and P400-TIP60 in melanoma remains unclear. Here, we show that individual subunit depletion of SRCAP, P400, and VPS72 (YL1) results in not only the loss of H2A.Z deposition into chromatin but also a reduction of H4 acetylation in melanoma cells. This loss of H4 acetylation is particularly found at the promoters of cell cycle genes directly bound by H2A.Z and its chaperones, suggesting a coordinated regulation between H2A.Z deposition and H4 acetylation to promote their expression. Knockdown of each of the three subunits downregulates E2F1 and its targets, resulting in a cell cycle arrest akin to H2A.Z depletion. However, unlike H2A.Z deficiency, loss of the shared H2A.Z chaperone subunit YL1 induces apoptosis. Furthermore, YL1 is overexpressed in melanoma tissues, and its upregulation is associated with poor patient outcome. Together, these findings provide a rationale for future targeting of H2A.Z chaperones as an epigenetic strategy for melanoma 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
    MCPH1已被确定为原发性小头畸形1型的致病基因,这是一种神经发育障碍,其特征是大脑尺寸减小和生长延迟。作为一种多功能蛋白质,据报道,MCPH1抑制TERT的表达并与转录调节因子E2F1相互作用。然而,目前尚不清楚MCPH1是否通过其转录调节功能调节大脑发育。这项研究表明,小鼠中Mcph1的敲除早在胚胎阶段E11.5就导致生长延迟。转录组分析(RNA-seq)显示,Mcph1的缺失导致有限数量基因的表达水平发生变化。虽然E2F1的一些表达靶点,例如Satb2和Cdkn1c,受到影响,差异表达基因(DEGs)作为E2F1靶基因没有显著富集。进一步的研究表明,原代和永生化的Mcph1敲除小鼠胚胎成纤维细胞(MEFs)表现出细胞周期停滞和细胞衰老表型。有趣的是,在Mcph1敲除MEF中检测到p19ARF的上调,沉默p19Arf可将细胞周期和生长停滞恢复到野生型水平。我们的研究结果表明,MCPH1不太可能通过E2F1介导的转录调节来调节神经发育。p19ARF依赖性细胞周期停滞和细胞衰老可能导致原发性小头畸形中观察到的发育异常。
    MCPH1 has been identified as the causal gene for primary microcephaly type 1, a neurodevelopmental disorder characterized by reduced brain size and delayed growth. As a multifunction protein, MCPH1 has been reported to repress the expression of TERT and interact with transcriptional regulator E2F1. However, it remains unclear whether MCPH1 regulates brain development through its transcriptional regulation function. This study showed that the knockout of Mcph1 in mice leads to delayed growth as early as the embryo stage E11.5. Transcriptome analysis (RNA-seq) revealed that the deletion of Mcph1 resulted in changes in the expression levels of a limited number of genes. Although the expression of some of E2F1 targets, such as Satb2 and Cdkn1c, was affected, the differentially expressed genes (DEGs) were not significantly enriched as E2F1 target genes. Further investigations showed that primary and immortalized Mcph1 knockout mouse embryonic fibroblasts (MEFs) exhibited cell cycle arrest and cellular senescence phenotype. Interestingly, the upregulation of p19ARF was detected in Mcph1 knockout MEFs, and silencing p19Arf restored the cell cycle and growth arrest to wild-type levels. Our findings suggested it is unlikely that MCPH1 regulates neurodevelopment through E2F1-mediated transcriptional regulation, and p19ARF-dependent cell cycle arrest and cellular senescence may contribute to the developmental abnormalities observed in primary microcephaly.
    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
    长链非编码RNA(lncRNA)表达的失调在胃癌(GC)的发生和发展中起着关键作用。然而,lncRNASNHG15在GC中的调控还没有得到很好的研究。SNHG15铁死亡的机制尚未揭示。这里,我们旨在探讨SNHG15介导的GC生物学功能和潜在的分子机制。通过分析来自我们队列和TCGA数据集的GC组织的RNA测序(RNA-seq)数据,鉴定了新的SNHG15。并通过qRT-PCR在GC细胞和组织中进一步验证。进行功能增益和功能丧失测定以检查SNHG15在体外和体内对GC的作用。SNHG15在GC中高度表达。增强的SNHG15与GC患者的恶性分期和不良预后呈正相关。功能增益和丧失研究表明,SNHG15是影响GC细胞生长所必需的,在体外和体内迁移和侵袭。机械上,致癌转录因子E2F1和MYC可以与SNHG15启动子结合并增强其表达。同时,SNHG15通过增强miR-24-3p增加E2F1和MYCmRNA表达。值得注意的是,SNHG15还可以通过竞争性结合HNRNPA1来增强SLC7A11在细胞质中的稳定性。此外,SNHG15通过SLC7A11/GPX4轴的HNRNPA1依赖性调节抑制铁凋亡。我们的结果支持了一个新的模型,其中E2F1-和MYC激活的SNHG15通过SLC7A11/GPX4轴的HNRNPA1依赖性调节铁凋亡,作为GC进展的关键效应,为GC的治疗提供了新的治疗方向。
    Dysregulation of long noncoding RNA (lncRNA) expression plays a pivotal role in the initiation and progression of gastric cancer (GC). However, the regulation of lncRNA SNHG15 in GC has not been well studied. Mechanisms for ferroptosis by SNHG15 have not been revealed. Here, we aimed to explore SNHG15-mediated biological functions and underlying molecular mechanisms in GC. The novel SNHG15 was identified by analyzing RNA-sequencing (RNA-seq) data of GC tissues from our cohort and TCGA dataset, and further validated by qRT-PCR in GC cells and tissues. Gain- and loss-of-function assays were performed to examine the role of SNHG15 on GC both in vitro and in vivo. SNHG15 was highly expressed in GC. The enhanced SNHG15 was positively correlated with malignant stage and poor prognosis in GC patients. Gain- and loss-of-function studies showed that SNHG15 was required to affect GC cell growth, migration and invasion both in vitro and in vivo. Mechanistically, the oncogenic transcription factors E2F1 and MYC could bind to the SNHG15 promoter and enhance its expression. Meanwhile, SNHG15 increased E2F1 and MYC mRNA expression by sponging miR-24-3p. Notably, SNHG15 could also enhance the stability of SLC7A11 in the cytoplasm by competitively binding HNRNPA1. In addition, SNHG15 inhibited ferroptosis through an HNRNPA1-dependent regulation of SLC7A11/GPX4 axis. Our results support a novel model in which E2F1- and MYC-activated SNHG15 regulates ferroptosis via an HNRNPA1-dependent modulation of the SLC7A11/GPX4 axis, which serves as the critical effectors in GC progression, and provides a new therapeutic direction in the treatment of GC.
    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:胰腺导管腺癌(PDAC)是治疗选择有限的最致命的癌症之一,说明迫切需要在PDAC中确定新的药物靶标。
    目的:利用肿瘤发育与正常胚胎发育的相似性,伴随着快速的细胞扩增,我们旨在鉴定和表征肿瘤形成和扩展过程中重新启动的胚胎信号通路.
    结果:这里,我们报道转录因子E2F1和E2F8是PDAC中潜在的关键调节因子.E2F1和E2F8RNA表达主要位于发育中的胰腺中的增殖细胞和PDAC中的恶性导管细胞中。PANC-1胰腺肿瘤细胞中E2F1和E2F8的沉默抑制了细胞增殖并损害了细胞扩散和迁移。此外,E2F1的缺失也影响细胞活力和凋亡,PDAC组织中E2F的表达与凋亡和有丝分裂途径基因的表达相关,提示E2F因子促进PDAC细胞的细胞周期调控和肿瘤发生。
    结论:我们的研究结果表明,E2F1和E2F8转录因子在胰腺祖细胞和PDAC细胞中表达,它们通过调节细胞增殖来促进肿瘤细胞的扩增,生存能力,和细胞迁移使这些基因成为胰腺癌的有吸引力的治疗靶标和潜在的预后标志物。
    BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers with limited treatment options, illustrating an urgent need to identify new drugable targets in PDACs.
    OBJECTIVE: Using the similarities between tumor development and normal embryonic development, which is accompanied by rapid cell expansion, we aimed to identify and characterize embryonic signaling pathways that were reinitiated during tumor formation and expansion.
    RESULTS: Here, we report that the transcription factors E2F1 and E2F8 are potential key regulators in PDAC. E2F1 and E2F8 RNA expression is mainly localized in proliferating cells in the developing pancreas and in malignant ductal cells in PDAC. Silencing of E2F1 and E2F8 in PANC-1 pancreatic tumor cells inhibited cell proliferation and impaired cell spreading and migration. Moreover, loss of E2F1 also affected cell viability and apoptosis with E2F expression in PDAC tissues correlating with expression of apoptosis and mitosis pathway genes, suggesting that E2F factors promote cell cycle regulation and tumorigenesis in PDAC cells.
    CONCLUSIONS: Our findings illustrate that E2F1 and E2F8 transcription factors are expressed in pancreatic progenitor and PDAC cells, where they contribute to tumor cell expansion by regulation of cell proliferation, viability, and cell migration making these genes attractive therapeutic targets and potential prognostic markers for pancreatic 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
    杀伤力,癌症的化学抗性和转移特征与表型可塑性癌症干细胞(CSC)相关。非细胞自主信号通路和细胞自主转录机制如何协调CSC的干细胞样特征仍然知之甚少。在这里,我们使用定量蛋白质组学方法来鉴定胰腺癌中CSC的分泌蛋白。我们发现细胞自主E2F1/4-pRb/RBL2轴平衡健康导管细胞中的非细胞自主信号,但在KRAS突变后变得失调。E2F1和E2F4诱导,而pRb/RBL2减少WNT配体表达(例如WNT7A,WNT7B,WNT10A,WNT4)从而调节自我更新,PDAC和乳腺癌中CSC的化学抗性和侵袭性,和成纤维细胞增殖。表观遗传学酶的筛选将GCN5鉴定为CSC的调节剂,其将H3K9ac沉积到WNT启动子和增强子上。总的来说,在多种癌症中,旁分泌信号通路受E2F-GCN5-RB轴控制,这可能是消除CSC的治疗靶标。
    The lethality, chemoresistance and metastatic characteristics of cancers are associated with phenotypically plastic cancer stem cells (CSCs). How the non-cell autonomous signalling pathways and cell-autonomous transcriptional machinery orchestrate the stem cell-like characteristics of CSCs is still poorly understood. Here we use a quantitative proteomic approach for identifying secreted proteins of CSCs in pancreatic cancer. We uncover that the cell-autonomous E2F1/4-pRb/RBL2 axis balances non-cell-autonomous signalling in healthy ductal cells but becomes deregulated upon KRAS mutation. E2F1 and E2F4 induce whereas pRb/RBL2 reduce WNT ligand expression (e.g. WNT7A, WNT7B, WNT10A, WNT4) thereby regulating self-renewal, chemoresistance and invasiveness of CSCs in both PDAC and breast cancer, and fibroblast proliferation. Screening for epigenetic enzymes identifies GCN5 as a regulator of CSCs that deposits H3K9ac onto WNT promoters and enhancers. Collectively, paracrine signalling pathways are controlled by the E2F-GCN5-RB axis in diverse cancers and this could be a therapeutic target for eliminating CSCs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号