E2F transcription factor 1 (E2F1)

  • 文章类型: Journal Article
    近年来,关于骨肉瘤中非SMC凝缩素I复合物G亚基(NCAPG)的报道很少。本研究旨在探讨NCAPG在骨肉瘤中的生物学作用及其潜在的分子机制,并进一步阐明NCAPG在骨肉瘤中异常表达的原因。
    这里,我们通过生物信息学方法,分析NCAPG在肉瘤组织和正常组织中的差异表达,探讨NCAPG表达水平与肉瘤组织分化的关系,包括肿瘤复发,转移,和病人的生存。接下来,通过多个在线网站工具预测骨肉瘤中NCAPG异常表达的转录因子,并通过细胞实验进行验证.随后,进行功能丧失和细胞表型实验以证实NCAPG对骨肉瘤细胞恶性生物学行为的影响。机械上,通过回顾文献,我们发现NCAPG可以通过调节Wnt/β-catenin信号通路影响多种实体肿瘤的恶性进展。因此,我们通过westernblot实验初步研究了NCAPG在骨肉瘤中对该通路的潜在作用。
    与正常组织相比,在肉瘤中发现NCAPG的表达增加,与分化差呈正相关,转移,预后不良。结合转录因子预测结果,相关分析,和TCGA公共数据库中的表达水平,以及体外细胞测定的验证结果,我们发现E2F转录因子1(E2F1)调节骨肉瘤中NCAPG的表达增加。细胞表型实验结果表明沉默NCAPG可以抑制细胞增殖,迁移,和骨肉瘤细胞的侵袭。初步机制研究表明,NCAPG可能通过Wnt/β-catenin途径影响骨肉瘤的进展。
    我们的数据表明,E2F1通过调节NCAPG基因的转录促进骨肉瘤中NCAPG的表达。NCAPG的上调通过Wnt/β-连环蛋白信号轴促进骨肉瘤进展。
    UNASSIGNED: In recent years, there are few reports on non-SMC condensin I complex subunit G (NCAPG) in osteosarcoma. Our study aims to explore the biological role of NCAPG in osteosarcoma and its underlying molecular mechanism and to further clarify the reasons for the abnormal expression of NCAPG in osteosarcoma.
    UNASSIGNED: Here, we mined The Cancer Genome Atlas (TCGA) Program public database through bioinformatics methods, analyzed the differential expression of NCAPG in sarcoma tissue and normal tissue, and explored the relationship between NCAPG expression level and sarcoma tissue differentiation, including tumor recurrence, metastasis, and patient survival. Next, the transcription factors responsible for the abnormal expression of NCAPG in osteosarcoma tumors were predicted by multiple online website tools and verified via cellular experiments. Subsequently, loss of function and cell phenotype experiments were performed to confirm the effect of NCAPG on the malignant biological behavior of osteosarcoma cells. Mechanistically, by reviewing the literature, we found that NCAPG can affect the malignant progression of many solid tumors by regulating the Wnt/β-catenin signaling pathway. Therefore, we preliminarily investigated the potential effect of NCAPG on this pathway via western blot experiments in osteosarcoma.
    UNASSIGNED: Increased expression of NCAPG was found in sarcoma compared to normal tissues, which was positively correlated with poor differentiation, metastasis, and poor prognosis. Combining the transcription factor prediction results, correlation analysis, and expression level in the TCGA public database with validation outcomes of in vitro cell assays, we found that E2F transcription factor 1 (E2F1) regulated the increased expression of NCAPG in osteosarcoma. The results of cell phenotype experiments showed that silencing NCAPG could inhibit the proliferation, migration, and invasion of osteosarcoma cells. The preliminary mechanistic investigation suggested that NCAPG may affect osteosarcoma progression through the Wnt/β-catenin pathway.
    UNASSIGNED: Our data reveal that E2F1 facilitates NCAPG expression in osteosarcoma by regulating the transcription of the NCAPG gene. Up-regulation of NCAPG promotes osteosarcoma progression via the Wnt/β-catenin 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
    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
    各种研究表明,赖氨酸乙酰转移酶2A(KAT2A),E2F转录因子1(E2F1),泛素接合酶E2C(UBE2C)基因通过调控细胞周期调控肿瘤细胞的增殖和迁移。然而,对它们的作用机制缺乏深入和系统的研究。这项研究分析了癌症基因组图谱(TCGA),以筛选潜在的候选基因以及泛癌症中KAT2A和E2F1复合物的调控网络。定量实时PCR(qRT-PCR)和蛋白质印迹(WB),细胞表型检测,免疫荧光共定位,染色质免疫沉淀测定(ChIP),和RNA-Seq技术用于探索候选基因的功能,UBE2C。我们发现,与正常组织相比,这三个基因在超过10种肿瘤类型中的表达明显更高。此外,UBE2C主要在肿瘤细胞中表达,其中强调了UBE2C作为一种特定治疗策略的影响。此外,KAT2A和E2F1可以通过增强UBE2C的表达来促进细胞增殖和癌细胞的迁移。机械上,发现KAT2A与E2F1合作,并被E2F1募集到UBE2C启动子,通过增加H3K9的乙酰化水平来提高UBE2C的表达。
    Various studies have shown that lysine acetyltransferase 2A (KAT2A), E2F transcription factor 1 (E2F1), and ubiquitin conjugating enzyme E2 C (UBE2C) genes regulated the proliferation and migration of tumor cells through regulating the cell cycle. However, there is a lack of in-depth and systematic research on their mechanisms of action. This study analyzed The Cancer Genome Atlas (TCGA) to screen potential candidate genes and the regulation network of KAT2A and E2F1 complex in pan-cancer. Quantitative real-time PCR (qRT-PCR) and Western blotting (WB), cell phenotype detection, immunofluorescence co-localization, chromatin immunoprecipitation assay (ChIP), and RNA-Seq techniques were used to explore the functional of a candidate gene, UBE2C. We found that the expression of these three genes was significantly higher in more than 10 tumor types compared to normal tissue. Moreover, UBE2C was mainly expressed in tumor cells, which highlighted the impacts of UBE2C as a specific therapeutic strategy. Moreover, KAT2A and E2F1 could promote cell proliferation and the migration of cancer cells by enhancing the expression of UBE2C. Mechanically, KAT2A was found to cooperate with E2F1 and be recruited by E2F1 to the UBE2C promoter for elevating the expression of UBE2C by increasing the acetylation level of H3K9.
    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
    Considering the increase in the proportion of lung adenocarcinoma (LUAD) cases among all lung cancers and its considerable contribution to cancer-related deaths worldwide, we sought to identify novel oncogenes to provide potential targets and facilitate a better understanding of the malignant progression of LUAD.
    The results from the screening of transcriptome and survival analyses according to the integrated Gene Expression Omnibus (GEO) datasets and The Cancer Genome Atlas (TCGA) data were combined, and a promising risk biomarker called meiotic nuclear divisions 1 (MND1) was selectively acquired. Cell viability assays and subcutaneous xenograft models were used to validate the oncogenic role of MND1 in LUAD cell proliferation and tumor growth. A series of assays, including mass spectrometry, co-immunoprecipitation (Co-IP), and chromatin immunoprecipitation (ChIP), were performed to explore the underlying mechanism.
    MND1 up-regulation was identified to be an independent risk factor for overall survival in LUAD patients evaluated by both tissue microarray staining and third party data analysis. In vivo and in vitro assays showed that MND1 promoted LUAD cell proliferation by regulating cell cycle. The results of the Co-IP, ChIP and dual-luciferase reporter assays validated that MND1 competitively bound to tumor suppressor Kruppel-like factor 6 (KLF6), and thereby protecting E2F transcription factor 1 (E2F1) from KLF6-induced transcriptional repression. Luciferase reporter and ChIP assays found that E2F1 activated MND1 transcription by binding to its promoter in a feedback manner.
    MND1, KLF6, and E2F1 form a positive feedback loop to regulate cell cycle and confer DDP resistance in LUAD. MND1 is crucial for malignant progression and may be a potential therapeutic target in LUAD patients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号