RuvBL1

RUVBL1
  • 文章类型: Journal Article
    子宫内膜癌(EC)是妇科恶性肿瘤中日益关注的问题。易洛魁Homeobox2(IRX2),易洛魁异型盒基因家族的一员,在不同类型的癌症中表现出不同的效果,强调需要广泛探索其参与欧共体进程。利用TCGA和GEO数据库,以及对临床样本进行免疫组织化学(IHC)分析,我们评估了IRX2及其启动子甲基化在EC中的表达水平.为了了解IRX2的功能作用,我们进行了各种测定,包括体外CCK-8测定,集落形成试验,细胞侵袭试验,和细胞凋亡测定。此外,我们使用了体内皮下异种移植小鼠模型。此外,我们进行了KEGG通路和基因集富集分析,以深入了解其潜在机制.为了验证IRX2和RUVBL1之间的调节关系,我们采用了染色质免疫沉淀和荧光素酶报告基因测定。我们的结果表明,在EC中IRX2表达水平显着降低,与较高的组织学等级相关,晚期临床阶段,总体生存率下降。我们观察到IRX2启动子的DNA甲基化抑制其在EC中的表达,cg26333652和cg11793269作为甲基化位点起关键作用。相比之下,IRX2的异位过表达显著抑制细胞增殖和侵袭,促进细胞凋亡。此外,我们发现IRX2对RUVBL1的表达有负调控作用,RUVBL1在EC中上调,与预后较差有关。总之,我们的发现表明,IRX2的表达降低通过调节RUVBL1的表达促进EC细胞的生长,从而促进EC的发展。因此,靶向IRX2-RUVBL1轴有望成为EC治疗的潜在治疗策略.
    Endometrial carcinoma (EC) is a rising concern among gynecological malignancies. Iroquois Homeobox 2 (IRX2), a member of the Iroquois homeobox gene family, demonstrates variable effects in different cancer types, emphasizing the need for extensive exploration of its involvement in EC progression. Utilizing TCGA and GEO databases, as well as performing immunohistochemistry (IHC) analysis on clinical samples, we assessed the expression levels of IRX2 and its promoter methylation in EC. To understand the functional roles of IRX2, we conducted various assays including in vitro CCK-8 assays, colony formation assays, cell invasion assays, and cell apoptosis assays. Moreover, we utilized in vivo subcutaneous xenograft mouse models. Additionally, we performed KEGG pathway and gene set enrichment analyses to gain insights into the underlying mechanisms. To validate the regulatory relationship between IRX2 and RUVBL1, we employed chromatin immunoprecipitation and luciferase reporter assays. Our results indicate significantly reduced levels of IRX2 expression in EC, correlating with higher histological grades, advanced clinical stages, and diminished overall survival. We observed that DNA methylation of the IRX2 promoter suppresses its expression in EC, with cg26333652 and cg11793269 playing critical roles as methylated sites. In contrast, ectopic overexpression of IRX2 substantially inhibits cell proliferation and invasion, and promotes cell apoptosis. Additionally, we discovered that IRX2 exerts negative regulation on the expression of RUVBL1, which is upregulated in EC and associated with a poorer prognosis. In conclusion, our findings indicate that decreased expression of IRX2 facilitates EC cell growth through the regulation of RUVBL1 expression, thereby contributing to the development of EC. Hence, targeting the IRX2-RUVBL1 axis holds promise as a potential therapeutic strategy for EC treatment.
    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
    全基因组关联研究以及表达数量性状基因座(eQTL)作图已经确定了数百个单核苷酸多态性(SNP)及其在前列腺癌(PCa)中的靶基因。然而,这些风险位点的功能表征仍具有挑战性.要筛选潜在的监管SNP,我们设计了一个包含9,133个向导RNA(gRNA)的CRISPRi文库,以覆盖2,166个与PCa相关的候选SNP位点,并鉴定出117个能够调节90个基因的SNPs,以获得PCa细胞生长优势.其中,rs60464856被筛选中显著耗竭的多个gRNA覆盖(FDR<0.05)。PRACTICAL和FinnGen队列中汇总的SNP关联分析显示,rs60464856G等位基因的PCa风险明显更高(p值分别为1.2×10-16和3.2×10-7)。随后的eQTL分析揭示G等位基因与多个数据集中的RUVBL1表达增加相关。进一步的CRISPRi和xCas9碱基编辑证实rs60464856G等位基因导致RUVBL1表达升高。此外,基于SILAC的蛋白质组分析证明了在rs60464856区域的粘附蛋白亚基的等位基因结合,其中HiC数据集显示前列腺细胞系中一致的染色质相互作用。在异种移植小鼠模型中,RUVBL1耗竭抑制PCa细胞增殖和肿瘤生长。基因集富集分析提示RUVBL1表达与细胞周期相关通路相关。在TCGA数据集中,RUVBL1的表达增加和细胞周期通路的激活与PCa的低存活率相关。我们的CRISPRi筛查优先考虑了约100个对前列腺细胞增殖至关重要的调节性SNP。结合蛋白质组学和功能研究,我们描述了rs60464856和RUVBL1在PCa进展中的机制作用。
    Genome-wide association studies along with expression quantitative trait locus (eQTL) mapping have identified hundreds of single-nucleotide polymorphisms (SNPs) and their target genes in prostate cancer (PCa), yet functional characterization of these risk loci remains challenging. To screen for potential regulatory SNPs, we designed a CRISPRi library containing 9,133 guide RNAs (gRNAs) to cover 2,166 candidate SNP loci implicated in PCa and identified 117 SNPs that could regulate 90 genes for PCa cell growth advantage. Among these, rs60464856 was covered by multiple gRNAs significantly depleted in screening (FDR < 0.05). Pooled SNP association analysis in the PRACTICAL and FinnGen cohorts showed significantly higher PCa risk for the rs60464856 G allele (p value = 1.2 × 10-16 and 3.2 × 10-7, respectively). Subsequent eQTL analysis revealed that the G allele is associated with increased RUVBL1 expression in multiple datasets. Further CRISPRi and xCas9 base editing confirmed that the rs60464856 G allele leads to elevated RUVBL1 expression. Furthermore, SILAC-based proteomic analysis demonstrated allelic binding of cohesin subunits at the rs60464856 region, where the HiC dataset showed consistent chromatin interactions in prostate cell lines. RUVBL1 depletion inhibited PCa cell proliferation and tumor growth in a xenograft mouse model. Gene-set enrichment analysis suggested an association of RUVBL1 expression with cell-cycle-related pathways. Increased expression of RUVBL1 and activation of cell-cycle pathways were correlated with poor PCa survival in TCGA datasets. Our CRISPRi screening prioritized about one hundred regulatory SNPs essential for prostate cell proliferation. In combination with proteomics and functional studies, we characterized the mechanistic role of rs60464856 and RUVBL1 in PCa progression.
    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)是导致癌症死亡的主要原因。近年来,AHNAK2在LUAD中的致瘤功能引起了越来越多的关注,虽然很少有研究报道其高分子量。
    方法:分析了AHNAK2的mRNA-seq数据以及来自UCSCXena和GEO的相应临床数据。用sh-NC和sh-AHNAK2转染LUAD细胞系,然后通过体外实验检测迁移和侵袭。我们进行了RNA测序和质谱分析,以探索AHNAK2的下游机制和相互作用蛋白。最后,westernblot,使用细胞周期分析和CO-IP来证实我们对先前实验的假设.
    结果:我们的研究表明,AHNAK2在肿瘤中的表达明显高于正常肺组织,而较高的AHNAK2表达导致预后不良,尤其是晚期肿瘤患者。通过shRNA抑制AHNAK2降低了LUAD细胞系的增殖,迁移和入侵,并诱导DNA复制的显着变化,NF-κB信号通路与细胞周期.AHNAK2敲低也引起G1/S期细胞周期停滞,这可能归因于AHNAK2和RUVBL1的相互作用。此外,基因集富集分析(GSEA)和RNA测序的结果表明,AHNAK2可能在有丝分裂细胞周期中起作用。
    结论:AHNAK2促进增殖,LUAD中的迁移和侵袭,并通过与RUVBL1的相互作用调节细胞周期。AHNAK2还需要更多的研究来揭示其上游机制。
    Lung adenocarcinoma (LUAD) is the leading cause of death among cancer diseases. The tumorigenic functions of AHNAK2 in LUAD have attracted more attention in recent years, while there are few studies which have reported its high molecular weight.
    The mRNA-seq data of AHNAK2 and corresponding clinical data from UCSC Xena and GEO was analyzed. LUAD cell lines were transfected with sh-NC and sh-AHNAK2, and cell proliferation, migration and invasion were then detected by in vitro experiments. We performed RNA sequencing and mass spectrometry analysis to explore the downstream mechanism and interacting proteins of AHNAK2. Finally, western blot, cell cycle analysis and CO-IP were used to confirm our assumptions regarding previous experiments.
    Our study revealed that AHNAK2 expression was significantly higher in tumors than in normal lung tissues and higher AHNAK2 expression led to a poor prognosis, especially in patients with advanced tumors. AHNAK2 suppression via shRNA reduced the LUAD cell lines proliferation, migration and invasion and induced significant changes in DNA replication, NF-kappa B signaling pathway and cell cycle. AHNAK2 knockdown also caused G1/S phase cell cycle arrest, which could be attributed to the interaction of AHNAK2 and RUVBL1. In addition, the results from gene set enrichment analysis (GSEA) and RNA sequencing suggested that AHNAK2 probably plays a part in the mitotic cell cycle.
    AHNAK2 promotes proliferation, migration and invasion in LUAD and regulates the cell cycle via the interaction with RUVBL1. More studies of AHNAK2 are still needed to reveal its upstream mechanism.
    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:由于EIF3D在结直肠癌(CRC)中是致癌的,并且与多药耐药有关,本研究旨在探讨EIF3D是否以及如何调节CRC对5-氟尿嘧啶(5-Fu)的耐药性。
    方法:使用生物信息学工具预测CRC中EIF3D相关基因。CRC细胞和裸小鼠接受5-Fu处理。然后,EIF3D以及EIF3D和RUVBL1之间的相互作用对细胞活力的影响,菌落形成,凋亡,通过MTT检测DNA损伤,菌落形成,流式细胞术,和免疫荧光分析,以及通过鼠异种移植试验对体内肿瘤发生的研究。通过probit回归分析确定5-Fu对CRC细胞的IC50值。EIF3D的表达式,eIF4E,EIF3D相关基因,γH2AX,Bcl-2,Bax,以及在CRC组织中裂解的Caspase-3/Caspase-3,细胞,和/或通过qRT-PCR和/或Western印迹分析异种移植肿瘤。
    结果:EIF3D和RUVBL1高表达,与CRC组织/细胞呈正相关。在CRC细胞中,除了eIF4E,EIF3D和RUVBL1水平均被5-Fu处理上调;除此之外,RUVBL1水平被EIF3D沉默而不是eIF4E下调。同时,EIF3D沉默降低了5-Fu的IC50值,并增强了5-Fu诱导的生存力降低,集落形成抑制,凋亡促进,Bcl-2下调,和γH2AX,Bax,和裂解的Caspase-3/Caspase-3上调,但逆转了5-Fu触发的RUVBL1上调。RUVBL1过表达抵消了EIF3D沉默诱导的5-Fu处理的CRC细胞的活力降低和凋亡促进,以及5-Fu处理小鼠的肿瘤发生抑制和凋亡促进。
    结论:EIF3D通过上调RUVBL1水平促进CRC对5-Fu的耐药性。
    BACKGROUND: As EIF3D is oncogenic in colorectal cancer (CRC) and is associated with multidrug resistance, this study aims to investigate whether and how EIF3D regulates resistance to 5-fluorouracil (5-Fu) in CRC.
    METHODS: EIF3D-associated genes in CRC were predicted using bioinformatics tools. CRC cells and nude mice received 5-Fu treatment. Then, the impacts of EIF3D and the interaction between EIF3D and RUVBL1 on cell viability, colony formation, apoptosis, and DNA damage were detected through MTT, colony formation, flow cytometry, and immunofluorescence assays, and those on in vivo tumorigenesis through murine xenograft assay. IC50 value of 5-Fu for CRC cells was determined by probit regression analysis. Expressions of EIF3D, eIF4E, EIF3D-associated genes, γH2AX, Bcl-2, Bax, and Cleaved Caspase-3/Caspase-3 in CRC tissues, cells, and/or xenograft tumors were analyzed by qRT-PCR and/or Western blot.
    RESULTS: EIF3D and RUVBL1 were highly expressed and positively correlated with CRC tissues/cells. In CRC cells, except for eIF4E, both EIF3D and RUVBL1 levels were upregulated by 5-Fu treatment; in addition to that, RUVBL1 level was downregulated by EIF3D silencing rather than eIF4E. Meanwhile, EIF3D silencing diminished IC50 value of 5-Fu and potentiated 5-Fu-induced viability decrease, colony formation inhibition, apoptosis promotion, Bcl-2 downregulation, and γH2AX, Bax, and Cleaved Caspase-3/Caspase-3 upregulation but reversed 5-Fu-triggered RUVBL1 upregulation. RUVBL1 overexpression offsets EIF3D silencing-induced viability decrease and apoptosis promotion of 5-Fu-treated CRC cells, and tumorigenesis suppression and apoptosis promotion in 5-Fu-treated mice.
    CONCLUSIONS: EIF3D promotes resistance to 5-Fu in CRC through upregulating RUVBL1 level.
    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
    原理:肝细胞癌(HCC)是世界上最严重的癌症之一,很少有有效的肝癌靶向治疗。脂质代谢重编程是癌症代谢的标志,指导对抗肿瘤治疗的反应。癌症中的这种脂质代谢改变受到哺乳动物雷帕霉素mTOR靶的关键调节,这被认为是一个有希望的治疗目标。尽管努力,mTOR抑制剂(mTORi)在临床上产生的反应有限,部分原因是对癌症中mTORC1成瘾的不完全了解。方法:采用CRISPR-Cas9系统建立Hpcal1空小鼠。使用具有二乙基亚硝胺(DEN)/CCL4或MYC/Trp53-/-的Hpcal1缺陷型小鼠通过流体动力学尾静脉注射产生小鼠中的肝癌模型。RNA测序(RNA-seq)用于鉴定潜在的信号传导途径。使用定量聚合酶链反应(qPCR)确定HPCAL1和mTOR信号的表达,蛋白质印迹和免疫组织化学。通过CCK-8测量评估Hpcal1在肝脏肿瘤发生中的作用及其对mTORi的反应,集落形成测定和小鼠模型。结果:在这项研究中,我们确定海马蛋白样蛋白1(HPCAL1)是从头脂质生物合成和mTOR信号激活的重要负调节因子,在小鼠中限制肝脏肿瘤发生并建立HCC的代谢脆弱性。HPCAL1的遗传丢失使HCCmTORC1成瘾,并在体外和体内对mTORiAZD-8055敏感。重要的是,HPCAL1的表达与人类标本中mTOR磷酸化和几种关键脂质生物合成酶的水平呈负相关。机械上,HPCAL1直接与RuvB类似AAAATP酶1(RUVBL1)结合,抑制TEL2-TTI1-TTI2(TTT)-RUVBL复合物的组装,随后导致mTOR信号抑制。结论:我们发现了HPCAL1缺失的HCC中的代谢脆弱性和mTOR成瘾,这为使用mTORi的mTORC1过度激活的HCC提供了选择性治疗窗口。
    Rationale: Hepatocellular carcinoma (HCC) is one of the most severe cancers worldwide, with few effective targeted therapies for HCC. Lipid metabolic reprogramming is emerged as a hallmark of cancer metabolism that guides response to antitumoral therapies. Such lipid metabolic alteration in cancers is critically regulated by the mammalian target of rapamycin mTOR, which is considered as a promising therapeutic target. Despite efforts, mTOR inhibitors (mTORi) have produced limited response clinically, partly due to incomplete knowledge of mTORC1 addiction in cancers. Methods: CRISPR-Cas9 system was used to establish Hpcal1 null mice. The liver cancer model in mice was generated using Hpcal1-deficient mice with diethylnitrosamine (DEN) /CCL4 or MYC/Trp53-/- via hydrodynamic tail-vein injection. RNA-sequencing (RNA-seq) was used to identify potential signaling pathways. The expression of HPCAL1 and mTOR signaling were determined using quantitative polymerase chain reaction (qPCR), western blot and immunohistochemistry. The role of Hpcal1 in liver tumorigenesis and its response to mTORi was assessed by CCK-8 measurements, colony formation assay and in mouse model. Results: In this study, we identified hippocalcin-like protein 1 (HPCAL1) as an important negative regulator of de novo lipid biosynthesis and mTOR signaling activation, limiting liver tumorigenesis and establishing a metabolic vulnerability of HCC in mice. Genetic loss of HPCAL1 rendered HCC mTORC1-addicted and sensitive to mTORi AZD-8055 in vitro and in vivo. Importantly, HPCAL1 expression was inversely correlated with the levels of mTOR phosphorylation and several critical lipid biosynthesis enzymes in human specimens. Mechanistically, HPCAL1 directly bound to RuvB Like AAA ATPase 1 (RUVBL1), inhibiting the assembly of TEL2-TTI1-TTI2 (TTT)-RUVBL complex and subsequent leading the mTOR signaling suppression. Conclusion: We uncover a metabolic vulnerability and mTOR addiction in HCC with HPCAL1 loss that provides a selective therapeutic window for HCC with mTORC1 hyperactivation using mTORi.
    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
    RUVBL1和RUVBL2是高度保守的AAAATP酶(与各种细胞活性相关的ATP酶),与癌症的进展高度相关,这使得它们成为新型治疗性抗癌药物的有吸引力的靶标。在这项工作中,进行基于对接的虚拟筛选以鉴定对RUVBL1/2复合物具有活性的化合物.七个化合物在酶和细胞测定中均显示出对复合物的抑制活性。基于化合物15的支架,合成了一系列吡唑并[1,5-a]嘧啶-3-甲酰胺类似物,具有抑制活性和良好的结构操作潜力。对结构-活性关系的分析确定R2上的苄基和R4上的芳环取代的哌嗪基是对RUVBL1/2复合物的抑制活性所必需的。其中,化合物18对RUVBL1/2复合物和RUVBL1的IC50值分别为6.0±0.6μM和7.7±0.9μM,在细胞系A549,H1795,HCT116和MDA-MB-231中显示出最有效的抑制作用,IC50值为15±1.2μM,15±1.8μM,11±1.0μM,和8.9±0.9μM。进行化合物的对接研究以预测吡唑并[1,5-a]嘧啶-3-甲酰胺的结合模式。此外,采用基于质谱的蛋白质组分析来探索通过用化合物16、18和19处理而失调的细胞蛋白质。一起,这些分析的数据表明,化合物18可以作为结构修饰的起点,以提高效力,选择性,和潜在治疗分子的药代动力学参数。
    RUVBL1 and RUVBL2 are highly conserved AAA ATPases (ATPases Associated with various cellular Activities) and highly relevant to the progression of cancer, which makes them attractive targets for novel therapeutic anticancer drugs. In this work, docking-based virtual screening was performed to identify compounds with activity against the RUVBL1/2 complex. Seven compounds showed inhibitory activity against the complex in both enzymatic and cellular assays. A series of pyrazolo[1,5-a]pyrimidine-3-carboxamide analogs were synthesized based on the scaffold of compound 15 with inhibitory activity and good potential for structural manipulation. Analysis of the structure-activity relationship identified the benzyl group on R2 and aromatic ring-substituted piperazinyl on R4 as essential for inhibitory activity against the RUVBL1/2 complex. Of these, compound 18, which has IC50 values of 6.0 ± 0.6 μM and 7.7 ± 0.9 μM against RUVBL1/2 complex and RUVBL1 respectively, showed the most potent inhibition in cell lines A549, H1795, HCT116, and MDA-MB-231 with IC50 values of 15 ± 1.2 μM, 15 ± 1.8 μM, 11 ± 1.0 μM, and 8.9 ± 0.9 μM respectively. A docking study of the compound was performed to predict the binding mode of pyrazolo[1,5-a]pyrimidine-3-carboxamides. Furthermore, mass spectrometry-based proteomic analysis was employed to explore cellular proteins dysregulated by treatment with compounds 16, 18, and 19. Together, the data from these analyses suggest that that compound 18 could serve as a starting point for structural modifications in order to improve potency, selectivity, and pharmacokinetic parameters of potential therapeutic molecules.
    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
    肺癌是世界上逝世亡率最高的常见恶性肿瘤。肿瘤坏死因子相关的凋亡诱导配体(TRAIL)作为潜在的抗癌剂可诱导人类癌细胞的选择性凋亡。不幸的是,大约一半的肺癌细胞系对TRAIL诱导的细胞死亡具有内在抗性。在这项研究中,我们将RuvBL1鉴定为c-Jun/AP-1活性的阻遏物,有助于肺癌细胞中的TRAIL抗性。敲除RuvBL1有效致敏TRAIL抗性细胞,RuvBL1的过表达抑制TRAIL诱导的细胞凋亡。此外,通过Oncomine分析,RuvBL1和c-Jun在肺腺癌中的表达呈负相关。肺癌中RuvBL1的高表达与c-Jun的低表达呈负相关,与整体预后不良相关。一起来看,我们的研究拓宽了TRAIL耐药的分子机制,并提示沉默RuvBL1与TRAIL协同成为肺癌治疗的一种新的治疗策略.
    Lung cancer is the common malignant tumor with the highest death rate in the world. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) as a potential anticancer agent induces selective apoptotic death of human cancer cells. Unfortunately, approximately half of lung cancer cell lines are intrinsically resistant to TRAIL-induced cell death. In this study, we identified RuvBL1 as a repressor of c-Jun/AP-1 activity, contributing to TRAIL resistance in lung cancer cells. Knocking down RuvBL1 effectively sensitized resistant cells to TRAIL, and overexpression of RuvBL1 inhibited TRAIL-induced apoptosis. Moreover, there was a negative correlation expression between RuvBL1 and c-Jun in lung adenocarcinoma by Oncomine analyses. High expression of RuvBL1 inversely with low c-Jun in lung cancer was associated with a poor overall prognosis. Taken together, our studies broaden the molecular mechanisms of TRAIL resistance and suggest the application of silencing RuvBL1 synergized with TRAIL to be a novel therapeutic strategy in lung 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
    CircMYO10 is a circular RNA generated by back-splicing of gene MYO10 and is upregulated in osteosarcoma cell lines, but its functional role in osteosarcoma is still unknown. This study aimed to clarify the mechanism of circMYO10 in osteosarcoma.
    CircMYO10 expression in 10 paired osteosarcoma and chondroma tissues was assessed by quantitative reverse transcription polymerase chain reaction (PCR). The function of circMYO10/miR-370-3p/RUVBL1 axis was assessed regarding two key characteristics: proliferation and endothelial-mesenchymal transition (EMT). Bioinformatics analysis, western blotting, real-time PCR, fluorescence in situ hybridization, immunoprecipitation, RNA pull-down assays, luciferase reporter assays, chromatin immunoprecipitation, and rescue experiments were used to evaluate the mechanism. Stably transfected MG63 cells were injected via tail vein or subcutaneously into nude mice to assess the role of circMYO10 in vivo.
    CircMYO10 was significantly upregulated, while miR-370-3p was downregulated, in osteosarcoma cell lines and human osteosarcoma samples. Silencing circMYO10 inhibited cell proliferation and EMT in vivo and in vitro. Mechanistic investigations revealed that miR-370-3p targets RUVBL1 directly, and inhibits the interaction between RUVBL1 and β-catenin/LEF1 complex while circMYO10 showed a contrary effect via the inhibition of miR-370-3p. RUVBL1 was found to be complexed with chromatin remodeling and histone-modifying factor TIP60, and lymphoid enhancer factor-1 (LEF1) to promote histone H4K16 acetylation (H4K16Ac) in the vicinity of the promoter region of gene C-myc. Chromatin immunoprecipitation methods showed that miR-370-3p sponge promotes H4K16Ac in the indicated region, which is partially abrogated by RUVBL1 small hairpin RNA (shRNA) while circMYO10 showed a contrary result via the inhibition of miR-370-3p. Either miR-370-3p sponge or ShRUVBL1 attenuated circMYO10-induced phenotypes in osteosarcoma cell lines. MiR-370-3p inhibition abrogated the inhibition of proliferation, EMT of osteosarcoma cells in vitro and in vivo seen upon circMYO10 suppression via Wnt/β-catenin signaling.
    CircMYO10 promotes osteosarcoma progression by regulating miR-370-3p/RUVBL1 axis to promote chromatin remodeling and thus enhances the transcriptional activity of β-catenin/LEF1 complex, which indicates that circMYO10 may be a potential therapeutic target for osteosarcoma treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Lung cancer remains the leading cause of cancer-related deaths in the world. The RAF/MEK/ERK pathway controls many fundamental cellular functions and plays key roles in lung carcinogenesis. However, the proteins that regulate this pathway remain largely unknown. Here, we identified a novel C-RAF-binding protein, RUVBL1, which activates the RAF/MEK/ERK pathway by inhibiting phosphorylation of the C-RAF protein at serine 259. RUVBL1 expression was elevated in lung adenocarcinoma tissues. In addition, knocking out RUVBL1 effectively inhibited the proliferation and invasion of A549 cells. In vivo experiments, RUVBL1 deficiency significantly decreased the tumorigensis of lung cancer. In conclusion, we have shown that RUVBL1 could activate the RAF/MEK/ERK pathway by inhibiting phosphorylation of the C-RAF protein at serine 259, to promote lung cancer progression. Therefore, RUVBL1 could represent a novel therapeutic target for lung cancer treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    The mechanisms of breast cancer collective invasion are poorly understood limiting the metastasis therapy. The ATPase RUVBL1 is frequently overexpressed in various cancers and plays a crucial role in oncogenic process. We further investigated the role of RUVBL1 in promoting collective invasion and uncovered that targeting RUVBL1 could inhibit metastatic progression.
    The expression levels of RUVBL1 and ITFG1 were examined by Western blot and qRT-PCR. Co-localization and interaction of RUVBL1 and ITFG1 were determined by immunofluorescence and co-immunoprecipitation. The invasive ability was examined by transwell assay and microfluidic assay. The metastatic and tumorigenic abilities of breast cancer cells were revealed in BALB/c nude mice by xenograft and tail vein injection.
    ATPase RUVBL1 is highly expressed in breast cancer and predicts the poor prognosis. Elevated expression of RUVBL1 is found in high metastatic breast cancer cells. Silencing RUVBL1 suppresses cancer cell expansion and invasion in vitro and in vivo. RUVBL1 interacts with a conserved transmembrane protein ITFG1 in cytoplasm and plasma membrane to promote the collective invasion. Using a microfluidic model, we demonstrated that silencing RUVBL1 or ITFG1 individually compromises collective invasion of breast cancer cells.
    RUVBL1 is a vital regulator for collective invasion. The interaction between RUVBL1 and ITFG1 is required for breast cancer cell collective invasion and progression.
    Targeting collective invasion promoted by RUVBL1-ITFG1 complex provides a novel therapeutic strategy to improve the prognosis of invasive breast cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号