首页> 外文期刊>Drug Design, Development and Therapy >Quinoline-based clioquinol and nitroxoline exhibit anticancer activity inducing FoxM1 inhibition in cholangiocarcinoma cells
【24h】

Quinoline-based clioquinol and nitroxoline exhibit anticancer activity inducing FoxM1 inhibition in cholangiocarcinoma cells

机译:喹啉基氯喹醇和硝氧嘧啶具有抗癌活性,可诱导胆管癌细胞中的FoxM1抑制

获取原文
           

摘要

Purpose: Fork head box M1 (FoxM1) is an oncogenic transcription factor frequently elevated in numerous cancers, including cholangiocarcinoma (CCA). A growing body of evidence documents its diverse functions contributing to tumorigenesis and cancer progression. As such, discovery of agents that can target FoxM1 would be valuable for the treatment of CCA. The quinoline-based compounds, namely clioquinol (CQ) and nitroxoline (NQ), represent a new class of anticancer drug. However, their efficacy and underlying mechanisms have not been elucidated in CCA. In this study, anticancer activities and inhibitory effects of CQ and NQ on FoxM1 signaling were explored using CCA cells.Methods: The effects of CQ and NQ on cell viability and proliferation were evaluated using the colorimetric 3-(4,5-dimethylthiazol-2yl)-5-(3-carboxymethoxyphenyl)-(4-sulfophenyl)-2H-tetrazolium (MTS assay). Colony formation and cell migration affected by CQ and NQ were investigated using a clonogenic and a wound healing assay, respectively. To demonstrate the agents’ effects on FoxM1 signaling, expression levels of the target genes were quantitatively determined using real-time polymerase chain reaction.Results: CQ and NQ significantly inhibited cell survival of HuCCT1 and Huh28 in a dose- and a time-dependent fashion. Further investigations using the rapidly proliferating HuCCT1 cells revealed significant suppression of cell proliferation and colony formation induced by low doses of the compounds. Treatment of CQ and NQ repressed expression of cyclin D1 but enhanced expression of p21. Most importantly, upon CQ and NQ treatment, expression of oncogenic FoxM1 was markedly decreased concomitant with downregulation of various FoxM1’s downstream targets including cdc25b, CENP-B, and survivin. In addition, the compounds distinctly impaired HuCCT1 migration as well as inhibited expression of matrix metalloproteinase (MMP)-2 and MMP-9.Conclusion: Collectively, this study reports for the first time the anticancer effects of CQ and NQ against CCA cells, and highlights new insights into the mechanism of actions of the quinoline-based compounds to disrupt FoxM1 signaling.
机译:目的:叉头盒M1(FoxM1)是一种致癌转录因子,在包括胆管癌(CCA)在内的许多癌症中经常升高。越来越多的证据表明其多种功能促成肿瘤发生和癌症进展。因此,发现可以靶向FoxM1的药物对于CCA的治疗将是有价值的。以喹啉为基础的化合物,即氯喹(CQ)和硝氧(NQ),代表了一类新的抗癌药物。但是,CCA尚未阐明其功效和潜在机制。本研究利用CCA细胞探索CQ和NQ对FoxM1信号传导的抗癌活性和抑制作用。方法:用比色法3-(4,5-二甲基噻唑-2yl)评估CQ和NQ对细胞活力和增殖的影响。 )-5-(3-羧基甲氧基苯基)-(4-磺基苯基)-2H-四唑鎓(MTS分析)。分别使用克隆形成和伤口愈合试验研究了受CQ和NQ影响的菌落形成和细胞迁移。为了证明这些因子对FoxM1信号传导的影响,使用实时聚合酶链反应定量测定了靶基因的表达水平。结果:CQ和NQ以剂量和时间依赖性方式显着抑制HuCCT1和Huh28的细胞存活。 。使用迅速增殖的HuCCT1细胞进行的进一步研究表明,低剂量的化合物可明显抑制细胞增殖和集落形成。 CQ和NQ的治疗抑制了cyclin D1的表达,但增强了p21的表达。最重要的是,经过CQ和NQ处理后,致癌的FoxM1的表达显着下降,同时下调了各种FoxM1的下游靶标,包括cdc25b,CENP-B和survivin。此外,这些化合物显着削弱了HuCCT1的迁移并抑制了基质金属蛋白酶(MMP)-2和MMP-9的表达。结论:本研究首次报道了CQ和NQ对CCA细胞的抗癌作用,并且重点介绍了基于喹啉的化合物破坏FoxM1信号传导的作用机理的新见解。

著录项

相似文献

  • 外文文献
  • 中文文献
  • 专利
获取原文

客服邮箱:kefu@zhangqiaokeyan.com

京公网安备:11010802029741号 ICP备案号:京ICP备15016152号-6 六维联合信息科技 (北京) 有限公司©版权所有
  • 客服微信

  • 服务号