...
首页> 外文期刊>The Prostate >Activation of silenced tumor suppressor genes in prostate cancer cells by a novel energy restriction-mimetic agent
【24h】

Activation of silenced tumor suppressor genes in prostate cancer cells by a novel energy restriction-mimetic agent

机译:新型能量限制模拟剂激活前列腺癌细胞中沉默的抑癌基因

获取原文
获取原文并翻译 | 示例

摘要

Background. Targeting tumor metabolism by energy restriction-mimetic agents (ERMAs) has emerged as a strategy for cancer therapy/prevention. Evidence suggests a mechanistic link between ERMA-mediated antitumor effects and epigenetic gene regulation. Methods. Microarray analysis showed that a novel thiazolidinedione-derived ERMA, CG-12, and glucose deprivation could suppress DNA methyltransferase (DNMT)1 expression and reactivate DNA methylation-silenced tumor suppressor genes in LNCaP prostate cancer cells. Thus, we investigated the effects of a potent CG-12 derivative, CG-5, vis-à-vis 2-deoxyglucose, glucose deprivation and/or 5-aza-deoxycytidine, on DNMT isoform expression (Western blotting, RT-PCR), DNMT1 transcriptional activation (luciferase reporter assay), and expression of genes frequently hypermethylated in prostate cancer (quantitative real-time PCR). Promoter methylation was assessed by pyrosequencing analysis. SiRNA-mediated knockdown and ectopic expression of DNMT1 were used to validate DNMT1 as a target of CG-5. Results. CG-5 and glucose deprivation upregulated the expression of DNA methylation-silenced tumor suppressor genes, including GADD45a, GADD45b, IGFBP3, LAMB3, BASP1, GPX3, and GSTP1, but also downregulated methylated tumor/invasion-promoting genes, including CD44, S100A4, and TACSTD2. In contrast, 5-aza-deoxycytidine induced global reactivation of these genes. CG-5 mediated these epigenetic effects by transcriptional repression of DNMT1, which was associated with reduced expression of Sp1 and E2F1. SiRNA-mediated knockdown and ectopic expression of DNMT1 corroborated DNMT1's role in the modulation of gene expression by CG-5. Pyrosequencing revealed differential effects of CG-5 versus 5-aza-deoxycytidine on promoter methylation in these genes. Conclusions. These findings reveal a previously uncharacterized epigenetic effect of ERMAs on DNA methylation-silenced tumor suppressor genes, which may foster novel strategies for prostate cancer therapy.
机译:背景。通过能量限制模拟剂(ERMA)靶向肿瘤代谢已成为癌症治疗/预防的策略。有证据表明,ERMA介导的抗肿瘤作用与表观遗传基因调控之间存在机械联系。方法。芯片分析表明,新型的噻唑烷二酮衍生的ERMA,CG-12和葡萄糖剥夺可以抑制LNCaP前列腺癌细胞中DNA甲基转移酶(DNMT)1的表达并重新激活DNA甲基化沉默的肿瘤抑制基因。因此,我们研究了有效的CG-12衍生物CG-5相对于2-脱氧葡萄糖,葡萄糖剥夺和/或5-氮杂脱氧胞苷对DNMT亚型表达的影响(Western blotting,RT-PCR) ,DNMT1转录激活(荧光素酶报告基因测定)和前列腺癌中经常发生甲基化的基因表达(实时定量PCR)。启动子甲基化通过焦磷酸测序分析进行评估。 SiRNA介导的敲低和异位表达的DNMT1被用来验证DNMT1作为CG-5的靶标。结果。 CG-5和葡萄糖剥夺上调了DNA甲基化沉默的抑癌基因的表达,包括GADD45a,GADD45b,IGFBP3,LAMB3,BASP1,GPX3和GSTP1,但还下调了甲基化肿瘤/侵袭促进基因,包括CD44,S100A4,和TACSTD2。相反,5-氮杂-脱氧胞苷诱导这些基因的整体活化。 CG-5通过DNMT1的转录抑制来介导这些表观遗传效应,这与Sp1和E2F1的表达降低有关。 SiRNA介导的DNMT1的敲低和异位表达证实了DNMT1在CG-5调控基因表达中的作用。焦磷酸测序揭示了CG-5与5-氮杂-脱氧胞苷对这些基因中启动子甲基化的不同作用。结论。这些发现揭示了ERMA对DNA甲基化沉默的肿瘤抑制基因的前所未有的表观遗传学作用,这可能为前列腺癌治疗树立新的策略。

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