首页> 外文OA文献 >Genome-wide DNA methylation and hydroxymethylation analysis reveal human menstrual blood-derived stem cells inhibit hepatocellular carcinoma growth through oncogenic pathway suppression via regulating 5-hmC in enhancer elements
【2h】

Genome-wide DNA methylation and hydroxymethylation analysis reveal human menstrual blood-derived stem cells inhibit hepatocellular carcinoma growth through oncogenic pathway suppression via regulating 5-hmC in enhancer elements

机译:基因组DNA甲基化和羟甲基化分析显示人性血液衍生的干细胞通过调节5-HMC在增强子元素中通过调节5-HMC来抑制肝细胞癌生长

代理获取
本网站仅为用户提供外文OA文献查询和代理获取服务,本网站没有原文。下单后我们将采用程序或人工为您竭诚获取高质量的原文,但由于OA文献来源多样且变更频繁,仍可能出现获取不到、文献不完整或与标题不符等情况,如果获取不到我们将提供退款服务。请知悉。

摘要

Abstract Background Epigenetic alteration is an important indicator of crosstalk between cancer cells and surrounding microenvironment components including mesenchymal stem cells (MSC). Human menstrual blood-derived stem cells (MenSCs) are novel source of MSCs which exert suppressive effects on cancers via multiple components of microenvironmental paracrine signaling. However, whether MenSCs play a crucial role in the epigenetic regulation of cancer cells remains unknown. Methods Epigenetic alterations of hepatocellular carcinoma (HCC) mediated by MenSCs were examined by immunofluorescence, ELISA, and RT-PCR assays. The suppressive impact of MenSCs on HCC was investigated in vitro using CCK8, apoptosis, wound healing, and invasion assays and in vivo using a xenograft mice model. MeDIP-seq, hMeDIP-seq, and RNA-seq were used to identify the genome-wide pattern of DNA methylation and hydroxymethylation in HCC cells after MenSC therapy. Results We show that HCC cells display distinct genome-wide alterations in DNA hydroxymethylation and methylation after MenSC therapy. MenSCs exert an inhibitory effect on HCC growth via regulating 5-hmC and 5-mC abundance in the regulatory regions of oncogenic pathways including PI3K/AKT and MAPK signaling, especially in enhancers and promoters. FOXO3 expression is rescued via reversal of 5-hmC and 5-mC levels in its enhancers and contributes to the activation of downstream apoptosis. Inactivation of the MAPK pathway further disrupts c-myc-mediated epithelial-mesenchymal transitions (EMT). Additionally, chemotherapy resistance-associated genes including ID4 and HMGA1 are suppressed via amending 5-hmC and 5-mC abundance at their regulatory regions. HMGA1 and BYSL might be potential targets for gene-modified MSC therapy. Conclusions Our results confirm that MSCs could regulate the epigenetic mechanism of HCC cells and provide a novel concept for a modified MSC strategy or combination therapy with chemotherapeutics based on epigenetics.
机译:摘要背景表观遗传改变是癌细胞和周围的微环境组分(包括间充质干细胞(MSC)之间的串扰的重要指标。人经期血液衍生的干细胞(Menscs)是MSC的新型MSC,其通过微环境甲烷信号传导的多种组分对癌症产生抑制作用。但是,Menscs是否在癌细胞的表观遗传调节中发挥至关重要的作用仍然未知。方法采用免疫荧光,ELISA和RT-PCR测定检查Menscs介导的肝细胞癌(HCC)的表观遗传改变。使用卵黄移植小鼠模型,在体外研究Menscs对HCC对HCC的抑制作用,并使用异种移植小鼠模型在体内进行侵袭测定。 MEDIP-SEQ,HMEDIP-SEQ和RNA-SEQ用于在MensC治疗后鉴定HCC细胞中DNA甲基化和羟甲基的基因组模式。结果表明,HCC细胞在Mensc治疗后显示DNA羟甲基化和甲基化的不同基因组改变。 Menscs通过调节5-HMC和5-MC丰度在包括PI3K / AKT和MAPK信号,特别是在增强剂和启动子的致癌途径中的5-HMC和5-MC丰度对HCC生长产生抑制作用。 FOXO3表达通过增强剂的5-HMC和5MC水平的逆转来拯救,并有助于激活下游细胞凋亡。 MAPK途径的失活进一步破坏了C-MYC介导的上皮 - 间充质转换(EMT)。另外,通过在其调节区修补5-HMC和5-MC丰度,抑制了包括ID4和HMGA1的化疗抗性相关基因。 HMGA1和BYSL可能是基因改性MSC治疗的潜在目标。结论我们的结果证实,MSCs可以调节HCC细胞的表观遗传机制,并为基于表观遗传学的化学治疗剂提供改进的MSC策略或组合治疗的新概念。

著录项

相似文献

  • 外文文献
  • 中文文献
  • 专利
代理获取

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