...
首页> 外文期刊>Journal of experimental & clinical cancer research : >Exosomal DLX6-AS1 from hepatocellular carcinoma cells induces M2 macrophage polarization to promote migration and invasion in hepatocellular carcinoma through microRNA-15a-5p/CXCL17 axis
【24h】

Exosomal DLX6-AS1 from hepatocellular carcinoma cells induces M2 macrophage polarization to promote migration and invasion in hepatocellular carcinoma through microRNA-15a-5p/CXCL17 axis

机译:来自肝细胞癌细胞的外泌体DLX6-AS1诱导M2巨噬细胞极化通过MicroRNA-15A-5P / CXCL17轴促进肝细胞癌中的迁移和侵袭

获取原文
           

摘要

Hepatocellular carcinoma (HCC) cells-secreted exosomes (exo) could stimulate M2 macrophage polarization and promote HCC progression, but the related mechanism of long non-coding RNA distal-less homeobox?6 antisense 1 (DLX6-AS1) with HCC-exo-mediated M2 macrophage polarization is largely ambiguous. Thereafter, this research was started to unearth the role of DLX6-AS1 in HCC-exo in HCC through M2 macrophage polarization and microRNA (miR)-15a-5p/C-X-C motif chemokine ligand 17 (CXCL17) axis. DLX6-AS1, miR-15a-5p and CXCL17 expression in HCC tissues and cells were tested. Exosomes were isolated from HCC cells with overexpressed DLX6-AS1 and co-cultured with M2 macrophages. MiR-15a-5p/CXCL17 down-regulation assays were performed in macrophages. The treated M2 macrophages were co-cultured with HCC cells, after which cell migration, invasion and epithelial mesenchymal transition were examined. The targeting relationships between DLX6-AS1 and miR-15a-5p, and between miR-15a-5p and CXCL17 were explored. In vivo experiment was conducted to detect the effect of exosomal DLX6-AS1-induced M2 macrophage polarization on HCC metastasis. Promoted DLX6-AS1 and CXCL17 and reduced miR-15a-5p exhibited in HCC. HCC-exo induced M2 macrophage polarization to accelerate migration, invasion and epithelial mesenchymal transition in HCC, which was further enhanced by up-regulated DLX6-AS1 but impaired by silenced DLX6-AS1. Inhibition of miR-15a-5p promoted M2 macrophage polarization to stimulate the invasion and metastasis of HCC while that of CXCL17 had the opposite effects. DLX6-AS1 mediated miR-15a-5p to target CXCL17. DLX6-AS1 from HCC-exo promoted metastasis in the lung by inducing M2 macrophage polarization in vivo. DLX6-AS1 from HCC-exo regulates CXCL17 by competitively binding to miR-15a-5p to induce M2 macrophage polarization, thus promoting HCC migration, invasion and EMT.
机译:肝细胞癌(HCC)细胞分泌的外泌体(EXO)可以刺激M2巨噬细胞偏振并促进HCC进展,但长期非编码RNA远端的相关机制与HCC-EXO - 介导的M2巨噬细胞极化在很大程度上是模糊的。此后,开始通过M2巨噬细胞极化和MICRRNA(MIR)-15A-5P / C-X-C型趋化因子17(CXCL17)轴线在HCC中的HCC-EXO中DLX6-AS1在HCC-EXO中的作用。测试HCC组织和细胞中的DLX6-AS1,miR-15a-5p和cxcl17表达。用过表达DLX6-AS1与HCC细胞分离出外泌体,并用M2巨噬细胞共培养。在巨噬细胞中进行miR-15a-5p / cxcl17下调测定。处理过的M2巨噬细胞与HCC细胞共培养,之后检查细胞迁移,侵袭和上皮间充质转换。探讨了DLX6-AS1和MIR-15A-5P之间的靶向关系,以及MIR-15A-5P和CXCL17之间的靶向关系。在体内实验中进行了检测外泌体DLX6-AS1诱导的M2巨噬细胞极化对HCC转移的影响。促进DLX6-AS1和CXCL17,并减少了HCC中展出的miR-15a-5p。 HCC-EXO诱导M2巨噬细胞极化加速HCC中的迁移,侵袭和上皮间充质转变,其通过上调的DLX6-AS1进一步增强,但通过沉默的DLX6-AS1损害。抑制miR-15a-5p的促进M2巨噬细胞极化,刺激HCC的侵袭和转移,而CXCL17具有相反的效果。 DLX6-AS1介导的MIR-15A-5P靶向CXCL17。来自HCC-EXO的DLX6-AS1通过在体内诱导M2巨噬细胞极化促进肺部转移。通过HCC-EXO的DLX6-AS1通过竞争性结合miR-15a-5p来调节CxCl17以诱导M2巨噬细胞极化,从而促进HCC迁移,侵袭和EMT。

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