首页> 外文期刊>Journal of immunology research. >KDM1A Promotes Immunosuppression in Hepatocellular Carcinoma by Regulating PD-L1 through Demethylating MEF2D
【24h】

KDM1A Promotes Immunosuppression in Hepatocellular Carcinoma by Regulating PD-L1 through Demethylating MEF2D

机译:通过去甲基化MEF2D调节PD-L1,KDM1A通过调节PD-L1来促进肝细胞癌免疫抑制

获取原文
           

摘要

Background . Immune checkpoint inhibitor therapy targeting antiprogrammed cell death-1 (anti-PD-1) or its ligand (anti-PD-L1) is effective in the treatment of some hepatocellular carcinomas (HCC). Hence, further identification of biological targets related to PD-L1 regulation in HCC is beneficial to improve the clinical efficacy of immunotherapy. Some HCC cells express lysine-specific demethylase 1A (KDM1A), which is implicated in the reduced survival time of patients. Here, we studied whether the level of PD-L1 and the immunosuppression are regulated by KDM1A and its miRNA in HCC cells. Methods . In the present study, we studied clinical data from The Cancer Genome Atlas (TCGA) database. We performed qPCR and western blotting assays to measure the expression level of genes of interest. PD-L1 expression was also analyzed by FACS. Clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 was used to generate gene knockout cells to investigate the relationships of genes of interest. We also developed a reporter gene assay (RGA) to explore the changes in T cell-induced antitumor immunity relative to PD-L1 expression in HCC cells. The binding between proteins and promoters or miRNAs and their target genes was explored by luciferase reporter assays. Results . The results showed that PD-L1 and KDM1A were increased in HCC patients and cells, and KDM1A promoted the expression of PD-L1 in HCC cells. Our findings showed that the enhancement of PD-L1 expression was not attributed to mitochondrial dysfunction caused by increases in KDM1A in HCC cells. Furthermore, we observed a lower level of MEF2D methylation in HCC cells than in normal human liver cells. Demethylated MEF2D could bind to the promoter of PD-L1 and activate its expression, while KDM1A interacted with MEF2D and acted as a demethylase to reduce its methylation. Moreover, a new miRNA, miR-329-3p, targeting KDM1A was found to regulate the PD-L1 expression profile in HCC cells. In the xenograft model, the tumors treated with miR-329-3p showed growth inhibition. Conclusions . Mechanistically, miR-329-3p inhibits tumor cellular immunosuppression and reinforces the response of tumor cells to T cell-induced cytotoxic effect by targeting KDM1A mRNA and downregulating its expression, which contributed to MEF2D demethylation and activation of PD-L1 expression.
机译:背景 。免疫检验点抑制剂疗法靶向antiprogrammed细胞死亡-1(抗PD-1)或它的配体(抗PD-L1)是有效的一些肝细胞癌(HCC)的治疗。因此,在HCC相关的PD-L1调节生物靶标进一步鉴定有利于提高免疫治疗的临床疗效。一些HCC细胞表达赖氨酸 - 特异性脱甲基酶1A(KDM1A),这是在患者的存活率降低时间牵连。在这里,我们研究是否PD-L1和免疫抑制的水平是由KDM1A及其中的miRNA肝癌细胞调控。方法 。在本研究中,我们研究了从癌症基因组图谱(TCGA)数据库的临床数据。我们进行了定量PCR和免疫印迹试验来测量的目的基因的表达水平。还通过FACS分析PD-L1表达。集群定期相互间隔短回文重复(CRISPR)/ Cas9用于产生基因敲除细胞研究的目的基因的关系。我们还开发了报告基因测定法(RGA),探讨在T细胞诱导的抗肿瘤免疫相对于PD-L1表达在HCC细胞中的变化。蛋白质和推动者或miRNA及其靶基因之间的结合是由荧光素酶报告基因测定研究。结果 。结果表明,PD-L1和KDM1A HCC患者和细胞增加,并且促进KDM1A HCC细胞中PD-L1的表达。我们的研究结果表明,PD-L1表达的增强不是归因于由肝癌细胞增加KDM1A线粒体功能障碍。此外,我们观察到MEF2D甲基化的较低水平在HCC细胞中比在正常的人肝细胞。脱甲基化MEF2D能结合PD-L1的启动子和激活其表达,而KDM1A相互作用与MEF2D和充当脱甲基酶,以减少它的甲基化。此外,一个新的miRNA,的miR-329-3p,靶向KDM1A发现调节在HCC细胞中的PD-L1表达谱。在该异种移植模型中,用的miR-329-3p处理的肿瘤显示出生长抑制。结论。在机制上,的miR-329-3p抑制肿瘤细胞的免疫抑制和增强的肿瘤细胞与T细胞诱导细胞毒性作用的通过靶向KDM1A mRNA和下调其表达,这有助于去甲基化MEF2D和PD-L1表达的激活的反应。

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