首页> 美国卫生研究院文献>Cancer Science >Hypoxia decreases macrophage glycolysis and M1 percentage by targeting microRNA‐30c and mTOR in human gastric cancer
【2h】

Hypoxia decreases macrophage glycolysis and M1 percentage by targeting microRNA‐30c and mTOR in human gastric cancer

机译:缺氧通过靶向microRNA-30c和mTOR降低人胃癌的巨噬细胞糖酵解和M1百分比

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

Macrophages are essential inflammatory cells which regulate the features of immune reactions within tumors. Many studies have reported their regulatory roles in immunity through cytokines and cell signaling. However, relatively few studies have focused on their metabolic features and mechanisms. We aimed to determine the signaling pathway regulating cell metabolism and the mechanism related to the regulation of human tumor‐associated macrophages ( s) in gastric cancer ( ). Tumor‐infiltrated macrophages were isolated from human tissues using magnetic beads, gene transcription was determined by real‐time , protein expression was monitored using western blots, metabolites were determined using , and transcriptional regulation was analyzed by the luciferase‐based reporter gene system. A significant decrease in microRNA (miR)‐30c and an increase in regulated in development and DNA damage responses 1 (REDD1) were detected in human s, the transcription of miR‐30c was negatively correlated with 1. MicroRNA‐30c expression was suppressed by hypoxia‐inducible factor‐1α activation and related to decreased activity as well as glycolysis in human s. Hypoxia‐regulated miR‐30c downregulated ‐1 expression by targeting its 3′ . Overexpression of miR‐30c or restored activity in macrophages with miR‐30c expression promoted M1 macrophage differentiation and function in s. Therefore, hypoxia in the human microenvironment suppressed the expression of miR‐30c, and decreased activity as well as glycolysis in s, thus inhibiting M1 differentiation and function. These results provide a novel metabolic strategy for tumor microenvironment‐based therapy.
机译:巨噬细胞是必需的炎性细胞,其调节肿瘤内免疫反应的特征。许多研究报告了它们通过细胞因子和细胞信号传导在免疫中的调节作用。然而,相对较少的研究集中在其代谢特征和机制上。我们旨在确定调节细胞代谢的信号传导途径以及与胃癌中与人类肿瘤相关的巨噬细胞调控有关的机制。使用磁珠从人体组织中分离出肿瘤浸润的巨噬细胞,通过实时测定基因转录,使用蛋白质印迹法监测蛋白质表达,使用蛋白质印迹法测定代谢物,并通过基于荧光素酶的报告基因系统分析转录调控。在人类s中检测到microRNA(miR)-30c的显着下降以及发育和DNA损伤反应1(REDD1)调控的增加,miR-30c的转录与1呈负相关。缺氧诱导因子-1α激活并与人类s活动减少以及糖酵解有关。低氧调节的miR-30c通过靶向其3'来下调-1表达。 miR-30c的过表达或具有miR-30c表达的巨噬细胞中活性的恢复促进了M1巨噬细胞的分化和功能。因此,人类微环境中的缺氧会抑制miR-30c的表达,并降低s的活性和糖酵解,从而抑制M1的分化和功能。这些结果为基于肿瘤微环境的治疗提供了新的代谢策略。

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