首页> 美国卫生研究院文献>other >Hypoxia mediates mitochondrial biogenesis in hepatocellular carcinoma to promote tumor growth via HMGB1 and TLR9 interaction
【2h】

Hypoxia mediates mitochondrial biogenesis in hepatocellular carcinoma to promote tumor growth via HMGB1 and TLR9 interaction

机译:低氧介导肝细胞癌的线粒体生物发生以通过HMGB1和TLR9相互作用促进肿瘤生长

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

The ability of cancer cells to survive and grow under hypoxic conditions has been known for decades, but the mechanisms remain poorly understood. Under certain conditions, cancer cells undergo changes in their bioenergetic profile to favor mitochondrial respiration by activating the peroxisome proliferator-activated receptor gamma (PGC-1α) and upregulating mitochondrial biogenesis. In this study, we hypothesized that augmented mitochondrial biogenesis plays a critical role for cancer cells to survive hypoxia. Consistent with this hypothesis, both hypoxic human hepatocellular carcinoma (HCC) tumors and HCC cell lines subjected to hypoxia increase mitochondrial biogenesis. Silencing of PGC-1α in hypoxic HCC cell lines halts their proliferation. Mechanistic investigations in vitro indicated that intracellular High Mobility Group Box (HMGB)-1 protein, a nuclear protein overexpressed in HCC, is essential for the process. Silencing of HMGB1 in hypoxic HCC cell lines resulted in a significant decrease in PGC-1α activation and mitochondrial biogenesis. Without HMGB1, hypoxic HCC cells had significantly reduced ATP production and decreased cellular proliferation and increased apoptosis. In a diethynitrosamine (DEN)-induced murine model of HCC, genetic blocking of HMGB1 in the hypoxic tumors resulted in a significant decrease in tumor growth. Tumors lacking HMGB1 had a significant reduction in mitochondrial biogenesis and a significant increase in mitochondrial dysfunction. Further in vitro mechanistic experiments indicated that, during hypoxia, HMGB1 translocates from the nucleus to the cytoplasm and binds to cytoplasmic Toll-like receptor (TLR)-9. This binding leads to the activation of p38 and subsequent phosphorylation of PGC-1α with resultant upregulation of mitochondrial biogenesis.ConclusionTaken together, our findings suggest that during hypoxia HMGB1 upregulates mitochondrial biogenesis in HCC cancer cells promoting tumor survival and proliferation.
机译:癌细胞在低氧条件下生存和生长的能力已有数十年的历史,但其机理仍知之甚少。在某些条件下,癌细胞会通过激活过氧化物酶体增殖物激活受体γ(PGC-1α)并上调线粒体的生物发生,从而改变其生物能谱,从而促进线粒体呼吸。在这项研究中,我们假设线粒体生物合成的增强对癌细胞生存缺氧起着至关重要的作用。与此假设一致,缺氧的人类肝细胞癌(HCC)肿瘤和遭受缺氧的HCC细胞系都会增加线粒体的生物发生。缺氧的HCC细胞株中PGC-1α的沉默会阻止其增殖。体外机制研究表明,细胞内高迁移率族盒(HMGB)-1蛋白是HCC中过表达的一种核蛋白,对于该过程至关重要。缺氧的HCC细胞系中HMGB1沉默导致PGC-1α激活和线粒体生物发生显着减少。没有HMGB1,低氧HCC细胞的ATP产生显着减少,细胞增殖减少,凋亡增加。在饮食亚硝胺(DEN)诱导的HCC鼠模型中,缺氧肿瘤中HMGB1的基因阻断导致肿瘤生长显着减少。缺乏HMGB1的肿瘤线粒体的生物发生显着减少,线粒体功能障碍的显着增加。进一步的体外机制实验表明,在缺氧期间,HMGB1从细胞核转移到细胞质,并与细胞质Toll样受体(TLR)-9结合。这种结合导致p38的激活和随后PGC-1α的磷酸化,从而导致线粒体生物发生的上调。结论,我们的研究结果表明,在缺氧期间HMGB1上调HCC癌细胞中的线粒体生物发生,从而促进肿瘤存活和增殖。

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