...
首页> 外文期刊>Hepatology: Official Journal of the American Association for the Study of Liver Diseases >Hypoxia Mediates Mitochondrial Biogenesis in Hepatocellular Carcinoma to Promote Tumor Growth Through HMGB1 and TLR9 Interaction
【24h】

Hypoxia Mediates Mitochondrial Biogenesis in Hepatocellular Carcinoma to Promote Tumor Growth Through HMGB1 and TLR9 Interaction

机译:缺氧在肝细胞癌中介导线粒体生物发生,以通过HMGB1和TLR9相互作用促进肿瘤生长

获取原文
获取原文并翻译 | 示例
           

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 coactivator 1 alpha (PGC-1 alpha) and up-regulating 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-1 alpha in hypoxic HCC cell lines halts their proliferation. Mechanistic investigations in vitro indicated that intracellular high mobility group box 1 (HMGB1) 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-1 alpha activation and mitochondrial biogenesis. Without HMGB1, hypoxic HCC cells had significantly reduced adenosine triphosphate production, decreased cellular proliferation, and increased apoptosis. In a diethylnitrosamine-induced murine model of HCC, genetic blocking of HMGB1 in 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-9. This binding leads to activation of p38 and subsequent phosphorylation of PGC-1 alpha, with resultant up-regulation of mitochondrial biogenesis. Conclusion: Taken together, our findings suggest that during hypoxia HMGB1 up-regulates mitochondrial biogenesis in HCC cancer cells, promoting tumor survival and proliferation.
机译:几十年来,癌细胞在缺氧条件下生存和生长的能力已经着名,但机制仍然明白。在某些条件下,癌细胞通过激活过氧化物体增殖物激活的受体γ1α(PGC-1α)和Up-Consemation线粒体生物发生来接受其生物能曲线的变化以偏及线粒体呼吸。在这项研究中,我们假设增强线粒体生物发生在癌细胞生存缺氧中发挥着关键作用。与该假设一致,缺氧人类肝细胞癌(HCC)肿瘤和HCC细胞系经受缺氧增加了线粒体生物发生。脱氧HCC细胞系中PGC-1α的沉默停止了它们的增殖。体外机械调查表明,细胞内高迁移率组箱1(HMGB1)蛋白是HCC中过表达的核蛋白对该过程至关重要。脱氧HCC细胞系中HMGB1的沉默导致PGC-1α激活和线粒体生物发生的显着降低。如果没有HMGB1,缺氧HCC细胞显着降低了三磷酸三磷酸盐产生,细胞增殖降低,细胞凋亡增加。在二乙基亚胺诱导的HCC鼠模型中,缺氧肿瘤中HMGB1的遗传阻断导致肿瘤生长显着降低。缺乏HMGB1的肿瘤对线粒体生物发生显着降低,并且线粒体功能障碍的显着增加。进一步的体外机械实验表明,在缺氧HMGB1期间从细胞核转移到细胞质并结合细胞质Toll样受体-9。该结合导致P38的活化和后续磷酸化PGC-1α,得到了线粒体生物的结果上调。结论:我们的研究结果表明,在缺氧HMGB1期间,在HCC癌细胞中调节线粒体生物发生,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
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