...
首页> 外文期刊>Journal of molecular medicine: Official organ of the "Gesellschaft Deutscher Naturforscher und Arzte." >Systemic oxygenation weakens the hypoxia and hypoxia inducible factor 1α-dependent and extracellular adenosine-mediated tumor protection
【24h】

Systemic oxygenation weakens the hypoxia and hypoxia inducible factor 1α-dependent and extracellular adenosine-mediated tumor protection

机译:全身性氧合削弱了缺氧和缺氧诱导因子1α依赖性和细胞外腺苷介导的肿瘤保护

获取原文
获取原文并翻译 | 示例
           

摘要

Intratumoral hypoxia and hypoxia inducible factor-1α (HIF-1-α)-dependent CD39/CD73 ectoenzymes may govern the accumulation of tumor-protecting extracellular adenosine and signaling through A2A adenosine receptors (A2AR) in tumor microenvironments (TME). Here, we explored the conceptually novel motivation to use supplemental oxygen as a treatment to inhibit the hypoxia/HIF-1α-CD39/CD73-driven accumulation of extracellular adenosine in the TME in order to weaken the tumor protection. We report that hyperoxic breathing (60 % O2) decreased the TME hypoxia, as well as levels of HIF-1α and downstream target proteins of HIF-1α in the TME according to proteomic studies in mice. Importantly, oxygenation also downregulated the expression of adenosine-generating ectoenzymes and significantly lowered levels of tumor-protecting extracellular adenosine in the TME. Using supplemental oxygen as a tool in studies of the TME, we also identified FHL-1 as a potentially useful marker for the conversion of hypoxic into normoxic TME. Hyperoxic breathing resulted in the upregulation of antigen-presenting MHC class I molecules on tumor cells and in the better recognition and increased susceptibility to killing by tumor-reactive cytotoxic T cells. Therapeutic breathing of 60 % oxygen resulted in the significant inhibition of growth of established B16.F10 melanoma tumors and prolonged survival of mice. Taken together, the data presented here provide proof-of principle for the therapeutic potential of systemic oxygenation to convert the hypoxic, adenosine-rich and tumor-protecting TME into a normoxic and extracellular adenosine-poor TME that, in turn, may facilitate tumor regression. We propose to explore the combination of supplemental oxygen with existing immunotherapies of cancer.Key messages: Oxygenation decreases levels of tumor protecting hypoxia. Oxygenation decreases levels of tumor protecting extracellular adenosine. Oxygenation decreases expression of HIF-1alpha dependent tumor-protecting proteins. Oxygenation increases MHC class I expression and enables tumor regression.
机译:肿瘤内缺氧和缺氧诱导因子-1α(HIF-1-α)依赖性CD39 / CD73外切酶可能控制肿瘤微环境(TME)中保护肿瘤的细胞外腺苷的积累并通过A2A腺苷受体(A2AR)进行信号传导。在这里,我们探索了在概念上新颖的动机,即使用补充氧气来抑制缺氧/HIF-1α-CD39/ CD73驱动的TME中细胞外腺苷的蓄积,从而削弱对肿瘤的保护。我们报告说,根据小鼠蛋白质组学研究,高氧呼吸(60%O2)降低了TME的低氧以及TME中HIF-1α和HIF-1α的下游靶蛋白的水平。重要的是,氧合作用还下调了TME中产生腺苷的胞外酶的表达,并显着降低了肿瘤保护细胞外腺苷的水平。使用补充氧气作为TME研究的工具,我们还确定FHL-1是将缺氧转化为常氧TME的潜在有用标记。高氧呼吸导致肿瘤细胞上呈递抗原的MHC I类分子上调,并导致更好的识别和增加对肿瘤反应性细胞毒性T细胞杀伤的敏感性。 60%氧气的治疗性呼吸会显着抑制已建立的B16.F10黑色素瘤肿瘤的生长,并延长小鼠的存活时间。综上所述,此处提供的数据为全身性氧合治疗将低氧,富含腺苷和肿瘤保护性TME转化为缺氧和细胞外腺苷贫乏TME的治疗潜力提供了证明依据,进而可促进肿瘤消退。我们建议探索补充氧气与现有的癌症免疫疗法的结合。重要信息:氧合可降低肿瘤保护性缺氧的水平。氧合降低了肿瘤保护细胞外腺苷的水平。氧合减少HIF-1alpha依赖性肿瘤保护蛋白的表达。氧合增加了I类MHC的表达,并使肿瘤消退。

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