首页> 美国卫生研究院文献>American Journal of Cancer Research >Respiratory hyperoxia reverses immunosuppression by regulating myeloid-derived suppressor cells and PD-L1 expression in a triple-negative breast cancer mouse model
【2h】

Respiratory hyperoxia reverses immunosuppression by regulating myeloid-derived suppressor cells and PD-L1 expression in a triple-negative breast cancer mouse model

机译:呼吸道高氧通过调节三阴性乳腺癌小鼠模型中髓样来源的抑制细胞和PD-L1表达来逆转免疫抑制

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

Hypoxia plays an extensive role in the development of the tumor microenvironment (TME), particularly in mediating immunosuppression. Respiratory hyperoxia therapy has the potential to improve the effects of conventional cancer therapies via molecular mechanisms mediating antitumor immunity. Here, we investigated whether hyperoxia therapy can restore tumor immunity and inhibit lung metastases in a mouse model of triple-negative breast cancer (TNBC) by treating a 4T1 mammary carcinoma mouse model with normoxia (21% oxygen) or hyperoxia (60% oxygen) therapy, after tumor development. Using flow cytometry analysis, we observed significant organ-specific expansion of myeloid-derived suppressor cells (MDSCs) and protein expression upregulation of the programmed death-ligand 1 (PD-L1) in the hypoxic TME of 4T1 tumor-bearing mice maintained under normoxia conditions, with the TME converting to a T-cell immune-suppressive state as early as the premetastatic phase. Markedly, hyperoxia treatments ameliorated hypoxia levels in the lung TME and decreased the proportion of MDSCs and the expression of PD-L1 in both the primary tumor and in the metastatic lung, when compared to animals treated with respiratory normoxia therapy. In addition, the number of lung metastatic nodes fell from 90 per lung in the normoxic treated group to 13 per lung in the hyperoxic treated group (P < 0.05), with the latter having limited hyperoxia effects on primary tumor growth (mammary glands). Notably, hyperoxia therapy was characterized by the differential recruitment of CD4+ and CD8+ T-cells. Thus, our study confirms that hyperoxia therapy may be used to overcome TME immunosuppression and control the extend of lung metastases in TNBC. Importantly, changes in immunosuppressive MDSCs frequency and PD-L1 expression levels may serve as biomarkers of hypoxia levels in cancer affected tissues that can benefit from hyperoxia treatments.
机译:缺氧在肿瘤微环境(TME)的发展中起着广泛的作用,尤其是在介导免疫抑制方面。呼吸性高氧疗法有潜力通过介导抗肿瘤免疫力的分子机制来改善常规癌症疗法的效果。在这里,我们研究了高氧疗法是否可以通过用常氧(21%氧气)或高氧(60%氧气)治疗4T1乳腺癌小鼠模型来恢复三阴性乳​​腺癌(TNBC)小鼠模型中的肿瘤免疫力并抑制肺转移。肿瘤发展后进行治疗。使用流式细胞仪分析,我们观察到常氧维持4T1荷瘤小鼠低氧TME中髓样来源的抑制细胞(MDSCs)的器官特异性显着扩增和程序性死亡配体1(PD-L1)的蛋白质表达上调在TME转移至T细胞免疫抑制状态之前(早于转移前阶段)。明显地,与用呼吸性正常氧疗法治疗的动物相比,高氧疗法可改善肺TME中的低氧水平,并降低原发性肿瘤和转移性肺中MDSC的比例和PD-L1的表达。此外,肺转移结点的数量从常氧治疗组的每肺90个降至高氧治疗组的每肺13个(P <0.05),而后者对原发肿瘤(乳腺)的高氧影响有限。值得注意的是,高氧疗法的特征在于CD4 + 和CD8 + T细胞的差异募集。因此,我们的研究证实高氧疗法可用于克服TME免疫抑制和控制TNBC中肺转移的扩展。重要的是,免疫抑制性MDSCs频率和PD-L1表达水平的变化可能会成为受癌症影响的组织缺氧水平的生物标记,可以从高氧治疗中受益。

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