首页> 外文OA文献 >Cytokine production and inflammation drive autophagy in the tumor microenvironment: Role of stromal caveolin-1 as a key regulator
【2h】

Cytokine production and inflammation drive autophagy in the tumor microenvironment: Role of stromal caveolin-1 as a key regulator

机译:细胞因子的产生和炎症驱动肿瘤微环境中的自噬:基质小窝蛋白-1作为关键调控因子的作用

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

Recently, we proposed a new paradigm for understanding the role of the tumor microenvironment in breast cancer onset and progression. In this model, cancer cells induce oxidative stress in adjacent fibroblasts. This, in turn, results in the onset of stromal autophagy, which produces recycled nutrients to “feed” anabolic cancer cells. However, it remains unknown how autophagy in the tumor microenvironment relates to inflammation, another key driver of tumorigenesis. To address this issue, here we employed a well-characterized co-culture system in which cancer cells induce autophagy in adjacent fibroblasts via oxidative stress and NFκB-activation. We show, using this co-culture system, that the same experimental conditions that result in an autophagic microenvironment, also drive in the production of numerous inflammatory mediators (including IL-6, IL-8, IL-10, MIp1α, IFNγ, RANTES (CCL5) and GMCSF). Furthermore, we demonstrate that most of these inflammatory mediators are individually sufficient to directly induce the onset of autophagy in fibroblasts. To further validate the in vivo relevance of these findings, we assessed the inflammatory status of Cav-1 (−/−) null mammary fat pads, which are a model of a bonafide autophagic microenvironment. Notably, we show that Cav-1 (−/−) mammary fat pads undergo infiltration with numerous inflammatory cell types, including lymphocytes, T-cells, macrophages and mast cells. Taken together, our results suggest that cytokine production and inflammation are key drivers of autophagy in the tumor microenvironment. These results may explain why a loss of stromal Cav-1 is a powerful predictor of poor clinical outcome in breast cancer patients, as it is a marker of both (1) autophagy and (2) inflammation in the tumor microenvironment. Lastly, hypoxia in fibroblasts was not sufficient to induce the full-blown inflammatory response that we observed during the co-culture of fibroblasts with cancer cells, indicating that key reciprocal interactions between cancer cells and fibroblasts may be required.
机译:最近,我们提出了一个新的范式,用于理解肿瘤微环境在乳腺癌发作和进展中的作用。在该模型中,癌细胞在邻近的成纤维细胞中诱导氧化应激。反过来,这会导致基质自噬的发生,基质自噬会产生回收的营养素,以“喂养”合成代谢癌细胞。然而,尚不清楚肿瘤微环境中的自噬与炎症的关系,炎症是肿瘤发生的另一个关键驱动因素。为了解决这个问题,我们在这里使用了一个特征明确的共培养系统,其中癌细胞通过氧化应激和NFκB激活在邻近的成纤维细胞中诱导自噬。我们显示,使用这种共培养系统,导致自噬微环境的相同实验条件也推动了许多炎症介质(包括IL-6,IL-8,IL-10,MIp1α,IFNγ,RANTES)的产生(CCL5)和GMCSF)。此外,我们证明大多数这些炎性介质单独足以直接诱导成纤维细胞中自噬的发作。为了进一步验证这些发现的体内相关性,我们评估了Cav-1(-/-)无效乳腺脂肪垫的炎症状态,该模型是一种真正自噬微环境的模型。值得注意的是,我们显示Cav-1(-/-)乳腺脂肪垫会浸润多种炎症细胞类型,包括淋巴细胞,T细胞,巨噬细胞和肥大细胞。两者合计,我们的结果表明,细胞因子的产生和炎症是肿瘤微环境中自噬的关键驱动力。这些结果可以解释为什么Cav-1的丢失是乳腺癌患者不良临床预后的有力预测因素,因为它既是肿瘤微环境中(1)自噬和(2)炎症的标志。最后,成纤维细胞中的缺氧不足以诱导我们在成纤维细胞与癌细胞共培养期间观察到的全面炎症反应,这表明癌细胞和成纤维细胞之间可能需要关键的相互作用。

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