...
首页> 外文期刊>Cell cycle >CTGF drives autophagy, glycolysis and senescence in cancer-associated fibroblasts via HIF1 activation, metabolically promoting tumor growth
【24h】

CTGF drives autophagy, glycolysis and senescence in cancer-associated fibroblasts via HIF1 activation, metabolically promoting tumor growth

机译:CTGF通过HIF1激活驱动癌症相关成纤维细胞的自噬,糖酵解和衰老,从而代谢促进肿瘤生长

获取原文
获取原文并翻译 | 示例

摘要

Previous studies have demonstrated that loss of caveolin-1 (Cav-1) in stromal cells drives the activation of the TGFβ signaling, with increased transcription of TGFβ target genes, such as connective tissue growth factor (CTGF). In addition, loss of stromal Cav-1 results in the metabolic reprogramming of cancer-associated fibroblasts, with the induction of autophagy and glycolysis. However, it remains unknown if activation of the TGFβ/CTGF pathway regulates the metabolism of cancerassociated fibroblasts. Therefore, we investigated whether CTGF modulates metabolism in the tumor microenvironment. For this purpose, CTGF was overexpressed in normal human fibroblasts or MDA-MB-231 breast cancer cells. Overexpression of CTGF induces HIF-1α-dependent metabolic alterations, with the induction of autophagy/mitophagy, senescence and glycolysis. Here, we show that CTGF exerts compartment-specific effects on tumorigenesis, depending on the cell-type. In a xenograft model, CTGF overexpressing fibroblasts promote the growth of co-injected MDA-MB-231 cells, without any increases in angiogenesis. Conversely, CTGF overexpression in MDA-MB-231 cells dramatically inhibits tumor growth in mice. Intriguingly, increased extracellular matrix deposition was seen in tumors with either fibroblast or MDA-MB-231 overexpression of CTGF. Thus, the effects of CTGF expression on tumor formation are independent of its extracellular matrix function, but rather depend on its ability to activate catabolic metabolism. As such, CTGF-mediated induction of autophagy in fibroblasts supports tumor growth via the generation of recycled nutrients, whereas CTGF-mediated autophagy in breast cancer cells suppresses tumor growth, via tumor cell self-digestion. Our studies shed new light on the compartment-specific role of CTGF in mammary tumorigenesis, and provide novel insights into the mechanism(s) generating a lethal tumor microenvironment in patients lacking stromal Cav-1. As loss of Cav-1 is a stromal marker of poor clinical outcome in women with primary breast cancer, dissecting the downstream signaling effects of Cav-1 are important for understanding disease pathogenesis, and identifying novel therapeutic targets.
机译:先前的研究表明,基质细胞中小窝蛋白1(Cav-1)的缺失会驱动TGFβ信号的激活,并增加TGFβ靶基因(例如结缔组织生长因子(CTGF))的转录。另外,间质Cav-1的丧失导致与癌症相关的成纤维细胞的代谢重编程,并诱导自噬和糖酵解。然而,尚不清楚TGFβ/ CTGF途径的激活是否调节与癌症相关的成纤维细胞的代谢。因此,我们调查了CTGF是否在肿瘤微环境中调节代谢。为此,CTGF在正常人成纤维细胞或MDA-MB-231乳腺癌细胞中过表达。 CTGF的过表达诱导HIF-1α依赖性代谢改变,并诱导自噬/有丝分裂,衰老和糖酵解。在这里,我们显示CTGF取决于细胞类型,在肿瘤发生上发挥区室特异性作用。在异种移植模型中,过度表达CTGF的成纤维细胞可促进共同注射的MDA-MB-231细胞的生长,而不会增加血管生成。相反,MDA-MB-231细胞中CTGF的过度表达可显着抑制小鼠的肿瘤生长。有趣的是,在CTGF成纤维细胞或MDA-MB-231过表达的肿瘤中,细胞外基质沉积增加。因此,CTGF表达对肿瘤形成的影响与其细胞外基质功能无关,而取决于其激活分解代谢的能力。因此,CTGF介导的成纤维细胞自噬诱导通过再生营养素的产生支持肿瘤的生长,而CTGF介导的乳腺癌细胞中的自噬通过肿瘤细胞的自我消化抑制肿瘤的生长。我们的研究为CTGF在乳腺肿瘤发生中的区室特异性作用提供了新的线索,并为缺乏基质Cav-1的患者产生致死性肿瘤微环境的机制提供了新见解。由于Cav-1的丧失是原发性乳腺癌妇女临床预后不良的根本标志,因此剖析Cav-1的下游信号传导作用对于理解疾病的发病机制和确定新的治疗靶点非常重要。

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