首页> 外文OA文献 >Microenvironmental Stiffness Enhances Glioma Cell Proliferation by Stimulating Epidermal Growth Factor Receptor Signaling
【2h】

Microenvironmental Stiffness Enhances Glioma Cell Proliferation by Stimulating Epidermal Growth Factor Receptor Signaling

机译:微环境僵硬通过刺激表皮生长因子受体信号增强胶质瘤细胞增殖

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

The aggressive and rapidly lethal brain tumor glioblastoma (GBM) is associated with profound tissue stiffening and genomic lesions in key members of the epidermal growth factor receptor (EGFR) pathway. Previous studies from our laboratory have shown that increasing microenvironmental stiffness in culture can strongly enhance glioma cell behaviors relevant to tumor progression, including proliferation, yet it has remained unclear whether stiffness and EGFR regulate proliferation through common or independent signaling mechanisms. Here we test the hypothesis that microenvironmental stiffness regulates cell cycle progression and proliferation in GBM tumor cells by altering EGFR-dependent signaling. We began by performing an unbiased reverse phase protein array screen, which revealed that stiffness modulates expression and phosphorylation of a broad range of signals relevant to proliferation, including members of the EGFR pathway. We subsequently found that culturing human GBM tumor cells on progressively stiffer culture substrates both dramatically increases proliferation and facilitates passage through the G1/S checkpoint of the cell cycle, consistent with an EGFR-dependent process. Western Blots showed that increasing microenvironmental stiffness enhances the expression and phosphorylation of EGFR and its downstream effector Akt. Pharmacological loss-of-function studies revealed that the stiffness-sensitivity of proliferation is strongly blunted by inhibition of EGFR, Akt, or PI3 kinase. Finally, we observed that stiffness strongly regulates EGFR clustering, with phosphorylated EGFR condensing into vinculin-positive focal adhesions on stiff substrates and dispersing as microenvironmental stiffness falls to physiological levels. Our findings collectively support a model in which tissue stiffening promotes GBM proliferation by spatially and biochemically amplifying EGFR signaling.
机译:侵袭性和快速致死性脑肿瘤胶质母细胞瘤(GBM)与表皮生长因子受体(EGFR)通路关键成员中的深部组织变硬和基因组损伤有关。我们实验室的先前研究表明,培养物中微环境硬度的增加可以强烈增强与肿瘤进展相关的神经胶质瘤细胞行为,包括增殖,但还不清楚硬度和EGFR是否通过共同或独立的信号传导机制调节增殖。在这里,我们测试的假设是微环境刚度通过改变EGFR依赖性信号传导来调节GBM肿瘤细胞的细胞周期进程和增殖。我们从进行无偏反相蛋白质阵列筛选开始,该筛选揭示了硬度调节与增殖相关的广泛信号的表达和磷酸化,包括EGFR途径的成员。我们随后发现,在逐渐变硬的培养底物上培养人GBM肿瘤细胞既可以显着增加增殖,又可以促进通过细胞周期的G1 / S检查点,这与EGFR依赖性过程一致。 Western Blot显示,微环境硬度的增加可增强EGFR及其下游效应物Akt的表达和磷酸化。药理学上的功能丧失研究表明,通过抑制EGFR,Akt或PI3激酶,增殖的刚度敏感性大大减弱。最后,我们观察到刚度强烈调节EGFR簇集,磷酸化的EGFR凝结在硬质底物上的纽蛋白阳性粘着斑上,并随着微环境刚度下降到生理水平而分散。我们的发现共同支持一种模型,其中组织变硬通过空间和生化放大EGFR信号传导促进GBM增殖。

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