首页> 美国卫生研究院文献>Proceedings of the National Academy of Sciences of the United States of America >PNAS Plus: Hypoxia-induced switch in SNAT2/SLC38A2 regulation generates endocrine resistance in breast cancer
【2h】

PNAS Plus: Hypoxia-induced switch in SNAT2/SLC38A2 regulation generates endocrine resistance in breast cancer

机译:PNAS Plus:缺氧诱导的SNAT2 / SLC38A2调节开关可在乳腺癌中产生内分泌抵抗力

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

Tumor hypoxia is associated with poor patient outcomes in estrogen receptor-α–positive (ERα+) breast cancer. Hypoxia is known to affect tumor growth by reprogramming metabolism and regulating amino acid (AA) uptake. Here, we show that the glutamine transporter, SNAT2, is the AA transporter most frequently induced by hypoxia in breast cancer, and is regulated by hypoxia both in vitro and in vivo in xenografts. SNAT2 induction in MCF7 cells was also regulated by ERα, but it became predominantly a hypoxia-inducible factor 1α (HIF-1α)–dependent gene under hypoxia. Relevant to this, binding sites for both HIF-1α and ERα overlap in SNAT2’s cis-regulatory elements. In addition, the down-regulation of SNAT2 by the ER antagonist fulvestrant was reverted in hypoxia. Overexpression of SNAT2 in vitro to recapitulate the levels induced by hypoxia caused enhanced growth, particularly after ERα inhibition, in hypoxia, or when glutamine levels were low. SNAT2 up-regulation in vivo caused complete resistance to antiestrogen and, partially, anti-VEGF therapies. Finally, high SNAT2 expression levels correlated with hypoxia profiles and worse outcome in patients given antiestrogen therapies. Our findings show a switch in the regulation of SNAT2 between ERα and HIF-1α, leading to endocrine resistance in hypoxia. Development of drugs targeting SNAT2 may be of value for a subset of hormone-resistant breast cancer.
机译:肿瘤缺氧与雌激素受体α阳性(ERα + )乳腺癌患者预后不良有关。缺氧通过重新编程代谢和调节氨基酸(AA)摄取来影响肿瘤的生长。在这里,我们显示出谷氨酰胺转运蛋白SNAT2是乳腺癌中低氧最常诱导的AA转运蛋白,在异种移植体内和体外均受到缺氧的调节。 ERα也调节了MCF7细胞中SNAT2的诱导,但在低氧条件下,它主要成为了一种缺氧诱导因子1α(HIF-1α)依赖性基因。与此相关,HIF-1α和ERα的结合位点在SNAT2的顺式调节元件中重叠。另外,在缺氧状态下,ER拮抗剂氟维司群对SNAT2的下调得以恢复。体外SNAT2的过表达可概括缺氧诱导的水平,导致生长增强,尤其是在ERα抑制后,缺氧或谷氨酰胺水平较低时。体内SNAT2的上调引起对抗雌激素剂的完全耐药,并部分导致抗VEGF治疗。最后,在接受抗雌激素治疗的患者中,高SNAT2表达水平与缺氧状况和较差的预后相关。我们的发现表明,ERNAT和HIF-1α之间SNAT2的调控发生改变,导致缺氧时的内分泌抵抗力。靶向SNAT2的药物的开发可能对一部分激素抵抗性乳腺癌有价值。

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