首页> 美国卫生研究院文献>Cell Regulation >ROCK and Nuclear Factor-κB–dependent Activation of Cyclooxygenase-2 by Rho GTPases: Effects on Tumor Growth and Therapeutic Consequences
【2h】

ROCK and Nuclear Factor-κB–dependent Activation of Cyclooxygenase-2 by Rho GTPases: Effects on Tumor Growth and Therapeutic Consequences

机译:ROCK和核因子-κB依赖性激活 Rho GTPases的环氧合酶-2:对肿瘤生长和治疗的影响 后果

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

Rho GTPases are overexpressed in a variety of human tumors contributing to both tumor proliferation and metastasis. Recently, several studies demonstrate an essential role of transcriptional regulation in Rho GTPases-induced oncogenesis. Herein, we demonstrate that RhoA, Rac1, and Cdc42 promote the expression of cyclooxygenase-2 (COX-2) at the transcriptional level by a mechanism that is dependent on the transcription factor nuclear factor-κB (NF-κB), but not Stat3, a transcription factor required for RhoA-induced tumorigenesis. With respect to RhoA, this effect is dependent on ROCK, but not PKN. Treatment of RhoA-, Rac1-, and Cdc42-transformed epithelial cells with Sulindac and NS-398, two well-characterized nonsteroid antiinflammatory drugs (NSAIDs), results in growth inhibition as determined by cell proliferation assays. Accordingly, tumor growth of RhoA-expressing epithelial cells in syngeneic mice is strongly inhibited by NS-398 treatment. The effect of NSAIDs over RhoA-induced tumor growth is not exclusively dependent on COX-2 because DNA-binding of NF-κB is also abolished upon NSAIDs treatment, resulting in complete loss of COX-2 expression. Finally, treatment of RhoA-transformed cells with Bay11-7083, a specific NF-κB inhibitor, leads to inhibition of cell proliferation. We suggest that treatment of human tumors that overexpress Rho GTPases with NSAIDs and drugs that target NF-κB could constitute a valid antitumoral strategy.
机译:Rho GTPases在多种人类肿瘤中过表达,导致肿瘤增殖和转移。最近,一些研究证明了转录调节在Rho GTPases诱导的肿瘤发生中的重要作用。在本文中,我们证明RhoA,Rac1和Cdc42在转录水平上通过依赖转录因子核因子-κB(NF-κB)而不是Stat3的机制促进环氧合酶2(COX-2)的表达。 ,是RhoA诱导的肿瘤发生所需的转录因子。对于RhoA,此效果取决于ROCK,但不取决于PKN。用Sulindac和NS-398(两种特征明确的非甾体抗炎药(NSAID))处理RhoA,Rac1-和Cdc42转化的上皮细胞,可通过细胞增殖试验确定其抑制生长。因此,NS-398治疗强烈抑制了同源小鼠中表达RhoA的上皮细胞的肿瘤生长。 NSAIDs对RhoA诱导的肿瘤生长的影响并不完全取决于COX-2,因为NSAIDs治疗也消除了NF-κB的DNA结合,导致COX-2表达完全丧失。最后,用特异性的NF-κBBay11-7083处理RhoA转化的细胞 抑制剂,导致细胞增殖的抑制。我们建议 NSAIDs和药物治疗过度表达Rho GTPases的人类肿瘤 靶向NF-κB可能构成有效的抗肿瘤策略。

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