...
首页> 外文期刊>Cell death & disease. >Subverted regulation of Nox1 NADPH oxidase-dependent oxidant generation by protein disulfide isomerase A1 in colon carcinoma cells with overactivated KRas
【24h】

Subverted regulation of Nox1 NADPH oxidase-dependent oxidant generation by protein disulfide isomerase A1 in colon carcinoma cells with overactivated KRas

机译:蛋白二硫键异构酶A1对KRas过度活化的结肠癌细胞Nox1 NADPH氧化酶依赖性氧化剂产生的颠覆调节

获取原文
   

获取外文期刊封面封底 >>

       

摘要

Protein disulfide isomerases including PDIA1 are implicated in cancer progression, but underlying mechanisms are unclear. PDIA1 is known to support vascular Nox1 NADPH oxidase expression/activation. Since deregulated reactive oxygen species (ROS) production underlies tumor growth, we proposed that PDIA1 is an upstream regulator of tumor-associated ROS. We focused on colorectal cancer (CRC) with distinct KRas activation levels. Analysis of RNAseq databanks and direct validation indicated enhanced PDIA1 expression in CRC with constitutive high (HCT116) vs. moderate (HKE3) and basal (Caco2) Ras activity. PDIA1 supported Nox1-dependent superoxide production in CRC; however, we first reported a dual effect correlated with Ras-level activity: in Caco2 and HKE3 cells, loss-of-function experiments indicate that PDIA1 sustains Nox1-dependent superoxide production, while in HCT116 cells PDIA1 restricted superoxide production, a behavior associated with increased Rac1 expression/activity. Transfection of Rac1G12V active mutant into HKE3 cells induced PDIA1 to become restrictive of Nox1-dependent superoxide, while in HCT116 cells treated with Rac1 inhibitor, PDIA1 became supportive of superoxide. PDIA1 silencing promoted diminished cell proliferation and migration in HKE3, not detectable in HCT116 cells. Screening of cell signaling routes affected by PDIA1 silencing highlighted GSK3β and Stat3. Also, E-cadherin expression after PDIA1 silencing was decreased in HCT116, consistent with PDIA1 support of epithelial–mesenchymal transition. Thus, Ras overactivation switches the pattern of PDIA1-dependent Rac1/Nox1 regulation, so that Ras-induced PDIA1 bypass can directly activate Rac1. PDIA1 may be a crucial regulator of redox-dependent adaptive processes related to cancer progression.
机译:包括PDIA1在内的蛋白质二硫键异构酶与癌症进展有关,但尚不清楚其潜在机制。已知PDIA1支持血管Nox1 NADPH氧化酶表达/激活。由于失活的活性氧(ROS)产生是肿瘤生长的基础,因此我们提出PDIA1是肿瘤相关ROS的上游调节剂。我们专注于具有不同KRas激活水平的结直肠癌(CRC)。对RNAseq数据库的分析和直接验证表明,CRC中的PDIA1表达增强,具有组成性高(HCT116),中度(HKE3)和基础(Caco2)Ras活性。 PDIA1支持CRC中Nox1依赖性超氧化物的产生;然而,我们首先报道了与Ras水平活性相关的双重效应:在Caco2和HKE3细胞中,功能丧失的实验表明PDIA1维持Nox1依赖性超氧化物的产生,而在HCT116细胞中PDIA1限制了超氧化物的产生,这种行为与增加Rac1表达/活性。 Rac1G12V活性突变体转染到HKE3细胞中会诱导PDIA1成为Nox1依赖性超氧化物的限制性酶,而在用Rac1抑制剂处理的HCT116细胞中,PDIA1成为超氧化物的支持物。 PDIA1沉默促进减少HKE3中的细胞增殖和迁移,而在HCT116细胞中则无法检测到。受PDIA1沉默影响的细胞信号传导途径的筛选突出显示了GSK3β和Stat3。此外,HCT116中PDIA1沉默后E-钙粘着蛋白表达降低,这与PDIA1支持上皮-间质转化有关。因此,Ras过度激活会切换依赖PDIA1的Rac1 / Nox1调节模式,因此Ras诱导的PDIA1旁路可以直接激活Rac1。 PDIA1可能是与癌症进展有关的氧化还原依赖性适应性过程的关键调节剂。

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