...
首页> 外文期刊>Oncogene >The p16INK4A tumor suppressor regulates cellular oxidative stress
【24h】

The p16INK4A tumor suppressor regulates cellular oxidative stress

机译:p16INK4A肿瘤抑制因子调节细胞氧化应激

获取原文
   

获取外文期刊封面封底 >>

       

摘要

Mutations or deletions in the cyclin-dependent kinase inhibitor p16 ~(INK4A ) are associated with multiple cancer types, but are more commonly found in melanoma tumors and associated with familial melanoma predisposition. Although p16 is thought to function as a tumor suppressor by negatively regulating the cell cycle, it remains unclear why the genetic compromise of p16 predisposes to melanoma over other cancers. Here we describe a novel role for p16 in regulating oxidative stress in several cell types, including melanocytes. Expression of p16 was rapidly upregulated following ultraviolet-irradiation and in response to H_(2)O_(2)-induced oxidative stress in a p38 stress-activated protein kinase-dependent manner. Knockdown of p16 using small interfering RNA increased intracellular reactive oxygen species (ROS) and oxidative (8-oxoguanine) DNA damage, which was further enhanced by H_(2)O_(2) treatment. Elevated ROS levels were also observed in p16-depleted human keratinocytes and in whole skin and dermal fibroblasts from Cdkn2a -deficient mice. Aberrant ROS and p38 signaling in Cdkn2a -deficient fibroblasts was normalized by expression of exogenous p16. The effect of p16 depletion on ROS was not recapitulated by the knockdown of retinoblastoma protein (Rb) and did not require Rb. Finally, p16-mediated suppression of ROS could not be attributed to the potential effects of p16 on cell cycle phase. These findings suggest a potential alternate Rb-independent tumor-suppressor function of p16 as an endogenous regulator of carcinogenic intracellular oxidative stress. Compared with keratinocytes and fibroblasts, we also found increased susceptibility of melanocytes to oxidative stress in the context of p16 depletion, which may explain why the compromise of p16 predisposes to melanoma over other cancers.
机译:细胞周期蛋白依赖性激酶抑制剂p16〜(INK4A)中的突变或缺失与多种癌症类型有关,但更常见于黑色素瘤肿瘤中,并与家族性黑色素瘤易感性有关。尽管人们认为p16通过负调控细胞周期来起到抑癌作用,但目前尚不清楚为什么p16的遗传危害比其他癌症更易导致黑色素瘤。在这里,我们描述了p16在调节包括黑素细胞在内的几种细胞类型的氧化应激中的新作用。 p16的表达在紫外线照射后迅速上调,并以p38应力激活的蛋白激酶依赖性方式响应H_(2)O_(2)诱导的氧化应激。使用小的干扰RNA敲低p16会增加细胞内活性氧(ROS)和氧化(8-氧代鸟嘌呤)DNA的损伤,这通过H_(2)O_(2)处理进一步增强。在缺乏p16的人角质形成细胞以及来自Cdkn2a缺陷小鼠的全皮肤和真皮成纤维细胞中也观察到ROS水平升高。 Cdkn2a缺陷的成纤维细胞中的异常ROS和p38信号转导通过外源p16的表达来标准化。视网膜母细胞瘤蛋白(Rb)的抑制不能概括p16耗竭对ROS的影响,因此不需要Rb。最后,p16介导的ROS抑制不能归因于p16对细胞周期期的潜在影响。这些发现表明,p16作为致癌性细胞内氧化应激的内源性调节剂,具有潜在的替代Rb依赖于Rb的肿瘤抑制功能。与角质形成细胞和成纤维细胞相比,我们还发现在p16耗竭的情况下黑素细胞对氧化应激的敏感性增加,这可以解释为什么p16的损害比其他癌症更易导致黑色素瘤。

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