...
首页> 外文期刊>Molecular cancer research: MCR >Loss of Keratinocytic RXR alpha Combined with Activated CDK4 or Oncogenic NRAS Generates UVB-Induced Melanomas via Loss of p53 and PTEN in the Tumor Microenvironment
【24h】

Loss of Keratinocytic RXR alpha Combined with Activated CDK4 or Oncogenic NRAS Generates UVB-Induced Melanomas via Loss of p53 and PTEN in the Tumor Microenvironment

机译:角化细胞RXRα的丧失与活化的CDK4或致癌的NRAS结合,通过在肿瘤微环境中丧失p53和PTEN产生了UVB诱导的黑素瘤。

获取原文
获取原文并翻译 | 示例
   

获取外文期刊封面封底 >>

       

摘要

Understanding the molecular mechanisms behind formation of melanoma, the deadliest form of skin cancer, is crucial for improved diagnosis and treatment. One key is to better understand the cross-talk between epidermal keratinocytes and pigment-producing melanocytes. Here, using a bigenic mouse model system combining mutant oncogenic NRAS(Q61K) (constitutively active RAS) or mutant activated CDK4(R24C/R24C) (prevents binding of CDK4 by kinase inhibitor p16(INK4A)) with an epidermis-specific knockout of the nuclear retinoid X receptor alpha (RXR alpha(ep-/-)) results in increased melanoma formation after chronic ultraviolet-B (UVB) irradiation compared with control mice with functional RXR alpha. Melanomas from both groups of bigenic RXR alpha(ep-/-) mice are larger in size with higher proliferative capacity, and exhibit enhanced angiogenic properties and increased expression of malignant melanoma markers. Analysis of tumor adjacent normal skin from these mice revealed altered expression of several biomarkers indicative of enhanced melanoma susceptibility, including reduced expression of tumor suppressor p53 and loss of PTEN, with concomitant increase in activated AKT. Loss of epidermal RXR alpha in combination with UVB significantly enhances invasion of melanocytic cells to draining lymph nodes in bigenic mice expressing oncogenic NRAS(Q61K) compared with controls with functional RXR alpha. These results suggest a crucial role of keratinocytic RXR alpha to suppress formation of UVB-induced melanomas and their progression to malignant cancers in the context of driver mutations such as activated CDK4(R24C/R24C) or oncogenic NRAS(Q61K). (C) 2014 AACR.
机译:了解黑色素瘤(皮肤癌的最致命形式)背后的分子机制,对于改善诊断和治疗至关重要。一个关键是更好地了解表皮角质形成细胞与产生色素的黑素细胞之间的串扰。在这里,使用双基因小鼠模型系统结合突变致癌的NRAS(Q61K)(组成性活性RAS)或突变激活的CDK4(R24C / R24C)(防止CDK4被激酶抑制剂p16(INK4A)结合)与表皮特异性敲除与具有功能性RXR alpha的对照小鼠相比,核类视黄醇X受体alpha(RXR alpha(ep-/-))导致慢性紫外线B(UVB)照射后黑色素瘤形成增加。来自两组双基因RXR alpha(ep-/-)小鼠的黑素瘤体积较大,具有较高的增殖能力,并表现出增强的血管生成特性和恶性黑色素瘤标志物的表达增加。对这些小鼠的肿瘤邻近正常皮肤的分析显示,几种生物标志物的表达发生了变化,表明黑素瘤易感性增强,包括肿瘤抑制因子p53的表达降低和PTEN的丧失,并伴随着活化AKT的增加。与具有功能性RXRα的对照相比,表皮RXRα与UVB的结合显着增强了表达致癌性NRAS(Q61K)的双基因小鼠中黑素细胞向排水淋巴结的侵袭。这些结果表明角质细胞RXRα在抑制诸如激活的CDK4(R24C / R24C)或致癌的NRAS(Q61K)等驱动基因突变的情况下,抑制UVB诱导的黑色素瘤的形成及其向恶性肿瘤的发展至关重要。 (C)2014 AACR。

著录项

相似文献

  • 外文文献
  • 中文文献
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