...
首页> 外文期刊>STEM CELLS >Ionizing Radiation Induces Ataxia Telangiectasia Mutated-Dependent Checkpoint Signaling and G2 But Not G1 Cell Cycle Arrest in Pluripotent Human Embryonic Stem Cells
【24h】

Ionizing Radiation Induces Ataxia Telangiectasia Mutated-Dependent Checkpoint Signaling and G2 But Not G1 Cell Cycle Arrest in Pluripotent Human Embryonic Stem Cells

机译:电离辐射诱导多能人类胚胎干细胞中共济失调毛细血管扩张突变依赖性检查点信号和G 2 而不是G 1 细胞周期阻滞

获取原文
获取原文并翻译 | 示例

摘要

Human embryonic stem (ES) cells are highly sensitive to environmental insults including DNA damaging agents, responding with high levels of apoptosis. To understand the response of human ES cells to DNA damage, we investigated the function of the ataxia telangiectasia mutated (ATM) DNA damage signaling pathway in response to -irradiation. Here, we demonstrate for the first time in human ES cells that ATM kinase is phosphorylated and properly localized to the sites of DNA double-strand breaks within 15 minutes of irradiation. Activation of ATM kinase resulted in phosphorylation of its downstream targets: Chk2, p53, and Nbs1. In contrast to murine ES cells, Chk2 and p53 were localized to the nucleus of irradiated human ES cells. We further show that irradiation resulted in a temporary arrest of the cell cycle at the G2, but not G1, phase. Human ES cells resumed cycling approximately 16 hours after irradiation, but had a fourfold higher incidence of aberrant mitotic figures compared to nonirradiated cells. Finally, we demonstrate an essential role of ATM in establishing G2 arrest since inhibition with the ATM-specific inhibitor KU55933 resulted in abolishment of G2 arrest, evidenced by an increase in the number of cycling cells 2 hours after irradiation. In summary, these results indicate that human ES cells activate the DNA damage checkpoint, resulting in an ATM-dependent G2 arrest. However, these cells re-enter the cell cycle with prominent mitotic spindle defects. STEM CELLS 2009;27:1822-1835
机译:人类胚胎干(ES)细胞对环境损伤(包括DNA破坏剂)高度敏感,并以高水平的细胞凋亡做出响应。为了了解人类ES细胞对DNA损伤的反应,我们调查了共济失调性毛细血管扩张突变(ATM)DNA损伤信号通路对辐射的反应。在这里,我们首次在人类ES细胞中证明ATM激酶被磷酸化并正确定位在辐射15分钟内的DNA双链断裂位点。 ATM激酶的激活导致其下游靶标Chk2,p53和Nbs1的磷酸化。与鼠胚胎干细胞相反,Chk2和p53定位于受辐照的人类胚胎干细胞的细胞核。我们进一步表明,辐射导致细胞周期暂时停滞在G2期,而不是G1期。人ES细胞在辐照后约16小时恢复循环,但与未辐照的细胞相比,有丝分裂异常象的发生率高四倍。最后,我们证明了ATM在建立G2阻滞中的重要作用,因为用ATM特异性抑制剂KU55933进行抑制会导致G2阻滞的消除,这可以通过照射2小时后循环细胞数量的增加来证明。总之,这些结果表明人类ES细胞激活了DNA损伤检查点,导致了ATM依赖性G2阻滞。但是,这些细胞重新进入具有明显有丝分裂纺锤体缺陷的细胞周期。干细胞2009; 27:1822-1835

著录项

  • 来源
    《STEM CELLS 》 |2009年第8期| 1822-1835| 共14页
  • 作者单位

    Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania|Pittsburgh Development Center, Pittsburgh, Pennsylvania;

    Medical Scientist Training Program and Molecular Pharmacology Graduate Training Program, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania;

    Pittsburgh Development Center, Pittsburgh, Pennsylvania|Centro de Neurociências e Biologia Celular, Departamento de Zoologia, Universidade de Coimbra, Portugal;

    Department of Radiation Oncology, University of Pittsburgh School of Medicine, Hillman Cancer Center, Pittsburgh, Pennsylvania;

    Pittsburgh Development Center, Pittsburgh, Pennsylvania;

    Pittsburgh Development Center, Pittsburgh, Pennsylvania|Department of Biology, University of Texas at San Antonio, San Antonio, Texas;

  • 收录信息
  • 原文格式 PDF
  • 正文语种 eng
  • 中图分类
  • 关键词

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