首页> 外文学位 >Differential expression of Helios in regulatory T cells.
【24h】

Differential expression of Helios in regulatory T cells.

机译:Helios在调节性T细胞中的差异表达。

获取原文
获取原文并翻译 | 示例

摘要

CD4+CD25+ T cells (Tregs) represent the dominant arm of immune regulation during an immune response. Dedicated FoxP3 + Tregs are generated in the thymus (nTregs) as well as induced in the periphery (iTregs). However, our understanding of conditions that promote the induction or expansion of these cells remains incomplete. In addition, deciphering molecular mechanisms that promote the stable expression of FoxP3+ has been challenging and confusing at best. In this body of work, we have used an autochthonous cancer model to evaluate the transcriptional profile of tumor-specific CD4 T cells encountering cognate antigen in vivo. Not surprisingly, tumor recognition led to the development of regulatory T cells as measured by upregulation of FoxP3 and in vitro suppressive ability. Interestingly, we found that Helios, a T cell restricted Ikaros transcription factor family member, was also upregulated in these cells. However, unlike FoxP3 expression, Helios up-regulation was absent in in vitro induced regulatory T cells and its ectopic expression induced apoptosis. On a functional level, suppression of Helios in CD4+CD25+ T cells by siRNA oligonucletides markedly abrogated the suppressive function of Human Tregs. Additionally, Helios expression was required for the maintenance of FoxP3 mRNA levels. Finally, we demonstrated that Helios specifically binds to the FoxP3 promoter region in EL4 cells. Taken together, these data suggest that tumors not only induce regulatory T cells but also support the expansion of thymic Tregs. Moreover, in light of other published work, these data suggest that Helios expression plays an important yet redundant role in maintaining a stable expression of FoxP3 in nTregs.
机译:CD4 + CD25 + T细胞(Tregs)代表免疫应答过程中免疫调节的主要部分。专用的FoxP3 + Tregs在胸腺中产生(nTregs),并在外周产生(iTregs)。但是,我们对促进这些细胞诱导或扩增的条件的理解仍然不完全。另外,破译促进FoxP3 +稳定表达的分子机制充其量是充满挑战和困惑的。在这项工作中,我们使用了一种本地癌症模型来评估体内遇到同源抗原的肿瘤特异性CD4 T细胞的转录谱。毫不奇怪,通过FoxP3的上调和体外抑制能力的测量,肿瘤识别导致调节性T细胞的发育。有趣的是,我们发现Helios是一种受T细胞限制的Ikaros转录因子家族成员,在这些细胞中也被上调。但是,与FoxP3表达不同,体外诱导的调节性T细胞不存在Helios上调,而异位表达诱导凋亡。在功能水平上,siRNA寡核苷酸抑制CD4 + CD25 + T细胞中的Helios明显废除了Human Tregs的抑制功能。此外,维持FoxP3 mRNA水平需要Helios表达。最后,我们证明了Helios与EL4细胞中的FoxP3启动子区域特异性结合。综上所述,这些数据表明肿瘤不仅诱导调节性T细胞,而且还支持胸腺Tregs的扩增。此外,根据其他已发表的工作,这些数据表明Helios表达在维持FoxP3在nTregs中的稳定表达中起着重要而又多余的作用。

著录项

  • 作者

    Getnet, Derese.;

  • 作者单位

    The Johns Hopkins University.;

  • 授予单位 The Johns Hopkins University.;
  • 学科 Health Sciences Oncology.
  • 学位 Ph.D.
  • 年度 2010
  • 页码 109 p.
  • 总页数 109
  • 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