...
首页> 外文期刊>Science Advances >Transcriptional regulation of APOBEC3 antiviral immunity through the CBF-β/RUNX axis
【24h】

Transcriptional regulation of APOBEC3 antiviral immunity through the CBF-β/RUNX axis

机译:通过CBF-β/ RUNX轴对APOBEC3抗病毒免疫的转录调控

获取原文

摘要

A diverse set of innate immune mechanisms protects cells from viral infections. The APOBEC3 family of DNA cytosine deaminases is an integral part of these defenses. For instance, APOBEC3D, APOBEC3F, APOBEC3G, and APOBEC3H would have the potential to destroy HIV-1 complementary DNA replication intermediates if not for neutralization by a proteasomal degradation mechanism directed by the viral protein Vif. At the core of this complex, Vif heterodimerizes with the transcription cofactor CBF-β, which results in fewer transcription complexes between CBF-β and its normal RUNX partners. Recent studies have shown that the Vif/CBF-β interaction is specific to the primate lentiviruses HIV-1 and SIV (simian immunodeficiency virus), although related nonprimate lentiviruses still require a Vif-dependent mechanism for protection from host species’ APOBEC3 enzymes. We provide a molecular explanation for this evolutionary conundrum by showing that CBF-β is required for expression of the aforementioned HIV-1–restrictive APOBEC3 gene repertoire. Knockdown and knockout studies demonstrate that CBF-β is required for APOBEC3 mRNA expression in the nonpermissive T cell line H9 and in primary CD4+ T lymphocytes. Complementation experiments using CBF-β separation-of-function alleles show that the interaction with RUNX transcription factors is required for APOBEC3 transcriptional regulation. Accordingly, the infectivity of Vif-deficient HIV-1 increases in cells lacking CBF-β, demonstrating the importance of CBF-β/RUNX–mediated transcription in establishing the APOBEC3 antiviral state. These findings demonstrate a major layer of APOBEC3 gene regulation in lymphocytes and suggest that primate lentiviruses evolved to hijack CBF-β in order to simultaneously suppress this potent antiviral defense system at both transcriptional and posttranslational levels.
机译:各种各样的先天免疫机制可以保护细胞免受病毒感染。 DNA胞嘧啶脱氨酶的APOBEC3家族是这些防御机制的组成部分。例如,APOBEC3D,APOBEC3F,APOBEC3G和APOBEC3H如果不被病毒蛋白Vif指导的蛋白酶体降解机制中和,可能会破坏HIV-1互补DNA复制中间体。 Vif是复合物的核心,它与转录辅因子CBF-β异源二聚体,从而导致CBF-β及其正常RUNX伴侣之间的转录复合物更少。最近的研究表明,Vif /CBF-β相互作用特定于灵长类慢病毒HIV-1和SIV(猿猴免疫缺陷病毒),尽管相关的非灵长类慢病毒仍需要Vif依赖性机制来保护宿主物种的APOBEC3酶。我们通过显示上述HIV-1限制性APOBEC3基因库表达所需的CBF-β,为这一进化难题提供了分子解释。敲除和敲除研究表明,APOBEC3 mRNA在非允许性T细胞系H9和原代CD4 + T淋巴细胞中的表达需要CBF-β。使用CBF-β功能分离等位基因的互补实验表明,APOBEC3转录调控需要与RUNX转录因子相互作用。因此,缺乏CBF-β的细胞缺乏Vif的HIV-1的感染力增加,表明CBF-β/ RUNX介导的转录在建立APOBEC3抗病毒状态中的重要性。这些发现证明了淋巴细胞中APOBEC3基因调控的主要层,并表明灵长类慢病毒进化为劫持CBF-β,以便同时在转录和翻译后水平上抑制这种有效的抗病毒防御系统。

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