首页> 外文期刊>Cell death and differentiation >CALCOCO2/NDP52 and SQSTM1/p62 differentially regulate coxsackievirus B3 propagation
【24h】

CALCOCO2/NDP52 and SQSTM1/p62 differentially regulate coxsackievirus B3 propagation

机译:Calcoco2 / NDP52和SQSTM1 / P62差异调节CoxSackeigirus B3传播

获取原文
获取原文并翻译 | 示例
           

摘要

Cell autonomous immunity is the ability of individual cells to initiate a first line of host defense against invading microbes, such as viruses. Autophagy receptors, a diverse family of multivalent proteins, play a key role in this host response by detecting, sequestering, and eliminating virus in a process termed virophagy. To counteract this, positive-stranded RNA viruses, such as enteroviruses, have evolved strategies to circumvent the host autophagic machinery in an effort to promote viral propagation; however, the underlying mechanisms remain largely unclear. Here we studied the interaction between coxsackievirus B3 (CVB3) and the autophagy receptor SQSTM1 (sequestosome 1)/p62 and CALCOCO2/NDP52 (calcium binding and coiled-coil domain-containing protein 2/nuclear dot 10 protein 52). We demonstrated that SQSTM1 and CALCOCO2 differentially regulate CVB3 infection. We showed that knockdown of SQSTM1 causes increased viral protein production and elevated viral titers, whereas depletion of CALCOCO2 results in a significant inhibition of viral growth. Both receptors appear to have a role in virophagy through direct interaction with the viral capsid protein VP1 that undergoes ubiquitination during infection. Further investigation of the proviral mechanism of CALCOCO2 revealed that CALCOCO2, but not SQSTM1, suppresses the antiviral type I interferon signaling by promoting autophagy-mediated degradation of the mitochondrial antiviral signaling (MAVS) protein. Moreover, we demonstrated that viral proteinase 2A-mediated cleavage of SQSTM1 at glycine 241 impairs its capacity to associate with viral capsid, whereas cleavage of CALCOCO2 by viral proteinase 3C at glutamine 139, generates a stable C-terminal fragment that retains the proviral function of full-length CALCOCO2. Altogether, our study reveals a mechanism by which CVB3 targets selective autophagy receptors to evade host virophagy.
机译:细胞自主免疫是个体细胞引发第一线宿主防御的能力,例如病毒。自噬受体是一种多样化的多价蛋白质,通过检测,螯合在过程中的过程中的病毒来发挥这种宿主响应的关键作用。为了抵消这一点,诸如肠病病毒的正链RNA病毒,具有促进宿主自噬机械的促进促进病毒繁殖的策略;然而,潜在机制仍然很大程度上不清楚。在这里,我们研究了Coxsackeivirus B3(CVB3)和自噬受体Sqstm1(封粒剂组1)/ p62和Calcoco2 / Ndp52之间的相互作用(螯合体1)/ Ndp52(钙结合和卷绕式卷曲域蛋白2 /核点10蛋白52)。我们证明SQSTM1和Calcoco2差异调节CVB3感染。我们表明,SQSTM1的敲低导致病毒蛋白质产生和病毒滴度升高,而核心2的耗尽导致病毒生长的显着抑制。通过与在感染期间经历泛素化的病毒衣壳蛋白VP1的直接相互作用,这两个受体似乎在疾病中具有作用。进一步调查Calcoco2的透过机制显示,通过促进线粒体抗病毒信号传导(MAVS)蛋白的自噬介导的降解抑制抗病毒型I干扰素信号,抑制抗病毒型I干扰素信号传导。此外,我们证明,甘氨酸241下Sqstm1的病毒蛋白酶2a介导的Sqstm1的切割损害其与病毒衣壳相关的能力,而通过病毒蛋白酶3c在谷氨酰胺139上切割Calcoco2,产生稳定的C末端片段,其保留透过的透顶函数全长计算。完全,我们的研究揭示了一种机制,CVB3靶向选择性自噬受体以逃避宿主的疾病。

著录项

  • 来源
    《Cell death and differentiation》 |2019年第6期|共15页
  • 作者单位

    Univ British Columbia Ctr Heart Lung Innovat St Pauls Hosp Vancouver BC V6Z 1Y6 Canada;

    Univ British Columbia Ctr Heart Lung Innovat St Pauls Hosp Vancouver BC V6Z 1Y6 Canada;

    Univ British Columbia Ctr Heart Lung Innovat St Pauls Hosp Vancouver BC V6Z 1Y6 Canada;

    Univ British Columbia Ctr Heart Lung Innovat St Pauls Hosp Vancouver BC V6Z 1Y6 Canada;

    Univ British Columbia Ctr Heart Lung Innovat St Pauls Hosp Vancouver BC V6Z 1Y6 Canada;

    Univ British Columbia Ctr Heart Lung Innovat St Pauls Hosp Vancouver BC V6Z 1Y6 Canada;

  • 收录信息
  • 原文格式 PDF
  • 正文语种 eng
  • 中图分类 细胞生物学;
  • 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