首页> 美国卫生研究院文献>Cell Death and Differentiation >CALCOCO2/NDP52 and SQSTM1/p62 differentially regulate coxsackievirus B3 propagation
【2h】

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

机译:CALCOCO2 / NDP52和SQSTM1 / p62差异调节柯萨奇病毒B3繁殖

代理获取
本网站仅为用户提供外文OA文献查询和代理获取服务,本网站没有原文。下单后我们将采用程序或人工为您竭诚获取高质量的原文,但由于OA文献来源多样且变更频繁,仍可能出现获取不到、文献不完整或与标题不符等情况,如果获取不到我们将提供退款服务。请知悉。

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 2uclear 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:细胞自主免疫是单个细胞启动针对入侵的微生物(如病毒)的第一道宿主防御能力。自噬受体是多种多样的多价蛋白家族,通过检测,隔离和消除病毒,在自噬过程中在这种宿主反应中起关键作用。为了解决这个问题,正链RNA病毒(例如肠病毒)已经进化出策略来规避宿主自噬机制,以促进病毒繁殖。但是,基本机制仍不清楚。在这里,我们研究了柯萨奇病毒B3(CVB3)和自噬受体SQSTM1(sequestosome 1)/ p62和CALCOCO2 / NDP52(钙结合和含卷曲螺旋结构域蛋白2 /核点10蛋白52)之间的相互作用。我们证明了SQSTM1和CALCOCO2差异调节CVB3感染。我们表明,敲低SQSTM1会导致病毒蛋白产量增加和病毒滴度升高,而CALCOCO2的耗尽会导致对病毒生长的显着抑制。两种受体似乎都通过与感染过程中发生泛素化的病毒衣壳蛋白VP1直接相互作用而在吞噬中起作用。 CALCOCO2的前病毒机制的进一步研究表明,CALCOCO2(而非SQSTM1)通过促进线粒体抗病毒信号(MAVS)蛋白的自噬介导的降解来抑制I型干扰素信号传导。此外,我们证明了病毒蛋白酶2A介导的SQSTM1在甘氨酸241上的切割削弱了其与病毒衣壳结合的能力,而谷氨酰胺139在病毒蛋白酶3C的作用下对CALCOCO2的切割产生了一个稳定的C末端片段,该片段保留了CV的前病毒功能全长CALCOCO2。总而言之,我们的研究揭示了CVB3靶向选择性自噬受体逃避宿主吞噬的机制。

著录项

相似文献

  • 外文文献
  • 中文文献
  • 专利
代理获取

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