中国农业科学 ›› 2023, Vol. 56 ›› Issue (7): 1401-1416.doi: 10.3864/j.issn.0578-1752.2023.07.016

• 畜牧·兽医 • 上一篇    下一篇

非洲猪瘟病毒MGF110-5L-6L蛋白诱导宿主细胞翻译阻滞和应激颗粒形成的作用机制

樊帅(), 钟函, 杨中元, 何文瑞, 万博, 魏战勇, 韩世充(), 张改平()   

  1. 河南农业大学动物医学院/动物免疫学国家国际联合研究中心,郑州 450046
  • 收稿日期:2022-02-28 接受日期:2022-06-30 出版日期:2023-04-01 发布日期:2023-04-03
  • 联系方式: 樊帅,Tel:15138698236;E-mail:fshuai9859@163.com。
  • 基金资助:
    国家自然科学基金(31941001); 国家自然科学基金(32102655); 河南省重点研发与推广专项(212102110369)

African Swine Fever Virus MGF110-5L-6L Induces Host Cell Translation Arrest and Stress Granule Formation by Activating the PERK/PKR-eIF2α Pathway

FAN Shuai(), ZHONG Han, YANG ZhongYuan, HE WenRui, WAN Bo, WEI ZhanYong, HAN ShiChong(), ZHANG GaiPing()   

  1. College of Veterinary Medicine, Henan Agricultural University/International Joint Research Center of National Animal Immunology, Zhengzhou 450046
  • Received:2022-02-28 Accepted:2022-06-30 Published:2023-04-01 Online:2023-04-03

摘要:

【背景】 非洲猪瘟(African swine fever,ASF)是由非洲猪瘟病毒(African swine fever virus,ASFV)感染引发的一种猪烈性传染病,是全球公认的养猪业“头号杀手”,至今尚无安全有效的疫苗和药物。病毒作为专性细胞内寄生物,必须通过“劫持”宿主翻译系统为病毒蛋白合成服务。其中翻译起始因子eIF2α作为翻译调控的核心节点,控制细胞应激反应和翻译重编程走向,对病毒毒力、嗜性、致病性及免疫逃逸等具有重要影响,eIF2α磷酸化调控无疑是病毒与宿主细胞竞争翻译资源的重要阵地之一。然而,关于ASFV编码蛋白与eIF2α磷酸化作用关系的认知极度匮乏。【目的】 探究非洲猪瘟病毒MGF110-5L-6L蛋白对宿主细胞翻译阻滞和促进应激颗粒形成的作用机制,为深入揭示非洲猪瘟病毒的致病机制研究提供科学依据。【方法】 在前期利用荧光素酶报告基因载体和绿色荧光报告载体,筛选发现外源表达MGF110-5L-6L极显著上调eIF2α磷酸化水平的基础上。选择猪肺泡巨噬细胞3D4/21和猪肾细胞PK-15作为研究用细胞系,利用质粒转染和特异性化学药物处理等方法,结合免疫印迹和激光共聚焦显微镜等检测技术,验证了MGF110-5L-6L蛋白与eIF2α磷酸化及其下游翻译效应之间的功能关系。随后,通过生物信息学网站预测、亚细胞定位和功能分析等,研究了MGF110-5L-6L蛋白与诱导细胞应激之间的相关性。【结果】 证实外源表达ASFV多基因家族蛋白MGF110-5L-6L能够显著增强细胞内eIF2α磷酸化水平及激活转录因子ATF4表达量,诱导综合应激反应的发生;还可诱导细胞发生内质网应激和未折叠蛋白反应,并通过活化PERK和PKR激酶来诱发eIF2α的磷酸化,进而导致细胞蛋白翻译阻滞和应激颗粒形成。进一步证实,MGF110-5L-6L蛋白含有两个高度保守的半胱氨酸基序,且主要定位于内质网,少量分布于高尔基体、线粒体和溶酶体,还可诱导高尔基体和过氧化物酶体的亚细胞定位及形态出现显著改变,提示其可能通过影响内质网腔中氧化还原稳态、蛋白分泌途径或相关膜性细胞器发生等创造应激条件。【结论】 报道了ASFV早期蛋白MGF110-5L-6L的结构、亚细胞定位及其潜在功能特征,揭示了MGF110-5L-6L蛋白与eIF2α磷酸化和宿主细胞蛋白翻译系统之间的功能关系,为深入认识ASFV的病原生物学与致病机制提供了新的科学参考。

关键词: 非洲猪瘟病毒, MGF110-5L-6L蛋白, eIF2α磷酸化, 翻译阻滞, 应激颗粒

Abstract:

【Background】 African swine fever (ASF) is an acute, highly contagious, and deadly infectious disease of pigs caused by ASF virus (ASFV), which is currently considered the biggest killer in global swine industry. To date, there is no effective vaccines or antiviral drugs for the prevention or treatment of ASF. As obligate intracellular parasites, the viruses are fully reliant on the host translation machinery to produce the polypeptides that are essential for viral replication. A central mechanism regulating translation initiation involves phosphorylation of the α subunit of eukaryotic initiation factor 2 (eIF2α), which directs host translational control and adaptation to cellular stress. The regulation of eIF2α phosphorylation has been regarded as a critical step for viral infection, with important effects on virulence, tissue tropism, pathogenicity, and immunoevasion. However, the molecular mechanisms by which most of the ASFV-encoded proteins affecting eIF2α phosphorylation have not been well studied. 【Objective】 The aim of this study was to explore the mechanism of ASFV MGF110-5L-6L protein on the host cell translation block and promote the formation of stress particles, so as to provide a scientific basis for further revealing the pathogenic mechanism of African swine fever virus.【Method】 The preliminary screening by luciferase reporter assays identified that ectopic expression of ASFV MGF110-5L-6L, a previously uncharacterized member of the multigene family 110, significantly increased eIF2α phosphorylation levels. To confirm and clarify the potential role of MGF110-5L-6L expression in mediating eIF2α phosphorylation and downstream of translation control, two continuous porcine cell lines, including 3D4/21 (porcine alveolar macrophage) and PK-15 (porcine kidney), were used for the plasmid transfection and/or drug treatment and subjected to immunoblotting or confocal immunofluorescence analysis. To investigate how the ectopic expression of MGF110-5L-6L triggers cellular stress, the structure, subcellular localization and function of the MGF110-5L-6L protein were further characterized by a combination of bioinformatic prediction, immunofluorescence and immunoblotting analysis.【Result】 Here, it was confirmed that ectopic expression of MGF110-5L-6L remarkably promoted eIF2α phosphorylation and the expression of ATF4, indicating that it functions in the integrated stress response. The subsequent analyses revealed that MGF110-5L-6L expression could trigger the ER stress and activate the unfolded protein response, and the phosphorylation of eIF2α was mediated via PERK and PKR, resulting in the suppression of host translation and stress granule formation. It was further observed that MGF110-5L-6L protein had two highly conserved central cysteine-rich domains and was mainly retained in the endoplasmic reticulum (ER), and also caused a significant reorganization of the subcellular distribution and morphological characteristics of the Golgi and peroxisome, suggesting that it might interfere with ER redox homeostasis, secretory pathway, and other membrane-bound organelles to trigger cellular stress.【Conclusion】 Together, these results demonstrated a previously uncharacterized role of ASFV MGF110-5L-6L and further defined several molecular interfaces by which ASFV MGF110-5L-6L hijacks the host cell translation, which expanded the view of ASFV in determining the fate of host-pathogen interactions.

Key words: African swine fever virus, MGF110-5L-6L protein, eIF2α phosphorylation, translation arrest, stress granules