1. Academic Validation
  2. The PERK/PKR-eIF2α Pathway Negatively Regulates Porcine Hemagglutinating Encephalomyelitis Virus Replication by Attenuating Global Protein Translation and Facilitating Stress Granule Formation

The PERK/PKR-eIF2α Pathway Negatively Regulates Porcine Hemagglutinating Encephalomyelitis Virus Replication by Attenuating Global Protein Translation and Facilitating Stress Granule Formation

  • J Virol. 2022 Jan 12;96(1):e0169521. doi: 10.1128/JVI.01695-21.
Junchao Shi  # 1 Zi Li  # 1 Rongyi Xu 1 Jing Zhang 1 Qianyu Zhou 1 Rui Gao 1 Huijun Lu 2 Yungang Lan 1 Kui Zhao 1 Hongbin He 3 Feng Gao 1 Wenqi He 1
Affiliations

Affiliations

  • 1 Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin Universitygrid.64924.3d, Changchun, China.
  • 2 Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, Jilin Universitygrid.64924.3d, Changchun, China.
  • 3 Key Laboratory of Animal Resistant Biology of Shandong, Ruminant Disease Research Center, College of Life Sciences, Shandong Normal Universitygrid.410585.d, Jinan, China.
  • # Contributed equally.
Abstract

The replication of coronaviruses, including severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome coronavirus (MERS-CoV), and the recently emerged severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is closely associated with the endoplasmic reticulum (ER) of infected cells. The unfolded protein response (UPR), which is mediated by ER stress (ERS), is a typical outcome in coronavirus-infected cells and is closely associated with the characteristics of coronaviruses. However, the interaction between virus-induced ERS and coronavirus replication is poorly understood. Here, we demonstrate that Infection with the betacoronavirus porcine hemagglutinating encephalomyelitis virus (PHEV) induced ERS and triggered all three branches of the UPR signaling pathway both in vitro and in vivo. In addition, ERS suppressed PHEV replication in mouse neuro-2a (N2a) cells primarily by activating the protein kinase R-like ER kinase (PERK)-eukaryotic initiation factor 2α (eIF2α) axis of the UPR. Moreover, another eIF2α phosphorylation kinase, interferon (IFN)-induced double-stranded RNA-dependent protein kinase (PKR), was also activated and acted cooperatively with PERK to decrease PHEV replication. Furthermore, we demonstrate that the PERK/PKR-eIF2α pathways negatively regulated PHEV replication by attenuating global protein translation. Phosphorylated eIF2α also promoted the formation of stress granules (SGs), which in turn repressed PHEV replication. In summary, our study presents a vital aspect of the host innate response to invading pathogens and reveals attractive host targets (e.g., PERK, PKR, and eIF2α) for Antiviral drugs. IMPORTANCE Coronavirus diseases are caused by different coronaviruses of importance in humans and Animals, and specific treatments are extremely limited. ERS, which can activate the UPR to modulate viral replication and the host innate response, is a frequent occurrence in coronavirus-infected cells. PHEV, a neurotropic betacoronavirus, causes nerve cell damage, which accounts for the high mortality rates in suckling piglets. However, it remains incompletely understood whether the highly developed ER in nerve cells plays an Antiviral role in ERS and how ERS regulates viral proliferation. In this study, we found that PHEV Infection induced ERS and activated the UPR both in vitro and in vivo and that the activated PERK/PKR-eIF2α axis inhibited PHEV replication through attenuating global protein translation and promoting SG formation. A better understanding of coronavirus-induced ERS and UPR activation may reveal the pathogenic mechanism of coronavirus and facilitate the development of new treatment strategies for these diseases.

Keywords

coronavirus; eIF2α; endoplasmic reticulum stress; porcine hemagglutinating encephalomyelitis virus; stress granule; translation attenuation.

Figures
Products