Picornavirus security proteins promote the release of extracellular vesicle enclosed viruses via the modulation of host kinases

PLoS Pathog. 2024 Apr 25;20(4):e1012133. doi: 10.1371/journal.ppat.1012133. eCollection 2024 Apr.

Abstract

The discovery that extracellular vesicles (EVs) serve as carriers of virus particles calls for a reevaluation of the release strategies of non-enveloped viruses. Little is currently known about the molecular mechanisms that determine the release and composition of EVs produced by virus-infected cells, as well as conservation of these mechanisms among viruses. We previously described an important role for the Leader protein of the picornavirus encephalomyocarditis virus (EMCV) in the induction of virus-carrying EV subsets with distinct molecular and physical properties. EMCV L acts as a 'viral security protein' by suppressing host antiviral stress and type-I interferon (IFN) responses. Here, we tested the ability of functionally related picornavirus proteins of Theilers murine encephalitis virus (TMEV L), Saffold virus (SAFV L), and coxsackievirus B3 (CVB3 2Apro), to rescue EV and EV-enclosed virus release when introduced in Leader-deficient EMCV. We show that all viral security proteins tested were able to promote virus packaging in EVs, but that only the expression of EMCV L and CVB3 2Apro increased overall EV production. We provide evidence that one of the main antiviral pathways counteracted by this class of picornaviral proteins, i.e. the inhibition of PKR-mediated stress responses, affected EV and EV-enclosed virus release during infection. Moreover, we show that the enhanced capacity of the viral proteins EMCV L and CVB3 2Apro to promote EV-enclosed virus release is linked to their ability to simultaneously promote the activation of the stress kinase P38 MAPK. Taken together, we demonstrate that cellular stress pathways involving the kinases PKR and P38 are modulated by the activity of non-structural viral proteins to increase the release EV-enclosed viruses during picornavirus infections. These data shed new light on the molecular regulation of EV production in response to virus infection.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Cardiovirus Infections / metabolism
  • Cardiovirus Infections / virology
  • Encephalomyocarditis virus / metabolism
  • Encephalomyocarditis virus / physiology
  • Extracellular Vesicles* / metabolism
  • Extracellular Vesicles* / virology
  • Humans
  • Mice
  • Picornaviridae* / metabolism
  • Picornaviridae* / physiology
  • Theilovirus / metabolism
  • Viral Proteins* / genetics
  • Viral Proteins* / metabolism
  • Virus Release / physiology
  • eIF-2 Kinase / metabolism

Substances

  • Viral Proteins
  • eIF-2 Kinase

Grants and funding

This work was supported by The Netherlands Organisation for Scientific Research (NWO-ALW grant number ALWOP.351 to ENMN-‘tH) (https://www.nwo.nl/en) and the China Scholarship Council (CSC, to XP). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.