A clinically attenuated double-mutant of porcine reproductive and respiratory syndrome virus-2 that does not prompt overexpression of proinflammatory cytokines during co-infection with a secondary pathogen

PLoS Pathog. 2024 Mar 28;20(3):e1012128. doi: 10.1371/journal.ppat.1012128. eCollection 2024 Mar.

Abstract

Porcine reproductive and respiratory syndrome virus (PRRSV) is known to suppress the type I interferon (IFNs-α/β) response during infection. PRRSV also activates the NF-κB signaling pathway, leading to the production of proinflammatory cytokines during infection. In swine farms, co-infections of PRRSV and other secondary bacterial pathogens are common and exacerbate the production of proinflammatory cytokines, contributing to the porcine respiratory disease complex (PRDC) which is clinically a severe disease. Previous studies identified the non-structural protein 1β (nsp1β) of PRRSV-2 as an IFN antagonist and the nucleocapsid (N) protein as the NF-κB activator. Further studies showed the leucine at position 126 (L126) of nsp1β as the essential residue for IFN suppression and the region spanning the nuclear localization signal (NLS) of N as the NF-κB activation domain. In the present study, we generated a double-mutant PRRSV-2 that contained the L126A mutation in the nsp1β gene and the NLS mutation (ΔNLS) in the N gene using reverse genetics. The immunological phenotype of this mutant PRRSV-2 was examined in porcine alveolar macrophages (PAMs) in vitro and in young pigs in vivo. In PAMs, the double-mutant virus did not suppress IFN-β expression but decreased the NF-κB-dependent inflammatory cytokine productions compared to those for wild-type PRRSV-2. Co-infection of PAMs with the mutant PRRSV-2 and Streptococcus suis (S. suis) also reduced the production of NF-κB-directed inflammatory cytokines. To further examine the cytokine profiles and the disease severity by the mutant virus in natural host animals, 6 groups of pigs, 7 animals per group, were used for co-infection with the mutant PRRSV-2 and S. suis. The double-mutant PRRSV-2 was clinically attenuated, and the expressions of proinflammatory cytokines and chemokines were significantly reduced in pigs after bacterial co-infection. Compared to the wild-type PRRSV-2 and S. suis co-infection control, pigs coinfected with the double-mutant PRRSV-2 exhibited milder clinical signs, lower titers and shorter duration of viremia, and lower expression of proinflammatory cytokines. In conclusion, our study demonstrates that genetic modification of the type I IFN suppression and NF-κB activation functions of PRRSV-2 may allow us to design a novel vaccine candidate to alleviate the clinical severity of PRRS-2 and PRDC during bacterial co-infection.

MeSH terms

  • Animals
  • Coinfection*
  • Cytokines / genetics
  • Cytokines / metabolism
  • Interferon Type I* / metabolism
  • Macrophages, Alveolar / metabolism
  • NF-kappa B / genetics
  • NF-kappa B / metabolism
  • Porcine Reproductive and Respiratory Syndrome* / genetics
  • Porcine Reproductive and Respiratory Syndrome* / metabolism
  • Porcine respiratory and reproductive syndrome virus* / metabolism
  • Swine

Substances

  • Cytokines
  • NF-kappa B
  • Interferon Type I

Grants and funding

This project was supported by Agriculture and Food Research Initiative (AFRI) Competitive Grants nos. 2018-67015-28287 and 2023-67015-39710 from the U.S. Department of Agriculture (USDA) National Institute of Food and Agriculture (NIFA) to DY and no. 2018-67016-28296 to YML. Part of this material is based upon work supported by the Cooperative State Research Service, USDA, under Project Number ILLU-888-944 awarded to DY. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. None of the authors received a salary from the funders.