Abstract

Porcine reproductive and respiratory syndrome virus (PRRSV) remains one of the most economically significant pathogens that seriously affect the global swine industry. Despite sustained efforts, the factors that affect PRRSV replication in host cells are far from being fully elucidated and thus warrants further investigation. In this study, we first demonstrated that PRRSV infection can cause downregulation of endogenous p21 protein in MARC-145 cells in a virus dose-dependent manner. Next, we analyzed the effect of p21 knockdown by RNA interference on cell cycle progression using flow cytometric analysis, and found that knockdown of p21 promotes MARC-145 cells entry into S phase of the cell cycle. Interestingly, we further discovered PRRSV infection is also able to promote MARC-145 cells entry into the S phase. Subsequently, we synchronized MARC-145 cells into G0/G1, S and G2/M phases, respectively, and then determined PRRSV replication in these cells. Results here show that the MARC-145 cells synchronized into the S phase exhibited the highest viral titer among the cells synchronized to different phases. Additionally, to reliably analyze the potential role of endogenous p21 protein in PRRSV replication, we constructed a p21 gene-knockout MARC-145 cell line (p21−/−) using CRISPR/Cas9 technology and evaluated its capability to support PRRSV replication. Our results indicate that knockout of p21 is conducive to PRRSV replication in MARC-145 cells. Furthermore, through construction of a series of eukaryotic plasmids expressing each of individual PRRSV proteins combined with cell transfection, we demonstrated that the nonstructural protein 11 (nsp11) of PRRSV mediates p21 degradation, which was further confirmed by generating a stable MARC-145 cell line constitutively expressing nsp11 using a lentivirus system. Notably, we further demonstrated that the endoribonuclease activity rather than the deubiquitinating activity of nsp11 is essential for p21 degradation via mutagenic analysis. Finally, we demonstrated that nsp11 mediates p21 degradation via a ubiquitin-independent proteasomal degradation manner. Altogether, our study not only uncovers a new pathogenesis of PRRSV, but also provides new insights into development of novel antiviral strategies.

Highlights

  • Porcine reproductive and respiratory syndrome (PRRS) has been one of the most economically important diseases affecting the global swine industry for three decades [1, 2]

  • Of the three multiplicity of infection (MOI) tested, MOI of 1 exhibited the highest inhibition effect on p21 expression (Figures 1A–C). This indicates that the effect of Porcine reproductive and respiratory syndrome virus (PRRSV) infection on p21 protein expression was in a dose-dependent manner

  • HB-1/3.9 was able to cause downregulation of p21 in MARC-145 cells, but to a lesser extent than did the HP-PRRSV strain JXwn06. These results indicate that PRRSV infection caused downregulation of p21 protein in MARC-145 cells in a dose-dependent manner

Read more

Summary

Introduction

Porcine reproductive and respiratory syndrome (PRRS) has been one of the most economically important diseases affecting the global swine industry for three decades [1, 2]. The causative agent of PRRS, porcine reproductive and respiratory syndrome virus (PRRSV), is classified in the family Arteriviridae in the order Nidovirales [3]. The two polyproteins are processed by viral encoded proteases to produce at least 16 distinct non-structural proteins (nsps), including nsp1α, nsp1β, nsp, nsp2N, nsp2TF, nsp, nsp, nsp, nsp, nsp7α, nsp7β, nsp, nsp, nsp, nsp, and nsp12 [4,5,6,7,8,9,10,11,12,13]. The remaining ORFs encode eight viral structural proteins, including five minor envelope proteins (GP2a, GP3, GP4, E, and ORF5a), two major envelope proteins (GP5 and M), and the nucleocapsid protein (N) [1, 16]

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call