Abstract

Peste des petits ruminants virus (PPRV), belonging to the genus Morbillivirus in the family Paramyxoviridae, causes severe infectious disease in small ruminants and has been rapidly spreading in many parts of Africa, the Middle East, and Asia. Although vaccination is considered to be an effective means of controlling PPR, the heat-sensitive nature of the vaccines against PPRV greatly limits their application in areas with a hot climate. In the present study, we investigated the anti-PPRV effects of favipiravir and sought to identify the underlying mechanisms in vitro using the Vero cell line. MTT assays, Western blotting, indirect immunofluorescence assays, virus plaque formation assays, and qRT-PCR were used to assess the effects of favipiravir on the life cycle of PPRV and the expression of RNA-dependent RNA polymerase (RdRp). Additionally, the expression levels of JAK1, STAT1, phosphorylated (p)-STAT1, PI3K, AKT, and p-AKT, as well as those of signaling molecules acting downstream of the JAK/STAT and PI3K/AKT signaling pathways, were determined by Western blotting and qRT-PCR. The results indicated that, in PPRV-infected, favipiravir-treated Vero cells, the attachment, invasion, replication, and release of PPRV were significantly inhibited, as was the expression of RdRp, when compared with that in untreated PPRV-infected cells. Furthermore, in favipiravir-treated cells, the expression of JAK1 and STAT1 was downregulated, whereas that of p-STAT1 was significantly upregulated. Similarly, the expression levels of PKR, IRF9, ISG54, and MxA proteins that are associated with innate antiviral activity in host cells were also markedly increased. Moreover, with favipiravir treatment, the expression of PI3K and p-AKT and the p-AKT/AKT ratio were significantly decreased, whereas the expression of AKT was noticeably upregulated. The expression of GSK3, NF-κB p65, p-NF-κB p65, and BAD was also increased with favipiravir treatment, while the expression of CREB, p-CREB, p-GSK3, and Bcl-2 was slightly decreased. In addition, all the p-GSK3/GSK3, p-CREB/CREB, p-NF-κB/NF-κB, and p-BAD/BAD ratios were significantly reduced in favipiravir-treated cells. These results implied that the antiviral effectivity of favipiravir against PPRV is mediated by the JAK/STAT and PI3K/AKT pathways and that favipiravir has potential for use as an effective antiviral agent against PPRV.

Highlights

  • Peste des petits ruminants (PPR) is a febrile, highly contagious, and often fatal disease, characterized by fever, mucopurulent secretions from the eyes and nose, necrotic stomatitis, bronchopneumonia, and necrohemorrhagic enteritis [1]

  • Analysis using Immunofluorescence assays (IFAs) indicated that substantially fewer virions were distributed in the cytoplasm of Peste des petits ruminants virus (PPRV)-infected, favipiravirtreated Vero cells when compared with control cells

  • Studies have demonstrated that favipiravir is converted into its active form, favipiravir-ribofuranosyl-5′-triphosphate, in vivo, and subsequently binds intracellular RNA-dependent RNA polymerase (RdRp) [23], leading to the inhibition of virus replication and transcription of the viral genome [24]

Read more

Summary

Introduction

Peste des petits ruminants (PPR) is a febrile, highly contagious, and often fatal disease, characterized by fever, mucopurulent secretions from the eyes and nose, necrotic stomatitis, bronchopneumonia, and necrohemorrhagic enteritis [1]. The scientific findings on the mechanism underlying the immunosuppressive effects induced by PPRV infection are based on comparisons with related morbilliviruses, such as rinderpest virus (RPV), measles virus (MV), and canine distemper virus (CDV) [2]. Immunosuppression may result from the inhibition of IFN synthesis, IFN-induced antiviral proteins, and immunoglobulin production, which blocks IFN signal transduction [6]. The N protein can bind to the type II receptor for the Fc region (FcγRII) of immunoglobulin G on human and murine B lymphocytes and inhibit antibody production in vitro [8]. These strategies allow morbilliviruses to evade the innate immune system and propagate, thereby increasing disease severity

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