Elsevier

Antiviral Research

Volume 156, August 2018, Pages 10-20
Antiviral Research

Research paper
Recombinant MYH9 protein C-terminal domain blocks porcine reproductive and respiratory syndrome virus internalization by direct interaction with viral glycoprotein 5

https://doi.org/10.1016/j.antiviral.2018.06.001Get rights and content

Highlights

  • MYH9 C-terminal domain protein (PRA) directly binds PRRSV GP5 protein.

  • PRA can capture PRRSV virions.

  • PRA inhibits PRRSV infection of susceptible cells in a dose-dependent manner.

  • PRA inhibits both genotype 1 and 2 PRRSV strains infection of susceptible cells.

Abstract

Porcine reproductive and respiratory syndrome (PRRS) is one of the most economically important infectious diseases impacting the swine industry worldwide. Prevention and control of PRRS have been problematic, as vaccination has achieved little success. MYH9 (encoded by the gene MYH9) is an essential cellular factor for PRRS virus (PRRSV) infection. The MYH9 C-terminal domain (designated PRA) interacts with viral glycoprotein 5 (GP5), a major PRRSV envelope protein. In this study, we investigated whether soluble PRA could serve as a novel blocking agent of PRRSV infection. Our data showed that preincubation of PRRSV with PRA inhibited virus infection of susceptible cells in a dose-dependent manner. Notably, PRA also exhibited broad-spectrum ability to inhibit infection with diverse strains of both PRRSV genotype 1 and 2. Analysis of the interaction between PRA and PRRSV GP5 revealed that PRA is able to capture PRRSV virions. In conclusion, our data suggest that PRA could serve as a novel broad-spectrum inhibitor of infection by heterogeneous PRRSV strains in vivo.

Introduction

Porcine reproductive and respiratory syndrome virus (PRRSV) is a positive-sense, single-stranded enveloped RNA virus belonging to the family Arteriviridae (Lunney et al., 2016). The genome of PRRSV is around 15 kb and contains more than 10 open reading frames (ORFs) (Dokland, 2010; Snijder and Meulenberg, 1998). ORF1a and ORF1b of PRRSV account for three-fourths of its entire genome size and encode all non-structural proteins required for viral replication, while ORFs 2–7 encode structural protein components of the virion (Lunney et al., 2016). Genetically, there are two PRRSV genotypes: Type 1, European-like (prototype Lelystad), and Type 2, North American-like (prototype VR-2332) (Mardassi et al., 1994; Wensvoort et al., 1991). Recently, based on a new proposed classification scheme, PRRSV Type 1 and Type 2 have been classified into two species in the genus Porartevirus, designated PRRSV-1 and PRRSV-2, respectively, under the new taxonomy (Adams et al., 2016; Kuhn et al., 2016).

PRRSV-1 and PRRSV-2 strains share approximately 60% nucleotide sequence identity and exhibit serotype difference (Forsberg, 2005; van Woensel et al., 1998). PRRSV infection is highly restricted to cells of the monocyte-macrophage lineage, such as porcine alveolar macrophages (PAMs) (Albina et al., 1998; Morgan et al., 2016). PRRSV propagation in vitro is generally conducted in MARC-145 cells, a subclone of the MA-104 epithelial cell line originating from monkey kidney (Kim et al., 1993). Numerous studies demonstrate that PRRSV infection is mediated by various cellular receptors or factors (Shi et al., 2015) such as heparin sulfate (HS) (Delputte et al., 2002), vimentin (Kim et al., 2006), CD151 (Wu et al., 2014), porcine CD163 (CD163) (Guo et al., 2014), sialoadhesin (CD169) (Delputte et al., 2007), DC-SIGN (CD209) (Pineyro et al., 2016), and MYH9 (Gao et al., 2016). However, only CD163 and MYH9 have been shown to be indispensable for PRRSV infection (Burkard et al., 2017; Gao et al., 2016).

Current strategies of PRRS control are inadequate despite substantial efforts by numerous researchers. Since its emergence in 1987, PRRS remains the major challenge to the swine industry globally, even though vaccines against PRRS have been commercially available for decades (Butler et al., 2014; Rowland et al., 2012; Tian et al., 2007). This lack of success underscores the urgent need for novel methods for PRRSV control and prevention.

Recently, by probing PRRSV permissive cells with an anti-idiotypic monoclonal antibody (Mab2-5G2), we identified MYH9 as the novel cellular host factor that binds to PRRSV major glycoprotein GP5 (Gao et al., 2016; Zhou et al., 2008). Subsequently, additional investigations demonstrated that MYH9 serves as a host factor for PRRSV and plays an indispensable role in PRRSV internalization by host cells (Gao et al., 2016). MYH9 is a member of the large myosin superfamily and is an endogenous non-muscle myosin II heterohexamer that is composed of two essential light chains (to stabilize the heavy chain structure), two regulatory light chains (that bind to the heavy chains to connect the head and rod domains), and two MYH9 heavy chains held together by interactions between coiled-coil rod domains (Chen et al., 2003; Fang et al., 2010). Generally, MYH9 functions as a motor protein involved in cell migration, integrin-mediated adhesion, epithelial cell polarization, cell-cell adhesion, and morphogenesis (Vicente-Manzanares et al., 2009).

MYH9 (also known as non-muscle myosin heavy chain 9) and its associated gene MYH9 have been heavily investigated for their associations with certain genetic diseases, such as MYH9-related platelet disorders and chronic kidney disease caused by mutations of MYH9 (Althaus and Greinacher, 2009; Althaus et al., 2011; Singh et al., 2009). The amino-terminal head domain of MYH9 possesses an ATPase activity and contains binding sites for both actin and the light chains of myosin (Obungu et al., 2003; Sellers, 2000). However, the roles that MYH9 plays during viral infection or other infectious diseases have not yet been thoroughly investigated. In recent years, data from other groups have demonstrated that MYH9 functions as a cellular factor for several viruses, including herpes simplex virus-1 (HSV-1), severe fever with thrombocytopenia syndrome virus (SFTSV), and Epstein-Barr virus (EBV) (Arii et al., 2010; Sun et al., 2014; Xiong et al., 2015), which suggest a novel role of MYH9 in virus infections. Meanwhile, our research has recently demonstrated that the C-terminal portion of MYH9 interacts with Mab2-5G2 (Gao et al., 2016).

The objective of this study was to investigate whether the C-terminal portion of MYH9 (hereafter referred to as PRA) is able to inhibit PRRSV replication in vitro. The recombinant PRA protein was expressed and purified from a prokaryotic expression system. Our results showed that PRA exhibited broad-spectrum anti-PRRSV activity in both susceptible monkey cells (MARC-145) and swine cells (PK-15CD163, CRL2843CD163, and PAMs). Further analysis revealed that PRA was able to capture PRRSV virions via GP5 interaction. Taken together, our data showed that PRA holds promise as a novel broad-spectrum inhibitor of infection by heterogeneous PRRSV isolates.

Section snippets

Cells, viruses, and chemicals

Porcine alveolar macrophages (PAMs) were collected from a 4-week-old PRRSV-negative pig as previously described (Xiao et al., 2014). The immortalized PAM cell line 3D4/21 (ATCC®CRL-2843™) was purchased from American Type Culture Collection (ATCC, Manassas, VA, USA). PRRSV-permissive CRL2843CD163 cells were generated by introducing porcine CD163 cDNA into ATCC®CRL-2843™ via lentiviral vector-mediated transduction under puromycin selection (Thermo Fisher Scientific, Waltham, MA, USA) at a

PRA inhibits PRRSV replication in vitro

We previously identified MYH9 as an indispensable factor for PRRSV infection in both MARC-145 and PAM cells (Gao et al., 2016). In that study, the C-terminal domain of MYH9 with aa 1651–1960 (designated PRA) (Fig. 1A) was found to bind Mab2-5G2 and was proposed to interact with PRRSV-GP5. Here, we hypothesize that PRA protein could serve as a novel blocker of PRRSV binding to permissive cells and subsequent infection. To test this hypothesis, recombinant PRA protein was produced by E. coli

Discussion

HSV-1 is the first virus reported to utilize MYH9 as its cellular receptor (Arii et al., 2010). Since then, several other viruses, including PRRSV, have been identified that enter host cells via MYH9 (Gao et al., 2016; Sun et al., 2014; Xiong et al., 2015). Moreover, interactions between MYH9 and viral proteins such as PRRSV GP5 or HSV-1 gB have been observed, as blocking of MYH9 with specific antibody inhibits virus infection in those systems as well (Arii et al., 2010; Gao et al., 2016).

Acknowledgement

We thank Dr. Ding Yu from School of Life Sciences, Fudan University for kindly providing sfGFP-TEV-His6Nd2 plasmid, Dr. Xiang-Jin Meng from Virginia Polytechnic Institute & State University for generously providing the pIR-VR2385-CA plasmid, Dr. Hanchun Yang from China Agriculture University and Dr. Kegong Tian from the National Research Center for Veterinary Medicine for kindly providing PRRSV-1 GZ11-G1 and PRRSV-1 P073-3 isolate, respectively. This study was financially supported by grants

References (58)

  • N. Singh et al.

    CKD in MYH9-related disorders

    Am. J. Kidney Dis.

    (2009)
  • M. Vanhee et al.

    Characterization of antigenic regions in the porcine reproductive and respiratory syndrome virus by the use of peptide-specific serum antibodies

    Vaccine

    (2011)
  • X. Wang et al.

    PK-15 cells transfected with porcine CD163 by PiggyBac transposon system are susceptible to porcine reproductive and respiratory syndrome virus

    J. Virol. Methods

    (2013)
  • E. Weiland et al.

    Monoclonal antibodies to the GP5 of porcine reproductive and respiratory syndrome virus are more effective in virus neutralization than monoclonal antibodies to the GP4

    Vet. Microbiol.

    (1999)
  • S. Xiao et al.

    Heme oxygenase-1 acts as an antiviral factor for porcine reproductive and respiratory syndrome virus infection and over-expression inhibits virus replication in vitro

    Antivir. Res.

    (2014)
  • Y. Zhang et al.

    Monoclonal antibodies against conformationally dependent epitopes on porcine reproductive and respiratory syndrome virus

    Vet. Microbiol.

    (1998)
  • Y.J. Zhang et al.

    Suppression of porcine reproductive and respiratory syndrome virus replication by morpholino antisense oligomers

    Vet. Microbiol.

    (2006)
  • E.M. Zhou et al.

    Generation of internal image monoclonal anti-idiotypic antibodies against idiotypic antibodies to GP5 antigen of porcine reproductive and respiratory syndrome virus

    J. Virol. Methods

    (2008)
  • M.J. Adams et al.

    Ratification vote on taxonomic proposals to the international committee on taxonomy of viruses

    Arch. Virol.

    (2016)
  • E. Albina et al.

    Interferon-alpha response to swine arterivirus (PoAV), the porcine reproductive and respiratory syndrome virus

    J. Interferon Cytokine Res.

    (1998)
  • K. Althaus et al.

    MYH9-related platelet disorders

    Semin. Thromb. Hemost.

    (2009)
  • K. Althaus et al.

    MYH9 related platelet disorders - often unknown and misdiagnosed

    Klin. Pädiatr.

    (2011)
  • J. Arii et al.

    Non-muscle myosin IIA is a functional entry receptor for herpes simplex virus-1

    Nature

    (2010)
  • L. Aschenbrenner et al.

    Uncoated endocytic vesicles require the unconventional myosin, Myo6, for rapid transport through actin barriers

    Mol. Biol. Cell

    (2004)
  • C. Burkard et al.

    Precision engineering for PRRSV resistance in pigs: macrophages from genome edited pigs lacking CD163 SRCR5 domain are fully resistant to both PRRSV genotypes while maintaining biological function

    PLoS Pathog.

    (2017)
  • J.E. Butler et al.

    Porcine reproductive and respiratory syndrome (PRRS): an immune dysregulatory pandemic

    Immunol. Res.

    (2014)
  • P.L. Delputte et al.

    Porcine arterivirus attachment to the macrophage-specific receptor sialoadhesin is dependent on the sialic acid-binding activity of the N-terminal immunoglobulin domain of sialoadhesin

    J. Virol.

    (2007)
  • P.L. Delputte et al.

    Involvement of the matrix protein in attachment of porcine reproductive and respiratory syndrome virus to a heparinlike receptor on porcine alveolar macrophages

    J. Virol.

    (2002)
  • Z. Fang et al.

    The membrane-associated protein, supervillin, accelerates F-actin-dependent rapid integrin recycling and cell motility

    Traffic (Copenhagen, Denmark)

    (2010)
  • Cited by (0)

    1

    These authors contributed equally to this work.

    View full text