Porcine 2′, 5′-oligoadenylate synthetase 2 inhibits porcine reproductive and respiratory syndrome virus replication in vitro

Porcine 2′, 5′-oligoadenylate synthetase 2 inhibits porcine reproductive and respiratory syndrome virus replication in vitro

Accepted Manuscript Porcine 2′, 5′-oligoadenylate synthetase 2 inhibits porcine reproductive and respiratory syndrome virus replication in vitro Mengm...

2MB Sizes 0 Downloads 69 Views

Accepted Manuscript Porcine 2′, 5′-oligoadenylate synthetase 2 inhibits porcine reproductive and respiratory syndrome virus replication in vitro Mengmeng Zhao, Bo Wan, Huawei Li, Jian He, Xinxin Chen, Linjian Wang, Yinbiao Wang, Sha Xie, Songlin Qiao, Gaiping Zhang PII:

S0882-4010(16)30646-5

DOI:

10.1016/j.micpath.2017.08.011

Reference:

YMPAT 2400

To appear in:

Microbial Pathogenesis

Received Date: 12 October 2016 Revised Date:

8 August 2017

Accepted Date: 9 August 2017

Please cite this article as: Zhao M, Wan B, Li H, He J, Chen X, Wang L, Wang Y, Xie S, Qiao S, Zhang G, Porcine 2′, 5′-oligoadenylate synthetase 2 inhibits porcine reproductive and respiratory syndrome virus replication in vitro, Microbial Pathogenesis (2017), doi: 10.1016/j.micpath.2017.08.011. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT

Porcine 2′, 5′-oligoadenylate synthetase 2 inhibits porcine reproductive and respiratory syndrome virus replication in vitro Mengmeng Zhao1†, Bo Wan1† Huawei Li2, Jian He1, Xinxin Chen2, Linjian Wang2, Yinbiao

College of Animal Husbandry and Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, People’s Republic of China

Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial

M AN U

2

SC

1

RI PT

Wang2, Sha Xie2, Songlin Qiao2*, Gaiping Zhang1, 2, 3*

Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, People’s Republic of China 3

Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious

TE D

Diseases and Zoonoses, Yangzhou 225009, People’s Republic of China

*Corresponding author: Gaiping Zhang [email protected]; Songlin Qiao

AC C

EP

[email protected]

Postal address: Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Huayuan Road 116, Jinshui District, 450002, Telephone number:(0371)65738179; Fax number: (0371)65738179

† These authors contributed equally to this work.

1

ACCEPTED MANUSCRIPT

Abstract Porcine reproductive and respiratory syndrome virus (PRRSV) is acknowledged a fulminating infectious

RI PT

pathogen affecting the pig farming industry, and current vaccines and drugs could hardly inhibit this virus. The 2′, 5′-oligoadenylate synthetase (OASs) have antiviral activities, but the role(s) played by porcine OAS2 in protection against PRRSV infection are unknown. Here we found that endogenous expression of the porcine

SC

OAS2 gene could be promoted by interferon (IFN)-beta or PRRSV infection in porcine alveolar macrophages.

M AN U

Knockdown of porcine OAS2 led to increases in PRRSV replication, and OAS2 expression suppressed replication of PRRSV in a retinoic acid inducible gene I (RIG-I)-dependent manner, anti-PRRSV activity of porcine OAS2 would be lost if RNase L and OAS2 were both silenced. This discovery illustrates a pathway that porcine OAS2 responses to host anti-PRRSV function.

TE D

Keywords: Porcine reproductive and respiratory syndrome virus; 2′, 5′-oligoadenylate synthetase; RIG-1;

AC C

EP

Replication; Porcine alveolar macrophages; RNase L

2

ACCEPTED MANUSCRIPT Introduction Porcine reproductive and respiratory syndrome (PRRS) is an important threat to pig farming. Since 2006 a highly pathological PRRS virus (PRRSV), of which main manifestations were high fever, late term abortions,

RI PT

reproductive failure, early farrowing, high mortality, respiratory distress, emerged in some Chinese pig farms [1].

PRRSV is the agent of PRRS; it belongs to Arteriviridae family, an enveloped positive-sense RNA virus.

SC

The length of PRRSV genome is about 15 kb; there are 10 open reading frames (ORFs), between them ORF1a

M AN U

and ORF1b encode about 80% of the genome, viral replicase polyproteins are encoded by ORF1a and ORF1b, the viral structural proteins GP2, E, GP3, GP4, GP5, GP5a, M and N were encoded by ORF2a, ORF2b and ORFs 3-7.

PRRSV modulates the host immune responses through suppression of innate immunity and persistent

TE D

infection, previous studies have shown that type I interferon could be inhibited by PRRSV, resulting in PRRSV replication and spread [2, 3]. Limited protection against PRRSV infection is provided by current vaccines and drugs; hence, developing new ways against this pathogen is in an urgent need. Type I interferons (IFNs), such as

EP

IFN-alpha and IFN-beta, play important roles in antiviral innate immunity and in modulating adaptive immune

AC C

responses to viral infection.

There are many factors effecting the antiviral innate immunity and adaptive immune responses, and interferons are one of the most important one, interferons could blind to the receptors on adjacent cell surfaces, and then JAK/STAT signaling pathway would be activated by interferons, whereby lots of IFN-stimulated genes (ISGs). For example, oligoadenylates synthetase (OAS), ISG15 are induced[4]. Among these ISGs, the 2′, 5′-OAS proteins induced by IFN are present in all vertebrates. The OAS proteins could catalyze the production of 2′, 5′-linked oligoadenylates, the 2′, 5′-linked oligoadenylates are presented

3

ACCEPTED MANUSCRIPT with formula pppA-(2, p5’A) n (n>1). RNase L was activated by 2′, 5′-linked oligoadenylates, intracellular 2′, 5′-linked oligoadenylates degraded viral and host RNAs [5]. The 2′, 5′-OAS belongs to a family with low amino acid sequence homology with proteins stimulated by

RI PT

interferons. OAS1, OAS2, OAS3, and OASL (OAS-like protein) are four genes encoding four proteins in the OAS family; there is one basal OAS functional unit in OAS1, two basal OAS functional units in OAS2, three basal OAS functional units in OAS3, a tandem ubiquitin unit in the C-terminal domain of OASL enzyme that

SC

lacks 2′, 5′-OAS activity [6-10]. OAS1 and OASL are distributed widely in vertebrates, whereas OAS2 and

M AN U

OAS3 proteins could only be found in mammals [11].

The porcine OAS family contains OAS1a, OAS1b, OAS2, and OASL, but no OAS3 [12]. The amino acid sequence homology between OAS1a and OAS1b is 89.1% [8], and porcine OAS1a was the first to be crystallized [8]. OAS1a shares 73% nucleotide sequence identity with human OAS1 , enzymatic characteristics

mouse OASs [13].

TE D

could be found in OAS1a, while high sequence homology was found between porcine OASs and human and

Antiviral effects of OAS proteins have been demonstrated in many researches. OAS1b, for example, was

EP

able to restrict West Nile virus (WNV) replication [14, 15], but this effect was not modulated by the OAS/RNase

AC C

L pathway [16, 17]. Additionally, while human OAS1 and OAS3 can inhibit dengue virus [18], OAS1 and OAS3 can inhibit hepatitis C virus [18, 19], but the effect would be lost without effect of RNase L. The single nucleotide polymorphism could be responsible for increasing the probability of WNV disease [18], and while extracellular OAS1 possesses strong antiviral activity [20]. Although the antiviral effects of other OAS proteins have been well-studied, it remains unclear whether porcine OAS2 protein can inhibit PRRSV. In this paper we report the discovery of antiviral characteristics of porcine OAS2 against PRRSV; the results indicate that porcine OAS2 has antiviral activity against PRRSV.

4

ACCEPTED MANUSCRIPT Material and methods 2.1. Cell lines and virus Porcine alveolar macrophages (PAMs) were obtained from the lungs of seven-week-old PRRSV-free pigs;

RI PT

they were maintained in RPMI 1640 medium with 10% fetal bovine serum (FBS). CRL-2843-CD163 cells were obtained from Professor Enmin Zhou of Northwest A & F University, China[21]. The CRL-2843-CD163 cells were maintained in RPMI 1640 medium supplemented with 6% FBS

SC

(GIBCO, Invitrogen, USA) at 37℃ in 5% CO2. The CRL-2843-CD163 cells were modified from CRL-2843

M AN U

cells, via CD163 gene transfection into CRL-2843 cells, to confer PRRSV with the ability to infect the CRL-2843-CD163 cells.

The PRRSV BJ-4 strain was provided by Professor Hanchun Yang from China Agricultural University. We strictly followed the rules for animal care which have been approved by the State Council of the People’s

TE D

Republic of China [22-24].

2.2 Analysis of OAS2 in PAMs which are infected with BJ-4 PRRSV PAMs were infected with PRRSV BJ-4 at a multiplicity of infection (MOI) of 1 or 0.1 for the times

EP

indicated (i.e., 0, 6, 24, 36, 48 h), after which PAMs were collected and subjected to quantitative real-time PCR

AC C

(qRT-PCR) to analyze the OAS2 expression levels.

2.3 Interferon treatment

The PAMs were treated with 1000 IU/ml of IFN-beta (Peprotech, Rocky Hill, NJ, USA) for the times indicated (i.e., 0, 6, 12, 24, 36, and 48), after which the cells were collected and subjected to qRT-PCR to analyze the OAS2 expression levels.

2.4 Plasmid construction The porcine OAS2 gene was amplified from cDNA from the PAMs using the following primer sequences:

5

ACCEPTED MANUSCRIPT 5′-TATGAATTCAATGGGAAACTGGGGGTCCC-3′ (OAS2-F) and 5′-CGCGACGACTCAGTCCATGAAT CTCC-3′ (OAS2-R). Purified PCR products were digested and inserted into the p3xFLAG-CMV -7.1 vector (Sigma-Aldrich, St. Louis, USA) to generate the pFLAG-pOAS2 expression vector.

RI PT

2.5 RNA interference Nonsense siRNA, OAS2 siRNA, retinoic acid inducible gene I (RIG-1) siRNA and RNase L siRNA were designed and synthesized by GenePharma Co., Ltd (Suzhou, China).

SC

2.6 Cell transfection and viral infections

M AN U

CRL-2843-CD163 cells were maintained in 24-well plates and, when the cell monolayer reached 60-70% confluence, 800 ng of the pFLAG-pOAS2 expression vector or the control expression vector (p3xFLAG-CMV -7.1) were transfected into the cells using lipofectamine 2000 reagent (Invitrogen, USA) according to the manufacturer’s instructions. After 24 h, the cells were infected with PRRSV BJ-4 (MOI=1) for another 24 h, and

TE D

the mRNA levels of PRRSV N (ORF7), OAS2, interferon regulatory factor (IRF)3, IRF7, Toll-like receptor (TLR)3, TLR7, RIG-1, melanoma differentiation-associated protein 5 (MDA5), myeloid differentiation primary response 88 (MyD88), TANK binding kinase 1 (TBK1), nuclear factor-κB (NF-κB), interleukin (IL)-8, IL-1

AC C

western blotting.

EP

beta, IFN-alpha, and IFN-beta mRNAs were detected by qRT-PCR. IRF3 and pIRF3 proteins were detected by

PAMs were transfected with 60 nM of OAS2 siRNA, RNase L siRNA, RIG-1 siRNA or nonsense siRNA, and then infected with PRRSV BJ-4 (MOI=1) for another 24 h, and PRRSV N mRNA was detected by qRT-PCR.

CRL-2843-CD163 cells were transfected with 60 nM of RNase L siRNA, RIG-1 siRNA or nonsense siRNA and, the CRL-2843-CD163 cells, which RNase L or RIG-1 were knockdown, were transfected with pFLAG-pOAS2 or the negative control vector, and infected with BJ-4 (MOI=1) for another 24 h, after which

6

ACCEPTED MANUSCRIPT PRRSV N mRNA was detected by qRT-PCR.

2.7 Cell viability assays The enhanced Cell Counting Kit-8 assay (Beyotime Biotech, China) here was used to evaluate cytotoxicity

RI PT

levels, whereby pFLAG-pOAS2 or p3xFLAG-CMV TM-7.1 expression vectors (800 ng each) were transfected individually into CRL-2843-CD163 cells plated in 24-well plates, and the cytotoxicity of OAS2 was evaluated. PAMs plated in 24-well plates were transfected with 60 nM of OAS2 siRNA, RNase L siRNA, RIG-1

SC

siRNA or nonsense siRNA for 48 h, the cytotoxicity was evaluated as stated above.

M AN U

2.8 Quantitative real-time polymerase chain reaction (qRT-PCR)

Total RNA from the cells was treated with TRIzol (Invitrogen, USA). Reverse transcriptase reactions were performed using a PrimeScript™ RT Reagent Kit with gDNA Eraser (Takara, Dalian, China), while SYBR Ex Taq™ (Takara, Dalian, China) was used in the polymerase chain reactions, and the machine was the 7500 Fast

2.9 Western blotting

TE D

Real-time PCR System (Applied Biosystems, Foster city, CA).

CRL-2843-CD163 cells were seeded in 24-well plates and then transfected with the pFLAG-OAS2

EP

expression vector, and then infected with PRRSV BJ-4 (MOI=1). 48 h later, western blotting was performed as

AC C

described previously [25] using either an anti-Flag monoclonal antibody (1:200, Abnova, Taipei, Taiwan) , an anti-GAPDH monoclonal antibody (1:1000, Beijing Biosynthesis Biotech Co., Ltd, China), an anti-IRF3 monoclonal antibody (1:1000, Proteintech Group, China), or an anti-pIRF3 monoclonal antibody (1:1000, Cell Signaling Technology, USA). The membranes were washed three times with Tris-buffered saline containing Tween-20 (TBST), and then reacted with

horseradish peroxide-conjugated goat anti-rabbit IgG (1:10000,

Zhongshan Golden bridge Biotechnology, China) or horseradish peroxide-conjugated goat anti-mouse IgG (1:10000, Jackson ImmunoResearch, West Grove, PA, USA) for one hour at 37℃, and then washed with TBST

7

ACCEPTED MANUSCRIPT five times, and then developed using enhanced Chemiluminescence (Cell Signaling Technology, Danvers, MA, USA).

2.10 Viral titers

RI PT

Supernatants from the transfected groups were obtained at 48 h post infection, and PRRSV titers were measured.

2.11 Luciferase reporter assays

SC

CRL-2843-CD163 cells were seed in 24 well-plates, when they reached 70% confluence, 200 ng of

M AN U

pIFN-Beta-Luc (luciferase reporter plasmid), 80 ng of pRL-TK, and the expression vector (500 ng) or the control vector were transfected into the cells with lipofectamine 2000 (Invitrogen, USA). 24 h later, these cells were transfected with 1.5 µg Poly (I:C) for 9 h, Luciferase Reporter System (Promega, Madison, WI, USA) was used to detect the luciferase activity, and the CRL-2843-CD163 cells were also

TE D

infected with PRRSV for 24 hours(MOI=1), and luciferase reporter assays were performed.

2.12 Secreted IFN-beta protein measurements

procedures.

EP

The secreted IFN-beta was detected with an ELISA kit (Cloud-Clone Group, China) by the manufacturer’s

AC C

2.13 Statistical analysis

Every experiment has three independent experiments; each independent experiment has three technical replicates. GraphPad Prism 5 was used to analyze the results using Student's t-test.

8

ACCEPTED MANUSCRIPT Results 3.1 IFN-beta and PRRSV induce porcine OAS2 mRNA expression in PAMs The results showed that from 6 hours post treatment with 1000 IU/ml IFN-beta, porcine OAS2 mRNA

RI PT

expression level increased by 40-fold in PAMs (Fig. 1A). The peak mRNA expression level for the OAS2 gene occurred at 12 h post treatment in the PAMs.

The expression kinetics of OAS2 in the PAMs stimulated by PRRSV was also investigated. 24 h later, the

M AN U

following PRRSV treatment resulted in only an 8-fold increase.

SC

highest level of OAS expression following PRRSV infection was detected (Fig. 1B). Induction of OAS2 mRNA

3.2 OAS2 overexpression inhibits PRRSV replication in CRL-2843-CD163 cells Immunoblotting revealed that porcine OAS2 was successfully expressed in the CRL-2843-CD163 cells transfected with the pFLAG-pOAS2 expression vector (Fig. 1C). The transfection efficiency of the plasmid (800

TE D

ng) in the 24-well-plate cells was about 25 %, and no significant cytotoxic effect was observed. Relative expression of PRRSV ORF7 mRNA of CRL-2843-CD163 cells transfected with pFLAG-pOAS2 was lower (Fig. 1D). We also found that the TCID50 of the supernatants from cells transfected with

EP

pFLAG-pOAS2 was lower (Fig. 1E).

AC C

As shown in Fig. 2A, after infection with PRRSV, OAS2 induced a 2.2-fold increase in IFN-alpha mRNA, a 2.0-fold increase in IFN-beta mRNA, a 2.4-fold increase in IRF3 mRNA, a 4.3-fold increase in TLR-3 mRNA, a 4.3-fold increase in RIG-1 mRNA, a 2.3-fold increase in NF-κB mRNA, a 2.6-fold increase in MDA5 mRNA, a 6.3-fold increase in MyD88 mRNA, a 1.9-fold increase in TBK1 mRNA, and no increases in TNF-alpha, IL-8, IL-1beta, IRF7 mRNA levels. In response to PRRSV infection, the supernatant from the CRL-2843-CD163 cells transfected with pFLAG-pOAS2 had a higher concentration of IFN-beta than that of the CRL-2843-CD163 cells transfected with

9

ACCEPTED MANUSCRIPT the p3xFLAG-CMV -7.1 vector (Fig. 2C). The active and latent forms of IRF-3 were also analyzed by western blotting, and as shown in Fig. 2D, OAS2 enhanced the phosphorylation level of IRF-3. IFN-beta promoter activity got significantly enhanced by OAS2 induced by Poly (I:C) and PRRSV (Fig. 2E).

RI PT

3.3 Knockdown of OAS2 led to increases in PRRSV replication in PAMs Efficiency of OAS2 gene knockdown in PAMs was approximately 70% (Fig. 3A), while PRRSV ORF7 mRNA increased significantly in the OAS2 knockdown group (Fig. 3D). The TCID50 of the supernatants from

SC

the OAS2 knockdown group was higher (Fig. 3C).

M AN U

In siRNA rescue experiments, 60 nM of si-OAS2, in combination with 800 ng of the pFLAG-pOAS2 plasmid were transfected into the CRL-2843-CD163 cells, and 24 h later, cells were infected with PRRSV. The results showed that no increase in the PRRSV ORF7 mRNA expression level was found.

3.4 RNase L- and RIG-1-dependent antiviral activities

TE D

In the PAMs, 60 nM of RNase L siRNA induced a 33% knockdown of RNase L mRNA (Fig. 4A), which in turn induced a 1.9-fold increase in PRRSV ORF7 mRNA (Fig. 4B). In order to detect whether the anti-PRRSV function rely on RNase L, 30 nM of OAS2 siRNA and 30 nM of RNase L siRNA were co-transfected into PAMs,

EP

followed by infection with PRRSV (MOI=1). In comparison with the control, no increase in PRRSV ORF7

AC C

mRNA expression in the OAS2/RNase L co-silenced group was found (Fig. 4C). For the PAMs, 60 nM of RIG-1 siRNA induced a 30% knockdown of RIG-1 mRNA (Fig.4D), and this facilitated a 1.7-fold increase in PRRSV ORF7 mRNA (Fig. 4E). To determine whether the anti-PRRSV activity was dependent upon the RIG-1 pathway, both of the OAS2 siRNA and RIG-1 siRNA were transfected into PAMs, and then PRRSV infect the cells (MOI=1). We found no increase in PRRSV ORF7 mRNA expression in the OAS2/ RIG-1 co-silencing group (Fig. 4F). In the CRL-2843-CD163 cells, 60 nM of RNase L siRNA induced a 29% knockdown of RNase L mRNA

10

ACCEPTED MANUSCRIPT (Fig. 4G), and this induced a 1.8-fold increase in PRRSV ORF7 mRNA (Fig. 4H). To determine whether the anti-PRRSV activity was dependent on the RNase L pathway, CRL-2843-CD163 cells with RNase L knocked-down were transfected with pFLAG-pOAS2 or the negative control vector. No increase in PRRSV

RI PT

ORF7 mRNA expression in the RNase L knockdown group was observed (Fig. 4I). In the CRL-2843-CD163 cells, 60 nM of RIG-1 siRNA induced a 28% knockdown of RIG-1 mRNA (Fig.4L), and this facilitated a 1.6-fold increase in PRRSV ORF7 mRNA (Fig. 4M). To determine whether the

SC

anti-PRRSV activity was dependent on the RIG-1 pathway, CRL-2843-CD163 cells with RIG-1 knocked-down

M AN U

were transfected with pFLAG-pOAS2 or the negative control vector. No increase in PRRSV ORF7 mRNA

AC C

EP

TE D

expression in the RNase L knockdown group was observed (Fig. 4N).

11

ACCEPTED MANUSCRIPT Discussion The current vaccines could hardly inhibit PRRSV infection, there are some factors: high frequency of PRRSV genome mutation, antibody dependent enhancement (ADE) effect, and persistence of virus in the

RI PT

infected pigs. So finding new antiviral therapy becomes a useful method to control PRRSV. Up to now, some ISGs are reported to inhibit viruses, for example, IFIT3[26, 27], ISG15[4], Mx2[28], but the effect of OAS were not well-known.

SC

This paper has shown PRRSV replication was significantly inhibited by overexpression of porcine OAS2,

M AN U

but increased following knockdown of porcine OAS2. The antiviral mechanism of OAS is shown in some other researches, but not well understood, overexpression of porcine OAS2 could trigger RIG-I and some other molecules, it has been shown human OASL binds RIG-I and has antiviral activities [29]. Porcine OAS2, which has also been found to have antiviral activities, can also activate the RIG-I pathway, and the innate immune

TE D

character could share sort of similarity for pigs and humans in this respect. During the experiments, we found that some antibodies used for human signaling molecules could not be used in the porcine signaling molecules, maybe some amino acids mutations change the recognition clues for porcine signaling molecules, so this

EP

become a question for the porcine innate immunes.

AC C

Zheng et al reported that porcine OAS2 can inhibit JEV, via porcine OAS2 functioning through OAS/RNase L signal channel [30], our result consistent with that found for JEV. For the porcine OAS1, we found that overexpressions of OAS1 also inhibit PRRSV, but the effect is not as significant as that of porcine OAS2. Knockdown of OAS1 could increase the replication of PRRSV, and also the effect was not as good as that of porcine OAS2 (date not shown), so we focused on antiviral effect of porcine OAS2 only. We first discovered that porcine OAS2 could induce the phosphorylation of IRF3 to enhance IFN pathway. The host gene could inhibit the virus through this way, there are reports showing that mouse OASL1 could

12

ACCEPTED MANUSCRIPT inhibit type I interferon [31], this is different from the porcine OAS, and the resultant innate immune character did not share similarity between pigs and mouse in this respect. Porcine OAS2 has enzymatic and antiviral activities, whether its antiviral activities connect with enzymatic activities needs further research. The porcine

RI PT

OAS2 located in the cytoplasm (date not shown), the PRRSV also replicate in the cytoplasm, whether the porcine OAS2 interacted with viral proteins in the cytoplasm needs more research, maybe the PRRSV could

are required for the antivirus activity needs more research.

SC

escape the innate immunity through inhibiting OAS2 or other OAS, and which critical regions or amino acids

M AN U

PRRSV can replicate in the Marc-145 cells which are derived from monkey kidney[32], and in PAMs which are target porcine cells in vivo [33], but PAMs are not easy for plasmid DNA to be transfected, so we used CRL-2843-CD163 cells to investigate the antiviral activity of OAS2 and found that the transfection efficiency of 800 ng of OAS2 in 24-well-plates cells was about 30%. The CRL-2843-CD163 cell line was

TE D

modified from CRL-2843 cells, which involved CD163 transfection into the CRL-2843 cells. Thus, PRRSV has the ability to infect CRL-2843-CD163 cells, a result consistent with that obtained for CRL-2843-CD163 cells and PAMs, which highlights the utility of CRL-2843-CD163 cells for studying the pathogenesis of PRRSV;

EP

therefore, use of the CRL-2843-CD163 line could sidestep the need for proper preparation of PAMs.

AC C

Given the antiviral activities of OAS2 against RNA viruses [19, 34, 35], it is possible that OAS2 is required for the prevention of RNA viruses [36]. Therefore, it seems that OAS2 is a universal pattern recognition receptor for the defense against invasion with DNA and RNA viruses. In summary, after cloning of porcine OAS2 gene, it was expressed in CRL-2843-CD163 cells. Porcine OAS2 is an inhibitor to PRRSV; it could increase IFN-beta expression. Further studies on the structure and function of porcine OAS2 mediated signaling pathway are needed, maybe these research could help find new ways to control PRRSV infection.

13

ACCEPTED MANUSCRIPT

Acknowledgment: Our work received funds from the National Natural Science Fund (No. 31490601,

RI PT

31502040), and the National Program on the Key Basic Research Project (2014CB542702).

SC

References

[1] Tian K, Yu X, Zhao T, Feng Y, Cao Z, Wang C, et al. Emergence of fatal PRRSV variants: unparalleled outbreaks of atypical PRRS in China and molecular dissection of the unique hallmark. PloS one. 2007;2:e526.

M AN U

[2] Nan Y, Wang R, Shen M, Faaberg KS, Samal SK, Zhang YJ. Induction of type I interferons by a novel porcine reproductive and respiratory syndrome virus isolate. Virology. 2012;432:261-70. [3] Buddaert W, Van Reeth K, Pensaert M. In vivo and in vitro interferon (IFN) studies with the porcine reproductive and respiratory syndrome virus (PRRSV). Advances in experimental medicine and biology. 1998;440:461-7.

[4] Sun Z, Li Y, Ransburgh R, Snijder EJ, Fang Y. Nonstructural protein 2 of porcine reproductive and respiratory syndrome virus inhibits the antiviral function of interferon-stimulated gene 15. Journal of virology.

TE D

2012;86:3839-50.

[5] Hovanessian AG. On the discovery of interferon-inducible, double-stranded RNA activated enzymes: the 2'-5'oligoadenylate synthetases and the protein kinase PKR. Cytokine & growth factor reviews. 2007;18:351-61. [6] Hartmann R, Olsen HS, Widder S, Jorgensen R, Justesen J. p59OASL, a 2'-5' oligoadenylate synthetase like protein: a novel human gene related to the 2'-5' oligoadenylate synthetase family. Nucleic acids research.

EP

1998;26:4121-8.

[7] Justesen J, Hartmann R, Kjeldgaard NO. Gene structure and function of the 2'-5'-oligoadenylate synthetase family. Cellular and molecular life sciences : CMLS. 2000;57:1593-612.

AC C

[8] Hartmann R, Justesen J, Sarkar SN, Sen GC, Yee VC. Crystal structure of the 2'-specific and double-stranded RNA-activated

interferon-induced

antiviral

protein

2'-5'-oligoadenylate

synthetase.

Molecular

cell.

2003;12:1173-85.

[9] Kristiansen H, Gad HH, Eskildsen-Larsen S, Despres P, Hartmann R. The oligoadenylate synthetase family: an ancient protein family with multiple antiviral activities. Journal of interferon & cytokine research : the official journal of the International Society for Interferon and Cytokine Research. 2011;31:41-7. [10] Hovanessian AG, Kerr IM. The (2'-5') oligoadenylate (pppA2'-5'A2'-5'A) synthetase and protein kinase(s) from interferon-treated cells. European journal of biochemistry / FEBS. 1979;93:515-26. [11] Kjaer KH, Poulsen JB, Reintamm T, Saby E, Martensen PM, Kelve M, et al. Evolution of the 2'-5'-oligoadenylate synthetase family in eukaryotes and bacteria. Journal of molecular evolution. 2009;69:612-24. [12] Perelygin AA, Zharkikh AA, Scherbik SV, Brinton MA. The mammalian 2'-5' oligoadenylate synthetase gene family: evidence for concerted evolution of paralogous Oas1 genes in Rodentia and Artiodactyla. Journal

14

ACCEPTED MANUSCRIPT of molecular evolution. 2006;63:562-76. [13] Lohofener J, Steinke N, Kay-Fedorov P, Baruch P, Nikulin A, Tishchenko S, et al. The Activation Mechanism of 2'-5'-Oligoadenylate Synthetase Gives New Insights Into OAS/cGAS Triggers of Innate Immunity. Structure. 2015;23:851-62. [14] Mashimo T, Lucas M, Simon-Chazottes D, Frenkiel MP, Montagutelli X, Ceccaldi PE, et al. A nonsense mutation in the gene encoding 2'-5'-oligoadenylate synthetase/L1 isoform is associated with West Nile virus susceptibility in laboratory mice. Proceedings of the National Academy of Sciences of the United States of

RI PT

America. 2002;99:11311-6.

[15] Perelygin AA, Scherbik SV, Zhulin IB, Stockman BM, Li Y, Brinton MA. Positional cloning of the murine flavivirus resistance gene. Proceedings of the National Academy of Sciences of the United States of America. 2002;99:9322-7.

[16] Elbahesh H, Jha BK, Silverman RH, Scherbik SV, Brinton MA. The Flvr-encoded murine oligoadenylate

SC

synthetase 1b (Oas1b) suppresses 2-5A synthesis in intact cells. Virology. 2011;409:262-70.

[17] Courtney SC, Di H, Stockman BM, Liu H, Scherbik SV, Brinton MA. Identification of novel host cell binding partners of Oas1b, the protein conferring resistance to flavivirus-induced disease in mice. Journal of virology. 2012;86:7953-63.

M AN U

[18] Lin RJ, Yu HP, Chang BL, Tang WC, Liao CL, Lin YL. Distinct antiviral roles for human 2',5'-oligoadenylate synthetase family members against dengue virus infection. J Immunol. 2009;183:8035-43. [19] Kwon YC, Kang JI, Hwang SB, Ahn BY. The ribonuclease L-dependent antiviral roles of human 2',5'-oligoadenylate synthetase family members against hepatitis C virus. FEBS letters. 2013;587:156-64. [20] Kristiansen H, Scherer CA, McVean M, Iadonato SP, Vends S, Thavachelvam K, et al. Extracellular 2'-5' oligoadenylate synthetase stimulates RNase L-independent antiviral activity: a novel mechanism of virus-induced innate immunity. Journal of virology. 2010;84:11898-904.

TE D

[21] Wang X, Ruifang. W, Shuqi. X, Enmin. Z. Generation of a porcine alveolar macrophage cell line stably expressing CD163 by lentiviral vector for the production of porcine reproductive and respiratory syndrome virus. acta veterinaria et zootechnica sinic. 2013;44:1797-804.

[22] Qiao S, Feng L, Bao D, Guo J, Wan B, Xiao Z, et al. Porcine reproductive and respiratory syndrome virus and bacterial endotoxin act in synergy to amplify the inflammatory response of infected macrophages.

EP

Veterinary microbiology. 2011;149:213-20.

[23] Zhao M, Ning Z, Wang H, Huang Z, Zhang M, Zhang G. Sequence analysis of NSP9 gene of 25 PRRSV strains from Guangdong province, subtropical southern China. Virus genes. 2013;46:88-96.

AC C

[24] Wang LJ, Xie W, Chen XX, Qiao S, Zhao M, Gu Y, et al. Molecular epidemiology of porcine reproductive and respiratory syndrome virus in Central China since 2014: The prevalence of NADC30-like PRRSVs. Microbial pathogenesis. 2017;109:20-8. [25] Zhao M, Qian J, Xie J, Cui T, Feng S, Wang G, et al. Characterization of polyclonal antibodies against nonstructural protein 9 from the porcine reproductive and respiratory syndrome virus. Frontiers of Agricultural Science and Engineering. 2016;3:153-60. [26] Miller LC, Jiang Z, Sang Y, Harhay GP, Lager KM. Evolutionary characterization of pig interferon-inducible transmembrane gene family and member expression dynamics in tracheobronchial lymph nodes of pigs infected with swine respiratory disease viruses. Veterinary immunology and immunopathology. 2014;159:180-91. [27] Zhang L, Liu J, Bai J, Du Y, Wang X, Liu X, et al. Poly(I:C) inhibits porcine reproductive and respiratory syndrome virus replication in MARC-145 cells via activation of IFIT3. Antiviral research. 2013;99:197-206. [28] Wang H, Bai J, Fan B, Li Y, Zhang Q, Jiang P. The Interferon-Induced Mx2 Inhibits Porcine Reproductive

15

ACCEPTED MANUSCRIPT and Respiratory Syndrome Virus Replication. Journal of interferon & cytokine research : the official journal of the International Society for Interferon and Cytokine Research. 2016;36:129-39. [29] Zhu J, Zhang Y, Ghosh A, Cuevas RA, Forero A, Dhar J, et al. Antiviral activity of human OASL protein is mediated by enhancing signaling of the RIG-I RNA sensor. Immunity. 2014;40:936-48. [30] Zheng S, Zhu D, Lian X, Liu W, Cao R, Chen P. Porcine 2', 5'-oligoadenylate synthetases inhibit Japanese encephalitis virus replication in vitro. Journal of medical virology. 2016;88:760-8. [31] Lee MS, Kim B, Oh GT, Kim YJ. OASL1 inhibits translation of the type I interferon-regulating

RI PT

transcription factor IRF7. Nature immunology. 2013;14:346-55.

[32] Kim HS, Kwang J, Yoon IJ, Joo HS, Frey ML. Enhanced replication of porcine reproductive and respiratory syndrome (PRRS) virus in a homogeneous subpopulation of MA-104 cell line. Archives of virology. 1993;133:477-83.

[33] Rossow KD, Collins JE, Goyal SM, Nelson EA, Christopher-Hennings J, Benfield DA. Pathogenesis of

SC

porcine reproductive and respiratory syndrome virus infection in gnotobiotic pigs. Veterinary pathology. 1995;32:361-73.

[34] Zhao L, Jha Babal K, Wu A, Elliott R, Ziebuhr J, Gorbalenya Alexander E, et al. Antagonism of the Interferon-Induced OAS-RNase L Pathway by Murine Coronavirus ns2 Protein Is Required for Virus

M AN U

Replication and Liver Pathology. Cell host & microbe. 2012;11:607-16.

[35] Zhao M, Wang L, Li S. Influenza A Virus-Host Protein Interactions Control Viral Pathogenesis. International journal of molecular sciences. 2017;18:1673.

[36] Wang J, Chu B, Du L, Han Y, Zhang X, Fan S, et al. Molecular cloning and functional characterization of

AC C

EP

TE D

porcine cyclic GMP-AMP synthase. Molecular immunology. 2015;65:436-45.

16

ACCEPTED MANUSCRIPT

TE D

M AN U

SC

RI PT

Figure Legends

EP

Fig. 1 OAS2 overexpression inhibits PRRSV replication (A) PAMs were stimulated by IFN-beta, at different time points (6, 12, 24, 36, and 48); PAMs were subjected to

AC C

qRT-PCR to quantify OAS2 mRNA levels. (B) PRRSV infected the PAMs, at different time points (6, 12, 24, 36, and 48); PAMs were subjected to qRT-PCR to quantify OAS2 mRNA levels. (C) CRL-2843-CD163 cells were transfected with the pFLAG-pOAS2 vector, 48 h later; western blotting was performed to detect expression. (D) pFLAG-pOAS2 or the negative control vectors were transfected into CRL-2843-CD163 cells; 24 h later, PRRSV infected the cells. 48 h later, qRT-PCR was used to measure the PRRSV ORF7 mRNA expression (* means a P value was less than 0.05, significant differences. ** means a P value was less than 0.01, very significant differences). (E) TCID50 was used to measure the viral titers (* means a P value was less than 0.05, 17

ACCEPTED MANUSCRIPT

Fig. 2 OAS2 enhances RIG-1 signaling

M AN U

SC

RI PT

significant differences. ** means a P value was less than 0.01, very significant differences).

(A) pFLAG-pOAS2 were transfected into CRL-2843-CD163 cells; 24 h later PRRSV infected the cells, 48 h

TE D

later qRT-PCR was used to measure IFN-alpha, IFN-beta, IL-8, IL-1beta, and TNF-alpha mRNA expression levels (* means a P value was less than 0.05, significant differences. ** means a P value was less than 0.01, very significant differences). (B) qRT-PCR was performed to measure IFR3, IFR7, TLR3, TLR7, RIG-1, NF-κB,

EP

MDA5, MyD88, and TBK1 mRNA expression levels (* means a P value was less than 0.05, significant

AC C

differences. ** means a P value was less than 0.01, very significant differences). (C) ELISA was performed to measure IFN-beta protein levels (* means a P value was less than 0.05, significant differences. ** means a P value was less than 0.01, very significant differences). (D) Immunoblotting was used to analyze the cells with anti-IRF-3 and anti-pIRF-3 antibodies. (E) OAS2 overexpression enhanced the activity of the IFN-beta promoter. On day 0, the CRL-2843-CD163 cells were cultured. On day 1, 200 ng of pIFN-Beta-Luc, 80 ng of pRL-TK, and the expression vector (500 ng) or the control were transfected into the cells. On day 2, 1.5 µg of Poly (I:C) treated the cells, 9 h later IFN-beta promoter activities were assessed (* means a P value was less than 0.05,

18

ACCEPTED MANUSCRIPT significant differences. ** means a P value was less than 0.01, very significant differences). PRRSV also infected cells (MOI=1), and on day 3, luciferase reporter assays were performed, followed by cell harvesting 24 h later (* means a P value was less than 0.05, significant differences. ** means a P value was less than 0.01,

AC C

EP

TE D

M AN U

SC

RI PT

very significant differences).

Fig. 3 OAS2 knockdown in PAMs enhances PRRSV replication (A) 60 nM of OAS2 siRNA (si-OAS2) or negative control siRNA (si-NC) were transfected into PAMs, 24 h later, PRRSV infected the cells for 48h (MOI=1). qRT-PCR was used to measure OAS2 expression (* means a P value was less than 0.05, significant differences. ** means a P value was less than 0.01, very significant differences). (B) Evaluation of cell cytotoxicity of si-OAS2 in PAMs. (C) Viral titers were investigated via 19

ACCEPTED MANUSCRIPT TCID50 (* means a P value was less than 0.05, significant differences. ** means a P value was less than 0.01, very significant differences). (D) qRT-PCR was used to measure PRRSV N expression levels (* means a P value

AC C

EP

TE D

M AN U

SC

RI PT

was less than 0.05, significant differences. ** means a P value was less than 0.01, very significant differences).

20

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

Fig. 4 The anti-PRRSV activity of OAS2 is dependent on RNase L and RIG-1

21

ACCEPTED MANUSCRIPT (A) PAMs were transfected with 60nM of RNase L siRNA (si- RNase L) or negative control siRNA (si-NC); At 24 h past transfection, PRRSV infected the cells, RNase L expression levels were measured by qRT-PCR (* means a P value was less than 0.05, significant differences. ** means a P value was less than 0.01, very

RI PT

significant differences). (B) RRSV N mRNA expression levels were measured by qRT-PCR (* means a P value was less than 0.05, significant differences. ** means a P value was less than 0.01, very significant differences). (C) 30nM of RNase L siRNA and 30nM of OAS2 siRNA were co-transfected into PAMs; 24 h later, cells were

SC

infected with PRRSV (MOI=1), RRSV N mRNA expression levels were measured by qRT-PCR. (D) PAMs

M AN U

were transfected with 60nM of RNase L siRNA (si- RIG-1) or negative control siRNA (si-Ctrl); 24 h later, cells were infected with PRRSV (MOI=1), RNase L mRNA expression levels were measured by qRT-PCR (* means a P value was less than 0.05, significant differences. ** means a P value was less than 0.01, very significant differences). (E) qRT-PCR was used to measure RRSV N mRNA expression levels (* means a P

TE D

value was less than 0.05, significant differences. ** means a P value was less than 0.01, very significant differences). (F) 30nM of RIG-1 siRNA and 30nM of OAS2 siRNA were co-transfected into PAMs; 24 h later, cells were infected with PRRSV (MOI=1), RRSV N mRNA expression levels were measured by qRT-PCR.

EP

(G) CRL-2843-CD163 cells were transfected with 60nM of RNase L siRNA (si- RNase L) or negative control

AC C

siRNA (si-NC); 24 h later, PRRSV was inoculated at a MOI of 1 for 48 h. qRT-PCR was used to measure RNase L expression levels (* means a P value was less than 0.05, significant differences. ** means a P value was less than 0.01, very significant differences). (H) qRT-PCR was used to measure RRSV N mRNA expression levels (* means a P value was less than 0.05, significant differences. ** means a P value was less than 0.01, very significant differences). (I) CRL-2843-CD163 cells were transfected with 60nM of RNase L siRNA; 24 h later, the CRL-2843-CD163 cells with RNase L knocked down were transfected with 400 ng of pFLAG-pOAS2 or the negative control vector, and cells were infected with PRRSV, qRT-PCR was used to measure RRSV N mRNA

22

ACCEPTED MANUSCRIPT expression levels. (L) CRL-2843-CD163 cells were transfected with 60nM of RNase L siRNA (si- RNase L) or negative control siRNA (si-NC); 24 h later, cells were infected with PRRSV. qRT-PCR was used to measure RNase L expression (* means a P value was less than 0.05, significant differences. ** means a P value was less

RI PT

than 0.01, very significant differences). (M) qRT-PCR was used to measure RRSV N mRNA expression levels (* means a P value was less than 0.05, significant differences. ** means a P value was less than 0.01, very significant differences). (N) CRL-2843-CD163 cells were transfected with 60nM of RIG-1 siRNA,24 h later, the

SC

CRL-2843-CD163 cells with RIG-1 knocked down were transfected with 400 ng of pFLAG-pOAS2 or the

M AN U

negative control vector, and cells were infected with PRRSV, qRT-PCR was used to measure RRSV N mRNA

AC C

EP

TE D

expression levels.

23

ACCEPTED MANUSCRIPT Table1 Primers used in the research primer sequence TATGAATTCAATGGGAAACTGGGGGTCCC

OAS2- R

CGCGACGACTCAGTCCATGAATCTCC

qOAS2-F

ATGGTGTGAACGCAAACTGA

qOAS2-R

AACCTTCAGCCGTGTCAAAG

qGAPDH-F

CTGCCGCCTGGAGAAACCT

qGAPDH-R

GCTGTAGCCAAATTCATTGTCG

RI PT

OAS2- F

qIRF3-F

CCACCAAACUGAAGGAUUU AAAUCCUUCAGUUUGGUGG GCCCUUAACCAAGCAGGUUTT AACCUGCUUGGUUAAGGGCTT UGGAAGAGAUGAAUGCAUA UAUGCAUUCAUCUCUUCCA AAGGTTGTCCCCATGTGTCTCCG

qIRF3-R

GGAAATGTGCAGGTCCACCGTG

qIRF7-F

TCCAGCCGAGATGCTAAGTG

qIRF7-R

GTCCAAGTCCTGCCCGATGT

qORF7-F

AAACCAGTCCAGAGGCAAGG

qORF7-R

GCAAACTAAACTCCACAGTGTAA

qIFN-beta -F

CTAGCACTGGCTGGAATGAGACT

Si-OAS2

SC

Si-RIG-1

qIFN-beta -R

GGCCTTCAGGTAATGCAGAATC

qTNF-alpha-F

qIL-8-R

ACGGGCTTATCTGAGGTTTGAG

GGCAGTTTTCCTGCTTTCT CAGTGGGGTCCACTCTCAAT

CAGAGCAGCGGCGGAATC

AC C

qRIG-1-F

CACCACGCTCTTCTGCCTAC

EP

qTNF-alpha-R qIL-8-F

TE D

M AN U

Si-RNase L

qRIG-1-R

ACTCAAGGTTGCCCAT

qTLR7-F

GAACTGTTTC TCTACAACA

qTLR7-R

AGACTTGTAATTCTGTCA

qTLR3-F

TACTGTACAC AACTTCTACC

qTLR3-R

TTAAATCCTCCATCCAAGG

qNF-κB-F

CCAGCACCTCCACTCCATTC

qNF-κB-R

ACATCAGCACCCAAAGACACC

qMDA5-F

CGAATTAACAGGCACCGATT

qMDA5-R

CGTCCAGACTTGGCTGATCT

qMyD88-F

CTCCGGAGCG GAGTCCGCG

ACCEPTED MANUSCRIPT GCCAGCCCAGTCCAGTCC

qTBK1-F

CCAGTGGAT GTTCAAAT

qTBK1-R

CTCCCACATGGACAAAAT

AC C

EP

TE D

M AN U

SC

RI PT

qMyD88-R

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

Highlights We firstly found that porcine 2’, 5’-oligoadenylate synthetase (OAS2) could inhibit the PRRSV replication, and the inhibition was dependent by RNase L. Porcine OAS2 suppressed replication of PRRSV in a RIG-I-dependent manner and enhanced RIG-I-mediated IFN induction. Co-silencing of RNase L and OAS2 revealed that the anti-PRRSV function of porcine OAS2 was RNase L dependent. Our findings show a mechanism by which porcine OAS2 contributes to host anti-PRRSV responses by enhancing RIG-I activation.