Skip to main content
Erschienen in: Virology Journal 1/2017

Open Access 01.12.2017 | Research

ORF1a of highly pathogenic PRRS attenuated vaccine virus plays a key role in neutralizing antibody induction in piglets and virus neutralization in vitro

verfasst von: Chaoliang Leng, Wuchao Zhang, Hongliang Zhang, Yunchao Kan, Lunguang Yao, Hongyue Zhai, Mingliang Li, Zhen Li, Chunxiao Liu, Tongqing An, Jinmei Peng, Qian Wang, Yumin Leng, Xuehui Cai, Zhijun Tian, Guangzhi Tong

Erschienen in: Virology Journal | Ausgabe 1/2017

Abstract

Background

Currently, porcine reproductive and respiratory syndrome virus (PRRSV) is one of the most economically important viral pathogens in swine in most countries, especially China. Two PRRSV attenuated live vaccine strains (HuN4-F112 and CH-1R) are currently widely used in China. Our previous study showed that HuN4-F112, but not CH-1R, induced high anti-nucleocapsid (N) antibody and neutralizing antibody (NA) titers. Additionally, sera from HuN4-F112 inoculated pigs induced low cross neutralization of CH-1R.

Methods

In the present study, 6 chimeric viruses through exchanging 5′ untranslated region (UTR) + open reading frame (ORF)1a, ORF1b, and ORF2–7 + 3’UTR between HuN4-F112 and CH-1R were constructed and rescued based on the infectious clones of rHuN4-F112 and rCH-1R. The characteristics of these viruses were investigated in vitro and vivo.

Results

All the three fragments, 5’UTR + ORF1a, ORF1b, and ORF2–7 + 3’UTR, could affect the replication efficiencies of rHuN4-F112 and rCH-1R in vitro. Additionally, both 5’UTR + ORF1a and ORF2–7 + 3’UTR affected the anti-N antibody and NA responses targeting rHuN4-F112 and rCH-1R in piglets.

Conclusions

The 5’UTR + ORF1a region of HuN4-F112 played a key role in inducing NAs in piglets. Furthermore, we confirmed for the first time that ORF1a contains a neutralization region. This study provides important information that can be used for further study of the generation of anti-PRRSV NAs.
Abkürzungen
CPE
Cytopathic effects
DMEM
Dulbecco’s modified Eagle’s medium
EU-type
European type
FBS
Fetal bovine serum
h
hours
HP-PRRSV
Highly pathogenic porcine reproductive and respiratory syndrome virus
IFA
Indirect immunofluorescence assay
LV
Lelystad virus
MOI
Multiplicity of infection
NA
Neutralizing antibody
NA-type
North American type
NSP
Non-structural protein
ORF
Open reading frame
PRRS
Porcine reproductive and respiratory syndrome
PRRSV
Porcine reproductive and respiratory syndrome virus
SPF
Specific pathogen-free
TCID50/mL
50% tissue culture infective doses per mL
UTR
Untranslated region

Background

Currently, porcine reproductive and respiratory syndrome (PRRS) is one of the most economically important diseases affecting the swine industry worldwide [1, 2]. The causative agent of this disease is the PRRS virus (PRRSV), which causes reproductive failure in sows and respiratory disease in pigs of all ages. This virus emerged in North America and central Europe in the late 1980s [35].
PRRSV, belonging to the family Arteriviridae in the order Nidovirales, is an enveloped single-stranded positive-sense RNA virus [6, 7]. Its genome is approximately 15 kb and consists of a 5’untranslated region (UTR), at least ten overlapping open reading frames (ORFs), and a 3’UTR [8]. Once the genome enters the cytoplasm, ORF1a and ORF1b are translated to produce two large polyproteins (pp1a and pp1ab) [9]. pp1ab expression is controlled by a − 1 ribosomal frameshift [10]. Autocatalytic processing of these precursors generates at least 14 non-structural proteins (NSPs) that are involved in virus replication and transcription [11, 12]. ORF1a encodes 10 NSPs (NSP1α, NSP1β, NSP2–6, NSP7α, NSP7β and NSP8), and ORF1b encodes 4 NSPs (NSP9–12) [1315]. Recently, an additional viral protein synthesized by a − 2 ribosomal frame shift in the NSP2 coding region has been described [16]. ORF2a, ORF2b and ORFs 3–7 encode the viral structural proteins GP2, E, GP3, GP4, GP5, M and N, respectively [8]. Additionally, a novel ORF overlapping the GP5 coding sequence has been discovered [17, 18].
PRRSV is divided into two major genotypes as follows: genotype 1 (European type, EU-type) and genotype 2 (North American type, NA-type) [19]. The representative strains of the two genotypes are Lelystad virus (LV) and VR-2332, respectively. These viruses share only approximately 70% nucleotide sequence identity [3, 4]. In 2006, an unparalleled, large-scale, atypical PRRS outbreak caused by a highly pathogenic PRRSV (HP-PRRSV) was reported in China [20, 21]. Currently, HP-PRRSV is recognized as the dominant virus in China and southeast Asian countries [22]. Phylogenetic analyses showed that both genotype 1 and 2 PRRSVs could be divided into three subtypes [23, 24]. Additionally, there have been frequent reports of a variety of atypical PRRSV strains in China recently [2426].
To control this severe disease, many PRRS attenuated live vaccines have been prepared. Among these vaccines, HuN4-F112 and CH-1R have been widely used in China. These strains were obtained by culture of the parental strains HP-PRRSV HuN4 and classical PRRSV CH-1a and passaged on MARC-145 cells for 112 and 165 passages, respectively [27, 28]. Our laboratory found that HuN4-F112 but not CH-1R induced high neutralizing antibody (NA) and N antibody titers in piglets [29]. Currently, the generation of anti-PRRSV NAs is poorly understood. A characteristic feature of PRRSV infection is that anti-PRRSV NAs appear late after infection, whereas natural infection or vaccination induces only low NA levels [7, 30]. Additionally, despite the attempts of many laboratories, a neutralizing monoclonal antibody against PRRSV has not been developed. Furthermore, our previous study found that the B epitope, which is the major neutralizing epitope of classical PRRSV, was not a neutralizing epitope in HP-PRRSV [29, 31]. Thus, exploration of the generation of anti-PRRSV NAs and the identification of the protective neutralizing antigenic region of PRRSV for potential use for the development of a vaccine are very useful and encouraging approaches.
In this study, we used the infectious clones (rHuN4-F112 and rCH-1R) of attenuated HP-PRRSV vaccine strain HuN4-F112 and classical PRRSV vaccine strain CH-1R as backbones and constructed a series of chimeric clones by individually exchanging the corresponding regions within the genome between the two parental clones. Then, we rescued the viruses and analyzed the growth kinetics, the ability to induce antibodies and NAs, and the viremia in piglets. Additionally, significant differences in cross NA titers of anti-rHuN4-F112 sera against the different rescued viruses were analyzed.

Methods

Viruses and cells

The attenuated live HP-PRRSV vaccine strain HuN4-F112 and classical PRRSV vaccine strain CH-1R (GenBank accession: EU807840) were obtained by culturing the parent strain HP-PRRSV HuN4 (GenBank accession: EF635006) and the classical strain PRRSV CH-1a (GenBank accession: AY032626) on MARC-145 cells [27, 28]. BHK-21 cells were used to rescue the virus by transfection within vitro transcribed RNA. MARC-145 cells were used for virus rescue and the subsequent experiments. The two cell lines were cultured in Dulbecco’s modified Eagle’s medium (DMEM) (Invitrogen) supplemented with 10% fetal bovine serum (FBS) (HyClone) at 37 °C in a humid atmosphere with 5% CO2.

Construction of PRRSV chimeric full-length cDNA clones

The PRRSV full-length infectious cDNA clone plasmids pHuN4-F112 and pCH-1R were previously constructed and identified by our laboratory [32, 33]. The strategy used for the construction of PRRSV chimeric full-length cDNA clones is illustrated in Fig. 1. The 5’UTR + ORF1a, ORF1b, or ORF2–7 + 3’UTR region was swapped between pHuN4-F112 and pCH-1R using the unique restriction enzymes (New England Biolabs) PacI and NheI, NheI and AscI, or AscI and NotI, respectively. The corresponding fragments were connected using the T4 DNA ligase (TaKaRa). The chimeric full-length cDNA clones with the pHuN4-F112 backbone were designated pHuN4-F112-C1a, pHuN4-F112-C1b and pHuN4-F112-C27. Correspondingly, the chimeric clones with the pCH-1R backbone were named pCH-1R-H1a, pCH-1R-H1b and pCH-1R-H27.

Recovery of the chimeric and parental viruses

The protocol for the rescue of the chimeric and parental viruses was performed as previously described [32]. First, each full-length cDNA clone plasmid was separately linearized using the restriction enzyme NotI (New England Biolabs). The linearized plasmid DNA was transcribed and capped using the mMessage High Yield Capped RNA Transcription kit (Ambion) according to the manufacturer’s instructions. The synthetic RNA was transfected into BHK-21 cells using the DMRIE-C reagent (Invitrogen) according to the manufacturer’s protocol. Finally, the supernatants were harvested 24 h post-transfection and serially passaged on MARC-145 cells.

Identification of the rescued viruses

The rescued viruses were examined by cytopathic effects (CPE) and indirect immunofluorescence assay (IFA). Briefly, the rescued virus suspensions were inoculated onto MARC-145 cell monolayers prepared in 6 well plates for 24 h (h) earlier. And then the cells were incubated at 37 °C and monitored daily for CPE. The IFA protocol was described previously [34]. Antigens of each rescued virus were prepared by inoculating MARC-145 cell monolayers in alternate rows of 96-well plates with 100 μL/well of the virus at the titer of 104 50% tissue culture infective doses per mL (TCID50/mL) and fixing infected cells with cold acetone-ethanol (8:2) mixture after a 20 h incubation at 37 °C. Uninfected cell monolayers used as cell control antigens were prepared in the identical manner using virus-free cell culture medium. All plates were dried and stored at −20 °C until use. The presence of PRRS viral antigen in each plate was confirmed by immunofluorescence microscopy using PRRSV specific monoclonal antibody 1C8 [35] and goat anti-mouse IgG antibody conjugated with FITC (Sigma) as the primary and second antibodies, respectively.
To further confirm the rescued PRRSVs, the complete genomes of the second-passage viruses were sequenced. The RT-PCR primers and protocols were previously described [24]. Briefly, the genomic RNAs were extracted using an RNA extraction QIAamp viral RNA Mini Kit (Qiagen) and RT-PCR was carried out with the One Step RT-PCR kit (Qiagen) according to the manufacturer’s instructions. The amplified PCR products were subjected to agarose gel electrophoresis and the target fragments were excised from the gels for purification at a later stage, which was performed using a Gel Extraction Kit (OMEGA, USA). The purified PCR products were cloned into a pMD18-T vector (TaKaRa, Japan). Recombinant clones were sent to Life Technologies (Shanghai, China) for sequencing. To further assess the replication stability, the rescued viruses were serially passaged on MARC-145 cells for 10 generations. And the complete genomes of the rescued PRRSVs of generations 5 and 10 were also sequenced.

Growth kinetics of the rescued viruses

To analyze the growth kinetics in vitro, MARC-145 cell monolayers in 6-well plates were individually infected with each chimeric virus and their parental viruses at the same multiplicity of infection (MOI) of 0.1. The viral titers in the supernatants at different time points were determined using the Reed-Mench method [36] and expressed as TCID50/mL. Briefly, MARC-145 cell monolayers were prepared in 96-well plates and inoculated with virus suspensions (100 μL/well) prepared by serial 10-fold dilution. Each diluted sample was run in eight parallel repeats in 96-well plates. And the plates were incubated for an additional 72 to 96 h. The viral titers were determined by the presence of visible CPE. The experiment was independently repeated three times and each time point was also independently repeated three times. The results represent the average of the duplicates.

Animal trials of the rescued viruses

Thirty-three 28-day-old PRRSV-free Bama piglets were obtained from the Laboratory Animal Center for specific pathogen-free (SPF) Swine Breeding and Management of the Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences. The piglets were randomly divided into nine groups (n = 3, 4, or 6). Each piglet in each experimental group was immunized intramuscularly with 2 mL of each virus containing 106.5 TCID50 (rHuN4-F112, rHuN4-F112-C1a, rHuN4-F112-C1b, rHuN4-F112-C27, rCH-1R, rCH-1R-H1a, rCH-1R-H1b, or rCH-1R-H27). Each piglet in the control group was mock-inoculated with the same dose of MARC-145 cell culture supernatant. Blood was collected from each animal at 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 and 11 weeks post-inoculation, and the sera were individually prepared and stored at −80 °C.

Serological analysis

The PRRSV-specific antibody levels in the sera were determined using a commercial IDEXX PRRS X3 ELISA Kit (IDEXX Laboratories, Westbrook, ME, USA). According to the manufacturer’s directions, samples with sample value/positive value (S/P) ratios ≥0.4 were considered positive for antibodies against PRRSV. To estimate the ability of the different rescued viruses in antibody induction in piglets, the S/P values of the serum samples collected from the piglets in the same group in different time points were accumulated and then the accumulated values were divided by the number of piglets in the group to obtain a value at last. The values can reflect the ability of the rescued viruses to induce antibodies in piglets to some extent.

Neutralization analysis

The sera neutralization assay was performed as previously described [29]. First, all tested sera were heat inactivated for 30 min at 56 °C prior to testing. Each serum sample was diluted using a two-fold serial dilution technique in DMEM. Then, 100 μL of each diluted sample was mixed with an equal volume of each virus (103 TCID50/mL). Finally, the mixtures were incubated for 1 h at 37 °C and inoculated onto MARC-145 cell monolayers prepared in 96-well plates 24 h earlier. Each diluted sample was run in four parallel repeats in 96-well plates. Thereafter, the cells were incubated at 37 °C and monitored daily for CPE. The presence of virus-specific CPE in each well was recorded after 5 days of incubation. The NA titer or cross NA titer of each serum sample against the different rescued PRRSVs was calculated using the Reed-Muench method [36]. The neutralization tests of each serum sample were repeated three times independently. The results represent the average of the duplicates. Similar to the previous description, to estimate the ability of the different rescued viruses in NA induction in piglets, the NA titers of the serum samples collected from the piglets in the same group in different time points were also accumulated and the accumulated values were also divided by the number of piglets in the group to obtain a value at last. And the values can also reflect the ability of the rescued viruses to induce NAs in piglets to some extent.

Viremia analysis

The viremia analysis was conducted using a virus isolation assay as previously described [24]. Briefly, the sera were diluted 10-fold with DMEM and transferred to MARC-145 cell monolayers prepared in 96-well plates 24 h earlier. Then, the cells were incubated at 37 °C for 3–5 days and monitored daily for CPE. All of the samples were tested three times independently.

Statistical analysis

The Student’s t-test was used to estimate the differences among the growth kinetics of the different rescued viruses, anti-N protein antibody and NA levels of the different rescued virus inoculated groups and cross NA titers of the anti-rHuN4-F112 sera against the different rescued viruses. Differences were considered significant at a P value <0.05 and extremely significant at values of P < 0.01 and P < 0.001.

Results

Recovery of the chimeric and parental viruses

The two rescued parental viruses were named rHuN4-F112 and rCH-1R (Fig. 1). Six chimeric viruses were successfully rescued from the chimeric infectious clones constructed by swapping the 5’UTR + ORF1a, ORF1b or ORF2–7 + 3’UTR regions between the pHuN4-F112 and pCH-1R plasmids and were individually designated rHuN4-F112-C1a, rHuN4-F112-C1b, rHuN4-F112-C27, rCH-1R-H1a, rCH-1R-H1b and rCH-1R-H27 (Fig. 1).
The MARC-145 cells infected with each rescued virus were positive for PRRSV based on CPE (Fig. 2) and the IFA results (Fig. 3). Furthermore, the sequencing results confirmed that the replaced regions and their flanking areas in the second-passage rescued viruses were consistent with the original design and that no additional mutations were introduced during construction (Fig. 4). No genetic variability was observed between the fifth and tenth passages of the rescued viruses (Fig. 4).

Growth kinetics of the rescued viruses

The growth kinetics of the chimeric viruses in parallel with their parental viruses (rHuN4-F112 and rCH-1R) were evaluated by infecting MARC-145 cells. rHuN4-F112-C1a had higher growth kinetics than its parental backbone virus rHuN4-F112 at 48 h, 60 h and 72 h (Fig. 5a). Interestingly, rHuN4-F112-C1b showed a reduced replication efficiency compared with the backbone virus rHuN4-F112 at 12 h and 36 h and an increased replication efficiency at 60 h and 72 h; conversely, rHuN4-F112-C27 exhibited an increased replication efficiency at 24 h and a reduced replication efficiency at 48 h and 96 h (Fig. 5a). Compared with the backbone virus rCH-1R, rCH-1R-H1a showed an increased replication efficiency at most time points, whereas rCH-1R-H1b and rCH-1R-H27 had reduced replication efficiencies at the same time points (Fig. 5b). These results indicated that exchanging the 5’UTR + ORF1a, ORF1b or ORF2–7 + 3’UTR affected the replication efficiencies of rHuN4-F112 and rCH-1R.

Antibody kinetics in the sera of piglets inoculated with the rescued viruses

Specific antibodies against the PRRSV N protein in the sera of the inoculated piglets were measured using a commercial PRRS X3 IDEXX ELISA Kit (Fig. 6). All of the piglets in the rHuN4-F112- and rHuN4-F112-C1b-inoculated groups seroconverted, whereas only half of the piglets in the rHuN4-F112-C1a- or rHuN4-F112-C27-inoculated groups had seroconverted at 2 weeks post-inoculation (Fig. 6a-6d). Conversely, the piglets in the rCH-1R-inoculated group remained seronegative throughout experiment. The piglets in the rCH-1R-H1a-, rCH-1R-H1b- and rCH-1R-H27-inoculated groups seroconverted at 4 weeks post-inoculation, and the rCH-1R-H1b- and rCH-1R-H27-inoculated groups each had one or two piglets that remained seronegative until the end of the experiment (Fig. 6e-6h). The control group remained seronegative for the duration of the experiment (Fig. 6i).

NA kinetics in the sera of piglets inoculated with the rescued viruses

The anti-PRRSV NAs in the sera of the inoculated piglets were measured using the neutralization test. As shown in Fig. 7, NAs were generated as soon as 3 weeks post-inoculation in the rHuN4-F112- and rHuN4-F112-C1b-inoculated groups, and the highest NA titer was greater than 45 (Fig. 7a). In contrast, the NAs of the rHuN4-F112-C1a-inoculated group were generated 9 weeks post-inoculation, and the highest NA titer was less than 10 (Fig. 7b). Additionally, the NAs of the rHuN4-F112-C27-inoculated group were generated 4 weeks post-inoculation, and the highest NA titer was approximately 10 (Fig. 7d). Throughout the experiment, no piglets in the rCH-1R-, rCH-1R-H1b- and rCH-1R-H27-inoculated groups produced NAs (Fig. 7e, g, h). However, one piglet in the rCH-1R-H1a-inoculated group generated NAs at 7 weeks post-inoculation (Fig. 7f). The control group piglets produced no NAs against rHuN4-F112 or rCH-1R during the experiment (Fig. 7i).

Ability of the rescued viruses in antibody and NA induction in piglets

The ability of the rescued viruses in anti-N antibody and NA induction in piglets were analyzed (Fig. 8). Results indicated that the ability of the rHuN4-F112 in anti-N antibody or NA induction in piglets was significantly better than that of rHuN4-F112-C1a or rHuN4-F112-C27, whereas the difference between rHuN4-F112 and rHuN4-F112-C1b was not obvious (Fig. 8). Correspondingly, we found that the ability of the rCH-1R in anti-N antibody induction in piglets was significantly worse than that of rCH-1R-H1a, whereas the difference between rCH-1R and rCH-1R-H1b or rCH-1R-H27 was not obvious (Fig. 8a). However, there was no significant difference of the ability in NA induction between rCH-1R and rCH-1R-1a, rCH-1R-1b or rCH-1R-27 (Fig. 8b).

Cross NA titers of the rHuN4-F112-inoculated piglet sera against the different rescued viruses

The NA titers or cross NA titers of the rHuN4-F112-inoculated piglet sera (#11, #12, #13, #14, #15 and #16) against rHuN4-F112, rHuN4-F112-C1a, rHuN4-F112-C1b, rHuN4-F112-C27, rCH-1R, rCH-1R-H1a, rCH-1R-H1b and rCH-1R-H27 were measured using the neutralization test (Fig. 9). All six serum samples were collected 9 weeks post-inoculation with rHuN4-F112. The results indicated that there was no significant difference between the NA titers against rHuN4-F112 and rHuN4-F112-C1b in the rHuN4-F112-inoculated piglet sera. However, the NA titers against rHuN4-F112 were significantly higher than the NA titers against rHuN4-F112-C1a, rHuN4-F112-C27 and rCH-1R-H27 in the rHuN4-F112-inoculated piglet sera. In contrast, the NA titers against rCH-1R were significantly lower than the NA titers against rCH-1R-H27 and rHuN4-F112-C27 in the rHuN4-F112-inoculated piglet sera. Moreover, no significant difference was found between the NA titers against rCH-1R and rCH-1R-H1a in the rHuN4-F112-inoculated piglet sera.

Viremia in the piglets inoculated with the rescued viruses

The viremia of the piglets inoculated with the rescued viruses was examined using the virus isolation assay (Table 1). For the rHuN4-F112-inoculated group, virus was isolated from all serum samples in the first two weeks post-inoculation and half of the serum samples in the third week post-inoculation. However, for the rHuN4-F112-C1a-, rHuN4-F112-C1b-, and rHuN4-F112-C27-inoculated groups, the virus was isolated from only a few serum samples. In contrast, the virus was not isolated in any serum sample from the rCH-1R-inoculated group. However, virus was isolated from three serum samples and one serum sample from the rCH-1R-H1a-inoculated and rCH-1R-H1b-inoculated groups, respectively. Additionally, virus was not isolated from any of the serum samples from the rCH-1R-H27-inoculated and control groups.
Table 1
Virus isolation results from the serum samples of piglets inoculated with the rescued viruses
Groups
Pig no.
Weeks post-inoculation
0
1
2
3
4
5
6
7
8
9
10
rHuN4-F112 inoculated group
#11
-a
+b
+
+
#12
+
+
#13
+
+
+
#14
+
+
#15
+
+
#16
+
+
+
rHuN4-F112-C1a inoculated group
#21
+
#22
#23
#24
+
rHuN4-F112-C1b inoculated group
#31
+
#32
#33
rHuN4-F112-C27 inoculated group
#41
#42
+
#43
#44
rCH-1R inoculated group
#51
#52
#53
rCH-1R-H1a inoculated group
#61
+
+
+
#62
#63
rCH-1R-H1b inoculated group
#71
#72
+
#73
rCH-1R-H27 inoculated group
#81
#82
#83
Control group
#91
#92
#93
#94
a “-” indicates that the virus was not isolated in the serum sample
b “+” indicates that the virus was isolated in the serum sample

Discussion

PRRSV continues to be one of the leading swine pathogens and causes great economic losses for the swine industry worldwide [1, 2, 24]. Our previous study found that the HP-PRRSV vaccine strain HuN4-F112 induced significantly higher NA titers than the classical PRRSV vaccine strain CH-1R [29]. That study confirmed that the B epitope, which is generally believed to be the major neutralizing epitope of PRRSV, was not a neutralizing epitope in HP-PRRSV [29]. Based on the significant differences in the NA titers induced by HuN4-F112 and CH-1R, we explored the generation of anti-PRRSV NAs and tried to determine the region(s) of the virus genome that contributed to neutralization of HP-PRRSV.
In the present study, large fragments of the genome were initially swapped between rHuN4-F112 and rCH-1R using a reverse genetic procedure to analyze the possible contributor to neutralization. The six chimeric viruses (rHuN4-F112-C1a, rHuN4-F112-C1b, rHuN4-F112-C27, rCH-1R-H1a, rCH-1R-H1b and rCH-1R-H27) were individually constructed, rescued and identified (Fig.1-4). The growth kinetics of the chimeric viruses in parallel with of the growth kinetics of their parental viruses (rHuN4-F112 or rCH-1R) were evaluated by infecting MARC-145 cells. Exchanging any one of the 5’UTR + ORF1a, ORF1b or ORF2–7 + 3’UTR regions could affect the replication efficiencies of rHuN4-F112 and rCH-1R (Fig. 5).
The anti-N protein antibody levels of the eight rescued PRRSVs used to infect piglets were determined by ELISA (Fig. 6). The data indicated that the seroconversion times of the rHuN4-F112- and rHuN4-F112-C1b-inoculated groups were shorter than the seroconversion times of the rHuN4-F112-C1a- and rHuN4-F112-C27-inoculated groups, although the latter two groups also produced high levels of antibodies (S/P ≥ 2.0) (Fig. 6). The replacement of 5’UTR + ORF1a or ORF2–7 + 3’UTR in rHuN4-F112 with the corresponding region from rCH-1R greatly decreased the anti-N protein antibody responses in the rHuN4-F112-infected piglets, whereas no difference was observed in the anti-N protein antibody responses following replacement of ORF1b of rHuN4-F112 with the corresponding region from rCH-1R (Fig. 8a). Additionally, most piglets in the rCH-1R-H1a-, rCH-1R-H1b- and rCH-1R-H27-inoculated groups seroconverted at 4 weeks post-inoculation, whereas the rCH-1R-inoculated groups remained seronegative for the duration of the experiment (Fig. 6). The replacement of 5’UTR + ORF1a, ORF1b and ORF2–7 + 3’UTR in rCH-1R with the corresponding regions in rHuN4-F112 increased the anti-N protein antibody responses (Fig. 6). However, we can find the significant difference in anti-N antibody responses between rCH-1R and rCH-1R-H1a, whereas there was no significant difference between rCH-1R and rCH-1R-H1b or rCH-1R-H27 (Fig. 8a).
The NA titers of the eight rescued PRRSVs used to infect piglets were also determined by the virus neutralization test (Fig. 7). The results were similar to the production of anti-N protein antibodies described above. The appearance times of the NAs were shorter in the rHuN4-F112- and rHuN4-F112-C1b-inoculated groups than in the rHuN4-F112-C1a- and rHuN4-F112-C27-inoculated groups (Fig. 7). Furthermore, the NA titers were higher in the rHuN4-F112- and rHuN4-F112-C1b-inoculated groups than in the rHuN4-F112-C1a- and rHuN4-F112-C27-inoculated groups (Fig. 7). Further analysis showed that the ability of the rHuN4-F112 in NA induction in piglets was significantly better than that of rHuN4-F112-C1a or rHuN4-F112-C27, whereas the difference between rHuN4-F112 and rHuN4-F112-C1b was not obvious (Fig. 8b). Thus, replacement of the 5’UTR + ORF1a or ORF2–7 + 3’UTR of rHuN4-F112 with the corresponding region in rCH-1R greatly delayed the appearance of NAs and decreased the NA titers in the rHuN4-F112-infected piglets, whereas no difference was observed in the NA responses when ORF1b of rHuN4-F112 was replaced with the corresponding region from rCH-1R. Additionally, no piglets produced NAs in the rCH-1R-, rCH-1R-H1b- and rCH-1R-H27-inoculated groups, and one piglet in the rCH-1R-H1a-inoculated group generated NAs at 7 weeks post-inoculation (Fig. 7). These data indicated that replacement of 5’UTR + ORF1a in rCH-1R with the corresponding region from rHuN4-F112 increased the NA responses. However, the differences of the ability in NA induction between rCH-1R and rCH-1R-1a, rCH-1R-1b or rCH-1R-27 was not obvious (Fig. 8b).
Based on the above results, we found that the 5’UTR + ORF1a and ORF2–7 + 3’UTR regions of PRRSV was closely related to the production of anti-N protein antibodies and NAs. As we all know, the 5′ and 3’UTRs of PRRSV may be involved in replicative and translational functionality, although the exact functions are poorly understood [37]. The NSPs encoded by ORF1a assemble into a membrane-associated enzyme complex that directs viral replication and transcription; however, the internal functions and associated mechanisms of this interaction are also unknown [38]. In addition, the nucleocapsid N protein of PRRSV builds the scaffold for the genomic RNA, and several membrane proteins are incorporated into the viral envelope [39]. GP2/3/4 forms a disulfide-linked heterotrimeric complex in virus particles that is required for virus entry and governs cell tropism [39, 40]. GP5 is disulfide-linked to M, and the heterodimer is required for virus budding [40]. In the present study, the replacements of the 5’UTR + ORF1a or ORF2–7 + 3’UTR region between rHuN4-F112 and rCH-1R may change the structure or function of these regions, alter the characteristics of the viruses further, and affect the replication and antibody responses of the viruses in piglets at last.
PRRSV NAs are directed against the surface proteins, including GP2, GP3, GP4, GP5 and M [4147]. GP5 was favored as the most relevant protein [30, 31, 4851]. In the present study, we found that the NA titers in the rHuN4-F112-inoculated piglet sera against rHuN4-F112-C27 were significantly lower than the NA titers against rHuN4-F112 itself (Fig. 9). Correspondingly, the NA titers from rHuN4-F112-inoculated piglet sera against rCH-1R-H27 were significantly higher than the NA titers against rCH-1R (Fig. 9). These results suggested that ORF2–7 was the neutralization region of PRRSV, which is consistent with previous reports. Additionally, no significant difference was found between the NA titers against rHuN4-F112 and the NA titers against rHuN4-F112-C1b or between the NA titers against rCH-1R and the NA titers against rCH-1R-H1b in the sera from the rHuN4-F112-inoculated piglets (Fig. 9). This result indicated that ORF1b was not the neutralization region of PRRSV. Surprisingly, the NA titers from the rHuN4-F112-inoculated piglet sera against rHuN4-F112-C1a were significantly lower than the NA titers against rHuN4-F112 itself. Conversely, the NA titers of the rHuN4-F112-inoculated piglet sera against rHuN4-F112-C27 were significantly higher than the NA titers against rCH-1R (Fig. 9). These results indicated that ORF1a was also a neutralization region of PRRSV. Most researchers believe that the neutralization region of PRRSV is located in ORF2–6, and many genetically engineered vaccines have been designed based on this region [5256]. At present, these vaccines are still in the experimental stage, possibly because the neutralization region remains unclear. Recently, it was described that NSP2 was a virion-associated structural PRRSV protein that existed in or on viral particles in multiple isoforms [57]. Further study demonstrated that NSP2 integrated into membranes with an unexpected topology in which the amino (N)-terminal (cytoplasmic) and C-terminal (luminal) domains were orientated on opposite sides of the membrane surface [58]. Taken together, these reports support our results showing that the neutralization region of ORF1a may be located at the NSP2 region. Of course, this issue requires further study.
The virus isolation results showed that rHuN4-F112 induced viremia in the first three weeks, whereas rCH-1R did not induce viremia during the experiment. The chimeric viruses rHuN4-F112-C1a, rHuN4-F112-C1b, rHuN4-F112-C27, rCH-1R-H1a and rCH-1R-H1b induced viremia only during limited time points. Moreover, rCH-1R-H27 did not induce viremia during the experiment. These results indicated that exchanging 5’UTR + ORF1a, ORF1b or ORF2–7 + 3’UTR affected the replication efficiencies of rHuN4-F112 and rCH-1R in vivo. Generally, a viremia that is higher and has a longer duration indicates a stronger replication ability of PRRSV in piglets and thus results in higher antibody production. We found consistent results in the rHuN4-F112 and rCH-1R inoculated groups. However, we did not find complete consistency in the other chimeric virus inoculation groups (Figs. 6-7; Table 1). This discrepancy was very confusing, and the internal mechanism requires further study.
In the present study, we found that rCH-1R strain could not induce antibody response and viremia in piglets (Figs. 6-7; Table 1). It seems that CH-1R can not grow in pigs. However, if given more immunization dosages or vaccination times of CH-1R, the pigs may produce low levels of immune response and viremia. Recently, our colleagues reported that CH-1R can induce lower levels of immune responses and viremia compared to HuN4-F112 and supply partial protection to the heterologous PRRSV challenge [28]. To date, the protective mechanism of CH-1R vaccine is unclear and future investigation is needed.

Conclusion

The present study constructed and rescued 6 chimeric viruses by exchanging 5’UTR + ORF1a, ORF1b, and ORF2–7 + 3’UTR between rHuN4-F112 and rCH-1R. All of these fragments affected the replication efficiencies of rHuN4-F112 and rCH-1R in vitro. Additionally, both 5’UTR + ORF1a and ORF2–7 + 3’UTR affected the anti-N antibody and NA responses to rHuN4-F112 and rCH-1R in piglets. Furthermore, we confirmed for the first time that ORF1a contains a neutralization region. This study provides important information for further study of the generation of anti-PRRSV NAs.

Acknowledgements

Not applicable.

Funding

The National Natural Science Foundation of China (No. 31502097) and the Heilongjiang Natural Science Funds for Distinguished Young Scholars (No. JC201314) supported this study. YML would like to thank the financial support from the National Natural Science Foundation of China (No. 21607083) and the Natural Science Foundation of Henan (162300410206).

Availability of data and materials

All data generated or analyzed during this study are included in this published article.
The study was approved by the Animal Ethics Committee of School of Harbin Veterinary Research Institute of the Chinese Academy of Agricultural Sciences and performed in accordance with animal ethics guidelines and approved protocols. The Animal Ethics Committee approval number was SYXK (Hei) 2,011,022.
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Neumann EJ, Kliebenstein JB, Johnson CD, Mabry JW, Bush EJ, Seitzinger AH, et al. Assessment of the economic impact of porcine reproductive and respiratory syndrome on swine production in the United States. J Am Vet Med Assoc. 2005;227(3):385–92.CrossRefPubMed Neumann EJ, Kliebenstein JB, Johnson CD, Mabry JW, Bush EJ, Seitzinger AH, et al. Assessment of the economic impact of porcine reproductive and respiratory syndrome on swine production in the United States. J Am Vet Med Assoc. 2005;227(3):385–92.CrossRefPubMed
2.
Zurück zum Zitat Nieuwenhuis N, Duinhof TF, van Nes A. Economic analysis of outbreaks of porcine reproductive and respiratory syndrome virus in nine sow herds. Vet Rec. 2012;170(9):225.CrossRefPubMed Nieuwenhuis N, Duinhof TF, van Nes A. Economic analysis of outbreaks of porcine reproductive and respiratory syndrome virus in nine sow herds. Vet Rec. 2012;170(9):225.CrossRefPubMed
3.
Zurück zum Zitat Wensvoort G, Terpstra C, Pol JM, ter Laak EA, Bloemraad M, de Kluyver EP, et al. Mystery swine disease in The Netherlands: the isolation of Lelystad virus. Vet Q. 1991;13(3):121–30.CrossRefPubMed Wensvoort G, Terpstra C, Pol JM, ter Laak EA, Bloemraad M, de Kluyver EP, et al. Mystery swine disease in The Netherlands: the isolation of Lelystad virus. Vet Q. 1991;13(3):121–30.CrossRefPubMed
4.
Zurück zum Zitat Benfield DA, Nelson E, Collins JE, Harris L, Goyal SM, Robison D, et al. Characterization of swine infertility and respiratory syndrome (SIRS) virus (isolate ATCC VR-2332). J Vet Diagn Investig. 1992;4(2):127–33.CrossRef Benfield DA, Nelson E, Collins JE, Harris L, Goyal SM, Robison D, et al. Characterization of swine infertility and respiratory syndrome (SIRS) virus (isolate ATCC VR-2332). J Vet Diagn Investig. 1992;4(2):127–33.CrossRef
5.
Zurück zum Zitat Paton DJ, Brown IH, Edwards S, Wensvoort G. 'Blue ear' disease of pigs. Vet Rec. 1991;128(26):617. Paton DJ, Brown IH, Edwards S, Wensvoort G. 'Blue ear' disease of pigs. Vet Rec. 1991;128(26):617.
6.
Zurück zum Zitat Cavanagh D. Nidovirales: a new order comprising Coronaviridae and Arteriviridae. Arch Virol. 1997;142(3):629–33.PubMed Cavanagh D. Nidovirales: a new order comprising Coronaviridae and Arteriviridae. Arch Virol. 1997;142(3):629–33.PubMed
7.
Zurück zum Zitat Meulenberg JJ. PRRSV, the virus. Vet Res. 2000;31(1):11–21.PubMed Meulenberg JJ. PRRSV, the virus. Vet Res. 2000;31(1):11–21.PubMed
8.
Zurück zum Zitat Meulenberg JJ. Petersen den Besten A, de Kluyver E, van Nieuwstadt A, Wensvoort G, Moormann RJ. Molecular characterization of Lelystad virus. Vet Microbiol. 1997;55(1–4):197–202.CrossRefPubMed Meulenberg JJ. Petersen den Besten A, de Kluyver E, van Nieuwstadt A, Wensvoort G, Moormann RJ. Molecular characterization of Lelystad virus. Vet Microbiol. 1997;55(1–4):197–202.CrossRefPubMed
9.
Zurück zum Zitat Snijder EJ, Meulenberg JJ. The molecular biology of arteriviruses. J Gen Virol. 1998;79(Pt 5):961–79.CrossRefPubMed Snijder EJ, Meulenberg JJ. The molecular biology of arteriviruses. J Gen Virol. 1998;79(Pt 5):961–79.CrossRefPubMed
10.
Zurück zum Zitat Yun SI, Lee YM. Overview: replication of porcine reproductive and respiratory syndrome virus. J Microbiol. 2013;51(6):711–23.CrossRefPubMed Yun SI, Lee YM. Overview: replication of porcine reproductive and respiratory syndrome virus. J Microbiol. 2013;51(6):711–23.CrossRefPubMed
11.
Zurück zum Zitat Ziebuhr J, Snijder EJ, Gorbalenya AE. Virus-encoded proteinases and proteolytic processing in the Nidovirales. J Gen Virol. 2000;81(Pt 4):853–79.CrossRefPubMed Ziebuhr J, Snijder EJ, Gorbalenya AE. Virus-encoded proteinases and proteolytic processing in the Nidovirales. J Gen Virol. 2000;81(Pt 4):853–79.CrossRefPubMed
12.
Zurück zum Zitat Fang Y, Snijder EJ. The PRRSV replicase: exploring the multifunctionality of an intriguing set of nonstructural proteins. Virus Res. 2010;154(1–2):61–76.CrossRefPubMed Fang Y, Snijder EJ. The PRRSV replicase: exploring the multifunctionality of an intriguing set of nonstructural proteins. Virus Res. 2010;154(1–2):61–76.CrossRefPubMed
13.
Zurück zum Zitat Snijder EJ, Wassenaar AL, Spaan WJ. Proteolytic processing of the replicase ORF1a protein of equine arteritis virus. J Virol. 1994;68(9):5755–64.PubMedPubMedCentral Snijder EJ, Wassenaar AL, Spaan WJ. Proteolytic processing of the replicase ORF1a protein of equine arteritis virus. J Virol. 1994;68(9):5755–64.PubMedPubMedCentral
14.
Zurück zum Zitat Chen Z, Lawson S, Sun Z, Zhou X, Guan X, Christopher-Hennings J, et al. Identification of two auto-cleavage products of nonstructural protein 1 (nsp1) in porcine reproductive and respiratory syndrome virus infected cells: nsp1 function as interferon antagonist. Virology. 2010;398(1):87–97.CrossRefPubMed Chen Z, Lawson S, Sun Z, Zhou X, Guan X, Christopher-Hennings J, et al. Identification of two auto-cleavage products of nonstructural protein 1 (nsp1) in porcine reproductive and respiratory syndrome virus infected cells: nsp1 function as interferon antagonist. Virology. 2010;398(1):87–97.CrossRefPubMed
15.
Zurück zum Zitat Li Y, Tas A, Snijder EJ, Fang Y. Identification of porcine reproductive and respiratory syndrome virus ORF1a-encoded non-structural proteins in virus-infected cells. J Gen Virol. 2012;93(Pt 4):829–39.CrossRefPubMed Li Y, Tas A, Snijder EJ, Fang Y. Identification of porcine reproductive and respiratory syndrome virus ORF1a-encoded non-structural proteins in virus-infected cells. J Gen Virol. 2012;93(Pt 4):829–39.CrossRefPubMed
16.
Zurück zum Zitat Fang Y, Treffers EE, Li Y, Tas A, Sun Z, van der Meer Y, et al. Efficient −2 frameshifting by mammalian ribosomes to synthesize an additional arterivirus protein. Proc Natl Acad Sci U S A. 2012;109(43):E2920–8.CrossRefPubMedPubMedCentral Fang Y, Treffers EE, Li Y, Tas A, Sun Z, van der Meer Y, et al. Efficient −2 frameshifting by mammalian ribosomes to synthesize an additional arterivirus protein. Proc Natl Acad Sci U S A. 2012;109(43):E2920–8.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Firth AE, Zevenhoven-Dobbe JC, Wills NM, Go YY, Balasuriya UB, Atkins JF, et al. Discovery of a small arterivirus gene that overlaps the GP5 coding sequence and is important for virus production. J Gen Virol. 2011;92(Pt 5):1097–106.CrossRefPubMedPubMedCentral Firth AE, Zevenhoven-Dobbe JC, Wills NM, Go YY, Balasuriya UB, Atkins JF, et al. Discovery of a small arterivirus gene that overlaps the GP5 coding sequence and is important for virus production. J Gen Virol. 2011;92(Pt 5):1097–106.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Johnson CR, Griggs TF, Gnanandarajah J, Murtaugh MP. Novel structural protein in porcine reproductive and respiratory syndrome virus encoded by an alternative ORF5 present in all arteriviruses. J Gen Virol. 2011;92(Pt 5):1107–16.CrossRefPubMedPubMedCentral Johnson CR, Griggs TF, Gnanandarajah J, Murtaugh MP. Novel structural protein in porcine reproductive and respiratory syndrome virus encoded by an alternative ORF5 present in all arteriviruses. J Gen Virol. 2011;92(Pt 5):1107–16.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Murtaugh MP, Stadejek T, Abrahante JE, Lam TT, Leung FC. The ever-expanding diversity of porcine reproductive and respiratory syndrome virus. Virus Res. 2010;154(1–2):18–30.CrossRefPubMed Murtaugh MP, Stadejek T, Abrahante JE, Lam TT, Leung FC. The ever-expanding diversity of porcine reproductive and respiratory syndrome virus. Virus Res. 2010;154(1–2):18–30.CrossRefPubMed
20.
Zurück zum Zitat 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(6):e526.CrossRefPubMedPubMedCentral 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(6):e526.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Tong GZ, Zhou YJ, Hao XF, Tian ZJ, An TQ, Qiu HJ. Highly pathogenic porcine reproductive and respiratory syndrome. China Emerg Infect Dis. 2007;13(9):1434–6.CrossRefPubMed Tong GZ, Zhou YJ, Hao XF, Tian ZJ, An TQ, Qiu HJ. Highly pathogenic porcine reproductive and respiratory syndrome. China Emerg Infect Dis. 2007;13(9):1434–6.CrossRefPubMed
22.
Zurück zum Zitat An TQ, Tian ZJ, Leng CL, Peng JM, Tong GZ. Highly pathogenic porcine reproductive and respiratory syndrome virus. Asia Emerg Infect Dis. 2011;17(9):1782–4.CrossRefPubMed An TQ, Tian ZJ, Leng CL, Peng JM, Tong GZ. Highly pathogenic porcine reproductive and respiratory syndrome virus. Asia Emerg Infect Dis. 2011;17(9):1782–4.CrossRefPubMed
23.
Zurück zum Zitat Stadejek T, Stankevicius A, Murtaugh MP, Oleksiewicz MB. Molecular evolution of PRRSV in Europe: current state of play. Vet Microbiol. 2013;165(1–2):21–8.CrossRefPubMed Stadejek T, Stankevicius A, Murtaugh MP, Oleksiewicz MB. Molecular evolution of PRRSV in Europe: current state of play. Vet Microbiol. 2013;165(1–2):21–8.CrossRefPubMed
24.
Zurück zum Zitat Leng CL, Tian ZJ, Zhang WC, Zhang HL, Zhai HY, An TQ, et al. Characterization of two newly emerged isolates of porcine reproductive and respiratory syndrome virus from Northeast China in 2013. Vet Microbiol. 2014;171(1–2):41–52.CrossRefPubMed Leng CL, Tian ZJ, Zhang WC, Zhang HL, Zhai HY, An TQ, et al. Characterization of two newly emerged isolates of porcine reproductive and respiratory syndrome virus from Northeast China in 2013. Vet Microbiol. 2014;171(1–2):41–52.CrossRefPubMed
25.
Zurück zum Zitat Zhou L, Wang Z, Ding Y, Ge X, Guo X, Yang H. NADC30-like strain of porcine reproductive and respiratory syndrome virus. China. Emerg Infect Dis. 2015;21(12):2256–7.CrossRefPubMed Zhou L, Wang Z, Ding Y, Ge X, Guo X, Yang H. NADC30-like strain of porcine reproductive and respiratory syndrome virus. China. Emerg Infect Dis. 2015;21(12):2256–7.CrossRefPubMed
26.
Zurück zum Zitat Zhao K, Ye C, Chang XB, Jiang CG, Wang SJ, Cai XH, et al. Importation and recombination are responsible for the latest emergence of highly pathogenic porcine reproductive and respiratory syndrome virus in China. J Virol. 2015;89(20):10712–6.CrossRefPubMedPubMedCentral Zhao K, Ye C, Chang XB, Jiang CG, Wang SJ, Cai XH, et al. Importation and recombination are responsible for the latest emergence of highly pathogenic porcine reproductive and respiratory syndrome virus in China. J Virol. 2015;89(20):10712–6.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Tian ZJ, An TQ, Zhou YJ, Peng JM, Hu SP, Wei TC, et al. An attenuated live vaccine based on highly pathogenic porcine reproductive and respiratory syndrome virus (HP-PRRSV) protects piglets against HP-PRRS. Vet Microbiol. 2009;138(1–2):34–40.CrossRefPubMed Tian ZJ, An TQ, Zhou YJ, Peng JM, Hu SP, Wei TC, et al. An attenuated live vaccine based on highly pathogenic porcine reproductive and respiratory syndrome virus (HP-PRRSV) protects piglets against HP-PRRS. Vet Microbiol. 2009;138(1–2):34–40.CrossRefPubMed
28.
Zurück zum Zitat Wang G, Yu Y, Zhang C, Tu Y, Tong J, Liu Y, et al. Immune responses to modified live virus vaccines developed from classical or highly pathogenic PRRSV following challenge with a highly pathogenic PRRSV strain. Dev Comp Immunol. 2016;62:1–7.CrossRefPubMed Wang G, Yu Y, Zhang C, Tu Y, Tong J, Liu Y, et al. Immune responses to modified live virus vaccines developed from classical or highly pathogenic PRRSV following challenge with a highly pathogenic PRRSV strain. Dev Comp Immunol. 2016;62:1–7.CrossRefPubMed
29.
Zurück zum Zitat Leng CL, An TQ, Chen JZ, Gong DQ, Peng JM, Yang YQ, et al. Highly pathogenic porcine reproductive and respiratory syndrome virus GP5 B antigenic region is not a neutralizing antigenic region. Vet Microbiol. 2012;159(3–4):273–81.CrossRefPubMed Leng CL, An TQ, Chen JZ, Gong DQ, Peng JM, Yang YQ, et al. Highly pathogenic porcine reproductive and respiratory syndrome virus GP5 B antigenic region is not a neutralizing antigenic region. Vet Microbiol. 2012;159(3–4):273–81.CrossRefPubMed
30.
Zurück zum Zitat Gonin P, Pirzadeh B, Gagnon CA, Dea S. Seroneutralization of porcine reproductive and respiratory syndrome virus correlates with antibody response to the GP5 major envelope glycoprotein. J Vet Diagn Investig. 1999;11(1):20–6.CrossRef Gonin P, Pirzadeh B, Gagnon CA, Dea S. Seroneutralization of porcine reproductive and respiratory syndrome virus correlates with antibody response to the GP5 major envelope glycoprotein. J Vet Diagn Investig. 1999;11(1):20–6.CrossRef
31.
Zurück zum Zitat Ostrowski M, Galeota JA, Jar AM, Platt KB, Osorio FA, Lopez OJ. Identification of neutralizing and nonneutralizing epitopes in the porcine reproductive and respiratory syndrome virus GP5 ectodomain. J Virol. 2002;76(9):4241–50.CrossRefPubMedPubMedCentral Ostrowski M, Galeota JA, Jar AM, Platt KB, Osorio FA, Lopez OJ. Identification of neutralizing and nonneutralizing epitopes in the porcine reproductive and respiratory syndrome virus GP5 ectodomain. J Virol. 2002;76(9):4241–50.CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Zhang S, Zhou Y, Jiang Y, Li G, Yan L, Yu H, et al. Generation of an infectious clone of HuN4-F112, an attenuated live vaccine strain of porcine reproductive and respiratory syndrome virus. Virol J. 2011;8:410.CrossRefPubMedPubMedCentral Zhang S, Zhou Y, Jiang Y, Li G, Yan L, Yu H, et al. Generation of an infectious clone of HuN4-F112, an attenuated live vaccine strain of porcine reproductive and respiratory syndrome virus. Virol J. 2011;8:410.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Zhang WC, Leng CL, Zhang HL, Chen JZ, Bai Y, Zhang QY, et al. The construction and immunogenic analysis of chimeric virus between attenuated classical and highly pathogenic porcine reproductive and respiratory syndrome virus vaccine strains. Chin J Pre Vet Med. 2015;37(4):5. Zhang WC, Leng CL, Zhang HL, Chen JZ, Bai Y, Zhang QY, et al. The construction and immunogenic analysis of chimeric virus between attenuated classical and highly pathogenic porcine reproductive and respiratory syndrome virus vaccine strains. Chin J Pre Vet Med. 2015;37(4):5.
34.
Zurück zum Zitat Kim WI, Lee DS, Johnson W, Roof M, Cha SH, Yoon KJ. Effect of genotypic and biotypic differences among PRRS viruses on the serologic assessment of pigs for virus infection. Vet Microbiol. 2007;123(1–3):1–14.CrossRefPubMed Kim WI, Lee DS, Johnson W, Roof M, Cha SH, Yoon KJ. Effect of genotypic and biotypic differences among PRRS viruses on the serologic assessment of pigs for virus infection. Vet Microbiol. 2007;123(1–3):1–14.CrossRefPubMed
35.
Zurück zum Zitat Wang Q, Chen J, Peng J, An T, Leng C, Sun Y, et al. Characterisation of novel linear antigen epitopes on north American-type porcine reproductive and respiratory syndrome virus M protein. Arch Virol. 2014;159(11):3021–8.CrossRefPubMed Wang Q, Chen J, Peng J, An T, Leng C, Sun Y, et al. Characterisation of novel linear antigen epitopes on north American-type porcine reproductive and respiratory syndrome virus M protein. Arch Virol. 2014;159(11):3021–8.CrossRefPubMed
37.
Zurück zum Zitat Kappes MA, Faaberg KS. PRRSV structure, replication and recombination: Origin of phenotype and genotype diversity. Virology. 2015;479–480:475–486. Kappes MA, Faaberg KS. PRRSV structure, replication and recombination: Origin of phenotype and genotype diversity. Virology. 2015;479–480:475–486.
38.
Zurück zum Zitat Snijder EJ, Kikkert M, Fang Y. Arterivirus molecular biology and pathogenesis. J Gen Virol. 2013;94(Pt 10):2141–63.CrossRefPubMed Snijder EJ, Kikkert M, Fang Y. Arterivirus molecular biology and pathogenesis. J Gen Virol. 2013;94(Pt 10):2141–63.CrossRefPubMed
39.
Zurück zum Zitat Veit M, Matczuk AK, Sinhadri BC, Krause E, Thaa B. Membrane proteins of arterivirus particles: structure, topology, processing and function. Virus Res. 2014;194:16–36.CrossRefPubMed Veit M, Matczuk AK, Sinhadri BC, Krause E, Thaa B. Membrane proteins of arterivirus particles: structure, topology, processing and function. Virus Res. 2014;194:16–36.CrossRefPubMed
40.
Zurück zum Zitat Wissink EH, Kroese MV, van Wijk HA, Rijsewijk FA, Meulenberg JJ, Rottier PJ. Envelope protein requirements for the assembly of infectious virions of porcine reproductive and respiratory syndrome virus. J Virol. 2005;79(19):12495–506.CrossRefPubMedPubMedCentral Wissink EH, Kroese MV, van Wijk HA, Rijsewijk FA, Meulenberg JJ, Rottier PJ. Envelope protein requirements for the assembly of infectious virions of porcine reproductive and respiratory syndrome virus. J Virol. 2005;79(19):12495–506.CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Yang L, Frey ML, Yoon KJ, Zimmerman JJ, Platt KB. Categorization of north American porcine reproductive and respiratory syndrome viruses: epitopic profiles of the N, M, GP5 and GP3 proteins and susceptibility to neutralization. Arch Virol. 2000;145(8):1599–619.CrossRefPubMed Yang L, Frey ML, Yoon KJ, Zimmerman JJ, Platt KB. Categorization of north American porcine reproductive and respiratory syndrome viruses: epitopic profiles of the N, M, GP5 and GP3 proteins and susceptibility to neutralization. Arch Virol. 2000;145(8):1599–619.CrossRefPubMed
42.
Zurück zum Zitat Vu HL, Kwon B, Yoon KJ, Laegreid WW, Pattnaik AK, et al. Immune evasion of porcine reproductive and respiratory syndrome virus through glycan shielding involves both glycoprotein 5 as well as glycoprotein 3. J Virol. 2011;85(11):5555–64.CrossRefPubMedPubMedCentral Vu HL, Kwon B, Yoon KJ, Laegreid WW, Pattnaik AK, et al. Immune evasion of porcine reproductive and respiratory syndrome virus through glycan shielding involves both glycoprotein 5 as well as glycoprotein 3. J Virol. 2011;85(11):5555–64.CrossRefPubMedPubMedCentral
43.
Zurück zum Zitat Vanhee M, Van Breedam W, Costers S, Geldhof M, Noppe Y, Nauwynck H. Characterization of antigenic regions in the porcine reproductive and respiratory syndrome virus by the use of peptide-specific serum antibodies. Vaccine. 2011;29(29–30):4794–804.CrossRefPubMed Vanhee M, Van Breedam W, Costers S, Geldhof M, Noppe Y, Nauwynck H. Characterization of antigenic regions in the porcine reproductive and respiratory syndrome virus by the use of peptide-specific serum antibodies. Vaccine. 2011;29(29–30):4794–804.CrossRefPubMed
44.
Zurück zum Zitat Karuppannan AK, Qiang J, Chang CC, Kwang J. A novel baculovirus vector shows efficient gene delivery of modified porcine reproductive and respiratory syndrome virus antigens and elicits specific immune response. Vaccine. 2013;31(46):5471–8.CrossRefPubMed Karuppannan AK, Qiang J, Chang CC, Kwang J. A novel baculovirus vector shows efficient gene delivery of modified porcine reproductive and respiratory syndrome virus antigens and elicits specific immune response. Vaccine. 2013;31(46):5471–8.CrossRefPubMed
45.
Zurück zum Zitat Fan B, Liu X, Bai J, Zhang T, Zhang Q, Jiang P. Influence of the amino acid residues at 70 in M protein of porcine reproductive and respiratory syndrome virus on viral neutralization susceptibility to the serum antibody. Virol J. 2016;13:51.CrossRefPubMedPubMedCentral Fan B, Liu X, Bai J, Zhang T, Zhang Q, Jiang P. Influence of the amino acid residues at 70 in M protein of porcine reproductive and respiratory syndrome virus on viral neutralization susceptibility to the serum antibody. Virol J. 2016;13:51.CrossRefPubMedPubMedCentral
46.
Zurück zum Zitat Fan B, Liu X, Bai J, Zhang T, Zhang Q, Jiang P. The amino acid residues at 102 and 104 in GP5 of porcine reproductive and respiratory syndrome virus regulate viral neutralization susceptibility to the porcine serum neutralizing antibody. Virus Res. 2015;204:21–30.CrossRefPubMed Fan B, Liu X, Bai J, Zhang T, Zhang Q, Jiang P. The amino acid residues at 102 and 104 in GP5 of porcine reproductive and respiratory syndrome virus regulate viral neutralization susceptibility to the porcine serum neutralizing antibody. Virus Res. 2015;204:21–30.CrossRefPubMed
47.
Zurück zum Zitat Trible BR, Popescu LN, Monday N, Calvert JG, Rowland RR. A single amino acid deletion in the matrix protein of porcine reproductive and respiratory syndrome virus confers resistance to a polyclonal swine antibody with broadly neutralizing activity. J Virol. 2015;89(12):6515–20.CrossRefPubMedPubMedCentral Trible BR, Popescu LN, Monday N, Calvert JG, Rowland RR. A single amino acid deletion in the matrix protein of porcine reproductive and respiratory syndrome virus confers resistance to a polyclonal swine antibody with broadly neutralizing activity. J Virol. 2015;89(12):6515–20.CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat Lopez OJ, Osorio FA. Role of neutralizing antibodies in PRRSV protective immunity. Vet Immunol Immunopathol. 2004;102(3):155–63.CrossRefPubMed Lopez OJ, Osorio FA. Role of neutralizing antibodies in PRRSV protective immunity. Vet Immunol Immunopathol. 2004;102(3):155–63.CrossRefPubMed
49.
Zurück zum Zitat Weiland E, Wieczorek-Krohmer M, Kohl D, Conzelmann KK, Weiland F. 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;66(3):171–86.CrossRefPubMed Weiland E, Wieczorek-Krohmer M, Kohl D, Conzelmann KK, Weiland F. 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;66(3):171–86.CrossRefPubMed
50.
Zurück zum Zitat Plagemann PG, Rowland RR, Faaberg KS. The primary neutralization epitope of porcine respiratory and reproductive syndrome virus strain VR-2332 is located in the middle of the GP5 ectodomain. Arch Virol. 2002;147(12):2327–47.CrossRefPubMed Plagemann PG, Rowland RR, Faaberg KS. The primary neutralization epitope of porcine respiratory and reproductive syndrome virus strain VR-2332 is located in the middle of the GP5 ectodomain. Arch Virol. 2002;147(12):2327–47.CrossRefPubMed
51.
Zurück zum Zitat Lee JA, Kwon B, Osorio FA, Pattnaik AK, Lee NH, Lee SW, et al. Protective humoral immune response induced by an inactivated porcine reproductive and respiratory syndrome virus expressing the hypo-glycosylated glycoprotein 5. Vaccine. 2014;32(29):3617–22.CrossRefPubMedPubMedCentral Lee JA, Kwon B, Osorio FA, Pattnaik AK, Lee NH, Lee SW, et al. Protective humoral immune response induced by an inactivated porcine reproductive and respiratory syndrome virus expressing the hypo-glycosylated glycoprotein 5. Vaccine. 2014;32(29):3617–22.CrossRefPubMedPubMedCentral
52.
Zurück zum Zitat Renukaradhya GJ, Meng XJ, Calvert JG, Roof M, Lager KM. Live porcine reproductive and respiratory syndrome virus vaccines: current status and future direction. Vaccine. 2015;33(33):4069–80.CrossRefPubMed Renukaradhya GJ, Meng XJ, Calvert JG, Roof M, Lager KM. Live porcine reproductive and respiratory syndrome virus vaccines: current status and future direction. Vaccine. 2015;33(33):4069–80.CrossRefPubMed
53.
Zurück zum Zitat Renukaradhya GJ, Meng XJ, Calvert JG, Roof M, Lager KM. Inactivated and subunit vaccines against porcine reproductive and respiratory syndrome: current status and future direction. Vaccine. 2015;33(27):3065–72.CrossRefPubMed Renukaradhya GJ, Meng XJ, Calvert JG, Roof M, Lager KM. Inactivated and subunit vaccines against porcine reproductive and respiratory syndrome: current status and future direction. Vaccine. 2015;33(27):3065–72.CrossRefPubMed
54.
Zurück zum Zitat Huang YW, Meng XJ. Novel strategies and approaches to develop the next generation of vaccines against porcine reproductive and respiratory syndrome virus (PRRSV). Virus Res. 2010;154(1–2):141–9.CrossRefPubMed Huang YW, Meng XJ. Novel strategies and approaches to develop the next generation of vaccines against porcine reproductive and respiratory syndrome virus (PRRSV). Virus Res. 2010;154(1–2):141–9.CrossRefPubMed
55.
Zurück zum Zitat Kimman TG, Cornelissen LA, Moormann RJ, Rebel JM, Stockhofe-Zurwieden N. Challenges for porcine reproductive and respiratory syndrome virus (PRRSV) vaccinology. Vaccine. 2009;27(28):3704–18.CrossRefPubMed Kimman TG, Cornelissen LA, Moormann RJ, Rebel JM, Stockhofe-Zurwieden N. Challenges for porcine reproductive and respiratory syndrome virus (PRRSV) vaccinology. Vaccine. 2009;27(28):3704–18.CrossRefPubMed
56.
Zurück zum Zitat Vu HL, Pattnaik AK, Osorio FA. Strategies to broaden the cross-protective efficacy of vaccines against porcine reproductive and respiratory syndrome virus. Vet Microbiol. 2017;206:29–34.CrossRefPubMed Vu HL, Pattnaik AK, Osorio FA. Strategies to broaden the cross-protective efficacy of vaccines against porcine reproductive and respiratory syndrome virus. Vet Microbiol. 2017;206:29–34.CrossRefPubMed
57.
Zurück zum Zitat Kappes MA, Miller CL, Faaberg KS. Highly divergent strains of porcine reproductive and respiratory syndrome virus incorporate multiple isoforms of nonstructural protein 2 into virions. J Virol. 2013;87(24):13456–65.CrossRefPubMedPubMedCentral Kappes MA, Miller CL, Faaberg KS. Highly divergent strains of porcine reproductive and respiratory syndrome virus incorporate multiple isoforms of nonstructural protein 2 into virions. J Virol. 2013;87(24):13456–65.CrossRefPubMedPubMedCentral
58.
Zurück zum Zitat Kappes MA, Miller CL, Faaberg KS. Porcine reproductive and respiratory syndrome virus nonstructural protein 2 (nsp2) topology and selective isoform integration in artificial membranes. Virology. 2015;481:51–62.CrossRefPubMed Kappes MA, Miller CL, Faaberg KS. Porcine reproductive and respiratory syndrome virus nonstructural protein 2 (nsp2) topology and selective isoform integration in artificial membranes. Virology. 2015;481:51–62.CrossRefPubMed
Metadaten
Titel
ORF1a of highly pathogenic PRRS attenuated vaccine virus plays a key role in neutralizing antibody induction in piglets and virus neutralization in vitro
verfasst von
Chaoliang Leng
Wuchao Zhang
Hongliang Zhang
Yunchao Kan
Lunguang Yao
Hongyue Zhai
Mingliang Li
Zhen Li
Chunxiao Liu
Tongqing An
Jinmei Peng
Qian Wang
Yumin Leng
Xuehui Cai
Zhijun Tian
Guangzhi Tong
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
Virology Journal / Ausgabe 1/2017
Elektronische ISSN: 1743-422X
DOI
https://doi.org/10.1186/s12985-017-0825-2

Weitere Artikel der Ausgabe 1/2017

Virology Journal 1/2017 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.