You are on page 1of 10

Virus Research 204 (2015) 21–30

Contents lists available at ScienceDirect

Virus Research
journal homepage: www.elsevier.com/locate/virusres

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
Baochao Fan a,1 , Xing Liu a,1 , Juan Bai a , Tingjie Zhang a , Qiaoya Zhang a , Ping Jiang a,b,∗
a
Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University,
Nanjing 210095, China
b
Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China

a r t i c l e i n f o a b s t r a c t

Article history: Porcine reproductive and respiratory syndrome virus (PRRSV) is mainly responsible for the heavy eco-
Received 10 January 2015 nomic losses in pig industry in the world. A number of neutralizing epitopes have been identified in the
Received in revised form 8 April 2015 viral structural proteins GP3, GP4, GP5 and M. In this study, the important amino acid (aa) residues of
Accepted 10 April 2015
HP-PRRSV strain BB affecting neutralization susceptibility of antibody were examined using resistant
Available online 20 April 2015
strains generated under neutralizing antibody (NAb) pressure in MARC-145 cells, reverse genetic tech-
nique and virus neutralization assay. HP-PRRSV strain BB was passaged under the pressure of porcine
Keywords:
NAb serum in vitro. A resistant strain BB34s with 102 and 104 aa substitutions in GP5, which have
PRRSV
102 and 104
been predicted to be the positive sites for pressure selection (Delisle et al., 2012), was cloned and
Neutralizing antibody identified. To determine the effect of the two aa residues on neutralization, eight recombinant PRRSV
strains were generated, and neutralization assay results confirmed that the aa residues 102 and 104
in GP5 played an important role in NAbs against HP-PRRSV in MARC-145 cells and porcine alveolar
macrophages. Alignment of GP5 sequences revealed that the variant aa residues at 102 and 104 were
frequent among type 2 PRRSV strains. It may be helpful for understanding the mechanism regulating
the neutralization susceptibility of PRRSV to the NAbs and monitoring the antigen variant strains in the
field.
© 2015 Elsevier B.V. All rights reserved.

1. Introduction the most economically important viruses in the swine industry


worldwide (Neumann et al., 2005; Pejsak et al., 1997). PRRSV
Porcine reproductive and respiratory syndrome virus (PRRSV) genome is a positive, single-stranded, 5 -capped and 3 -
is a single-stranded, positive-sense RNA virus that belongs to the polyadenylated mRNA molecule, with a length of approximately
Arteriviridae family of the order Nidovirales (Gorbalenya et al., 15,000 nucleotides (nt). It contains, in the direction 5 –3 , two
2006). Based on genetic and antigenic characteristics, two major large open reading frames (ORFs), ORF1a and 1b, which encode
genotypes of PRRSV, type 1 (European; prototype strain Lelystad) the viral replicase and constitute approximately three-quarters
and type 2 (North American; prototype strain VR-2332), have been of the genome, and seven smaller ORFs, designated 2a, 2b and 3
identified and share approximately 55–70% nucleotide identity through 7, which express structural proteins termed GP2a, E, GP3,
(Andreyev et al., 1997; Mateu et al., 2006; Meng et al., 1994; Nelsen GP4, GP5, M and N, respectively (Meulenberg, 2000). An additional
et al., 1999). PRRSV is responsible for reproduction problems in structural protein, GP5a, exists and is encoded by an alternative
sows and boars, and respiratory problems in pigs of all ages (Collins ORF in the subgenomic viral mRNA encoding GP5 (Johnson et al.,
et al., 1992; Wensvoort et al., 1991). It is considered to be one of 2011).
PRRSV can cause persistent infections in pigs. The NAbs in
PRRSV-infected animals are generated late and their titers remain
low (reviewed by Balasuriya and MacLachlan, 2004; Lopez and
∗ Corresponding author at: College of Veterinary Medicine, Nanjing Agricultural
Osorio, 2004). The delayed or weak induction of NAbs upon
University, Nanjing 210095, China. Tel.: +86 25 84395504; fax: +86 25 84396640.
E-mail address: jiangp@njau.edu.cn (P. Jiang). arterivirus infection has frequently been linked to GP5 glycosyl-
1
These authors contributed equally to this work. ation (Vu et al., 2011). Passive transfer of NAbs to pigs prior to

http://dx.doi.org/10.1016/j.virusres.2015.04.015
0168-1702/© 2015 Elsevier B.V. All rights reserved.
22 B. Fan et al. / Virus Research 204 (2015) 21–30

challenge with a homologous virulent PRRSV strain results in com- the serums were isolated and named N1, N2, N3, N4 and N5,
plete protection of the pigs against infection, demonstrating the respectively. The neutralizing antibody (NA) titers of these serums
important role of NAbs in protective immunity (Lopez et al., 2007; against HP-PRRSV BB were 1:34–1:48.
Osorio et al., 2002). However, the role of anti-PRRSV NAbs for the
virus diversity and evolution has not been understood completely.
The genomic variation sometimes results in the emergence of 2.2. Isolation of antibody-resistant variants
new PRRSV populations (Allende et al., 2000; Chang et al., 2002;
Rowland et al., 1999). It is expected that the adaptive immune To select variants resistant to NAb against HP-PRRSV, PRRSV
response of the host will act as an important source of selective strain BB (100 TCID50 ) was mixed with a 100-fold dilution of the
pressure in the evolutionary process of the virus (Al-Gelban, 2004; anti-PRRSV antibody and incubated at 37 ◦ C for 1 h before infecting
Domingo et al., 2001). Several studies have been published on the MARC-145 cell monolayers. The virus arising from the cells show-
examination of PRRSV sequence variability followed the selective ing a CPE was used for the subsequent passage. After passaged by
pressure on PRRSV genes during infection in vivo. But they did not 5–30 times under the antibody pressure, five NAb-resistant vari-
reveal a correlation between regions under high positive selection ant clones were selected by using plaque tests. The titers of the
and the location of neutralizing B-cell epitopes (Allende et al., 2000; resistant virus were 106 –108 TCID50 /ml.
Chang et al., 2002; Goldberg et al., 2003). Costers et al. (2010a,
2010b) isolated the antibody escape variants from the vaccinated 2.3. Sequencing of resistant variants
pigs and revealed that NAbs in pigs was a driving force in the rapid
evolution of the neutralizing epitope on GP4 of type 1 PRRSV strains Viral RNA was purified from culture supernatants taken from
(Costers et al., 2010a, 2010b). cells showing a CPE using the QIAmp viral RNA Mini Kit (Qia-
GP5 is the main target for NAbs in type 2 PRRSV strains (Gonin gen, Hilden, Germany), and reverse transcriptase PCR (RT-PCR)
et al., 1999; Ostrowski et al., 2002; Plagemann et al., 2002). A neu- was performed to generate cDNA, according to the manufacturer’s
tralizing epitope (aa 37–45: SHLQLIYNL) has also been identified protocol, using the SuperScript III First-Strand Synthesis Kit (Invi-
on GP5 (Plagemann, 2004). A recent report argued that the major trogen, Carlsbad, CA, USA). The complete genome of the virus was
envelope protein surface epitopes were disassociated with the virus divided into seven overlapping fragments for amplification, and
neutralization (Li and Murtaugh, 2012). Meanwhile, it was sug- the 5 and 3 termini of the genomic were synthesized using rapid
gested that the aa sites 102 and 104 in GP5 were likely positive amplification of the cDNA ends (RACE). The six structural protein
sites under some selective pressure by host immune cells based genes (GP2, GP3, GP4, GP5, M and N) were amplified using the
on a large dataset of 1301 sequences (1998–2009) analysis (Delisle Phanta Super Fidelity DNA Polymerase (Vazyme, China) with the
et al., 2012). In this study, a type 2 highly pathogenic PRRSV (HP- special primers.
PRRSV) strain BB0907 (tenth passage, termed BB) was passaged
in MARC-145 cells in the presence of porcine serum antibodies
against this PRRSV strain, and five resistant variants were gener- 2.4. Serum neutralization assay
ated. The sequencing results showed that BB30s and BB34s had aa
substitutions at 102 and 104 in GP5 gene. Then eight recombinant The NA titers of the antibody serums against the parent and
PRRSV strains containing these mutations were generated by site- mutant HP-PRRSVs were assayed in MARC-145 cells as described
directed mutagenesis using BB and BB34s infectious cDNA clones. previously (Jiang et al., 2006) with minor modifications. The
It was found that the aa residues at 102 and 104 in GP5 of PRRSV viruses were diluted to a concentration of 100 TCID50 per 50 ␮l
played important role in escaping from the NAbs against HP-PRRSV. (103.3 TCID50 /ml) in DMEM supplemented with 2% FBS. Serial dilu-
Our data may provide important information for understanding the tions of the neutralizing serums were mixed with each of the
molecular mechanism regulating the neutralization susceptibility viruses and incubated at 37 ◦ C for 1 h. The mixtures (100 ␮l/well)
of PRRSV to NAbs and may be helpful for monitoring the antigen were transferred to MARC-145 monolayers in 96-well plates and
variant strains in the field. incubated for an additional 4 days at 37 ◦ C with 5% CO2 . Every
serum dilution contained four replicate wells. Cells were then
examined for CPE. NA titers were expressed as the reciprocal of
2. Materials and methods the highest dilution that completely inhibited the appearance of
the CPE.
2.1. Cells, viruses and NAb serums In addition, the NA titers of the antibody serums against PRRSVs
were detected in porcine alveolar macrophages (PAMs) as previ-
MARC-145 cells were maintained in Dulbecco’s Modified Eagle’s ously description (Vanhee et al., 2010) with minor modifications.
medium (DMEM, GIBCO, Carlsbad, CA, USA) supplemented with In brief, PAM cells were prepared from 4-week-old piglets free of
10% fetal bovine serum (FBS, GIBCO) containing 100 U penicillin/ml PRRSV by lung lavage and adjusted to 5 × 106 /ml with RPMI-1640
and 100 ␮g streptomycin/ml at 37 ◦ C with 5% CO2 . (GIBCO) medium containing 10% fetal bovine serum, 100 units/ml
The HP-PRRSV isolate BB0907 (GenBank no. HQ315835, tenth of penicillin, and 100 ␮g/ml of streptomycin. The cells were added
passage by using MARC-145 cells, termed BB) used in this to 96-well culture flasks (Costar, Corning Incorporated, NY) and
study was isolated in Guangxi Province, China, in 2009. The incubated for 6 h at 37 ◦ C in a humidified compartment to allow
recombinant PRRSV strains (rBB, rBB/GP5s, rBB34s, rBB34s/GP5, cells to adhere to flasks. Twofold serial dilutions of serum N4 in
rBB/GP5(Y102C), rBB/GP5(G104R), rBB/GP5s-R and rBB34s/GP5-R) RPMI-1640 were mixed with equal volumes of 100 TCID50 PRRSV
were constructed by site-directed mutagenesis of the aa residues strains, and incubated for 1 h at 37 ◦ C and transferred to a 96-well
according to conventional methods (Fig. 1). The titers of the viral plate (100 ␮l/well) with PAMs. The inoculum was removed after
stocks were determined by measuring their cytopathic effect (CPE) 1 h and replaced by medium, after which the cells were further
in MARC-145 cells. incubated for another 10 h. The cells were fixed and stained with
Five anti-HP-PRRSV neutralizing serums were prepared from mAb against the N protein of PRRSV and FITC-conjugated goat anti-
five 45-day-old piglets free of PRRSV by inoculated with low dose mouse IgG (BOSTER, China). The NA titers were determined as the
(104 TCID50 ) of HP-PRRSV strain BB by 2 times with interval 28 reciprocal of the highest dilution that resulted in more than 90%
days. At 70 days post-inoculation, the bloods were collected and reduction of infected cells.
B. Fan et al. / Virus Research 204 (2015) 21–30 23

Fig. 1. The construction strategy of infectious cDNA clones of the recombinant PRRSVs using BB and BF34s strains.
a
The rBB/GP5s-R and rBB34s/GP5-R were two revertant viruses and constructed from the full-length infectious cDNA clone pCMV-BB/GP5s and rBB34s/GP5 by using the
site-directed mutagenesis, respectively.

2.5. Construction of infectious cDNA clones of PRRSVs Supplementary Table S1 related to this article can be found, in
the online version, at http://dx.doi.org/10.1016/j.virusres.2015.04.
The full-length PRRSV genome was amplified using the five 015
primer pairs listed in Table S1. A recombinant plasmid (pCMV-
BB) containing the full-length cDNA of the BB was constructed as 2.6. Rescue of recombinant viruses
shown in Fig. 1. To introduce the NAb-resistant mutations of the
structural protein GP5 into PRRSV infectious cDNA clone pCMV-BB, Plasmids carrying a full-length PRRSV cDNA were individually
site-directed mutagenesis was employed using the QuikChange® transfected into MARC-145 cells using Lipofectamine 2000 (Invitro-
II XL Site-Directed Mutagenesis kit (Stratagene, La Jolla, CA, USA) gen) according to the manufacturer’s instructions. Four days after
according to the manufacturer’s recommendations. First, fragment transfection, the rescues of infectious viruses were obtained and
D, which encodes the structural proteins, was amplified using cloned by the plaque assay. The mutations in the rescued viruses
pCMV-BB as template and cloned into the pEASY-Simple Blunt vec- were confirmed by RT-PCR and sequencing.
tor (Beijing TransGen Biotech Co., Ltd., Beijing, China) using AscI and
SpeI restriction endonucleases (New England Biolabs, USA), thereby 2.7. Viral plaque assay
yielding pEASY-BB-D, which was used as the intermediate plasmid.
Second, the site-directed mutagenesis constructs were prepared MARC-145 cells in 12-well plates were inoculated with 100 ␮l
using pEASY-BB-D as template. Briefly, the oligonucleotide primers of tenfold serially diluted PRRSV. After 1 h adsorption at 37 ◦ C, cell
were designed such that a foreign insertion sequence was incorpo- monolayers were washed with phosphate-buffered saline (PBS)
rated at the 5 end or in the middle of the primer, leaving at least and overlaid with 1% low melting agarose in DMEM (Invitrogen)
ten nucleotides at the 3 end that matched the template sequence. containing 2% FBS. After the gel overlay solidified, the plates were
PCRs were run according to the instructions of the QuickChange
mutagenesis kit using circular plasmid DNA as the template. The
Table 1
plasmid template was eliminated by DpnI digestion (New England The recombinant viruses used in this study and the related mutant aa sites.
Biolabs), followed by transformation of the digested PCR mixtures
Name aa mutant sites Name aa mutant sites
into Top10 competent cells (Invitrogen). The intermediate plas-
mids were screened and verified by restriction enzyme mapping rBB No rBB34s No
and nucleotide sequencing. Third, the mutations in pEASY-BB-D rBB/GP5s GP5(102Y/C; 104G/R) rBB34s/GP5 GP5(102C/Y;
104R/G)
and pCMV-BB were digested with AscI/SpeI, and fragment D in rBB/GP5(Y102C) GP5(102Y/C) rBB34s/GP5-Rb No
pCMV-BB was replaced by the analogous fragments derived from rBB/GP5(G104R) GP5(104G/R)
pEASY-BB-D, and the full-length mutant clones from pCMV-BB rBB/GP5s-Ra No
were obtained. Meanwhile, the plasmid pCMV-BB34s and its site- a
The recombinant virus rBB/GP5s-R was a revertant virus from rBB/GP5s and
directed mutation plasmids were also constructed according the contained 102Y and 104G in GP5. The plasmid was constructed from the full-
above methods (Fig. 1). Besides, two different reverse mutants length infectious cDNA clone pCMV-BB/GP5s by using the site-directed mutagenesis
that were constructed from the full-length infectious cDNA clones described.
b
The recombinant virus rBB34s/GP5-R was a revertant virus from rBB34s/GP5 and
pCMV-BB/GP5s and pCMV-BB34s/GP5 by the site-directed muta- contained 102C and 104R in GP5. The plasmid was constructed from the full-length
genesis described above, respectively (Fig. 1). All the recombinant infectious cDNA clone pCMV-BB34s/GP5 by using the site-directed mutagenesis
viruses and the aa mutations are summarized in Table 1. described.
24 B. Fan et al. / Virus Research 204 (2015) 21–30

inverted (top side down) and placed into an incubator at 37 ◦ C with Table 2
Amino acid mutations in the structural proteins of the different generation viruses
5% CO2 . At 4 days post-infection (dpi), plaques were visualized by
under immune pressure.
crystal violet staining.
AA mutations

BB BB5s BB10s BB20s BB30s BB34s


2.8. Antibody binding analyses
GP2 198 G D V V V V

Parent and recombinant mutant PRRSVs were purified by ultra- GP3 69 P P S S S S


centrifugation and diluted in coating buffer to a final concentration 71K K R R R R
226 S S P P P P
5 ␮g/ml. The antigens were added in triplicate to 96-well flat-
bottomed enzyme-linked immunosorbent assay (ELISA) plates. GP4 43 D N N N N N
After incubation at 4 ◦ C for at least 16 h, wells were blocked with 44 F S S S S S

PBS-5% FBS for 1 h at room temperature. A non-saturating con- GP5 102 Y Y Y Y C C


centration of the serum antibody (diluted 1:100), which was in 104 G G G G R R
the linear portion of the antibody titration curve determined pre- M 70 R R K K K K
viously, was added to each well, serially diluted twofold, and
incubated with the virus-coated plates for 1 h at room temperature.
After washing, anti-pig horseradish peroxidase (HRP)-conjugated 3. Results
antibody was added to the plates, and they were incubated
for 1 h at room temperature. After another round of washing, 3.1. Generation of resistant variants against NAb to PRRSV
3,3 ,5,5 -tetramethylbenzidine (TMB) substrate (KPL Biomedical,
Gaithersburg, MD) was added, and 2 M H2 SO4 was used to stop Five different NAb serums were prepared from the PRRSV-free
the reaction. The amount of HRP product was determined using a piglets infected with PRRSV strain BB. The NA titers of the serums
plate reader at 450 nm. The control plates were coated with ultra- against this strain were 1:34–1:48. We used the N4 NAb serum that
centrifuged non-infected MARC-145 cell lysates. had the highest NA titer (1:48) to generate the resistant variants
from the parental virus BB.
After 5–34 passages or purification of PRRSV BB in MARC-145
2.9. Western blot cells in the presence of N4 NAb to PRRSV, five resistant strains BB5s,
BB10s, BB20s, BB30s and BB34s, which could replicated in MARC-
In order to evaluate the consistence of PRRSV antigens bind- 145 under pressure of NAb to PRRSV, were obtained. As shown in
ing in the wells, the antigens were lysed from the coated 96-well Fig. 2, the plaque morphologies of BB30s and BB34s were larger
ELISA plate by using RIPA Lysis Buffer (Beyotime, China). Briefly, than that of the parental strain BB in size (Fig. 2A). Neutralization
100 ␮l lysis buffer was added into one coated well. After 5 min, the assays results showed that BB10s and BB20s had similar NA titers
lysate samples were obtained and added to another coated well. A that were significantly lower against that of BB (P < 0.05). The NA
total of 12 wells (for one strain antigen) were lysed and obtained titers of BB30s and BB34s were significantly lower than those of
as one sample. Then all the PRRSV strains antigen samples from BB20s (P < 0.01) and BB (P < 0.001). BB34s almost totally escaped
the coated plate and the original purified antigens were separated the neutralizing by N4 (Fig. 2B).
by 12% SDS-PAGE, and transferred onto nitrocellulose filter mem- Meanwhile, the viral replication kinetics results showed that
brane (PALL, New York, USA). The membranes were incubated with BB reached peak titer at 48 hpi. BB10s and BF30s reached peak
mAb N (made in our laboratory) or GP5 (provided by Dr GZ. Tong, titers at 36 and 24 hpi, respectively. BB34s had the highest titers
Shanghai Veterinary Research Institute, China) as the primary anti- (109 TCID50 /ml) in these six strains (Fig. 2C). It suggested that BB34s
body. After the membranes were rinsed with PBS, the membrane could replicate more efficiently in MARC-145 than the parental
was treated with goat anti-mouse IgG-HRP (BOSTER, China) as the virus BB.
secondary antibody. The proteins were visualized by scanning the Furthermore, the replication of those resistant strains viruses
membranes with the Tanon 5200 chemiluminescence imaging sys- in PAMs was detected with or without NAb to PRRSV. The results
tem (Tanon, China). showed that the NA titers of N4 serum with BB30s and BB34s
were significantly lower than those with their parental virus BB
(P < 0.001) (Fig. 2D). It indicated that they escaped efficiently from
2.10. Growth curves of viruses
the neutralizing by N4. But they could replicate efficiently in PAMs
as BB (Fig. 2E).
To determine viral one-step growth curves, PRRSV mutants
(105 TCID50 ) were inoculated into sub-confluent MARC-145 cells
3.2. Sequence analyses of the NAb resistant variants
or PAMs in six-well plates. Then, 200 ␮l of the supernatants of the
infected cells was collected and replenished with the same volume
To map the binding epitope defined by the NAb serum, the struc-
of fresh medium at 12, 24, 36, 48, and 72 h post-infection (hpi), and
tural protein genes of the resistant strains and parent strain BB were
stored at −70 ◦ C for virus titration. The virus titers for each time
sequenced and analyzed. As shown in Table 2, BB34s had a total of
point were determined in MARC-145 cells by TCID50 .
nine different aa substitutions in the GP2 (G198V), GP3 (P69S, K71R,
and S226P), GP4 (D43N and F44S), GP5 (Y102C and G104R) and M
2.11. Statistical analysis (R70K) proteins, compared with the parent PRRSV BB. Meanwhile,
BB34s had only different aa residues at 102 and 104 in GP5, when
All data were analyzed using GraphPad Prism (Version 5.03, compared with BB10s and BB20s.
San Diego, California) software. Differences among all groups were
examined using one-way analysis of variance (ANOVA), followed 3.3. Identification of the antibody binding sites
by Tukey’s tests. Differences between two groups were assessed
using unpaired two-tailed t-tests. Differences were considered sig- To elucidate the role of the two aa mutations at 102 and 104
nificant if P was <0.05. in GP5 of BB34s in the virus’s escape ability, four recombinant
B. Fan et al. / Virus Research 204 (2015) 21–30 25

Fig. 2. Generation and characterization of the resistant variants from PRRSV BB under NAb to HP-PRRSV in MARC-145 cells (A–C) and PAMs (D and E). The neutralization
assays of the variants were generated under N4 neutralizing serum in MARC-145 cells. The neutralization titer represents the inverse of the highest antibody dilution at
which an infectivity of 100 TCID50 of the viruses was neutralized. (A) Analysis of plaque morphologies. (B and D) Virus neutralization assays. (C and E) The growth kinetics.
The data are represented as the means ± s.d. of three independent experiments, and significant differences are shown (*P < 0.05 and ***P < 0.001) by using one-way analysis
of variance (ANOVA).

PRRSV strains, rBB, rBB/GP5s, rBB34s and rBB34s/GP5 were res- showed that rBB/GP5s and rBB34s had a higher replication capacity
cued by mutations of both aa residues at 102 and 104 by using than rBB and rBB34s/GP5 (Fig. 3C).
reverse genetic techniques as Fig. 1. In addition, in order to definite
that the results were due to the aa mutants, two revertant viruses 3.4. Binding analysis of resistant variants
rBB/GP5s-R and rBB34s/GP5-R were also generated.
Plaque assay results showed that the plaques of rBB/GP5s were ELISA binding was used to confirm that the resistant ability of
larger than those of rBB, and the plaques of rBB34s/GP5 were the viruses was due to the antigenic mutations. As shown in Fig. 4A,
smaller than those of rBB34s (Fig. 3A). The NA titer of the N4 the binding antigens in the plates that coated with the purified
serum with rBB/GP5s was significantly lower than that with rBB viruses at the same concentration almost had the same amount
(P < 0.001). The NA titers of the same serum with rBB34s/GP5 were of GP5 and N proteins. Meanwhile, the ELISA results showed that
significantly higher than that with rBB34s (P < 0.001). Meanwhile, the evasion virus BB34s demonstrated significantly lower bind-
the revertant viruses rBB/GP5s-R and rBB34s/GP5-R had the simi- ing ability to N4 serum compared with its parental virus BB. The
lar NA titers with rBB and rBB34s, respectively (Fig. 3B). These data rBB/GP5s had higher binding inability to N4 than did rBB. rBB34s
suggested that the two aa residues at 102 and 104 in GP5 were crit- and its parent virus BB34s had the lowest OD values compared with
ical for binding the NAb. Moreover, viral replication kinetic results other virus groups. However, the aa mutant viruses rBB34s/GP5
26 B. Fan et al. / Virus Research 204 (2015) 21–30

Fig. 3. Characterization of the recombinant PRRSV strains rBB, rBB/GP5s, rBB34s, rBB34s/GP5 and two revertant viruses rBB/GP5s-R and rBB34s/GP5-R. (A) Plaque morphology
assay. (B) Virus neutralization assays of recombinant viruses by using the N4 serum. (C) The growth kinetics. The data are represented as the means ± s.d. of three independent
experiments, and significant differences are shown (***P < 0.001) by using one-way analysis of variance (ANOVA).

significantly increased the N4 binding ability and had similar OD rBB34s/GP5 were significantly higher than those with BB34s. It con-
values to BB (Fig. 4B). firmed that the aa residues 102 and 104 in GP5 played important
role in the binding ability of PRRSV to its NAb.
3.5. Identification of aa mutations Y102C and G104R in GP5
3.7. Replication of the recombinant PRRSV in PAMs
In order to determine the key aa site in GP5 played greater
role in the neutralization abilities, we constructed another two The NA titers of N4 serums antibody were measured with the
recombinant viruses that contained only one aa mutation Y102C different mutations in PAMs. As shown in Fig. 6A, rBB34s signif-
or G104R in GP5 using plasmid pCMV-BB (Fig. 1). As shown in icantly reduced the NA titers of the serum when compared with
Fig. 5A, the recombinant viruses rBF10/sGP5(Y102C) formed larger rBB (P < 0.001). Meanwhile, the NA titer with rBB/GP5s was signif-
plaques than rBB/GP5s(G104R) and rBB. The neutralization abili- icantly lower than that with rBB (P < 0.01). And the NA titers of the
ties of rBB/GP5s(Y102C) and rBB/GP5s(G104R) were both obviously same serum with rBB34s/GP5 were significantly higher than that
decreased when compared with that of rBB (P < 0.05) (Fig. 5B). with rBB34s (P < 0.01). However, the viral replication kinetics of the
Meanwhile, rBB/GP5s(Y102C) had a higher replication capacity four virus strains rBB, rBB/GP5s, rBB34s, rBB34s/GP5 were similar
than rBB/GP5s(G104R) and rBB (Fig. 5C). to each other (Fig. 6B).

3.6. Resistances of the variants against different NAb serums to 3.8. Comparison of the aa sequences of PRRSV isolates in the field
HP-PRRSV
As shown in Fig. 7, some field isolates also had the same aa
The NA titers of five serums antibody samples were mea- mutants as BB34s and BB30s. For example, strains HUBEZ1 and
sured with the different strains obtained as above. As shown JMS01 possessed the Y102C substitution, while HN10 possessed
in Table 3, the NA titers with BB34s and rBB/GP5s were the G104R substitution in GP5, as did BB34s. PRRSV strains GD and
significantly reduced when compared to those with the parent SZ1-09 had some other aa substitutions at these two sites (Y102L
virus BB. Meanwhile, the neutralizing abilities of the serums with and G104E). It was also noticed that aa 102 of the classical PRRSV
strains VR2332 and S1 was valine (V), which is different from those
Table 3 of HP-PRRSV strains, such as SY0608 and JXA1.
The neutralizing antibody titers of the five serums against the four different
PRRSV strains. The data are represented as the means ± s.d. of three independent
experiments. 4. Discussion

Virus Neutralizing antibody titers (mean ± s.d.)


PRRSV has been proved to be highly genetically variable (Leng
N1 N2 N3 N4 N5 et al., 2014), and a number of antigenic sites recognized by NAbs
BB 38 ± 5.9 34 ± 3.8 42 ± 4.8 48 ± 6.7 40 ± 2.8 have been identified in the viral structural proteins (Meulenberg
BB34s 3 ± 2.9 2 ± 1.9 4 ± 3.6 4 ± 2.2 3 ± 0.9 et al., 1997; Oleksiewicz et al., 2001, 2005; Plagemann, 2004).
rBB/GP5s 8 ± 2.5 6 ± 2.9 8 ± 3.3 10 ± 3.3 6 ± 1.6 However, the key aa residues related to viral neutralization sus-
rBB34s/GP5 31 ± 3.2 28 ± 5.3 34 ± 4.3 34 ± 5.2 31 ± 1.8
ceptibility to the porcine serum neutralizing antibody have not
B. Fan et al. / Virus Research 204 (2015) 21–30 27

Fig. 4. Binding analysis of the escape and recombinant mutant strains. (A) The purified antigens and binding antigens of PRRSV strains from the coated 96-well ELISA plate
were detected by Western blot with the monoclonal antibodies against GP5 and N proteins of PRRSV. (B) A binding ELISA was performed using plates coated with each of the
indicated viruses in triplicate. Serial dilutions of N4 serum were added to the wells, followed by the addition of a secondary anti-pig antibody conjugated to HRP. OD values
were read at 450 nm. The negative control was wells that coated with ultracentrifuged non-infected MARC-145 cell lysates.

been understood completely. In this study, the HP-PRRSV BB strain Wissink et al., 2003; Yang et al., 2000). The specific sequences of
was passaged in MARC-145 cells under the pressure of a serum linear neutralizing epitopes in GP5 have been identified as aa 37–45
NAb against HP-PRRSV. And five resistant variants that signifi- (SHLQLIYNL) and aa 36–52 (SSHLQLIYNLTLCELNG) of the North
cantly reduced the NA titers compared with BB were generated. American strain VR-2332 (Ostrowski et al., 2002; Plagemann et al.,
The sequencing results showed that the BB34s and BB30s contained 2002). Synthetic peptides representing neutralizing epitopes from
two aa mutations Y102C and G104R in GP5 when compared to PRRSV GP5 did not induce virus neutralizing antibodies, suggesting
BB10s and BB20s, which being suggested as positive sites for evo- that additional residues or structural features are critical for anti-
lutionary selection by analyzed a large dataset of 1301 sequences genicity (Lopez and Osorio, 2004; Plagemann, 2001). In this study,
(1998–2009) (Delisle et al., 2012). The consistency of the aa sites we identified two aa sites, 102 and 104, which were the important
promoted us to investigate the importance of these two sites. By antibody binding sites in GP5 using an immune pressure system in
using the reverse genetic techniques and the neutralization assay, vitro. A TMHMM prediction shows that this region will be predicted
it was firstly confirmed that the two aa residues, 102 and 104 within to locate in one putative, short ectodomain region (aa 100–109)
GP5 of PRRSV played important role in escaping from NAbs against after two transmembrane domains prior to the third transmem-
PRRSV. brane region in GP5 (Li and Murtaugh, 2012; Mardassi et al., 1995,
The major structural proteins, GP5 and M, form disulfide-linked 1996). Considering the important role of the two aa substitutions
heterodimers that bind to heparin sulfate, which is required for (102 and 104) in preventing neutralization by antibodies, as well as
virus attachment (Delputte et al., 2005; Faaberg et al., 1995; Snijder the fact that no other aa substitutions occurred in GP5, especially
et al., 2003). Many reports implicate GP5 and M as key PRRSV neu- in the confirmed neutralizing epitopes, it is likely that the two aa
tralization targets (Ostrowski et al., 2002; Pirzadeh and Dea, 1997; substitutions may be one part for formation of the conformational
Plagemann, 2004; Plagemann et al., 2002; Weiland et al., 1999; neutralizing epitopes in GP5. Of course, this hypothesis should be
28 B. Fan et al. / Virus Research 204 (2015) 21–30

Fig. 5. Characterization of the recombinant PRRSV rBB/GP5(Y102C) and rBB/GP5(G104R). (A) Plaque morphology assays. (B) Virus neutralization assays of recombinant
viruses by using N4 neutralizing serum. (C) The growth kinetics. The data are represented as the means ± s.d. of three independent experiments, and significant differences
are shown (*P < 0.05, **P < 0.01 and ***P < 0.001) by using one-way analysis of variance (ANOVA).

investigated further after the three-dimensional model of PRRSV of the neutralizing epitope on GP4 of type 1 PRRSV strains (Costers
GP5 obtained. et al., 2010a, 2010b). In this study, the sequence results showed
The in vitro generation of escape mutants is unlikely to recapitu- that the structure proteins GP2, GP3, GP4 and M of the resistant
late drift in its entirety. This is largely due to the stochastic nature of strains also contained some aa substitutions compared with the
drift and the very large mutational space available. The exertion of parent viruses. In addition, the complete genome sequence analy-
antibody-mediated selective pressure onto a neutralizing epitope sis results of BB34s (GenBank no. KM453698) revealed that some aa
rapidly resulted in the selection of viable type 1 PRRSV variants that mutants also occurred in the non-structural proteins (Nsps) of this
were resistant to neutralization by these antibodies. It was found strain when compared to BB (data not shown). The resistant variant
that NAbs in pigs might be a driving force in the rapid evolution BB34s replicated more efficiently in MARC-145 than the parental

Fig. 6. Characterization of the recombinant PRRSV strains in PAMs. (A) Virus neutralization assays of recombinant viruses by using N4 neutralizing serum. (B) The growth
kinetics of recombinant viruses. The data are represented as the means ± s.d. of three independent experiments, and significant differences are shown (**P < 0.01 and
***P < 0.001) by using one-way analysis of variance (ANOVA).
B. Fan et al. / Virus Research 204 (2015) 21–30 29

Fig. 7. Alignment of partial GP5 aa sequences of BB34s with wild-type PRRSV isolates. Y102C and G104R in GP5 of BB34s and the same sites of the field PRRSV isolates are
indicated by black boxes.

virus BB. The changes in Nsps of BB34s upon long term passaging Acknowledgments
in a host cell might suggest further adaptation of the virus to the
host cell machinery. This work was mainly supported by the National Natural Science
Using monoclonal antibodies (mAbs) directed against a sepa- Foundation (31230071), grants from the Ministry of Education,
rate protein, especially the mAbs against conformational epitopes, China (313031, 2012009711004) for PRRSV immunology, a grant
may lead to the comprehensive identification of neutralizing epi- from the Ministry of Agriculture (CARS-36) for swine disease
topes. The antigenic sites of influenza virus have been identified controlling techniques, and the priority academic program devel-
using immune pressure method with the specific mAbs (Hensley opment of Jiangsu higher education institutions (PAPD).
et al., 2009; O’Donnell et al., 2012). In this study, we identified the
antigenic sites using an anti-PRRSV neutralizing polyclonal serum References
made in swine. In addition, during the passage of PRRSV strain BB,
we used one negative anti-PRRSV serum as negative pressure con- Al-Gelban, K.S., 2004. Intra-rator variability of primary care physicians in manage-
ment of hyperlipidaemia. West Afr. J. Med. 23 (1), 69–73.
trol. The sequence results indicated that the viruses at passage 10 Allende, R., Laegreid, W.W., Kutish, G.F., Galeota, J.A., Wills, R.W., Osorio, F.A., 2000.
and 34 do not have aa substitutions at aa102 and aa104 in GP5 Porcine reproductive and respiratory syndrome virus: description of persistence
(data not shown here). Of course, further studies should be needed in individual pigs upon experimental infection. J. Virol. 74 (22), 10834–10837.
Andreyev, V.G., Wesley, R.D., Mengeling, W.L., Vorwald, A.C., Lager, K.M., 1997.
to improve these techniques and to address the importance of other Genetic variation and phylogenetic relationships of 22 porcine reproductive and
putative antigenic sites using different anti-PRRSV serums. respiratory syndrome virus (PRRSV) field strains based on sequence analysis of
In addition, we noted that the significantly resistant virus BB30s open reading frame 5. Arch. Virol. 142 (5), 993–1001.
Balasuriya, U.B., MacLachlan, N.J., 2004. The immune response to equine arteritis
was generated on the passage 30. It takes a long time when com- virus: potential lessons for other arteriviruses. Vet. Immunol. Immunopathol.
pared with the mAb-resistant PRRSV strains of LV generated on 102 (3), 107–129.
passage 5 (Costers et al., 2010a). The possible reasons for this dif- Chang, C.C., Yoon, K.J., Zimmerman, J.J., Harmon, K.M., Dixon, P.M., Dvorak, C.M., Mur-
taugh, M.P., 2002. Evolution of porcine reproductive and respiratory syndrome
ference might be related to the techniques of cloning and passaging
virus during sequential passages in pigs. J. Virol. 76 (10), 4750–4763.
and the NAb serum. The mixed clones might be picked up during Collins, J.E., Benfield, D.A., Christianson, W.T., Harris, L., Hennings, J.C., Shaw, D.P.,
the passage of those virus clones. At passage 34s, there were still Goyal, S.M., McCullough, S., Morrison, R.B., Joo, H.S., et al., 1992. Isolation of
swine infertility and respiratory syndrome virus (isolate ATCC VR-2332) in North
virus particles present that were not resistant yet. The putative aa
America and experimental reproduction of the disease in gnotobiotic pigs. J. Vet.
residues in other proteins might regulate the viral neutralization Diagn. Invest. 4 (2), 117–126.
susceptibility to NAb, which should be investigated in the further. Costers, S., Lefebvre, D.J., Van Doorsselaere, J., Vanhee, M., Delputte, P.L., Nauwynck,
In conclusion, by passage of PRRSV under the viral specific NAb H.J., 2010a. GP4 of porcine reproductive and respiratory syndrome virus con-
tains a neutralizing epitope that is susceptible to immunoselection in vitro. Arch.
pressure, it was firstly found that the aa residues 102 and 104 in Virol. 155 (3), 371–378.
GP5 of PRRSV played important role in regulating neutralization Costers, S., Vanhee, M., Van Breedam, W., Van Doorsselaere, J., Geldhof, M.,
susceptibility to the porcine serum NAb. It may be helpful for mon- Nauwynck, H.J., 2010b. GP4-specific neutralizing antibodies might be a driving
force in PRRSV evolution. Virus Res. 154 (1/2), 104–113.
itoring the antigen variant strains in the field and developing new Delisle, B., Gagnon, C.A., Lambert, M.E., D’Allaire, S., 2012. Porcine reproductive and
vaccine against PRRSV in the future. respiratory syndrome virus diversity of Eastern Canada swine herds in a large
30 B. Fan et al. / Virus Research 204 (2015) 21–30

sequence dataset reveals two hypervariable regions under positive selection. O’Donnell, C.D., Vogel, L., Wright, A., Das, S.R., Wrammert, J., Li, G.M., McCausland, M.,
Infect. Genet. Evol. 12 (5), 1111–1119. Zheng, N.Y., Yewdell, J.W., Ahmed, R., Wilson, P.C., Subbarao, K., 2012. Antibody
Delputte, P.L., Costers, S., Nauwynck, H.J., 2005. Analysis of porcine reproductive and pressure by a human monoclonal antibody targeting the 2009 pandemic H1N1
respiratory syndrome virus attachment and internalization: distinctive roles for virus hemagglutinin drives the emergence of a virus with increased virulence
heparan sulphate and sialoadhesin. J. Gen. Virol. 86 (Pt 5), 1441–1445. in mice. mBio 3 (3).
Domingo, E., Mas, A., Yuste, E., Pariente, N., Sierra, S., Gutierrez-Riva, M., Menendez- Oleksiewicz, M.B., Botner, A., Toft, P., Normann, P., Storgaard, T., 2001. Epitope map-
Arias, L., 2001. Virus population dynamics, fitness variations and the control of ping porcine reproductive and respiratory syndrome virus by phage display: the
viral disease: an update. Prog. Drug Res. 57, 77–115. nsp2 fragment of the replicase polyprotein contains a cluster of B-cell epitopes.
Faaberg, K.S., Even, C., Palmer, G.A., Plagemann, P.G., 1995. Disulfide bonds between J. Virol. 75 (7), 3277–3290.
two envelope proteins of lactate dehydrogenase-elevating virus are essential for Oleksiewicz, M.B., Stadejek, T., Mackiewicz, Z., Porowski, M., Pejsak, Z., 2005. Dis-
viral infectivity. J. Virol. 69 (1), 613–617. criminating between serological responses to European-genotype live vaccine
Goldberg, T.L., Lowe, J.F., Milburn, S.M., Firkins, L.D., 2003. Quasispecies variation of and European-genotype field strains of porcine reproductive and respiratory
porcine reproductive and respiratory syndrome virus during natural infection. syndrome virus (PRRSV) by peptide ELISA. J. Virol. Methods 129 (2), 134–144.
Virology 317 (2), 197–207. Osorio, F.A., Galeota, J.A., Nelson, E., Brodersen, B., Doster, A., Wills, R., Zuckermann,
Gonin, P., Pirzadeh, B., Gagnon, C.A., Dea, S., 1999. Seroneutralization of porcine F., Laegreid, W.W., 2002. Passive transfer of virus-specific antibodies confers
reproductive and respiratory syndrome virus correlates with antibody response protection against reproductive failure induced by a virulent strain of porcine
to the GP5 major envelope glycoprotein. J. Vet. Diagn. Invest. 11 (1), 20–26. reproductive and respiratory syndrome virus and establishes sterilizing immu-
Gorbalenya, A.E., Enjuanes, L., Ziebuhr, J., Snijder, E.J., 2006. Nidovirales: evolving nity. Virology 302 (1), 9–20.
the largest RNA virus genome. Virus Res. 117 (1), 17–37. Ostrowski, M., Galeota, J.A., Jar, A.M., Platt, K.B., Osorio, F.A., Lopez, O.J., 2002. Identifi-
Hensley, S.E., Das, S.R., Bailey, A.L., Schmidt, L.M., Hickman, H.D., Jayaraman, A., cation of neutralizing and nonneutralizing epitopes in the porcine reproductive
Viswanathan, K., Raman, R., Sasisekharan, R., Bennink, J.R., Yewdell, J.W., 2009. and respiratory syndrome virus GP5 ectodomain. J. Virol. 76 (9), 4241–4250.
Hemagglutinin receptor binding avidity drives influenza A virus antigenic drift. Pejsak, Z., Stadejek, T., Markowska-Daniel, I., 1997. Clinical signs and economic
Science 326 (5953), 734–736. losses caused by porcine reproductive and respiratory syndrome virus in a large
Jiang, W., Jiang, P., Li, Y., Tang, J., Wang, X., Ma, S., 2006. Recombinant adenovirus breeding farm. Vet. Microbiol. 55 (1–4), 317–322.
expressing GP5 and M fusion proteins of porcine reproductive and respiratory Pirzadeh, B., Dea, S., 1997. Monoclonal antibodies to the ORF5 product of porcine
syndrome virus induce both humoral and cell-mediated immune responses in reproductive and respiratory syndrome virus define linear neutralizing deter-
mice. Vet. Immunol. Immunopathol. 113 (1/2), 169–180. minants. J. Gen. Virol. 78 (Pt 8), 1867–1873.
Johnson, C.R., Griggs, T.F., Gnanandarajah, J., Murtaugh, M.P., 2011. Novel structural Plagemann, P.G., 2001. Complexity of the single linear neutralization epitope of
protein in porcine reproductive and respiratory syndrome virus encoded by an the mouse arterivirus lactate dehydrogenase-elevating virus. Virology 290 (1),
alternative ORF5 present in all arteriviruses. J. Gen. Virol. 92 (Pt 5), 1107–1116. 11–20.
Leng, C.L., Tian, Z.J., Zhang, W.C., Zhang, H.L., Zhai, H.Y., An, T.Q., Peng, J.M., Ye, C., Sun, Plagemann, P.G., 2004. The primary GP5 neutralization epitope of North American
L., Wang, Q., Sun, Y., Li, L., Zhao, H.Y., Chang, D., Cai, X.H., Zhang, G.H., Tong, G.Z., isolates of porcine reproductive and respiratory syndrome virus. Vet. Immunol.
2014. Characterization of two newly emerged isolates of porcine reproductive Immunopathol. 102 (3), 263–275.
and respiratory syndrome virus from Northeast China in 2013. Vet. Microbiol. Plagemann, P.G., Rowland, R.R., Faaberg, K.S., 2002. The primary neutralization
171 (1/2), 41–52. epitope of porcine respiratory and reproductive syndrome virus strain VR-
Li, J., Murtaugh, M.P., 2012. Dissociation of porcine reproductive and respiratory syn- 2332 is located in the middle of the GP5 ectodomain. Arch. Virol. 147 (12),
drome virus neutralization from antibodies specific to major envelope protein 2327–2347.
surface epitopes. Virology 433 (2), 367–376. Rowland, R.R., Steffen, M., Ackerman, T., Benfield, D.A., 1999. The evolution of porcine
Lopez, O.J., Oliveira, M.F., Garcia, E.A., Kwon, B.J., Doster, A., Osorio, F.A., 2007. Pro- reproductive and respiratory syndrome virus: quasispecies and emergence of a
tection against porcine reproductive and respiratory syndrome virus (PRRSV) virus subpopulation during infection of pigs with VR-2332. Virology 259 (2),
infection through passive transfer of PRRSV-neutralizing antibodies is dose 262–266.
dependent. Clin. Vaccine Immunol. 14 (3), 269–275. Snijder, E.J., Dobbe, J.C., Spaan, W.J., 2003. Heterodimerization of the two major
Lopez, O.J., Osorio, F.A., 2004. Role of neutralizing antibodies in PRRSV protective envelope proteins is essential for arterivirus infectivity. J. Virol. 77 (1), 97–104.
immunity. Vet. Immunol. Immunopathol. 102 (3), 155–163. Vanhee, M., Costers, S., Van Breedam, W., Geldhof, M.F., Van Doorsselaere, J.,
Mardassi, H., Massie, B., Dea, S., 1996. Intracellular synthesis, processing, and trans- Nauwynck, H.J., 2010. A variable region in GP4 of European-type porcine repro-
port of proteins encoded by ORFs 5 to 7 of porcine reproductive and respiratory ductive and respiratory syndrome virus induces neutralizing antibodies against
syndrome virus. Virology 221 (1), 98–112. homologous but not heterologous virus strains. Viral Immunol. 23 (4), 403–413.
Mardassi, H., Mounir, S., Dea, S., 1995. Molecular analysis of the ORFs 3 to 7 of porcine Vu, H.L., Kwon, B., Yoon, K.J., Laegreid, W.W., Pattnaik, A.K., Osorio, F.A., 2011.
reproductive and respiratory syndrome virus, Quebec reference strain. Arch. Immune evasion of porcine reproductive and respiratory syndrome virus
Virol. 140 (8), 1405–1418. through glycan shielding involves both glycoprotein 5 as well as glycoprotein 3.
Mateu, E., Diaz, I., Darwich, L., Casal, J., Martin, M., Pujols, J., 2006. Evolution of ORF5 J. Virol. 85 (11), 5555–5564.
of Spanish porcine reproductive and respiratory syndrome virus strains from Weiland, E., Wieczorek-Krohmer, M., Kohl, D., Conzelmann, K.K., Weiland, F., 1999.
1991 to 2005. Virus Res. 115 (2), 198–206. Monoclonal antibodies to the GP5 of porcine reproductive and respiratory
Meng, X.J., Paul, P.S., Halbur, P.G., 1994. Molecular cloning and nucleotide sequencing syndrome virus are more effective in virus neutralization than monoclonal anti-
of the 3 -terminal genomic RNA of the porcine reproductive and respiratory bodies to the GP4. Vet. Microbiol. 66 (3), 171–186.
syndrome virus. J. Gen. Virol. 75 (Pt 7), 1795–1801. Wensvoort, G., Terpstra, C., Pol, J.M., ter Laak, E.A., Bloemraad, M., de Kluyver, E.P.,
Meulenberg, J.J., 2000. PRRSV, the virus. Vet. Res. 31 (1), 11–21. Kragten, C., van Buiten, L., den Besten, A., Wagenaar, F., et al., 1991. Mystery
Meulenberg, J.J., van Nieuwstadt, A.P., van Essen-Zandbergen, A., Langeveld, J.P., swine disease in The Netherlands: the isolation of Lelystad virus. Vet. Q. 13 (3),
1997. Posttranslational processing and identification of a neutralization domain 121–130.
of the GP4 protein encoded by ORF4 of Lelystad virus. J. Virol. 71 (8), 6061–6067. Wissink, E.H., van Wijk, H.A., Kroese, M.V., Weiland, E., Meulenberg, J.J., Rottier, P.J.,
Nelsen, C.J., Murtaugh, M.P., Faaberg, K.S., 1999. Porcine reproductive and respiratory van Rijn, P.A., 2003. The major envelope protein, GP5, of a European porcine
syndrome virus comparison: divergent evolution on two continents. J. Virol. 73 reproductive and respiratory syndrome virus contains a neutralization epitope
(1), 270–280. in its N-terminal ectodomain. J. Gen. Virol. 84 (Pt 6), 1535–1543.
Neumann, E.J., Kliebenstein, J.B., Johnson, C.D., Mabry, J.W., Bush, E.J., Seitzinger, Yang, L., Frey, M.L., Yoon, K.J., Zimmerman, J.J., Platt, K.B., 2000. Categorization
A.H., Green, A.L., Zimmerman, J.J., 2005. Assessment of the economic impact of North American porcine reproductive and respiratory syndrome viruses:
of porcine reproductive and respiratory syndrome on swine production in the epitopic profiles of the N, M, GP5 and GP3 proteins and susceptibility to neu-
United States. J. Am. Vet. Med. Assoc. 227 (3), 385–392. tralization. Arch. Virol. 145 (8), 1599–1619.

You might also like