Accepted Manuscript Title: 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 Author: Baochao Fan Xing Liu Juan Bai Tingjie Zhang Qiaoya Zhang Ping Jiang PII: DOI: Reference:

S0168-1702(15)00149-5 http://dx.doi.org/doi:10.1016/j.virusres.2015.04.015 VIRUS 96591

To appear in:

Virus Research

Received date: Revised date: Accepted date:

10-1-2015 8-4-2015 10-4-2015

Please cite this article as: 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 Research (2015), http://dx.doi.org/10.1016/j.virusres.2015.04.015 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

1

Highlights

2



The NAb resistant PRRSV strains were generated under NAb pressure in vitro.

3



The 102 and 104 aa sites in GP5 regulate viral neutralization susceptibilities to porcine serum NAbs in MARC-145 and PAM cells.

5



The aa mutants Y102C and G104R also appear in wild type 2 PRRSV strains.

ip t

4

cr

6

Ac ce

pt

ed

M

an

us

7

1

Page 1 of 39

The amino acid residues at 102 and 104 in GP5 of porcine

8

reproductive and respiratory syndrome virus regulate viral

9

neutralization susceptibility to the porcine serum

10

neutralizing antibody

ip t

7

11

Baochao Fana*, Xing Liua*, Juan Baia, Tingjie Zhanga, Qiaoya Zhanga, Ping Jianga、b#

13

a. Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of

14

Agriculture, College of Veterinary Medicine, Nanjing Agricultural University,

15

Nanjing 210095, China

16

b. Jiangsu Co-innovation Center for Prevention and Control of Important Animal

17

Infectious Diseases and Zoonoses, Yangzhou, China

M

an

us

cr

12

21 22 23 24

These authors contributed equally to this work.

#

Corresponding author:

pt

20

*

Dr. Ping Jiang

Ac ce

19

ed

18

College of Veterinary Medicine Nanjing Agricultural University, Nanjing 210095, P.R. China

25

Tel.: +86 25 84395504

26

Fax: + 86 25 84396640

27

[email protected]

28 29 2

Page 2 of 39

ABSTRACT

31

Porcine reproductive and respiratory syndrome virus (PRRSV) is mainly responsible

32

for the heavy economic losses in pig industry in the world. A number of neutralizing

33

epitopes have been identified in the viral structural proteins GP3, GP4, GP5 and M. In

34

this study, the important amino acid (aa) residues of HP-PRRSV strain BB affecting

35

neutralization susceptibility of antibody were examined using resistant strains

36

generated under neutralizing antibody (NAb) pressure in MARC-145 cells, reverse

37

genetic technique and virus neutralization assay. HP-PRRSV strain BB was passaged

38

under the pressure of porcine NAb serum in vitro. A resistant strain BB34s with 102

39

and 104 aa substitutions in GP5, which have been predicted to be the positive sites for

40

pressure selection (Delisle et al., 2012), was cloned and identified. To determine the

41

effect of the two aa residues on neutralization, 8 recombinant PRRSV strains were

42

generated, and neutralization assay results confirmed that the aa residues 102 and 104

43

in GP5 played an important role in NAbs against HP-PRRSV in MARC-145 cells and

44

porcine alveolar macrophages. Alignment of GP5 sequences revealed that the variant

45

aa residues at 102 and 104 were frequent among type 2 PRRSV strains. It may be

46

helpful for understanding the mechanism regulating the neutralization susceptibility of

47

PRRSV to the NAbs and monitoring the antigen variant strains in the field.

48

Key words: PRRSV; 102 and 104; neutralizing antibody.

Ac ce

pt

ed

M

an

us

cr

ip t

30

49 50 51 3

Page 3 of 39

52

53

1. Introduction Porcine reproductive and respiratory syndrome virus (PRRSV) is a single-stranded, positive-sense

RNA

virus

that

belongs

to

the

Arteriviridae

family

of

55

the order Nidovirales (Gorbalenya et al., 2006). Based on genetic and antigenic

56

characteristics, two major genotypes of PRRSV, type 1 (European; prototype strain

57

Lelystad) and type 2 (North American; prototype strain VR-2332), have been

58

identified and share approximately 55–70% nucleotide identity (Andreyev et al., 1997;

59

Mateu et al., 2006; Meng et al., 1994; Nelsen et al., 1999). PRRSV is responsible for

60

reproduction problems in sows and boars, and respiratory problems in pigs of all ages

61

(Collins et al., 1992; Wensvoort et al., 1991). It is considered to be one of the most

62

economically important viruses in the swine industry worldwide (Neumann et al.,

63

2005; Pejsak et al., 1997). PRRSV genome is a positive, single-stranded, 5′-capped

64

and 3′-polyadenylated mRNA molecule, with a length of approximately 15,000

65

nucleotides (nt). It contains, in the direction 5′-3′, two large open reading frames

66

(ORFs), ORF1a and 1b, which encode the viral replicase and constitute approximately

67

three-quarters of the genome, and seven smaller ORFs, designated 2a, 2b and 3

68

through 7, which express structural proteins termed GP2a, E, GP3, GP4, GP5, M and

69

N, respectively (Meulenberg, 2000). An additional structural protein, GP5a, exists and

70

is encoded by an alternative ORF in the subgenomic viral mRNA encoding GP5

71

(Johnson et al., 2011).

Ac ce

pt

ed

M

an

us

cr

ip t

54

72

PRRSV can cause persistent infections in pigs. The NAbs in PRRSV-infected

73

animals are generated late and their titers remain low (reviewed by (Balasuriya and 4

Page 4 of 39

MacLachlan, 2004; Lopez and Osorio, 2004). The delayed or weak induction of NAbs

75

upon arterivirus infection has frequently been linked to GP5 glycosylation (Vu et al.,

76

2011). Passive transfer of NAbs to pigs prior to challenge with a homologous virulent

77

PRRSV strain results in complete protection of the pigs against infection,

78

demonstrating the important role of NAbs in protective immunity (Lopez et al., 2007;

79

Osorio et al., 2002). However, the role of anti-PRRSV NAbs for the virus diversity

80

and evolution has not been understood completely.

us

cr

ip t

74

The genomic variation sometimes results in the emergence of new PRRSV

82

populations (Allende et al., 2000; Chang et al., 2002; Rowland et al., 1999). It is

83

expected that the adaptive immune response of the host will act as an important

84

source of selective pressure in the evolutionary process of the virus (Al-Gelban, 2004;

85

Domingo et al., 2001). Several studies have been published on the examination of

86

PRRSV sequence variability followed the selective pressure on PRRSV genes during

87

infection in vivo. But they did not reveal a correlation between regions under high

88

positive selection and the location of neutralizing B-cell epitopes (Allende et al., 2000;

89

Chang et al., 2002; Goldberg et al., 2003). Costers et al (2010) isolated the antibody

90

escape variants from the vaccinated pigs and reveled that NAbs in pigs was a driving

91

force in the rapid evolution of the neutralizing epitope on GP4 of type 1 PRRSV

92

strains (Costers et al., 2010a; Costers et al., 2010b).

Ac ce

pt

ed

M

an

81

93

GP5 is the main target for NAbs in type 2 PRRSV strains (Gonin et al., 1999;

94

Ostrowski et al., 2002; Plagemann et al., 2002). A neutralizing epitope (aa 37 to 45:

95

SHLQLIYNL) has also been identified on GP5 (Plagemann, 2004). A recent report

5

Page 5 of 39

argued that the major envelope protein surface epitopes were disassociated with the

97

virus neutralization (Li and Murtaugh, 2012). Meanwhile, it was suggested that the aa

98

sites 102 and 104 in GP5 were likely positive sites under some selective pressure by

99

host immune cells based on a large dataset of 1301 sequences (1998–2009) analysis

100

(Delisle et al., 2012). In this study, a type 2 highly pathogenic PRRSV (HP-PRRSV)

101

strain BB0907 (tenth passage, termed BB) was passaged in MARC-145 cells in the

102

presence of porcine serum antibodies against this PRRSV strain, and five resistant

103

variants were generated. The sequencing results showed that BB30s and BB34s had

104

aa substitutions at 102 and 104 in GP5 gene. Then 8 recombinant PRRSV strains

105

containing these mutations were generated by site-directed mutagenesis using BB and

106

BB34s infectious cDNA clones. It was found that the aa residues at 102 and 104 in

107

GP5 of PRRSV played important role in escaping from the NAbs against HP-PRRSV.

108

Our data may provide important information for understanding the molecular

109

mechanism regulating the neutralization susceptibility of PRRSV to NAbs and may be

110

helpful for monitoring the antigen variant strains in the field.

111

2. Materials and methods

112

2.1. Cells, viruses and NAb serums

Ac ce

pt

ed

M

an

us

cr

ip t

96

113

MARC-145 cells were maintained in Dulbecco's Modified Eagle's medium

114

(DMEM, GIBCO, Carlsbad, CA, USA) supplemented with 10% fetal bovine serum

115

(FBS, GIBCO) containing 100 U penicillin/ml and 100 μg streptomycin/ml at 37°C

116

with 5% CO2. 6

Page 6 of 39

The HP-PRRSV isolate BB0907 (GenBank no. HQ315835, tenth passage by using

118

MARC-145 cells,termed BB) used in this study was isolated in Guangxi Province,

119

China, in 2009. The recombinant PRRSV strains (rBB, rBB/GP5s, rBB34s,

120

rBB34s/GP5, rBB/GP5(Y102C), rBB/GP5(G104R), rBB/GP5s-R and rBB34s/GP5-R)

121

were constructed by site-directed mutagenesis of the aa residues according to

122

conventional methods (Fig. 1). The titers of the viral stocks were determined by

123

measuring their cytopathic effect (CPE) in MARC-145 cells.

us

cr

ip t

117

Five anti-HP-PRRSV neutralizing serums were prepared from five 45-day-old

125

piglets free of PRRSV by inoculated with low dose (104 TCID50) of HP-PRRSV strain

126

BB by 2 times with interval 28 days. At 70 days post inoculation, the bloods were

127

collected and the serums were isolated and named N1, N2, N3, N4 and N5,

128

respectively. The neutralizing antibody (NA) titers of these serums against

129

HP-PRRSV BB were 1: 34-1:48.

130

2.2. Isolation of antibody-resistant variants

M

ed

pt

Ac ce

131

an

124

To select variants resistant to NAb against HP-PRRSV, PRRSV strain BB (100

132

TCID50) was mixed with a 100-fold dilution of the anti-PRRSV antibody and

133

incubated at 37°C for 1 h before infecting MARC-145 cell monolayers. The virus

134

arising from the cells showing a CPE was used for the subsequent passage. After

135

passaged by 5-30 times under the antibody pressure, five NAb-resistant variant clones

136

were selected by using plaque tests. The titers of the resistant virus were 106–108

137

TCID50/mL.

7

Page 7 of 39

138

2.3. Sequencing of resistant variants Viral RNA was purified from culture supernatants taken from cells showing a CPE

140

using the QIAmp viral RNA Mini Kit (Qiagen, Hilden, Germany), and reverse

141

transcriptase PCR (RT-PCR) was performed to generate cDNA, according to the

142

manufacturer’s protocol, using the SuperScript III First-Strand Synthesis Kit

143

(Invitrogen, Carlsbad, CA, USA). The complete genome of the virus was divided into

144

seven overlapping fragments for amplification, and the 5′ and 3′ termini of the

145

genomic were synthesized using rapid amplification of the cDNA ends (RACE). The

146

six structural protein genes (GP2, GP3, GP4, GP5, M and N) were amplified using the

147

Phanta Super Fidelity DNA Polymerase (Vazyme, China) with the special primers.

148

2.4. Serum neutralization assay

ed

M

an

us

cr

ip t

139

The NA titers of the antibody serums against the parent and mutant HP-PRRSVs

150

were assayed in MARC-145 cells as described previously (Jiang et al., 2006) with

151

minor modifications. The viruses were diluted to a concentration of 100 TCID50 per

152

50 μl (103.3 TCID50/ml) in DMEM supplemented with 2% FBS. Serial dilutions of the

153

neutralizing serums were mixed with each of the viruses and incubated at 37°C for 1 h.

154

The mixtures (100 μl/well) were transferred to MARC-145 monolayers in 96-well

155

plates and incubated for an additional 4 days at 37°C with 5% CO2. Every serum

156

dilution contained four replicate wells. Cells were then examined for CPE. NA titers

157

were expressed as the reciprocal of the highest dilution that completely inhibited the

158

appearance of the CPE.

Ac ce

pt

149

8

Page 8 of 39

In addition, the NA titers of the antibody serums against PRRSVs were detected

160

in porcine alveolar macrophages (PAMs) as previously description (Vanhee et al.,

161

2010) with minor modifications. In brief, PAM cells were prepared from 4-week-old

162

piglets free of PRRSV by lung lavage and adjusted to 5×106/mL with RPMI-1640

163

(GIBCO) medium containing 10% fetal bovine serum, 100 units/mL of penicillin, and

164

100 μg/mL of streptomycin. The cells were added to 96-well culture flasks (Costar,

165

Corning Incorporated, NY) and incubated for 6 h at 37°C in a humidified

166

compartment to allow cells to adhere to flasks. Two-fold serial dilutions of serum N4

167

in RPMI-1640 were mixed with equal volumes of 100 TCID50 PRRSV strains, and

168

incubated for 1 h at 37°C and transferred to a 96-well plate (100 μl/well) with PAMs.

169

The inoculum was removed after 1h and replaced by medium, after which the cells

170

were further incubated for another 10 h. The cells were fixed and stained with mAb

171

against the N protein of PRRSV and FITC-conjugated goat anti-mouse IgG (BOSTER,

172

China). The NA titers were determined as the reciprocal of the highest dilution that

173

resulted in more than 90% reduction of infected cells.

174

2.5. Construction of infectious cDNA clones of PRRSVs

cr

us

an

M

ed

pt

Ac ce

175

ip t

159

The full-length PRRSV genome was amplified using the five primer pairs listed in

176

Table S1. A recombinant plasmid (pCMV-BB) containing the full-length cDNA of the

177

BB was constructed as shown in Fig. 1. To introduce the NAb-resistant mutations of

178

the structural protein GP5 into PRRSV infectious cDNA clone pCMV-BB,

179

site-directed mutagenesis was employed using the QuikChange® II XL Site-Directed

180

Mutagenesis kit (Stratagene, La Jolla, CA, USA) according to the manufacturer's 9

Page 9 of 39

181

recommendations. First, fragment D, which encodes the structural proteins, was

182

amplified using pCMV-BB as template and cloned into the pEASY-Simple Blunt

183

vector (Beijing TransGen Biotech Co., Ltd., Beijing, China) using AscI and SpeI

184

restriction

185

pEASY-BB-D, which was used as the intermediate plasmid. Second, the site-directed

186

mutagenesis constructs were prepared using pEASY-BB-D as template. Briefly, the

187

oligonucleotide primers were designed such that a foreign insertion sequence was

188

incorporated at the 5′ end or in the middle of the primer, leaving at least ten

189

nucleotides at the 3′ end that matched the template sequence. PCRs were run

190

according to the instructions of the QuickChange mutagenesis kit using circular

191

plasmid DNA as the template. The plasmid template was eliminated by DpnI

192

digestion (New England Biolabs), followed by transformation of the digested PCR

193

mixtures into Top10 competent cells (Invitrogen). The intermediate plasmids were

194

screened and verified by restriction enzyme mapping and nucleotide sequencing.

195

Third, the mutations in pEASY-BB-D and pCMV-BB were digested with AscI/SpeI,

196

and fragment D in pCMV-BB was replaced by the analogous fragments derived from

197

pEASY-BB-D, and the full-length mutant clones from pCMV-BB were obtained.

198

Meanwhile, the plasmid pCMV-BB34s and its site-directed mutation plasmids were

199

also constructed according the above methods (Fig. 1). Besides, two different reverse

200

mutants that were constructed from the full-length infectious cDNA clones

201

pCMV-BB/GP5s and pCMV-BB34s/GP5 by the site-directed mutagenesis described

202

above, respectively (Fig. 1). All the recombinant viruses and the aa mutations were

(New

England

Biolabs,

USA),

thereby

yielding

Ac ce

pt

ed

M

an

us

cr

ip t

endonucleases

10

Page 10 of 39

203

summarized in Table 1.

204

2.6. Rescue of recombinant viruses Plasmids carrying a full-length PRRSV cDNA were individually transfected into

206

MARC-145 cells using Lipofectamine 2000 (Invitrogen) according to the

207

manufacturer’s instructions. Four days after transfection, the rescues of infectious

208

viruses were obtained and cloned by the plaque assay. The mutations in the rescued

209

viruses were confirmed by RT-PCR and sequencing.

210

2.7. Viral plaque assay

an

us

cr

ip t

205

MARC-145 cells in 12-well plates were inoculated with 100 μl of tenfold serially

212

diluted PRRSV. After 1 h adsorption at 37°C, cell monolayers were washed with

213

phosphate-buffered saline (PBS) and overlaid with 1% low melting agarose in DMEM

214

(Invitrogen) containing 2% FBS. After the gel overlay solidified, the plates were

215

inverted (top side down) and placed into an incubator at 37°C with 5% CO2. At 4 days

216

post-infection (dpi), plaques were visualized by crystal violet staining.

217

2.8. Antibody binding analyses

ed

pt

Ac ce

218

M

211

Parent and recombinant mutant PRRSVs were purified by ultracentrifugation and

219

diluted in coating buffer to a final concentration 5 g/ml. The antigens were added in

220

triplicate to 96-well flat-bottomed enzyme-linked immunosorbent assay (ELISA)

221

plates. After incubation at 4°C for at least 16 h, wells were blocked with PBS-5% FBS

222

for 1 h at room temperature. A non-saturating concentration of the serum antibody

223

(diluted 1:100), which was in the linear portion of the antibody titration curve 11

Page 11 of 39

determined previously, was added to each well, serially diluted twofold, and incubated

225

with the virus-coated plates for 1 h at room temperature. After washing, anti-pig

226

horseradish peroxidase (HRP)-conjugated antibody was added to the plates, and they

227

were incubated for 1 h at room temperature. After another round of washing,

228

3,3’,5,5’-tetramethylbenzidine (TMB) substrate (KPL Biomedical, Gaithersburg, MD)

229

was added, and 2 M H2SO4 was used to stop the reaction. The amount of HRP product

230

was determined using a plate reader at 450 nm. The control plates were coated with

231

ultracentrifuged non-infected MARC-145 cell lysates.

232

2.9. Western blot

an

us

cr

ip t

224

In order to evaluate the consistence of PRRSV antigens binding in the wells, the

234

antigens were lysed from the coated 96-well ELISA plate by using RIPA Lysis Buffer

235

(Beyotime, China). Briefly, 100μl lysis buffer was added into one coated well. After 5

236

minutes, the lysate samples were obtained and added to another coated well. A total of

237

12 wells (for one strain antigen) were lysed and obtained as one sample. Then all the

238

PRRSV strains antigen samples from the coated plate and the original purified

239

antigens were separated by 12% SDS-PAGE, and transferred onto nitrocellulose filter

240

membrane (PALL, New York, USA). The membranes were incubated with mAb N

241

(made in our laboratory) or GP5 (provided by Dr GZ. Tong, Shanghai Veterinary

242

Research Institute, China) as the primary antibody. After the membranes were rinsed

243

with PBS, the membrane was treated with goat anti-mouse IgG-HRP (BOSTER,

244

China) as the secondary antibody. The proteins were visualized by scanning the

245

membranes with the Tanon 5200 chemiluminescence imaging system (Tanon, China).

Ac ce

pt

ed

M

233

12

Page 12 of 39

246

2.10. Growth curves of viruses To determine viral one-step growth curves, PRRSV mutants (105 TCID50) were

248

inoculated into sub-confluent MARC-145 cells or PAMs in six-well plates. Then, 200

249

μl of the supernatants of the infected cells was collected and replenished with the

250

same volume of fresh medium at 12, 24, 36, 48, and 72 h post-infection (hpi), and

251

stored at −70 °C for virus titration. The virus titers for each time point were

252

determined in MARC-145 cells by TCID50.

253

2.11. Statistical analysis

an

us

cr

ip t

247

All data were analyzed using GraphPad Prism (Version 5.03, San Diego,

255

California) software. Differences among all groups were examined using one-way

256

analysis of variance (ANOVA), followed by Tukey’s tests. Differences between two

257

groups were assessed using unpaired two-tailed t-tests. Differences were considered

258

significant if P was < 0.05.

259

3. Results

260

3.1. Generation of resistant variants against NAb to PRRSV

ed

pt

Ac ce

261

M

254

Five different NAb serums were prepared from the PRRSV-free piglets infected

262

with PRRSV strain BB. The NA titers of the serums against this strain were 1:34-1:48.

263

We used the N4 NAb serum that had the highest NA titer (1:48) to generate the

264

resistant variants from the parental virus BB.

265

After 5-34 passages or purification of PRRSV BB in MARC-145 cells in the

13

Page 13 of 39

presence of N4 NAb to PRRSV, five resistant strains BB5s, BB10s, BB20s, BB30s

267

and BB34s, which could replicated in MARC-145 under pressure of NAb to PRRSV,

268

were obtained. As shown in Fig. 2, the plaque morphologies of BB30s and BB34s

269

were larger than that of the parental strain BB in size (Fig. 2A). Neutralization assays

270

results showed that BB10s and BB20s had a similar NA titers that were significantly

271

lower against that of BB (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.

Porcine reproductive and respiratory syndrome virus (PRRSV) is mainly responsible for the heavy economic losses in pig industry in the world. A number...
466KB Sizes 2 Downloads 11 Views