JGV Papers in Press. Published June 5, 2015 as doi:10.1099/vir.0.000213

Journal of General Virology In vitro Growth, Pathogenicity, and Serological Characteristics of the Japanese Encephalitis Virus Genotype V Muar strain. --Manuscript Draft-Manuscript Number:

VIR-D-15-00245R1

Full Title:

In vitro Growth, Pathogenicity, and Serological Characteristics of the Japanese Encephalitis Virus Genotype V Muar strain.

Short Title:

Characterization of the Japanese Encephalitis Virus Genotype V Muar strain.

Article Type:

Standard

Section/Category:

Animal - Positive-strand RNA Viruses

Corresponding Author:

Shigeru Tajima, Ph.D. National Institute of Infectious Diseases Shinjuku, Tokyo JAPAN

First Author:

Shigeru Tajima, Ph.D.

Order of Authors:

Shigeru Tajima, Ph.D. Kazumi Yagasaki Akira Kotaki Takumi Tomikawa Eri Nakayama Meng Ling Moi Chang-Kweng Lim Masayuki Saijo Ichiro Kurane Tomohiko Takasaki

Abstract:

The characteristics of the genotype V Japanese encephalitis virus (GV JEV) remain poorly understood since only two strains have been isolated, thus far. In this study, we examined the effects of the GV JEV Muar strain on in vitro growth and pathogenicity in mice; we also evaluated the efficacy of inactivated Japanese encephalitis (JE) vaccines against the Muar strain. Although the growth of the Muar strain in mouse neuroblastoma N18 cells was clearly worse than those of the GIII Beijing-1 and GI Mie/41/2002 strains, neuroinvasiveness of the Muar strain was similar to that of the Beijing-1 strain and significantly higher than that of the Mie/41/2002 strain. The results of a plaque reduction neutralization test suggest that the neutralization ability of the JE vaccines against the Muar strain is reduced compared with GI and GIII strains. However, the protection potency of the JE vaccine against the Muar strain was similar to that for the Beijing-1 strain in mice. Our data indicate that GV JEV has unique growth, virulence, and antigenicity features.

Powered by Editorial Manager® and ProduXion Manager® from Aries Systems Corporation

Manuscript Including References (Word document) Click here to download Manuscript Including References (Word document): Text(PRNT)150603JGV.docx

1

In vitro Growth, Pathogenicity, and Serological Characteristics of the Japanese

2

Encephalitis Virus Genotype V Muar strain.

3 4

Shigeru Tajima*, Kazumi Yagasaki, Akira Kotaki, Takumi Tomikawa, Eri Nakayama, Meng

5

Ling Moi, Chang-Kweng Lim, Masayuki Saijo, Ichiro Kurane and Tomohiko Takasaki.

6 7

Department of Virology I, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku,

8

Tokyo 162-8640, Japan

9 10

Key words:

11

Japanese encephalitis virus / Genotype / Pathogenicity / Neutralization / Vaccine

12 13

Correspondence: Shigeru Tajima, Ph.D.

14

Department of Virology 1, National Institute of Infectious Diseases

15

1-23-1 Toyama, Shinjuku, Tokyo 162-8640, Japan

16

E-mail: [email protected]

17

Phone/Fax: +81-3-5285-1188

18 19 20

Footnote

21

The GenBank accession numbers for the JEV E gene sequence we determined in this study are as

22

follow: Nakayama strain, AB920348; Beijing-1 strain, AB920347; Kumamoto/65/2005 strain,

23

AB920445; Kumamoto/81/2006 strain, AB920446; Kumamoto/104/2006 strain, AB920447;

24

Chiba/150/2007 strain, AB920448; Chiba/103/2008, AB920449.

25

Summary

26

The characteristics of the genotype V Japanese encephalitis virus (GV JEV) remain poorly

27

understood since only two strains have been isolated, thus far. In this study, we examined the

28

effects of the GV JEV Muar strain on in vitro growth and pathogenicity in mice; we also

29

evaluated the efficacy of inactivated Japanese encephalitis (JE) vaccines against the Muar strain.

30

Although the growth of the Muar strain in mouse neuroblastoma N18 cells was clearly worse

31

than those of the GIII Beijing-1 and GI Mie/41/2002 strains, neuroinvasiveness of the Muar

32

strain was similar to that of the Beijing-1 strain and significantly higher than that of the

33

Mie/41/2002 strain. The results of a plaque reduction neutralization test suggest that the

34

neutralization ability of the JE vaccines against the Muar strain is reduced compared with GI and

35

GIII strains. However, the protection potency of the JE vaccine against the Muar strain was

36

similar to that for the Beijing-1 strain in mice. Our data indicate that GV JEV has unique growth,

37

virulence, and antigenicity features.

38 39

40

Introduction

41

Japanese encephalitis (JE), resulting from an infection of the Japanese encephalitis virus

42

(JEV), is a significant public health problem in many Asian countries. JEV causes serious

43

nervous disorders such as meningitis and encephalitis. There are an estimated 68,000 cases of JE

44

per year, occurring mainly in China, India, and Southeast Asian countries, which result in 15,000

45

fatalities, mostly in children (Campbell et al., 2011; Erlanger et al., 2009; Tsai, 2000). There is

46

no specific treatment available for JE, other than preventive vaccination with JE vaccines. In

47

Japan, the number of reported JE cases has dramatically decreased since the late 1960s, and less

48

than 10 JE cases have occurred in Japan over the past two decades (Arai et al., 2008). This

49

decrease is thought to be due to active vaccination, a decrease in the number of mosquitoes that

50

can transmit JEV, and urbanization. However, a study on the prevalence of JEV that used

51

domestic pigs indicated that, in most regions of Japan, naive pigs are still seroconverted to JEV

52

every year, which suggests that JEV is still a serious threat (Arai et al., 2008).

53

JEV belongs to the genus Flavivirus in the family Flaviviridae and is amplified in a bird

54

and pig-mosquito transmission cycle (Pierson, 2013). The infected mosquitoes, mainly Culex

55

tritaeniorhynchus, transmit JEV to dead-end hosts: either humans or horses. JEV has a single-

56

stranded positive-sense RNA genome, which encodes three structural proteins (C, prM, and E)

57

and seven non-structural proteins (NS1, NS2A, NS2B, NS3, NS4A, NS4B, and NS5). JEV is

58

classified into five genotypes (GI - GV) based on its genomic sequence (Solomon et al., 2003),

59

of which the GIII strain was the most widely distributed and most frequently isolated in JE

60

endemic areas until the 1990s. Consequently, all licensed inactivated and live-attenuated JE

61

vaccines are derived from GIII JEV. However, the major genotype of JEV isolated in Japan

62

changed from GIII to GI strain in the early 1990s (Ma et al., 2003; Yoshida et al., 2005). In

63

recent years, a similar genotype shift has been observed in South Korea, northern Vietnam,

64

China, Taiwan, and Thailand (Chen et al., 2011; Nam et al., 1996; Nga et al., 2004; Nitatpattana

65

et al., 2008; Pan et al., 2011; Wang et al., 2007; Yang et al., 2004; Yun et al., 2010; Zhang et al.,

66

2011; Zhang et al., 2009). GI JEV has also been found in India since 2009 (Fulmali et al., 2011).

67

Thus, the dominant genotype in JEV endemic areas is now GI, although the reasons for the broad

68

shift from GIII to GI remain unclear. The Indonesian/Malaysian region is thought to be the origin

69

of all genotypes of JEV (Solomon et al., 2003), and in several Southeast Asian countries, GI was

70

isolated as a minor JEV strain before the 1990s. Comprehensive phylogenetic analysis of the GI

71

JEV strains demonstrates that the old and the new GI strains form distinct lineages (Gao et al.,

72

2013; Schuh et al., 2014), suggesting that genetic differences in the new GI genome might be

73

responsible for the spread of GI viruses. Recent findings indicate that GI JEV exhibits a higher

74

replicative ability in mosquitoes than does GIII JEV (Schuh et al., 2014).

75

A GV strain of JEV was first isolated from an encephalitis patient in Malaysia in 1952

76

(also referred to as the Muar strain), and it remained the only known instance of this genotype for

77

over 50 years (Hasegawa et al., 1994; Solomon et al., 2003). Recently, the GV JEV genome has

78

been identified in Culex mosquitoes in China in 2009 and in South Korea in 2010 and 2012 (Kim

79

et al., 2015; Li et al., 2011; Takhampunya et al., 2011). A group in China succeeded in isolating

80

a new GV strain (referred to as the XZ0934 strain) from C. tritaeniorhynchus (Li et al., 2011). In

81

South Korea, genomes of GV JEV were detected in Culex bitaeniorhynchus, Culex orientalis,

82

and Culex pipiens, but none were identified in the major JEV vector C. tritaeniorhynchus (Kim

83

et al., 2015; Takhampunya et al., 2011). These findings raise the possibility that the other species

84

of Culex mosquitoes may be involved in the natural transmission cycle of the GV strain of JEV.

85

Thus, GV JEV may also be emerging in other endemic areas, and more attention needs to be

86

focused on investigating the dynamics of circulating JEV strains in these areas.

87

Since only two GV strains, Muar and XZ0934, have been isolated thus far and biological

88

studies on the features of GV JEV have been very limited, the growth and pathogenic properties

89

of GV remain poorly understood. The spread of JEV strains, other than GIII, into endemic areas

90

makes it imperative that systematic evaluation is conducted on the protective efficacy of current

91

GIII-derived JE vaccines against other JEV genotypes. Previous reports call attention to the

92

reduced neutralizing potency of inactivated and live-attenuated GIII JE vaccines against

93

circulating GI JEV (Fan et al., 2013; Fan et al., 2012). On the other hand, other researchers have

94

shown that inactivated and live-attenuated JE vaccines are effective against both GIII and GI

95

JEV strains that are prevalent in China (Liu et al., 2011), and inactivated vaccines used for

96

European travelers induce protective levels of neutralizing antibodies against GI - GIV JEV

97

(Erra et al., 2013a; b). Thus, the efficacy of the current JE vaccines against heterologous

98

genotypes remains unresolved. Furthermore, these studies did not assess the Beijing-1 strain-

99

derived vaccine that is currently available in Japan, the Nakayama strain-derived vaccine, or a

100

GV JEV strain as a target virus.

101

To uncover the characteristics of GV JEV, we investigated in vitro growth and

102

pathogenicity in mice for the GV JEV Muar strain. We also evaluated the protective efficacy of

103

the inactivated JE vaccines against the Muar strain as well as several GI and GIII JEV strains.

104 105 106 107 108

109

Results

110

In vitro growth properties of the Muar strain

111

To characterize the growth of the GV Muar strain in vitro, we first compared the plaques

112

induced by the Muar strain to those of GI Mie/41/2002 strain and GIII Beijing-1 strains in Vero

113

cells (Fig. 1A). The plaque size induced by Muar (average diameter and SEM: 0.98±0.03 mm)

114

was less than that of Mie/41/2002 (1.89±0.05 mm; P < 0.001) but was similar to that of Beijing-

115

1 (1.02±0.03 mm; P = 0.37). Similar results were obtained using a replication kinetics assay for

116

these viruses in Vero cells and BHK-21 cells (Fig. 1B and C). These results suggest that the

117

growth of Muar is similar to that of Beijing-1 in interferon response-deficient cells. Our previous

118

report showed that, in contrast to the results in Vero cells, the steady state number of infectious

119

particles for Beijing-1 was apparently higher than that of Mie/41/2002 in mouse neuroblastoma-

120

derived N18 cells (Tajima et al., 2010). We next examined the growth properties of Muar in N18

121

cells (Fig. 1D). However, the growth rate of Muar in N18 cells was slower than the other strains,

122

and the infectious particles of Muar at steady state were 100-fold and 10-fold less than those of

123

Beijing-1 and Mie/41/2002, respectively. These data indicate that Muar might exhibit a reduced

124

amplification in N18 cells, and the characteristics of the Muar strain are clearly different from

125

those of the Beijing-1 and Mie/41/2002 strains in vitro. Using real-time RT-PCR, we also

126

confirmed in all three cultured cells that the quantity of viral genomes in the supernatant and in

127

the cells infected the viruses corresponded to those of the virus titers (data not shown), indicating

128

that the reduced growth of Muar in cultured cells does not reflect defective particle assembly and

129

secretion.

130 131

Virulence of the Muar strain in mice

132

Next, we examined the pathogenicity of the Muar strain in mice. Mice were infected

133

intraperitoneally (i.p.) with Muar, Beijing-1, or Mie/41/2002 and observed for 3 weeks (Table 1).

134

Our previous report using the Beijing-1 and Mie/41/2002 strains demonstrated that the growth

135

characteristics of some JEV strains in N18 cells were positively correlated with

136

neuroinvasiveness in mice (Tajima et al., 2010); therefore, we speculated that the Muar strain

137

might exhibit a lower virulence in mice compared with the Beijing-1 and Mie/41/2002 strains. In

138

the 103, 104, and 105 plaque forming units (p.f.u.)-infected groups, more than 90% of the mice

139

infected with Muar or Beijing-1 died; however, less than 30% of the mice infected with

140

Mie/41/2002 died. The survival curves for the Muar-infected groups were similar to those of the

141

Beijing-1-infceted groups. These results indicate that, in contrast to the growth kinetics in N18

142

cells, neuroinvasiveness of Muar is equivalent to that of Beijing-1 and significantly higher than

143

that of Mie/41/2002 in mice.

144 145

The neutralizing ability of inactivated JE vaccines against Muar and recombinant

146

intertypic JEVs.

147

We next evaluated the ability of inactivated Nakayama and Beijing-1 vaccines, which

148

were produced in Japan, to induce neutralizing antibodies against Muar. We also used three GIII

149

and 11 GI viruses with differing amino acid sequences in the E protein as target viruses

150

(Supplementary table 1). The E protein is the major structural protein that constitutes the surface

151

structure of the virus particles, and a majority of neutralizing antibodies bind to the E protein and

152

can inhibit viral infection (Wu et al., 2003). Serum samples collected from mice at 14 days after

153

initial immunization with 2-fold-diluted and 8-fold-diluted JE vaccines were used for the plaque

154

reduction neutralization test (PRNT), and the value of 50% reduction relative to the non-serum

155

control (PRNT50) for each virus was determined (Table 2). In the sera of mice that were

156

immunized with 2-fold dilutions, the PRNT titer against GI and GIII viruses was between 1:80

157

and 1:320. In those with 8-fold dilutions, it was between 1:40 and 1:160. We statistically

158

compared the PRNT titers of sera against GI viruses to those against GIII viruses using a

159

parametric Welch’s t-test and a non-parametric Mann-Whitney U-test. No significant differences

160

(P > 0.05) were observed between the GI group and GIII group in all cases, indicating that the

161

neutralizing efficacy of inactivated Nakayama and Beijing-1 vaccines against the GI strains is

162

similar to that for the GIII strains. In contrast to the GI and GIII strains, the PRNT titers in the

163

sera of mice immunized against GV Muar were 4-fold less than those against the GI and GIII

164

strains. These results suggest that the neutralizing capacity of the JE vaccines against GV JEV

165

was considerably lower than that against GI and GIII JEV. We also examined the neutralizing

166

potency (PRNT) of sera obtained from mice at 2 and 4 months after the initial immunization for

167

the 2-fold-diluted and 8-fold-diluted Nakayama and Beijing-1 vaccines (Table 3). Two vaccine

168

strains, two GI strains, three GIII strains, and one GV strain were used as target viruses. The

169

PRNT titers for GV Muar were less than those for the GI and GIII JEV, indicating that the

170

neutralization capacity of the inactivated JE vaccines is relatively weak against the GV Muar

171

strain.

172

A comparison of the E protein amino acid sequences amount the viral genotypes showed a

173

higher homology between GI and GIII than that between GI and GV or that between GIII and

174

GV (data not shown). There were 40 residues (8.0%) that differed between Beijing-1 and Muar,

175

and 44 residues (8.8%) were different between Nakayama and Muar. In contrast, less than 12

176

residues (2.4%) were different between the vaccine strains and the GI strains, and less than 8

177

residues (1.6%) were different between the vaccine strains and the GIII strains (Supplementary

178

table 2 and 3). Other GV strains, XZ0934 and 10-1827, also showed similar homology to the

179

vaccine strains, and only two residues (0.4%) were different between the XZ0934 and 10-1827

180

strains. These findings raise the possibility that the neutralization potency of the inactivated

181

vaccines against the other GV strains is also relatively weak. To examine the efficacy of the

182

vaccines against GV strains other than Muar, we generated recombinant intertypic JEVs rJEV-

183

EMuar-M41 and rJEV-EXZ0934-M41, which contain the majority of the E region of Muar and

184

XZ0934, respectively, in the backbone of the Mie/41/2002 genome. We also generated rJEV-

185

ENakayama-M41 and rJEV-EBeijing-1-M41 as control viruses for use in the neutralization assay

186

(Tajima et al., 2010). The PRNT titers in the sera from mice immunized with the vaccines

187

against rJEV-EMuar-M41 and rJEV-EXZ0934-M41 were clearly less than those against the parent

188

virus Mie/41/2002, rJEV-ENakayama-M41, and rJEV-EBeijing-1-M41 (Table 4). These results

189

indicate that the neutralization ability of the vaccines may be weak against both Muar and other

190

GV JEV strains.

191 192

Protection efficacy of the JE vaccines against Muar infection in mice

193

To examine whether immunization of mice with the inactivated JE vaccines inhibits the

194

onset of Muar-induced disease, mice that were vaccinated with the inactivated Beijing-1 vaccine

195

were inoculated i.p. with 104 PFU of Muar or Beijing-1 strains (Table 5). Mice vaccinated with

196

2-fold diluted vaccine had a 100% survival rate after inoculation with Beijing-1 or Muar.

197

Fourteen of 16 mice (87.5%) and 12 of 16 mice (75%) that were vaccinated with 32-fold diluted

198

vaccine survived after infection with Beijing-1 or Muar, respectively. There was no significant

199

difference in the survival rates between Beijing-1-infected and Muar-infected groups. These

200

results suggest that the protection efficacy of the Beijing-1 vaccine against infection with Muar is

201

similar to that against infection with Beijing-1 in vivo.

202 203

The neutralizing ability of the sera from Muar-infected mice against various JEVs

204

Finally, we assessed the ability of GV Muar-infected mouse sera to block the infection of

205

GI and GIII JEVs in vitro using the neutralization assay (Table 6). The PRNT titer against Muar

206

was 2- or 4-fold higher than those against two GI, three GIII, and Beijing-1 strains, though the

207

titer against Nakayama was equivalent to that against Muar. Thus, we confirmed that sera from

208

Muar-infected mice were able to neutralize the infection of GI and GIII strains; however, the

209

neutralization potency of these sera against GI and GIII viruses was weaker than that against

210

Muar.

211 212

213

Discussion

214

To date, only two GV JEV strains have been isolated and amplified in vitro. Therefore, the

215

characteristics of GV JEV remains poorly understood. In this study, we examined the in vitro

216

growth properties and in vivo pathogenicity of the GV strain Muar. Furthermore, we investigated

217

the potential of inactivated JE vaccines that are currently available in Japan to induce a

218

protective immune response against GV JEV using mouse models. Our results indicate that GV

219

JEV has different growth, pathogenic, and antigenic characteristics compared with GI and GIII

220

JEV.

221

Our previous report suggested that there is a correlation between the growth characteristics

222

of JEV in mouse neuroblastoma N18 cells and virulence in mice (Tajima et al., 2010). The

223

growth rate of Muar was less than that of the highly virulent GIII Beijing-1 strain and the

224

moderately virulent GI Mie/41/2002 strain in N18 cells; however, the neuroinvasiveness of Muar

225

was similar to that of Beijing-1. These results suggest that the growth properties of JEV in N18

226

cells do not necessarily reflect pathogenicity in mice. There is little information regarding the

227

regions of the Muar genome that are associated with virulence in mice. We previously

228

demonstrated that the E protein contributes to the difference in virulence between Mie/41/2002

229

and Beijing-1 (Tajima et al., 2010). Other reports have also highlighted the importance of the E

230

protein as a determinant of pathogenicity (Monath et al., 2002; Sumiyoshi et al., 1995; Zhao et

231

al., 2005). On the other hand, our group determined that an amino acid substitution in the JEV

232

NS4A protein alters virulence in mice (Yamaguchi et al., 2011). To clarify the regions

233

responsible for increased pathogenicity in the Muar genome, various GI-GV intertypic and point

234

mutant viruses may be required. Recently, Ishikawa et al. identified the infectious molecular

235

clone and reporter replicon of the Muar strain, which is also beneficial for generally

236

understanding the characteristics of GV JEV (Ishikawa et al., 2015). In addition, de Wispelaere

237

et al. constructed a molecular clone of the GV XZ0934 JEV strain and demonstrated that this

238

recombinant GV virus exhibited a greater pathogenicity in BALB/c mice than that of GIII JEV

239

(de Wispelaere et al., 2015).

240

The neutralization tests using the vaccinated mouse sera indicated that both the Nakayama

241

and Beijing-1 vaccines induce sufficient neutralizing antibodies against GI JEV. However, these

242

vaccines induce fewer neutralizing antibodies against GV Muar or the recombinant intertypic

243

JEVs, which contain the E protein from the GV Muar or XZ0934 strains. Furthermore, the sera

244

from Muar-infected mice had a lower neutralization titer against GI and GIII viruses compared

245

with the Muar strain. These data indicate that GV JEV may be distinct from GI and GIII JEV in

246

antigenicity. The E protein sequences between GI and GIII JEV differ by approximately 3%

247

(Solomon et al., 2003). However, this number increases to 8.8 - 9.2% between GV strains and

248

the Nakayama strain, and 8.2 - 8.6% between GV strains and the Beijing-1 strain

249

(Supplementary table 2 and 3). The decrease in homology between the vaccine strains and the

250

GV strains might be responsible for the observed weak efficacy of the vaccines against the Muar

251

strain and recombinant JEVs that contain GV E proteins. The amino acid differences between the

252

vaccine strains and the GI strains did not exceed 2.4%, indicating that the difference may not

253

influence the efficacy of the vaccines. Previous studies have identified individual residues within

254

the E protein that are related to the recognition of JEV by neutralizing antibodies (Crill & Chang,

255

2004; Goncalvez et al., 2008; Kobayashi et al., 1985; Morita et al., 2001; Vogt et al., 2009; Wu

256

et al., 2003). Although the sequence variation between the vaccine strains and GV strains do not

257

correspond to epitope residues, only a limited number of residues could be responsible for the

258

decreased neutralization ability of the vaccines against GV strains. All inactivated and live-

259

attenuated JE vaccines that are currently available are derived from GIII strains. Our results

260

indicate that live-attenuated GIII-derived vaccines may have a reduced potency in inducing

261

neutralizing antibodies against GV JEV. In this report, we used vaccinated mouse sera for all

262

neutralization analyses. It is generally accepted that neutralizing antibodies with a PRNT of 1:10

263

or greater are sufficient to protect from the onset of JE (Kitano, 1996; Kurane & Takasaki, 2000).

264

Further studies using human sera from JE vaccine-immunized healthy and JE case individuals

265

will be needed in order to validate our findings in humans.

266

In contrast to the results of the neutralization tests, the challenge analysis in mice

267

immunized with the inactivated Beijing-1 vaccine showed no clear difference in mortality

268

between Beijing-1-infected and Muar-infected groups for either vaccine concentration. This

269

indicates that the ability of the Beijng-1 vaccine to block the onset of encephalitis by Muar might

270

be comparable to that by Beijing-1 in vivo. Our results indicated that the PRNT titers of 2-fold

271

and 8-fold diluted Beijing-1 vaccine against Muar were 1:20 and 1:10, respectively, which were

272

16-fold less than those against Beijing-1 (1:320 and 1:160, respectively) (Table 2). These

273

findings suggest that although the neutralization potency of the Beijing-1 vaccine against GV

274

JEV is weaker than those against GI and GIII JEV, the potency of the vaccine is not critical to

275

inhibit disease caused by GV JEV, as long as the titer exceeds a threshold of 1:10. We inoculated

276

104 PFU of Muar and Beijing-1 per 5-week-old mouse in the protection assay as this virus titer

277

was sufficient to kill all 3-week-old mice, as shown in Table 1. However, only 50% of the mice

278

were deceased in the non-vaccinated Beijing-1 inoculated group, while over 90% of mice were

279

deceased in the non-vaccinated Muar inoculated group. It is possible that Muar may exhibit a

280

modestly increased virulence compared with Beijing-1 when 5-week-old mice are used for the

281

evaluation of neuroinvasiveness.

282

For more than 50 years, the Muar strain was the only example of GV. Recently, several

283

GV genomes were detected in Culex mosquitoes in China and South Korea (Kim et al., 2015; Li

284

et al., 2011; Takhampunya et al., 2011), but there has been no GV JEV detected in Japan, thus

285

far. However, in both Japan and South Korea, a GIII-to-GI shift occurred in the early 1990s,

286

raising the possibility that GV JEV has already invaded Japan. Recently, Kim et al. suggested

287

that two new vector mosquito species might play a role in the transmission of GV JEV (Kim et

288

al., 2015). Continuous and careful monitoring of the spread of GV viruses must be undertaken.

289

In South Korea, 26 JE patients were reported in 2010, representing a 12-fold increase compared

290

with the mean number that was reported over the previous 26 years (Seo et al., 2013). The

291

relationship between the increase in the number of JE patients in South Korea in 2010 and the

292

emergence of GV JEV remains unknown. Continuous isolation of JEV from JE patients,

293

mosquitoes, and other amplifying hosts is important to understand the effects of the GV

294

emergence.

295 296 297

298

Methods

299

Cells, virus strains, and genetic analysis

300

Vero cells (strain 9013) and BHK-21 cells were maintained at 37 C with 5% CO2 in

301

Eagle’s Minimal Essential Medium supplemented with 10% heat-inactivated fetal bovine serum

302

and 100 U Penicillin-Streptomycin mL-1. JEV strains used in this study are listed in

303

Supplementary table S1. Briefly, we used two JE vaccine strains (Nakayama and Beijing-1),

304

eleven GI strains, three GIII strains, and one GV strain. All of the GI and GIII strains were

305

isolated in Japan, while the GV Muar strain was isolated from a patient with encephalitis in

306

Malaysia in 1952. Working virus stocks were prepared in Vero cells. The nucleotide sequence of

307

the E region containing 1,500 bp were determined for all strains used in this study, including the

308

two vaccine strains (Nakayama and Beijing-1). These nucleotide sequences and their

309

corresponding amino acid sequences were subjected to sequence alignments using GENETYX

310

Ver. 11 genetic software (Genetyx Corporation, Japan).

311 312

Analysis of growth kinetics and plaque morphology

313

The growth properties of the JEV strains were analyzed as described previously (Tajima et

314

al., 2010). Briefly, cells were plated into 6-well culture plates and infected with the JEV strains

315

at a multiplicity of infection of 0.01 p.f.u./cell. Aliquots of the media were removed periodically

316

and their viral titers were determined using a plaque assay on Vero cells. To evaluate the plaque

317

size, Vero cells were plated in 12-well plates and inoculated with the viruses. Four days after

318

inoculation, cells were fixed using a 10% formalin-PBS solution and subsequently stained with

319

methylene blue. The diameter of 20 plaques was measured and the mean plaque size in mm and

320

standard error (SEM) was calculated.

321 322

Production of recombinant JEV

323

Four recombinant molecular clones, rJEV-EBeijing-1-M41/pMW119, rJEV-ENakayama-

324

M41/pMW119, rJEV-EMuar-M41/pMW119, and rJEV-EXZ0934-M41/pMW119, were constructed

325

in the GI JEV strain Mie/41/2002 backbone as described previously (Tajima et al., 2010). Briefly,

326

the E region, from position 927 to 2410, of rJEV(Mie/41/2002)/pMW119 was replaced with the

327

corresponding region of the Beijing-1, Nakayama, Muar, or XZ0934 strain. The recombinant

328

viruses were recovered by transfection of in vitro-transcribed recombinant viral RNA into Vero

329

cells, as previously described (Tajima et al., 2010).

330 331

Mouse vaccination

332

Mouse experiments were performed in accordance with the “Guidelines for animal

333

experiments performed at National Institute of Infectious Diseases” published by the Animal

334

Welfare and Animal Care Committee of the National Institute of Infectious Diseases, Japan.

335

Mouse brain-derived inactivated JE Nakayama vaccine (Biken: lot no. 101-2004) and Vero cell-

336

derived inactivated JE Beijing-1 vaccine (Biken: lot no. 106VC-2009) were used for

337

immunization of the mice. Both vaccines were diluted 1:2 and 1:8 with PBS. Groups of inbred

338

ddY mice (4 weeks old, n=30; Japan SLC) were vaccinated i.p. with 0.5 mL of the diluted

339

vaccines and were immunized again one week after the initial immunization. The immunized

340

mice were sacrificed at 14 days, 2 months, or 4 months after the initial immunization, and their

341

sera were pooled independently (10 mice/pool) for each period. These sera were used to measure

342

neutralization antibody titers as described below.

343

344

Mouse challenge test

345

For evaluation of neuroinvasiveness, groups of ddY mice (3 weeks old, n=10) were

346

inoculated i.p. with 100 l of serially-diluted recombinant virus in 0.9% NaCl solution. The mice

347

were observed for 3 weeks after inoculation to determine survival rates. Survival curve

348

comparisons were performed using Prism software (GraphPad software) statistical analysis with

349

a log-rank (Mantel-Cox) test. To evaluate the protection efficacy of the vaccines, groups of ddY

350

mice (3 weeks old, n=14 or 15) were vaccinated i.p. with 0.5 mL of the inactivated Beijing-1

351

vaccine diluted 1:2 or 1:32 and were immunized again one week after the initial immunization.

352

The immunized mice were inoculated i.p. with 100 l of 104 p.f.u. Beijing-1 and Muar strains.

353

The mice were observed for 3 weeks after inoculation to determine survival rates, and survival

354

curve comparisons were performed as described above. To obtain anti-Muar antisera, ddY mice

355

(3 weeks old, n=4) were inoculated i.p. with 100 l of 10 p.f.u. Muar and were inoculated

356

subsequently with 100 l of 102 p.f.u. Muar at 11 days after the initial inoculation. The Muar-

357

infected mice were sacrificed 22 days after the first inoculation to collect pooled antisera, and the

358

sera were used for neutralization tests.

359 360

Plaque reduction neutralization test (PRNT)

361

Neutralizing antibodies to JEV were measured using the PRNT method. Each JEV strain

362

was combined at a 1:1 ratio with 2-fold serial dilutions (1:10 to 1:640) of the sera from mice

363

immunized with the Nakayama vaccine or the Beijing-1 vaccine and then incubated at 35 C for

364

90 min. Vero cell monolayers were inoculated with these mixtures in 12-well plates and

365

incubated at 35 C for 90 min. Subsequently, overlay medium containing 1% methylcellulose

366

was added and cells were incubated at 35 C for 4 to 6 days. The cells were fixed using a 10%

367

formalin-PBS solution and stained using methylene blue. The PRNT titer was defined as the

368

reciprocal of the highest dilution that resulted in 50% (PRNT50) reduction relative to the non-

369

serum control.

370 371

Acknowledgements

372

This work was supported by a Health and Labour Sciences Research Grant for Research on

373

Emerging and Re-emerging Infectious Diseases by the Ministry of Health, Labor, and Welfare in

374

Japan (H24-Shinkou-007), a Grant-in-Aid for Scientific Research (C) from the JSPS (25460577),

375

and a grant from the Japan Foundation for Pediatric Research (13-008).

376 377

The authors declare that they have no conflict of interest.

378 379

References

380

Arai, S., Matsunaga, Y., Takasaki, T., Tanaka-Taya, K., Taniguchi, K., Okabe, N., Kurane,

381

I. & Vaccine Preventable Diseases Surveillance Program of, J. (2008). Japanese

382

encephalitis: surveillance and elimination effort in Japan from 1982 to 2004. Jpn J Infect

383

Dis 61, 333-338.

384

Campbell, G. L., Hills, S. L., Fischer, M., Jacobson, J. A., Hoke, C. H., Hombach, J. M.,

385

Marfin, A. A., Solomon, T., Tsai, T. F., Tsu, V. D. & Ginsburg, A. S. (2011).

386

Estimated global incidence of Japanese encephalitis: a systematic review. Bull World

387

Health Organ 89, 766-774, 774A-774E.

388

Chen, Y. Y., Fan, Y. C., Tu, W. C., Chang, R. Y., Shih, C. C., Lu, I. H., Chien, M. S., Lee,

389

W. C., Chen, T. H., Chang, G. J. & Chiou, S. S. (2011). Japanese encephalitis virus

390

genotype replacement, Taiwan, 2009-2010. Emerg Infect Dis 17, 2354-2356.

391 392

Crill, W. D. & Chang, G. J. (2004). Localization and characterization of flavivirus envelope glycoprotein cross-reactive epitopes. Journal of virology 78, 13975-13986.

393

de Wispelaere, M., Frenkiel, M. P. & Despres, P. (2015). A Japanese Encephalitis virus

394

genotype 5 molecular clone is highly neuropathogenic in a mouse model: implication of

395

the structural proteins region in virulence. Journal of virology.

396 397 398

Erlanger, T. E., Weiss, S., Keiser, J., Utzinger, J. & Wiedenmayer, K. (2009). Past, present, and future of Japanese encephalitis. Emerg Infect Dis 15, 1-7. Erra, E. O., Askling, H. H., Yoksan, S., Rombo, L., Riutta, J., Vene, S., Lindquist, L.,

399

Vapalahti, O. & Kantele, A. (2013a). Cross-protection elicited by primary and booster

400

vaccinations against Japanese encephalitis: a two-year follow-up study. Vaccine 32, 119-

401

123.

402

Erra, E. O., Askling, H. H., Yoksan, S., Rombo, L., Riutta, J., Vene, S., Lindquist, L.,

403

Vapalahti, O. & Kantele, A. (2013b). Cross-protective capacity of Japanese encephalitis

404

(JE) vaccines against circulating heterologous JE virus genotypes. Clin Infect Dis 56,

405

267-270.

406

Fan, Y. C., Chen, J. M., Chen, Y. Y., Lin, J. W. & Chiou, S. S. (2013). Reduced neutralizing

407

antibody titer against genotype I virus in swine immunized with a live-attenuated

408

genotype III Japanese encephalitis virus vaccine. Vet Microbiol 163, 248-256.

409

Fan, Y. C., Chen, J. M., Chiu, H. C., Chen, Y. Y., Lin, J. W., Shih, C. C., Chen, C. M.,

410

Chang, C. C., Chang, G. J. & Chiou, S. S. (2012). Partially neutralizing potency

411

against emerging genotype I virus among children received formalin-inactivated Japanese

412

encephalitis virus vaccine. PLoS Negl Trop Dis 6, e1834.

413

Fulmali, P. V., Sapkal, G. N., Athawale, S., Gore, M. M., Mishra, A. C. & Bondre, V. P.

414

(2011). Introduction of Japanese encephalitis virus genotype I, India. Emerg Infect Dis 17,

415

319-321.

416

Gao, X., Liu, H., Wang, H., Fu, S., Guo, Z. & Liang, G. (2013). Southernmost Asia is the

417

source of Japanese encephalitis virus (genotype 1) diversity from which the viruses

418

disperse and evolve throughout Asia. PLoS Negl Trop Dis 7, e2459.

419

Goncalvez, A. P., Chien, C. H., Tubthong, K., Gorshkova, I., Roll, C., Donau, O., Schuck,

420

P., Yoksan, S., Wang, S. D., Purcell, R. H. & Lai, C. J. (2008). Humanized

421

monoclonal antibodies derived from chimpanzee Fabs protect against Japanese

422

encephalitis virus in vitro and in vivo. Journal of virology 82, 7009-7021.

423

Hasegawa, H., Yoshida, M., Fujita, S. & Kobayashi, Y. (1994). Comparison of structural

424

proteins among antigenically different Japanese encephalitis virus strains. Vaccine 12,

425

841-844.

426

Ishikawa, T., Abe, M. & Masuda, M. (2015). Construction of an infectious molecular clone of

427

Japanese encephalitis virus genotype V and its derivative subgenomic replicon capable of

428

expressing a foreign gene. Virus research 195, 153-161.

429

Kim, H., Cha, G. W., Jeong, Y. E., Lee, W. G., Chang, K. S., Roh, J. Y., Yang, S. C., Park,

430

M. Y., Park, C. & Shin, E. H. (2015). Detection of Japanese encephalitis virus genotype

431

V in Culex orientalis and Culex pipiens (Diptera: Culicidae) in Korea. PloS one 10,

432

e0116547.

433

Kitano, T. O., A. (1996). Japanese encephalitis vaccine. In Vaccine handbook, pp. 103-113.

434

Edited by Y. A. Arai, T.; Chino, T.; Katow, S.; Miyamura, T.; Nakamura, R.; Oya, A.;

435

Sato, H. Tokyo: Maruzen.

436

Kobayashi, Y., Hasegawa, H. & Yamauchi, T. (1985). Studies on the antigenic structure of

437

Japanese encephalitis virus using monoclonal antibodies. Microbiol Immunol 29, 1069-

438

1082.

439

Kurane, I. & Takasaki, T. (2000). Immunogenicity and protective efficacy of the current

440

inactivated Japanese encephalitis vaccine against different Japanese encephalitis virus

441

strains. Vaccine 18 Suppl 2, 33-35.

442

Li, M. H., Fu, S. H., Chen, W. X., Wang, H. Y., Guo, Y. H., Liu, Q. Y., Li, Y. X., Luo, H. M.,

443

Da, W., Duo Ji, D. Z., Ye, X. M. & Liang, G. D. (2011). Genotype v Japanese

444

encephalitis virus is emerging. PLoS Negl Trop Dis 5, e1231.

445

Liu, X., Yu, Y., Li, M., Liang, G., Wang, H., Jia, L. & Dong, G. (2011). Study on the

446

protective efficacy of SA14-14-2 attenuated Japanese encephalitis against different JE

447

virus isolates circulating in China. Vaccine 29, 2127-2130.

448

Ma, S. P., Yoshida, Y., Makino, Y., Tadano, M., Ono, T. & Ogawa, M. (2003). Short report:

449

a major genotype of Japanese encephalitis virus currently circulating in Japan. Am J Trop

450

Med Hyg 69, 151-154.

451

Monath, T. P., Arroyo, J., Levenbook, I., Zhang, Z. X., Catalan, J., Draper, K. &

452

Guirakhoo, F. (2002). Single mutation in the flavivirus envelope protein hinge region

453

increases neurovirulence for mice and monkeys but decreases viscerotropism for

454

monkeys: relevance to development and safety testing of live, attenuated vaccines.

455

Journal of virology 76, 1932-1943.

456

Morita, K., Tadano, M., Nakaji, S., Kosai, K., Mathenge, E. G., Pandey, B. D., Hasebe, F.,

457

Inoue, S. & Igarashi, A. (2001). Locus of a virus neutralization epitope on the Japanese

458

encephalitis virus envelope protein determined by use of long PCR-based region-specific

459

random mutagenesis. Virology 287, 417-426.

460

Nam, J. H., Chung, Y. J., Ban, S. J., Kim, E. J., Park, Y. K. & Cho, H. W. (1996). Envelope

461

gene sequence variation among Japanese encephalitis viruses isolated in Korea. Acta

462

Virol 40, 303-309.

463

Nga, P. T., del Carmen Parquet, M., Cuong, V. D., Ma, S. P., Hasebe, F., Inoue, S., Makino,

464

Y., Takagi, M., Nam, V. S. & Morita, K. (2004). Shift in Japanese encephalitis virus

465

(JEV) genotype circulating in northern Vietnam: implications for frequent introductions

466

of JEV from Southeast Asia to East Asia. The Journal of general virology 85, 1625-1631.

467

Nitatpattana, N., Dubot-Peres, A., Gouilh, M. A., Souris, M., Barbazan, P., Yoksan, S., de

468

Lamballerie, X. & Gonzalez, J. P. (2008). Change in Japanese encephalitis virus

469

distribution, Thailand. Emerg Infect Dis 14, 1762-1765.

470

Pan, X. L., Liu, H., Wang, H. Y., Fu, S. H., Liu, H. Z., Zhang, H. L., Li, M. H., Gao, X. Y.,

471

Wang, J. L., Sun, X. H., Lu, X. J., Zhai, Y. G., Meng, W. S., He, Y., Wang, H. Q.,

472

Han, N., Wei, B., Wu, Y. G., Feng, Y., Yang, D. J., Wang, L. H., Tang, Q., Xia, G.,

473

Kurane, I., Rayner, S. & Liang, G. D. (2011). Emergence of genotype I of Japanese

474

encephalitis virus as the dominant genotype in Asia. Journal of virology 85, 9847-9853.

475

Pierson, T. C. D., M. S. (2013). Flaviviruses. In Fields Virology, 6 edn, pp. 747-794. Edited by

476 477

D. M. H. Knipe, P. M. Philadelphia: Lippincott Williams & Wilkins. Schuh, A. J., Ward, M. J., Leigh Brown, A. J. & Barrett, A. D. (2014). Dynamics of the

478

Emergence and Establishment of a Newly Dominant Genotype of Japanese Encephalitis

479

Virus throughout Asia. Journal of virology 88, 4522-4532.

480

Seo, H. J., Kim, H. C., Klein, T. A., Ramey, A. M., Lee, J. H., Kyung, S. G., Park, J. Y., Cho,

481

Y. S., Cho, I. S. & Yeh, J. Y. (2013). Molecular detection and genotyping of Japanese

482

encephalitis virus in mosquitoes during a 2010 outbreak in the Republic of Korea. PloS

483

one 8, e55165.

484

Solomon, T., Ni, H., Beasley, D. W., Ekkelenkamp, M., Cardosa, M. J. & Barrett, A. D.

485

(2003). Origin and evolution of Japanese encephalitis virus in southeast Asia. Journal of

486

virology 77, 3091-3098.

487

Sumiyoshi, H., Tignor, G. H. & Shope, R. E. (1995). Characterization of a highly attenuated

488

Japanese encephalitis virus generated from molecularly cloned cDNA. The Journal of

489

infectious diseases 171, 1144-1151.

490

Tajima, S., Nerome, R., Nukui, Y., Kato, F., Takasaki, T. & Kurane, I. (2010). A single

491

mutation in the Japanese encephalitis virus E protein (S123R) increases its growth rate in

492

mouse neuroblastoma cells and its pathogenicity in mice. Virology 396, 298-304.

493

Takhampunya, R., Kim, H. C., Tippayachai, B., Kengluecha, A., Klein, T. A., Lee, W. J.,

494

Grieco, J. & Evans, B. P. (2011). Emergence of Japanese encephalitis virus genotype V

495

in the Republic of Korea. Virol J 8, 449.

496

Tsai, T. F. (2000). New initiatives for the control of Japanese encephalitis by vaccination:

497

minutes of a WHO/CVI meeting, Bangkok, Thailand, 13-15 October 1998. Vaccine 18

498

Suppl 2, 1-25.

499

Vogt, M. R., Moesker, B., Goudsmit, J., Jongeneelen, M., Austin, S. K., Oliphant, T.,

500

Nelson, S., Pierson, T. C., Wilschut, J., Throsby, M. & Diamond, M. S. (2009).

501

Human monoclonal antibodies against West Nile virus induced by natural infection

502

neutralize at a postattachment step. Journal of virology 83, 6494-6507.

503

Wang, H. Y., Takasaki, T., Fu, S. H., Sun, X. H., Zhang, H. L., Wang, Z. X., Hao, Z. Y.,

504

Zhang, J. K., Tang, Q., Kotaki, A., Tajima, S., Liang, X. F., Yang, W. Z., Kurane, I.

505

& Liang, G. D. (2007). Molecular epidemiological analysis of Japanese encephalitis

506

virus in China. The Journal of general virology 88, 885-894.

507

Wu, K. P., Wu, C. W., Tsao, Y. P., Kuo, T. W., Lou, Y. C., Lin, C. W., Wu, S. C. & Cheng,

508

J. W. (2003). Structural basis of a flavivirus recognized by its neutralizing antibody:

509

solution structure of the domain III of the Japanese encephalitis virus envelope protein. J

510

Biol Chem 278, 46007-46013.

511

Yamaguchi, Y., Nukui, Y., Tajima, S., Nerome, R., Kato, F., Watanabe, H., Takasaki, T. &

512

Kurane, I. (2011). An amino acid substitution (V3I) in the Japanese encephalitis virus

513

NS4A protein increases its virulence in mice, but not its growth rate in vitro. The Journal

514

of general virology 92, 1601-1606.

515

Yang, D. K., Kim, B. H., Kweon, C. H., Kwon, J. H., Lim, S. I. & Han, H. R. (2004).

516

Molecular characterization of full-length genome of Japanese encephalitis virus

517

(KV1899) isolated from pigs in Korea. J Vet Sci 5, 197-205.

518

Yoshida, Y., Tabei, Y., Hasegawa, M., Nagashima, M. & Morozumi, S. (2005). Genotypic

519

analysis of Japanese encephalitis virus strains isolated from swine in Tokyo, Japan. Jpn J

520

Infect Dis 58, 259-261.

521

Yun, S. M., Cho, J. E., Ju, Y. R., Kim, S. Y., Ryou, J., Han, M. G., Choi, W. Y. & Jeong, Y.

522

E. (2010). Molecular epidemiology of Japanese encephalitis virus circulating in South

523

Korea, 1983-2005. Virol J 7, 127.

524

Zhang, J. S., Zhao, Q. M., Guo, X. F., Zuo, S. Q., Cheng, J. X., Jia, N., Wu, C., Dai, P. F. &

525

Zhao, J. Y. (2011). Isolation and genetic characteristics of human genotype 1 Japanese

526

encephalitis virus, China, 2009. PloS one 6, e16418.

527 528 529

Zhang, J. S., Zhao, Q. M., Zhang, P. H., Jia, N. & Cao, W. C. (2009). Genomic sequence of a Japanese encephalitis virus isolate from southern China. Arch Virol 154, 1177-1180. Zhao, Z., Date, T., Li, Y., Kato, T., Miyamoto, M., Yasui, K. & Wakita, T. (2005).

530

Characterization of the E-138 (Glu/Lys) mutation in Japanese encephalitis virus by using

531

a stable, full-length, infectious cDNA clone. The Journal of general virology 86, 2209-

532

2220.

533 534 535

536

Table 1. Mouse neuroinvasiveness of the GI, GIII, and GV JEV strains.

Virus Survival† Muar/1952 9/10 Beijing-1 8/10 Mie/41/2002 10/10 103 Muar/1952 1/10 Beijing-1 0/10 Mie/41/2002 8/10 4 10 Muar/1952 0/10 Beijing-1 0/10 Mie/41/2002 7/10 105 Muar/1952 0/10 Beijing-1 0/10 Mie/41/2002 7/10 † No. of mice surviving/no. mice inoculated. Dose 102

537 538 539 540



Average time to death (day) 9 8 7.8 7.2 9 6.6 7.2 10.7 7.7 8.7 11.3

P value‡ (vs. Mie/41/2002)

P value‡ (vs. Muar/1952)

0.32 0.15

In vitro growth, pathogenicity and serological characteristics of the Japanese encephalitis virus genotype V Muar strain.

The characteristics of genotype V Japanese encephalitis virus (GV JEV) remain poorly understood as only two strains have been isolated to date. In thi...
1013KB Sizes 0 Downloads 9 Views