JGV Papers in Press. Published April 28, 2015 as doi:10.1099/vir.0.000168

Journal of General Virology Delayed interferon response differentiates replication of West Nile virus and Japanese encephalitis virus in human neuroblastoma and glioblastoma cells --Manuscript Draft-Manuscript Number:

VIR-D-15-00215R1

Full Title:

Delayed interferon response differentiates replication of West Nile virus and Japanese encephalitis virus in human neuroblastoma and glioblastoma cells

Short Title:

Delayed IFN response in WNV- or JEV- infected human neuronal cells

Article Type:

Short Communication

Section/Category:

Animal - Positive-strand RNA Viruses

Corresponding Author:

Yuki Takamatsu, Ph.D., M.D. Institute of Tropical Medicine, Nagasaki University Nagasaki city, Nagasaki JAPAN

First Author:

Yuki Takamatsu, Ph.D., M.D.

Order of Authors:

Yuki Takamatsu, Ph.D., M.D. Leo Uchida, Ph.D., D.V.M. Kouichi Morita, Prof., Ph.D., M.D.

Abstract:

West Nile virus (WNV) and Japanese encephalitis virus (JEV) are important causes of human encephalitis cases that resulted in high-mortality ratio and neurological sequelae after recovery. The mechanism for neuro-pathogenicity in these viruses infection is an important research interest to develop specific anti-viral therapy. Here, we focused on human derived neuronal and glial cells to understand the cellular responses against WNV- and JEV- infection. It was demonstrated that the early IFN-β induction regulates viral replication in glioblastoma T98G cells, whereas delayed IFN-β induction resulted in efficient virus replication in neuroblastoma SK-N-SH cells. Moreover, the concealing of viral dsRNA in the intracellular membrane resulted in the delayed IFN response in SK-N-SH cells. These results which showed different IFN response between human neuronal and glial cells after WNV or JEV infection are expected to contribute in understanding the molecular mechanisms for neuropathology in these viruses infection.

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): revised manuscript.docx

Delayed interferon response differentiates replication of West Nile

virus

and

Japanese

encephalitis

virus

in

human

neuroblastoma and glioblastoma cells

Yuki Takamatsu1, Leo Uchida1, and Kouichi Morita1*

1) Department of Virology, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan

*Corresponding author: Kouichi Morita Address: 1-12-4, Sakamoto, Nagasaki, Nagasaki 852-8524, Japan Phone: +81-95-819-7829, Fax; +81-95-819-7830 E-mail: [email protected]

The word count of abstract: 144 The word count of the main text: 2441

1

1

Abstract

2

West Nile virus (WNV) and Japanese encephalitis virus (JEV) are important

3

causes of human encephalitis cases that resulted in high-mortality ratio and

4

neurological sequelae after recovery. The mechanism for neuro-pathogenicity in

5

these viruses infection is an important research interest to develop specific

6

anti-viral therapy. Here, we focused on human derived neuronal and glial cells to

7

understand the cellular responses against WNV- and JEV- infection. It was

8

demonstrated that the early IFN- induction regulates viral replication in

9

glioblastoma T98G cells, whereas delayed IFN- induction resulted in efficient

10

virus replication in neuroblastoma SK-N-SH cells. Moreover, the concealing of

11

viral dsRNA in the intracellular membrane resulted in the delayed IFN response

12

in SK-N-SH cells. These results which showed different IFN response between

13

human neuronal and glial cells after WNV or JEV infection are expected to

14

contribute in understanding the molecular mechanisms for neuropathology in

15

these viruses infection.

16 17

Short communications

18

West

Nile

virus (WNV) and

Japanese

2

encephalitis virus

(JEV) are

19

mosquito-borne viruses that belong to genus Flavivirus, family Flaviviridae

20

(Gubler et al., 2007; Sips et al., 2012). Most infections due to either viruses are

21

asymptomatic or cause febrile illness in humans, and may lead to encephalitis

22

resulting in high-mortality ratio and neurological sequelae after recovery

23

(Solomon, 2004). The specific anti-viral therapy has not yet been developed

24

(Solomon, 2004). There have been few reports focused on WNV and JEV

25

infection in human cells derived from brain (Kleinschmidt et al., 2007; Kumar et

26

al., 2010), and it has not yet been revealed how these viruses spread in these

27

cells. The mechanism of neuro-pathogenicity in WNV and JEV infection is an

28

important research interest to develop specific anti-viral therapy.

29

WNV and JEV can multiply in several cultured cells which exhibit cytopathic

30

effect (CPE) upon infection (Gubler et al., 2007; Kleinschmidt et al., 2007; Kumar

31

et al., 2010; Parquet et al., 2001; Stim & Henderson, 1969). To understand the

32

cellular responses against WNV- and JEV- infection, we used two kinds of cells

33

which showed different interferon (IFN) response and viral replication after WNV

34

and JEV infection.

35

3

36

The IFN response is the important defense against the early phase of viral

37

infections (Randall & Goodbourn, 2008). The type-I IFN induction is triggered by

38

viral components called pathogen-associated molecular patterns (PAMPs) such

39

as virus derived double-stranded RNA (dsRNA). The endoplasmic reticulum

40

(ER) provides the membrane platform for the formation of the flavivirus

41

replication complex that houses the nonstructural (NS) proteins and the

42

accumulating viral dsRNAs during viral genome synthesis (Gillespie et al., 2010;

43

Westaway et al., 1997). The dsRNA is recognized by pattern recognition

44

receptors (PRR), namely retinoic acid-inducible gene-I (RIG-I) and melanoma

45

differentiation-associated protein 5 (MDA5), and induces the IFN response (Loo

46

& Gale, 2011; Quicke & Suthar, 2013). The induction of IFN- is critical for

47

controlling WNV and JEV replication during the course of infection (Lin et al.,

48

2004; Quicke & Suthar, 2013). It was firstly reported in tick borne encephalitis

49

virus (TBEV) infection that the dsRNA is enclosed in intracellular membrane

50

vesicles, and as a consequence leads to an escape from the host immune

51

system (Overby et al., 2010). It was reported previously in JEV infected porcine

52

cells that viral dsRNA is concealed in intracellular membrane resulting in delayed

53

cytosolic exposure (Espada-Murao & Morita, 2011). The concealing mechanism

4

54

of dsRNA was also reported in Dengue virus (DENV) infected HeLa cells, which

55

resulted in the evasion of IFN response (Uchida et al., 2014). These findings

56

clearly suggest that the concealing of dsRNA plays a crucial role on viral

57

replication by escaping from the host immune system in flaviviruses infection.

58

This study aims to reveal the molecular mechanisms for WNV or JEV

59

dissemination and replication in human neuronal and glial cells.

60 61

Human

62

glioblastoma T98G cells (ATCC; CRL-1690) were used for viral infection. The

63

role of glial cells in WNV and JEV infection has been unrevealed, and this is the

64

first attempt to compare WNV and JEV infection between neuronal cells and glial

65

cells. WNV strains NY99 and Egypt101, JEV strains JaOArS982 and JaNAr0102

66

were used in this study. The viruses were propagated in the African green

67

monkey kidney cells (Vero cells) to generate working stocks. At 72 hour

68

post-inoculation, the culture supernatants were collected and stored in aliquots

69

at -80oC. WNV or JEV infection was performed on a monolayer of cells on

70

24-well plates at a multiplicity of infection (MOI) of 5, and supernatants were

71

harvested at the indicated time points. Virus titers were determined by

neuroblastoma

SK-N-SH

cells

5

(ATCC;

HTB-11),

and

human

72

plaque-forming assays on BHK cells and expressed as PFU /mL (Hayasaka et

73

al., 2009; Takamatsu et al., 2014). The clear CPE (rounding of cells, detachment

74

from the monolayer, and cell shrinkage) was observed in WNV or JEV infected

75

SK-N-SH cells from 3 days post-inoculation. On the other hand, no clear

76

morphology changes were observed in WNV or JEV infected T98G cells up to 5

77

days post-inoculation (data not shown). SK-N-SH cells showed higher virus titers

78

than T98G cells after infection with WNV or JEV. A slower viral growth and a

79

lower peak of viral titer were observed in T98G cells (Fig. 1a). These

80

experiments suggested that SK-N-SH cells are permissive for viral replication,

81

whereas T98G cells have some mechanisms to regulate virus propagation.

82

Next, we focused on cellular innate immune response against viral infection.

83

Real-time quantitative RT-PCR assay for IFN- was performed as described

84

previously (Takamatsu et al., 2014). The primer information was indicated in

85

Table S1. The early IFN- up-regulation was observed both in WNV- and JEV-

86

infected T98G cells from 8 hour post-inoculation (Fig. 1b). The timing of

87

increased viral RNA level and virus production was similar in SK-N-SH cells and

88

T98G cells (Fig. S1). Interestingly, the delayed IFN- induction was shown in

89

SK-N-SH cells, although sufficient viral replication was observed from early time

6

90

of infection. On the other hand, immediate IFN- induction was shown in the

91

absence of sufficient viral replication in T98G cells. There was no significant

92

difference in the basal levels of IFN- (absolute amount) between SK-N-SH and

93

T98G cells prior to infection (data not shown).

94 95

To clarify the role of type-I IFN in virus dissemination and growth, it was blocked

96

using an anti-IFN antibody cocktail. The cells were treated with a combination of

97

an anti-human IFN- (PBL Interferon Source) and an anti-human IFN-R2

98

(PBL Interferon Source) at different concentrations 1 hour before the viral

99

inoculation (Uchida et al., 2014). Focus formation assay was performed on

100

SK-N-SH cells and T98G cells as described previously (Espada-Murao & Morita,

101

2011). Foci of infected cells were larger in WNV- or JEV- infected SK-N-SH and

102

T98G cells treated with anti-IFN cocktail compared to the non-treated group (Fig.

103

2a). These results indicate that IFN response restricts viral replication both in

104

SK-N-SH and T98G cells. To elucidate the role of IFN- on viral replication, an

105

immediate IFN- treatment with 100 or 1000 units/well was performed

106

(Espada-Murao & Morita, 2011). The results showed that an early IFN-

107

treatment significantly reduced viral titers both in SK-N-SH cells and T98G cells

7

108

(Fig. 2b). It is suggested that the delayed IFN- induction impaired IFN response

109

at early time of infection, thereby enhancing WNV and JEV replication in

110

SK-N-SH cells. The cytosolic PRRs are reported to regulate IFNexpression

111

in flavivirus infection (Kato et al., 2006; Loo & Gale, 2011). To compare protein

112

expression in WNV- and JEV- infected cells, cellular extracts were subjected to

113

immunoblotting for RIG-I, MDA5, and for -Actin as an internal control

114

(Takamatsu et al., 2014; Uchida et al., 2014). The RIG-I expression was

115

detectable from 24 hour post-inoculation both in SK-N-SH and T98G cells (Fig.

116

2c), whereas the MDA5 expression was not detectable in the two cells (data not

117

shown). It indicates that the RIG-I is required but not MDA-5 for IFN-induction

118

in JEV infection (Kato et al., 2006), and this is in agreement with the previous

119

report (Uchida et al., 2014).

120

121

The timing of the dsRNA exposure in cytosol has been reported to be important

122

for inducing IFN response in TBEV-, DENV-, and JEV- infected cells

123

(Espada-Murao & Morita, 2011; Overby et al., 2010; Uchida et al., 2014). Two

124

permeabilization methods were applied to differentiate the localization of the

125

dsRNA, either exposed in cytoplasm or concealed in intracellular membrane. 8

126

Nonidet P-40 (NP-40) treatment permeabilizes all cellular membrane structure

127

like the plasma membrane, and ER; digitonin permeabilizes only the plasma

128

membrane (Uchida et al., 2014). WNV or JEV infection was performed on a

129

monolayer of cells on eight-well chamber slides (Nunc) at an MOI of 10. At

130

indicated time points, the cells were subjected to immunostaining as described

131

previously (Espada-Murao & Morita, 2011; Uchida et al., 2014). The mouse

132

IgG2a K1 monoclonal Ab (English & Scientific Consulting Kit) was used to

133

visualize viral dsRNA. The images were captured by LSM 780 confocal laser

134

scanning microscope (Carl Zeiss). The cell number in a field was counted by

135

ImageJ software (Schneider et al., 2012). Interestingly, the dsRNA was

136

predominantly concealed in intra-cellular membrane structures in SK-N-SH cells

137

after WNV infection (Fig. 3a). The ratio of exposed dsRNA in cytoplasm was

138

lower (approximately 10%) in WNV infected SK-N-SH cells, whereas that in

139

T98G cells was higher (approximately 50%) during the course of infection (Fig.

140

3b). A similar finding of concealed dsRNA was noted in JEV- infected SK-N-SH

141

cells, and the ratio of exposed dsRNA was also higher in T98G cells than that in

142

SK-N-SH cells (Fig. 3c). The results suggested that the delayed cytosolic

143

exposure of dsRNA was correlated with the delayed IFN- induction in SK-N-SH

9

144

cells. In our previous report, the dsRNA of DENV was concealed in intracellular

145

membrane but JEV was exposed in the cytoplasm of a human cervical-derived

146

HeLa cells at early infection (Uchida et al., 2014). A similar finding was observed

147

in WNV- infected HeLa cells (data not shown). The specific observation of

148

concealed dsRNA in neuronal cells may contribute to the viral pathogenicity in

149

WNV and JEV infection. It is suggested that the dsRNA leaking from the small

150

pore of replication vesicles at the later phase of infection is recognized by the

151

cytosolic PRRs in TBEV infection (Overby & Weber, 2011). It is possible that the

152

difference in formation of vesicle packets where dsRNA is likely to be concealed

153

(Gillespie et al., 2010) is related to the difference in the levels of dsRNA

154

exposure in cytoplasm between neuronal and glial cells. However, the

155

mechanism of triggering the cytosolic dsRNA exposure has not yet been

156

identified. Therefore, we need further investigations to reveal how WNV and JEV

157

utilize the mechanism for concealing viral dsRNA in association with host cellular

158

factors.

159 160

In conclusion, this is the first report to indicate the concealing mechanisms of

161

dsRNA in WNV- and JEV- infected human neuronal cells. The early IFN-

10

162

induction regulates viral replication in glioblastoma T98G cells, whereas delayed

163

IFN- induction resulted in efficient virus replication in neuroblastoma SK-N-SH

164

cells. These results which showed a difference in the IFN response between

165

human neuronal and glial cells after WNV or JEV infection, are expected to

166

contribute to understanding the molecular mechanisms for neuropathology due

167

to these viruses.

168 169

Acknowledgements

170

We would like to thank Dr. Corazon C. Buerano, and Mr. Gianne Eduard L.

171

Ulanday from the Department of Virology, Institute of Tropical Medicine,

172

Nagasaki University for helping the revision of our manuscript, and all of the

173

members and the recent alumni of the Department of Virology, Institute of

174

Tropical Medicine, Nagasaki University for their support. This study was

175

supported by a Grant-in-Aid for Scientific Research from the Ministry of

176

Education, Culture, Sports, Science and Technology (MEXT), Japan; the

177

Grant-in-Aid for JSPS Fellows (Japan Society for the Promotion of Science),

178

MEXT, Japan; the Global COE program, MEXT, Japan, the Japan Initiative for

179

Global Network on Infectious Diseases (J-GRID), MEXT, Japan; and the

11

180

Grant-in-aid for scientific research from the Ministry of Health, Labour, and

181

Welfare, Japan.

182

12

183

References

184

Espada-Murao, L. A. & Morita, K. (2011). Delayed cytosolic exposure of Japanese

185

encephalitis virus double-stranded RNA impedes interferon activation and enhances

186

viral dissemination in porcine cells. Journal of virology 85, 6736-6749.

187

Gillespie, L. K., Hoenen, A., Morgan, G. & Mackenzie, J. M. (2010). The endoplasmic

188

reticulum provides the membrane platform for biogenesis of the flavivirus

189

replication complex. Journal of virology 84, 10438-10447.

190

Gubler, D. J., Kuno, G. & Markoff, L. (2007). Field's Virology. Philadelphia: Wolters Kluwer

191

Lippincott Williams and Wilkins.

192

Hayasaka, D., Nagata, N., Fujii, Y., Hasegawa, H., Sata, T., Suzuki, R., Gould, E. A.,

193

Takashima, I. & Koike, S. (2009). Mortality following peripheral infection with

194

tick-borne encephalitis virus results from a combination of central nervous system

195

pathology, systemic inflammatory and stress responses. Virology 390, 139-150.

196

Kato, H., Takeuchi, O., Sato, S., Yoneyama, M., Yamamoto, M., Matsui, K., Uematsu, S.,

197

Jung, A., Kawai, T., Ishii, K. J., Yamaguchi, O., Otsu, K., Tsujimura, T., Koh, C. S.,

198

Reis e Sousa, C., Matsuura, Y., Fujita, T. & Akira, S. (2006). Differential roles of

199

MDA5 and RIG-I helicases in the recognition of RNA viruses. Nature 441, 101-105.

200

Kleinschmidt, M. C., Michaelis, M., Ogbomo, H., Doerr, H. W. & Cinatl, J., Jr. (2007).

201

Inhibition of apoptosis prevents West Nile virus induced cell death. BMC

202

microbiology 7, 49.

203

Kumar, M., Verma, S. & Nerurkar, V. R. (2010). Pro-inflammatory cytokines derived from

204

West Nile virus (WNV)-infected SK-N-SH cells mediate neuroinflammatory markers

205

and neuronal death. Journal of neuroinflammation 7, 73.

206

Lin, R. J., Liao, C. L., Lin, E. & Lin, Y. L. (2004). Blocking of the alpha interferon-induced

207

Jak-Stat signaling pathway by Japanese encephalitis virus infection. Journal of

208

virology 78, 9285-9294.

209 210

Loo, Y. M. & Gale, M., Jr. (2011). Immune signaling by RIG-I-like receptors. Immunity 34, 680-692.

211

Overby, A. K., Popov, V. L., Niedrig, M. & Weber, F. (2010). Tick-borne encephalitis virus

212

delays interferon induction and hides its double-stranded RNA in intracellular

213

membrane vesicles. Journal of virology 84, 8470-8483.

214 215 216 217

Overby, A. K. & Weber, F. (2011). Hiding from intracellular pattern recognition receptors, a passive strategy of flavivirus immune evasion. Virulence 2, 238-240. Parquet, M. C., Kumatori, A., Hasebe, F., Morita, K. & Igarashi, A. (2001). West Nile virus-induced bax-dependent apoptosis. FEBS letters 500, 17-24.

13

218

Quicke, K. M. & Suthar, M. S. (2013). The innate immune playbook for restricting West Nile

219

virus infection. Viruses 5, 2643-2658.

220

Randall, R. E. & Goodbourn, S. (2008). Interferons and viruses: an interplay between

221

induction, signalling, antiviral responses and virus countermeasures. The Journal of

222

general virology 89, 1-47.

223

Schneider, C. A., Rasband, W. S. & Eliceiri, K. W. (2012). NIH Image to ImageJ: 25 years of

224 225

image analysis. Nature methods 9, 671-675. Sips, G. J., Wilschut, J. & Smit, J. M. (2012). Neuroinvasive flavivirus infections. Reviews in

226 227

medical virology 22, 69-87. Solomon, T. (2004). Flavivirus encephalitis. The New England journal of medicine 351,

228 229

370-378. Stim, T. B. & Henderson, J. R. (1969). Arbovirus plaquing in a clonal line (PS Y-15) of

230

porcine kidney. Applied microbiology 17, 246-249.

231

Takamatsu, Y., Okamoto, K., Dinh, D. T., Yu, F., Hayasaka, D., Uchida, L., Nabeshima, T.,

232

Buerano, C. C. & Morita, K. (2014). NS1' protein expression facilitates production of

233

Japanese encephalitis virus in avian cells and embryonated chicken eggs. The

234

Journal of general virology 95, 373-383.

235

Uchida, L., Espada-Murao, L. A., Takamatsu, Y., Okamoto, K., Hayasaka, D., Yu, F.,

236

Nabeshima, T., Buerano, C. C. & Morita, K. (2014). The dengue virus conceals

237

double-stranded RNA in the intracellular membrane to escape from an interferon

238

response. Scientific reports 4, 7395.

239

Westaway, E. G., Mackenzie, J. M., Kenney, M. T., Jones, M. K. & Khromykh, A. A. (1997).

240

Ultrastructure of Kunjin virus-infected cells: colocalization of NS1 and NS3 with

241

double-stranded RNA, and of NS2B with NS3, in virus-induced membrane

242

structures. Journal of virology 71, 6650-6661.

243 244 245

Figure legend

246

Figure 1 Virus replication and IFN- induction in SK-N-SH cells and T98G

247

cells

248

(a) Each virus (WNV; NY99, Egyot101, JEV; JaOArS982, JaNAr0102) was

14

249

inoculated at MOI of 5 onto SK-N-SH cells and T98G cells. The cell supernatants

250

were harvested at 1, 4, 8, 12, 24, 36, and 48 hour post-inoculation. The results of

251

PFU titration are expressed as the mean of three independent experiments, the

252

error bars indicate standard deviations of the mean. (b) Each virus (WNV; NY99,

253

Egyot101, JEV; JaOArS982, JaNAr0102) was infected on a monolayer of

254

SK-N-SH cells and T98G cells on 24-well plates at an MOI of 5. The number of

255

copies of IFN- mRNA, glyceraldehyde 3-phosphate dehydrogenase (GAPDH)

256

mRNAs was calculated by absolute quantification based on in vitro-transcribed

257

viral RNA, and GAPDH standards. The results for IFN- mRNA were normalized

258

to GAPDH, and expressed as the fold increase over non-infected cells. The

259

results are expressed as the mean of three independent experiments, and the

260

error bars indicate standard deviations of the mean. A group of samples was

261

assessed by Student’s t-tests or Welch tests. A P value of less than 0.05 was

262

considered statistically significant. Double asterisks and a number indicated a P

263

value.

264 265

Figure 2 IFN response and cytosolic PRRs expression

266

(a) WNV (NY99) or JEV (JaOArS982) infection was performed on a monolayer

15

267

of cells on 24-well plates with or without anti-IFN cocktail treatment. The cells

268

were treated by anti-IFN cocktail 1 h before viral inoculation. Culture medium

269

was used for control group of cells. At 5 days post-inoculation, the size of foci

270

was compared by focus forming assay. a; 400 neutralization U/ml of anti-human

271

IFN- and 4 mg/ml of anti-human IFN-R2. b; 2,000 neutralization U/ml of

272

anti-human IFN- and 20 mg/ml of anti-human IFN-R2. (b) WNV (NY99) or

273

JEV (JaOArS982) infection was performed on a monolayer of cells on 24-well

274

plates at an MOI of 5 with or without immediate IFN- treatment (100 or 1000

275

units/well). Culture medium was used for control group of cells. At 24 and 48

276

hour post-inoculation, viral titers were measured by PFU titration. The results are

277

expressed as the mean of three independent experiments, and the error bars

278

indicate standard deviations of the mean. A group of samples was assessed by

279

Student’s t-tests or Welch tests. A P value of less than 0.01 was indicated as

280

asterisks. (c) WNV (NY99) or JEV (JaOArS982) infection was performed on a

281

monolayer of cells on 12-well plates at an MOI of 5. At 24 and 48 hour

282

post-inoculation, the infected cells were harvested and the target proteins were

283

detected by the immunoblotting assay. The -Actin was used as an internal

284

control. All the samples were derived from the same experiment and blotting was

16

285

processed in parallel.

286 287

Figure 3 Delayed cytosolic exposure of dsRNA in SK-N-SH cells

288

WNV (NY99) infection was performed on a monolayer of cells on eight-well

289

chamber slides (Nunc) at an MOI of 10. (a) At the indicated time points, the

290

infected cells were fixed and permeabilized by 1% NP-40 or 0.5mM digitonin.

291

The viral dsRNA was visualized by using the immunofluorescence assay.

292

Nuclear staining was achieved using DAPI. (b, c) The exposed dsRNA ratio in

293

(b) WNV (NY99)- or (c) JEV (JaOArS982)- infected SK-N-SH cells and T98G

294

cells. The ratio expressed in percentage was determined by dividing the number

295

of digitonin-treated cells positive for dsRNA with the number of NP-40 treated

296

cells positive for dsRNA. The images showed comparable numbers of total cells

297

from both NP-40- and digitonin- treated cells. The percentage of cells was

298

calculated in 3 different fields. The error bars indicate standard error of the mean.

299

A group of samples was assessed by Mann Whitney test. A P value of less than

300

0.05 was considered statistically significant, and indicated as asterisks.

17

Figure Click here to download Figure: Figure 1.tif

Figure Click here to download Figure: Figure 2.tif

Figure Click here to download Figure: Figure 3.tif

Click here to download Supplementary Material Files: revised Supplementary matterial.pdf

Delayed IFN response differentiates replication of West Nile virus and Japanese encephalitis virus in human neuroblastoma and glioblastoma cells.

West Nile virus (WNV) and Japanese encephalitis virus (JEV) are important causes of human encephalitis cases, which result in a high mortality ratio a...
1MB Sizes 0 Downloads 8 Views