JGV Papers in Press. Published June 19, 2014 as doi:10.1099/vir.0.062562-0

1 2

Production of a neutralizing antibody against envelope protein of dengue virus type 2 using linear array epitope (LAE) technique

3

Peng-Yeh Laia, Chia-Tse Hsub, Shao-Hung Wangc, Jin-Ching Leed, Min-Jen Tsenga, Jaulang

4

Hwanga, e, Wen-Tsai Jia and Hau-Ren Chena,*

5

a

6

Science, College of Science, National Chung Cheng University, Min-Hsiung, Chia-Yi 62102,

7

Taiwan.

8

b

9

Biochemical Engineering, Kao Yuan University, Luzhu District, Kaohsiung City 82151,

Department of Life Science, Institute of Molecular Biology and Institute of Biomedical

Department of Chemical and Biochemical Engineering and Institute of Chemical and

10

Taiwan

11

c

12

University, Chiayi City 60004, Taiwan.

13

d

14

80708,Taiwan

15

e

16

11031, Taiwan.

17 18

*Correspondence to Dr. Hau-Ren Chen, Department of Life Science, National Chung Cheng University.

19

168, Sec. 1, University Road, Min-Hsiung, Chia-Yi 62102, Taiwan.

20

Tel, 886-5-2720411 ext 66503; Fax, 886-5-2722871; e-mail, [email protected]

21

Running title: Novel antibody generated by LAE technique

22

Abstract: 245

23

Text including figure legends: 4832

24

Total 5 figures and 0 table

25

Category: Animal virus-Positive-strand RNA

26

Department of Microbiology, Immunology and Biopharmaceuticals, National Chiayi

Department

of

Biotechnology,

Kaohsiung

Medical

University,

Kaohsiung

city

Department of Biochemistry, School of Medicine, Taipei Medical University, Taipei City

27

Abstract

28

Dengue virus belongs to Flavivirus and contains a positive-stranded RNA genome

29

Binding of dengue virus to host cells was mediated through domain III of the viral

30

envelope protein. Many therapeutic monoclonal antibodies (mAbs) against domain III

31

have been generated and characterized because of its high antigenicity. We have

32

previously established a novel PCR method named linear array epitope (LAE)

33

technique for producing monoclone-like polyclonal antibodies. To prove this method

34

could be utilized to produce antibody against epitopes with low antigenicity, a region

35

of 10 amino acids (V365NIEAEPPFG374) from domain III of the envelope protein in

36

dengue virus serotype 2 (DENV2) was selected to design the primers for LAE

37

technique. A DNA fragment encoding 10 directed repeats of these 10 amino acids for

38

producing the tandem repeated peptides was obtained and fused it with

39

GST-containing vector. This fusion protein (GST-Den EIII10-His6) was purified from

40

E. coli and used as antigen for immunizing rabbits to obtain polyclonal antibody.

41

Furthermore, this EIII antibody could recognize envelope proteins either ectopically

42

overexpressed or synthesized by DENV2 infection using immunoblot and

43

immunofluorescence assays. Most importantly, this antibody was also capable of

44

detecting DENV2 virions by ELISA assay and could block viral entry into BHK-21

45

cells as shown by immunofluorescence and qRT-PCR assays. Taken together, LAE

46

technique could be applied for production of antibody against antigen with low

47

antigenicity successfully and carries a high potential to produce antibodies with good

48

quality for academic research, diagnosis and even therapeutic application in the

49

future.

50

Abbreviations: LAE: linear array epitope, TR-PCR: template-repeated polymerase

51

chain reaction, PEIa: exotoxin receptor-binding domain A of Pseudomonas

52

531. Introduction 54

Dengue virus is a considerable threat to public health all worldwide. The outbreaks

55

and epidemics caused by virus infection become a serious public health issue in many

56

tropical and subtropical countries. Dengue viruses are classified into four serotypes

57

(DENV1 to DENV4) and are transmitted to human by mosquito vectors such as Aedes

58

aegypti and Aedes albopictus. Virus infection may cause different kinds of diseases,

59

including dengue fever (DF) with febrile illness, dengue haemorrhagic fever (DHF)

60

with plasma leakage syndrome and dengue shock syndrome (DSS). Approximately

61

100 million cases of DF and 500,000 cases of DHF and DSS occur every year, with a

62

mortality rate of 5% (Gubler, 2002; Wilder-Smith & Gubler, 2008). Therefore, it is

63

important and urgent to develop proper vaccines for prevention of the disease. In the

64

past decades, there are several approaches attempting to develop dengue virus

65

vaccines, including live attenuated viruses, chimeric viruses, recombinant subunit

66

antigens, expression vector-based vaccines and DNA vaccines (Chokephaibulkit &

67

Perng, 2013; Heinz & Stiasny, 2012; Lee et al., 2012; Leng et al., 2009; Schmitz et al.,

68

2011). However, there is currently no commercially available vaccine against dengue

69

virus.

70

Dengue virus belongs to the family of Flaviviridae, genus Flavivirus, with a

71

positive-sense single-stranded RNA genome. The RNA genome of 10.7 kb in length

72

has a 5’ cap structure but lacks 3’ poly A tail, and encodes a polyprotein which is

73

cleaved into three structural proteins (core, premembrane and envelope) and seven

74

non-structural proteins (NS1, NS2A, NS2B, NS3, NS4A, NS4B and NS5) by host

75

and/or viral proteases. Among the three structural proteins, the premembrane and

76

envelope proteins are the primary antigenic targets of humoral immune response in

77

human (Wan et al., 2013). The neutralizing antibodies are usually induced by the

78

envelope protein (Murphy & Whitehead, 2011). The envelope protein is consisted of

79

495 amino acids with a molecular weight of about 60 kDa. It has been suggested that

80

the major role of the envelope protein is host cell attachment and entry (Bhardwaj et

81

al., 2001). The ectodomain of envelope protein is composed of three domains,

82

including domain I, II and III (Modis et al., 2003; 2004; Rey et al., 1995). Domain I

83

contains mainly type-specific non-neutralizing epitopes and is thought to be the

84

molecular hinge region involved in low-pH-triggered conformational changes

85

(Roehrig et al., 1998). Domain II is the dimerization domain, and is involved in

86

virus-mediated membrane fusion. It contains many cross-reactive epitopes eliciting

87

neutralizing and non-neutralizing monoclonal antibodies (Rey et al., 1995; Roehrig et

88

al., 1998). Domain III is an immunoglobulin-like structure containing the most distal

89

projecting loops from the virion surface. According to X-ray crystal structure, domain

90

III contains 10 β-strands (A to G and a small extra sheet, AxCxDx). In addition,

91

domain III carries multiple type- and subcomplex-specific epitopes eliciting

92

neutralizing monoclonal antibodies that have been hypothesized to block virus

93

binding to the host cell receptor (Bhardwaj et al., 2001; Chiu & Yang, 2003; Modis et

94

al., 2003; 2004; Rey et al., 1995; Sukupolvi-Petty et al., 2007; Volk et al., 2004; Yu et

95

al., 2004). Since envelope domain III plays important roles in viral entry, it is a good

96

target for generating neutralizing antibody.

97

Linear array epitope technique (LAE) is a method for preparation of an immunogen

98

containing multiple copies of a defined peptide in a linear alignment. This method

99

was designed to overcome poor immune response induced by traditional

100

immunogenic peptide/protein antigens. The tandem repeated epitopes are able to

101

efficiently induce a strong immune response in vivo (Yi et al., 2006) and prime CD4+

102

T cells in vitro (Barratt-Boyes et al., 1999). Immunization of female rabbits with the

103

LAE-produced immunogen that contained the exotoxin receptor-binding domain A of

104

Pseudomonas (PEIa) and 12 copies of gonadotropin-releasing hormone (GnRH)

105

resulted in the generation of high-titer antibodies specific for GnRH. The anti-GnRH

106

antibodies effectively neutralized GnRH activity in vivo, as demonstrated by the

107

degeneration of the ovaries in the injected rabbits (Hsu et al., 2000). The

108

LAE-produced gastrin-releasing peptide (GRP) immunogen also induced antibody

109

potentially inhibiting breast cancer cell proliferation in vitro and in vivo (Guojun et al.,

110

2008). A rabbit anti-mouse EKLF (erythroid Krűppel-like factor) antibody induced by

111

LAE-produced immunogen could be used to detect EKLF in mouse embryonic

112

samples by Western blot and immunostaining, and was applicable for chromatin

113

immunoprecipitation (ChIP) assay (Shyu et al., 2007).

114

In this study, we used LAE technique to produce a tandem-repeated domain III region

115

(V365NIEAEPPFG374)10 of DENV2 envelope protein as the immunogen (referred as

116

EIII epitope hereafter). We demonstrated that anti-EIII antibody induced by this novel

117

immunogen could be used to detect the envelope protein either ectopically

118

overexpressed or synthesized by DENV2 infection in cells. Importantly, the antibody

119

could be used to detect dengue virions and to block dengue viral entry into BHK-21

120

cells successfully.

1212. Results 122

Epitope selection and the principle of linear array epitope (LAE) technique

123

To test whether LAE technique could be used to induce antibodies against DENV2

124

envelope domain III, we chose an epitope (V365NIEAEPPFG374) consisting of

125

β-strand and random coil secondary structures, and was not co-localized with previous

126

neutralizing epitopes (Crill & Chang, 2004; Gromowski & Barrett, 2007; Lin et al.,

127

1994; Thullier et al., 2001; Wan et al., 2013). According to the three-dimensional

128

structures

129

(www.rcsb.org/pdb), this epitope seemed to be localized on the surface of the protein

130

(Fig. S1). To synthesize repeated EIII epitope fused with GST, two oligonucleotides

131

(oligo Den E-A and Den E-B) were designed for template-repeated PCR (TR-PCR).

132

Oligo Den E-A encodes VNIEAEPPFG peptide, and its 5’ and 3’ halves are

133

complementary to those of oligo Den E-B, respectively (Figure 1). The TR-PCR

134

products containing repeated EIII epitope were used as the templates for the second

135

PCR (adapter-PCR) to introduce the restriction enzyme sites and stop codon. The

136

products of both TR-PCR and adapter-PCR showed the ladder pattern in 8% PAGE

137

(Figure 1d). The lowest band (the major one in lane 3) among the ladders was the

138

monomer, and the higher bands were multimers. The DNA products were eluted and

139

cloned into T vectors, followed by screening for positive clones. Finally, a clone with

140

10 tandem repeated epitopes was obtained.

141

Preparation of tandem repeated antigen for generating rabbit antiserum against

142

domain III of the envelope protein

143

The DNA fragment with 10 repeats of EIII epitope was subcloned into two expression

144

plasmids for producing tandem repeated antigens, generating GST-Den EIII10-His6

145

fusion protein and PEIa-Den EIII10-His6 fusion protein (data not shown) in E. coli

146

BL21(DE3). The induced proteins were purified by glutathione Sepharose and nickel

147

column, respectively. The Coomassie blue staining for purification of GST-Den

148

EIII10-His6 protein is shown in Figure 2a. Elution fractions 3 to 7 were collected as the

149

antigen to immunize rabbits. The harvested antiserum against GST-Den EIII10-His6

150

protein was named EIII antibody, and the antibody titer against recombinant

151

full-length domain III fused with His6 tag (referred as EIII-His6 hereafter) was

152

monitored by ELISA every two weeks. Even as low as 10 ng coated EIII-His6 could

of

the

envelope

protein

obtained

from

protein

data

bank

153

be significantly detected by this antibody; the OD410 values of 102- and 103-fold

154

diluted sera were much higher than those of control (Fig. 2b). The titer of the

155

antiserum achieved plateau after 6 weeks (data not shown).

156

Evaluation of the specificity of EIII antibody by various assays

157

To further confirm the specificity of the harvested EIII antibody, ELISA and

158

immunoblot assay was performed. Recombinant EIII-His6 proteins from four

159

serotypes of DENV were used to test whether EIII Ab could recognize EIII protein of

160

all serotypes. In ELISA assay, Fig. 2c showed that EIII Ab mainly detected EIII-His6

161

of DENV2 with 104-fold dilution, but cross-reacted with EIII-His6 of all four

162

serotypes with 102-fold dilution. In immunoblot assay, EIII antibody could recognize

163

purified EIII-His6, PEIa-Den EIII10-His6 and GST-Den EIII10-His6, but not the

164

unrelated proteins such as PEIa-His6 (the protein product of empty pPEIa-His6 vector),

165

TAF7-His6 (a transcriptional factor) and NS3-C-His6 (the N-terminal truncated NS3

166

protein of dengue virus), suggesting the high specificity of LAE-induced antibody

167

(Fig. 3a). We next examined if EIII antibody could detect full-length DENV2

168

envelope protein in cells. Figure 3b and 3c showed that EIII antibody could

169

specifically detect the DENV2 envelope proteins ectopically overexpressed in

170

BHK-21 or HEK293T cells by Western blot analysis and immunofluorescence assay,

171

respectively. Furthermore, we tested if the antibody could be used in

172

immunoprecipitation assay. The cell lysates from HEK293T cells transfected with

173

vector only or with pCMV-3FLAG-DENV2 envelope were incubated with EIII

174

antibody, resulting in immunoprecipitation of ectopically expressed FLAG-tagged

175

DENV2 envelope proteins (Fig. 3d).

176

Application of EIII antibody for detecting virions and blocking viral entry

177

Since EIII antibody could recognize recombinant envelope proteins in BHK-21 and

178

HEK293T cells, we examined if it could recognize the envelope protein of dengue

179

virus virions. C6/36 cells were infected with DENV2 for 10 to 12 days and the cell

180

lysates or the media were collected for Western blot analysis. EIII antibody could

181

clearly detect dengue virus premembrane/envelope heterodimer protein in

182

virus-infected cell lysates as indicated in the filled arrow (Sukupolvi-Petty et al., 2010;

183

Zhang et al., 2003). EIII antibody also detected envelope protein in the medium

184

fraction of virus-infected culture weakly as indicated in the open arrow (Fig. 4a). To

185

further confirm this result, EIII antibody was used to detect virus-infected BHK-21

186

cells at different time points after infection. As shown in Figure 4b, EIII antibody

187

could detect virus envelope proteins in the infected cells and the signal increased

188

commensurate to the infection time, which correlated with the replication cycle of

189

dengue virus. Since the envelope protein is located on the surface of the virus, the

190

antibody was tested for virions recognition. Different amounts of dengue viruses were

191

coated on the ELISA plate and serially diluted EIII antibody was used to detect the

192

virions. The results showed that EIII antibody with 10- to 103-fold dilutions could

193

detect virions in a dosage-dependent manner (Fig. 4c).

194

Finally we tested if EIII antibody could be applied for blocking dengue virus infection.

195

Dengue viruses were pre-incubated with serially diluted EIII antibody and the

196

virus-antibody mixture was used to infect BHK-21 cells. Viruses were detected for

197

NS3 by immunofluorescence and qRT-PCR assays. The immunofluorescence signals

198

were quantified by counting green fluorescence-positive cells in different fields, and

199

the results showed that EIII antibody with 10-fold dilution significantly reduced the

200

fluorescence signals (Fig. 5a and 5b). The levels of NS3 RNA were also decreased by

201

EIII antibody (Fig 5c), suggesting that viral entry might be blocked effectively. In

202

order to further verify the process, we treated EIII Ab either before or after dengue

203

virus infection and found that EIII Ab affected viral infection significantly only in

204

pretreated condition, indicating that EIII Ab blocked virus infection through blocking

205

of viral attachment (Fig. 5d). Since EIII Ab could block DENV2 infection, we further

206

test if this Ab would block viral entry of other serotypes. All fours serotypes of

207

DENV were used to evaluate the blocking ability as done in Fig. 5c. The result

208

suggested EIII Ab specifically blocked DENV2 infection although DENV4 could not

209

be determined (Fig. S3)

2103. Discussion 211

Infection by dengue virus is a global concern. However, there is still no approved

212

antiviral treatment available so far. Several candidate vaccines remain in preclinical or

213

clinical evaluation (Durbin & Whitehead, 2010). The main strategy of these candidate

214

vaccines is to induce neutralizing antibodies to block virus infection. The best target

215

for blocking virus infection is the structural proteins on the virion’s surface.

216

According to previous studies, many neutralizing antibodies were induced by the

217

envelope proteins. Based on epitope mapping data, many epitopes of the type-specific

218

neutralizing antibodies against individual flaviviruses are localized to domain III,

219

whereas neutralizing monoclonal antibodies that cross-react with other flaviviruses

220

are localized primarily to domain II (Beasley & Barrett, 2002; Cecilia & Gould, 1991;

221

Chavez et al., 2010; Crill & Chang, 2004; Crill & Roehrig, 2001; Fonseca et al., 1994;

222

Goncalvez et al., 2004a; Goncalvez et al., 2004b; Lin et al., 1994; Oliphant et al.,

223

2005; Oliphant et al., 2006; Stiasny et al., 2006). Some studies further defined contact

224

residues for serotype-specific, subcomplex-specific and cross-reactive monoclonal

225

antibodies that recognize domain III of the envelope protein in dengue virus; the

226

epitopes of serotype-specific neutralizing monoclonal antibodies are localized to the

227

BC, DE and FG loop on the lateral ridge of domain III, whereas subcomplex-specific

228

monoclonal antibodies recognize A strand of domain III (Gromowski & Barrett, 2007;

229

Gromowski et al., 2008; Lok et al., 2008; Rajamanonmani et al., 2009;

230

Sukupolvi-Petty et al., 2007; Thullier et al., 2001). To prove that LAE technique

231

could improve epitopes of poor antigenicity, we chose the epitope (V365 N I E A E P P

232

F G374) localized between E-strand and F-strand of domain III, which has not been

233

reported to induce any neutralizing antibody. During the antigen purification process,

234

there were some ladder-like minor bands located under the major bands (Fig. 2a). We

235

speculate that the tandem repeated sequences might affect antigen translation or

236

degradation. This tandem repeated antigen indeed induces EIII antibody production,

237

which could be utilized for basic biochemical assays, such as Western blot analysis,

238

immunofluorescence and co-immunoprecipitation. In addition, this antibody could

239

specifically detect EIII-His6 (Fig. 3a), ruling out the possibility that it recognized the

240

junction sequences between two repeated epitopes. More importantly, this antibody

241

could be also applied for detecting dengue virus virions and blocking viral infection

242

into BHK-21 cells. According to literature search, this is the first report of application

243

of LAE technique in virus-related research.

244

From epidemiological studies, it is generally believed that heterologous antibodies

245

that fail to neutralize the infecting serotype contribute primarily to the development of

246

DHF/DSS in both infants and adults (Halstead, 2003). Those sub-neutralizing

247

antibodies enhance viral uptake by Fcγ receptor (FcγR)-dependent or Fcγ-independent

248

mechanisms called antibody-dependent enhancement (ADE) (Halstead & O'Rourke,

249

1977; Huang et al., 2006). To reduce the risk of ADE, the neutralizing antibodies

250

must be protective against each of the four dengue virus serotypes. This might be

251

achieved by choosing a conserved epitope in different serotypes to produce tetravalent

252

neutralizing antibodies by LAE technique. Therefore, this is the second reason why

253

we chose a region of 10 amino acids (V365NIEAEPPFG374) from domain III of the

254

envelope protein in DENV2 as the target of epitope. This sequence is 100% conserved

255

in DENV1, DENV2 and DENV3 but is only with two amino acids difference in

256

DENV4 (Fig. S2a). Interestingly, even this minor divergence might reflect in the

257

generated EIII Ab: 2000-fold diluted EIII Ab recognizes recombinant EIII-His6 of

258

DENV1 to DENV3, but not DENV4 (Fig. S2b). However, 100-fold diluted EIII Ab

259

could detect all the recombinant EIII-His6 of four serotypes (data not shown).

260

Similarly, EIII Ab majorly recognizes recombinant EIII-His6 of DENV2 under low

261

concentration (104-fold dilution), but cross-reacts with EIII-His6 of all four serotypes

262

under high concentration (102-fold dilution) in ELISA (Fig. 2c). Surprisingly, Fig. 5

263

and Fig. S3 also show that EIII Ab specifically blocks DENV2 infection to BHK-21

264

cells although the epitope are identical in DENV1, DENV2 and DENV3. We

265

speculate this specific blockage might derive from higher avidity of EIII Ab with

266

envelope protein of DENV2, suggesting that the the antibody generated by LAE not

267

only recognizes the sequence but also discriminate structural arrange of corresponding

268

antigen.

269

To further delineate the process of blockage, we further examined whether that EIII

270

Ab blocked virus infection through the blockage of viral attachment to host receptor

271

(Fig. 5d). Our result is consistent with previous studies, which suggests that domain

272

III also play important roles in binding to the host cell receptors (Bhardwaj et al.,

273

2001; Chiu & Yang, 2003; Crill & Roehrig, 2001; Hung et al., 2004).

274

Taken together, LAE technique can be used to develop antigens with higher

275

antigenicity or antibodies with higher specificity through simple PCR-based method

276

even without any DNA template. Moreover, the LAE-produced polyclonal antibody

277

recognized the same epitope, displaying the property of monoclonal antibody and may

278

be considered as monoclone-like polyclonal antibody. Such antigens and antibodies

279

have high potential for uses in academic research, diagnosis and therapeutic

280

application in the future.

2814. Methods 282

Template-repeated PCR (TR-PCR) and adapter-PCR

283

KOD Hot Start DNA Polymerase (Novagen) was used in TR-PCR. Two

284

oligonucleotides were used in this reaction: Den E-A: 5’-GTT AAC ATC GAA GCC

285

GAA CCT CCT TTT GGT-3’ and Den E-B: 5’-GGC TTC GAT GTT AAC ACC

286

AAA AGG AGG TTC-3’. In TR-PCR, the two primers were also functional as

287

templates. The PCR programs were 30 cycles of denaturation at 94°C for 1 minute,

288

annealing at 44°C for 1 minute, and polymerization at 72°C for 1 minute, followed by

289

a final polymerization step at 72°C for 10 minutes. After TR-PCR, the PCR product

290

was then used as template for adapter-PCR reaction catalyzed by Taq polymerase

291

(Fermentas). Two oligonucleotides were used in this reaction: Den E-5b: 5’-GGA

292

TCC GTT AAC ATC GAA GCC-3’ and Den E-3he: 5’-GAA TTC ATT AAA GCT

293

TAC CAA AAG GAG GTT C-3’. The PCR condition were 5 cycles of denaturation at

294

94°C for 1 minute, annealing at 44°C for 1 minute, and polymerization at 72°C for 1

295

minute, followed by 10 cycles of denaturation at 94°C for 1 minute, annealing at 48°C

296

for 1 minute, and polymerization at 72°C for 1 minute. Finally the reaction was

297

terminated with polymerization step at 72°C for 10 minutes. The product of adaptor

298

PCR was cloned into T vector for sequencing. The correct construct was then

299

subcloned into expression vectors pGEX-His6 or pPEIa-His6.

300

Antigen and antibody preparation

301

Recombinant GST-Den EIII10-His6 protein was expressed in BL21 (DE3) with 1 mM

℃ for 3hrs followed by breaking cells with sonication. After centrifugation,

302

IPTG at 37

303

the soluble protein was purified with Glutathione-Sepharose 4B (Amersham

304

Pharmacia). One hundred or five hundred micrograms of purified GST-Den

305

EIII10-His6 proteins were mixed with Freund’s complete adjuvant (Chemicon) and

306

then subcutaneously injected into female New Zealand white rabbits four times with

307

two weeks intervals. Sera were collected from rabbits’ ears biweekly and titer was

308

tested by ELISA. The sera were purified by Millipore montage® antibody purification

309

kit.

310

Enzyme-linked immunosorbent assay (ELISA)

311

The recombinant EIII-His6 proteins are the courtesy of Dr. Chih-Hsiang Leng in

312

National Health Research Institute, Taiwan (Leng et al., 2009). The consensus

313

sequence for EIII from DENV1 was obtained by alignment of the amino acid

314

sequences from five different isolates of DENV1, including Brazil/97-11/1997,

315

Jamaica/CV1636/1977,

316

Thailand/AHF 82-80/1980. The consensus sequences for EIII from DENV2, DENV3

317

and DENV4 were obtained by alignment of the amino acid sequences from different

318

isolates, respectively. For DENV2, 16681-PDK53, China/D2-04, Jamaica/1409/1983,

319

Malaysia M2, Malaysia M3, Peru/IQT2913/1996, Thailand/0168/1979, Puerto

320

Rico/PR159-S1/1969,

321

Thailand/16681/84,

322

referenced. Strains for DENV3 included Philippines/H87/1956, China/80-2/1980,

323

Singapore/8120/1995, Martinique/1243/1999, and Sri Lanka/1266/2000. Strains for

324

DENV4

325

Thailand/0348/1991 Singapore/8976/1995, and Thailand/0476/1997. 10 ng, 100 ng

Nauru/West

Pac/1974,

Tonga/EKB194/1974 Thailand/NGS-C/1944,

included

Singapore/S275/1990

P27914, and

Philippines/H241/1956,

and

Thailand/PUO-218/1980,

Thailand/TH-36/1958

were

Dominica/814669/1981,

326

or 1000 ng of EIII-His6 proteins or dengue virus virion were coated on 96-well plates

327

with coating buffer (10 mM Na2CO3, 35 mM NaHCO3, pH 9.6) at 37

328

4

℃ for 2 hrs or

330

℃ overnight. After coating, samples were blocked by blocking solution (1% BSA and 0.0375% saponin in PBS) at 37℃ for 2 hrs or at 4℃ for overnight. The samples were incubated with EIII antibody at 37℃ for 2 hrs and washed with 120 μl PBS

331

three times. Then the samples were incubated with goat anti-rabbit IgG-HRP second

332

antibody at 37

333

substrate solution [2,2-azino-diethylbenzthiazoline sulphonic acid (ABTS) 0.54mg/ml,

334

0.1M citric acid pH 4.2, 3μl 30% H2O2/ml] was incubated with samples at room

335

temperature for 15 minutes. The signals were detected by multiscan ELISA reader

336

with A410 nm (Dynatech).

337

Cell cultures, transfection, virus strain and infection:

338

Baby hamster kidney (BHK-21) cells and human embryonic kidney (HEK293T) cells

339

were cultured at 37

340

medium (Gibco) supplemented with 10% fetal bovine serum (Biowest). C6/36, a

341

mosquito cell line established from Aedes albopictus was grown at 28

342

in RPMI 1640 medium (Gibco) supplemented with 10% FBS. Cells were transfected

343

with pCMV-3FLAG or pCMV-3FLAG-envelope using TurbofectTM reagent

344

(Fermentas). DENV1 (Myanmar 3886201 strain), DENV2 (PL046, NGC strain),

345

DENV3 (98TW503 strain) and DENV4 (H241 strain) stock was prepared in C6/36

346

cells and titrated by plaque assay. In infection experiments, appropriated cells were

347

seeded on culture plates and incubated at 37

329

℃ for 1 hour and washed three times with 120 ul PBS. Finally,

℃ with 5% CO in Dulbecco's modified Eagle's medium (DMEM) 2

℃ with 5% CO

2

℃ overnight. The next day, cells were

℃ for 2 hrs by adding dengue virus-containing culture supernatant of a

348

infected at 37

349

MOI of 0.02 or 12. After infection, cells were washed with PBS and cultured in fresh

350

medium.

351

Western blot and Co-immunoprecipitation (Co-IP):

352

Samples and pre-stained marker were subjected to a 10% or 12.5% polyacrylamide

353

gel for electrophoresis. The proteins were then electrophoretically transferred from gel

354

onto PVDF membrane by a semi-dry transfer system (Biometra). After transferring

355

onto the PVDF membranes and subsequent blocking, the blots were incubated with

356

the respective primary antibodies, followed by the secondary antibody conjugated

357

with horseradish peroxidase (HRP). Finally the signal was detected by adding

358

chemiluminescence reagent (PerkinElmer) and visualized using Chemi Genius2

359

CG2/D2A

360

immunoprecipitation, 150 micrograms cell lysates from HEK293T cells transfected

361

with empty vector or pCMV-3FLAG-envelope plasmid were immunoprecipitated with

362

EIII antibody using Dynabeads protein A system (Invitrogen), followed by Western

363

blot with FLAG antibody.

364

Immunofluorescence assay (IFA):

365

In dengue virus blocking assay, BHK-21 cells were seeded on cover glasses in

366

six-well plates (105 or 3X 105 cells per well) and incubated at 37°C for overnight. EIII

367

antibody at different dilutions was incubated with dengue virus with a MOI of 0.02 at

368

4°C for 1 hour. The virus-antibody mixture was then added to BHK-21 and incubated

369

for an additional hour at 4°C. Negative controls received PBS instead of antibody.

370

After infection, BHK-21 cells were washed three times with 1 ml cold PBS and

machine

(Syngene)

or

using

traditional

X-ray

film.

For

371

cultured in DMEM medium supplemented with 10% FBS. At different time points

372

after infection, the cells on cover glasses were then permeabilized with 0.25% Triton

373

X-100 followed by 100% methanol fixation at -20°C for 5 minutes. After fixation,

374

cells were incubated with rabbit-anti-Den2 NS3 antibody followed by incubation with

375

anti-rabbit-IgG-Alexa488 (Molecular Probes) secondary antibody. Finally, cover

376

glasses were mounted on slide with Vectashield mounting medium (Vector Labs) with

377

4, 6-diamidino-2-phenylindole (DAPI). The slides were observed by fluorescence

378

microscope or confocal microscope. In transfection experiment, pCMV-3FLAG or

379

pCMV-3FLAG-envelope plasmid was transfected to HEK293T cells by using

380

TurbofectTM transfection reagent (Fermentas). The primary antibody was EIII or

381

FLAG and the secondary antibody was anti-rabbit-IgG-Alexa488 or anti-mouse

382

IgG-Alexa555 (Molecular Probes) in this experiment.

383

Quantitative reverse transcription polymerase chain reaction (qRT-PCR):

384

Total RNA was isolated from mock infected or dengue virus infected cells by REzol

385

C&T reagent (PROTECH). 100 ng or 1μg RNA was used as template for qRT-PCR

386

using KAPA SYBR® FAST One-Step qRT-PCR kit (KAPA Biosystem) or ABI

387

qRT-PCR kit and ABI StepOne detection system and analysis software (Applied

388

Biosystems). Levels of NS3 were normalized to β-actin mRNA levels using a

389

comparative threshold cycle (2–[delta] Ct) method which converts differences of cycle

390

numbers to test gene/β-actin ratios.

391

Statistical Analysis

392

The data were analyzed using either t-test or Mann–Whitney U test and the results

393

with p value less than 0.05 were considered significant.

394 3955. Acknowledgements 396

We thank Dr. Wen Chang (Institute of Molecular Biology, Academia Sinica, Taiwan)

397

for providing plasmid pET-21C-EIII for expression of domain III protein and Dr.

398

Huey-Nan Wu (Institute of Molecular Biology, Academia Sinica, Taiwan) for

399

providing plasmid pMR-SpeI-prME-HA1-RzD for construction of full-length

400

envelope protein and NS3 antibody for Western blot. We highly appreciate

401

Chih-Hsiang Leng and Hsin-Wei Chen (National Institute of Infectious Diseases and

402

Vaccinolog, National Health Research Institute) for providing purified recombinant

403

EIII-His6 proteins of four serotypes DENV. We also thank Dr. Ying-Ray Lee, (Chia-Yi

404

Christian Hospital) for providing Dengue virus type 2 (PL046 strain) for infection

405

experiments. This study is based in part on sequence data from the Taiwan Pathogenic

406

Microorganism Genome Database provided by Centers for Disease Control, Ministry

407

of Health and Welfare, Taiwan (supported by National Research Program for Genome

408

Medicine 99-0324-01-F-12). The interpretation and conclusions contained herein do

409

not represent those of Centers for Disease Control, Ministry of Health and Welfare,

410

Taiwan. This work was supported by National Science Council grants

411

NSC93-2311-B-194-002 for Hau-Ren Chen.

4126. References 413 414 415 416

Barratt-Boyes, S. M., Vlad, A. & Finn, O. J. (1999). Immunization of chimpanzees with tumor antigen MUC1 mucin tandem repeat peptide elicits both helper and cytotoxic T-cell responses. Clinical cancer research : an official journal of the American Association for Cancer Research 5, 1918-1924.

417 418 419

Beasley, D. W. & Barrett, A. D. (2002). Identification of neutralizing epitopes within structural domain III of the West Nile virus envelope protein. J Virol 76, 13097-13100.

420 421 422

Bhardwaj, S., Holbrook, M., Shope, R. E., Barrett, A. D. & Watowich, S. J. (2001). Biophysical characterization and vector-specific antagonist activity of domain III of the tick-borne flavivirus envelope protein. J Virol 75, 4002-4007.

423 424 425

Cecilia, D. & Gould, E. A. (1991). Nucleotide changes responsible for loss of neuroinvasiveness in Japanese encephalitis virus neutralization-resistant mutants. Virology 181, 70-77.

426 427 428

Chavez, J. H., Silva, J. R., Amarilla, A. A. & Moraes Figueiredo, L. T. (2010). Domain III peptides from flavivirus envelope protein are useful antigens for serologic diagnosis and targets for immunization. Biologicals 38, 613-618.

429 430 431

Chiu, M. W. & Yang, Y. L. (2003). Blocking the dengue virus 2 infections on BHK-21 cells with purified recombinant dengue virus 2 E protein expressed in Escherichia coli. Biochem Biophys Res Commun 309, 672-678.

432 433

Chokephaibulkit, K. & Perng, G. C. (2013). Challenges for the formulation of a universal vaccine against dengue. Exp Biol Med (Maywood) 238, 566-578.

434 435

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

436 437 438

Crill, W. D. & Roehrig, J. T. (2001). Monoclonal antibodies that bind to domain III of dengue virus E glycoprotein are the most efficient blockers of virus adsorption to Vero cells. J Virol 75, 7769-7773.

439 440

Durbin, A. P. & Whitehead, S. S. (2010). Dengue vaccine candidates in development. Current topics in microbiology and immunology 338, 129-143.

441 442 443

Fonseca, B. A., Pincus, S., Shope, R. E., Paoletti, E. & Mason, P. W. (1994). Recombinant vaccinia viruses co-expressing dengue-1 glycoproteins prM and E induce neutralizing antibodies in mice. Vaccine 12, 279-285.

444 445 446 447

Goncalvez, A. P., Men, R., Wernly, C., Purcell, R. H. & Lai, C. J. (2004a). Chimpanzee Fab fragments and a derived humanized immunoglobulin G1 antibody

448 449 450

Goncalvez, A. P., Purcell, R. H. & Lai, C. J. (2004b). Epitope determinants of a chimpanzee Fab antibody that efficiently cross-neutralizes dengue type 1 and type 2 viruses map to inside and in close proximity to fusion loop of the dengue type 2 virus

that efficiently cross-neutralize dengue type 1 and type 2 viruses. J Virol 78, 12910-12918.

451

envelope glycoprotein. J Virol 78, 12919-12928.

452 453 454

Gromowski, G. D. & Barrett, A. D. (2007). Characterization of an antigenic site that contains a dominant, type-specific neutralization determinant on the envelope protein domain III (ED3) of dengue 2 virus. Virology 366, 349-360.

455 456 457

Gromowski, G. D., Barrett, N. D. & Barrett, A. D. (2008). Characterization of dengue virus complex-specific neutralizing epitopes on envelope protein domain III of dengue 2 virus. J Virol 82, 8828-8837.

458 459

Gubler, D. J. (2002). Epidemic dengue/dengue hemorrhagic fever as a public health, social and economic problem in the 21st century. Trends in microbiology 10, 100-103.

460 461 462 463

Guojun, W., Wei, G., Kedong, O., Yi, H., Yanfei, X., Qingmei, C., Yankai, Z., Jie, W., Hao, F., Taiming, L.& other authors (2008). A novel vaccine targeting gastrin-releasing peptide: efficient inhibition of breast cancer growth in vivo. Endocrine-related cancer 15, 149-159.

464 465

Halstead, S. B. (2003). Neutralization and antibody-dependent enhancement of dengue viruses. Adv Virus Res 60, 421-467.

466 467

Halstead, S. B. & O'Rourke, E. J. (1977). Antibody-enhanced dengue virus infection in primate leukocytes. Nature 265, 739-741.

468 469

Heinz, F. X. & Stiasny, K. (2012). Flaviviruses and flavivirus vaccines. Vaccine 30, 4301-4306.

470 471 472 473

Hsu, C. T., Ting, C. Y., Ting, C. J., Chen, T. Y., Lin, C. P., Whang-Peng, J. & Hwang, J. (2000). Vaccination against gonadotropin-releasing hormone (GnRH) using toxin receptor-binding domain-conjugated GnRH repeats. Cancer Res 60, 3701-3705.

474 475 476 477

Huang, K. J., Yang, Y. C., Lin, Y. S., Huang, J. H., Liu, H. S., Yeh, T. M., Chen, S. H., Liu, C. C. & Lei, H. Y. (2006). The dual-specific binding of dengue virus and target cells for the antibody-dependent enhancement of dengue virus infection. J Immunol 176, 2825-2832.

478 479 480 481

Hung, J. J., Hsieh, M. T., Young, M. J., Kao, C. L., King, C. C. & Chang, W. (2004). An external loop region of domain III of dengue virus type 2 envelope protein is involved in serotype-specific binding to mosquito but not mammalian cells. J Virol 78, 378-388.

482 483 484

Lee, H. C., Butler, M. & Wu, S. C. (2012). Using recombinant DNA technology for the development of live-attenuated dengue vaccines. Enzyme and microbial technology 51, 67-72.

485 486 487 488

Leng, C. H., Liu, S. J., Tsai, J. P., Li, Y. S., Chen, M. Y., Liu, H. H., Lien, S. P., Yueh, A., Hsiao, K. N., Lai, L. W.& other authors (2009). A novel dengue vaccine candidate that induces cross-neutralizing antibodies and memory immunity. Microbes and infection / Institut Pasteur 11, 288-295.

489 490 491

Lin, B., Parrish, C. R., Murray, J. M. & Wright, P. J. (1994). Localization of a neutralizing epitope on the envelope protein of dengue virus type 2. Virology 202, 885-890.

492 493 494 495 496

Lok, S. M., Kostyuchenko, V., Nybakken, G. E., Holdaway, H. A., Battisti, A. J., Sukupolvi-Petty, S., Sedlak, D., Fremont, D. H., Chipman, P. R., Roehrig, J. T.& other authors (2008). Binding of a neutralizing antibody to dengue virus alters the arrangement of surface glycoproteins. Nature structural & molecular biology 15, 312-317.

497 498 499

Modis, Y., Ogata, S., Clements, D. & Harrison, S. C. (2003). A ligand-binding pocket in the dengue virus envelope glycoprotein. Proc Natl Acad Sci U S A 100, 6986-6991.

500 501

Modis, Y., Ogata, S., Clements, D. & Harrison, S. C. (2004). Structure of the dengue virus envelope protein after membrane fusion. Nature 427, 313-319.

502 503

Murphy, B. R. & Whitehead, S. S. (2011). Immune response to dengue virus and prospects for a vaccine. Annual review of immunology 29, 587-619.

504 505 506 507

Oliphant, T., Engle, M., Nybakken, G. E., Doane, C., Johnson, S., Huang, L., Gorlatov, S., Mehlhop, E., Marri, A., Chung, K. M.& other authors (2005). Development of a humanized monoclonal antibody with therapeutic potential against West Nile virus. Nat Med 11, 522-530.

508 509 510 511

Oliphant, T., Nybakken, G. E., Engle, M., Xu, Q., Nelson, C. A., Sukupolvi-Petty, S., Marri, A., Lachmi, B. E., Olshevsky, U., Fremont, D. H.& other authors (2006). Antibody recognition and neutralization determinants on domains I and II of West Nile Virus envelope protein. J Virol 80, 12149-12159.

512

Rajamanonmani, R., Nkenfou, C., Clancy, P., Yau, Y. H., Shochat, S. G.,

513 514 515

Sukupolvi-Petty, S., Schul, W., Diamond, M. S., Vasudevan, S. G. & Lescar, J. (2009). On a mouse monoclonal antibody that neutralizes all four dengue virus serotypes. J Gen Virol 90, 799-809.

516 517 518

Rey, F. A., Heinz, F. X., Mandl, C., Kunz, C. & Harrison, S. C. (1995). The envelope glycoprotein from tick-borne encephalitis virus at 2 A resolution. Nature 375, 291-298.

519 520

Roehrig, J. T., Bolin, R. A. & Kelly, R. G. (1998). Monoclonal antibody mapping of the envelope glycoprotein of the dengue 2 virus, Jamaica. Virology 246, 317-328.

521 522

Schmitz, J., Roehrig, J., Barrett, A. & Hombach, J. (2011). Next generation dengue vaccines: a review of candidates in preclinical development. Vaccine 29, 7276-7284.

523 524 525

Shyu, Y. C., Lee, T. L., Wen, S. C., Chen, H., Hsiao, W. Y., Chen, X., Hwang, J. & Shen, C. K. (2007). Subcellular transport of EKLF and switch-on of murine adult beta maj globin gene transcription. Molecular and cellular biology 27, 2309-2323.

526 527 528

Stiasny, K., Kiermayr, S., Holzmann, H. & Heinz, F. X. (2006). Cryptic properties of a cluster of dominant flavivirus cross-reactive antigenic sites. J Virol 80, 9557-9568.

529 530 531 532

Sukupolvi-Petty, S., Austin, S. K., Engle, M., Brien, J. D., Dowd, K. A., Williams, K. L., Johnson, S., Rico-Hesse, R., Harris, E., Pierson, T. C.& other authors (2010). Structure and function analysis of therapeutic monoclonal antibodies against dengue virus type 2. J Virol 84, 9227-9239.

533 534 535 536 537

Sukupolvi-Petty, S., Austin, S. K., Purtha, W. E., Oliphant, T., Nybakken, G. E., Schlesinger, J. J., Roehrig, J. T., Gromowski, G. D., Barrett, A. D., Fremont, D. H.& other authors (2007). Type- and subcomplex-specific neutralizing antibodies against domain III of dengue virus type 2 envelope protein recognize adjacent epitopes. J Virol 81, 12816-12826.

538 539 540 541

Thullier, P., Demangel, C., Bedouelle, H., Megret, F., Jouan, A., Deubel, V., Mazie, J. C. & Lafaye, P. (2001). Mapping of a dengue virus neutralizing epitope critical for the infectivity of all serotypes: insight into the neutralization mechanism. J Gen Virol 82, 1885-1892.

542 543

Volk, D. E., Beasley, D. W., Kallick, D. A., Holbrook, M. R., Barrett, A. D. & Gorenstein, D. G. (2004). Solution structure and antibody binding studies of the

544 545

envelope protein domain III from the New York strain of West Nile virus. J Biol Chem 279, 38755-38761.

546 547 548

Wan, S. W., Lin, C. F., Wang, S., Chen, Y. H., Yeh, T. M., Liu, H. S., Anderson, R. & Lin, Y. S. (2013). Current progress in dengue vaccines. Journal of biomedical science 20, 37.

549 550

Wilder-Smith, A. & Gubler, D. J. (2008). Geographic expansion of dengue: the impact of international travel. The Medical clinics of North America 92, 1377-1390, x.

551 552 553 554

Yi, H., Rong, Y., Yankai, Z., Wentao, L., Hongxia, Z., Jie, W., Rongyue, C., Taiming, L. & Jingjing, L. (2006). Improved efficacy of DNA vaccination against breast cancer by boosting with the repeat beta-hCG C-terminal peptide carried by mycobacterial heat-shock protein HSP65. Vaccine 24, 2575-2584.

555 556 557

Yu, S., Wuu, A., Basu, R., Holbrook, M. R., Barrett, A. D. & Lee, J. C. (2004). Solution structure and structural dynamics of envelope protein domain III of mosquito- and tick-borne flaviviruses. Biochemistry 43, 9168-9176.

558 559 560

Zhang, Y., Corver, J., Chipman, P. R., Zhang, W., Pletnev, S. V., Sedlak, D., Baker, T. S., Strauss, J. H., Kuhn, R. J. & Rossmann, M. G. (2003). Structures of immature flavivirus particles. EMBO J 22, 2604-2613.

561 562 563

564

Figure legends

565 566

Fig. 1. The linear array epitope (LAE) technique includes TR-PCR and adapter-PCR steps

567

(a) Template-repeated PCR (TR-PCR): Oligo Den E-A and oligo Den E-B are used as

568

primers and templates for TR-PCR. (b) Primers designed for TR-PCR. The peptide

569

sequences of epitope chosen are VNIEAEPPFG. Oligo Den E-A encodes this peptide

570

and oligo Den E-B is partially complementary to oligo Den E-A as indicated. The 5’

571

half of oligo Den E-A (shown as a) is complementary to the 5’ half of oligo Den E-B

572

(shown as a’) and the 3’ half of oligo Den E-A (shown as b) is complementary to the

573

3’ half of oligo Den E-B (shown as b’). (c) Adapter-PCR: The products of TR-PCR

574

(3μl) are subjected to adapter-PCR as templates. Adapter primer Den E-5b and Den

575

E-3he introduced proper restriction enzyme sites, leading to have a Bam HI site at 5’

576

end, a Hind III/Eco RI site at 3’ end, and two stop codons at the end of coding region.

577

The PCR products are then subcloned into a T vector. (d) The products of TR-PCR

578

(lane 2) and adapter-PCR (lane 3) are analyzed on 8% PAGE with ethidium bromide

579

stain. Lane 1 and 4 indicate 100 bp DNA markers.

580 581

Fig. 2. Novel tandem-repeated antigen was used to generate anti-EIII antiserum from rabbit

582

(a) Recombinant GST-Den EIII10-His6 protein was expressed in E. coli BL21 (DE3)

583

and was purified through Glutathione-Sepharose affinity column. Elution fractions

584

were analyzed on 10% SDS-PAGE and stained with Coomassie blue. Lane N, total

585

lysates before induction. Lane I, total lysates after 3 hrs IPTG induction. Lane Wash,

586

wash fraction during column purification. Elution fractions 1 to 8 indicated fractions

587

eluted by 10 mM glutathione from affinity column. (b) The titer of anti-EIII antiserum

588

from rabbit was measured by ELISA. 10 ng, 100 ng and 1000 ng of EIII-His 6 proteins

589

from DENV2 PL046 were coated onto ELISA plate and serial dilutions of EIII Ab

590

were added into the plate to measure OD410. No antigen (Ag) coated, no primary (1’)

591

antibody and no secondary (2’) antibody are all negative control sets. (c) The titer of

592

anti-EIII antiserum from rabbit was measured by ELISA. 10 ng, 100 ng and 1000 ng

593

of recombinant EIII-His6 proteins from four serotypes were coated onto ELISA plate

594

and serial dilutions of EIII Ab were added into the plate to measure OD410. No antigen

595

(Ag) coated, no primary (1’) antibody and no secondary (2’) antibody are all negative

596

control sets.

597 598

Fig. 3. The specificity of EIII antibody was evaluated in various assays

599

(a) The specificity of EIII antibody was first examined by Western blot. Lane 1:

600

purified EIII-His6, lane 2: PEIa-His6, lane 3: PEIa-Den EIII10-His6, lane 4: TAF7-His6,

601

lane 5: NS3-C-His6, lane 6: GST-Den EIII10-His6. Arrow indicated PEIa-Den

602

EIII10-His6 and arrow head indicated GST-Den EIII10-His6, respectively. (b) BHK-21

603

cells were transfected with empty vector or FLAG-envelope plasmid. Then, cell

604

lysates were analyzed by Western blot using either anti-EIII or anti-FLAG antibodies.

605

(c) Overexpression of FLAG-envelope in HEK293T cells were analyzed by

606

immunofluorescence assay using either anti-EIII or anti-FLAG antibodies. (d)

607

HEK293T cells were transfected with empty vector or pCMV-3FLAG-envelope

608

plasmid. Then, 150 μg total protein of cell lysates were immunoprecipitated with 10,

609

20 or 50 μl EIII antibody, followed by Western blot using anti-FLAG antibody. Input

610

was loaded with 15 μg of total protein.

611

Fig. 4. EIII antibody could recognize viral envelope proteins and virions

612

(a) C6/36 cells were infected by dengue virus and the medium was collected post 10

613

to 12 days infection. The cells were lysed by 2X sample buffer and the proteins either

614

in medium fraction (med) or in the cell fraction were analyzed by Western blot. The

615

first two lanes indicate mock infection. The other six lanes indicate three independent

616

experiments (Exp.1, 2 and 3) for dengue virus infection. Filled Arrow, open arrow and

617

arrow head indicated the band of premembrane/envelope heterodimer, envelope and

618

NS3 proteins, respectively. (b) BHK-21 cells were infected by dengue virus (MOI: 12)

619

and cells were harvested after 12 or 24 hrs infection, followed by Western blot

620

analysis with EIII or NS3 antibody. (c) Virions with different dilution folds were

621

coated in ELISA plate and were detected by anti-EIII antibody.

622

Fig. 5. EIII antibody could block dengue viral entry

623

(a) Purified EIII antibodies with serial dilution were pre-mixed with DENV2 PL046

624

strain, and then the antibody-virus mixtures were used to infect BHK-21 cells. After

625

infection for 48 hrs, cells were fixed with methanol and then dengue viruses were

626

detected by anti-NS3 (1:200) antibody. The signals were quantified in (b) by counting

627

the green fluorescence positive cells/DAPI positive cells in different fields. The

628

percentage of green fluorescence positive cells were measured based on over 180

629

DAPI-positive cells examined). The data was analyzed by Mann–Whitney U test (*

630

p

Production of a neutralizing antibody against envelope protein of dengue virus type 2 using the linear array epitope technique.

Dengue virus (DENV; genus Flavivirus) contains a positive-stranded RNA genome. Binding of DENV to host cells is mediated through domain III of the vir...
759KB Sizes 0 Downloads 4 Views