G Model

ARTICLE IN PRESS

JVAC 16501 1–8

Vaccine xxx (2015) xxx–xxx

Contents lists available at ScienceDirect

Vaccine journal homepage: www.elsevier.com/locate/vaccine

A novel linear neutralizing epitope of hepatitis E virus

1

Q1

2 3

Zi-Min Tang a,1 , Ming Tang b,1 , Min Zhao b , Gui-Ping Wen b , Fan Yang a , Wei Cai b , Si-Ling Wang a , Zi-Zheng Zheng a,∗ , Ning-Shao Xia a,b,∗ a State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, Fujian 361005, PR China b School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, PR China

4 5 6 7

8 22

a r t i c l e

i n f o

a b s t r a c t

9 10 11 12 13 14

Article history: Received 17 January 2015 Received in revised form 14 May 2015 Accepted 23 May 2015 Available online xxx

15

21

Keywords: Hepatitis E virus Linear epitope Nonimmunodominant Vaccine Neutralization

23

1. Introduction

16 17 18 19 20

Hepatitis E virus (HEV) is a serious public health problem that causes acute hepatitis in humans and is primarily transmitted through fecal and oral routes. The major anti-HEV antibody responses are against conformational epitopes located in a.a. 459–606 of HEV pORF2. All reported neutralization epitopes are present on the dimer domain constructed by this peptide. While looking for a neutralizing monoclonal antibody (MAb)-recognized linear epitope, we found a novel neutralizing linear epitope (L2) located in a.a. 423–437 of pORF2. Moreover, epitope L2 is proved non-immunodominant in the HEV-infection process. Using the hepatitis B virus core protein (HBc) as a carrier to display this novel linear epitope, we show herein that this epitope could induce a neutralizing antibody response against HEV in mice and could protect rhesus monkeys from HEV infection. Collectively, our results showed a novel nonimmunodominant linear neutralizing epitope of hepatitis E virus, which provided additional insight of HEV vaccine. © 2015 Published by Elsevier Ltd.

Q2 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38

Hepatitis E is one of the major acute viral hepatitides and is caused by hepatitis E virus (HEV). Outbreaks usually occur in developing countries [1,2], but sporadic cases have also been identified in Europe, the United States and Japan [3–7]. HEV is a naked, positivestrand RNA virus. HEV has nonenveloped, 32–34 nm, icosahedral virions, and the native virion has a T = 3 symmetry and was composed of 180 copies of the capsid protein [8,9]. The 7.2 kb RNA genome contains 3 open reading frames (ORFs), of which ORF2 (nt 5147–7127) encodes the main structural protein constructing the viral capsid, pORF2 [10]. The capsids of HEV constructed with pORF2 are the main target of host antibody responses. Both prokaryotic- and eukaryoticexpressed recombinant pORF2 antigens efficiently induce a host antibody response and immune protection against HEV [11,12]. The main antibody responses induced by recombinant pORF2 depend

∗ Corresponding authors at: State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, 422nd Siming South Road, Xiamen 361005, PR China. Tel.: +86 592 2880626; fax: +86 592 2181258. E-mail addresses: [email protected] (Z.-Z. Zheng), [email protected] (N.-S. Xia). 1 These authors contributed equally to this work.

on the presence of conformational epitopes located in a.a. 459–606 of pORF2 [13,14]. Additionally, in a dimeric domain constructed by pORF2, a.a. 459–606 was the target in sera from HEV infected patients [15–17]. A study of HEV capsid structures (PDB, 2ZTN, 3YIO, 2ZZQ, and 3HAG) further showed that the dimeric domain (E2s/P2 domain) constructed by a.a. 459–606 of pORF2 was highly exposed on the surface of the HEV capsid [18–22]. A prokaryotic-expressed recombinant pORF2 peptide (a.a. 368–606), p239, can self-assemble a 23 nm virus-like particle (VLP) by first constructing dimers [12]. The HEV vaccine, which uses p239 recombinant particle as a unique antigen, was demonstrated to be safe and efficacious during a large scale clinical trial [23]. In this study, we generated 30 conformational dependent and 6 linear dependent HEV-specific mouse monoclonal antibodies (MAbs) by immunization with p239 VLP (Table 1S) and found a novel neutralizing MAb 12A10. The neutralization sites had been characterized in HEV VLPs with a large panel of monoclonal antibodies [14,16,24–29]. Different from all identified neutralizing sites were conformational and were mapped in the a.a. 459–606 of pORF2 and are composed with discontinuous peptide stretches, the epitope recognized by MAb 12A10 was a linear determinant (L2) and located in a.a. 423–437 of pORF2. To improve the immunogenicity of antigens, virus-like particles (VLPs) were employed as particulate carriers. Among these, HBc protein VLP has been well characterized and used as carrier for different foreign sequences [30–32]. HBc protein consists roughly

http://dx.doi.org/10.1016/j.vaccine.2015.05.065 0264-410X/© 2015 Published by Elsevier Ltd.

Please cite this article in press as: Tang Z-M, et al. A novel linear neutralizing epitope of hepatitis E virus. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.05.065

39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64

G Model JVAC 16501 1–8

ARTICLE IN PRESS Z.-M. Tang et al. / Vaccine xxx (2015) xxx–xxx

2

76

of an assembly domain (residues 1–149) and an arginine-rich protamine-like domain (residues 150–183) [33]. The C-terminal deletion form which is truncated at 149 still can self-assemble into practically indistinguishable particles from full-length HBc particles [34,35]. And the immunogenicity of inserted foreign epitope can be considerably enhanced owing to repetitive arrays and multiple potent T helper epitopes of HBc VLPs [36]. As the 15-mer peptide lacks of immunogenicity, we used the HBc protein as a carrier to display L2 epitope as the procedure described in other studies [37,38]. Further investigation in this study indicated that the L2 epitope is a novel linear neutralizing non-immunodominant determinant on the HEV capsid.

replaced by a linker (G4 SG4 T-GS-G4 SG4 ), in which GS-encoding sequence was a BamH I site [40]. To express the HBcL1/L2/L3 fusion protein, the L1/L2/L3 gene was inserted into the pC149/mut plasmid vector [40] by anneal oligos to generate a plasmid HBcL1/L2/L3 with the primers (Table 2S), which inserted 15 amino acids between a.a. 78 and 83 of pC149/mut. Assemble the annealing reaction by mixing 1 ␮L of each oligo with 48 ␮L annealing buffer (100 mM NaCl and 50 mM HEPES pH 7.4), Incubate the mixture at 90 ◦ C for 4 min, and then at 70 ◦ C for 10 min. Slowly cool the annealed oligos to 10 ◦ C. The annealed oligo inserts can be used immediately in a ligation reaction. Recombinant HBcL1/L2/L3 and pC149/mut were prepared as described previously [40].

77

2. Materials and methods

2.6. Murine polyclonal antibody preparation

78

2.1. Cell lines, antibodies and hepatitis E virus

Six-week-old BALB/c female mice were immunized subcutaneously with 100 ␮g of the peptide fusion protein mixed with Freund’s complete adjuvant (Sigma, St. Louis, USA). This was followed by two booster immunizations with 100 ␮g of protein in an incomplete adjuvant (Sigma, St. Louis, USA) at 1 week intervals. Blood samples were taken from immunized mice before every booster immunization.

65 66 67 68 69 70 71 72 73 74 75

90

HepG2 cells (HB-8065, obtained from the ATCC, MD, USA) were grown in Dulbecco’s modified Eagle’s medium (DMEM) containing 10% fetal calf serum (both from GIBCO, California, USA) and antibiotics (100 unit/ml ampicillin and 100 unit/ml streptomycin) (Lukang, Shandong, China) at 37 ◦ C with 5% CO2 . HEV capsid protein-specific MAbs (15B2, 12A10, 1B7, and 8G12) were produced in our laboratory. Mouse MAb anti-␤-tubulin was from Santa Cruz Biotechnology. Goat anti-mouse polyclone antibody (PAb) conjugated with Alexa Fluor 680 was from Invitrogen. The virus source was stool samples from rhesus monkeys infected with genotype 1 virus (strain Xinjiang), and genotype 4 virus (strain Ch-S-1), respectively.

91

2.2. In vitro neutralization test of HEV by MAb/PAb in HepG2 cells

79 80 81 82 83 84 85 86 87 88 89

96

MAb/PAb (100 ␮g) were mixed with 105 copies of HEV virus and incubated at 37 ◦ C for 30 min. Then, the mixture was added to HepG2 cells (106 cells per well) and incubated at 37 ◦ C for 30 min. After washing three times with PBS, the presence of HEV RNA in the cells was detected by RT-PCR [39].

97

2.3. Indirect enzyme-linked immunosorbent assay (ELISA)

92 93 94 95

106

Purified recombinant proteins were used as coating antigens and were added (100 ng per well) to carbonate buffer (pH 9.6) at 37 ◦ C for 4 h. After washing once with 0.025% (v/v) Tween 20 in PBS (PBST), the proteins were blocked with 2% BSA in PBS at 37 ◦ C for 2 h. MAbs (100 ␮l) were added to the wells and incubated at 37 ◦ C for 30 min. The plates were washed five times and incubated with HRP-conjugated goat anti-mouse antibodies (Wantai, Beijing, China) at 37 ◦ C for 30 min. After washing five times, the color was developed. The absorbance was measured at 450 nm and 620 nm.

107

2.4. Competitive ELISA assay of MAb and serum

98 99 100 101 102 103 104 105

115

Microtiter plate wells were coated with 100 ng per well of purified recombinant protein as indirect ELISA. Patient serum was diluted 1:100 (v/v) in PBS prior to use. The serum (100 ␮l per well) was added to the well and incubated at 37 ◦ C for 1 h. After washing five times with PBST, the plates were incubated with 100 ␮l/well HRP-conjugated competition antibody (8G12HRP and 12A10-HRP) at 37 ◦ C for 40 min. The method of detection is described in the Indirect ELISA section.

116

2.5. Plasmid construction and production of proteins

108 109 110 111 112 113 114

117 118 119

N-terminus of HBc antigen (a.a.1-149) protein was used for present the linear epitope. In pC149/mut plasmid vector, amino acid 79–81-encoding sequence of HBc antigen (a.a. 1–149) was

2.7. In vitro neutralization test of genotype 1 p239 (p239(1)) and genotype 4 p239 (p239(4)) by MAb and PAb in HepG2 cells Antibodies (100 ␮g) were mixed with 15 ␮g of p239(1) or p239(4) and incubated at 37 ◦ C for 30 min. The mixture was added to the HepG2 cells and incubated at 4 ◦ C for 30 min. The cells were washed three times with PBS, and the proteins in the cells were tested by Western blotting. 2.8. Western blotting The protein concentration was assessed using the Bradford assay (Bio-Rad, Hercules, CA). Equal amounts of proteins were separated by SDS-PAGE and subsequently blotted onto a nitrocellulose membrane (Whatman, Dassel, Germany). After transfer, the membrane was immersed for 30 min in a blocking solution (5% non-fat milk in PBS) and washed with PBST. The membrane was incubated with the primary antibody and then washed three times with PBST. Alex Fluor 680 conjugated polyclonal antibodies (PAbs) were used as the secondary antibody. The reaction signals were quantitatively examined with an Odyssey imaging system (Li-COR, Lincoln, Nebraska). 2.9. Protection test in rhesus monkeys by using HBcL2 recombinant chimeric particles Six rhesus monkeys were used in this study. The animals were screened using an HEV Ab ELISA kit (Wantai, Beijing, China), and none showed detectable serum HEV antibodies at 1:10 serum dilution. The 6 rhesus monkeys were divided into 3 groups and immunized with 20 ␮g of p239 particles, HBc-L2 particles or HBc particles separately, in 3 doses by a deltoid muscle injection on the left forelimb (0, 30 and 120 d). Proteins were mixed with Freund’s complete adjuvant in the first dose and with an incomplete adjuvant for the two remaining doses. All of the monkeys were infected with 107 copies of genotype 1 HEV by intravenous injection. Stool samples were collected every day and tested for HEV viral genomes by RT-PCR as described. Serum samples were collected twice a week to assess levels of alanine aminotransferase (ALT) and anti-HEV IgM and IgG by ELISA (Wantai, Beijing, China). The animal experiment was designed based on the principles expressed in the “Guide for the Care and Use of Laboratory Animals” by the National Research Council of the National Academies and “Guidance for Experimental

Please cite this article in press as: Tang Z-M, et al. A novel linear neutralizing epitope of hepatitis E virus. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.05.065

120 121 122 123 124 125 126 127 128 129 130 131

132

133 134 135 136 137 138 139

140 141

142 143 144 145 146

147

148 149 150 151 152 153 154 155 156 157 158

159 160

161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 177

G Model JVAC 16501 1–8

ARTICLE IN PRESS Z.-M. Tang et al. / Vaccine xxx (2015) xxx–xxx

3

Fig. 1. Characterization of a typical antibody. (A) The typical MAbs used in this study and the location of the epitopes recognized by these MAbs. 15B2 is the MAb recognizing the linear epitope L1 located in a.a. 403–417, 12A10 is the MAb recognizing the linear epitope L2 located in a.a. 423–437, 1B7 is the MAb recognizing the linear epitope L3 located in a.a. 443–457, and 8G12 is the MAb recognizing the conformational epitope located in a.a. 459–606. (B) The capacity to block HEV infection of HepG2 cells, as described in the text. HEV infection was shown by detection of a 190 bp product from infected cells by RT-PCR. The G3PDH gene was co-amplified to serve as an internal control.

178 179 180 181

Animal Welfare and Ethical Treatment” by the Ministry of Science and Technology of the People’s Republic of China. The experimental procedures and the animal use and care protocols were approved by the Committee on Ethical Use of Animals of Xiamen University.

182

3. Results

183

3.1. Linear determinant located at a.a. 394–458 on HEV ORF2

184 185 186 187 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203

MAbs recognizing the HEV capsid were obtained using a standard murine MAb preparation protocol [25] from mice immunized with the recombinant-expressed HEV capsid VLP p239 (amino acids 368–606) (Table 1S). MAbs recognizing the linear epitopes were identified by detecting the reaction of these MAbs with completely denatured p239 and 15-mer peptides (Figs. 1S and 2S). The reactions with the 15-mer peptides showed the major linear epitopes recognized by these MAbs. These linear epitopes included 3 major independent peptides, a.a. 403–417 of pORF2, a.a. 423–437 of pORF2 and a.a. 443–457 pORF2, which were named linear epitopes L1 (a.a. 403–417), L2 (a.a. 423–437) and L3 (a.a. 443–457) in this study. Three MAbs, 15B2, 12A10 and 1B7,were chosen as the typical MAbs for further detection (Fig. 1A). 15B2 recognizes linear epitope L1, 12A10 recognizes linear epitope L2, and 1B7 recognizes linear epitope L3. 8G12, a protective and neutralizing MAb which recognizes the conformational determinant located at the dimeric domain (E2s domain, a.a. 459–606), was used as a typical antibody control [41,42]. We first detected the neutralization of these MAbs in HEV genotypes 1 and 4 after in vitro HEV infection in HepG2 cells. As shown

in Fig. 1B, 12A10 and 8G12 significantly blocked the infections, but 15B2 and 1B7 did not. The results show that 12A10 neutralized HEV genotypes 1 and 4 as well as the reported neutralizing MAb 8G12, which recognizes the conformational epitope located in a.a. 459–606 (E2s domain) of pORF2.

Fig. 2. MAb 12A10 and 8G12 binding to p239 in the presence of HEV-infected or uninfected donor sera. Competitive ELISA test for MAb 12A10 binding to p239. The sera were incubated with p239 in plates prior to the addition of HRP-conjugated 12A10 or 8G12 to block their binding. Acute- and convalescent-phase patient sera have been shown to block more than 50% of MAb 8G12 binding to p239. However, the sera could not block MAb 12A10. The negative sera were used as negative controls.

Please cite this article in press as: Tang Z-M, et al. A novel linear neutralizing epitope of hepatitis E virus. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.05.065

204 205 206 207 208

G Model JVAC 16501 1–8 4 209 210

211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229

230 231

232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 248 249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264

265 266

267 268 269

ARTICLE IN PRESS Z.-M. Tang et al. / Vaccine xxx (2015) xxx–xxx

3.2. Characterization of reactivity against epitope L2 and E2s domain in sera from HEV-infected patients We determined the variation in reactivity of 12A10 and 8G12 in the presence of HEV- or non-HEV-donor sera to determine whether the reactivity against epitope L2 and/or E2s domain in the sera. As shown in Fig. 2, pre-incubation with both acute phase and convalescent HEV patient sera interfered with the reaction of 8G12 with pORF2 (at 47.29% with acute phase HEV patient sera and 23.9% with convalescent HEV patient sera), but showed no effect on the reactivity of 12A10 (at 93.6% with acute phase HEV patient sera and 97.7% with convalescent HEV patient sera). This indicates that there was no competition between 12A10 and the HEV-infected patient sera when binding to HEV pORF2. Thus, in both acute phase and convalescent HEV patient sera, the major anti-pORF2 antibodies were against the immunodominant conformational determinant located in the E2s domain (a.a. 459–606), and the novel neutralization linear epitope L2 (recognized by 12A10) was non-immunodominant. The anti-L2 responses in sera from HEV infected patients and vaccinated donors were also detected. The result showed that sera from HEV infected patients and vaccinated donors had no responses with L2 peptide (Fig. 3S). 3.3. Immunogenicity of recombinant chimeric particles (vector: HBc 149) displaying the linear epitopes (L1, L2 and L3) To further understand the effect of epitopes L1, L2 and L3 on the active immunization process, the peptides were displayed on the HBc149 vector as described [43] to induce an anti-peptide specific antibody response in vivo. The chimeric proteins were expressed in E. coli (the identification process of the chimeric particles is described in supplemental material 6, Fig. 4S) and were used to immunize mice via subcutaneous injection. The p239(1) and p239(4) particles were used for immunization as an antiHEV immunogenicity positive control, and the HBc 149 particle was used as the negative control. The sera were obtained after 2 booster doses for further analysis of the reaction and neutralization. The reaction of sera with peptides (L1/L2/L3) was analyzed by ELISA (Fig. 3A). The reaction of sera induced by HBcL1/L2/L3 particle with their corresponding linear peptide yielded optical density (OD) readings between 1.389 and 1.907, while the sera induced by HBc particle with L1/L2/L3 peptides yielded OD readings under 0.1. The results showed that the PAbs specifically recognized the corresponding L1, L2 and L3 peptides. The neutralization of PAbs to p239 particle and the virus were analyzed as described previously. The p239 particles and virus on and in the cells were detected by Western blotting (Fig. 3B) and RTPCR, respectively (Fig. 3C). The p239 particles binding on cells were interfered to 21.85 ± 14.6% (p239(1)) and 22.97 ± 16.6% (p239(4)) by anti-HBcL2 PAbs compared with control group in Fig. 3B. The anti-HBcL2 PAb significantly blocked both the binding of p239 particle (Fig. 3B) and infection of the virus (Fig. 3C) on cells as well as in the sera of mice immunized with p239(1) and p239(4). The anti-HBcL1, anti-HBcL3, and anti-HBc-vector PAbs showed no significant influence on the binding of p239 particle and viral infection. The results from both genotypes 1 and 4 were similar, which suggested that the HBc 149 vector displayed epitope L2 well, and the novel potential neutralizing epitope L2 successfully induced an anti-HEV neutralizing antibody response in vivo. 3.4. HEV challenge of rhesus monkeys immunized with HBcL2 particles As mentioned above, the L2 peptide displayed on the HBc 149 particle showed good immunogenicity in mouse immunization. However, mice do not provide an HEV-infectable model.

Therefore, this study evaluated the protection of the HBcL2 particle against HEV infection in rhesus monkeys, which are sensitive to HEV infection. Six rhesus monkeys were divided into 3 groups and immunized with p239(1) particle (the only antigen in the HEV vaccine Hecolin® ), HBcL2 particle, or HBc particle in 3 doses by deltoid muscle injection in the left forelimb. The sera of the monkeys were collected weekly, and the anti-HEV IgG, anti-HEV IgM, anti-L2 Abs and anti-HBc Abs were examined to identify the effect of immunization. As shown in Fig. 4, immunization with HBcL2 particle induced specific anti-L2 (Fig. 4C, blue points and lines) and anti-HBc antibodies (Fig. 4D, blue points and lines) in monkeys as expected. The HBcL2 group sera got positive with L2 peptide at −5 log10 dilution at day 120. P239 particle also exhibited a low-level anti-L2 response (positive at 1:100 or 1:1000 dilution at day 120) (Fig. 4C, red points and lines). Anti-HEV IgG was efficiently induced both in monkeys immunized with p239 particle and those immunized with HBcL2 particle, and the sera from these monkeys got positive at least −4 log10 dilution after second boost. This result indicated that the HBc 149 particle displaying the L2 peptide showed good immunogenicity in rhesus monkey immunization. Thirty days after the second booster dose, all of the monkeys were infected with 107 copies of genotype 1 HEV virus by intravenous injection. The sera were collected twice per week for ALT detection, and viral RNA in the stools was detected every day until all of the parameters became negative. As shown in Fig. 5, HBcL2 particle did not protect as well as p239(1) particle. In the monkeys immunized with HBcL2 particle, viral RNA was still detected in the stools 3–15 d.p.i. (days post infection) and 5–23 d.p.i., but the fecal shedding period of HEV was much shorter than in the monkeys immunized with HBc particle (2–49 d.p.i. and 1–34 d.p.i.). The number of HEV virus copies present in stools from the rhesus monkeys immunized with HBcL2 particle was also lower than that of the HBc particle-immunized control group. The pre-peak ALT values for each monkey were: 1.27 (p239(1)-1), 1.18 (p239(1)-2), 3.81 (HBcL2-1), 2.71 (HBcL2-2), 4.53 (HBc-1), and 6.44 (HBc-2). The ALT elevation in the HBcL2 group was much lower than in the HBc particle group. The protection offered by HBcL2 particle was not as effective as candidate vaccine p239 particle; we did not use the virus in a further titer experiment. However, the results showed that active immunization with HBcL2 particle protected monkeys from HEV infection to some degree.

4. Discussion In summary, we found a novel neutralizing linear epitope L2 (a.a. 423–437) of HEV. The novel neutralizing site present at a.a. 423–437 of pORF2 was different from the reported neutralizing epitopes on the HEV capsid located in the E2s domain (a.a. 459–606). MAb 12A10, which recognized the L2 epitope, significantly neutralized the virus in an in vitro study to the same level as the neutralizing MAbs that recognized the E2s domain. Using HBc as a carrier, L2-epitope-immunized mouse serum blocked the entry of HEV into HepG2 cells, indicating that HBcL2 particle could produce antibodies similar to 12A10. Further investigation showed that the HBcL2 particle efficiently stimulated immunogenicity and protection against HEV infection in the rhesus monkey to some degree. Fecal shedding period of HEV was 16 days and the pre-peak ALT value was 3.21 in average in HBcL2 group. In the control group, fecal shedding period of HEV was 41 days and the pre-peak ALT value was 5.4 in average, which was more severe than the HBcL2 group. HBcL2 particle immunization shortened the period of HEV presentation by 60% in rhesus monkeys. Compared with p239 particle, which is the effective protein in the HEV vaccine, HBcL2 particle did not provide complete protection to immunized monkeys. However, considering that HBcL2 particle included only a single epitope and p239

Please cite this article in press as: Tang Z-M, et al. A novel linear neutralizing epitope of hepatitis E virus. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.05.065

270 271 272 273 274 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309 310

311

312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330 331 332

G Model JVAC 16501 1–8

ARTICLE IN PRESS Z.-M. Tang et al. / Vaccine xxx (2015) xxx–xxx

5

Fig. 3. Characterization of PAbs. (A) The reaction of PAbs with peptides (L1/L2/L3) as detected by ELISA. (B) The PAbs anti-p239(1), anti-p239(4), blank sera, anti-HBcL1, anti-HBcL2 and anti-HBcL3 were pre-mixed with genotype 1 HEV VLP p239(1) (left figure) and genotype 4 HEV VLP p239(4) (right figure), and then the mixture was added to HepG2 cells. Residual VLP binding was detected by Western blotting, and the data were calculated as a percentage of the binding without sera. ␤-Tubulin was detected to serve as an internal control. (C) The PAbs were pre-mixed with genotypes 1 and 4 HEV. Residual HEV binding was detected by RT-PCR. The G3PDH gene was co-amplified to serve as an internal control. Compared with the control group (without sera and with blank sera), the PAbs anti-p239(1), anti-p239(4) and anti-HBcL2 significantly blocked the binding of particles (B) and viral infection (C) of the cells.

333 334 335 336 337 338

particle includes multiple epitopes and that multi-epitope synergies protect more completely, the effect of the L2 epitope may be better than the results indicate. Synthetic peptides derived from other viruses, such as HAV [44], HBV [45,46], HCV [47], and EV71 [38], have been shown to elicit neutralizing antibodies, but this phenomenon has not been shown

for HEV. In a previous study by Meng’s group, 30-mer peptides spanning the a.a. 221–660 region of HEV pORF2 were tested in an in vitro neutralization assay; however, none of the pooled sera antipeptides demonstrated any neutralizing activity [16]. This result may be lack of immunogenicity with synthetic peptides, which is inefficient for stimulating high-titer antibody responses. In a

Please cite this article in press as: Tang Z-M, et al. A novel linear neutralizing epitope of hepatitis E virus. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.05.065

339 340 341 342 343 344

G Model JVAC 16501 1–8 6

ARTICLE IN PRESS Z.-M. Tang et al. / Vaccine xxx (2015) xxx–xxx

Fig. 4. Antibody response of rhesus monkeys to HBcL2. Six rhesus monkeys were divided into 3 groups and immunized with 20 ␮g of three proteins: p239(1) (red), HBcL2

Q3 (blue) or HBc (black) in 3 doses by deltoid muscle injection in the left forelimb (0, 30 and 120 d). The serum levels of anti-HEV IgG (A), anti-HEV IgM (B), anti-L2 Abs (C) and anti-HBc Abs (D) were detected weekly after primary immunization. The arrows mean the day when the booster injection was performed. (For interpretation of the references to color in this figure legend, the reader is referred to the web version of this article.)

345 346 347 348 349 350 351 352 353 354 355 356 357 358

previous study, HBcL2 particle could induce anti-p239 antibodies with efficient immunization, but the peptides could not (Fig. 5S). And for analyze the conformational stability of peptides and HBcL2, a comparison of the binding activity of p239, HBcL2 and L2 peptide with MAb 12A10 was conducted in a blocking assay (Fig. 6S). It indicated that the HBcL2 displayed similar epitope L2 as p239. In this study, the HBcL2 should be a better antigen to stabilize the conformation of L2 epitope and enhance immunogenicity for stimulating 12A10-like antibodies responses. Epitope L2 is non-immunodominant in the natural HEVinfection process (Fig. 2), making it difficult to find this neutralizing epitope. However, the non-immunodominance also indicates that this epitope induces low immune responses in the natural infection process, thereby preventing mutations on the epitope sequence

more than immunodominant epitopes. Such phenomenon has been found in the influenza virus [48]. The L2 epitope is linear epitope, and the linear epitopes could be displayed and constructed according to methods which had been described and been extensively applied in numbers of investigations [31,34,36–38,40,43,49–54]. Although the immune response against immunodominant epitopes is effective in the prevention of HEV [25], epitope L2 is an alternative target for HEV vaccine development that may prevent future viral mutations with the extensive use of vaccines and the increasing anti-HEV immune pressure in the population. Moreover, this study exemplified a non-immunodominant linear neutralizing epitope against a virus. In the prevention of viruses with a single serotype, such as HEV, non-immunodominant neutralizing linear epitopes show no advantage compared with immunodominant

Fig. 5. Protection by HBcL2 assessed in a rhesus monkey infection model. The course of six infected rhesus monkeys immunized with three particles (p239(1) (red), HBcL2 (blue) or HBc (black)) was monitored for 7.5 weeks by serum ALT levels, duration of virus excretion in stool and HEV antibody responses. (A) shows that no viral shedding was found in Group p239(1). However, the viral shedding in Group HBcL2 was detected from 3 to 15 and 5 to 23 days after inoculation, and viral shedding in Group HBc was detected from 2 to 49 and 1 to 34 days after inoculation. (B) shows the levels of liver enzymes (ALT) in the serum. The baseline liver enzymes were 15–27 U of ALT per liter for rhesus monkey. The pre-peak values of ALT for each monkey were: 1.27 (p239(1)-1), 1.18 (p239(1)-2), 3.81 (HBcL2-1), 2.71 (HBcL2-2), 4.53 (HBc-1), and 6.44 (HBc-2). The serum levels of anti-HEV IgG (C) and anti-HEV IgM (D) were detected after challenge. (For interpretation of the references to color in this figure legend, the reader is referred to the web version of this article.)

Please cite this article in press as: Tang Z-M, et al. A novel linear neutralizing epitope of hepatitis E virus. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.05.065

359 360 361 362 363 364 365 366 367 368 369 370 371 372

G Model JVAC 16501 1–8

ARTICLE IN PRESS Z.-M. Tang et al. / Vaccine xxx (2015) xxx–xxx

373 374 375 376 377

378

379 380 381 382 383 384 385 386 387 388 389 390 391 392

393

394

395

neutralizing epitopes. However, in viral vaccine research with a variety of mutations in the immunodominant neutralizing epitopes, such as flu, HIV, and HCV, the search for viral nonimmunodominant linear neutralizing epitopes is a potentially effective direction. 5. Conclusion In summary, a novel non-immunodominant neutralizing linear epitope L2 present in a.a. 423–437 of HEV pORF2 was identified using a neutralizing MAb 12A10. To stabilize terminates of the peptide and to enhance immunogenicity, we used HBc as a carrier. The HBcL2 recombinant chimeric particles could elicit mouse antibodies to block the entry of HEV into HepG2 cells. And the HBcL2 chimeric particle efficiently stimulated immunogenicity and protection against HEV infection in the rhesus monkey to some degree. This study suggested that the epitope L2 was nonimmunodominant in the HEV infection, it provided an example of means to identify non-immunodominant neutralizing linear epitopes. This identification process could be a potential efficient way for vaccine research of the virus with variable immunodominant neutralizing epitopes. Conflict of interest The authors declare no conflicts of interests. Acknowledgements

399

This work was supported by Chinese National High-tech R&D Program (863 program; 2011AA02A101), Xiamen Science and Technology Platform Project (3502Z20131001) and Xiamen Science and Technology Plan Project (3502Z201410045).

400

Appendix A. Supplementary data

396 397 398

401 402 403

404

405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433

Supplementary data associated with this article can be found, in the online version, at http://dx.doi.org/10.1016/j.vaccine.2015.05. 065 References [1] Chandra V, Taneja S, Kalia M, Jameel S. Molecular biology and pathogenesis of hepatitis E virus. J Biosci 2008;33:451–64. [2] Acharya SK, Panda SK, Saxena A, Gupta SD. Acute hepatic failure in India: a perspective from the East. J Gastroenterol Hepatol 2000;15:473–9. [3] Pina S, Buti M, Cotrina M, Piella J, Girones R. HEV identified in serum from humans with acute hepatitis and in sewage of animal origin in Spain. J Hepatol 2000;33:826–33. [4] Wang Y, Levine DF, Bendall RP, Teo CG, Harrison TJ. Partial sequence analysis of indigenous hepatitis E virus isolated in the United Kingdom. J Med Virol 2001;65:706–9. [5] Mast EE, Kuramoto IK, Favorov MO, Schoening VR, Burkholder BT, Shapiro CN, et al. Prevalence of and risk factors for antibody to hepatitis E virus seroreactivity among blood donors in Northern California. J Infect Dis 1997;176:34–40. [6] Thomas DL, Yarbough PO, Vlahov D, Tsarev SA, Nelson KE, Saah AJ, et al. Seroreactivity to hepatitis E virus in areas where the disease is not endemic. J Clin Microbiol 1997;35:1244–7. [7] Suzuki K, Aikawa T, Okamoto H. Fulminant hepatitis E in Japan. N Engl J Med 2002;347:1456. [8] Xing L, Kato K, Li T, Takeda N, Miyamura T, Hammar L, et al. Recombinant hepatitis E capsid protein self-assembles into a dual-domain T= 1 particle presenting native virus epitopes. Virology 1999;265:35–45. [9] Mori Y, Matsuura Y. Structure of hepatitis E viral particle. Virus Res 2011;161:59–64. [10] Tam AW, Smith MM, Guerra ME, Huang C-C, Bradley DW, Fry KE, et al. Hepatitis E virus (HEV): Molecular cloning and sequencing of the full-length viral genome. Virology 1991;185:120–31. [11] Li T-C, Yamakawa Y, Suzuki K, Tatsumi M, Razak M, Uchida T, et al. Expression and self-assembly of empty virus-like particles of hepatitis E virus. J Virol 1997;71:7207–13.

7

[12] Li SW, Zhang J, Li YM, Ou SH, Huang GY, He ZQ, et al. A bacterially expressed particulate hepatitis E vaccine: antigenicity, immunogenicity and protectivity on primates. Vaccine 2005;23:2893–901. [13] Li T-C, Suzaki Y, Ami Y, Dhole TN, Miyamura T, Takeda N. Protection of cynomolgus monkeys against HEV infection by oral administration of recombinant hepatitis E virus-like particles. Vaccine 2004;22:370–7. [14] Zhao Q, Zhang J, Wu T, Li SW, Ng MH, Xia NS, et al. Antigenic determinants of hepatitis E virus and vaccine-induced immunogenicity and efficacy. J Gastroenterol 2013;48:159–68. [15] Riddell MA, Li F, Anderson DA. Identification of immunodominant and conformational epitopes in the capsid protein of hepatitis E virus by using monoclonal antibodies. J Virol 2000;74:8011–7. [16] Meng J, Dai X, Chang JC, Lopareva E, Pillot J, Fields HA, et al. Identification and characterization of the neutralization epitope (s) of the hepatitis E virus. Virology 2001;288:203–11. [17] Gu Y, Zhang J, Wang Y-B, Li S-W, Yang H-J, Luo W-X, et al. Selection of a peptide mimicking neutralization epitope of hepatitis E virus with phage peptide display technology. World J Gastroenterol 2004;10:1583–8. [18] Yamashita T, Mori Y, Miyazaki N, Cheng RH, Yoshimura M, Unno H, et al. Biological and immunological characteristics of hepatitis E virus-like particles based on the crystal structure. Proc Natl Acad Sci 2009;106:12986–91. [19] Li S, Tang X, Seetharaman J, Yang C, Gu Y, Zhang J, et al. Dimerization of hepatitis E virus capsid protein E2s domain is essential for virus–host interaction. PLoS Pathogens 2009;5:e1000537. [20] Xing L, Li T-C, Mayazaki N, Simon MN, Wall JS, Moore M, et al. Structure of hepatitis E virion-sized particle reveals an RNA-dependent viral assembly pathway. J Biol Chem 2010;285:33175–83. [21] Guu TS, Liu Z, Ye Q, Mata DA, Li K, Yin C, et al. Structure of the hepatitis E viruslike particle suggests mechanisms for virus assembly and receptor binding. Proc Natl Acad Sci USA 2009;106:12992–7. [22] Tang X, Yang C, Gu Y, Song C, Zhang X, Wang Y, et al. Structural basis for the neutralization and genotype specificity of hepatitis E virus. Proc Natl Acad Sci USA 2011;108:10266–71. [23] Zhu F-C, Zhang J, Zhang X-F, Zhou C, Wang Z-Z, Huang S-J, et al. Efficacy and safety of a recombinant hepatitis E vaccine in healthy adults: a largescale, randomised, double-blind placebo-controlled, phase 3 trial. The Lancet 2010;376:895–902. [24] Schofield D, Glamann J, Emerson S, Purcell R. Identification by phage display and characterization of two neutralizing chimpanzee monoclonal antibodies to the hepatitis E virus capsid protein. J Virol 2000;74:5548–55. [25] Zhang J, Gu Y, Ge SX, Li SW, He ZQ, Huang GY, et al. Analysis of hepatitis E virus neutralization sites using monoclonal antibodies directed against a virus capsid protein. Vaccine 2005;23:2881–92. [26] Zhang H, Dai X, Shan X, Meng J. The Leu477 and Leu613 of ORF2-encoded protein are critical in forming neutralization antigenic epitope of hepatitis E virus genotype 4. Cell Mol Immunol 2008;5:447–56. [27] Takahashi M, Hoshino Y, Tanaka T, Takahashi H, Nishizawa T, Okamoto H. Production of monoclonal antibodies against hepatitis E virus capsid protein and evaluation of their neutralizing activity in a cell culture system. Archiv Virol 2008;153:657–66. [28] Xing L, Wang JC, Li T-C, Yasutomi Y, Lara J, Khudyakov Y, et al. Spatial configuration of hepatitis E virus antigenic domain. J Virol 2011;85: 1117–24. [29] He J, Kuschner RA, Dewar V, Voet P, Asher LV, Vaughn DW. Characterization of monoclonal antibodies to hepatitis E virus (HEV) capsid protein and identification of binding activity. J Biomed Sci 2007;14:555–63. [30] Ludwig C, Wagner R. Virus-like particles—universal molecular toolboxes. Curr Opin Biotechnol 2007;18:537–45. [31] Pumpens P, Grens E. HBV core particles as a carrier for B cell/T cell epitopes. Intervirology 2000;44:98–114. [32] Peek LJ, Middaugh CR, Berkland C. Nanotechnology in vaccine delivery. Adv Drug Del Rev 2008;60:915–28. [33] Böttcher B, Wynne S, Crowther R. Determination of the fold of the core protein of hepatitis B virus by electron cryomicroscopy; 1997. [34] Pumpens P, Grens E. Hepatitis B core particles as a universal display model: a structure-function basis for development. FEBS Lett 1999;442:1–6. [35] Crowther R, Kiselev N, Böttcher B, Berriman J, Borisova G, Ose V, et al. Threedimensional structure of hepatitis B virus core particles determined by electron cryomicroscopy. Cell 1994;77:943–50. [36] GuangDi L, Bin W, Zuo-Yi C, Yuan W, Yue-Ting G. The Expression of HCG epitope fused to hepatitis B virus core antigen. Acta Biochim Biophys Sin 1996;28:177–86. [37] Yoshikawa A, Tanaka T, Hoshi Y, Kato N, Tachibana K, Iizuka H, et al. Chimeric hepatitis B virus core particles with parts or copies of the hepatitis C virus core protein. J Virol 1993;67:6064–70. [38] Xu L, He D, Li Z, Zheng J, Yang L, Yu M, et al. Protection against Lethal Enterovirus 71 challenge in mice by a recombinant vaccine candidate containing a broadly cross-neutralizing epitope within the VP2 EF Loop. Theranostics 2014;4:498. [39] Jothikumar N, Cromeans TL, Robertson BH, Meng X, Hill VR. A broadly reactive one-step real-time RT-PCR assay for rapid and sensitive detection of hepatitis E virus. J Virol Methods 2006;131:65–71. [40] Yang H-J, Chen M, Cheng T, He S-Z, Li S-W, Guan B-Q, et al. Expression and immunoactivity of chimeric particulate antigens of receptor binding site-core antigen of hepatitis B virus. World J Gastroenterol 2005;11:492–7. [41] Gu Y, Tang X, Zhang X, Song C, Zheng M, Wang K, et al. Structural basis for the neutralization of hepatitis E virus by a cross-genotype antibody. Cell Res 2015.

Please cite this article in press as: Tang Z-M, et al. A novel linear neutralizing epitope of hepatitis E virus. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.05.065

434 435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519

G Model JVAC 16501 1–8 8 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539

ARTICLE IN PRESS Z.-M. Tang et al. / Vaccine xxx (2015) xxx–xxx

[42] He S, Miao J, Zheng Z, Wu T, Xie M, Tang M, et al. Putative receptor-binding sites of hepatitis E virus. J Gen Virol 2008;89:245–9. [43] Pumpens P, Borisova G, Crowther R, Grens E. Hepatitis B virus core particles as epitope carriers. Intervirology 1995;38:63–74. [44] Emini EA, Hughes JV, Perlow DS, Boger J. Induction of hepatitis A virus-neutralizing antibody by a virus-specific synthetic peptide. J Virol 1985;55:836–9. [45] Przewlocki G, Audibert F, Jolivet M, Chedid L, Kent SBH, Neurath AR. Production of antibodies recognizing a hepatitis B virus (HBV) surface antigen by administration of Murabutide associated to a synthetic pre-S HBV peptide conjugated to a toxoid carrier. Biochem Biophys Res Commun 1986;140: 557–64. [46] Neurath A, Adamowicz P, Kent S, Riottot M, Strick N, Parker K, et al. Characterization of monoclonal antibodies specific for the pre-S2 region of the hepatitis B virus envelope protein. Mol Immunol 1986;23:991–7. [47] Farci P, Shimoda A, Wong D, Cabezon T, De-Gioannis D, Strazzera A, et al. Prevention of hepatitis C virus infection in chimpanzees by hyperimmune serum against the hypervariable region 1 of the envelope 2-protein. Proc Natl Acad Sci 1996;93:15394–9. [48] Rathore U, Kesavardhana S, Mallajosyula VVA, Varadarajan R. Immunogen design for HIV-1 and influenza. Biochim Biophys Acta 2014;1844:1891–906.

[49] Zhang H, Qian P, Liu L, Qian S, Chen H, Li X. Virus-like particles of chimeric recombinant porcine circovirus type 2 as antigen vehicle carrying foreign epitopes. Viruses 2014;6:4839–55. [50] Wang J, Michel V, Leclerc C, Hofnung M, Charbit A. Immunogenicity of viral B-cell epitopes inserted into two surface loops of the Escherichia coli K12 LamB protein and expressed in an attenuated aroA strain of Salmonella typhimurium. Vaccine 1999;17:1–12. [51] Langeveld JP, Martinez-Torrecuadrada J, Boshuizen RS, Meloen RH, Ignacio Casal J. Characterisation of a protective linear B cell epitope against feline parvoviruses. Vaccine 2001;19:2352–60. [52] Bhatia B, Solanki AK, Kaushik H, Dixit A, Garg LC. B-cell epitope of beta toxin of Clostridium perfringens genetically conjugated to a carrier protein: expression, purification and characterization of the chimeric protein. Protein Expr Purification 2014;102:38–44. [53] Ye X, Ku Z, Liu Q, Wang X, Shi J, Zhang Y, et al. Chimeric virus-like particle vaccines displaying conserved enterovirus 71 epitopes elicit protective neutralizing antibodies in mice through divergent mechanisms. J Virol 2014;88:72–81. [54] Stepanova LA, Kotlyarov RY, Kovaleva AA, Potapchuk MV, Korotkov AV, Sergeeva MV, et al. Protection against multiple influenza A virus strains induced by candidate recombinant vaccine based on heterologous M2e peptides linked to flagellin. PLoS One 2015;10:e0119520.

Please cite this article in press as: Tang Z-M, et al. A novel linear neutralizing epitope of hepatitis E virus. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.05.065

540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560

A novel linear neutralizing epitope of hepatitis E virus.

Hepatitis E virus (HEV) is a serious public health problem that causes acute hepatitis in humans and is primarily transmitted through fecal and oral r...
2MB Sizes 0 Downloads 9 Views