1,569
Views
14
CrossRef citations to date
0
Altmetric
Research Papers

Tandem truncated rotavirus VP8* subunit protein with T cell epitope as non-replicating parenteral vaccine is highly immunogenic

, , , &
Pages 2483-2489 | Received 16 Apr 2015, Accepted 19 May 2015, Published online: 16 Sep 2015

Abstract

The two currently available live oral rotavirus vaccines, Rotarix® and RotaTeq®, are highly efficacious in the developed countries. However, the efficacy of such vaccines in resource deprived countries in Africa and Southeast Asia is low. We reported previously that a bacterially-expressed rotavirus P2-P[8] ΔVP8* subunit vaccine candidate administered intramuscularly elicited high-titers of neutralizing antibodies in guinea pigs and mice and significantly shortened the duration of diarrhea in neonatal gnotobiotic pigs upon oral challenge with virulent human rotavirus Wa strain. To further improve its vaccine potential and provide wider coverage against rotavirus strains of global and regional epidemiologic importance, we constructed 2 tandem recombinant VP8* proteins, P2-P[8] ΔVP8*-P[8] ΔVP8* and P2-P[8] ΔVP8*-P[6] ΔVP8* based on Escherichia coli expression system. The two resulting recombinant tandem proteins were highly soluble and P2-P[8] ΔVP8*-P[8] ΔVP8* was generated with high yield. Moreover, guinea pigs immunized intramuscularly by 3 doses of the P2-P[8] ΔVP8*-P[8] ΔVP8* or P2-P[8] ΔVP8*-P[6] ΔVP8* vaccine with aluminum phosphate adjuvant developed high titers of homotypic and heterotypic neutralizing antibodies against human rotaviruses bearing G1-G4, G8, G9 and G12 with P[8], P[4] or P[6] combination. The results suggest that these 2 subunit vaccines in monovalent or bivalent formulation can provide antigenic coverage to almost all the rotavirus G (VP7) types and major P (VP4) types of global as well as regional epidemiologic importance.

Introduction

Rotaviruses are the single most important cause of severe diarrheal disease in infants and young children under 5 years of age worldwide, responsible for an estimated 453,000 deaths annually, predominantly in low-income or middle-income countries.Citation1 In 2009 and again in 2013, the World Health Organization (WHO) Strategic Advisory Group of Experts (SAGE) on Immunization recommended that 2 live oral rotavirus vaccines, Rotarix® (RV1; GlaxoSmithKline Biologicals) and RotaTeq® (RV5; Merck Vaccines), be introduced into all national immunization programs for children, especially in nations where diseases related to rotaviruses indicate major threat.Citation2-4 Seventy-5 countries have introduced Rotarix® and/or RotaTeq® vaccine to the national immunization program by January 2015 according to WHO record.Citation5 These 2 vaccines have been shown to be safe and efficacious in both developed and some developing countries.Citation6-8 However, the immunogenicity and efficacy of these vaccines are substantially reduced in low-income countries of Africa, Asia and Central America,Citation9-11 where the high mortality due to rotavirus infections occurs.Citation12,13 In addition, formation of reassortant rotaviruses between vaccine strains and circulating wild-type strains or among vaccine strains can potentially increase virulence as reported recently.Citation14-17 PATH (Program for Appropriate Technology in Health, an international nonprofit organization) has launched a non-replicating rotavirus vaccine (NRRV) program (including the P2-P[8] ΔVP8* recombinant protein vaccine we reported previously)Citation18 aiming to develop next generation, non-oral rotavirus vaccines. Vaccines administered through non-oral routes may overcome factors that can weaken an oral vaccine's efficacy including co-infections in the digestive system or the presence of maternal antibodies, and remove the slight risk of intussusception associated with live-rotavirus vaccines entir-ely.Citation19-23 The latest phase I trial launched by PATH showed that healthy adult recipients parenterally administered with 10, 30 or 60 µg P2-Wa ΔVP8* (aa 65–223) using aluminum hydroxide adjuvant demonstrated >4-fold IgG and IgA antibody titer rises in almost all recipients to the immunogenic by ELISA after 3 doses vaccinations (only one recipient out of 12 did not demonstrate an IgA response).Citation24 Furthermore, NRRVs could easily be introduced into an Expanded Program on Immunization (EPI), which can be given concomitantly with other children vaccines without affecting or being affected by them, as well as being safe for the immunocompromised children.

In general, the magnitude of protection triggered by a parenteral vaccine against orally transmitted diseases depends on the levels of protective antibodies.Citation25,26 Westerman and his colleagues demonstrated that high or medium titer of specific IgG or neutralizing antibodies passively transferred to naive non-human primate by blood infusion delayed the rotavirus infection after oral challenge with virulent simian rotavirus.Citation26 It suggests that circulating IgG can provide protection against pathogens infecting the host via mucosal route and the development of parenteral rotavirus vaccine candidates is applicable for infants and children in the underdeveloped countries, where rotavirus-related diarrhea is a life-threat disease.Citation27,28 We reported previously that bacterially-expressed P2-P[8] ΔVP8* induced high levels of both homotypic and heterotypic neutralizing antibodies in guinea pigs and significantly delayed the onset and shortened the duration of diarrhea in gnotobiotic pigs vaccinated with purified P2-P[8] ΔVP8* after oral challenge with the virulent human rotavirus Wa strain.Citation18 The above mentioned phase I clinical trial launched by PATH demonstrated moderate immunogenicity of the P2-P[8] ΔVP8*(aa 65–223) vaccine in humans.Citation24 Therefore, we aimed to further increase the immunogenicity of such P2-P[8] ΔVP8* candidate vaccine. In this study, we constructed 2 fusion subunit recombinant proteins, P2-P[8] ΔVP8*-P[8] ΔVP8* and P2-P[8] ΔVP8*-P[6] ΔVP8* to provide wide coverage for human rotaviruses combined with P[8], P[4] or P[6] types of global as well as regional epidemiologic importance including new and emerging G8, G9 and G12 rotaviruses, since we previously reported that P[8] ΔVP8* induced a cross-neutralizing antibodies against human rotaviruses with P[4] specificity.Citation29 We then characterized the immunogenicity of these recombinant fusion proteins in guinea pigs. In consideration of concerns associated with live oral rotavirus vaccines stated above as well as potential advantages of non-replicating rotavirus vaccines, our tandem VP8* proteins demonstrate a high potential for a successful parenteral vaccine.

Results

Expression, purification and specificity of recombinant proteins

Each of the 2 recombinant VP8* proteins, P2-P[8] ΔVP8*-P[8] ΔVP8* and P2-P[8] ΔVP8*-P[6] ΔVP8* was expres-sed both in soluble and insoluble (inclusion bodies) forms. The expression and specificity of each purified recombinant protein was confirmed by SDS-PAGE () and Western blot analyses (). The molecular weight of each recombinant protein analyzed by SDS-PAGE was identical as expected, around 45 kilodolton. Both of recombinant proteins reacted with polyclonal antisera against Wa rotavirus strain () and P2-P[8] ΔVP8*-P[6] ΔVP8* recombinant protein reacted with polyclonal antisera against ST3 (P[6]) rotavirus strain (). An additional band at around 70 kilodolton molecular weight was observed in Western blot assay in which P2-P[8] ΔVP8*-P[6] ΔVP8* recombinant protein was recognized by antisera against Wa or ST3 (the lower band). The upper band is likely the residual bacterial proteins that reacted with the antisera.

Figure 1. Analysis by SDS-PAGE and Western blot of purified recombinant proteins P2-P[8] ΔVP8*-P[8] ΔVP8* (A and B) and P2-P[8] ΔVP8*-P[6] ΔVP8* (C, D and E). The proteins were expressed in E. coli at 18°C and purified as described in Materials and Methods. Lane M: molecular size markers. (A) SDS-PAGE analysis. Lane 1: purified P2-P[8] ΔVP8*-P[8] ΔVP8*; (B) Western blot analysis probed with guinea pig anti-Wa strain antiserum. Lane 1: P2-P[8] ΔVP8*-P[8] ΔVP8*. (C) SDS-PAGE analysis. Lane 1: purified P2-P[8] ΔVP8*-P[6] ΔVP8*. (D) Western blot analysis probed with guinea pig anti-Wa strain antiserum. Lane 1: purified P2-P[8] ΔVP8*-P[6] ΔVP8* against anti-Wa hyperimmune sera; (E) Western blot analysis probed with guinea pig anti-ST3 strain antiserum. Lane 1: purified P2-P[8] ΔVP8*-P[6] ΔVP8* against anti-ST3 hyperimmune sera. Arrows indicate the recombinant proteins.

Figure 1. Analysis by SDS-PAGE and Western blot of purified recombinant proteins P2-P[8] ΔVP8*-P[8] ΔVP8* (A and B) and P2-P[8] ΔVP8*-P[6] ΔVP8* (C, D and E). The proteins were expressed in E. coli at 18°C and purified as described in Materials and Methods. Lane M: molecular size markers. (A) SDS-PAGE analysis. Lane 1: purified P2-P[8] ΔVP8*-P[8] ΔVP8*; (B) Western blot analysis probed with guinea pig anti-Wa strain antiserum. Lane 1: P2-P[8] ΔVP8*-P[8] ΔVP8*. (C) SDS-PAGE analysis. Lane 1: purified P2-P[8] ΔVP8*-P[6] ΔVP8*. (D) Western blot analysis probed with guinea pig anti-Wa strain antiserum. Lane 1: purified P2-P[8] ΔVP8*-P[6] ΔVP8* against anti-Wa hyperimmune sera; (E) Western blot analysis probed with guinea pig anti-ST3 strain antiserum. Lane 1: purified P2-P[8] ΔVP8*-P[6] ΔVP8* against anti-ST3 hyperimmune sera. Arrows indicate the recombinant proteins.

The yield of recombinant proteins and concentration of endotoxin

The yield of purified P2-P[8] ΔVP8*-P[8] ΔVP8* recombinant protein reached ∼90mg per liter (L) of culture, whereas that of purified P2-P[8] ΔVP8*-P[6] ΔVP8* was much lower at∼5 mg/L. The endotoxin level in each tandem ΔVP8* protein was around 1.8 EU/ml (1.853 EU/ml for P2-P[8]ΔVP8*-P[8]ΔVP8* and 1.837 EU/ml for P2-P[8]ΔVP8*-P[6]ΔVP8*), which was far less than maximum recommended endotoxin levels in recombinant subunit vaccine (<20 EU/ml).Citation30

Humoral antibody measurements

Neutralizing antibody responses induced by the P2-P[8] ΔVP8*-P[8] ΔVP8* and P2-P[8] ΔVP8*-P[6] ΔVP8* protein with AP in guinea pigs after IM immunization are shown in . The recombinant protein P2-P[8] ΔVP8*-P[8] ΔVP8* induced high titers neutralizing antibodies against P[8]-specific rotavirus with G1 combination (ranged from 1:5120˜40960; GMT,1:13279.64), G3 combination (ranged from 1:1280˜20480; GMT,1:3620.38), G4 combination (ranged from 1:1280˜10240; GMT,1:3620.38), and G9 combination (ranged from 1:1280˜20480; GMT,1:3620.38). Thus, P2-P[8] ΔVP8*-P[8] ΔVP8* elicited the identical GMT neutralizing antibodies against P[8] specificity with G3, G4 or G9 combination, which was lower than G1 (GMT, 1:3620.38 vs. 1:13279.64). In addition, P2-P[8] ΔVP8*-P[8] ΔVP8* also elicited high levels of heterotypic neutralizing antibodies against P[4]-specificity with G2 (ranged from 1:5120˜20480; GMT,1:7240.77), G8 (ranged from 1:1280˜5120; GMT,1:1659.96), and G12 (ranged from 1:1280˜5120; GMT,1:2347.53). Furthermore, P2-P[8] ΔVP8*-P[6] ΔVP8* raised high titer of neutralizing antibodies against P[8] strains with G1 (ranged from 1:2560˜20480; GMT,1:4305.39), G3 (ranged from 1:1280˜5120; GMT,1:2152.69), G4 (ranged from 1:1280˜10240; GMT,1: 2560), and G9 (ranged from 1:1280˜5120; GMT, 1:1659.96), and against P[4]-specificity with G2 combination (ranged from 1:1280˜5120; GMT,1:1522.19), G8 (GMT,1:1280), and G12 (ranged from 1:640˜5120; GMT,1:1395.85). Additionally, P2-P[8] ΔVP8*-P[6] ΔVP8* induced antibodies against P[4]-specificity with G2 (GMT,1:1280) or G4 (ranged from 1:640˜1280; GMT,1:987).

Table 1. Tandem P2-P[8] ΔVP8*-P[8] ΔVP8* and P2-P[8]ΔVP8*-P[6] ΔVP8* subunit vaccine with aluminum phosphate adjuvant induced varied levels of neutralizing antibodies to indicated strains in guinea pigs

Discussion

In our previous study, recombinant P2-P[8] ΔVP8* was demonstrated to be of high immunogenicity and induced high levels of neutralizing antibodies in guinea pigs and gnotobiotic pigs and provided protection against rotavirus diarrhea in gnotobiotic pigs.Citation18 In this study, we generated tandem rotavirus VP8* recombinant protein with the universal tetanus toxoid (TT) T cell epitope P2-P[8] ΔVP8*-P[8] ΔVP8* and P2-P[8] ΔVP8*-P[6] ΔVP8* in an attempt to further improve the immunogenicity of the recombinant subunit vaccine candidates. Our study demonstrated that both of the recombinant proteins induced high titers of neutralizing antibodies against homotypic and hetertypic human rotavirus strains, which are of global and regional epidemiologic importance, such as G1, G2, G3, G4, G8, G9 and G12.

We previously reported that P[8] ΔVP8* induced high titers of cross neutralizing antibodies against P[4] rotaviruses.Citation29 In this study, we demonstrated that the tandem P2-P[8] ΔVP8*-P[8] ΔVP8* mounted higher titers of cross neutralizing antibodies against P[4] rotaviruses than P2-P[8] ΔVP8*-P[6] ΔVP8*(combined with G2, GMT7240.77 vs. 1522.19; G8, GMT1569.96 vs. 1280; G12, GMT2347.53 vs. 1395.85). The results suggests that 2 copies of P[8] ΔVP8* in tandem further enhanced cross neutralizing immune response, which was independent of the increase of molecular mass, since the P2-P[8] ΔVP8*-P[6] ΔVP8* induced the same titer of neutralizing antibody against P[6] rotavirus with P[6] ΔVP8* recombinant subunit proteinCitation29. The transform of P[6] ΔVP8* into P2-P[8] ΔVP8*-P[6] ΔVP8* did not enhance the immune response against P[6] specific rotaviruses.

It is well know that the individual B cell antigen receptors (BCR) (Ig structure) on the surface of B cells must be cross-linked simultaneously by an antigen to activate immature B cells to trigger efficent humoral immunity.Citation31,32 Repeated copies of an epitope in one antigen are able to induce the cross-linkage of BCR to enhance the specificity and levels of antibodies produced following immunization with such constructs.Citation33 For example, in a study by Kovacs-Nolan, et al. it was reported that 3 copies of human rotavirus VP8* epitope (1–10 amino acids) peptide with P2 T cell epitope induced a more potent neutralizing antibodies than one single copy of VP8* (1–10 amino acids) or even than the full-size VP8 recombinant protein.Citation34 Actually, one copy of VP8* (1–10 amino acids) itself without P2 could not induce epitope-specific immune response. Thus, repeated epitope in a single protein molecule significantly enhances antigenicity. It appears that the same mechanism applies to both peptides and subunit protein. It is speculated that 2 copies of tandem P[8] ΔVP8* with repeated epitopes may induce the cross-linkage of IgM receptors on the surface of immuture B cells and help antigen presentation in the absence of T-cell assistance, presumably enhance antigen presentation or antigen processing, resulting in increased immune response. This explains why 2 copies of tandem P[8] ΔVP8* induced higher titers of cross-neutralizing antobodies against P[4]-specific rotaviruses. Conversely, the hybrid P[8] ΔVP8*-P[6] ΔVP8* did not enhance the titer of cross-neutralizing antibodies, since it did not contain repeated epitopes in a single protein molecule. The effect of structure constraint on antigen presentation was considered when we constructed the tandem rotavirus recombinant VP8 protein. A flexible linker “GSGSG” was introduced between T cell epitope P2 and truncated VP8*, as well as in the middle of the 2 truncated VP8*s (2 copies of truncated P[8] VP8* or P[8] VP8*-P[6] VP8*) to avoid possible structure constraint. The mechanism underlying the enhancement of immunogenicity by tandem copies of the subunit proteins remains to be explored.

Both P2-P[8] ΔVP8*-P[8] ΔVP8* and P2-P[8] ΔVP8-P[6] ΔVP8* were expressed in soluble and insoluble (inclusion bodies) forms. The yield of purified P2-P[8] ΔVP8*-P[8] ΔVP8* reached 90 mg/L of culture, whereas that of P2-P[8] ΔVP8-P[6] ΔVP8* was only ∼5 mg/L culture. The difference in yield between the 2 tandem fusion proteins was in part due to the different expression systems used. The former used a pET expression system and the latter employed a SUMO expression system which required an additional purification step: a cleavage and removal of the SUMO tag off the fusion protein, resulting in additional loss of target protein. Of note, P2-P[8] ΔVP8-P[6] ΔVP8* recombinant protein from pET expression system existed exclusively in insoluble form (data not shown). In contrast, introduction of the one addtional copy of P[8] ΔVP8* to P2-P[8] ΔVP8* recombinant protein enhanced the yield of soluble protein; the yield of P2-P[8] ΔVP8* was ∼46 mg/L culture as we previously reported.Citation18 Of interest, introduction of P[6] ΔVP8* to P2-P[8] ΔVP8* in pET expression system compromised the expression of soluble protein. In this study, we mainly focused on evaluation of the immunogenicity of tandem fusion proteins, rather than the production of target proteins. Presumably, the yield of P2-P[8] ΔVP8-P[6] ΔVP8* recombinant protein can be further enhanced by optimization for pharmaceutical production. For example, the phase I trial launched by PATH has proven that the yield of P2-P[8] ΔVP8 recombinant protein can be significantly enhanced under the optimizaiton of expression system and the culture of bacteria. Based on our understanding, the low yield of P2-P[8] ΔVP8-P[6] ΔVP8* recombinant protein obtained in this study will not preclude the potential of its application later.

Despite current 2 oral human rotavirus vaccines, RotaTeq and Rotarix are proven to be safe and efficacious in selected countries the efficacy of the 2 vaccines still needs to be further enhanced, especially in resource deprived countries. Furthermore, more intensive surveillance for evaluation of the risk of intussusceptions and for applicability to immunocompromised humans are required, since both of the 2 live oral rotavirus vaccines slightly increase the risk of intussusception and RotaTeq® vaccination caused vaccine-acquired rotavirus shedding or diarrhea in severe combined immunodeficient (SCID) patients.Citation35-37 Of note, evidence from licensed vaccines has suggested that parenteral vaccines can be successful in preventing orally transmitted diseases due to poliovirus, hepatitis A and B viruses, Vibrio cholera and Salmonella typhi.Citation38-42 Accordingly, the parenteral vaccines, such as subunit vaccines, could be alternative vaccine candidates for prevention of rotavirus diseases, especially in countries where rotaviruses threaten the lives of infants and young children. Importantly, such parenteral vaccines are free from various concerns associated with live oral rotavirus vaccines and may work better in underdeveloped regions of the world where the vaccine is needed the most.

It is well known that (i) the incidence of individual G and/or P types in the same region can show a yearly fluctuation; (ii) the incidence of rotavirus G and/or P types in different regions within the same country can differ during the same year; and (iii) multiple G and P types can co-circulate within the same year.Citation43 In addition, extensive global epidemiological surveys have confirmed that G1-G4 as well as P[8] and P[4] genotypic strains are of global epidemiologic importance.Citation43,44 It is of note that uncommon G and P types causing a high incidence of human infection have been reported in various regions worldwide including G8, G9, G10, G12, and P[6].Citation45–49 Since in nature, almost all human rotavirus G types have been detected in combination with P[8], P[4] or P[6] specificity, the P2-P[8]ΔVP8*-P[8]ΔVP8* and P2-P[8]ΔVP8*-P[6]ΔVP8* vaccines generated in this study could be used at a minimum singly or preferably in bivalent formulation to provide antigenic coverage against almost all the rotavirus G (VP7) types and the 2 major P types of global as well as regional epidemiologic importance, including new and emerging G8, G9 and G12 rotaviruses.

Materials and Methods

Viruses and cell culture

Human rotavirus strains Wa (G1P[8])Citation50 and 1076 (G2P[6])Citation51 were grown in primary Africa green monkey kidney (AGMK) cells (Diagnostic Hybrids, Athens, OH). Eagle's minimum essential medium (EMEM) supplemented with 0.5 µg/ml of trypsin [2 mg/ml (W/V)] (Sigma γ-irradiated trypsin, Sigma Chemical, St. Louiis, MO), 100 IU/ml of Penicillin, 100 µg/ml of Streptomycin and 2.5 µg/ml of Amphotericin B was used as maintenance medium. Additional rotavirus strains P (G3P[8]), VA70 (G4P[8]), W161 (G9P[8]), DS-1 (G2P[4]), 1290 (G8P[4]), L26 (G12P[4]), ST3 (G4P[6]) and 69M (G8P[10]) were cultured with medium above to be used in the neutralizing assay.

Vaccine plasmid construction

Rotaviral gene cDNA of human rotavirus Wa and 1076 strains were obtained by a reverse transcription-polymerase chain reaction (RT-PCR) procedure from viral RNAs extracted using TRIzol-LS (Thermo Fisher Scientific). The primers used were designed according to the genomic sequence of the RNA segment 4 of each rotavirus strain [GenBank accession numbers: FJ423116 (Wa) and M88480 (1076)]. Oligo nucleotide sequences included the restriction endonuclease sites Nde I and Sac I to facilitate the cloning of the inserts in multiple clone sites in an expression vector. The primers for constructing truncated P2-P[8] ΔVP8* were as follows: Forward:5′-tactcatatgcagtatataaaagcaaattctaaatttataggtataactgaactaggctccggctcaggcttagatggtccttatcagcc-3′, T cell epitope P2 encoding aa 830–844 of TT is italicized, and the linker sequence encoding GSGSG is in boldface, Nde I sites (underlined), Rerverse:5′- atggatcccagaccattattaatatattcattac-3′, BamH I site (underlined). The primers for P[8] ΔVP8* were as follows: Forward: 5′-atggatccggctcaggcttagatggtccttatcagccaactac-3′ BamH I site (underlined), Rerverse: 5′-atctaagcttgacagaccattattaatatattcattac-3′, Hind III site (underlined). The P[6] ΔVP8* fragment was amplified and firstly cloned into pET28 vector to facilitate the following the process. The primers were as follows: Forward: 5′-aatggatccggctcaggcgtactcgatggtccttatcaaccaac-3′, the linker sequence encoding GSGSG is in boldface, BamH Isite (underlined), Rerverse: 5′-tactcgagtaacccagtatttatatattcattacac-3′, Xho I site (underlined). The construction of recombinant plasmid for expression of a fusion gene P2-P[8] ΔVP8*-P[6] ΔVP8* was conducted according to manufacturer's instruction (pETite N-His SUMO Kan Vector, Lucigen). The construction of expression vector, pETite- P2-P[8] ΔVP8*-P[6] ΔVP8*, was conducted with following primers: 5′- cgcgaacagattggaggtcagtatataaaagcaaattc-3′(Forward), 5′-gtggcggccgctctattataacccagtatttatatattcattac-3′(Reverse) (The detailed information for primers used in this study is also shown in Table S1). VP8* cDNA was synthesized by using Superscript III reverse transcriptase (Thermo Fisher Scientific) and reaction parameters were as follows: 100–200 ng of genomic RNA was denatured with final concentration of 15% DMSO and incubated at 94°C for 3 min, followed by chilling on ice immediately. The first strand cDNA was synthesized under manufacturer's instruction. The fusion fragment was amplified with cDNA as the template by a Phusion Hot Start High-Fidelity DNA polymerase system (Thermo Fisher Scientific). Each PCR product was purified by a Wizard® SV Gel and PCR Clean-Up System (Promega) by agarose gel electrophoresis. The two copies of P[8] ΔVP8* or P[8] ΔVP8*-P[6] ΔVP8*with P2 T cell epitope fusion fragment was cloned into pET28a vector (Novagen) harboring a 6×histidine tag for affinity column isolation of recombinant protein, creating pET28a-P2-P[8] ΔVP8*-P[8] ΔVP8* or pET28a-P2-P[8] ΔVP8*-P[6] ΔVP8*plasmid. Then the P2-P[8] ΔVP8*-P[6] ΔVP8* product was cloned into a linearized pETite vector (Lucigen) with homologous recombination strategy. The integrity and fidelity of the amplicons were confirmed by DNA sequencing.

Expression of recombinant protein

The expression plasmids pET28a-P2-P[8] ΔVP8*-P[8] ΔVP8* or pETite-P2-P[8] ΔVP8*-P[6] ΔVP8* were transformed into competent E. coli BL21(DE3)pLysS cells (pET system) or HI-control BL21(DE3) cells (pETite system) by the heat shock method. A single colony in a transform agar plate was inoculated into LB broth-containing 50 µg/ml kanamycin. When absorbance reading at 600 nm reached 0.5, the expression of each fusion protein was induced by the addition of 0.5 mM isopropyl-1-thio-β-D-galactoside at 18°C overnight. The recombinant E. coli cells were harvested by centrifugation at 10,000 ×g for 15 min at 4°C and stored at −80°C until use.

Purification of P2-P[8] ΔVP8*-P[8] ΔVP8* and P2-P[8] ΔVP8*-P[6] ΔVP8* proteins

Each protein was purified by affinity chromatography as described previously.Citation29 The SUMO tag at N-terminus of P2-P[8] ΔVP8*-P[6] ΔVP8* was removed using the SUMO express protease (Lucigen) according to the manufacturer's instruction. The purity of each recombinant protein was confirmed by SDS-PAGE, followed by a removal of imidazole in solution using a centrifugal filter unit (Millipore). The concentration of purified recombinant protein was measured by BCA assay (Thermo Fisher Scientific) according to the manufacturer's instruction. The level of endotoxin in each purified protein was quantified as previously described.Citation29 The proteins were stored at −80°C until use.

Western blot

Proteins were analyzed by 4–12% NuPAGE gel and transferred electrophoretically onto a nitrocellulose membrane (Whatman). The membrane was incubated at 4°C overnight with a hyperimmune guinea pig antiserum (1:50) raised against Wa (P[8]) or ST3 (P[6]) strain. Following washing 3 times, the membrane was incubated with a peroxidase-conjugated goat anti-guinea pig IgG (H+L) (KPL) (1:3,000) for 1 h at room temperature. The membrane was developed by the addition of 3,3′-diaminobenzidine (DAB) substrate (Sigma) as previously described.Citation29

Immunization of guinea pigs

Outbred female Hartley guinea pigs weighing 500–550 grams were purchased from Charles River Laboratories (Wilmington, MA). Guinea pigs were maintained under animal biosafety level 2 conditions in isolator cages for the entire period of each experiment. All animal experiments were done at the NIH, in compliance with the guidelines of the Institutional Animal Care and Use Committee of the National Institute of Allergy and Infectious Diseases or the National Institute of Child Health. Guinea pigs were immunized intramuscularly (IM) 3 times at 2-week intervals with 20µg of P2-P[8] ΔVP8*-P[8] ΔVP8* or P2-P[8] ΔVP8*-P[6] ΔVP8* protein with aluminum phosphate (AP) adjuvant (ADJU-PHOS™, Brenntag Biosector) (aluminum content of 100 µg/dose). Blood samples were collected prior to each immunization as well as 7 days after the third immunization.

Neutralization assay

Neutralizing antibody titer of each serum sample was determined by 60% plaque reduction neutralization (PRN) assay by using MA104 cells and 6-well plates as previously described,Citation29 except that the virus-serum mixture, after a one hour adsorption period, was left in the agarose overlay.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Supplemental material

Supplemental_Table.doc

Download MS Word (41 KB)

Acknowledgments

We thank Elias Gonzales for technical assistance. We extend our appreciation to late Dr. Albert Z. Kapikian for his support of this project.

Funding

This work was partially supported by the Intramural Research Program of the National Institute of Allergy and Infectious Diseases, National Institutes of Health, USA and National Natural Science Foundation of China (Grant No. 31201909) and Doctoral Initiating Project of Heilongjiang Province (No. LBH-Q13134).

Supplemental Material

Supplemental data for this article can be accessed on the publisher's website.

References

  • Tate JE, Burton AH, Boschi-Pinto C, Steele AD, Duque J, Parashar UD. 2008 estimate of worldwide rotavirus-associated mortality in children younger than 5 years before the introduction of universal rotavirus vaccination programmes: a systematic review and meta-analysis. Lancet Infect Dis 2012; 12:136-41; PMID:22030330; http://dx.doi.org/10.1016/S1473-3099(11)70253-5
  • World Health Organization. Meeting of the immunization Strategic Advisory Group of Experts, April 2009-conclusions and recommendations. Wkly Epidemiol Rec 2009; 84:220-36; PMID:19499606; Available at http://www.who.int/wer/2009/wer8423.pdf
  • World Health Organization. Meeting of the Strategic Advisory Group of Experts on immunization, October 2009-conclusions and recommendations. 2009; 84:517-32; PMID:19999831; Available at http://www.who.int/wer/2009/wer8450.pdf
  • World Health Organization. Rotavirus vaccines. WHO position paper-January 2013. Wkly Epidemiol Rec 2013; 84:49-64; PMID:23424730; Available at http://www.who.int/wer/2013/wer8805.pdf
  • Rota Council [Internet]. National and Regional Rotavirus Vaccine Introductions. Availabe from http://rotacouncil.org/national-and-regional-rotavirus-introductions/. Last accessed 04.05.2015
  • McCormack PL, Keam SJ. Rotavirus vaccine RIX4414 (Rotarix): a review of its use in the prevention of rotavirus gastroenteritis. Paediatr Drugs 2009; 11:75-88; PMID:19127963; http://dx.doi.org/10.2165/0148581-200911010-00025
  • Chandran A, Santosham M. RotaTeq: a three-dose oral pentavalent reassortant rotavirus vaccine. Expert Rev Vaccines 2008; 7:1475-80; PMID:19053204; http://dx.doi.org/10.1586/14760584.7.10.1475
  • Soares-Weiser K, Maclehose H, Bergman H, Ben-Aharon I, Nagpal S, Goldberg E, Pitan F, Cunliffe N. Vaccines for preventing rotavirus diarrhoea: vaccines in use. Cochrane Database Syst Rev 2012; 11:CD008521
  • Armah GE, Sow SO, Breiman RF, Dallas MJ, Tapia MD, Feikin DR, Binka FN, Steele AD, Laserson KF, Ansah NA, et al. Efficacy of pentavalent rotavirus vaccine against severe rotavirus gastroenteritis in infants in developing countries in sub-Saharan Africa: a randomised, double-blind, placebo-controlled trial. Lancet 2010; 376:606-14; PMID:20692030; http://dx.doi.org/10.1016/S0140-6736(10)60889-6
  • Madhi SA, Cunliffe NA, Steele D, Witte D, Kirsten M, Louw C, Ngwira B, Victor JC, Gillard PH, Cheuvart BB, et al. Effect of human rotavirus vaccine on severe diarrhea in African infants. N Engl J Med 2010; 362:289-98; PMID:20107214; http://dx.doi.org/10.1056/NEJMoa0904797
  • Zaman K, Dang DA, Victor JC, Shin S, Yunus M, Dallas MJ, Podder G, Vu DT, Le TP, Luby SP, et al. Efficacy of pentavalent rotavirus vaccine against severe rotavirus gastroenteritis in infants in developing countries in Asia: a randomised, double-blind, placebo-controlled trial. Lancet 2010; 376:615-23; PMID:20692031; http://dx.doi.org/10.1016/S0140-6736(10)60755-6
  • Jiang V, Jiang B, Tate J, Parashar UD, Patel MM. Performance of rotavirus vaccines in developed and developing countries. Hum Vaccin 2010; 6:532-42; PMID:20622508; http://dx.doi.org/10.4161/hv.6.7.11278
  • Chandran A, Fitzwater S, Zhen A, Santosham M. Prevention of rotavirus gastroenteritis in infants and children: rotavirus vaccine safety, efficacy, and potential impact of vaccines. Biologics 2010; 4:213-29; PMID:20714358
  • Payne DC, Edwards KM, Bowen MD, Keckley E, Peters J, Esona MD, Teel EN, Kent D, Parashar UD, Gentsch JR. Sibling transmission of vaccine-derived rotavirus (RotaTeq) associated with rotavirus gastroenteritis. Pediatrics 2010; 125:e438-41; PMID:20100758; http://dx.doi.org/10.1542/peds.2009-1901
  • Bucardo F, Rippinger CM, Svensson L, Patton JT. Vaccine-derived NSP2 segment in rotaviruses from vaccinated children with gastroenteritis in Nicaragua. Infect Genet Evol 2012; 12:1282-94; PMID:22487061; http://dx.doi.org/10.1016/j.meegid.2012.03.007
  • Boom JA, Sahni LC, Payne DC, Gautam R, Lyde F, Mijatovic-Rustempasic S, Bowen MD, Tate JE, Rench MA, Gentsch JR, et al. Symptomatic infection and detection of vaccine and vaccine-reassortant rotavirus strains in 5 children: a case series. J Infect Dis 2012; 206:1275-9; PMID:22872730; http://dx.doi.org/10.1093/infdis/jis490
  • Hemming M, Vesikari T. Detection of rotateq vaccine-derived, double-reassortant rotavirus in a 7-year-old child with acute gastroenteritis. Pediatr Infect Dis J 2014; 33:655-6; PMID:24326415; http://dx.doi.org/10.1097/INF.0000000000000221
  • Wen X, Wen K, Cao D, Li G, Jones RW, Li J, Szu S, Hoshino Y, Yuan L. Inclusion of a universal tetanus toxoid CD4(+) T cell epitope P2 significantly enhanced the immunogenicity of recombinant rotavirus DeltaVP8* subunit parenteral vaccines. Vaccine 2014; 32:4420-7; PMID:24962749; http://dx.doi.org/10.1016/j.vaccine.2014.06.060
  • Victoria JG, Wang C, Jones MS, Jaing C, McLoughlin K, Gardner S, Delwart EL. Viral nucleic acids in live-attenuated vaccines: detection of minority variants and an adventitious virus. J Virol 2010; 84:6033-40; PMID:20375174; http://dx.doi.org/10.1128/JVI.02690-09
  • Yih WK, Lieu TA, Kulldorff M, Martin D, McMahill-Walraven CN, Platt R, Selvam N, Selvan M, Lee GM, Nguyen M. Intussusception risk after rotavirus vaccination in U.S. infants. N Engl J Med 2014; 370:503-12; PMID:24422676; http://dx.doi.org/10.1056/NEJMoa1303164
  • Weintraub ES, Baggs J, Duffy J, Vellozzi C, Belongia EA, Irving S, Klein NP, Glanz JM, Jacobsen SJ, Naleway A, et al. Risk of intussusception after monovalent rotavirus vaccination. N Engl J Med 2014; 370:513-9; PMID:24422678; http://dx.doi.org/10.1056/NEJMoa1311738
  • Rha B, Tate JE, Weintraub E, Haber P, Yen C, Patel M, Cortese MM, DeStefano F, Parashar UD. Intussusception following rotavirus vaccination: an updated review of the available evidence. Expert Rev Vaccines 2014; 13:1339-48; PMID:25066368; http://dx.doi.org/10.1586/14760584.2014.942223
  • PATH. Vaccines for the Future, News from PATH's Vaccine Development Program. Seattle, WA: PATH; 2012; April:2-3; Available at http://sites.path.org/vaccinedevelopment/diarrhea-rotavirus-shigella-etec/ Last accessed 07.02.2015.
  • Cryz S, Hrro C, McNeal MM, Meyer N, DeNearing B, Cage A, Adams D, Flores J, Boslego JW, Fix A. Safety and immunogenicity of a parenterally administered rotavirus VP8 subunit vaccine in healthy adults. Eleventh International Rotavirus Symposium. New Delhi, India, 2014; 80-1
  • Offit PA, Clark HF. Maternal antibody-mediated protection against gastroenteritis due to rotavirus in newborn mice is dependent on both serotype and titer of antibody. J Infect Dis 1985; 152:1152-8; PMID:2999254; http://dx.doi.org/10.1093/infdis/152.6.1152
  • Westerman LE, McClure HM, Jiang B, Almond JW, Glass RI. Serum IgG mediates mucosal immunity against rotavirus infection. Proc Natl Acad Sci U S A 2005; 102:7268-73; PMID:15883382; http://dx.doi.org/10.1073/pnas.0502437102
  • Crawford SE, Estes MK, Ciarlet M, Barone C, O'Neal CM, Cohen J, Conner ME. Heterotypic protection and induction of a broad heterotypic neutralization response by rotavirus-like particles. J Virol 1999; 73:4813-22; PMID:10233942
  • Ciarlet M, Crawford SE, Barone C, Bertolotti-Ciarlet A, Ramig RF, Estes MK, Conner ME. Subunit rotavirus vaccine administered parenterally to rabbits induces active protective immunity. J Virol 1998; 72:9233-46; PMID:9765471
  • Wen X, Cao D, Jones RW, Li J, Szu S, Hoshino Y. Construction and characterization of human rotavirus recombinant VP8* subunit parenteral vaccine candidates. Vaccine 2012; 30:6121-6; PMID:22885016; http://dx.doi.org/10.1016/j.vaccine.2012.07.078
  • Brito LA, Singh M. Acceptable levels of endotoxin in vaccine formulations during preclinical research. J Pharm Sci 2011; 100:34-7; PMID:20575063; http://dx.doi.org/10.1002/jps.22267
  • Siemasko K, Skaggs BJ, Kabak S, Williamson E, Brown BK, Song W, Clark MR. Receptor-facilitated antigen presentation requires the recruitment of B cell linker protein to Igalpha. J Immunol 2002; 168:2127-38; PMID:11859098; http://dx.doi.org/10.4049/jimmunol.168.5.2127
  • Rosenberg AS. Effects of protein aggregates: an immunologic perspective. AAPS J 2006; 8:E501-7; PMID:17025268; http://dx.doi.org/10.1208/aapsj080359
  • Partidos C, Stanley C, Steward M. The influence of orientation and number of copies of T and B cell epitopes on the specificity and affinity of antibodies induced by chimeric peptides. Eur J Immunol 1992; 22:2675-80; PMID:1382995; http://dx.doi.org/10.1002/eji.1830221030
  • Kovacs-Nolan J, Mine Y. Tandem copies of a human rotavirus VP8 epitope can induce specific neutralizing antibodies in BALB/c mice. Biochim Biophys Acta 2006; 1760:1884-93; PMID:16978788; http://dx.doi.org/10.1016/j.bbagen.2006.07.015
  • Patel NC, Hertel PM, Estes MK, de la Morena M, Petru AM, Noroski LM, Revell PA, Hanson IC, Paul ME, Rosenblatt HM, et al. Vaccine-acquired rotavirus in infants with severe combined immunodeficiency. N Engl J Med 2010; 362:314-9; PMID:20107217; http://dx.doi.org/10.1056/NEJMoa0904485
  • Uygungil B, Bleesing JJ, Risma KA, McNeal MM, Rothenberg ME. Persistent rotavirus vaccine shedding in a new case of severe combined immunodeficiency: A reason to screen. J Allergy Clin Immunol 2010; 125:270-1; PMID:20109754; http://dx.doi.org/10.1016/j.jaci.2009.10.029
  • Klinkenberg D, Blohm M, Hoehne M, Mas Marques A, Malecki M, Schildgen V, Schneppenheim R, Muller I, Schildgen O, Kobbe R. Risk of rotavirus vaccination for children with SCID. Pediatr Infect Dis J 2015; 34:114-5; PMID:25741807; http://dx.doi.org/10.1097/INF.0000000000000507
  • Ma Y, Qin M, Hu HQ, Ji G, Feng L, Gao N, Gu J, Xie BF, He JH, Sun MB. [Immunogenicity of sabin inactivated poliovirus vaccine induced by diphtheria-tetanus-acellular pertussis and Sabin inactivated poliovirus combined vaccine]. Zhonghua Shi Yan He Lin Chuang Bing Du Xue Za Zhi 2011; 25:197-200; PMID:21977591
  • Lee SY, Hwang HS, Kim JH, Kim HH, Lee HS, Chung EH, Park SE, Ma SH, Chang JK, Guitton F, et al. Immunogenicity and safety of a combined diphtheria, tetanus, acellular pertussis, and inactivated poliovirus vaccine (DTaP-IPV) compared to separate administration of standalone DTaP and IPV vaccines: a randomized, controlled study in infants in the Republic of Korea. Vaccine 2011; 29:1551-7; PMID:21215828; http://dx.doi.org/10.1016/j.vaccine.2010.12.094
  • Kouiavskaia D, Collett MS, Dragunsky EM, Sarafanov A, Chumakov KM. Immunogenicity of inactivated polio vaccine with concurrent antiviral V-073 administration in mice. Clin Vaccine Immunol 2011; 18:1387-90; PMID:21715577; http://dx.doi.org/10.1128/CVI.05147-11
  • Frampton JE. DTaP(5)-IPV-Hib Vaccine (Pediacel(R)). Paediatr Drugs 2011; 13:401-15; PMID:21999652; http://dx.doi.org/10.2165/11207740-000000000-00000
  • Hewlett AT. Combined hepatitis A and B vaccine: providing a bright future for preventing hepatitis. Expert Opin Biol Ther 2009; 9:1235-40; PMID:19601727; http://dx.doi.org/10.1517/14712590903160639
  • Santos N, Hoshino Y. Global distribution of rotavirus serotypes/genotypes and its implication for the development and implementation of an effective rotavirus vaccine. Rev Med Virol 2005; 15:29-56; PMID:15484186; http://dx.doi.org/10.1002/rmv.448
  • Iturriza-Gomara M, Dallman T, Banyai K, Bottiger B, Buesa J, Diedrich S, Fiore L, Johansen K, Koopmans M, Korsun N, et al. Rotavirus genotypes co-circulating in Europe between 2006 and 2009 as determined by EuroRotaNet, a pan-European collaborative strain surveillance network. Epidemiol Infect 2011; 139:895-909; PMID:20707941; http://dx.doi.org/10.1017/S0950268810001810
  • Matthijnssens J, Rahman M, Van Ranst M. Two out of the 11 genes of an unusual human G6P[6]rotavirus isolate are of bovine origin. J Gen Virol 2008; 89:2630-5; PMID:18796733; http://dx.doi.org/10.1099/vir.0.2008/003780-0
  • Sanz JC, Barbas JF, Lasheras MD, Jimenez M, Ramos B, Sanchez-Fauquier A. [Detection of a rotavirus G9P[8]outbreak causing gastroenteritis in a geriatric nursing home]. Enferm Infecc Microbiol Clin 2009; 27:219-21; PMID:19249129; http://dx.doi.org/10.1016/j.eimc.2008.06.005
  • Mukherjee A, Dutta D, Ghosh S, Bagchi P, Chattopadhyay S, Nagashima S, Kobayashi N, Dutta P, Krishnan T, Naik TN, et al. Full genomic analysis of a human group A rotavirus G9P[6]strain from Eastern India provides evidence for porcine-to-human interspecies transmission. Arch Virol 2009; 154:733-46; PMID:19333549; http://dx.doi.org/10.1007/s00705-009-0363-3
  • Cunliffe NA, Ngwira BM, Dove W, Nakagomi O, Nakagomi T, Perez A, Hart CA, Kazembe PN, Mwansambo CC. Serotype g12 rotaviruses, Lilongwe, Malawi. Emerg Infect Dis 2009; 15:87-90; PMID:19116060; http://dx.doi.org/10.3201/eid1501.080427
  • Rahman M, Matthijnssens J, Yang X, Delbeke T, Arijs I, Taniguchi K, Iturriza-Gomara M, Iftekharuddin N, Azim T, Van Ranst M. Evolutionary history and global spread of the emerging g12 human rotaviruses. J Virol 2007; 81:2382-90; PMID:17166908; http://dx.doi.org/10.1128/JVI.01622-06
  • Wyatt RG, James WD, Bohl EH, Theil KW, Saif LJ, Kalica AR, Greenberg HB, Kapikian AZ, Chanock RM. Human rotavirus type 2: cultivation in vitro. Science 1980; 207:189-91; PMID:6243190; http://dx.doi.org/10.1126/science.6243190
  • Hoshino Y, Wyatt RG, Flores J, Midthun K, Kapikian AZ. Serotypic characterization of rotaviruses derived from asymptomatic human neonatal infections. J Clin Microbiol 1985; 21:425-30; PMID:2984247

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.