Clin Exp Vaccine Res.  2017 Jul;6(2):135-145. 10.7774/cevr.2017.6.2.135.

Preferential production of IgM-secreting hybridomas by immunization with DNA vaccines coding for Ebola virus glycoprotein: use of protein boosting for IgG-secreting hybridoma production

Affiliations
  • 1BK21 Plus Graduate Program and Department of Microbiology, School of Medicine, Kangwon National University, Chuncheon, Korea. jsin1964@hanmail.net

Abstract

PURPOSE
The goal of this study was to investigate the utility of DNA vaccines encoding Ebola virus glycoprotein (GP) as a vaccine type for the production of GP-specific hybridomas and antibodies.
MATERIALS AND METHODS
DNA vaccines were constructed to express Ebola virus GP. Mice were injected with GP DNA vaccines and their splenocytes were used for hybridoma production. Enzyme-linked immunosorbent assays (ELISAs), limiting dilution subcloning, antibody purification methods, and Western blot assays were used to select GP-specific hybridomas and purify monoclonal antibodies (MAbs) from the hybridoma cells.
RESULTS
Twelve hybridomas, the cell supernatants of which displayed GP-binding activity, were selected by ELISA. When purified MAbs from 12 hybridomas were tested for their reactivity to GP, 11 MAbs, except for 1 MAb (from the A6-9 hybridoma) displaying an IgG2a type, were identified as IgM isotypes. Those 11 MAbs failed to recognize GP. However, the MAb from A6-9 recognized the mucin-like region of GP and remained reactive to the antigen at the lowest tested concentration (1.95 ng/mL). This result suggests that IgM-secreting hybridomas are predominantly generated by DNA vaccination. However, boosting with GP resulted in greater production of IgG-secreting hybridomas than GP DNA vaccination alone.
CONCLUSION
DNA vaccination may preferentially generate IgM-secreting hybridomas, but boosting with the protein antigen can reverse this propensity. Thus, this protein boosting approach may have implications for the production of IgG-specific hybridomas in the context of the DNA vaccination platform. In addition, the purified monoclonal IgG antibodies may be useful as therapeutic antibodies for controlling Ebola virus infection.

Keyword

Ebola virus; Hybridoma; DNA vaccines; Glycoprotein; Antibody production

MeSH Terms

Animals
Antibodies
Antibodies, Monoclonal
Antibody Formation
Blotting, Western
Clinical Coding*
DNA*
Ebolavirus*
Enzyme-Linked Immunosorbent Assay
Glycoproteins*
Hemorrhagic Fever, Ebola
Hybridomas*
Immunization*
Immunoglobulin G
Immunoglobulin M
Mice
Vaccination
Vaccines, DNA*
Antibodies
Antibodies, Monoclonal
DNA
Glycoproteins
Immunoglobulin G
Immunoglobulin M
Vaccines, DNA

Figure

  • Fig. 1 Production of glycoprotein (GP) DNA vaccines (A), immunization schemes using GP DNA vaccines (B), and the evaluation of GP-specific antibody production by GP DNA vaccination (C, D). (A) pcDNA3-GP encoding Zaire Ebola virus strain Gabon-94 virion spike GP was constructed as described in the “Materials and Methods.” (B, C) BALB/c mice (n = 5/group) were injected by intramuscular-electroporation with pcDNA3-GP (50 µg/mouse) at 0, 1, and 3 weeks. The mice were bled at 1 week following the final injection and sera were collected (B). The sera were reacted with recombinant Ebola virus GP (0.5 µg/mL) in an enzyme-linked immunosorbent assay (C). The optical density (OD) values were measured at 405 nm. The values and bars represent OD values and standard deviation, respectively. (D) Recombinant Ebola virus GP (3 µg) was separated on a 15% sodium dodecyl sulfate–polyacrylamide gel. The right arrow indicates a 140-kDa GP. *p < 0.05 compared with the negative control.

  • Fig. 2 Production of monoclonal antibodies–secreting hybridomas by Ebola virus glycoprotein (GP) DNA vaccination. BALB/c mice were injected by intramuscular-electroporation with pcDNA3-GP (50 µg/muse) at 0, 1, and 3 weeks. The mice were sacrificed at 1, 2, and 3 weeks following the final injection and the splenocytes were used for fusion (as shown in Fig. 1B). Among a total of 960 hybridomas, 12 (A6, A13, C21, C22, E53, G3, G29, G44, J10, M65, N47, and N52) with reactivity to Ebola virus GP in an enzyme-linked immunosorbent assay (secondary Ab, horseradish peroxidase [HRP]–conjugated anti-mouse IgG [H+L]) were selected. These hybridomas were subcloned with the limiting dilution subcloning method. The resulting monoclonal hybridoma clones were designated A6-9, A13-9, C21-2, C22-2, E53-2, G3-2, G29-5, G44-2, J10-2, M65-16, N47-9, and N52-7. For enzyme-linked immunosorbent assay, cell supernatants from the 12 monoclonal clones were reacted with GP, followed by detection with HRP-conjugated anti-mouse IgG (H+L) (A), HRP-conjugated anti-mouse IgA and HRP-conjugated IgM (B), and HRP-conjugated anti-mouse IgG (Fc-specific) (C) secondary antibodies. The positive control was GP-specific sera (1:10 dilution) from Fig. 1C, whereas the negative control was naive sera (1:10 dilution). (D) Cell supernatants (20 µL) were loaded onto a 12% sodium dodecyl sulfate–polyacrylamide gel, followed by Western blot assay using HRP-conjugated anti-mouse IgG (H+L). OD, optical density.

  • Fig. 3 The structure of the whole Ebola virus glycoprotein (GP) and the amino acid sequence numbers of recombinant GPs spanning GP1 region. The signal peptide (SP), the putative receptor-binding domain, mucin-like region, furin cleavage site and trans-membrane region (TMR) are shown. GP1a, GP1b, GP1c, and GP1d are recombinant proteins spanning the putative receptor-binding domains of GP1, whereas GP1e is a recombinant protein spanning the mucin-like region of GP1. The amino acid sequence numbers are also displayed.

  • Fig. 4 Production of recombinant GP1a-e (A), the binding activity of monoclonal antibodies (MAbs) in cell supernatants to the recombinant GP1a and GP1e (B), purification of IgG and IgM (C), and their binding to the recombinant GP1a and GP1e (D). (A) The recombinant proteins GP1a, GP1b, GP1c, GP1d, and GP1e were produced as described in the “Materials and Methods.” GP1a, GP1b, GP1c, GP1d, and GP1e (2 µg) were loaded on an 8% sodium dodecyl sulfate–polyacrylamide gel. (B) We assessed the binding activity of the MAbs in the hybridoma cell supernatants to the recombinant GP1a and GP1e by enzyme-linked immunosorbent assay. Horseradish peroxidase (HRP)–conjugated anti-mouse IgG (H+L) was used as the secondary antibody (Ab). (C) Hybridoma cells that produced MAbs were cultured, and the cell supernatants were collected for Ab purification. Some hybridoma cells (C21-2, C22-2, G44-2, and M65-16) were injected into nude mice for ascites fluid production. IgG was purified from cell supernatants using the protein G-resin column (for A6-9). IgM was purified either from ascites fluids using the HiTrap IgM purification kit (for C21-2, C22-2, G44-2, and M65-16) or from cell supernatants using the protein L resin columns (for A13-9, E53-2, G3-2, G29-5, J10-2, N47-9, and N52-7). The purified Abs (2 µg) were run on a 12% sodium dodecyl sulfate–polyacrylamide gel for Brilliant Blue R250 staining. (D) The purified IgG and IgM (2 µg/mL) Abs were reacted with GP1a and GP1e in an enzyme-linked immunosorbent assay using HRP-conjugated anti-IgG (H+L) as the secondary antibody. The positive control was GP-specific sera (1:10 dilution) from Fig. 1C, whereas the negative control was naive sera (1:10 dilution). OD, optical density.

  • Fig. 5 Evaluation of glycoprotein (GP)-specific IgG subclass (A) and its lowest concentration for GP binding (B). (A) Purified IgG (2 µg/mL) from clone A6-9 was reacted with GP1e, followed by incubation with horseradish peroxidase (HRP)–conjugated anti-IgG1, HRP-conjugated anti-IgG2a, HRP-conjugated anti-IgG2b and HRP-conjugated anti-IgG3 by enzyme-linked immunosorbent assay (ELISA), to determine its IgG subclass. (B) IgG from clone A6-9 (2 µg/mL) was serially diluted by 2-fold, and then reacted with GP1e in parallel with control IgG (2 µg/mL) to determine its lowest concentration for GP binding using ELISA. OD, optical density.

  • Fig. 6 Effects of booster-injections with glycoprotein (GP) DNA vaccines vs. GP on the production of IgM- and IgG-secreting hybridomas. BALB/c mice were immunized by intramuscular-electroporation (IM-EP) with pcDNA3-GP (50 mg/mouse) at 0 and 4 weeks. The mice received a booster-injection at 12 weeks with either GP DNA vaccines by IM-EP or with GP by intravenous delivery. (A, B) One week after booster injection with GP DNA vaccines, the mice were sacrificed and their splenocytes were used for hybridoma production. Among the numerous hybridomas, six were selected, which had cell supernatants with optical density (OD) values at least 6× higher than those of the negative control in an enzyme-linked immunosorbent assay (ELISA) with horseradish peroxidase (HRP)–conjugated anti-mouse IgG (H+L). Cell supernatants from the six hybridomas were next reacted with GP in an ELISA using HRP-conjugated anti-mouse IgM. The OD values of the six hybridomas are shown (A). Cell supernatants from the six hybridomas were tested for the presence of IgG and IgM by Western blot assay as in Fig. 2D (B). (C, D) Experiments were performed as in Fig. 6A on splenocytes harvested 3 days after intravenous injection with GP. Subsequently, 12 hybridomas were selected. The OD values of the 12 hybridomas are shown (C). (D) Cell supernatants (20 µL) from the 12 hybridomas were loaded onto a 12% sodium dodecyl sulfate–polyacrylamide gel, followed by Western blot assay using HRP-conjugated anti-mouse IgG (H+L). The positive control was GP-specific sera (1:10 dilution) from Fig. 1C, whereas the negative control was naive sera (1:10 dilution).


Cited by  1 articles

Purification and characterization of monoclonal IgG antibodies recognizing Ebola virus glycoprotein
Baek-Sang Han, Ho-Young Jang, Trina Racine, Xiangguo Qiu, Jeong-Im Sin
Clin Exp Vaccine Res. 2018;7(2):119-128.    doi: 10.7774/cevr.2018.7.2.119.


Reference

1. Roshania R, Mallow M, Dunbar N, et al. Successful implementation of a multicountry clinical surveillance and data collection system for Ebola virus disease in west Africa: findings and lessons learned. Glob Health Sci Pract. 2016; 4:394–409. PMID: 27688716.
Article
2. Wilson JA, Hevey M, Bakken R, et al. Epitopes involved in antibody-mediated protection from Ebola virus. Science. 2000; 287:1664–1666. PMID: 10698744.
Article
3. Parren PW, Geisbert TW, Maruyama T, Jahrling PB, Burton DR. Pre- and postexposure prophylaxis of Ebola virus infection in an animal model by passive transfer of a neutralizing human antibody. J Virol. 2002; 76:6408–6412. PMID: 12021376.
Article
4. Leroy EM, Baize S, Volchkov VE, et al. Human asymptomatic Ebola infection and strong inflammatory response. Lancet. 2000; 355:2210–2215. PMID: 10881895.
Article
5. Qiu X, Audet J, Wong G, et al. Successful treatment of ebola virus-infected cynomolgus macaques with monoclonal antibodies. Sci Transl Med. 2012; 4:138ra81.
Article
6. Qiu X, Audet J, Wong G, et al. Sustained protection against Ebola virus infection following treatment of infected nonhuman primates with ZMAb. Sci Rep. 2013; 3:3365. PMID: 24284388.
Article
7. Aihara H, Miyazaki J. Gene transfer into muscle by electroporation in vivo. Nat Biotechnol. 1998; 16:867–870. PMID: 9743122.
Article
8. Rizzuto G, Cappelletti M, Maione D, et al. Efficient and regulated erythropoietin production by naked DNA injection and muscle electroporation. Proc Natl Acad Sci U S A. 1999; 96:6417–6422. PMID: 10339602.
Article
9. Ahlen G, Soderholm J, Tjelle T, et al. In vivo electroporation enhances the immunogenicity of hepatitis C virus nonstructural 3/4A DNA by increased local DNA uptake, protein expression, inflammation, and infiltration of CD3+ T cells. J Immunol. 2007; 179:4741–4753. PMID: 17878373.
Article
10. Lee IH, Park JB, Cheong M, Choi YS, Park D, Sin JI. Antitumor therapeutic and antimetastatic activity of electroporation-delivered human papillomavirus 16 E7 DNA vaccines: a possible mechanism for enhanced tumor control. DNA Cell Biol. 2011; 30:975–985. PMID: 21649506.
Article
11. Kohler G, Milstein C. Continuous cultures of fused cells secreting antibody of predefined specificity. 1975. J Immunol. 2005; 174:2453–2455. PMID: 15728446.
12. Harlow E, Lane D. Antibodies: a laboratory manual. Cold Spring Harbor, NY: Cold Spring Harbor Laboratory Press;1988.
13. Krieg AM, Yi AK, Matson S, et al. CpG motifs in bacterial DNA trigger direct B-cell activation. Nature. 1995; 374:546–549. PMID: 7700380.
Article
14. Klinman DM, Yi AK, Beaucage SL, Conover J, Krieg AM. CpG motifs present in bacteria DNA rapidly induce lymphocytes to secrete interleukin 6, interleukin 12, and interferon gamma. Proc Natl Acad Sci U S A. 1996; 93:2879–2883. PMID: 8610135.
Article
15. Sin JI, Bagarazzi M, Pachuk C, Weiner DB. DNA priming-protein boosting enhances both antigen-specific antibody and Th1-type cellular immune responses in a murine herpes simplex virus-2 gD vaccine model. DNA Cell Biol. 1999; 18:771–779. PMID: 10541436.
Article
16. Sin JI, Ayyavoo V, Boyer J, Kim J, Ciccarelli RB, Weiner DB. Protective immune correlates can segregate by vaccine type in a murine herpes model system. Int Immunol. 1999; 11:1763–1773. PMID: 10545480.
Article
17. Alvarez CP, Lasala F, Carrillo J, Muniz O, Corbi AL, Delgado R. C-type lectins DC-SIGN and L-SIGN mediate cellular entry by Ebola virus in cis and in trans. J Virol. 2002; 76:6841–6844. PMID: 12050398.
18. Ito H, Watanabe S, Sanchez A, Whitt MA, Kawaoka Y. Mutational analysis of the putative fusion domain of Ebola virus glycoprotein. J Virol. 1999; 73:8907–8912. PMID: 10482652.
Article
Full Text Links
  • CEVR
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr