LDH as an adjuvant makes Brucella outer-membrane vesicles and outer-membrane vesicle-associated proteins highly protective in mice

Document Type : Original Article

Authors

1 School of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China

2 Collaborative Innovation Center for Prevention and Control of High Incidence Zoonotic Infectious Diseases in Western China, College of Animal Science and Technology, Shihezi University, Shihezi, China

3 Department of Basic Medicine, Xinjiang Second Medical College, Kelamayi, Xinjiang, China

4 Wuwei vocational college, Wuwei, Gansu 733000, China

Abstract

Objective(s): Existing Brucella vaccines are attenuated and can cause vaccine-associated brucellosis; and these safety concerns have affected their application. Although subunit vaccines have the advantages of safety, efficacy, low cost, and rapid production, they are usually poorly immunogenic and insufficient to trigger persistent immunity. Therefore, we added layered double hydroxide (LDH) as an adjuvant to Brucella subunit vaccine formulations to enhance the immune response to the antigen.
Materials and Methods: LDH and Freund’s adjuvant were combined with Brucella outer-membrane vesicles (OMVs) and OMV-associated proteins to form a subunit vaccine, respectively. The immunogenicity of LDH as an adjuvant was assessed in BALB/c mice. We examined levels of immunoglobulin G, G1, and G2a (IgG, IgG1, and IgG2a) antibodies (aBs); percentages of Cluster of Differentiation 4-positive (CD4+) and CD8+ T cells in peripheral-blood lymphocytes; and secretion of cytokines in mouse spleen lymphocytes. Finally, splenic index and splenic bacterial load were assessed via Brucella challenge experiments on mice.
Results: The LDH subunit vaccine also produced high levels of specific aBs in mice compared with Freund’s adjuvant subunit vaccine and induced mainly T-helper 1 cell (Th1)-type immune responses. In addition, mice in the LDH subunit vaccine group had significantly lower bacterial loads in their spleens than those in the Freund’s adjuvant subunit vaccine group, and the LDH-OMV vaccine offered a higher level of protection against Brucella attack.
Conclusion: LDH as an adjuvant-paired vaccine provided a high level of protection against Brucella infection.

Keywords


1. Seleem MN, Boyle SM, Sriranganathan N. Brucellosis: A re-emerging zoonosis. Vet Microbiol 2010; 140:392-398.
2. Galinska EM, Zagórski J. Brucellosis in humans-etiology, diagnostics, clinical forms. Ann Agric Environ Med 2013; 20:233-238.
3. Doganay M, Aygen B. Human brucellosis: An overview. Int J Infec Dis 2003; 7:173-182.
4. Hou H, Liu X, Peng Q. The advances in brucellosis vaccines. Vaccine 2019; 37:3981-3988.
5. Knox K, Vesk M, Work E. Relation between excreted lipopolysaccharide complexes and surface structures of a lysine-limited culture of Escherichia coli. J Bacteriol 1966; 92:1206-1217.
6. del Socorro Ruiz-Palma M, Avila-Calderón ED, Aguilera-Arreola MG, López-Merino A, Ruiz EA, del Rosario Morales-García M, et al. Comparative proteomic analysis of outer membrane vesicles from Brucella suis, Brucella ovis, Brucella canis and Brucella neotomae. Arch Microbiol 2021; 203:1611-1626.
7. Muralinath M, Kuehn MJ, Roland KL, Curtiss III R. Immunization with Salmonella enterica serovar Typhimurium-derived outer membrane vesicles delivering the pneumococcal protein PspA confers protection against challenge with Streptococcus pneumoniae. Infect Immun 2011; 79:887-894.
8. Hong G-E, Kim D-G, Park E-M, Nam B-H, Kim Y-O, Kong I-S. Identification of Vibrio anguillarum outer membrane vesicles related to immunostimulation in the Japanese flounder, Paralichthys olivaceus. Biosci Biotechnol Biochem 2009; 73:437-439.
9. Post DM, Zhang D, Eastvold JS, Teghanemt A, Gibson BW, Weiss JP. Biochemical and functional characterization of membrane blebs purified from Neisseria meningitidis serogroup B. J Biol Chem 2005; 280:38383-38394.
10. Klieve AV, Yokoyama MT, Forster RJ, Ouwerkerk D, Bain PA, Mawhinney EL. Naturally occurring DNA transfer system associated with membrane vesicles in cellulolytic Ruminococcus spp. of ruminal origin. Appl Environ Microbiol 2005; 71:4248-4253.
11. Kuehn MJ, Kesty NC. Bacterial outer membrane vesicles and the host–pathogen interaction. Genes Dev 2005; 19:2645-2655.
12. Zhang X, Yang F, Zou J, Wu W, Jing H, Gou Q, et al. Immunization with Pseudomonas aeruginosa outer membrane vesicles stimulates protective immunity in mice. Vaccine 2018; 36:1047-1054.
13. Mancini F, Rossi O, Necchi F, Micoli F. OMV vaccines and the role of TLR agonists in immune response. Int J Mol Sci 2020; 21:4416.
14. Liu Q, Li X, Zhang Y, Song Z, Li R, Ruan H, et al. Orally-administered outer-membrane vesicles from Helicobacter pylori reduce H. pylori infection via Th2-biased immune responses in mice. Pathog Dis 2019; 77:ftz050.
15. Pulido MR, García-Quintanilla M, Pachón J, McConnell MJ. A lipopolysaccharide-free outer membrane vesicle vaccine protects against Acinetobacter baumannii infection. Vaccine 2020; 38:719-724.
16. Wang H, Liang K, Kong Q, Liu Q. Immunization with outer membrane vesicles of avian pathogenic Escherichia coli O78 induces protective immunity in chickens. Vet Microbiol 2019; 236:108367.
17. Araiza-Villanueva M, Avila-Calderón ED, Flores-Romo L, Calderón-Amador J, Sriranganathan N, Qublan HA, et al. Proteomic analysis of membrane blebs of Brucella abortus 2308 and RB51 and their evaluation as an acellular vaccine. Front Microbiol 2019; 10:2714.
18. Olsen S. Recent developments in livestock and wildlife brucellosis vaccination. Rev Sci Tech 2013; 32:207-217.
19. Isaacs A, Li Z, Cheung S, Wijesundara DK, McMillan CL, Modhiran N, et al. Adjuvant selection for influenza and RSV prefusion subunit vaccines. Vaccines 2021; 9:71.
20. Zhang L-X, Hu J, Jia Y-B, Liu R-T, Cai T, Xu ZP. Two-dimensional layered double hydroxide nanoadjuvant: Recent progress and future direction. Nanoscale 2021;13:7533-7549.
21. Li A, Qin L, Zhu D, Zhu R, Sun J, Wang S. Signalling pathways involved in the activation of dendritic cells by layered double hydroxide nanoparticles. Biomaterials 2010; 31:748-756.
22. Yan S, Xu K, Li L, Gu W, Rolfe BE, Xu ZP. The pathways for layered double hydroxide nanoparticles to enhance antigen (cross)-presentation on immune cells as adjuvants for protein vaccines. Front Pharmacol 2018; 9:1060.
23. Chen W, Zuo H, Li B, Duan C, Rolfe B, Zhang B, et al. Clay nanoparticles elicit long‐term immune responses by forming biodegradable depots for sustained antigen stimulation. Small 2018; 14:1704465.
24. Williams GR, Fierens K, Preston SG, Lunn D, Rysnik O, De Prijck S, et al. Immunity induced by a broad class of inorganic crystalline materials is directly controlled by their chemistry. J  Exp Med 2014; 211:1019-1025.
25. Yan S, Gu W, Zhang B, Rolfe BE, Xu ZP. High adjuvant activity of layered double hydroxide nanoparticles and nanosheets in anti-tumour vaccine formulations. Dalton Trans 2018; 47:2956-2964.
26. Wu P, Zhang Y, Yin X, He Y, Zhang Q, Chen C. Layered double hydroxide nanoparticles as an adjuvant for inactivated foot-and-mouth disease vaccine in pigs. BMC Vet Res 2020; 16:1-9.
27. Pollak CN, Delpino MV, Fossati CA, Baldi PC. Outer membrane vesicles from Brucella abortus promote bacterial internalization by human monocytes and modulate their innate immune response. PloS one 2012; 7:e50214.
28. Verstreate D, Creasy M, Caveney N, Baldwin C, Blab M, Winter A. Outer membrane proteins of Brucella abortus: Isolation and characterization. Infect Immun 1982; 35:979-989.
29. Zhu L, Wang Q, Wang Y, Xu Y, Peng D, Huang H, et al. Comparison of immune effects between Brucella recombinant Omp10-Omp28-L7/L12 proteins expressed in eukaryotic and prokaryotic systems. Front Vet Sci 2020; 7:576.
30. Shi D, Fan B, Sun B, Zhou J, Zhao Y, Guo R, et al. LDH nanoparticle adjuvant subunit vaccine induces an effective immune response for porcine epidemic diarrhea virus. Virology 2022; 565:58-64.
31. Mukherjee F, Prasad A, Bahekar V, Rana S, Rajendra L, Sharma G, et al. Evaluation of immunogenicity and protective efficacy of a liposome containing Brucella abortus S19 outer membrane protein in BALB/c mice. Iran J Vet Res 2016; 17:1.
32. Swietnicki W. Secretory system components as potential prophylactic targets for bacterial pathogens. Biomolecules 2021; 11:892.
33. Goel D, Bhatnagar R. Intradermal immunization with outer membrane protein 25 protects BALB/c mice from virulent B. abortus 544. Mol Immun 2012; 51:159-168.
34. Vanzini V, Aguirre N, Lugaresi C, De Echaide S, De Canavesio V, Guglielmone A, et al. Evaluation of an indirect ELISA for the diagnosis of bovine brucellosis in milk and serum samples in dairy cattle in Argentina. Prev Vet Med 1998; 36:211-217.
35. Al-Mariri A, Tibor A, Mertens P, De Bolle X, Michel P, Godfroid J, et al. Induction of immune response in BALB/c mice with a DNA vaccine encoding bacterioferritin or P39 of Brucella spp. Infect Immun 2001; 69:6264-6270.
36. Ghasemi A, Jeddi-Tehrani M, Mautner J, Salari MH, Zarnani A-H. Simultaneous immunization of mice with Omp31 and TF provides protection against Brucella melitensis infection. Vaccine 2015; 33:5532-5538.
37. Foged C. Subunit vaccines of the future: The need for safe, customized and optimized particulate delivery systems. Ther Deliv 2011; 2:1057-1077.
38. Al-Mariri A, Mahmoud NH, Hammoud R. Efficacy evaluation of live Escherichia coli expression Brucella P39 protein combined with CpG oligodeoxynucleotides vaccine against Brucella melitensis 16M, in BALB/c mice. Biologicals 2012; 40:140-145.
39. Kaushik P, Singh DK, Kumar SV, Tiwari AK, Shukla G, Dayal S, et al. Protection of mice against Brucella abortus 544 challenge by vaccination with recombinant OMP28 adjuvanted with CpG oligonucleotides. Vet Res Commun 2010; 34:119-132.
40. Fu S, Xu J, Li X, Xie Y, Qiu Y, Du X, et al. Immunization of mice with recombinant protein CobB or AsnC confers protection against Brucella abortus infection. PloS One 2012; 7:e29552.
41. Vitry M-A, De Trez C, Goriely S, Dumoutier L, Akira S, Ryffel B, et al. Crucial role of gamma interferon-producing CD4+ Th1 cells but dispensable function of CD8+ T cell, B cell, Th2, and Th17 responses in the control of Brucella melitensis infection in mice. Infect Immun 2012; 80:4271-4280.
42. Zhang K, Wang H, Guo F, Yuan L, Zhang W, Wang Y, et al. OMP31 of Brucella melitensis 16M impairs the apoptosis of macrophages triggered by TNF-α. Exp Ther Med 2016; 12:2783-2789.
43. Corsetti PP, de Almeida LA, Carvalho NB, Azevedo V, Silva TM, Teixeira HC, et al. Lack of endogenous IL-10 enhances production of proinflammatory cytokines and leads to Brucella abortus clearance in mice. PLoS One 2013; 8:e74729.
44. Ghasemi A, Jeddi-Tehrani M, Mautner J, Salari MH, Zarnani A-H. Immunization of mice with a novel recombinant molecular chaperon confers protection against Brucella melitensis infection. Vaccine 2014; 32:6659-6666.
45. Avila-Calderón ED, Lopez-Merino A, Jain N, Peralta H, López-Villegas EO, Sriranganathan N, et al. Characterization of outer membrane vesicles from Brucella melitensis and protection induced in mice. Clin Dev Immunol 2012;2012:352493.