Bacillus subtilis spores as vaccine adjuvants: further insights into the mechanisms of action

Renata Damásio de Souza, Milene Tavares Batista, Wilson Barros Luiz, Rafael Ciro Marques Cavalcante, Jaime Henrique Amorim, Raíza Sales Pereira Bizerra, Eduardo Gimenes Martins, Luís Carlos de Souza Ferreira, Renata Damásio de Souza, Milene Tavares Batista, Wilson Barros Luiz, Rafael Ciro Marques Cavalcante, Jaime Henrique Amorim, Raíza Sales Pereira Bizerra, Eduardo Gimenes Martins, Luís Carlos de Souza Ferreira

Abstract

Bacillus subtilis spores have received growing attention regarding potential biotechnological applications, including the use as probiotics and in vaccine formulations. B. subtilis spores have also been shown to behave as particulate vaccine adjuvants, promoting the increase of antibody responses after co-administration with antigens either admixed or adsorbed on the spore surface. In this study, we further evaluated the immune modulatory properties of B. subtilis spores using a recombinant HIV gag p24 protein as a model antigen. The adjuvant effects of B. subtilis spores were not affected by the genetic background of the mouse lineage and did not induce significant inflammatory or deleterious effects after parenteral administration. Our results demonstrated that co-administration, but not adsorption to the spore surface, enhanced the immunogenicity of that target antigen after subcutaneous administration to BALB/c and C57BL/6 mice. Spores promoted activation of antigen presenting cells as demonstrated by the upregulation of MHC and CD40 molecules and enhanced secretion of pro-inflammatory cytokines by murine dendritic cells. In addition, in vivo studies indicated a direct role of the innate immunity on the immunomodulatory properties of B. subtilis spores, as demonstrated by the lack of adjuvant effects on MyD88 and TLR2 knockout mouse strains.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Adsorption of the p24 protein…
Figure 1. Adsorption of the p24 protein to spores of the B. subtilis WW02 strain.
(A) Evaluation of protein adsorption to spores at different pHs. Spores (2×109) were suspended in PBS buffer at pH 4, pH 7 or pH 10, mixed with purified p24 protein (2 µg) and kept for 1 h at room temperature. The samples were centrifuged, washed twice with PBS and the spore coat proteins were extracted and submitted to electrophoresis in SDS-PAGE at 15%. The control lane (Con) represents spore samples without incubation with the protein and the control lane (Pr) shows 2 µg of purified p24. Image is representative of four independent experiments. (B) Detection of protein released from spores after adsorption. Spores adsorbed with p24 (PBS at pH 4) were resuspended in PBS at pH 4, pH 7 or pH 10 and incubated for different times. At indicated time points (15, 30, 45, and 60 min) samples were centrifuged and the supernatants and pellet (spores) were loaded onto 15% SDS-PAGE gel. (C) Determination of the saturation curve of p24 protein adsorption to B. subtilis spores. Increasing amounts of protein (0.5 to 4 µg) were added to B. subtilis spores (2×109) following the procedure described before. The amount of protein adsorbed to the spores coat was quantified by dot-blot assay.
Figure 2. Evaluation of antibody responses following…
Figure 2. Evaluation of antibody responses following subcutaneous immunization with p24 protein adsorbed or mixed with spores.
Mouse groups (n = 5) were immunized (three doses on days 0, 14 and 28) with 2×109 spores (□), 700 ng of p24 only (○), 700 ng of p24 in combination with 2×109 spores (▪) or 700 ng of p24 adsorbed at 2×109 spores (•). Levels of anti-p24 (A and B) and anti-spores (C and D) specific IgG antibodies were detected in serum samples at 13, 27 and 41 days after immunization in BALB/c and C57 BL/6 mice. The results are presented as the mean ± s.e.m. of three independent experiments, and the values obtained from PBS immunized mice were subtracted from each group. *** p<0.001 when compared with mice immunized with p24 only.
Figure 3. Humoral responses after subcutaneous immunization…
Figure 3. Humoral responses after subcutaneous immunization with p24 protein mixed with live or heat-killed spores.
Detection of anti-p24 specific IgG antibodies in serum samples of immunized BALB/c (A) and C57 BL/6 mice (B). Mouse groups (n = 5) were immunized (three doses on days 0, 14 and 28) with viable spores (), heat-killed spores (HkSp) (), p24 protein (○), p24 protein in combination with Alum (▪), p24 protein in combination with spores (•) or p24 protein in combination with heat-killed spores (▴). *** p<0.001 when compared with group immunized with p24 only or with each other. Specific IgG subclasses elicited in BALB/c (C) and C57BL/6 (D) mice after complete immunization regimen. The IgG2a/IgG1 or IgG2c/IgG1 ratios are indicated at the top of the figure. * p<0.05 or *** p<0.001. All the results are presented as the mean ± s.e.m. of three independent experiments, and the values obtained from PBS immunized mice were subtracted from each group.
Figure 4. Intracellular cell staining and flow…
Figure 4. Intracellular cell staining and flow cytometry analysis of p24-specific CD4+ and CD8+ T cells.
BALB/c mice were immunized (three doses on days 0, 14 and 28) with p24 protein (700 ng), p24 (10 µg) admixed with LT1 (1 µg), p24 (700 ng) admixed with spores (2×109) or p24 (700 ng) admixed with heat-killed spores (2×109). Spleen cells were collected two weeks after the last dose, and the detection of IL-4 producing p24-specific CD4+ T cells (A) and IFN-γ producing p24-specific CD4+ (B) and CD8+ T cells (C) was carried out after stimulation with MHC class I or II restricted p24 peptide and cell surface staining for CD8 (FITC), CD4 (Cy) and intracellular staining for IL-4 or IFN-γ (PE). The percentage of each population is indicated in the upper right corners and the frequencies in all groups in the absence of stimulus were below 0.70% (data not shown). Measurements were performed in duplicate for each individual sample, and the data are representative of two independent experiments.
Figure 5. Maturation of dendritic cells after…
Figure 5. Maturation of dendritic cells after stimulation with B. subtilis spores.
(A) Induction of pro-inflammatory cytokines after stimulation with spores. BMDCs from C57BL/6 mice were treated with PBS (control group), LT1 and live or heat-killed spores at a MOI of 10.000. Supernatants were collected after 24 h, and the IL-1β, IL12p70 and TNF-α concentrations were quantified by ELISA. (B) Expression of surface molecules by CD11c+ cells following exposure to spores. BMDCs were treated as described above and the surface expression levels of CD40, MHC-I and MHC-II were measured by flow cytometry. Gray-filled histograms represent samples incubated with PBS and open histograms represent samples incubated with spores. The MFI for both conditions is shown in the upper left in each histogram. (C) Graphical comparison of the expression of CD40, MHC-I and MHC-II. Data represent mean values ± s.e.m. of two independent experiments. *p<0.05, **p<0.01 or *** p<0.001 when compared with control group or with each other. Sp: B. subtilis WW02 spores; HkSp: Heat-killed B. subtilis WW02 spores.
Figure 6. The adjuvant effects of B.…
Figure 6. The adjuvant effects of B. subtilis spores require stimulation of TLR2.
MyD88−/− (A) and TLR2−/− (B) C57 BL/6 mice (n = 5) were immunized (three doses on days 0, 14 and 28) with p24 protein only (700 ng) or p24 (700 ng) in combination with live or heat-killed spores (2×109). Levels of anti-p24 specific IgG antibodies were detected in serum samples 13 days after the third dose. The results represent the means ± s.e.m. of two experiments, and the values obtained from PBS immunized mice were subtracted from each group. *** p<0.001.

References

    1. Reed SG, Bertholet S, Coler RN, Friede M (2008) New horizons in adjuvants for vaccine development. Trends Immunol 30: 23–32.
    1. Lambrecht BN, Kool M, Willart MA, Hammad H (2009) Mechanism of action of clinically approved adjuvants. Curr Opin Immunol 21: 23–29.
    1. O'Hagan DT, Valiante NM (2003) Recent advances in the discovery and delivery of vaccine adjuvants. Nat Rev Drug Discov 2: 727–735.
    1. Montomoli E, Piccirella S, Khadang B, Mennitto E, Camerini R, et al. (2011) Current adjuvants and new perspectives in vaccine formulation. Expert Rev Vaccines 10: 1053–1061.
    1. Heegaard PM, Dedieu L, Johnson N, Le Potier MF, Mockey M, et al. (2011) Adjuvants and delivery systems in veterinary vaccinology: current state and future developments. Arch Virol 156: 183–202.
    1. Miyaji EN, Carvalho E, Oliveira ML, Raw I, Ho PL (2011) Trends in adjuvant development for vaccines: DAMPs and PAMPs as potential new adjuvants. Braz J Med Biol Res 44: 500–513.
    1. Huang JM, Hong HA, VanTong H, Hoang TH, Brisson A, et al. (2010) Mucosal delivery of antigens using adsorption to bacterial spores. Vaccine 28: 1021–1030.
    1. Barnes AGC, Cerovic V, Hobson PS, Klavinskis LS (2007) Bacillus subtilis spores: a novel microparticle adjuvant which can instruct a balanced Th1 and Th2 immune response to specific antigen. Eur. J. Immunol 37: 1538–1547.
    1. Song M, Hong HA, Huang JM, Colenutt C, Khang DD, et al. (2012) Killed Bacillus subtilis spores as a mucosal adjuvant for an H5N1 vaccine. Vaccine 30: 3266–3277.
    1. Cutting SM (2011) Bacillus probiotics. Food Microbiol 28: 214–220.
    1. Tavares MB, Silva BM, Cavalcante RCM, Souza RD, Luiz WB, et al. (2010) Induction of neutralizing antibodies in mice immunized with an amino-terminal polypeptide of Streptococcus mutans P1 protein produced by a recombinant Bacillus subtilis strain. FEMS Immunol Med Microbiol 59: 131–142.
    1. Amuquni H, Tzipori S (2012) Bacillus subtilis: a temperature resistant and needle free delivery system of immunogens. Hum Vaccin Immunother 8: 979–986.
    1. Paccez JD, Nguyen HD, Luiz WB, Ferreira RCC, Sbrogio-Almeida ME, et al. (2007) Evaluation of different promoter sequences and antigen sorting signals on the immunogenicity of Bacillus subtilis vaccine vehicles. Vaccine 25: 4671–4680.
    1. Negri A, Potocki W, Iwanicki A, Obuchowski M, Hinc K (2013) Expression and display of Clostridium difficile protein FliD on the surface of Bacillus subtilis spores. J Med Microbiol 62: 1379–1385.
    1. Nicholson WL, Munakata H, Horneck G, Melosh HJ, Setlow P (2000) Resistance of Bacillus endospores to extreme terrestrial and extraterrestrial environments. Microbiol Mol Biol Rev 64: 548–572.
    1. Li X, Xu W, Xiong S (2012) A novel tuberculosis DNA vaccine in an HIV-1 p24 protein backbone confers protection against Mycobacterium tuberculosis and simultaneously elicits robust humoral and cellular responses to HIV-1. Clin Vaccine Immunol 19: 723–730.
    1. Bozzaco L, Yu H, Dengjel J, Trumpfheller C, Zebroski HA 3rd, et al. (2012) Strategy for identifying dendritic cell-processed CD4+ T cell epitopes from the HIV gag p24 protein. PLoS One 7: e41897 10.1371/journal.pone.0041897
    1. Wehrl W, Niederweis M, Schumann W (2000) The FtsH protein accumulates at the septum of Bacillus subtilis during cell division and sporulation. J Bacteriol 182: 3870–3873.
    1. Deuerling E, Paeslack B, Schumann W (1995) The ftsH gene of Bacillus subtilis is transiently induced after osmotic and temperature upshift. J Bacteriol 177: 4105–4112.
    1. Tavares MB, Souza RD, Luiz WB, Cavalcante RCM, Casaroli C, et al. (2013) Bacillus subtilis endospores at high purity and recovery yields: optimization of growth conditions and purification method. Curr Microbiol 66: 279–285.
    1. Aida Y, Pabst MJ (1990) Removal of endotoxin from protein solutions by phase separation using Triton X-114. J Immunol Methods 132: 191–195.
    1. Breyhan H, Wirths O, Duan K, Marcello A, Retting J, et al. (2009) APP/PS1KI bigenic mice develop early synaptic deficits and hippocam pus atrophy. Acta Neuropathol 117: 667–685.
    1. Liard C, Munier S, Arias M, Joulin-Giet A, Bonduelle O, et al. (2011) Targeting of HIV-p24 particle based vaccine into differential skin layers induces distinct arms of the immune responses. Vaccine 29: 6379–6391.
    1. Tagliamonte M, Marasco D, Ruggiero A, De Stradis A, Tornesello ML, et al. (2012) HIV p24 as scaffold for presenting conformational HIV Env antigens. PLoS One 7: e43318 10.1371/journal.pone.0043318
    1. Zanoni I, Ostuni R, Capuano G, Collini M, Caccia M, et al. (2009) CD14 regulates the dendritic cell life cycle after LPS exposure through NFAT activation. Nature 460: 264–268.
    1. Marim FM, Silveira TN, Lima DSJ, Zamboni DS (2010) A method for generation of bone marrow-derived macrophages from cryopreserved mouse bone marrow cells. PLoS One 5: e15263 10.1371/journal.pone.0015263
    1. Amorim JH, Diniz MO, Cariri FA, Rodrigues JF, Bizerra RS, et al. (2012) Protective immunity to DENV2 after immunization with a recombinant NS1 protein using a genetically detoxified heat-labile toxin as an adjuvant. Vaccine 5: 837–845.
    1. Visciano ML, Tangliamonte M, Tornesello ML, Buonaguro FM, Buonaguro L (2012) Effects of adjuvants on IgG subclasses elicited by virus-like particles. J. Tranl. Med 10: 4 10.1186/1479-5876-10-4
    1. Fierens K, Kool M (2012) The mechanism of adjuvanticity of aluminium-contaning formulas. Curr Pharm Des 18: 2305–2313.
    1. Rodrigues JF, Mathias-Santos C, Sbrogio-Almeida ME, Amorim JH, Cabrera-Crespo J, et al. (2011) Functional diversity of heat-labile toxin (LT) produced by enterotoxigenic Escherichia coli: differential enzymatic and immunological activities of LT1 (hLT) and LT4 (pLT). J Biol Chem 286: 5222–5233.
    1. Christmas P (2010) Toll-Like Receptors: Sensors that Detect Infection. Nature Education 3: 85.
    1. Huang JM, La Ragione RM, Nunez A, Cutting SM (2008) Immunostimulatory activity of Bacillus spores. FEMS Immunol Med Microbiol 53: 195–203.
    1. Watanabe H, Numata K, Ito T, Takagi K, Matsukawa A (2004) Innate immune response in Th1- and Th2-dominant mouse strains. Shock 22: 460–466.

Source: PubMed

3
Subscribe