Immune Netw.  2017 Oct;17(5):326-342. 10.4110/in.2017.17.5.326.

Distinct Effects of Monophosphoryl Lipid A, Oligodeoxynucleotide CpG, and Combination Adjuvants on Modulating Innate and Adaptive Immune Responses to Influenza Vaccination

Affiliations
  • 1Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
  • 2Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA. skang24@gsu.edu

Abstract

Monophosphoryl lipid A (MPL) and oligodeoxynucleotide CpG are toll-like receptor (TLR) 4 and 9 agonist, respectively. Here, we investigated the effects of MPL, CpG, and combination adjuvants on stimulating in vitro dendritic cells (DCs), in vivo innate and adaptive immune responses, and protective efficacy of influenza vaccination. Combination of MPL and CpG was found to exhibit distinct effects on stimulating DCs in vitro to secrete IL-12p70 and tumor necrosis factor (TNF)-α and proliferate allogeneic CD8 T cells. Prime immunization of mice with inactivated split influenza vaccine in the presence of low dose MPL+CpG adjuvants increased the induction of virus-specific IgG and IgG2a isotype antibodies. MPL and CpG adjuvants contribute to improving the efficacy of prime influenza vaccination against lethal influenza challenge as determined by body weight monitoring, lung function, viral titers, and histology. A combination of MPL and CpG adjuvants was effective in improving vaccine efficacy as well as in reducing inflammatory immune responses locally and in inducing cellular immune responses upon lethal influenza virus challenge. This study demonstrates unique adjuvant effects of MPL, CpG, and combination adjuvants on modulating innate and adaptive immune responses to influenza prime vaccination.

Keyword

Monophosphoryl lipid A; Oligonucleotide CpG; Adjuvant; Influenza vaccine

MeSH Terms

Animals
Antibodies
Body Weight
Dendritic Cells
Immunity, Cellular
Immunization
Immunoglobulin G
In Vitro Techniques
Influenza Vaccines
Influenza, Human*
Lipid A*
Lung
Mice
Orthomyxoviridae
T-Lymphocytes
Toll-Like Receptors
Tumor Necrosis Factor-alpha
Vaccination*
Antibodies
Immunoglobulin G
Influenza Vaccines
Lipid A
Toll-Like Receptors
Tumor Necrosis Factor-alpha

Figure

  • Figure 1 In vitro activation of bone marrow-derived DCs by adjuvant stimulation. DCs were enriched from mouse bone marrow cells by treatment with mGM-CSF. (A-C) Cytokine levels secreted into the culture supernatants of DCs treated with different concentrations of MPL and CpG were measured by ELISA. For statistical analysis, Two-way ANOVA and Bonferroni post-multiple comparison tests were performed. (D-F) The immature DCs were cultured with MPL (0.2 μg/ml), CpG (1 μg/ml), or MPL (0.2 μg/ml)+CpG (1 μg/ml) for 2 days. Expression levels of DC activation markers were determined by flow cytometry. All results were shown in mean±SEM. For statistical analysis, One-way ANOVA and Tukey's post-multiple comparison tests were performed. mGM-CSF, mouse granulocyte-macrophage colony stimulating factor. *p<0.033; **p<0.002; ***p<0.001 between the indicated groups.

  • Figure 2 In vitro proliferation and activation of T cells by adjuvant-activated DCs. DCs enriched from bone marrow cells were pre-activated by MPL (0.2 μg/ml), CpG (1 μg/ml), or MPL (0.2 μg/ml)+CpG (1 μg/ml) for 2 days. Allogeneic lymphocytes were harvested from spleens of C57BL/6 mice. CFSE-labeled lymphocytes and pre-activated DCs were co-cultured for 5 days. T cell proliferation and cytokine producing cells were determined by flow cytometry. (A) Proliferated CD4+ T cells. (B) Proliferated CD8+ T cells. (C) IFN-γ producing CD4+ T cells. (D) IFN-γ producing CD8+ T cells. (E) IL-4 producing CD4+ T cells. (F) IL-4 producing CD8+ T cells. All results were shown in mean±SEM. For statistical analysis, One-way ANOVA and Tukey's post-multiple comparison tests were performed. ns, not significant between the indicated groups. *p<0.033; **p<0.002; ***p<0.001.

  • Figure 3 TLR agonist adjuvant effects on inducing IgG antibodies specific for influenza vaccine antigen. BALB/c mice (n=5) were immunized with sPR8 virus vaccine only or sPR8 virus vaccine in the presence of TLR agonist adjuvants (MPL, CpG, or MPL+CpG). Immune sera were taken 2 weeks after immunization and PR8 virus antigen-specific IgG antibody levels were measured by ELISA. IgG (A), IgG1 (B) and IgG2a (C) levels were shown in OD values at 450 nm. (D) IgG2a/IgG1 ratio was calculated at 102 times sera dilution. All results were shown in mean±SEM. For statistical analysis, One-way ANOVA and Tukey's post-multiple comparison tests were performed. *p<0.033; **p<0.002; ***p<0.001 between the indicated groups or compared to sPR8 group; †p<0.033 compared to sPR8+MPL.

  • Figure 4 MPL and CpG adjuvant effects on improving protective efficacy of influenza vaccination after lethal virus infection. The immunized mice (n=5) were infected with A/PR8 virus (2×LD50) after 6 weeks of immunization. BW (A) and PenH (B) were measured for 7 days after infection and % changes were calculated based on the day 0. (C) Lung samples were harvested day 7 post infection. Lung virus titers of each immunized mice were measured by using embryonated eggs. EID50 were shown. All results were shown in mean±SEM. For statistical analysis, One-way ANOVA and Tukey's post-multiple comparison tests were performed. (D) Lung histopathology. Intact lungs were harvested at day 7 post infection, fixed, processed, and stained with hematoxylin and eosin. EID50, 50% egg infection dose; inf., infection. ***p<0.001 compared to naïve infection group; †††p<0.001 compared to sPR8 group.

  • Figure 5 Cytokines and chemokines in lung samples after lethal virus infection of mice. Lung samples were harvested from the immunized mice (n=5) day 7 post A/PR8 virus infection. Cytokine and chemokine levels of each lung samples were measured by ELISA. All results were shown in mean±SEM. For statistical analysis, One-way ANOVA and Tukey's post-multiple comparison test were performed. *p<0.033; **p<0.002; ***p<0.001 compared to Naïve infection group; †p<0.033; ††p<0.002; †††p<0.001 compared to sPR8 group.

  • Figure 6 Cellular infiltration in lungs after lethal virus infection. The immunized mice (n=5) were infected with A/PR8 virus (2×LD50) after 6 weeks of immunization. Lung samples were harvested day 7 post infection, and cell phenotypes were determined by flow cytometry and calculated by multiplying cell percentages with total cell numbers. (A) Total lung cells. (B) AMs; CD11b−CD11c+F4/80+. (C) Monocytes; CD11b+F4/80+Ly6Chigh. (D) Neutrophils; CD11b+F4/80−Ly6c+. (E) pDCs; CD45+F4/80−CD11c+MHCIIhighB220+. (F) CD103 + DC; CD45+F4/80−CD11c+MHCIIhighCD11b−CD103+. (G) cDC; CD45+F4/80−CD11c+MHCIIhighCD11b+. All results were shown in mean±SEM. For statistical analysis, One-way ANOVA and Tukey's post-multiple comparison tests were performed. pDC, plasmacytoid dendritic cell; cDC, conventional DC. *p<0.033; **p<0.002 compared to naïve infection group; †p<0.033 compared to sPR8 group.

  • Figure 7 Cytokine producing T cells after immunization and lethal virus infection. The immunized mice were infected with a lethal dose (2×LD50) of A/PR8 virus after 6 weeks of immunization. Lung and BAL samples were harvested day 7 post infection. Intracellular cytokine staining was performed after incubation with MHCI and II-restricted peptides for CD8 and CD4 T cell stimulation as described in the Materials and Methods section. The cytokine producing cell numbers were calculated by multiplying cell percentages with total cell numbers. All results were shown in mean±SEM. For statistical analysis, One-way ANOVA and Tukey's post-multiple comparison tests were performed. *p<0.033; **p<0.002 compared to naïve infection group; †p<0.033 compared to sPR8 group.

  • Figure 8 Cytokine production of spleen cells from the immunized mice after in vitro antigen stimulation. Spleen cells were harvested from the immunized mice day 7 post infection and then cultured with inactivated A/PR8 virus stimulation. After 3 days culture, cytokine levels in supernatants were determined by ELISA. All results were shown in mean±SEM. For statistical analysis, One-way ANOVA and Tukey's post-multiple comparison tests were performed. nd, not detected or values below detection limit. **p<0.002; ***p<0.001 compared to naïve infection group; ††p<0.002; †††p<0.001 compared to sPR8 group.


Reference

1. Centers for Disease Control and Prevention (CDC). Estimates of deaths associated with seasonal influenza --- United States, 1976–2007. MMWR Morb Mortal Wkly Rep. 2010; 59:1057–1062.
2. Domínguez A, Godoy P, Torner N. The effectiveness of influenza vaccination in different groups. Expert Rev Vaccines. 2016; 15:751–764.
Article
3. Heikkinen T, Heinonen S. Effectiveness and safety of influenza vaccination in children: European perspective. Vaccine. 2011; 29:7529–7534.
Article
4. Kawai T, Akira S. The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nat Immunol. 2010; 11:373–384.
Article
5. Coffman RL, Sher A, Seder RA. Vaccine adjuvants: putting innate immunity to work. Immunity. 2010; 33:492–503.
Article
6. Heer AK, Shamshiev A, Donda A, Uematsu S, Akira S, Kopf M, Marsland BJ. TLR signaling fine-tunes anti-influenza B cell responses without regulating effector T cell responses. J Immunol. 2007; 178:2182–2191.
Article
7. Steinhagen F, Kinjo T, Bode C, Klinman DM. TLR-based immune adjuvants. Vaccine. 2011; 29:3341–3355.
Article
8. Querec T, Bennouna S, Alkan S, Laouar Y, Gorden K, Flavell R, Akira S, Ahmed R, Pulendran B. Yellow fever vaccine YF-17D activates multiple dendritic cell subsets via TLR2, 7, 8, and 9 to stimulate polyvalent immunity. J Exp Med. 2006; 203:413–424.
Article
9. Godaly G, Young DB. Mycobacterium bovis bacille Calmette Guerin infection of human neutrophils induces CXCL8 secretion by MyD88-dependent TLR2 and TLR4 activation. Cell Microbiol. 2005; 7:591–601.
Article
10. Kundi M. New hepatitis B vaccine formulated with an improved adjuvant system. Expert Rev Vaccines. 2007; 6:133–140.
Article
11. Szarewski A. Cervarix®: a bivalent vaccine against HPV types 16 and 18, with cross-protection against other high-risk HPV types. Expert Rev Vaccines. 2012; 11:645–657.
Article
12. McAleer JP, Vella AT. Educating CD4 T cells with vaccine adjuvants: lessons from lipopolysaccharide. Trends Immunol. 2010; 31:429–435.
Article
13. Chu RS, Targoni OS, Krieg AM, Lehmann PV, Harding CV. CpG oligodeoxynucleotides act as adjuvants that switch on T helper 1 (Th1) immunity. J Exp Med. 1997; 186:1623–1631.
Article
14. Heeg K, Zimmermann S. CpG DNA as a Th1 trigger. Int Arch Allergy Immunol. 2000; 121:87–97.
Article
15. Ioannou XP, Gomis SM, Karvonen B, Hecker R, Babiuk LA, van Drunen Littel-van den Hurk S. CpG-containing oligodeoxynucleotides, in combination with conventional adjuvants, enhance the magnitude and change the bias of the immune responses to a herpesvirus glycoprotein. Vaccine. 2002; 21:127–137.
Article
16. Vollmer J, Krieg AM. Immunotherapeutic applications of CpG oligodeoxynucleotide TLR9 agonists. Adv Drug Deliv Rev. 2009; 61:195–204.
Article
17. Reed LJ, Muench H. A simple method of estimating fifty per cent endpoints. Am J Epidemiol. 1938; 27:493–497.
18. Quan FS, Yoo DG, Song JM, Clements JD, Compans RW, Kang SM. Kinetics of immune responses to influenza virus-like particles and dose-dependence of protection with a single vaccination. J Virol. 2009; 83:4489–4497.
Article
19. Ko EJ, Byon YY, Jee Y, Shin T, Park SC, Hahn TW, Joo HG. Maturation of bone marrow-derived dendritic cells by a novel β-glucan purified from Paenibacillus polymyxa JB115. J Vet Sci. 2011; 12:187–189.
Article
20. Timmermans K, Plantinga TS, Kox M, Vaneker M, Scheffer GJ, Adema GJ, Joosten LA, Netea MG. Blueprints of signaling interactions between pattern recognition receptors: implications for the design of vaccine adjuvants. Clin Vaccine Immunol. 2013; 20:427–432.
Article
21. Mata-Haro V, Cekic C, Martin M, Chilton PM, Casella CR, Mitchell TC. The vaccine adjuvant monophosphoryl lipid A as a TRIF-biased agonist of TLR4. Science. 2007; 316:1628–1632.
Article
22. Yamamoto M, Sato S, Hemmi H, Uematsu S, Hoshino K, Kaisho T, Takeuchi O, Takeda K, Akira S. TRAM is specifically involved in the Toll-like receptor 4-mediated MyD88-independent signaling pathway. Nat Immunol. 2003; 4:1144–1150.
Article
23. Kawai T, Akira S. TLR signaling. Semin Immunol. 2007; 19:24–32.
Article
24. Hancock GE, Heers KM, Pryharski KS, Smith JD, Tiberio L. Adjuvants recognized by toll-like receptors inhibit the induction of polarized type 2 T cell responses by natural attachment (G) protein of respiratory syncytial virus. Vaccine. 2003; 21:4348–4358.
Article
25. Prince GA, Mond JJ, Porter DD, Yim KC, Lan SJ, Klinman DM. Immunoprotective activity and safety of a respiratory syncytial virus vaccine: mucosal delivery of fusion glycoprotein with a CpG oligodeoxynucleotide adjuvant. J Virol. 2003; 77:13156–13160.
Article
26. Didierlaurent AM, Morel S, Lockman L, Giannini SL, Bisteau M, Carlsen H, Kielland A, Vosters O, Vanderheyde N, Schiavetti F, et al. AS04, an aluminum salt- and TLR4 agonist-based adjuvant system, induces a transient localized innate immune response leading to enhanced adaptive immunity. J Immunol. 2009; 183:6186–6197.
Article
27. Prince GA, Denamur F, Deschamps M, Garçon N, Prieels JP, Slaoui M, Thiriart C, Porter DD. Monophosphoryl lipid A adjuvant reverses a principal histologic parameter of formalin-inactivated respiratory syncytial virus vaccine-induced disease. Vaccine. 2001; 19:2048–2054.
Article
28. Blanco JC, Boukhvalova MS, Pletneva LM, Shirey KA, Vogel SN. A recombinant anchorless respiratory syncytial virus (RSV) fusion (F) protein/monophosphoryl lipid A (MPL) vaccine protects against RSV-induced replication and lung pathology. Vaccine. 2014; 32:1495–1500.
Article
29. Wack A, Baudner BC, Hilbert AK, Manini I, Nuti S, Tavarini S, Scheffczik H, Ugozzoli M, Singh M, Kazzaz J, et al. Combination adjuvants for the induction of potent, long-lasting antibody and T-cell responses to influenza vaccine in mice. Vaccine. 2008; 26:552–561.
Article
30. Grossmann C, Tenbusch M, Nchinda G, Temchura V, Nabi G, Stone GW, Kornbluth RS, Uberla K. Enhancement of the priming efficacy of DNA vaccines encoding dendritic cell-targeted antigens by synergistic toll-like receptor ligands. BMC Immunol. 2009; 10:43.
Article
31. Smith AM, Smith AP. A critical, nonlinear threshold dictates bacterial invasion and initial kinetics during influenza. Sci Rep. 2016; 6:38703.
Article
32. Tate MD, Pickett DL, van Rooijen N, Brooks AG, Reading PC. Critical role of airway macrophages in modulating disease severity during influenza virus infection of mice. J Virol. 2010; 84:7569–7580.
Article
33. Tate MD, Schilter HC, Brooks AG, Reading PC. Responses of mouse airway epithelial cells and alveolar macrophages to virulent and avirulent strains of influenza A virus. Viral Immunol. 2011; 24:77–88.
Article
34. Liu Q, Zhou YH, Yang ZQ. The cytokine storm of severe influenza and development of immunomodulatory therapy. Cell Mol Immunol. 2016; 13:3–10.
Article
35. Yokota S, Imagawa T, Miyamae T, Ito S, Nakajima S, Nezu A, Mori M. Hypothetical pathophysiology of acute encephalopathy and encephalitis related to influenza virus infection and hypothermia therapy. Pediatr Int. 2000; 42:197–203.
Article
36. Russell JH, Ley TJ. Lymphocyte-mediated cytotoxicity. Annu Rev Immunol. 2002; 20:323–370.
Article
37. Topham DJ, Tripp RA, Doherty PC. CD8+ T cells clear influenza virus by perforin or Fas-dependent processes. J Immunol. 1997; 159:5197–5200.
38. Hufford MM, Kim TS, Sun J, Braciale TJ. Antiviral CD8+ T cell effector activities in situ are regulated by target cell type. J Exp Med. 2011; 208:167–180.
Article
39. Sun J, Madan R, Karp CL, Braciale TJ. Effector T cells control lung inflammation during acute influenza virus infection by producing IL-10. Nat Med. 2009; 15:277–284.
Article
40. Hua L, Yao S, Pham D, Jiang L, Wright J, Sawant D, Dent AL, Braciale TJ, Kaplan MH, Sun J. Cytokine-dependent induction of CD4+ T cells with cytotoxic potential during influenza virus infection. J Virol. 2013; 87:11884–11893.
Article
Full Text Links
  • IN
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