Vaccine-Induced Carbohydrate-Specific Memory B Cells Reactivate During Rodent Malaria Infection.
Journal: 2019/August - Frontiers in Immunology
ISSN: 1664-3224
Abstract:
A long-standing challenge in malaria is the limited understanding of B cell immunity, previously hampered by lack of tools to phenotype rare antigen-specific cells. Our aim was to develop a method for identifying carbohydrate-specific B cells within lymphocyte populations and to determine whether a candidate vaccine generated functional memory B cells (MBCs) that reactivated upon challenge with Plasmodium (pRBCs). To this end, a new flow cytometric probe was validated and used to determine the kinetics of B cell activation against the candidate vaccine glycosylphosphatidylinositol conjugated to Keyhole Limpet Haemocyanin (GPI-KLH). Additionally, immunized C57BL/6 mice were rested (10 weeks) and challenged with pRBCs or GPI-KLH to assess memory B cell recall against foreign antigen. We found that GPI-specific B cells were detectable in GPI-KLH vaccinated mice, but not in Plasmodium-infected mice. Additionally, in previously vaccinated mice GPI-specific IgG1 MBCs were reactivated against both pRBCs and synthetic GPI-KLH, which resulted in increased serum levels of anti-GPI IgG in both challenge approaches. Collectively our findings contribute to the understanding of B cell immunity in malaria and have important clinical implications for inclusion of carbohydrate conjugates in malaria vaccines.
Relations:
Content
References
(32)
Diseases
(1)
Chemicals
(2)
Organisms
(2)
Processes
(3)
Anatomy
(2)
Similar articles
Articles by the same authors
Discussion board
Front Immunol 10: 1840

Vaccine-Induced Carbohydrate-Specific Memory B Cells Reactivate During Rodent Malaria Infection

Supplementary Figure 1

Flow cytometry gating strategy. This is the flow cytometry gating strategy using an example of a spleen isolated from a GPI-immunized mouse at day 14.

Click here for additional data file.(1.3M, TIF)

Supplementary Figure 2

Representative plots for experimental groups; one mouse from each group.

Click here for additional data file.(1.2M, TIF)

Supplementary Figure 3

Challenge with synthetic GPI-KLH or pRBCs induced rapid activation of GPI-specific MBCs in previously GPI-vaccinated mice, whilst Plasmodium mice do not generate GPI-specific MBC. Mice were vaccinated and boosted with GPI-KLH (n = 24) or infected twice with pRBCs i.p. (n = 24, referred to as Plasmodium mice). Naïve mice were included as a control group (n = 24). After resting for a minimum of 10 weeks, mice were injected with GPI-KLH i.p. or 1 × 10 pRBCs i.p. to assess reactivation of MBCs at day 5. The supplementary figure outlines FACS plots.

Click here for additional data file.(995K, TIF)

Supplementary Table 1

Vaccination/Infection protocol for assessing B cell activation kinetics and memory B cell recall.

Click here for additional data file.(20K, DOCX)
Division of Population Health and Immunity, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, QLD, Australia
Edited by: Wanderley De Souza, Federal University of Rio de Janeiro, Brazil
Reviewed by: Moriya Tsuji, Aaron Diamond AIDS Research Center, United States; Ricardo Fujiwara, Federal University of Minas Gerais, Brazil
*Correspondence: Emily M. Eriksson ua.ude.ihew@nosskire
This article was submitted to Microbial Immunology, a section of the journal Frontiers in Immunology
Edited by: Wanderley De Souza, Federal University of Rio de Janeiro, Brazil
Reviewed by: Moriya Tsuji, Aaron Diamond AIDS Research Center, United States; Ricardo Fujiwara, Federal University of Minas Gerais, Brazil
Received 2019 Jan 18; Accepted 2019 Jul 22.
This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

Abstract

A long-standing challenge in malaria is the limited understanding of B cell immunity, previously hampered by lack of tools to phenotype rare antigen-specific cells. Our aim was to develop a method for identifying carbohydrate-specific B cells within lymphocyte populations and to determine whether a candidate vaccine generated functional memory B cells (MBCs) that reactivated upon challenge with Plasmodium (pRBCs). To this end, a new flow cytometric probe was validated and used to determine the kinetics of B cell activation against the candidate vaccine glycosylphosphatidylinositol conjugated to Keyhole Limpet Haemocyanin (GPI-KLH). Additionally, immunized C57BL/6 mice were rested (10 weeks) and challenged with pRBCs or GPI-KLH to assess memory B cell recall against foreign antigen. We found that GPI-specific B cells were detectable in GPI-KLH vaccinated mice, but not in Plasmodium-infected mice. Additionally, in previously vaccinated mice GPI-specific IgG1 MBCs were reactivated against both pRBCs and synthetic GPI-KLH, which resulted in increased serum levels of anti-GPI IgG in both challenge approaches. Collectively our findings contribute to the understanding of B cell immunity in malaria and have important clinical implications for inclusion of carbohydrate conjugates in malaria vaccines.

Keywords: malaria, B cell immunity, vaccines, memory failure, glycosylphosphatidylinositol (GPI)
Abstract

Acknowledgments

The authors would like to thank Prof. David Tarlinton and Dr. Kim Jacobson for their invaluable input and the Bundoora Antibody facility for generating IgD-A680 and Gr-A680.

Acknowledgments

Glossary

Abbreviations

BIObiotin labelling
BSAbovine serum albumin
GPIglycosylphosphatidylinositol
KLHKeyhole Limpet Haemocyanin
MBCmemory B cells
NPFour-hydroxy-3-nitrophenyl acetyl-Osu
pRBCsparasitized red blood cells.
Glossary

Footnotes

Funding. This work was supported by the National Health and Medical Research Council [GNT1052580 to HJ and GNT1093311] and the Bill and Melinda Gates Foundation [OPP1136441].

Footnotes
Click here for additional data file.(1.3M, TIF)Click here for additional data file.(1.2M, TIF)Click here for additional data file.(995K, TIF)Click here for additional data file.(20K, DOCX)

References

  • 1. Krishnamurty AT, Thouvenel CD, Portugal S, Keitany GJ, Kim KS, Holder A, et al. . Somatically hypermutated plasmodium-specific IgM(+) memory B cells are rapid, plastic, early responders upon malaria rechallenge. Immunity. (2016) 45:402–14. 10.1016/j.immuni.2016.06.014 ] [
  • 2. Wipasa J, Suphavilai C, Okell LC, Cook J, Corran PH, Thaikla K, et al. . Long-lived antibody and B cell memory responses to the human malaria parasites, Plasmodium falciparum and plasmodium vivax. PLoS Pathog. (2010) 6:e1000770. 10.1371/journal.ppat.1000770 ] [
  • 3. Weiss GE, Traore B, Kayentao K, Ongoiba A, Doumbo S, Doumtabe D, et al. . The plasmodium falciparum-specific human memory B cell compartment expands gradually with repeated malaria infections. PLoS Pathog. (2010) 6:e1000912. 10.1371/journal.ppat.1000912 ] [
  • 4. McGregor IA. Malarial immunity: current trends and prospects. Ann Trop Med Parasitol. (1987) 81:647–56. 10.1080/00034983.1987.11812166 [] [[PubMed]
  • 5. Vekemans J. Major global vaccine challenges: recent progress in malaria vaccine development A2 – bloom. In: Lambert PH, editor. , editor. The Vaccine Book, 2nd ed Cambridge, MA: Academic Press; (2016). p. 385–99. 10.1016/B978-0-12-802174-3.00019-9 [[PubMed]
  • 6. Gordon DM, McGovern TW, Krzych U, Cohen JC, Schneider I, LaChance R, et al. . Safety, immunogenicity, and efficacy of a recombinantly produced Plasmodium falciparum circumsporozoite protein-hepatitis B surface antigen subunit vaccine. J Infect Dis. (1995) 171:1576–85. 10.1093/infdis/171.6.1576 [] [[PubMed]
  • 7. Campo JJ, Sacarlal J, Aponte JJ, Aide P, Nhabomba AJ, Dobaño C, et al. . Duration of vaccine efficacy against malaria: 5th year of follow-up in children vaccinated with RTS,S/AS02 in Mozambique. Vaccine. (2014) 32:2209–16. 10.1016/j.vaccine.2014.02.042 [] [[PubMed]
  • 8. Keitany GJ, Kim KS, Krishnamurty AT, Hondowicz BD, Hahn WO, Dambrauskas N, et al. . Blood stage malaria disrupts humoral immunity to the pre-erythrocytic stage circumsporozoite protein. Cell Rep. (2016) 17:3193–205. 10.1016/j.celrep.2016.11.060 ] [
  • 9. Fisher CR, Sutton HJ, Kaczmarski JA, McNamara HA, Clifton B, Mitchell J, et al. . T-dependent B cell responses to Plasmodium induce antibodies that form a high-avidity multivalent complex with the circumsporozoite protein. PLoS pathogens. (2017) 13:e1006469. 10.1371/journal.ppat.1006469 ] [
  • 10. Yang C, Lu F, Wang B, Li J, Han J-H, Ito D, et al Plasmodium vivax GPI-anchored micronemal antigen (PvGAMA) binds human erythrocytes independent of Duffy antigen status. Sci Rep. (2016) 6:35581 10.1038/srep35581 ] [[Google Scholar]
  • 11. Kitt AJ, Leigh JA. Synthetic GPI as a candidate anti-toxic vaccine in a model of malaria. Nature. (2002) 418:785-9. 10.1038/nature00937 [] [[PubMed]
  • 12. Schofield L, Hackett F. Signal transduction in host cells by a glycosylphosphatidylinositol toxin of malaria parasites. J Exp Med. (1993) 177:145–53. 10.1084/jem.177.1.145 ] [
  • 13. Tachado SD, Gerold P, McConville MJ, Baldwin T, Quilici D, Schwarz RT, et al. . Glycosylphosphatidylinositol toxin of Plasmodium induces nitric oxide synthase expression in macrophages and vascular endothelial cells by a protein tyrosine kinase-dependent and protein kinase C-dependent signaling pathway. J Immunol. (1996) 156:1897–907. [[PubMed]
  • 14. Schofield L, Novakovic S, Gerold P, Schwarz RT, McConville MJ, Tachado SD. Glycosylphosphatidylinositol toxin of Plasmodium up-regulates intercellular adhesion molecule-1, vascular cell adhesion molecule-1, and E-selectin expression in vascular endothelial cells and increases leukocyte and parasite cytoadherence via tyrosine kinase-dependent signal transduction. J Immunol. (1996) 156:1886–96. [[PubMed]
  • 15. Tamborrini M, Liu X, Mugasa JP, Kwon YU, Kamena F, Seeberger PH, et al. . Synthetic glycosylphosphatidylinositol microarray reveals differential antibody levels and fine specificities in children with mild and severe malaria. Bioorg Med Chem. (2010) 18:3747–52. 10.1016/j.bmc.2010.04.059 [] [[PubMed]
  • 16. Kapoor N, Vanjak I, Rozzelle J, Berges A, Chan W, Yin G, et al. . Malaria derived glycosylphosphatidylinositol anchor enhances anti-Pfs25 functional antibodies that block malaria transmission. Biochemistry. (2018) 57:516–9. 10.1021/acs.biochem.7b01099 ] [
  • 17. Berhe S, Schofield L, Schwarz RT, Gerold P. Conservation of structure among glycosylphosphatidylinositol toxins from different geographic isolates of Plasmodium falciparum. Mol Biochem Parasitol. (1999) 103:273–8. 10.1016/S0166-6851(99)00125-5 [] [[PubMed]
  • 18. Kouba DJ, Miller SJ. Immunogenicity of glycosylphosphatidylinositol-anchored micronemal antigen in natural Plasmodium vivax exposure. Malar J. (2017) 16–348.. 10.1186/s12936-017-1967-9 ] [
  • 19. Kouba DJ, Miller SJ. IgG antibodies to synthetic GPI are biomarkers of immune-status to both Plasmodium falciparum and Plasmodium vivax malaria in young children. Malar J. (2017) 16–386.. 10.1186/s12936-017-2042-2 ] [
  • 20. Burda PC, Caldelari R, Heussler VT. Manipulation of the host cell membrane during plasmodium liver stage egress. MBio. (2017) 8:e00139–17. 10.1128/mBio.00139-17 ] [
  • 21. Mathias DK, Jardim JG, Parish LA, Armistead JS, Trinh HV, Kumpitak C, et al. . Differential roles of an Anopheline midgut GPI-anchored protein in mediating Plasmodium falciparum and Plasmodium vivax ookinete invasion. Infect Genet Evol. (2014) 28:635–47. 10.1016/j.meegid.2014.05.025 ] [
  • 22. Schofield L, Ioannidis LJ, Karl S, Robinson LJ, Tan QY, Poole DP, et al. . Synergistic effect of IL-12 and IL-18 induces TIM3 regulation of γδ T cell function and decreases the risk of clinical malaria in children living in Papua New Guinea. BMC Med. (2017) 15:114. 10.1186/s12916-017-0883-8 ] [
  • 23. Lalor PA, Nossal GJ, Sanderson RD, McHeyzer-Williams MG. Functional and molecular characterization of single, (4-hydroxy-3-nitrophenyl)acetyl (NP)-specific, IgG1+ B cells from antibody-secreting and memory B cell pathways in the C57BL/6 immune response to NP. Eur J Immunol. (1992) 22:3001–11. 10.1002/eji.1830221136 [] [[PubMed]
  • 24. Ridderstad A, Tarlinton DM. Kinetics of establishing the memory B cell population as revealed by CD38 expression. J Immunol. (1998) 160:4688–95. [[PubMed]
  • 25. Kalia V, Sarkar S, Gourley TS, Rouse BT, Ahmed R. Differentiation of memory B and T cells. Curr Opin Immunol. (2006) 18:255–64. 10.1016/j.coi.2006.03.020 [] [[PubMed]
  • 26. Shinde R, Chaudhary K, Shimoda M, Pacholczyk G, McGaha T. Indoleamine 2,3-dioxygenase inhibits B cell immune response to T independent antigens (IRM8P.710). J Immunol. (2014) 192:127.11–127.11. [PubMed]
  • 27. Tetsutani K, To H, Torii M, Hisaeda H, Himeno K. Malaria parasite induces tryptophan-related immune suppression in mice. Parasitology. (2007) 134:923–30. 10.1017/S0031182007002326 [] [[PubMed]
  • 28. Su S, Miska KB, Fetterer RH, Jenkins MC, Wong EA. Plasmodium falciparum: asexual erythrocytic stages synthesize two structurally distinct free and protein-bound glycosylphosphatidylinositols in a maturation-dependent manner. Exp Parasitol. (1998) 8895–102. 10.1006/expr.1998.4241 [] [[PubMed]
  • 29. Jaurigue JA, Seeberger PH. Parasite carbohydrate vaccines. Front Cell Infect Microbiol. (2017) 7:248. 10.3389/fcimb.2017.00248 ] [
  • 30. Janeway CA, Travers P, Walport M, Shlomchik MJ. Janeway's Immunobiology, 7th edition. New York, NY: Garland Publishing; (2008). [PubMed]
  • 31. Sash L, Yang ASP, John A, Scott NE, Lingford JP, O'Neill MT, et al Protein O-fucosylation in Plasmodium falciparum ensures efficient infection of mosquito and vertebrate hosts. Nat Commun. (2017) 8:561 10.1038/s41467-017-00571-y ] [[Google Scholar]
  • 32. Buchanan RM, Arulanandam BP, Metzger DW. IL-12 enhances antibody responses to T-independent polysaccharide vaccines in the absence of T and NK Cells. J Immunol. (1998) 161:5525–33. [[PubMed]
  • 33. Mamchak AA, Hodgkin PD. Regulation of lipopolysaccharide-induced B-cell activation: evidence that surface immunoglobulin mediates two independently regulated signals. Immunol Cell Biol. (2000) 78:142–8. 10.1046/j.1440-1711.2000.00896.x [] [[PubMed]
  • 34. Moens L, Verbinnen B, Covens K, Wuyts G, Johnson M, Roalfe L, et al. . Anti-pneumococcal capsular polysaccharide antibody response and CD5 B lymphocyte subsets. Infect Immun. (2015) 83:2889–96. 10.1128/IAI.00068-15 ] [
  • 35. Rubinstein LJ, García-Ojeda PA, Michon F, Jennings HJ, Stein KE. Murine immune responses to Neisseria meningitidis group C capsular polysaccharide and a thymus-dependent toxoid conjugate vaccine. Infect Immun. (1998) 66:5450–6.
Collaboration tool especially designed for Life Science professionals.Drag-and-drop any entity to your messages.