Our laboratory has been investigating the immunobiology of the Neisserial porin PorB for the last two decades and, through our work regarding its use as a vaccine candidate, we found that PorB had unique immune stimulatory abilities above and beyond its own potential use as a vaccine. This was due to its property as a pathogen associated molecular patter (PAMP) inducing significant antigen presenting cell activation via recognition by TOLL-like Receptor 2 (TLR2) along with heterodimerization with TLR1 (but not TLR6). More recent work from our laboratory has greatly enhanced our understanding of the breadth and mechanism of the immune stimulating activity of PorB and allowed comparison to other potential immune adjuvants, including both TLR and non-TLR ligands. PorB adjuvant activity is absolutely dependent on MyD88 expression and almost totally dependent on TLR2 expression as shown by immunizations of MyD88 or TLR2 knockout (KO) mice with PorB/Ovalbumin (OVA - as a test antigen). We have further demonstrated that PorB can enhance antigen uptake in dendritic cells (DC) and macrophages, along with improved APC trafficking to draining lymph nodes (Platt et al., submitted Journal of Immunology). In addition, immunizations of MyD88 conditional KO mice (MyD88 floxed mice) have shown that macrophage, B cell or DC MyD88 signaling is essential for PorB adjuvant activity. Moreover, we have demonstrated that PorB is a potent inducer of germinal center (GC) formation in spleens and lymph nodes of immunized mice, and this induction is MyD88 dependent. Surprisingly, we found that PorB/OVA immunization of macrophage specific MyD88 conditional KO mice failed to induce a robust germinal center response or anti-OVA antibody as normally seen in WT mice. This defect in antibody responses appears to be greater when compared to either B cell or DC MyD88 conditional KO mice, demonstrating that macrophages may be the most important APC involved with PorB adjuvanticity. Based on the information obtained from our preliminary data, we have developed the following aims to further delineate the mechanisms of adjuvant activity PorB and other TLR ligand based immune adjuvants.
Aim 1 will examine the overall effect of PorB and other vaccine adjuvants on GC formation and function using OVA as a test antigen and Influenza Nucleoprotein as a pathogen related antigen;
Aim 2 will investigate the ability of PorB and other adjuvants to directly influence SHM and antibody affinity;
and Aim 3 will delineate the role and potential function of macrophages in the adjuvant activity of PorB, especially in relation to GC formation. Importantly, the impact of findings from the studies in thi proposal go beyond just describing the use of PorB as an adjuvant, but will elucidate here-to-fore under studied mechanisms for the induction of immune responses by immune adjuvants, in general, and will have a great bearing on our overall understanding regarding germinal center formation and effective vaccine responses.

Public Health Relevance

The development of vaccines to prevent current and emerging infectious diseases is of utmost importance and the single most significant development in public health in the last half-century, and the current outbreak of Ebola in western Africa emphasizes this need. The development of new adjuvants and the understanding of their mechanisms of action are essential to make improved vaccines. The goal of this proposal is to better understand the mechanism of the potential vaccine adjuvant PorB (along with other TLR ligand based adjuvants) so that their use in vaccines can be optimized to induce the types of protective immune responses that are needed.

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
Research Project (R01)
Project #
5R01AI040944-21
Application #
9540782
Study Section
Vaccines Against Microbial Diseases Study Section (VMD)
Program Officer
Lapham, Cheryl K
Project Start
1997-04-01
Project End
2020-08-31
Budget Start
2018-09-01
Budget End
2019-08-31
Support Year
21
Fiscal Year
2018
Total Cost
Indirect Cost
Name
Boston Medical Center
Department
Type
DUNS #
005492160
City
Boston
State
MA
Country
United States
Zip Code
Mosaheb, Munir; Wetzler, Lee M (2018) Meningococcal PorB induces a robust and diverse antigen specific T cell response as a vaccine adjuvant. Vaccine 36:7689-7699
Mosaheb, Munir M; Reiser, Michael L; Wetzler, Lee M (2017) Toll-Like Receptor Ligand-Based Vaccine Adjuvants Require Intact MyD88 Signaling in Antigen-Presenting Cells for Germinal Center Formation and Antibody Production. Front Immunol 8:225
Reiser, Michael L; Mosaheb, Munir M; Lisk, Christina et al. (2017) The TLR2 Binding Neisserial Porin PorB Enhances Antigen Presenting Cell Trafficking and Cross-presentation. Sci Rep 7:736
Toussi, Deana N; Wetzler, Lee M; Liu, Xiuping et al. (2016) Neisseriae internalization by epithelial cells is enhanced by TLR2 stimulation. Microbes Infect 18:627-638
Kattner, Christof; Pfennig, Sabrina; Massari, Paola et al. (2015) One-step purification and porin transport activity of the major outer membrane proteins P2 from Haemophilus influenzae, FomA from Fusobacterium nucleatum and PorB from Neisseria meningitidis. Appl Biochem Biotechnol 175:2907-15
Kattner, Christof; Toussi, Deana N; Zaucha, Jan et al. (2014) Crystallographic analysis of Neisseria meningitidis PorB extracellular loops potentially implicated in TLR2 recognition. J Struct Biol 185:440-7
Platt, Andrew; MacLeod, Heather; Massari, Paola et al. (2013) In vivo and in vitro characterization of the immune stimulating activity of the Neisserial porin PorB. PLoS One 8:e82171
Massari, Paola; Wetzler, Lee M (2012) Analysis of parameters associated with prevention of cellular apoptosis by pathogenic Neisseriae and purified porins. Methods Mol Biol 799:319-41
Toussi, Deana N; Carraway, Margaretha; Wetzler, Lee M et al. (2012) The amino acid sequence of Neisseria lactamica PorB surface-exposed loops influences Toll-like receptor 2-dependent cell activation. Infect Immun 80:3417-28
Arjunaraja, Swadhinya; Massari, Paola; Wetzler, Lee M et al. (2012) The nature of an in vivo anti-capsular polysaccharide response is markedly influenced by the composition and/or architecture of the bacterial subcapsular domain. J Immunol 188:569-77

Showing the most recent 10 out of 25 publications