ECU Libraries Catalog

A single-chain GMCSF-MOG tolerogenic vaccine expands MOG-specific CD25+ FOXP3+ regulatory T cells through low-efficiency antigen recognition events to inhibit experimental autoimmune encephalomyelitis / by Cody Deumont Moorman.

Author/creator Moorman, Cody Deumont author.
Other author/creatorMannie, Mark D., degree supervisor.
Other author/creatorEast Carolina University. Department of Microbiology and Immunology.
Format Theses and dissertations, Electronic, and Book
Publication Info [Greenville, N.C.] : [East Carolina University], 2019.
Description187 pages : illustrations (some color).
Supplemental Content Access via ScholarShip
Subject(s)
Series ECU Brody School of Medicine dissertation
ECU Brody School of Medicine dissertation. UNAUTHORIZED
Summary Previous studies showed that tolerogenic vaccines comprised of single-chain GMCSF-neuroantigen (NAg) fusion proteins inhibited experimental autoimmune encephalomyelitis (EAE) in rodents. The studies detailed here provide evidence that GMCSF-NAg vaccines elicited tolerance through the expansion of preexisting NAg-specific regulatory T cells (Tregs) via low-efficiency antigen recognition that was below the CD40/CD40L activation threshold. GMCSF-NAg-induced tolerance was dependent upon vaccine-induced Tregs, because treatment of mice with a Treg-depleting mAb reversed vaccine-induced tolerance. Vaccine-induced T cell responses were investigated using T cell receptor (TCR) transgenic OTII-FIG mice which recognize OVA323-337 as a high-efficiency antigen, and 2D2-FIG mice which recognize MOG35-55 as a low-efficiency antigen and NFM13-37 as a high-efficiency antigen. Subcutaneous vaccination of 2D2-FIG mice with the low-efficiency GMCSF-MOG vaccine elicited a major Treg population that appeared within 3 days, was sustained over several weeks, expressed canonical Treg markers, and was present systemically in the blood, spleen, and lymph nodes. The GMCSF-MOG vaccine required covalent linkage because a vaccine that contained GM-CSF and MOG35-55 as separate molecules did not elicit Treg responses. GMCSF-MOG vaccination elicited Tregs when introduced either subcutaneously or intravenously as well as in the proinflammatory adjuvants CFA and alum. The GMCSF-MOG-induced Tregs were immunosuppressive and prevented the proliferation of MOG35-55-specific T cells. The GMCSF-MOG vaccine not only elicited Tregs but also induced a desensitized MOG35-55-specific (2D2) T cell repertoire because the vaccine decreased the number of 2D2 CD3+ T cells, reduced the overall expression of the 2D2 TCR, and increased the CD4- T cell compartment. The ability of GMCSF-NAg vaccines to induce Tregs was dependent upon the efficiency of T cell antigen recognition, because treatment of OTII-FIG and 2D2-FIG mice with the high-efficiency GMCSF-OVA and GMCSF-NFM vaccines respectively, did not elicit Treg responses. The high-efficiency GMCSF-NFM vaccine induced a vigorous T conventional cell (Tcon) memory response and activated the CD40L/CD40 co-stimulatory pathway. In contrast, the low-efficiency GMCSF-MOG vaccine elicited Tregs and lacked sufficient TCR signal strength to activate CD40L/CD40 pathway. Activation of the CD40L/CD40 pathway using an agonistic anti-CD40 mAb precluded Treg expansion with the low-efficiency GMCSF-MOG vaccine in 2D2-FIG mice. Therefore, the strength of the TCR stimulus and the downstream activation or exclusion of the CD40L/CD40 costimulatory pathway was the switch that controlled Tcon versus Treg responses respectively. Remarkably, the low-efficiency GMCSF-MOG vaccine retained Treg expansive activity when co-administered with the high-efficiency GMCSF-NFM vaccine in 2D2-FIG mice. The GMCSF-MOG vaccine appeared to predominantly drive Treg expansion rather than Treg induction because the emergence of Tregs was delayed in 2D2-FIG-Rag-/- mice which have reduced frequencies of pre-existing Tregs as compared to 2D2-FIG mice. Pre-existing Tregs were also required for tolerance because GMCSF-MOG was encephalitogenic in 2D2-FIG-Rag1-/- mice but not in 2D2-FIG mice. Likewise, GMCSF-MOG was an effective prophylactic in Treg-sufficient C57BL/6 mice and prevented active EAE. Overall, these studies provide evidence that GM-CSF is an effective tolerogenic adjuvant when combined with low-efficiency peptides that fall below the CD40L/CD40 triggering threshold. Thus, a subthreshold CD40L/ CD40 response delimits a critical parameter needed for antigen-specific tolerance and expansion of pre-existing Treg populations.
General notePresented to the faculty of the Department of Microbiology and Immunology
General noteAdvisor: Mark D. Mannie
General noteTitle from PDF t.p. (viewed April 3, 2020).
Dissertation notePh.D. East Carolina University 2019.
Bibliography noteIncludes bibliographical references.
Technical detailsSystem requirements: Adobe Reader.
Technical detailsMode of access: World Wide Web.

Available Items

Library Location Call Number Status Item Actions
Electronic Resources Access Content Online ✔ Available