Experimental autoimmune encephalomyelitis (EAE) is a CD4+ Th1/17 cell-mediated inflammatory CNS demyelinating disease that serves as a model for human multiple sclerosis (MS). Disease progression (i.e., relapses) in the SJL/J mouse is characterized by de novo activation of CD4+ T cell responses against non- crossreactive endogenous epitopes on the same or different myelin proteins (intramolecular or intermolecular epitope spreading). Effective specific therapy of established disease requires tolerogenic targeting of T cells specific for spread myelin epitopes. Tolerance can be effectively induced to ameliorate pre-established disease by spread myelin peptide-pulsed, ethylene carbodiimide (ECDI)-fixed splenic APCs (Ag-SP) indicating that responses to spread epitopes play the major pathologic role in mediating disease relapses. Ag-SP- induced tolerance is significantly more efficient and safer than tolerance induced by free peptide or peptide multimers, works primarily by indirect (cross-tolerance) representation of peptide coupled to apoptotic Ag-SP by splenic marginal zone (MZ) APCs which induce unresponsiveness via two separate but complimentary mechanisms: PD-L1/IL-10-dependent cell intrinsic anergy and activation of induced Tregs (iTregs). The experiments proposed in this renewal will test the hypothesis that the uptake of apoptotic Ag-SP by scavenger receptors on splenic MZ macrophages (MZM) induces a 'tolerogenic signature'in the APCs leading to antigen representation (cross-tolerance) and induction of both clonal anergy and activation of antigen-specific CD4+CD25+ iTregs which together regulate both na?ve and activated encephalitogenic Th1/17 cells.
Aim 1 of the proposal will build on our productive studies from the previous funding period by further defining the phenotype and functional capacity of MZ APCs involved in interaction with and tolerogenic cross-presentation of Ag-SP resulting in the antigen-specific regulation of encephalitogenic Th1 and Th17 responses. The role of scavenger receptors in Ag-SP uptake and the innate response of the tolerogenic APCs will be elucidated.
Aim 2 will test the hypothesis that maintenance of Ag-SP tolerance is primarily due to the activity of antigen-specific iTregs. The ability of Ag-SP t activate iTregs both in vivo and in vitro will be assessed, as will their role in maintenance of Ag SP tolerance and their potential ability to differentially regulate effector functions of na?ve and activated Th1 vs. Th17 cells.
Aim 3 will test the hypothesis that uptake of Ag-SP by tolerogenic splenic MZ APCs induces a cell intrinsic anergy program in na?ve and activated Th1/17 cells due to engagement of regulatory costimulatory molecules (CTLA-4/PD-L1) and/or cytokines (IL-10/TGF-?) which results in inhibition of normal Th1/17 signaling pathways resulting in inhibition of their differentiation/effector function. Collectively, these studies should enhance the understanding of the cellular/molecular mechanisms underlying a highly efficient antigen-specific tolerance therapy which is currently being tested in a MRI-controlled Phase I/IIa clinical trial in new-onset relapsing-remitting MS patients.

Public Health Relevance

The proposed studies are designed to determine the innate and adaptive immune mechanism(s) by which the i.v. infusion of antigen-coupled, ECDI-fixed apoptotic leukocytes induce antigen-specific tolerance for the prevention and treatment of mouse relapsing and progressive EAE models by specifically suppressing the activity of autoreactive Th1/17 T cells. This work has important implications for the etiology and treatment of multiple sclerosis as well as other autoimmune diseases particularly since we are currently carrying out a Phase I/IIa clinical trial testing the ability of autologous PBMCs cross-linked witha cocktail of immunodominant myelin peptides using ECDI to ameliorate disease progression in new-onset relapsing-remitting MS patients.

Agency
National Institute of Health (NIH)
Institute
National Institute of Neurological Disorders and Stroke (NINDS)
Type
Research Project (R01)
Project #
5R01NS026543-25
Application #
8662808
Study Section
Clinical Neuroimmunology and Brain Tumors Study Section (CNBT)
Program Officer
Utz, Ursula
Project Start
1988-07-01
Project End
2017-05-31
Budget Start
2014-06-01
Budget End
2015-05-31
Support Year
25
Fiscal Year
2014
Total Cost
$334,588
Indirect Cost
$118,026
Name
Northwestern University at Chicago
Department
Microbiology/Immun/Virology
Type
Schools of Medicine
DUNS #
005436803
City
Chicago
State
IL
Country
United States
Zip Code
60611
Prasad, Suchitra; Neef, Tobias; Xu, Dan et al. (2018) Tolerogenic Ag-PLG nanoparticles induce tregs to suppress activated diabetogenic CD4 and CD8 T cells. J Autoimmun 89:112-124
Pearson, Ryan M; Casey, Liam M; Hughes, Kevin R et al. (2017) Controlled Delivery of Single or Multiple Antigens in Tolerogenic Nanoparticles Using Peptide-Polymer Bioconjugates. Mol Ther 25:1655-1664
Ifergan, Igal; Davidson, Todd S; Kebir, Hania et al. (2017) Targeting the GM-CSF receptor for the treatment of CNS autoimmunity. J Autoimmun 84:1-11
Podojil, Joseph R; Miller, Stephen D (2017) Potential targeting of B7-H4 for the treatment of cancer. Immunol Rev 276:40-51
Kuo, Robert; Saito, Eiji; Miller, Stephen D et al. (2017) Peptide-Conjugated Nanoparticles Reduce Positive Co-stimulatory Expression and T Cell Activity to Induce Tolerance. Mol Ther 25:1676-1685
Pearson, Ryan M; Casey, Liam M; Hughes, Kevin R et al. (2017) In vivo reprogramming of immune cells: Technologies for induction of antigen-specific tolerance. Adv Drug Deliv Rev 114:240-255
Kang, Hee Kap; Wang, Shusen; Dangi, Anil et al. (2017) Differential Role of B Cells and IL-17 Versus IFN-? During Early and Late Rejection of Pig Islet Xenografts in Mice. Transplantation 101:1801-1810
Jeong, Su Ji; Cooper, John G; Ifergan, Igal et al. (2017) Intravenous immune-modifying nanoparticles as a therapy for spinal cord injury in mice. Neurobiol Dis 108:73-82
Edwards, Rebecca G; Kopp, Sarah J; Ifergan, Igal et al. (2017) Murine Corneal Inflammation and Nerve Damage After Infection With HSV-1 Are Promoted by HVEM and Ameliorated by Immune-Modifying Nanoparticle Therapy. Invest Ophthalmol Vis Sci 58:282-291
McCarthy, Derrick P; Yap, Jonathan Woon-Teck; Harp, Christopher T et al. (2017) An antigen-encapsulating nanoparticle platform for TH1/17 immune tolerance therapy. Nanomedicine 13:191-200

Showing the most recent 10 out of 150 publications