Identification of antigens that are involved in the pathogenesis of type 1 diabetes (T1D) is crucial to develop biomarkers and therapies for T1D. There have been recent efforts focused on identifying antigens recognized by T cells within the residual islets of T1D organ donors due to the premise that such T cells are likely to be involved in disease pathogenesis. Our preliminary data studying islet-infiltrating CD8 T cells demonstrates that a large proportion, 1/3 to 1/2 of T1D organ donors, are reactive to preproinsulin (PPI) peptides, whereas the remaining donors have only few or no PPI-reactive CD8 T cells in the islets. Epitopes recognized by the PPI- reactive CD8 T cells are presented by many HLA class I molecules spanning HLA-A, B, and C, and are spread throughout the PPI protein with several hot spots that are preferentially recognized by multiple T cell receptors (TCR) expressed by the T cells. Thus, preproinsulin is a major self-antigen and contains epitopes for T1D- associated CD8 T cells in a subset of T1D patients. Yet antigens targeted by islet-infiltrating T cells in the remaining patients exist outside PPI, suggesting heterogeneity in antigen specificity targeted by islet-resident CD8 T cells. Towards an ultimate goal of developing antigen-specific biomarkers and therapies for T1D, two important questions remain; (1) What antigens are recognized by islet-infiltrating CD8 T cells that do not react with PPI? (2) By measuring reactivity in peripheral blood, can we identify patients that have a particular antigen specificity in order to design appropriate therapy for each patient? The goal of this proposal is to uncover unknown antigen specificities recognized by islet-derived CD8 T cells and to evaluate the association between PPI reactivity in peripheral blood and the response to oral insulin as a proof of principle for personalized antigen- specific therapies. Our central hypothesis is that heterogeneity in self-antigen specificity for islet-derived CD8 T cells exists and determines the response to antigen-specific immunotherapy. If this hypothesis is correct, self- antigens targeted by CD8 T cells can be utilized to identify patients with particular antigen specificity to select for responders receiving antigen-specific immunotherapies for T1D prevention efforts. To test the hypothesis, we will identify antigens that are recognized by islet-infiltrating CD8 T cells either in a protein-targeted manner (Aim 1) and in an unbiased manner (Aim 2). Given our preliminary results that PPI is a major antigen for islet-infiltrating CD8 T cells in a subset of patients, we will focus on PPI to test whether there is an association between CD8 PPI reactivity and the response to oral insulin treatment (Aim 3). The successful completion of this proposal will result in identification of major self-antigens for islet-resident CD8 T cells in T1D patients. The identification of antigens can be used to classify heterogeneous T1D patients based on CD8 T cell antigen specificity with the goal to ultimately providing appropriate personalized antigen-specific immunotherapy for T1D prevention efforts.

Public Health Relevance

Type 1 diabetes occurs due to autoimmune destruction of insulin-secreting beta cells in the pancreas. T immune cells attack beta cells by recognizing molecules that are expressed by the cell; however such targeted molecules, antigens, are not fully elucidated. This project aims to characterize antigen profiles for T immune cells that are associated with the development of type 1 diabetes. Insights obtained from this project will guide us to identify novel biomarkers for type 1 diabetes and to develop approaches to therapy through the induction of antigen- specific immune tolerance to ultimately prevent and cure type 1 diabetes.

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
Research Project (R01)
Project #
2R01DK099317-06A1
Application #
9981284
Study Section
Hypersensitivity, Autoimmune, and Immune-mediated Diseases Study Section (HAI)
Program Officer
Spain, Lisa M
Project Start
2013-08-20
Project End
2024-05-31
Budget Start
2020-08-19
Budget End
2021-05-31
Support Year
6
Fiscal Year
2020
Total Cost
Indirect Cost
Name
University of Colorado Denver
Department
Pediatrics
Type
Schools of Medicine
DUNS #
041096314
City
Aurora
State
CO
Country
United States
Zip Code
80045
Baker, Rocky L; Jamison, Braxton L; Wiles, Timothy A et al. (2018) CD4 T Cells Reactive to Hybrid Insulin Peptides Are Indicators of Disease Activity in the NOD Mouse. Diabetes 67:1836-1846
Burrack, Adam L; Landry, Laurie G; Siebert, Janet et al. (2018) Simultaneous Recognition of Allogeneic MHC and Cognate Autoantigen by Autoreactive T Cells in Transplant Rejection. J Immunol 200:1504-1512
Michels, Aaron W; Landry, Laurie G; McDaniel, Kristen A et al. (2017) Islet-Derived CD4 T Cells Targeting Proinsulin in Human Autoimmune Diabetes. Diabetes 66:722-734
Marrack, Philippa; Krovi, Sai Harsha; Silberman, Daniel et al. (2017) The somatically generated portion of T cell receptor CDR3? contributes to the MHC allele specificity of the T cell receptor. Elife 6:
Seay, Howard R; Yusko, Erik; Rothweiler, Stephanie J et al. (2016) Tissue distribution and clonal diversity of the T and B cell repertoire in type 1 diabetes. JCI Insight 1:e88242
Nakayama, Maki; Simmons, Kimberly M; Michels, Aaron W (2015) Molecular Interactions Governing Autoantigen Presentation in Type 1 Diabetes. Curr Diab Rep 15:113
Nakayama, Maki; McDaniel, Kristen; Fitzgerald-Miller, Lisa et al. (2015) Regulatory vs. inflammatory cytokine T-cell responses to mutated insulin peptides in healthy and type 1 diabetic subjects. Proc Natl Acad Sci U S A 112:4429-34
Zhang, Li; Crawford, Frances; Yu, Liping et al. (2014) Monoclonal antibody blocking the recognition of an insulin peptide-MHC complex modulates type 1 diabetes. Proc Natl Acad Sci U S A 111:2656-61
Bettini, Maria; Blanchfield, Lori; Castellaw, Ashley et al. (2014) TCR affinity and tolerance mechanisms converge to shape T cell diabetogenic potential. J Immunol 193:571-9
Bettini, Matthew L; Bettini, Maria; Nakayama, Maki et al. (2013) Generation of T cell receptor-retrogenic mice: improved retroviral-mediated stem cell gene transfer. Nat Protoc 8:1837-40

Showing the most recent 10 out of 11 publications