The β-cell plays a crucial role in the pathogenesis of type 1 diabetes, in part through the posttranslational modification of self-proteins by biochemical processes such as deamidation. These neoantigens are potential triggers for breaking immune tolerance. We report the detection by LC-MS/MS of 16 novel Gln and 27 novel Asn deamidations in 14 disease-related proteins within inflammatory cytokine–stressed human islets of Langerhans. T-cell clones responsive against one Gln- and three Asn-deamidated peptides could be isolated from peripheral blood of individuals with type 1 diabetes. Ex vivo HLA class II tetramer staining detected higher T-cell frequencies in individuals with the disease compared with control individuals. Furthermore, there was a positive correlation between the frequencies of T cells specific for deamidated peptides, insulin antibody levels at diagnosis, and duration of disease. These results highlight that stressed human islets are prone to enzymatic and biochemical deamidation and suggest that both Gln- and Asn-deamidated peptides can promote the activation and expansion of autoreactive CD4+ T cells. These findings add to the growing evidence that posttranslational modifications undermine tolerance and may open the road for the development of new diagnostic and therapeutic applications for individuals living with type 1 diabetes.

Article Highlights
  • Posttranslationally modified neoantigens are formed in β-cells under conditions of inflammation, but studies to date have focused on a limited number of antigens.

  • We sought to identify disease-relevant β-cell neoantigens through unbiased proteomic analysis and confirmation by antigen-specific T-cell assays.

  • We detected novel Gln and Asn deamidations within 14 type 1 diabetes–related proteins in cytokine-stressed human islets and verified disease-associated T-cell recognition for three corresponding epitopes.

  • These results demonstrate that both enzymatic and biochemical deamidation in cytokine-exposed islets undermine self-tolerance and lay a foundation for new diagnostic and therapeutic approaches for individuals living with type 1 diabetes.

Enzymatic deamidation is a posttranslational modification (PTM) mediated by transglutaminase (TGM) enzymes. The result is conversion of a glutamine (Gln) to a glutamic acid (Glu) residue, introducing a negative charge and a mass increase of 0.98 Da. The role of enzymatic deamidation is well established in celiac disease (1), where deamidation enhances the presentation of gliadin peptides by HLA molecules and induces robust T-cell activation (2). Type 1 diabetes is an autoimmune disease that shares susceptible HLA genes with celiac disease and for which a major hallmark is development of islet cell autoantibodies (3). Susceptible HLA class II molecules are thought to promote thymic selection of a potentially autoreactive T-cell repertoire (4). For HLA class I molecules, this hypothesis was borne out by an observed state of benign autoimmunity, i.e., the presence of islet-reactive CD8+ T cells at similar circulating frequencies in individuals with type 1 diabetes and HLA-matched control individuals (5,6). Likewise, the type 1 diabetes–associated DR4 haplotype is sufficient to select a CD4+ T-cell repertoire that includes high-affinity self-reactive T cells (7). The most common HLA class II haplotype in celiac disease is DR3/DQ2 (>90% of patients with celiac disease harbor at least one copy) (8). The second most predominant haplotype in celiac disease and most highly susceptible haplotype in type 1 diabetes is DR4/DQ8. Both DQ2 and DQ8 anchor negatively charged peptides tightly in their binding groove, with DQ2 preferring negative charges at position 4 (P4) or P6 and DQ8 at P1, P6, and P9 (4,9). HLA-DRB1*04:01, the HLA-DRB1*04 allotype with the highest odds ratio for type 1 diabetes development, accepts negatively charged amino acids at P4 (4). As deamidation induces a negative charge, this modification can increase peptide binding to these susceptible HLA class II haplotypes. A CD4+ T-cell line against deamidated insulin C-peptide was isolated from a patient with new-onset type 1 diabetes homozygous for HLA-DQ8, and similar immune responses could be observed in newly diagnosed individuals (10). Of note, a more recent research report has challenged this conclusion, showing no evidence that deamidation increased the immunogenicity of C-peptide (11). T cells specific for a deamidated GAD65 peptide were more frequent in the peripheral blood of individuals with type 1 diabetes compared with healthy control individuals (12), and T-cell receptors that recognized the deamidated peptide were shown to more readily escape thymic selection in HLA transgenic mice (13). The deamidated insulin peptide showed increased binding to HLA-DQ8, while the deamidated GAD65 peptide showed increased binding to HLA-DRB1*04:01 compared with their native forms (10,12).

In addition to enzymatic deamidation, nonenzymatic deamidation occurs, converting asparagine (Asn) to aspartic acid (Asp) (14) or isoAsp and Gln to Glu or isoGlu. Nonenzymatic conversion of Glu or isoGlu occurs through the formation of a six-member-ring glutarimide intermediate, which is less favored than the corresponding succinimide intermediate formed in Asp deamidation, making this reaction much slower (15). We previously showed that nonenzymatic Asn deamidation is significantly increased in cytokine-exposed murine MIN6 β-cells (15). To date, no published study has investigated the role of nonenzymatic Asn deamidation in type 1 diabetes. Given that Asn deamidation is increased in stressed β-cells and results in negative charges that can enhance peptide binding and presentation by type 1 diabetes–susceptible HLA molecules, we hypothesized that this modification may generate neoantigens in type 1 diabetes.

Although deamidation of insulin C-peptide was documented by mass spectrometry (MS) (16), deamidation of GAD65 and other putative PTM autoantigens has never been directly shown in the islets of Langerhans. Here, we exposed human islets of Langerhans (primary islets) to a mixture of inflammatory cytokines or to interferon-α (IFN-α), to mimic inflammation and viral infection, and performed proteomic analysis to identify enzymatically and nonenzymatically deamidated peptides. Tetramer-based assays were then used to show that T cells recognizing deamidated peptides were present in peripheral blood of individuals with type 1 diabetes.

Islet Culture and Treatment

Human islets were obtained from Pisa University with approval by its local ethics committee (#2615). Characteristics of human islet donors are listed in Supplementary Table 1. Islets were cultured in DMEM supplemented with 1% l-glutamine (Gibco), 10% FBS, and 100 units/mL penicillin-streptomycin and exposed to human interleukin 1β (IL-1β) (50 units/mL; R&D Systems), murine tumor necrosis factor-α (TNF-α) (1,000 units/mL; R&D Systems), and human IFN-γ (1,000 units/mL; PeproTech) or human IFN-α (2,000 units/mL; PBL Assay Science) for 24 or 72 h.

Human Subjects and PBMC Isolation

Heparinized blood was obtained from control individuals and individuals with type 1 diabetes. All procedures were approved by the ethics committee of UZ Leuven and the institutional review board at the Benaroya Research Institute. Subject attributes are summarized in Supplementary Tables 2 and 3. Peripheral blood mononuclear cells (PBMCs) were isolated using Lymphoprep and frozen in AIM V Medium (Gibco) with 10% DMSO.

RNA Extraction and Quantitative RT-PCR

RNA was extracted from primary islets using the RNeasy Micro Kit (QIAGEN), and cDNA was made using oligo-d(T) and Superscript II Reverse Transcriptase (Invitrogen). Quantitative RT-PCR was performed on a StepOnePlus RT-PCR System (Applied Biosystems, Santa Clara, CA). The relative fold gene expression was calculated using the delta-delta Ct method.

Cell Death Assay

Human islets were incubated with propidium iodide (Invitrogen) and Hoechst 33342 (Invitrogen) for 15 min at 37°C. At least 10 islets were assessed for apoptosis by two independent researchers on a Nikon Eclipse TI microscope.

Analysis of Single-Cell RNA Sequencing and Pseudobulk Differential Expression Data

Cell Ranger (version 6.1.2) (17) was used for alignment (reference genome hg38) and filtering of raw sequencing reads retrieved from the Human Pancreas Analysis Program (HPAP) (https://hpap.pmacs.upenn.edu) (donor characteristics listed in Supplementary Table 4). Decontamination of ambient mRNA was performed for each sample using SoupX (version 1.6.1) (18), using genes associated with major cell types identified in the initial Seurat (version 4.1.1) (19) clustering. Single-cell gene expression profiles were imported into Seurat for quality control, retaining genes expressed in at least three cells expressing a minimum of 200 genes. Potential doublet cells were identified and removed using the R package scDbIFinder (version 3.16) (20). Additional filtering criteria included nFeature_RNA <9,000, mitochondrial genes <10%, and nCount_RNA <10,000, resulting in high-quality single cells for downstream analysis. Seurat’s SCTransform function was applied to measure differences in sequencing depth per cell and normalize counts by removing variation due to sequencing depth (number of unique molecular identifiers). The top 3,000 variable features were selected for principal component analysis and uniform manifold approximation and projection embedding. To assign cells to known cell types based on marker genes, we used the R package scSorter (version 0.0.2) (21). Subsequently, we extracted pancreatic endocrine cells (α-cells, β-cells, δ-cells, and γ-cells) and pancreatic nonendocrine cells (acinar cells, ductal cells, and endothelial cells) for downstream pseudobulk differential expression analysis. To evaluate the mRNA expression changes of TGM2 within endocrine and nonendocrine cells, we used a pseudobulk analysis strategy as previously described (22). Specifically, we extracted the counts matrix and aggregated the counts for each gene from individual cells to the sample level using the aggregate.Matrix function. This involved combining the counts of all endocrine (13,159 cells for individuals without diabetes and 22,040 cells for individuals with diabetes) or nonendocrine cells (12,417 cells for individuals without diabetes and 18,967 cells for individuals with diabetes) at the sample level. Subsequently, we imported the aggregated counts matrix into DESeq2 (23) for the analysis of differential gene expression across the specified conditions. Statistical significance was assessed by DESeq2, and P values were corrected for multiple testing using the Benjamini-Hochberg method. Finally, the normalized counts matrix by DESeq2 for TGM2 was selected for visualization using the R package ggplot2.

Functional Enrichment Analysis

Functional enrichment analysis to identify enriched biological pathways associated with upregulated genes in endocrine cells of individuals with diabetes, focusing on the Reactome database, was conducted using g:Profiler (https://biit.cs.ut.ee/gprofiler/gost) (a list of genes differentially expressed is provided in Supplementary Table 5). The g:SCS algorithm was applied for multiple testing correction of P values obtained from a hypergeometric test–based pathway enrichment analysis. The number of genes contributing to the significance of the gene set enrichment are depicted on the x-axis of Fig. 1K.

Figure 1

Primary islets exposed to inflammatory cytokines or IFN-α and islets from individuals affected by type 1 diabetes (T1D) upregulate TGM2. A: Cell death after 72 h in control and inflammatory cytokine–treated primary islets. BD: CHOP, ATF3, and TGM2 mRNA levels after 24 h in control and inflammatory cytokine–treated human islets. EH: CXCL10, MX1, HLA-I, and TGM2 mRNA levels after 24 h in control- and IFN-α–treated human islets. I: TGM2 expression levels in transcriptomic data obtained from endocrine cells (α-cells, β-cells, δ-cells, and γ-cells) from islets from control individuals and individuals with diabetes from the HPAP-T1D cohort after pseudobulk analysis. J: TGM2 expression levels in transcriptomic data obtained from nonendocrine cells (acinar cells, ductal cells, and endothelial cells) from control individuals and individuals with diabetes from the HPAP-T1D cohort after pseudobulk analysis. K: Functional enrichment analysis for upregulated genes in endocrine cells of individuals with T1D after pseudobulk analysis identified enriched biological pathways associated with the 125 upregulated genes. Inflammatory cytokine treatment: 50 units/mL IL-1β, 1,000 units/mL TNF-α, and 1,000 units/mL IFN-γ. IFN-α treatment: 2,000 units/mL IFN-α. Data are mean ± SEM. Each dot represents one unique donor. *P < 0.05 by Wilcoxon test (AH); ***P < 0.001 by DESeq2 with correction for multiple testing using the Benjamini-Hochberg method (I and J). The g:SCS algorithm was applied for multiple testing correction of P values (K). CTR, control; CYT, cytochrome; ER, endoplasmic reticulum; padj, adjusted P value; rel., relative.

Figure 1

Primary islets exposed to inflammatory cytokines or IFN-α and islets from individuals affected by type 1 diabetes (T1D) upregulate TGM2. A: Cell death after 72 h in control and inflammatory cytokine–treated primary islets. BD: CHOP, ATF3, and TGM2 mRNA levels after 24 h in control and inflammatory cytokine–treated human islets. EH: CXCL10, MX1, HLA-I, and TGM2 mRNA levels after 24 h in control- and IFN-α–treated human islets. I: TGM2 expression levels in transcriptomic data obtained from endocrine cells (α-cells, β-cells, δ-cells, and γ-cells) from islets from control individuals and individuals with diabetes from the HPAP-T1D cohort after pseudobulk analysis. J: TGM2 expression levels in transcriptomic data obtained from nonendocrine cells (acinar cells, ductal cells, and endothelial cells) from control individuals and individuals with diabetes from the HPAP-T1D cohort after pseudobulk analysis. K: Functional enrichment analysis for upregulated genes in endocrine cells of individuals with T1D after pseudobulk analysis identified enriched biological pathways associated with the 125 upregulated genes. Inflammatory cytokine treatment: 50 units/mL IL-1β, 1,000 units/mL TNF-α, and 1,000 units/mL IFN-γ. IFN-α treatment: 2,000 units/mL IFN-α. Data are mean ± SEM. Each dot represents one unique donor. *P < 0.05 by Wilcoxon test (AH); ***P < 0.001 by DESeq2 with correction for multiple testing using the Benjamini-Hochberg method (I and J). The g:SCS algorithm was applied for multiple testing correction of P values (K). CTR, control; CYT, cytochrome; ER, endoplasmic reticulum; padj, adjusted P value; rel., relative.

Close modal

Orbitrap Liquid Chromatography-Tandem MS

Human islets were prepared for liquid chromatography-tandem MS (LC-MS/MS) as previously described (24). Digestion was performed with modified trypsin (trypsin:protein ratio of 1:10 w/w; Pierce) for 90 min to minimize artifactual deamidations (15). Solvent-evaporated peptide mixtures subjected to desalting with C18 ZipTip pipet tips (Millipore) were dissolved in 0.1% formic acid and 5% acetonitrile and run on a Q Exactive Orbitrap Mass Spectrometer (Thermo Fisher Scientific). Peptides were identified by Mascot (Matrix Science) using UniProt (Homo sapiens, 169,779 entries) as a database through Proteome Discoverer 2.2, incorporating Percolator for peptide validation. Oxidation (M), deamidation (N/Q), and citrullination (R), resulting in the same mass increase as deamidation, were included as variable modifications. Carbamidomethylation (C) was included as a fixed modification. Two missed cleavages were allowed, and peptide tolerance was set at 5 parts per million and MS/MS tolerance at 20 millimass units. Spectra of predicted deamidated peptides were manually checked as previously described (15).

Peptide-HLA Binding Prediction and Affinity Assay

Predicted binding of 27 peptides containing deamidated glutamine residues and 30 peptides containing deamidated Asn residues (as detected by LC-MS/MS) to HLA-DRB1*04:01 was performed using a previously described approach (25,26). Supplementary Table 6 shows predicted binding for all deamidated peptides. Peptides with a relative binding affinity score of ≥0.075 were predicted to bind. Selected deamidated peptides were synthesized (Synpeptide), and their binding was measured by incubating increasing concentrations of each peptide with 0.02 μmol/L biotinylated influenza hemagglutinin (HA) 306–318 in wells coated with recombinant DRB1*04:01 protein (purified from insect cell cultures as previously described [27]). Residual biotin-HA was detected after washing with europium-conjugated streptavidin on a Victor Nivo time-resolved fluorometer (PerkinElmer). IC50 values were calculated as the concentration of peptide needed to displace 50% of the biotin-HA peptide. The sequences and IC50 values of peptides with measurable binding are listed in Table 3. For peptides subsequently shown to be immunogenic, the corresponding wild-type peptides were synthesized, and their binding was measured using the same competition assay. Comparisons of IC50 values for deamidated and wild-type peptides are listed in Supplementary Table 7.

HLA Class II Protein and Tetramer Reagents

Recombinant DRB1*04:01 monomers were purified from insect cell cultures as previously described (28) and loaded with peptide (0.2 mg/mL) in the presence of n-dodecyl-β-maltoside (0.2 mg/mL) and Pefabloc (1 mmol/L; Sigma) for 72 h at 37°C. Peptide-loaded monomers were conjugated into tetramers using streptavidin at a molar ratio of 8:1.

In Vitro Tetramer Assay and T-Cell Clone Isolation

PBMCs (5 × 106) were stimulated with pools of deamidated peptides and a negative control peptide (WGTLTDCVVMXDPQT, with X being a citrulline residue shown to elicit consistent negative T-cell responses) in 48-well plates for 14 days. Starting on day 7, human IL-2 (10 units/mL; Roche) was added every 2 days. Expanded cells were stained with tetramers for 1 h at 37°C and with CD3-BV510, CD4-APC, CD14-PerCPCy5.5, CD19-PerCPCy5.5, and CD25-FITC antibodies for 20 min at 4°C. Cells were acquired on an LSR II Flow Cytometer (BD Biosciences) and analyzed using FlowJo (Treestar Inc.). The gating strategy is shown in Supplementary Fig. 1A. Clones were isolated from cultures by sorting single tetramer-positive CD4+ T cells using FACSAria (BD Biosciences) and expanded in the presence of 105 irradiated PBMCs and phyto-HA (2 μg/mL; Remel Inc.) in 96-well plates.

T-Cell Clone Specificity Assessment

To assess preferential recognition of deamidated peptides, 104 T cells were plated in triplicate in the presence of 105 irradiated HLA-DRB1*04:01+ PBMCs as antigen-presenting cells (APCs) and stimulated with 10 μg/mL native or deamidated peptide. After incubation for 72 h at 37°C, cells were pulsed with medium containing 3H-thymidine (1 μCi/well). Incorporation was measured 18 h later with a scintillation counter. Data are represented as stimulation index, calculated by normalizing the 3H-thymidine incorporation of each clone with that of nonstimulated wells.

Ex Vivo Tetramer Assay

Cryopreserved PBMCs (20–30 × 106) were thawed and treated with dasatinib (50 nmol/L; Cell Signaling Technology) for 10 min at 37°C and stained with 20 μg/mL phycoerythrin (PE)-labeled, PECF594-labeled, SPRD-labeled, PECy7-labeled, and APC-labeled tetramers for 90 min at room temperature. Cells were then washed and labeled with anti-PE and anti-APC magnetic beads for 20 min at 4°C. A preenriched fraction (2%) of the cells was reserved to determine the total cell number. Remaining cells were enriched using Miltenyi Biotec MS magnetic columns according to the manufacturer’s instructions. Both fractions were stained for 30 min at 4°C with CD4-BUV395, CD14-FITC, CD19-FITC, CD45RA-AF700, CXCR3-BV786, CCR7-BV650, CCR6-BV510, and CCR4-BUV605 antibodies. After washing, cells were labeled with Sytox Green (Thermo Fisher Scientific), run on an LSR II Flow Cytometer, and analyzed using FlowJo. The gating strategy is shown in Supplementary Fig. 1B.

Islet-Derived T-Cell Assays

Cell lines from islet-infiltrating T cells were recovered and expanded from Network for Pancreatic Organ Donors With Diabetes (nPOD) donor 6551 (Supplementary Table 8). For generation of lines, 150-μm live slices of pancreas were digested with 1 mg/mL collagenase-P (Sigma-Aldrich) in Hanks’ balanced salt solution with Mg2+/Ca2+ (Thermo Fisher Scientific) for 10 min in a 37°C water bath with vigorous shaking. Digestion was confirmed by visual inspection. Digested tissue was washed and placed in medium (AIM V supplemented with 2 mmol/L l-glutamine, 5 mmol/L HEPES, 100 units/mL penicillin, 100 μg/mL streptomycin, 0.1 mmol/L of each nonessential amino acid, and 1 mmol/L sodium pyruvate; all from Lonza, Walkersville, MD) with 5% heat-inactivated Human Male AB Serum (Access Biologicals, Vista, CA). Individual islets were handpicked and placed in culture with irradiated allogeneic PBMCs as feeders, a blocking anti-Fas antibody (1 μg/mL; Applied Biosystems); anti-PD1 antibody (1 μg/mL; BD Biosciences), and mifepristone (100 nmol/L; Invitrogen). After 12 days of culture, T-cell lines were collected, further expanded, and phenotyped by flow cytometry for T-cell subsets. Epstein-Barr virus–transformed B-lymphoblastoid cell lines were generated from autologous splenocytes as previously described (29). Responsiveness was assessed as previously described (30). Briefly, irradiated autologous B-lymphoblastoid cell lines were plated (20,000/well) after pulsing with peptides at 50 μg/mL, no peptide, or DMSO volume control and then cultured with T cells (75,000/well) and anti-CD28 (5 μg/mL) for costimulation. After 48 h of coculture, responses were detected by Cytometric Bead Array Human IFN-γ Enhanced Sensitivity Set (BD Biosciences), with a lower limit of 274 fg/mL sensitivity.

Statistical Analyses

Data were analyzed using GraphPad Prism 9 (GraphPad, La Jolla, CA). Statistical tests included Wilcoxon test, paired t test, unpaired t test with Welch correction, and one-way ANOVA. Analysis of quantitative RT-PCR data was performed on delta CT values. All significant differences are reported.

Data and Resource Availability

MS proteomics data have been deposited to the ProteomeXchange Consortium via the Proteomics Identifications (PRIDE) (31) partner repository with the data set identifier PXD038758. Other data sets are available from the corresponding author upon reasonable request.

Primary Islets Exposed to Cytokines and Pancreatic Endocrine Cells From Individuals With Type 1 Diabetes Upregulate TGM2

Primary islets from donors without diabetes were exposed to cytokines (IL-1β, TNF-α, and IFN-γ) to mimic inflammation or IFN-α to mimic antiviral response. IFN-α is crucial in innate and adaptive immunity and mediates HLA class I overexpression in human islets in type 1 diabetes (32,33). Inflammatory cytokines induced apoptosis after 72 h (24.90% vs. 5.84% in control islets) (Fig. 1A), and cell death was preceded by endoplasmic reticulum stress, indicated by increased C/EBP homologous protein (CHOP) and activating transcription factor 3 (ATF3) mRNA levels (1.60- and 3.41-fold increase compared with control islets) (Fig. 1B and C). IFN-α induced upregulation of chemokine C-X-C motif chemokine ligand 10 (CXCL10), the antiviral MX dynamin-like GTPase 1 (MX1), and MHC class I mRNA after 24 h (233.85-, 34.18-, and 2.40-fold compared with control islets, respectively) (Fig. 1E–G), as previously described (34). IFN-α did not induce apoptosis (data not shown). Exposure to inflammatory cytokines or IFN-α resulted in significant upregulation of TGM2 mRNA levels, with a 5.17- and 2.15-fold increase compared with control islets, respectively (Fig. 1D and H). Next, TGM2 expression was investigated in RNA sequencing data retrieved from the HPAP database in endocrine and nonendocrine cells from islets obtained from individuals with and without diabetes. While there was no difference in nonendocrine cells, TGM2 expression was significantly increased in islet endocrine cells of individuals with diabetes compared with control individuals (Fig. 1I and J). A functional enrichment analysis of genes enriched in endocrine cells of individuals with diabetes showed the presence of inflammation-induced gene signatures, mostly downstream of IFNs (Fig. 1K), similar to our previous findings (35).

Novel Deamidations Occur in Stressed Primary Islets

Protein lysates from primary islets exposed to inflammatory cytokines or IFN-α were subjected to LC-MS/MS and searched for Gln and Asn deamidations. Digestion and proteomic analysis of cytokine-exposed islets resulted in the detection of 3,587 ± 823 and 4,245 ± 801 proteins, respectively (identified with high confidence), corresponding to 27,475 ± 5,564 and 30,658 ± 2,930 unique peptides, respectively. Of these unique peptides, 1,510 ± 315 (or 5.52% ± 0.61%) and 1,586 ± 293 (or 5.13% ± 0.55%) were predicted to have one or more deamidated or citrullinated residues. MS/MS spectra were manually inspected to confirm deamidation, as previously described (15). A total of 16 novel enzymatic Gln deamidations were confirmed, with 8 only occurring in the inflammatory cytokine–stressed islets (donors 1–4), 4 only occurring in the IFN-α–stressed islets (donors 5–8), and 4 occurring in both sets of islets (Table 1). Nonenzymatic Asn deamidations were less influenced by the kind of stress; out of 25 confirmed Asn-deamidated peptides, 11 only occurred in inflammatory cytokine–stressed islets, 2 only in IFN-α–stressed islets, and 12 in both sets of islets (Table 2). Of note, nonenzymatic deamidations were restricted to fewer proteins than enzymatic deamidations, but more residues within each protein were deamidated (5 proteins with up to 9 Asn-deamidated residues per protein vs. 14 proteins with a maximum of 2 Gln-deamidated residues per protein).

Table 1

Enzymatic Gln deamidations present in stressed primary islets

ProteinPeptideModified residueSequenceaDonorb
CHGA 78–88 E80 ELEDLALQGAK 3, 4 
CTSD 314–331 E322 AIGAVPLIEGEYMIPCEK 5, 6, 7 
GRP78 448–464 E449 SEIFSTASDNQPTVTIK 
GRP78 620–633 E628 KKELEEIVEPIISK 
HNRNPA2B1 191–200 E194 QEMEEVQSSR 
HNRNPH1 17–29 E28 GLPWSCSADEVE
HNRNPK 104–139 E122 IIPTLEEGLQLPSPTATSELPLESDAVECLNYQHYK 3, 5 
HNRNPU 424–433 E426 NGEDLGVAFK 
HSPB1 28–37 E31 LFDEAFGLPR 2, 6 
HSPB1 172–184 E175 LATESNEITIPVTFESR 
HYOU1 901–908 E903 EVEYLLNK 
P4HB 197–207 E197 YELDKDGVVLFK 
PCSK1 655–669 E665 RDELEEGAPSEAMLR 4, 6, 7 
PDIA3 472–482 E481 ELSDFISYLE2, 4, 6, 7 
SCG2 491–510 E502 QMAYENLNDKDEELGEYLAR 
YWHAZ 140–157 E144 E146 GIVDESEQAYQEAFEISK 
ProteinPeptideModified residueSequenceaDonorb
CHGA 78–88 E80 ELEDLALQGAK 3, 4 
CTSD 314–331 E322 AIGAVPLIEGEYMIPCEK 5, 6, 7 
GRP78 448–464 E449 SEIFSTASDNQPTVTIK 
GRP78 620–633 E628 KKELEEIVEPIISK 
HNRNPA2B1 191–200 E194 QEMEEVQSSR 
HNRNPH1 17–29 E28 GLPWSCSADEVE
HNRNPK 104–139 E122 IIPTLEEGLQLPSPTATSELPLESDAVECLNYQHYK 3, 5 
HNRNPU 424–433 E426 NGEDLGVAFK 
HSPB1 28–37 E31 LFDEAFGLPR 2, 6 
HSPB1 172–184 E175 LATESNEITIPVTFESR 
HYOU1 901–908 E903 EVEYLLNK 
P4HB 197–207 E197 YELDKDGVVLFK 
PCSK1 655–669 E665 RDELEEGAPSEAMLR 4, 6, 7 
PDIA3 472–482 E481 ELSDFISYLE2, 4, 6, 7 
SCG2 491–510 E502 QMAYENLNDKDEELGEYLAR 
YWHAZ 140–157 E144 E146 GIVDESEQAYQEAFEISK 

CHGA, chromogranin A; CTSD, cathepsin D; HNRNPA2B1, heterogeneous nuclear ribonucleoproteins A2/B1; HNRNPH1, heterogeneous nuclear ribonucleoprotein H; HNRNPK, heterogeneous nuclear ribonucleoprotein K; HNRNPU, heterogeneous nuclear ribonucleoprotein U; HYOU1, hypoxia upregulated protein 1; PCSK1, neuroendocrine convertase 1; PDIA3, protein disulfide-isomerase A3; SCG2, secretogranin-2.

a

Modified residues underlined and in boldface.

b

Donors 1–4 cytokine mix; donors 5–8 IFN-α.

Table 2

Nonenzymatic Asn deamidations present in stressed primary islets

ProteinPeptideModified residueSequenceaDonorb
GRP78 47–60 D47 DGRVEIIANDQGNR 1, 2, 3, 4, 5, 7 
GRP78 82–96 D82 DQLTSNPENTVFDAK 3, 4, 6 
GRP78 82–96 D87/D90 NQLTSDPEDTVFDAK 1, 2, 3, 7, 8 
GRP78 167–181 D177 VTHAVVTPAYDDAQR 1, 2, 3, 4, 6, 7, 8 
GRP78 325–336 D331 AKFEELDMDLFR 2, 3 
GRP78 377–386 D380 EFFDGKEPSR 1, 2, 3, 4, 6, 8 
GRP78 448–464 D457 SQIFSTASDDQPTVTIK 2, 4 
GRP78 524–532 D528 ITITDDQNR 2, 3, 4, 6 
GRP78 563–573 D563 DELESYAYSLK 2, 3, 7, 8 
GRP94 44–67 D62 TDDEVVQREEEAIQLDGLDASQIR 
GRP94 88–95 D91 LIIDSLYK 
GRP94 103–116 D107 ELISDASDALDKIR 
GRP94 117–135 D124 LISLTDEDALSGNEELTVK 
GRP94 329–348 D337 TVWDWELMDDIKPIWQRPSK 1, 4 
GRP94 609–623 D620 EAVEKEFEPLLDWMK 3, 6, 7 
GRP94 672–683 D676 DISTDYYASQKK 1, 2 
HSPA9 108–121 D114 QAVTNPDNTFYATK 1, 2, 3, 4, 5 
HSPA9 147–159 D149 ASDGDAWVEAHGK 2, 3, 4, 6, 8 
HSPA9 189–202 D198 NAVITVPAYDDSQR 3, 8 
PDIA4 516–527 D522/D523 SQPVPKDDGPVK 
PDIA4 560–570 D566 QLEPVYDSLAK 1, 4 
SCG2 92–112 D93 EDGDESHLPERDSLSEEDWMR 
SCG2 371–381 D376 TGEKPDGSVEPER 1, 2, 3, 4, 7 
SCG2 450–461 D458 TSYFPNPYDQEK 
SCG2 491–510 D496/D498 QMAYEDLDDKDQELGEYLAR 2, 3 
ProteinPeptideModified residueSequenceaDonorb
GRP78 47–60 D47 DGRVEIIANDQGNR 1, 2, 3, 4, 5, 7 
GRP78 82–96 D82 DQLTSNPENTVFDAK 3, 4, 6 
GRP78 82–96 D87/D90 NQLTSDPEDTVFDAK 1, 2, 3, 7, 8 
GRP78 167–181 D177 VTHAVVTPAYDDAQR 1, 2, 3, 4, 6, 7, 8 
GRP78 325–336 D331 AKFEELDMDLFR 2, 3 
GRP78 377–386 D380 EFFDGKEPSR 1, 2, 3, 4, 6, 8 
GRP78 448–464 D457 SQIFSTASDDQPTVTIK 2, 4 
GRP78 524–532 D528 ITITDDQNR 2, 3, 4, 6 
GRP78 563–573 D563 DELESYAYSLK 2, 3, 7, 8 
GRP94 44–67 D62 TDDEVVQREEEAIQLDGLDASQIR 
GRP94 88–95 D91 LIIDSLYK 
GRP94 103–116 D107 ELISDASDALDKIR 
GRP94 117–135 D124 LISLTDEDALSGNEELTVK 
GRP94 329–348 D337 TVWDWELMDDIKPIWQRPSK 1, 4 
GRP94 609–623 D620 EAVEKEFEPLLDWMK 3, 6, 7 
GRP94 672–683 D676 DISTDYYASQKK 1, 2 
HSPA9 108–121 D114 QAVTNPDNTFYATK 1, 2, 3, 4, 5 
HSPA9 147–159 D149 ASDGDAWVEAHGK 2, 3, 4, 6, 8 
HSPA9 189–202 D198 NAVITVPAYDDSQR 3, 8 
PDIA4 516–527 D522/D523 SQPVPKDDGPVK 
PDIA4 560–570 D566 QLEPVYDSLAK 1, 4 
SCG2 92–112 D93 EDGDESHLPERDSLSEEDWMR 
SCG2 371–381 D376 TGEKPDGSVEPER 1, 2, 3, 4, 7 
SCG2 450–461 D458 TSYFPNPYDQEK 
SCG2 491–510 D496/D498 QMAYEDLDDKDQELGEYLAR 2, 3 

PDIA4, protein disulfide-isomerase A4; SCG2, secretogranin-2.

a

Modified residues underlined and in boldface.

b

Donors 1–D cytokine mix; donors 5–8 IFN-α.

Deamidated Peptides Are Presented by HLA-DRB1*04:01

A previously developed algorithm (25,26) was used to predict binding of deamidated peptide sequences to HLA-DRB1*04:01. Peptides with predicted binding (Supplementary Table 6) were synthesized and their binding affinity measured. Five peptides with enzymatic deamidations and four with nonenzymatic deamidations had detectable binding to DRB1*04:01 (Table 3). Of the five Gln-deamidated peptides, two peptides, including the 78-kDa glucose-regulated protein (GRP78) 445–459 with deamidation at residue 449 (GRP78 445–459 E449) and prolyl-4-hydroxylase β (P4HB) 193–207 with deamidation at residue 197 (P4HB 193–207 E197), bound with high affinity (IC50 <10 μmol/L). One peptide, P4HB 188–202 with deamidation at residue 197 (the same residue as the other P4HB peptide) (P4HB 188–202 E197) had an intermediate binding affinity. Two peptides bound weakly (IC50 >30 μmol/L), including HSP β-1 (HSPB1) 166–180 with deamidation at position 175 (HSPB1 166–180 E175) and 14-3-3 protein ζ/δ (YWHAZ) 138–152 with deamidations at positions 144 and 146 (YWHAZ 138–152 E144E146). Of the four Asn-deamidated peptides, three bound with high affinity: GRP78 547–566 with deamidation at residue 457 (GRP78 447–466 D457), GRP94 114–133 with deamidation at residue 124 (GRP94 114–133 D124), and HSPA9 139–158 with deamidation at residue 149 (HSPA9 139–158 D149). One peptide, GRP94 328–347 with deamidation at residue 337 (GRP94 328–347 D337), had an intermediate binding affinity.

Table 3

Motif analysis for Gln- and Asn-deamidated DRB1*04:01 sequences

ProteinDeamidationPeptideNameSequencea,bIC50 (μmol/L)c
GRP78 Gln 445–459 GRP78 445–459 E449 TKKSEIFSTASDNQP 1.48 
HSPB1 Gln 166–180 HSPB1 166–180 E175 EAPMPKLATESNEIT 41.81 
P4HB Gln 188–202 P4HB 188–202 E197 SNSDVFSKYELDKD19.51 
P4HB Gln 193–207 P4HB 193–207 E197 FSKYELDKDGVVLFK 4.39 
YWHAZ Gln 138–152 YWHAZ 138–152 E144E146 KKGIVDESEQAYQEA 30.22 
GRP78 Asn 547–566 GRP78 447–466 D457 KSQIFSTASDDQPTVTIKVY 10.01 
GRP94 Asn 114–133 GRP94 114–133 D124 KIRLISLTDEDALSGNEELT 9.11 
GRP94 Asn 328–347 GRP94 328–347 D337 KTVWDWELMDDIKPIWQRPS 28.02 
HSPA9 Asn 139–158 HSPA9 139–158 D149 NVPFKIVRASDGDAWVEAHG 3.35 
ProteinDeamidationPeptideNameSequencea,bIC50 (μmol/L)c
GRP78 Gln 445–459 GRP78 445–459 E449 TKKSEIFSTASDNQP 1.48 
HSPB1 Gln 166–180 HSPB1 166–180 E175 EAPMPKLATESNEIT 41.81 
P4HB Gln 188–202 P4HB 188–202 E197 SNSDVFSKYELDKD19.51 
P4HB Gln 193–207 P4HB 193–207 E197 FSKYELDKDGVVLFK 4.39 
YWHAZ Gln 138–152 YWHAZ 138–152 E144E146 KKGIVDESEQAYQEA 30.22 
GRP78 Asn 547–566 GRP78 447–466 D457 KSQIFSTASDDQPTVTIKVY 10.01 
GRP94 Asn 114–133 GRP94 114–133 D124 KIRLISLTDEDALSGNEELT 9.11 
GRP94 Asn 328–347 GRP94 328–347 D337 KTVWDWELMDDIKPIWQRPS 28.02 
HSPA9 Asn 139–158 HSPA9 139–158 D149 NVPFKIVRASDGDAWVEAHG 3.35 
a

Modified residues shown in boldface.

b

Predicted minimal motifs underlined.

c

A lower IC50 indicates better binding.

T Cells Recognizing Deamidated Peptides Are Present in Individuals With Type 1 Diabetes

Immunogenicity of the deamidated peptides was assessed by stimulating PBMCs from individuals with type 1 diabetes (summarized in Supplementary Table 2) in vitro with peptides for 14 days, followed by staining with DRB1*04:01 tetramers. The percentage of CD4+Tmr+ cells was compared with background (a control peptide with measurable DRB1*04:01 binding but no immunogenicity). We observed a significant increase in CD4+Tmr+ cells compared with background for six deamidated peptides and trends toward a higher percentage for three others (Fig. 2A). Representative staining results for each deamidated peptide are depicted in Fig. 2B. Responses were considered positive when the percentage of CD4+Tmr+ cells was >0.10% and at least threefold above background tetramer staining (CD4Tmr+). Notably, all deamidated peptides with high or intermediate HLA binding elicited positive in vitro T-cell responses except for P4HB 193–207 E197. The two deamidated peptides with weak binding did not generate detectable responses in our assays. Responses against one individual without diabetes are shown in Supplementary Fig. 2.

Figure 2

CD4+ T cells responding against deamidated peptides are present in in vitro cultures of PBMCs of individuals with type 1 diabetes. A: Percentage of CD4+Tmr+ cells of each deamidated peptide with HLA-DRB1*04:01 binding and background staining, i.e., staining with a tetramer that is not an epitope. B: One representative tetramer staining image for each deamidated peptide and a negative (Neg) background staining. Clear positives (>0.10 and threefold above background) are underlined. Data are mean ± SEM (A). Each dot represents one donor. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001 by paired t test.

Figure 2

CD4+ T cells responding against deamidated peptides are present in in vitro cultures of PBMCs of individuals with type 1 diabetes. A: Percentage of CD4+Tmr+ cells of each deamidated peptide with HLA-DRB1*04:01 binding and background staining, i.e., staining with a tetramer that is not an epitope. B: One representative tetramer staining image for each deamidated peptide and a negative (Neg) background staining. Clear positives (>0.10 and threefold above background) are underlined. Data are mean ± SEM (A). Each dot represents one donor. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001 by paired t test.

Close modal

T Cells From Individuals With Type 1 Diabetes Preferentially Recognize Deamidated Peptides

To confirm that the above-described responses were bona fide, we sorted and expanded Tmr+CD4+ cells to obtain T-cell clones. Clones specific for P4HB 193–207 E197, GRP78 447–466 D457, GRP94 114–133 D124, and HSPA9 139–158 D149 were successfully isolated. Each clone stained as a single population of T cells with a mean fluorescence intensity above background (Supplementary Fig. 3). Because unmodified versions of several deamidated peptides of interest also had detectable binding to DRB1*04:01, we assessed whether T-cell clones preferentially responded to deamidated peptide by activating each clone in vitro using APCs pulsed with either native or deamidated peptide and assessing their proliferation through thymidine incorporation. In each case, the T-cell clone proliferated more robustly in response to deamidated peptide than the corresponding native peptide (Fig. 3). Specifically, respective proliferation was 23-, 45-, 26-, and 4-fold higher in response to the deamidated vs. native peptide. To clarify whether these observed differences were caused by altered HLA binding, we compared binding of each deamidated and native peptide to DRB1*04:01 (Supplementary Table 7). Deamidation only improved binding for GRP94 328–347, for which the native peptide had no detectable binding. This suggests WDWELMDIK as the minimal binding motif, placing the deamidated residue in P7. For P4HB 193–207, the deamidated residue is predicted to occupy P2, a T-cell receptor contact. For GRP78 547–566, GRP94 114–133, and HSPA9 139–158, deamidation negatively affects binding but is required for recognition. This implies conformational alterations of the bound peptide, leading to altered recognition. In any case, preferred recognition of the deamidated peptides establishes these peptides as bona fide neoepitopes.

Figure 3

Isolated T-cell clones have a preference for their cognate deamidated peptide. AD: Proliferation of T-cell clones reactive against P4HB 193–207 E197, GRP78 447–466 D457, GRP94 114–133 D124, and HSPA9 139–158 D149 in response to nonpulsed APCs, APCs pulsed with native peptide, or deamidated peptide (10 μg/mL). Data are stimulation index values calculated by normalizing the proliferation of each clone based on 3H-thymidine incorporation of the control nonpulsed APC-stimulated wells. P4HB 193–207 E197–specific clones did not proliferate (stimulation index <3) in response to APCs pulsed with native peptide. All peptides proliferated (stimulation index >3) in response to APCs pulsed with deamidated peptide. Data are mean ± SEM (technical error bars). Ag, antigen.

Figure 3

Isolated T-cell clones have a preference for their cognate deamidated peptide. AD: Proliferation of T-cell clones reactive against P4HB 193–207 E197, GRP78 447–466 D457, GRP94 114–133 D124, and HSPA9 139–158 D149 in response to nonpulsed APCs, APCs pulsed with native peptide, or deamidated peptide (10 μg/mL). Data are stimulation index values calculated by normalizing the proliferation of each clone based on 3H-thymidine incorporation of the control nonpulsed APC-stimulated wells. P4HB 193–207 E197–specific clones did not proliferate (stimulation index <3) in response to APCs pulsed with native peptide. All peptides proliferated (stimulation index >3) in response to APCs pulsed with deamidated peptide. Data are mean ± SEM (technical error bars). Ag, antigen.

Close modal

Deamidated Peptide-Specific T Cells Have Elevated Frequencies in Individuals With Type 1 Diabetes and Are Present in Islets

We then selected a set of high-interest epitopes based on positivity after in vitro stimulation with peptide and/or the isolation of clones, representing both Gln and Asn-deamidated peptides. Corresponding HLA class II tetramers were used to assess the frequency and phenotype of CD4+ T cells specific for these novel deamidated peptides in PBMCs from healthy control individuals and individuals with type 1 diabetes (representative results are shown in Supplementary Fig. 4, positive and negative control individuals in Supplementary Fig. 5, and donor characteristics in Supplementary Table 3). Previous studies set the limit of detection for tetramer staining as 0.5–1 cell per million (36). Detectable frequencies were observed for GRP94 114–133 D124 and HSPA9 139–158 D149, but there was no significant difference between control individuals and individuals with type 1 diabetes (Fig. 4A and B). In contrast, significantly higher frequencies were observed in individuals with type 1 diabetes compared with control individuals for P4HB 193–207 E197, GRP78 447–466 D457, and GRP94 328–347 D337 (Fig. 4A and B). As illustrated by heat map visualization, diverse patterns of T-cell frequency were observed in our cohort. Donors 26 and 27 had high frequencies for GRP94 328–347 D337 but low frequencies for other epitopes. In contrast, donors 35 and 36 had comparatively high frequencies for all deamidated epitopes (Fig. 4C). We observed no significant differences in T-cell phenotype between control individuals and individuals with type 1 diabetes (Supplementary Fig. 6) probably because very few control individuals had adequate frequencies (five or more tetramer-positive cells per million CD4+ T cells) to permit phenotypic analysis. Next, to investigate whether the deamidated peptides are present in islets of individuals with diabetes at sufficient concentrations to elicit T-cell responses, cell lines were generated from T cells infiltrating the islets of nPOD donor 6551, and IFN-γ production upon exposure to deamidated peptide was measured. Significant IFN-γ production above background (a control HIV peptide) was measured in one (P4HB 193–207 E197) or two (all other peptides) out of three generated T-cell lines for each deamidated peptide.

Figure 4

Identification of CD4+ T-cell frequencies specific for novel Gln- and Asn-deamidated peptides in peripheral blood of healthy control individuals and individuals with type 1 diabetes (T1D). A: Total CD4+ T-cell frequencies in the enriched fraction directed against P4HB 193–207 E197, GRP78 447–466 D457, GRP94 114–133 D124, GRP94 328–347 D337, and HSPA9 139–158 D149 in control (CTR) individuals (6) and in individuals with T1D subdivided by short (≤5 year; squares) and long (>5 year; circles) duration of disease. CD4+ T-cell frequencies are considered positive when ≥5 (indicated in red). The dotted line indicates the frequency of negative control YWHAZ 138-152 E144 E146 specific T cells. B: Representative FACS plots depict CD4+ T cells in the enriched fraction directed against P4HB 193–207 E197, GRP78 447–466 D457, GRP94 114–133 D124, GRP94 328–347 D337, and HSPA9 139–158 D149. C: A heat map analysis of the T-cell frequencies shown in (A), indicating patterns of reactivity. D: IFN-γ production of islet T-cell lines exposed to deamidated peptides (normalized to a control HIV peptide). The dotted line indicates a normalized IFN-γ production value of 1. Data are mean ± SEM. Each dot represents one donor or repeat. **P < 0.01 by unpaired t test with Welch correction (A); one-way ANOVA against negative control peptide with correction for multiple testing (D).

Figure 4

Identification of CD4+ T-cell frequencies specific for novel Gln- and Asn-deamidated peptides in peripheral blood of healthy control individuals and individuals with type 1 diabetes (T1D). A: Total CD4+ T-cell frequencies in the enriched fraction directed against P4HB 193–207 E197, GRP78 447–466 D457, GRP94 114–133 D124, GRP94 328–347 D337, and HSPA9 139–158 D149 in control (CTR) individuals (6) and in individuals with T1D subdivided by short (≤5 year; squares) and long (>5 year; circles) duration of disease. CD4+ T-cell frequencies are considered positive when ≥5 (indicated in red). The dotted line indicates the frequency of negative control YWHAZ 138-152 E144 E146 specific T cells. B: Representative FACS plots depict CD4+ T cells in the enriched fraction directed against P4HB 193–207 E197, GRP78 447–466 D457, GRP94 114–133 D124, GRP94 328–347 D337, and HSPA9 139–158 D149. C: A heat map analysis of the T-cell frequencies shown in (A), indicating patterns of reactivity. D: IFN-γ production of islet T-cell lines exposed to deamidated peptides (normalized to a control HIV peptide). The dotted line indicates a normalized IFN-γ production value of 1. Data are mean ± SEM. Each dot represents one donor or repeat. **P < 0.01 by unpaired t test with Welch correction (A); one-way ANOVA against negative control peptide with correction for multiple testing (D).

Close modal

Clinical Characteristics Correlate With Frequencies of Deamidated Peptide–Specific T Cells

We next asked if the frequency of P4HB 193–207 E197–, GRP78 447–466 D457–, and GRP94 328–347 D337–reactive T cells correlates with characteristics such as autoantibody status, age at diagnosis, or disease duration. Individuals positive for insulin autoantibodies (IAAs) at diagnosis (≥0.6% binding) had a trend toward higher frequencies of deamidated peptide–reactive T cells (Fig. 5A). Considering the three deamidated peptides individually, the frequency of T cells responsive against P4HB 193–207 E197 tended to be increased in IAA-positive individuals (Fig. 5B). Because low numbers of individuals were negative for GAD antibody and islet antigen 2 antibodies at diagnosis, we grouped individuals with low (including negative) versus high antibody titers, but no significant differences were observed (Fig. 5A). Zinc transporter 8 antibody levels were not determined and therefore could not be analyzed. A significant negative correlation was observed between Tmr+ T-cell frequencies and age at diagnosis (Fig. 5C). A significant positive correlation was present between Tmr+ T-cell frequencies and disease duration. No correlation was observed between age and Tmr+ T-cell frequencies, suggesting that the correlation between frequencies and disease duration is independent of the generally older age of individuals with longer disease duration (Fig. 5C). Positive individual correlations were also present for P4HB 193–207 E197, GRP78 447–466 D457, and GRP94 328–347 D337, providing extra strength to the observed correlation between disease duration and Tmr+ T-cell frequency (Fig. 5D).

Figure 5

Correlations between Tmr+ CD4+ T-cell frequencies specific for novel Gln- and Asn-deamidated peptides in peripheral blood and insulin antibody levels at disease onset and duration. A: Tmr+ frequencies in individuals negative (<0.6% binding) and positive (≥0.6% binding) for insulin antibodies, low (<52 units/mL) and high (≥52 units/mL) for GAD antibodies, and low (<62 units/mL) and high (≥62 units/mL) for islet antigen 2 (IA2) antibodies. B: Tmr+ frequencies in individuals negative and positive for insulin antibodies for P4HB 193–207 E197, GRP78 447–466 D457, and GRP94 328–347 D337. C: Applying linear regression analysis indicated a significant inverse correlation between the Tmr+ frequency of peptides with significantly higher Tmr+ frequencies in individuals with type 1 diabetes and age at diagnosis, a significant positive correlation between Tmr+ frequency and disease duration, and no correlation between Tmr+ frequency and age. D: Correlations between frequency and disease duration for each peptide individually were also significant. Data are mean ± SEM (A and B). Unpaired t test (A and B) and simple linear regression (C and D) were used. Each dot represents one unique donor. Neg, negative; pos, positive.

Figure 5

Correlations between Tmr+ CD4+ T-cell frequencies specific for novel Gln- and Asn-deamidated peptides in peripheral blood and insulin antibody levels at disease onset and duration. A: Tmr+ frequencies in individuals negative (<0.6% binding) and positive (≥0.6% binding) for insulin antibodies, low (<52 units/mL) and high (≥52 units/mL) for GAD antibodies, and low (<62 units/mL) and high (≥62 units/mL) for islet antigen 2 (IA2) antibodies. B: Tmr+ frequencies in individuals negative and positive for insulin antibodies for P4HB 193–207 E197, GRP78 447–466 D457, and GRP94 328–347 D337. C: Applying linear regression analysis indicated a significant inverse correlation between the Tmr+ frequency of peptides with significantly higher Tmr+ frequencies in individuals with type 1 diabetes and age at diagnosis, a significant positive correlation between Tmr+ frequency and disease duration, and no correlation between Tmr+ frequency and age. D: Correlations between frequency and disease duration for each peptide individually were also significant. Data are mean ± SEM (A and B). Unpaired t test (A and B) and simple linear regression (C and D) were used. Each dot represents one unique donor. Neg, negative; pos, positive.

Close modal

Here, we report on the enzymatic formation of novel Gln-deamidated peptides and nonenzymatic formation of Asn-deamidated peptides in stressed primary human islets. Higher frequencies of CD4+ T cells reactive against deamidated peptides were present in the peripheral blood of individuals with type 1 diabetes compared with control individuals. In particular, nonenzymatically deamidated targets have not been previously investigated as neoepitopes in this disease. Therefore, our findings implicate a new class of relevant targets of autoimmunity.

Protein deamidation was potentiated in pancreatic islets through treatment with either IFN-α or a blend of inflammatory cytokines (IL-1β, TNF-α, and IFN-γ). Notably, these cytokines also influenced the surface expression of HLA molecules. HLA class I genes harbor interferon consensus sequences in their regulatory region, allowing MHC class I molecules to be induced by both IFN-α and IFN-γ. In contrast, HLA class II genes have a motif present in their regulatory region that does not respond to IFN-α, explaining why IFN-γ is a better inducer of MHC class II proteins (37). Correspondingly, we observed a highly significant induction of the MHC class II DR α-chain in cytokine-stimulated (IL-1β, TNF-α, and IFN-γ) islets (6.63-fold, P < 0.001 [data not shown]) and no significant induction of MHC class II proteins in IFN-α–exposed islets. In parallel, we observed a higher increase in TGM2 expression after cytokine exposure compared with IFN-α alone, as well as more deamidated peptides. These data suggest that inflammatory cytokines promote the formation and presentation of deamidated peptides to T cells. However, a notable limitation of our study is that because data were generated using unfractionated islets, deamidation may occur not only in β-cells but also in other cell types present in the islets. IFN-α is induced early in the immune response, providing a priming mechanism that can orchestrate subsequent pathways in innate and adaptive immunity (32). Since IFN-γ is part of a later stage in type 1 diabetes progression (38), deamidation might contribute to epitope spreading and the acceleration of autoimmune attack following initial immune priming by other classes of antigens.

We previously reported that GRP78 (involved in the unfolded protein response in endoplasmic reticulum stress) is an autoantigen in its citrullinated form in both murine and human diabetes (24,39). Autoantibodies against GRP78 are a diagnostic marker in rheumatoid arthritis, a disease that is also linked to HLA-DRB1*04:01 (40). Furthermore, autoimmunity in rheumatoid arthritis has been observed against citrullinated and carbamylated GRP78 (41,42). Therefore, modified versions of GRP78 are involved in the loss of self-tolerance in multiple disease settings. Notably, GRP78 is translocated to the cell surface upon inflammatory cytokine exposure, followed by secretion in the extracellular space, facilitating presentation to the immune system (43). We previously detected enzymatically deamidated GRP78 in murine islets of Langerhans (44). We now provide evidence that GRP78 is both enzymatically (two Gln residues) and nonenzymatically (nine Asn residues) deamidated in human islets. The frequencies of GRP78 447–466 D457–responsive T cells were increased in individuals with type 1 diabetes compared with healthy control individuals, and there was a positive correlation between Tmr+ T-cell frequency and disease duration.

P4HB, a chaperone essential for proper insulin folding, was recently shown to be an antigen in murine models of type 1 diabetes and in humans (45,46). Stressed human islets had amplified levels of carbonylated P4HB, coinciding with altered proinsulin/insulin ratios and decreased glucose-stimulated insulin secretion (GSIS), suggesting a loss of function of P4HB due to carbonylation (45). In our work, the frequencies of deamidated P4HB 193–207 E197–responsive T cells were significantly increased in individuals with type 1 diabetes, and there was a positive correlation between T-cell frequency and disease duration. Deamidation of proteins may alter their structure, stability, or function and contribute to protein degradation. It was recently shown that citrullination, a modification similar to deamidation, of glucokinase altered activity of the enzyme and suppressed GSIS (47). Therefore, in addition to activating autoreactive CD4+ T cells, deamidation of P4HB could contribute to altered proinsulin/insulin ratios and decreased GSIS, aggravating the disease.

In vitro responses toward the nonenzymatically deamidated peptides GRP94 114–133 D124 and HSPA9 139–158 D149 were observed, and clones were successfully isolated. However, T cells recognizing these peptides were not present at higher frequencies in individuals with type 1 diabetes than in healthy control individuals, suggesting that these peptides are a component of a benign autoimmune repertoire (as previously noted for islet-reactive CD8+ T cells) and not uniquely involved in type 1 diabetes pathogenesis (5,6).

Individuals with type 1 diabetes showed increased CD4+ T-cell frequencies directed against GRP94 328–347 D337 and a positive correlation between T-cell frequency and disease duration. GRP94 has not been described as an autoantigen in type 1 diabetes. However, a recent study investigated sequence similarities between GRP94 and known islet antigens, including insulin and islet amyloid polypeptide (48). Interestingly, the region encompassing GRP94 114–133, for which we isolated clones that recognize the deamidated form of the peptide, has high sequence similarity with INS 6–20 (48). This INS signal peptide region is targeted by CD8+ T cells (49) and has also been described as a DRB4-restricted CD4+ T-cell epitope that is detectable among the circulating T cells of individuals with type 1 diabetes (50). Therefore, GRP94 and other HSP peptides with similarity to islet peptides are of potential immunogenic relevance in type 1 diabetes (48), making GRP94 114–133 D124 an interesting candidate for further study.

Overall, we report on the recognition of five deamidated self-peptides, originating from four different HSPs: GRP78, GRP94, P4HB, and HSPA9. Importantly, several of these specificities were also recognized by islet infiltrating T-cell lines from a pancreatic organ donor. Antibodies against HSPs have been detected in various autoimmune diseases, such as systemic lupus erythematosus, rheumatoid arthritis, and type 1 diabetes (51), suggesting an important shared mechanism in human autoimmune disease. Besides the above-mentioned autoantibodies detected against (modified) GRP78 and P4HB in the context of type 1 diabetes, autoantibodies against HSP65 and HSP90 have also been reported (52,53). Here, we add two more HSPs, GRP94 and HSPA9, and a novel modification to the list, further supporting their importance in autoimmunity and type 1 diabetes.

We observed a negative correlation between T-cell frequencies and age at diagnosis, a positive correlation between frequencies and disease duration, and a trend for lower frequencies in individuals negative for IAA. A similar negative relationship between frequencies and age at diagnosis has been reported for hybrid insulin peptide–reactive cells, as well as lower frequencies in two IAA-negative individuals (54). Although a confirmatory study with more individuals is warranted, the observation of lower frequencies of CD4+ T cells reactive against PTM peptides (deamidated peptides and hybrid insulin peptides) in IAA-negative individuals suggests that IAA status at diagnosis might be an indicator of different disease endotypes or pathotypes.

Besides the above-mentioned deamidated peptides, we detected several other deamidated peptides in stressed human islets with LC-MS/MS, including chromogranin-A, neuroendocrine convertase 1, and secretogranin-2. Although none of these were identified as targets of DRB1*04:01-restricted T cells, both HLA-DQ2 and HLA-DQ8 (the HLA-DQ haplotypes most strongly associated with type 1 diabetes) favor presentation of negatively charged peptides (4,9). Consequently, it would be of great interest to investigate the recognition of deamidated targets in the context of HLA-DQ2 and HLA-DQ8.

In summary, we report the detection of novel deamidated peptides in stressed human islets and show that CD4+ T cells responsive against those deamidated peptides are present in individuals with type 1 diabetes. These results demonstrate that deamidation of self-proteins occurs within primary islets under conditions of cytokine stress and that deamidated peptides can promote the activation of CD4+ T cells. These findings add to the growing list of evidence that PTMs undermine tolerance in type 1 diabetes and may open the road for the development of new diagnostic or therapeutic applications for patients.

Acknowledgments. The authors thank Martine Gilis and Eline Desager (KU Leuven) for expert assistance and the SyBioMa Mass Spectrometry Facility of KU Leuven. The authors also acknowledge Hilde Morobé (UZ Leuven) for patient recruitment and collection of blood samples and thank all the patients and control individuals who donated blood samples. The authors thank the Benaroya Research Institute Flow Cytometry Core Facility for assistance with flow cytometry.

Funding. This work was supported by Innovative Medicines Initiative 2 Joint Undertaking grant 115797 (INNODIA) and 948268 (INNODIA HARVEST). This joint undertaking received support from the European Union’s Horizon 2020 research and innovation program and through the European Federation of Pharmaceutical Industries and Associations (EFPIA), JDRF, and The Leona M. and Harry B. Helmsley Charitable Trust. This research was performed with the support of nPOD (Research Resource Identifier SCR_014641), a collaborative type 1 diabetes research project supported by JDRF grant 5-SRA-2018-557-Q-R and The Leona M. and Harry B. Helmsley Charitable Trust grants 2018PG-T1D053 and G-2108-04793. The work was funded by the KU Leuven grant C16/18/006), Flemish Research Foundation predoctoral fellowship 3-PDF-2018-593-A-N (to A.C.), EFPIA postdoctoral fellowship 1189518N (to M.B.), JDRF grant 2-SRA-2018-551-S-B (to E.A.J.), and National Institute of Diabetes and Digestive and Kidney Diseases grants U01 DK104218 and UC4 DK116284.

The content and views expressed are the responsibility of the authors and do not necessarily reflect the official view of nPOD. Organ Procurement Organizations partnering with nPOD to provide research resources are listed at https://npod.org/for-partners/npod-partners/.

Duality of Interest. No potential conflicts of interest relevant to this article were reported.

Author Contributions. A.C., P.G., R.D., M.B., F.M.C.S., S.V., and E.A.J. contributed to the design, conduct, analysis, and interpretation of the data. A.M. and S.C.K. conducted the islet T-cell line experiments. X.Y. and D.L.E. conducted the RNA sequencing data analysis. M.S. and P.M. collected human islets. C.M., E.A.J., and L.O. helped design experiments. All authors contributed to the writing of the manuscript and approved the final version. C.M., E.A.J., and L.O. are the guarantors of this work and, as such, had full access to all the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis.

Prior Presentation. Parts of this study were presented in abstract form at the 19th Immunology of Diabetes Society Congress, Paris, France, 23–27 May 2023. An earlier version of the work appears as a chapter in A.C.’s doctoral dissertation at KU Leuven.

This article contains supplementary material online at https://doi.org/10.2337/figshare.25253794.

E.A.J. and L.O. share senior authorship.

1.
Zheng
GXY
,
Terry
JM
,
Belgrader
P
, et al
.
Massively parallel digital transcriptional profiling of single cells
.
Nat Commun
2017
;
8
:
14049
2.
van de Wal
Y
,
Kooy
Y
,
van Veelen
P
, et al
.
Selective deamidation by tissue transglutaminase strongly enhances gliadin-specific T cell reactivity
.
J Immunol
1998
;
161
:
1585
1588
3.
Noble
JA
,
Valdes
AM.
Genetics of the HLA region in the prediction of type 1 diabetes
.
Curr Diab Rep
2011
;
11
:
533
542
4.
McGinty
JW
,
Marré
ML
,
Bajzik
V
,
Piganelli
JD
,
James
EA.
T cell epitopes and post-translationally modified epitopes in type 1 diabetes
.
Curr Diab Rep
2015
;
15
:
90
5.
Culina
S
,
Lalanne
AI
,
Afonso
G
, et al;
ImMaDiab Study Group
.
Islet-reactive CD8+ T cell frequencies in the pancreas, but not in blood, distinguish type 1 diabetic patients from healthy donors
.
Sci Immunol
2018
;
3
:
eaao4013
6.
Gonzalez-Duque
S
,
Azoury
ME
,
Colli
ML
, et al
.
Conventional and neo-antigenic peptides presented by β cells are targeted by circulating naïve CD8+ T cells in type 1 diabetic and healthy donors
.
Cell Metab
2018
;
28
:
946
960.e6
7.
Danke
NA
,
Koelle
DM
,
Yee
C
,
Beheray
S
,
Kwok
WW.
Autoreactive T cells in healthy individuals
.
J Immunol
2004
;
172
:
5967
5972
8.
Cecilio
LA
,
Bonatto
MW.
The prevalence of HLA DQ2 and DQ8 in patients with celiac disease, in family and in general population
.
Arq Bras Cir Dig
2015
;
28
:
183
185
9.
Klöck
C
,
Diraimondo
TR
,
Khosla
C.
Role of transglutaminase 2 in celiac disease pathogenesis
.
Semin Immunopathol
2012
;
34
:
513
522
10.
van Lummel
M
,
Duinkerken
G
,
van Veelen
PA
, et al
.
Posttranslational modification of HLA-DQ binding islet autoantigens in type 1 diabetes
.
Diabetes
2014
;
63
:
237
247
11.
Foster
A
,
Bhattacharjee
P
,
Tresoldi
E
,
Pakusch
M
,
Cameron
FJ
,
Mannering
SI.
Glutamine deamidation does not increase the immunogenicity of C-peptide in people with type 1 diabetes
.
J Transl Autoimmun
2022
;
6
:
100180
12.
McGinty
JW
,
Chow
IT
,
Greenbaum
C
,
Odegard
J
,
Kwok
WW
,
James
EA.
Recognition of posttranslationally modified GAD65 epitopes in subjects with type 1 diabetes
.
Diabetes
2014
;
63
:
3033
3040
13.
Jing
Y
,
Kong
Y
,
McGinty
J
, et al
.
T-cell receptor/HLA humanized mice reveal reduced tolerance and increased immunogenicity of posttranslationally modified GAD65 epitope
.
Diabetes
2022
;
71
:
1012
1022
14.
DeVoss
J
,
Hou
Y
,
Johannes
K
, et al
.
Spontaneous autoimmunity prevented by thymic expression of a single self-antigen [published correction appears in J Exp Med 2007;204:203]
.
J Exp Med
2006
;
203
:
2727
2735
15.
Callebaut
A
,
Derua
R
,
Vig
S
,
Delong
T
,
Mathieu
C
,
Overbergh
L.
Identification of deamidated peptides in cytokine-exposed MIN6 cells through LC-MS/MS using a shortened digestion time and inspection of MS2 spectra
.
J Proteome Res
2021
;
20
:
1405
1414
16.
McLaughlin
RJ
,
de Haan
A
,
Zaldumbide
A
, et al
.
Human islets and dendritic cells generate post-translationally modified islet autoantigens
.
Clin Exp Immunol
2016
;
185
:
133
140
17.
Erlich
H
,
Valdes
AM
,
Noble
J
, et al;
Type 1 Diabetes Genetics Consortium
.
HLA DR-DQ haplotypes and genotypes and type 1 diabetes risk: analysis of the type 1 diabetes genetics consortium families
.
Diabetes
2008
;
57
:
1084
1092
18.
Young
MD
,
Behjati
S.
SoupX removes ambient RNA contamination from droplet-based single-cell RNA sequencing data
.
Gigascience
2020
;
9
:
giaa151
19.
Hao
Y
,
Stuart
T
,
Kowalski
MH
, et al
.
Dictionary learning for integrative, multimodal and scalable single-cell analysis
.
Nat Biotechnol
2024
;
42
:
293
304
20.
Germain
P-L
,
Lun
A
,
Garcia Meixide
C
,
Macnair
W
,
Robinson
MD.
Doublet identification in single-cell sequencing data using scDblFinder
.
F1000 Res
2021
;
10
:
979
21.
Guo
H
,
Li
J.
scSorter: assigning cells to known cell types according to marker genes
.
Genome Biol
2021
;
22
:
69
22.
Coomans De Brachène
A
,
Alvelos
MI
,
Szymczak
F
, et al
. Interferons are the key cytokines acting on pancreatic islets in type 1 diabetes. 1 July
2023
[preprint]. bioRxiv:2023.06.29.547000
23.
Love
MI
,
Huber
W
,
Anders
S.
Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2
.
Genome Biol
2014
;
15
:
550
24.
Buitinga
M
,
Callebaut
A
,
Marques Câmara Sodré
F
, et al
.
Inflammation-induced citrullinated glucose-regulated protein 78 elicits immune responses in human type 1 diabetes
.
Diabetes
2018
;
67
:
2337
2348
25.
James
EA
,
Moustakas
AK
,
Bui
J
, et al
.
HLA-DR1001 presents “altered-self” peptides derived from joint-associated proteins by accepting citrulline in three of its binding pockets
.
Arthritis Rheum
2010
;
62
:
2909
2918
26.
James
EA
,
Rieck
M
,
Pieper
J
, et al
.
Citrulline-specific Th1 cells are increased in rheumatoid arthritis and their frequency is influenced by disease duration and therapy
.
Arthritis Rheumatol
2014
;
66
:
1712
1722
27.
Ettinger
RA
,
Papadopoulos
GK
,
Moustakas
AK
,
Nepom
GT
,
Kwok
WW.
Allelic variation in key peptide-binding pockets discriminates between closely related diabetes-protective and diabetes-susceptible HLA-DQB1*06 alleles
.
J Immunol
2006
;
176
:
1988
1998
28.
Novak
EJ
,
Liu
AW
,
Nepom
GT
,
Kwok
WW.
MHC class II tetramers identify peptide-specific human CD4(+) T cells proliferating in response to influenza A antigen
.
J Clin Invest
1999
;
104
:
R63
R67
29.
Babon
JAB
,
DeNicola
ME
,
Blodgett
DM
, et al
.
Analysis of self-antigen specificity of islet-infiltrating T cells from human donors with type 1 diabetes
.
Nat Med
2016
;
22
:
1482
1487
30.
Delong
T
,
Wiles
TA
,
Baker
RL
, et al
.
Pathogenic CD4 T cells in type 1 diabetes recognize epitopes formed by peptide fusion
.
Science
2016
;
351
:
711
714
31.
Perez-Riverol
Y
,
Bai
J
,
Bandla
C
, et al
.
The PRIDE database resources in 2022: a hub for mass spectrometry-based proteomics evidences
.
Nucleic Acids Res
2022
;
50
(
D1
):
D543
D552
32.
Lombardi
A
,
Tsomos
E
,
Hammerstad
SS
,
Tomer
Y.
Interferon alpha: the key trigger of type 1 diabetes
.
J Autoimmun
2018
;
94
:
7
15
33.
Marroqui
L
,
Dos Santos
RS
,
Op de Beeck
A
, et al
.
Interferon-α mediates human beta cell HLA class I overexpression, endoplasmic reticulum stress and apoptosis, three hallmarks of early human type 1 diabetes
.
Diabetologia
2017
;
60
:
656
667
34.
Coomans de Brachène
A
,
Dos Santos
RS
,
Marroqui
L
, et al
.
IFN-α induces a preferential long-lasting expression of MHC class I in human pancreatic beta cells
.
Diabetologia
2018
;
61
:
636
640
35.
Szymczak
F
,
Colli
ML
,
Mamula
MJ
,
Evans-Molina
C
,
Eizirik
DL.
Gene expression signatures of target tissues in type 1 diabetes, lupus erythematosus, multiple sclerosis, and rheumatoid arthritis
.
Sci Adv
2021
;
7
:
eabd7600
36.
Kwok
WW
,
Tan
V
,
Gillette
L
, et al
.
Frequency of epitope-specific naive CD4(+) T cells correlates with immunodominance in the human memory repertoire
.
J Immunol
2012
;
188
:
2537
2544
37.
Muegge
K
,
Durum
SK.
Cytokines and transcription factors
.
Cytokine
1990
;
2
:
1
8
38.
Atkinson
MA
,
Bluestone
JA
,
Eisenbarth
GS
, et al
.
How does type 1 diabetes develop?: the notion of homicide or β-cell suicide revisited
.
Diabetes
2011
;
60
:
1370
1379
39.
Rondas
D
,
Crèvecoeur
I
,
D’Hertog
W
, et al
.
Citrullinated glucose-regulated protein 78 is an autoantigen in type 1 diabetes
.
Diabetes
2015
;
64
:
573
586
40.
Bläss
S
,
Union
A
,
Raymackers
J
, et al
.
The stress protein BiP is overexpressed and is a major B and T cell target in rheumatoid arthritis
.
Arthritis Rheum
2001
;
44
:
761
771
41.
Shoda
H
,
Fujio
K
,
Shibuya
M
, et al
.
Detection of autoantibodies to citrullinated BiP in rheumatoid arthritis patients and pro-inflammatory role of citrullinated BiP in collagen-induced arthritis
.
Arthritis Res Ther
2011
;
13
:
R191
42.
Yu
HC
,
Lai
PH
,
Lai
NS
,
Huang
HB
,
Koo
M
,
Lu
MC.
Increased serum levels of anti-carbamylated 78-kDa glucose-regulated protein antibody in patients with rheumatoid arthritis
.
Int J Mol Sci
2016
;
17
:
1510
43.
Vig
S
,
Buitinga
M
,
Rondas
D
, et al
.
Cytokine-induced translocation of GRP78 to the plasma membrane triggers a pro-apoptotic feedback loop in pancreatic beta cells
.
Cell Death Dis
2019
;
10
:
309
44.
Callebaut
A
,
Bruggeman
Y
,
Zamit
C
, et al
.
Aberrant expression of transglutaminase 2 in pancreas and thymus of NOD mice underscores the importance of deamidation in neoantigen generation
.
Front Endocrinol (Lausanne)
2022
;
13
:
908248
45.
Yang
ML
,
Connolly
SE
,
Gee
RJ
, et al
.
Carbonyl posttranslational modification associated with early-onset type 1 diabetes autoimmunity
.
Diabetes
2022
;
71
:
1979
1993
46.
Yang
ML
,
Doyle
HA
,
Clarke
SG
,
Herold
KC
,
Mamula
MJ.
Oxidative modifications in tissue pathology and autoimmune disease
.
Antioxid Redox Signal
2018
;
29
:
1415
1431
47.
Yang
M-L
,
Horstman
S
,
Gee
R
, et al
.
Citrullination of glucokinase is linked to autoimmune diabetes
.
Nat Commun
2022
;
13
:
1870
48.
Finotti
P
,
Pagetta
A.
A mutant α1antitrypsin in complex with heat shock proteins as the primary antigen in type 1 diabetes in silico investigation
.
Sci Rep
2021
;
11
:
3002
49.
Kronenberg-Versteeg
D
,
Eichmann
M
,
Russell
MA
, et al
.
Molecular pathways for immune recognition of preproinsulin signal peptide in type 1 diabetes
.
Diabetes
2018
;
67
:
687
696
50.
James
EA
,
Gillette
L
,
Durinovic-Bello
I
, et al
.
DRB4*01:01 has a distinct motif and presents a proinsulin epitope that is recognized in subjects with type 1 diabetes
.
J Immunol
2018
;
201
:
3524
3533
51.
Wu
T
,
Tanguay
RM.
Antibodies against heat shock proteins in environmental stresses and diseases: friend or foe
?
Cell Stress Chaperones
2006
;
11
:
1
12
52.
Weitgasser
R
,
Lechleitner
M
,
Koch
T
, et al
.
Antibodies to heat-shock protein 65 and neopterin levels in patients with type 1 diabetes mellitus
.
Exp Clin Endocrinol Diabetes
2003
;
111
:
127
131
53.
Qin
HY
,
Mahon
JL
,
Atkinson
MA
,
Chaturvedi
P
,
Lee-Chan
E
,
Singh
B.
Type 1 diabetes alters anti-hsp90 autoantibody isotype
.
J Autoimmun
2003
;
20
:
237
245
54.
Arribas-Layton
D
,
Guyer
P
,
Delong
T
, et al
.
Hybrid insulin peptides are recognized by human T cells in the context of DRB1*04:01
.
Diabetes
2020
;
69
:
1492
1502
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. More information is available at https://www.diabetesjournals.org/journals/pages/license.