Key requirements in type 1 diabetes (T1D) are in setting up new assays as diagnostic biomarkers that will apply to prediabetes, likely T-cell assays, and in designing antigen-specific therapies to prevent T1D development. New preclinical models of T1D will be required to help with advancing both aims. By crossing mouse strains that lack either murine MHC class I and class II genes and insulin genes, we developed YES mice that instead express human HLA-A*02:01, HLA-DQ8, and insulin genes as transgenes. The metabolic and immune phenotype of YES mice is basically identical to that of the parental strains. YES mice remain insulitis and diabetes free up to 1 year of follow-up, maintain normoglycemia to an intraperitoneal glucose challenge in the long-term range, have a normal β-cell mass, and show normal immune responses to conventional antigens. This new model has been designed to evaluate adaptive immune responses to human insulin on a genetic background that recapitulates a human high-susceptibility HLA-DQ8 genetic background. Although insulitis free, YES mice develop T1D when challenged with polyinosinic-polycytidylic acid. They allow the characterization of preproinsulin epitopes recognized by CD8+ and CD4+ T cells upon immunization against human preproinsulin or during diabetes development.

The NOD mouse has been instrumental in deciphering mechanisms involved in type 1 diabetes (T1D), but it fails to cover clinical T1D heterogeneity and shows phenotypic differences with human T1D and limitations when developing T-cell assays or therapies to be applied to humans (1). Immunosuppression has shown not only efficacy in preserving β-cells in patients with recent-onset T1D but also side effects precluding its long-term use (2). Strategies to restore immune tolerance to β-cells should thus be prioritized (3) as preserving responses to unrelated antigens. We lack rodent models to evaluate antigen-specific immunotherapy that directly applies to humans as well as rodent models to explore environmental factors involved in triggering diabetes on conventional genetic backgrounds.

We designed a mouse that expresses both a human β-cell autoantigen and a human antigen-presenting module to characterize autoantigen-derived epitopes that translate to humans. On the basis of evidence that insulin is a key autoantigen, we introgressed a human insulin (hINS) transgene in our model. Indeed, insulin is targeted by autoreactive T cells in T1D (4,5). Anti-insulin autoantibodies are the first to be detected in children at risk and carry a high positive predictive value for T1D in patient siblings (6). The variable number tandem repeat located at 5′ of the hINS gene predisposes to T1D. In the NOD mouse, the lack of either the mouse insulin (mINS) 1 or 2 gene markedly alters the diabetes phenotype (4,7). We selected HLA-A*02:01 and HLA-DQ A1*0301/B1*03:02 (i.e., DQ8) as human MHC genes to be expressed in addition to the hINS gene. HLA-DQ8 carries the highest risk for T1D in man (8) and presumably presents epitopes that drive the autoimmunity to β-cells. Class I HLA-A*02:01 also modifies the risk for T1D and is the most common class I gene expressed in Caucasians (9).

We thus generated humanized mice that lack the expression of murine MHC class I and class II genes and insulin genes and express HLA-A*02:01, HLA-DQ8, and hINS transgenes, hereafter called YES mice. YES mice develop T1D upon injection of polyinosinic-polycytidylic acid (pI:C).

Mice

YES mice were obtained by crossing mINS−/− H-2k mice that express an hINS transgene under the control of the 353-base pair human insulin gene promoter on a mix C57BL/6JxCBA background (10) with double transgenic HLA-A*02:01/HLA-DQ8 mice carrying a dominant C57BL/6J background. The HLA-A*02:01/HLA-DQ8 mouse was obtained by crossing transgenic H-2Db mouse-β2-microglobulin (mß2m) double-knockout HLA-A*02:01 (11) and IAbIEb double-knockout HLA-DQ8 mice (12,13). It expresses a chimeric HLA-A*02:01 HHD monochain containing the HLA-A*02:01 α1 and α2 and H-2Db α3 domains linked to human β2m by a 15-mer linker under the control of the HLA-A*02:01 promoter (11) and the HLA-DQ8 α and β-chains under the control of their promoter (13) (Supplementary Fig. 1). Mice were maintained under specific pathogen-free conditions, and experiments were performed following Institutional Animal Care and Use Guidelines accreditation CEEA34.CB.024.11 by the ethics committee of Université Paris Descartes.

Genotyping

Insulin PCRs were performed with REDTaq (Sigma-Aldrich) using primers listed in Supplementary Table 1. Screening for mß2m and human HLA transgenes was performed with Advantage GC 2 Polymerase Mix and PCR Kit (Clontech). Real-time quantitative PCRs that were based on three-point dilutions using QuantiTect SYBR Green PCR Kit (QIAGEN) on a LightCycler 480 (Roche) allowed the discrimination of mice carrying a single mINS1 and/or mINS2 allele for further crosses. Cell surface stainings were performed on blood cells for the loss of H-2Dk and IAkIEk using anti-mouse T-cell receptor-allophycocyanin (APC)–labeled and anti-H-2Dk-fluorescein isothiocyanate (FITC)–labeled antibodies (BD Pharmingen) or using anti-mouse CD19-APC–labeled and anti-H-2IAk/IEk-FITC–labeled antibodies (BD Pharmingen). Data were collected with an LSRFortessa cytometer and analyzed using FlowJo version 9.2 software (Tree Star).

Thymic Epithelial Cell Isolation

Thymi from 6-week-old mice were digested with 0.125% collagenase D with 0.1% DNase I followed by two-step digestions with 0.5 units/mL Liberase TM and 0.1% DNase I (14). Thymic epithelial cell (TEC) enrichments were then depleted using mouse CD45 MicroBeads (Miltenyi Biotec) on an LD column according to the manufacturer’s protocol.

Flow Cytometry

Cell suspensions in RPMI medium/FCS 10% (106 cells/mL) were stained with anti-mouse CD3ε-PE, CD3ε-APC, CD8α-PerCP, CD4-efluor450, CD44-FITC, CD45-APC, CD19-APC, CD11c-APC, CD11b-efluor450 or CD11b-APC, anti-human β2m-FITC (clone TÜ99; BD Biosciences), anti-HLA-DQ-Biot (hybridoma SVPL3), streptavidin-FITC, anti-H-2Dk-FITC, anti-H-2IAk-FITC, anti-mouse NKp46-PE, CD25-PE, FoxP3-FITC, TCRβ-APC, NK1.1-APC, LY6G-PE-labeled, TER119-FITC (homemade), CD45.2-PE or CD90.2-FITC, biotin-conjugated anti-insulin (DAKO), biotin-conjugated anti-mouse CD62L and SAV-PE-Cy7 from eBioscience if not specified. Data were collected with an LSRFortessa cytometer and analyzed using FlowJo version 9.2 software.

Medullary TECs (mTECs) and cortical TECs (cTECs) were sorted with a FACSAria II using an anti-mouse CD45-Pe-Cy7, an anti-H-2IAb-FITC or anti-H-2IAk-FITC or anti-HLA-DQ-FITC, depending on the mouse genetic background; an anti-mouse Epcam-APC; an anti-mouse BP1-PE; an anti-mouse UEA-I-biot (Ulex europaeus agglutinin I); and a Brilliant Violet 650 Streptavidin (eBioscience, except for UEA-I-Biot [VECTOR Laboratories]). CD45 MHC class II+ Epcam+ cells define TECs. BP1 and UEA-I markers define cTECs as BP1+ UEA-I and BP1 UEA-I+ as mTECS (14).

Gene Expression

After RNAlater stabilization, RNA extractions were performed on solid organs and inguinal lymph nodes (iLNs) with RNeasy Mini Kit and DNase I treatment (QIAGEN). Sorted cTECs and mTECs were lysed in QIAzol Lysis Reagent, and RNA was extracted with miRNeasy Mini Kit and DNase I treatment (QIAGEN). Gene-specific primers were used for reverse transcription on 10–15 ng RNA (Supplementary Table 2). First-round PCRs were performed on 5 μL amplified cDNA (for ßactin, Aire, and Caseinß, 30 cycles/Tm = 54°C; for insulin genes, 30 cycles/Tm = 58°C) (Supplementary Table 3), and second-round PCRs were performed on 2 μL of the PCR1 product (for ßactin, Aire, and Caseinß, 40 cycles/Tm = 54°C; for all insulin genes, 40 cycles/Tm = 58°C) (15) (Supplementary Table 4). Gene expression data have been deposited in the National Center for Biotechnology Information Gene Expression Omnibus and are accessible through Gene Expression Omnibus series accession number GSE101551 (www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE101551).

Immunizations

Mice were immunized against either 100 μg HLA-A*02:01–restricted influenza matrix protein 2 peptide GILGFVFTL (MatA258–66) in Complete Freund’s Adjuvant. For hPPI analysis, YES mice were immunized with 100 μg hPPI peptides (Supplementary Table 5) with 140 μg HLA-DQ8–restricted helper Nef66–97 peptide or with 100 μg hPPI protein (Supplementary Methods 1) in Incomplete Freund’s Adjuvant (IFA) subcutaneously at the base of the tail followed by two recall injections in IFA every other week. Control immunizations were realized by injecting PBS 1× or KLH in adjuvant.

Enzyme-Linked Immunospot

Interferon-γ (IFN-γ)-enzyme-linked immunospot (ELIspot) assays were performed as previously reported (16). Spots were counted using Bioreader 5000 ProSF (BIO-SYS). Data are the mean of triplicate wells and expressed as spot-forming cells per 106 cells, evaluating the background IFN-γ responses in the absence of peptide. Positive controls were cells stimulated by 1 μg/mL ConA (Sigma-Aldrich) and negative controls by irrelevant pyruvate dehydrogenase (PDHase208–216) peptide. When indicated, responses were inhibited by preincubating splenocytes for 20 min with 50 μg/mL anti-HLA-A*02:01 antibody (BB7.2). For CD4+ IFN-γ-ELIspot assays, splenocytes were depleted using mouse anti-CD8 MicroBeads (Miltenyi) on an LD column according to the manufacturer’s protocol. When indicated, responses were inhibited by preincubating CD8+ T-cell–depleted splenocytes for 20 min with 50 µg/mL anti-HLA-DQ antibody (hybridoma SVPL3).

Cell Proliferations

In triplicate, 105 spleen cells/well were incubated with 0.5 μg antigen/well or 0.1 μg peptide/well (Supplementary Table 6) for 72 h at 37°C. Proliferation was evaluated with BrdU Cell Proliferation Assay Kit (Cell Signaling) and expressed as the proliferation index. Background and positive controls were evaluated in triplicate wells containing 105 cells/well incubated without antigen or in the presence of 10 μg/mL final concentration of anti-mouse CD3ε antibody. When indicated, the response was inhibited by adding 50 μg/mL anti-HLA-DQ8 antibody (hybridoma SVPL3).

Metabolic Evaluations

Body weight was monitored weekly for 20 weeks. Intraperitoneal glucose tolerance tests (2 g/kg body weight) were performed after 12–16 h of fasting. Intraperitoneal insulin tolerance tests (0.75 units/kg body weight) were performed by injecting Actrapid (Novo Nordisk) after 4 h of fasting. Blood glucose was measured using a glucometer (BG Star) at 0, 30, 60, and 120 min.

Immunostainings

Immunostainings were performed on formalin-fixed paraffin pancreas sections deparaffinized in xylene and dehydrated by ethanol. After washing, antigen retrieval was realized by hot incubation followed by permeabilization (20 min in PBS 1×/0.4% Triton X-100) and saturation (20 min PBS 1×/1% horse serum) before immunostaining with polyclonal guinea pig anti-insulin, polyclonal rabbit antiglucagon (Dako), anti-PDX1, anti-TLR3 (FITC, TLR3.7; Hycult Biotech), or rat anti-human CD3-Biot (AbD Serotec) antibodies overnight. Slides were washed with PBS 1×/1% BSA/0.1% Triton X-100 and stained with an anti-rabbit Ig-FITC antibody, a goat anti-guinea pig-CyA3 (Abcam), or SAV-Cy3 at room temperature. Sections were mounted in VECTASHIELD Antifade Mounting Medium for fluorescence with DAPI (Vector Laboratories). Observations were made with a Zeiss Axio Observer Z1 fluorescence microscope coupled with a Zeiss MRm AxioCam, and pictures were analyzed with ImageJ software. Assessment of β-cell mass was performed on stained microscope slide scans with ImageJ software using a guinea pig anti-hINS antibody (Dako) and calculated as the ratio between β-cell surface (μm2) / pancreas surface (μm2) × pancreas weight (mg) (17).

TLR Ligand Treatment and Additional Controls

Six- to 8-week-old mice were daily injected i.p. with 100 μg vac-pI:C (InvivoGen), or 50 μg CpG (ODN 2395; InvivoGen), or 10 μg lipopolysacharide (LPS) (Sigma) for 6 days, tested for glycosuria every 2 days, and diagnosed as diabetic on the basis of blood glucose levels >250 mg/dL. YES mice were injected with six daily doses of 2 mg corticosterone/kg as stress control or with five daily injections of 50 mg streptozotocin (STZ)/kg as diabetes-induced control. To deplete in vivo CD4+ and CD8+ T cells, we injected 500 μg of GK1.5 and 500 µg of YTS169.4 48 h before the pI:C injection, and we maintain CD4+ T-cell depletion for 3 more days (18,19).

HLA-A*02:01/HLA-DQ8/hINS YES Mice

YES mice lack the expression of mINS genes and express the hINS transgene in the pancreas and thymus (Fig. 1A and B). The hINS transgene was expressed in thymic mTEC, coexpressed with the AIRE gene, but not expressed in cortical cTEC (Fig. 1C), a pattern similar to that of hINS in parental mice and of mINS2 in C57BL/6 mice (15,20). We confirmed the loss of mouse MHC class I (H-2Dk and H-2Kk) (Fig. 1D, upper panel) and class II (IAk) (Fig. 1D, lower panel) and the presence of the transgenic HLA-A*02:01 molecule (Fig. 1E, upper panel) on T cells and the transgenic HLA-DQ8 molecule (Fig. 1E, lower panel) on B cells. We observed the expression of HLA-A*02:01 on CD45 insulin+ islet cells from YES mice as of murine H-2Db in C57BL/6 mice (Fig. 1F).

Figure 1

Insulin and MHC class I and class II expression in YES mice. A: RT-PCR analysis of mIns expression in thymus and pancreas of YES mice compared with the pancreas of C57BL/6 mice. GeneRuler 1 kb Plus DNA Ladder to 500–70 bp. B: Nested RT-PCR analysis of hINS expression in various organs of YES mice. GeneRuler 1 kb Plus DNA Ladder to 500–70 bp. C: Nested RT-PCR analysis of hINS transgene and mIns expression in thymocyte fractions, cTECs, and mTECs of YES mice, parental hINS mice, and control C57BL/6 mice compared with mAire and mCsn2 (casein2) gene expression. GeneRuler 1 kb plus DNA ladder to 500–70 bp. D: Flow cytometry evaluation of the expression of mouse class I H-2Dk and class II IAk, respectively on T (upper panel) and B cells (lower panel), in YES and parental hINS transgenic mice. E: Flow cytometry evaluation of the expression of HLA-A*02:01/HHD and HLA-DQ8, respectively on T (upper panel) and B cells (lower panel), in YES and parental hINS transgenic mice. F: Flow cytometry evaluation of the expression of HLA-A*02:01/HHD and H-2Db on islets cells (CD45Ins+ cells) in YES and C57BL/6 conventional mice. FSC, forward scatter; PLN, peripheric lymph node; SSC, side scatter.

Figure 1

Insulin and MHC class I and class II expression in YES mice. A: RT-PCR analysis of mIns expression in thymus and pancreas of YES mice compared with the pancreas of C57BL/6 mice. GeneRuler 1 kb Plus DNA Ladder to 500–70 bp. B: Nested RT-PCR analysis of hINS expression in various organs of YES mice. GeneRuler 1 kb Plus DNA Ladder to 500–70 bp. C: Nested RT-PCR analysis of hINS transgene and mIns expression in thymocyte fractions, cTECs, and mTECs of YES mice, parental hINS mice, and control C57BL/6 mice compared with mAire and mCsn2 (casein2) gene expression. GeneRuler 1 kb plus DNA ladder to 500–70 bp. D: Flow cytometry evaluation of the expression of mouse class I H-2Dk and class II IAk, respectively on T (upper panel) and B cells (lower panel), in YES and parental hINS transgenic mice. E: Flow cytometry evaluation of the expression of HLA-A*02:01/HHD and HLA-DQ8, respectively on T (upper panel) and B cells (lower panel), in YES and parental hINS transgenic mice. F: Flow cytometry evaluation of the expression of HLA-A*02:01/HHD and H-2Db on islets cells (CD45Ins+ cells) in YES and C57BL/6 conventional mice. FSC, forward scatter; PLN, peripheric lymph node; SSC, side scatter.

Close modal

HLA-A*02:01/HHD expression was confirmed on immune cells in spleen, blood, and iLNs by flow cytometry (Supplementary Fig. 2). In all subsets analyzed, HLA-A*02:01 expression was comparable in YES and in parental HLA-A*02:01/HLA-DQ8 double transgenic mice (data not shown). Expression of both HLA class I and class II transgenic molecules was lower than expression of H-2Db (Supplementary Fig. 2A) and H-2IAb (Supplementary Fig. 2B) molecules in C57BL/6 mice.

Because the YES genetic background is a mix of C57BL/6 and CBA strains, we characterized the YES genome relative to C57BL/6 and NOD genomes (Supplementary Methods 2). NOD and C57BL/6 mice showed 80.4% identity. YES mice showed 79.3% identity with NOD but 90.9% identity with C57BL/6 mice (Table 1). We studied T1D-associated regions in the YES genome. Mouse linkage regions associated with T1D were listed and updated on the University of California, Santa Cruz, Genome Browser (https://genome.ucsc.edu/index.html). Single-nucleotide polymorphisms (SNPs) that were identical in NOD and C57BL/6 mice were not further considered. Between YES and C57BL/6 mice, 0.21% of SNPs were different. SNPs were listed according to each chromosome and corresponding Idd loci when relevant. We identified SNPs for which at least one variation was identified. Using the Mouse Genome Informatics Web site (www.informatics.jax.org), we found that 86% of SNPs associated to a CBA genetic background. Fourteen percent of SNPs were not described or covered regions for which PCR probes do not discriminate NOD, C57BL/6, and CBA Idd profiles or could not be defined in the absence of a marker for the relevant region (Supplementary Table 7).

Table 1

Similarity of genetic backgrounds of NOD, C57BL/6, YES mice

Identity NOD (%)C57BL/6 (%)YES (%)
NOD ND 80.4 79.3 
C57BL/6 80.4 ND 90.9 
YES 79.3 90.9 99.9 
Identity NOD (%)C57BL/6 (%)YES (%)
NOD ND 80.4 79.3 
C57BL/6 80.4 ND 90.9 
YES 79.3 90.9 99.9 

ND, not determined.

Distribution of Immune Cell Subsets in YES Mice

Absolute numbers of spleen, iLN, and thymic cells were comparable in YES and in parental HLA-A*02:01/HLA-DQ8 double transgenic mice (data not shown). Distributions of most immune cell subsets were comparable in YES and C57BL/6 mice, with few differences (Fig. 2). Spleen T-cell frequencies were lower in YES than in C57BL/6 mice (13.0 ± 2.4% vs. 29.0 ± 2.1%), with an inverted CD3+CD4+/CD3+CD8+ T-cell ratio (0.7 and 1.35, respectively) (Fig. 3A). A significant fraction of cells (9.0 ± 3.9%) were CD3CD4 CD8CD19CD11bCD11cNk1.1 and not observed in parental HLA-A*02:01/HLA-DQ8 double transgenic mice (Supplementary Fig. 3) but were present in parental hINS transgenic mice (8.0 ± 2.9%). Corresponding cells were hematopoietic cells expressing CD45. In YES spleens, a majority of cells were analogous to double-negative DN3 (CD44CD25+) pre-T cells (59.3%), whereas a minority was analogous to DN1 (CD44+CD25, 17.2%), DN2 (CD44+CD25+, 5.2%) pro-T cells, and DN4 (CD44CD25, 16.3%) pre-T cells compared with 3.4%, 32.2%, 3.1%, and 61.2%, respectively, in hINS transgenic mice (21). In YES spleens, those cells were CD62L, suggesting that they were immature cells incapable of homing in peripheral lymph nodes. In iLN cells (Fig. 2B), an inverted CD3+CD4+/CD3+CD8+ T-cell ratio was also observed in YES compared with C57BL/6 mice (0.5 and 1.25, respectively), with a slight difference in T-cell frequency (47.0 ± 8.5% vs. 53.45 ± 5.6%). In peripheral blood, T cells were underrepresented in YES compared with C57BL/6 mice (12.0 ± 9.8% vs. 48.0 ± 7.6%; P = 0.0238 by Mann-Whitney test), with an inverted CD3+CD4+/CD3+CD8+ T-cell ratio (0.69 and 1.3, respectively). Dendritic cells (DCs) (12.0 ± 1.83% vs. 0.89 ± 1.1%) and macrophages (30.0 ± 8.1% vs. 8.83 ± 3.4%) were overrepresented (Fig. 2C). Thymus analyses showed no difference in YES compared with C57BL/6 mice. The dominant CD3+low T-cell subset, which mainly consists of CD3+lowCD4+CD8+ double-positive cells, was comparable in YES and C57BL/6 mice (90 ± 3.19% and 95 ± 3.09%, respectively) (Fig. 2D). CD4+CD25+Foxp3+ regulatory T cells were not significantly different in YES and C57BL/6 mice.

Figure 2

Immune cell subsets in YES mice. Splenocytes (A), iLN cells (B), peripheral blood mononuclear cells (PBMCs) (C), and thymocytes (D) were stained using the following antibodies: anti-CD19 (B cells), anti-CD11c (DCs), anti-CD11b (macrophages), anti-NKp46 (natural killer [NK] cells), and anti-CD3ε (T cells; CD3low, CD3high). T-cells subsets were further stained with anti-CD4 and anti-CD8α monoclonal antibodies (gray). A population of immature cells (ICs) was detected in YES mice (gold and Supplementary Fig. 3).

Figure 2

Immune cell subsets in YES mice. Splenocytes (A), iLN cells (B), peripheral blood mononuclear cells (PBMCs) (C), and thymocytes (D) were stained using the following antibodies: anti-CD19 (B cells), anti-CD11c (DCs), anti-CD11b (macrophages), anti-NKp46 (natural killer [NK] cells), and anti-CD3ε (T cells; CD3low, CD3high). T-cells subsets were further stained with anti-CD4 and anti-CD8α monoclonal antibodies (gray). A population of immature cells (ICs) was detected in YES mice (gold and Supplementary Fig. 3).

Close modal
Figure 3

Metabolic characterization of YES mice. A and B: Body weight of male (●, n = 6) and female (○, n = 6) YES mice, male (▲, n = 6) and female (△, n = 6) HLA-A*02:01/HLA-DQ8 parental mice, and male (■, n = 6) and female (□, n = 6) hINS parental mice up to 20 weeks of age. C: Insulin (Ins, red), glucagon (Glc, green), PDX1 (green), and cell nuclei (DAPI blue) staining of YES and HLA-A*02:01/HLA-DQ8 pancreatic sections. Magnification ×20. D: β-Cell mass: male (●, n = 6) and female (○, n = 6) YES mice, male (▲, n = 3) and female (△, n = 3) HLA-A*02:01/HLA-DQ8 parental mice, and male (■, n = 3) and female (□, n = 3) hINS parental mice. β-Cell mass is expressed in milligrams for the whole pancreas. E: Intraperitoneal glucose tolerance (IGT) test after a 2 g/kg glucose injection and areas under the curve in 6-month-old male (n = 4, ● and black bars, respectively) and female (n = 4, ○ and black checkerboard bars, respectively) YES mice and male (n = 4, ▲ and gray bars, respectively) and female (n = 4, △ and gray checkerboard bars, respectively) HLA-A*02:01/HLA-DQ8 transgenic mice. F: Intraperitoneal insulin tolerance (IIT) test curves after 0.75 units/kg insulin injection and areas under the curve in 15-week-old male (n = 4, ● and black bars, respectively) and female (n = 6, ○ and black checkerboard bars, respectively) YES mice and male (n = 5, ▲ and gray bars, respectively) and female (n = 5, △ and gray checkerboard bars, respectively) HLA-A*02:01/HLA-DQ8 parental mice. *P ≤ 0.05 by Mann-Whitney test. ns, nonsignificant.

Figure 3

Metabolic characterization of YES mice. A and B: Body weight of male (●, n = 6) and female (○, n = 6) YES mice, male (▲, n = 6) and female (△, n = 6) HLA-A*02:01/HLA-DQ8 parental mice, and male (■, n = 6) and female (□, n = 6) hINS parental mice up to 20 weeks of age. C: Insulin (Ins, red), glucagon (Glc, green), PDX1 (green), and cell nuclei (DAPI blue) staining of YES and HLA-A*02:01/HLA-DQ8 pancreatic sections. Magnification ×20. D: β-Cell mass: male (●, n = 6) and female (○, n = 6) YES mice, male (▲, n = 3) and female (△, n = 3) HLA-A*02:01/HLA-DQ8 parental mice, and male (■, n = 3) and female (□, n = 3) hINS parental mice. β-Cell mass is expressed in milligrams for the whole pancreas. E: Intraperitoneal glucose tolerance (IGT) test after a 2 g/kg glucose injection and areas under the curve in 6-month-old male (n = 4, ● and black bars, respectively) and female (n = 4, ○ and black checkerboard bars, respectively) YES mice and male (n = 4, ▲ and gray bars, respectively) and female (n = 4, △ and gray checkerboard bars, respectively) HLA-A*02:01/HLA-DQ8 transgenic mice. F: Intraperitoneal insulin tolerance (IIT) test curves after 0.75 units/kg insulin injection and areas under the curve in 15-week-old male (n = 4, ● and black bars, respectively) and female (n = 6, ○ and black checkerboard bars, respectively) YES mice and male (n = 5, ▲ and gray bars, respectively) and female (n = 5, △ and gray checkerboard bars, respectively) HLA-A*02:01/HLA-DQ8 parental mice. *P ≤ 0.05 by Mann-Whitney test. ns, nonsignificant.

Close modal

YES Responses to Conventional Antigens

Because differences were seen in immune cell distributions, we evaluated immune responses to conventional antigens. After immunization against MatA258–66, YES splenocytes showed a strong IFN-γ-ELIspot response (Supplementary Fig. 4A) that was inhibited in the presence of the anti-HLA-A*02:01BB7.2 antibody. CD8+ T-cell–depleted HLA-DQ8–restricted CD4+ spleen T cells from YES mice immunized against KLH showed significant IFN-γ responses (Supplementary Fig. 4B) that were inhibited by an anti-HLA-DQ antibody (P ≤ 0.03). Altogether, these experiments indicate that YES mice respond to conventional antigens as expected.

YES Mice Show Conserved Glucose Homeostasis

Because hINS binding to the mouse insulin receptor is in the normal range (22), YES mice are expected to remain normoglycemic. They maintained a normal weight in the long-term range (Fig. 3A and B). Their overall islet morphology was comparable in YES and HLA-A*02:01/HLA-DQ8 parental mice, as were insulin, glucagon, and PDX1 staining (Fig. 3C). The β-cell mass was comparable in YES mice and parental hINS and HLA-A*02:01/HLA-DQ8 mice at 15 weeks of age (Fig. 3D). No significant difference was observed in YES responses to the 2 mg/kg intraperitoneal glucose tolerance test at 6 months of age compared with HLA-A*02:01/HLA-DQ8 parental mice (Fig. 3E). Glycemic responses to 0.75 units/kg i.p. insulin were similar in female YES mice and parental HLA-A*02:01/HLA-DQ8 females, whereas a difference was observed in male mice (Fig. 3F). In the long-term range, YES mice developed neither diabetes nor insulitis.

YES Immune Responses to Preproinsulin

We previously identified CD8+ T-cell responses to hPPI peptides in human T1D (23). After immunization of YES mice against individual HLA-A*02:01–restricted hPPI peptides, a significant response was detected against hPPI2–11 (P ≤ 0.03) and hPPI6–14 (P ≤ 0.03) using an IFN-γ-ELIspot assay. A significant, but weak response was detected against hPPI33–42 (P ≤ 0.04) and against hPPI101–109 in an individual mouse (Fig. 4A).

Figure 4

MHC class I and class II restricted T-cell responses against hPPI in YES mice. A: IFN-γ responses against individual hPPI peptides restricted to MHC class I. YES mice were immunized against hPPI2–11, hPPI6–14, hPPI15–24, hPPI30–39, hPPI33–42, hPPI34–42, hPPI42–51, and hPPI101–109 peptides (100 µg/mouse, Complete Freund’s Adjuvant, HLA-DQ–restricted helper Nef66–97 peptide) or PBS and boosted (IFA, two times). IFN-γ-ELIspot assays were performed in the presence of the immunizing peptide, PDHase208–216 as irrelevant peptide, or ConA as positive control. Numbers of spot-forming cells were expressed for 106 cells. B: IFN-γ responses to hPPI. Splenocytes from YES mice immunized against hPPI full-length protein or PBS were depleted in CD8+ T cells and IFN-γ-ELIspot assays performed in the presence of individual hPPI peptide, Nef66–97 as irrelevant peptide, or ConA as positive control. Numbers of spot-forming cells were expressed for 106 cells. C: Proliferative responses to full-length hPPI and hPPI peptides hPPI1–15, hPPI8–23, hPPI16–30, hPPI18–30, hPPI20–35, hPPI25–40, hPPI33–47, hPPI40–55, hPPI46–61, hPPI55–70, hPPI61–76, hPPI70–86, hPPI80–97, and hPPI92–110. Splenocytes from YES mice immunized against hPPI full-length protein or PBS were stimulated in vitro for 3 days, and proliferation was evaluated by measuring BrdU incorporation. Each dot represents an individual mouse. *P ≤ 0.05, **P ≤ 0.01, ***P ≤ 0.0008 by Mann-Whitney test. Nb, number.

Figure 4

MHC class I and class II restricted T-cell responses against hPPI in YES mice. A: IFN-γ responses against individual hPPI peptides restricted to MHC class I. YES mice were immunized against hPPI2–11, hPPI6–14, hPPI15–24, hPPI30–39, hPPI33–42, hPPI34–42, hPPI42–51, and hPPI101–109 peptides (100 µg/mouse, Complete Freund’s Adjuvant, HLA-DQ–restricted helper Nef66–97 peptide) or PBS and boosted (IFA, two times). IFN-γ-ELIspot assays were performed in the presence of the immunizing peptide, PDHase208–216 as irrelevant peptide, or ConA as positive control. Numbers of spot-forming cells were expressed for 106 cells. B: IFN-γ responses to hPPI. Splenocytes from YES mice immunized against hPPI full-length protein or PBS were depleted in CD8+ T cells and IFN-γ-ELIspot assays performed in the presence of individual hPPI peptide, Nef66–97 as irrelevant peptide, or ConA as positive control. Numbers of spot-forming cells were expressed for 106 cells. C: Proliferative responses to full-length hPPI and hPPI peptides hPPI1–15, hPPI8–23, hPPI16–30, hPPI18–30, hPPI20–35, hPPI25–40, hPPI33–47, hPPI40–55, hPPI46–61, hPPI55–70, hPPI61–76, hPPI70–86, hPPI80–97, and hPPI92–110. Splenocytes from YES mice immunized against hPPI full-length protein or PBS were stimulated in vitro for 3 days, and proliferation was evaluated by measuring BrdU incorporation. Each dot represents an individual mouse. *P ≤ 0.05, **P ≤ 0.01, ***P ≤ 0.0008 by Mann-Whitney test. Nb, number.

Close modal

YES mice were immunized against hPPI and tested for responses against a panel of hPPI-overlapping peptides. They remained insulitis free upon immunization (data not shown). Significant IFN-γ responses (Fig. 4B) were detected against peptides overlapping the whole hPPI sequence. When considering individual mouse responses, 93.75% of mice showed IFN-γ responses against at least one hPPI peptide (Supplementary Table 8). Observed predominant individual IFN-γ responses were to hPPI20–35 (62.5% of mice; P ≤ 0.008), hPPI25–40 (56.25% of mice; P ≤ 0.02), hPPI46–61 (56.25% of mice; P ≤ 0.02), hPPI55–70 (68.75% of mice; P ≤ 0.005), hPPI61–76 (62.5% of mice; P ≤ 0.008), hPPI92–110 (68.75% of mice; P ≤ 0.005), and hPPI (81.25% of mice; P ≤ 0.0005).

The number of epitopes that led to proliferative responses in vitro after hPPI immunization was restricted compared with IFN-γ responses (Fig. 4C). As for IFN-γ responses, proliferative responses were diversified. After determination of the threshold value for each peptide in YES mice (Supplementary Table 9) using pairs of measurements and the Bland-Altman test (Supplementary Table 10), responses were seen against both hPPI55–70 peptide (58.3% of mice; P ≤ 0.006) and hPPI (66.6% of mice; P ≤ 0.02) in a significant number of mice. Proliferative responses were seen against at least one hPPI peptide in 91.67% mice upon immunization against hPPI. These experiments show responses that preferentially cluster around a region covering the leader, B-chain, and C-peptide sequences.

Induction of Autoimmune Diabetes in YES Mice

We addressed whether YES mice were amenable to T1D induction. After six daily 100-μg pI:C injections (24), diabetes developed within 1 week after the last injection (P ≤ 0.0003) but not after LPS or CpG injection (Fig. 5A). As expected, diabetes occurred after multi-STZ low-dose stimulation (P ≤ 0.0001) but was not induced under cortisol stress conditions that were tested as a control. Administration of anti-CD8 and -CD4 monoclonal antibodies prevented diabetes induced by pI:C stimulation (P ≤ 0.04) (Fig. 5B). An islet infiltration was observed in diabetic mice (Fig. 5C), with islets showing a loss of cells expressing insulin (Fig. 5D) and the presence of CD3+ T cells (Fig. 5E). No significant TLR3 expression was observed in control mice that remained untreated (Fig. 5F). TLR3 expression in diabetic pI:C-treated mice showed a speckled distribution within the islet cell cytoplasm, which mostly colocalized with insulin (Fig. 5G). A few glucagon-stained cells showed TLR3 colocalization. Spleen CD8+ T cells from diabetic pI:C-treated mice showed IFN-γ responses to hPPI6–14 (P ≤ 0.016) and hPPI15–24 (P ≤ 0.02), although not to hPPI2–11 or hPPI33–42 as observed in hPPI-immunized YES mice (Fig. 6A). Individual responses were observed against at least one hPPI peptide in 80% of mice. No CD8+ T-cell responses were observed in nondiabetic pI:C-treated mice. We next evaluated CD4+ T-cell responses in pI:C-treated and control YES mice (Fig. 6B). Diabetic pI:C-treated mice showed significant proliferative responses compared with control YES mice as follows: hPPI16–30 (P ≤ 0.048), hPPI18–30 (P ≤ 0.008), hPPI33–47 (P ≤ 0.05), hPPI40–55 (P ≤ 0.016), hPPI46–61 (P ≤ 0.022), hPPI61–76 (P ≤ 0.031), hPPI80–97 (P ≤ 0.041), and hPPI92–110 (P ≤ 0.005) and the hPPI protein (P ≤ 0.011). As for CD4+ T-cell responses in hPPI-immunized YES mice, individual responses were observed against at least one hPPI peptide in 86.7% of mice (Supplementary Tables 10 and 11). Responses were against hPPI1–15 (53.3%; P ≤ 0.02), hPPI33–47 (46.7%; P ≤ 0.05), hPPI46–61 (46.7%; P ≤ 0.05), hPPI70–86 (53.3%; P ≤ 0.02), hPPI92–110 (73.3%; P ≤ 0.02), and hPPI (53.3%; P ≤ 0.05) (Fig. 6C). Responses clustered in a region covering the leader sequence and C-peptide sequence. A strong correlation was observed between the responses against hPPI protein and most of all hPPI peptides in diabetic pI:C-treated YES mice (Supplementary Fig. 5). In contrast with CD8+ T cells, we detected proliferative responses against hPPI peptides in splenocytes from nondiabetic pI:C-treated YES mice (Fig. 6B). All mice also were tested for ZnT8 and GAD T-cell responses (Fig. 6D). Significant proliferative responses were observed against ZnT8166–179 and GAD536–550 peptides that are common to humans and mice. Stronger CD8+ T-cell (Fig. 7A) and CD4+ T-cell (Fig. 7B) responses were observed in pancreatic infiltrates in diabetic versus nondiabetic pI:C-treated YES mice. Infiltrating cells from nondiabetic pI:C-treated mice showed an increased frequency of regulatory T cells but a decreased frequency in CD11b+ and DCs compared with diabetic pI:C-treated mice (Fig. 7C). In pancreatic lymph nodes, populations maintained similar frequencies with the exception of DCs expressing the activator marker CD103 specifically in diabetic pI:C-treated YES mice (Fig. 7D).

Figure 5

pI:C induction of diabetes in YES mice. A: Diabetes incidence upon pI:C stimulation (six daily injections i.p./100 μg pI:C) in YES mice compared with the CpG-stimulated YES mice (six daily injections i.p./50 μg ODN2395), LPS-stimulated YES mice (six daily injections i.p./10 μg LPS), or untreated YES mice. B: Diabetes incidence upon pI:C stimulation (six daily injections i.p./100 μg pI:C) in YES mice compared with the STZ-stimulated YES mice (five daily injections i.p./50 mg/kg STZ), corticosterone-stimulated YES mice (six daily injections i.p./2 mg/kg corticosterone), PBS-treated YES mice (six daily injections i.p./100 μL PBS 1×), and pI:C-stimulated depleted YES mice (two daily injection i.p. of 500 μg GK1.5 and 500 μg YTS 169.4 followed by three daily injections i.p. of 100 μg pI:C and 500 μg GK1.5 then three daily injections i.p. of 100 μg pI:C). C: Paraffin pancreas sections stained by hematoxylin-eosin in pI:C-induced diabetic YES mice. Magnification ×10. D: Glucagon and insulin immunofluorescent staining of pancreatic section from pI:C-induced diabetic YES mice and YES control mice. E: Glucagon and CD3 immunofluorescent staining of pancreatic section from pI:C-induced diabetic YES mice and YES control mice. F and G: Glucagon (gray), insulin (red), and TLR 3 (green) staining of pancreatic sections from YES mice and from pI:C-induced diabetic YES mice, respectively. D and E: Magnification ×20. F and G: Magnification ×40. *P ≤ 0.05, ***P ≤ 0.003 by log-rank (Mantel-Cox) test.

Figure 5

pI:C induction of diabetes in YES mice. A: Diabetes incidence upon pI:C stimulation (six daily injections i.p./100 μg pI:C) in YES mice compared with the CpG-stimulated YES mice (six daily injections i.p./50 μg ODN2395), LPS-stimulated YES mice (six daily injections i.p./10 μg LPS), or untreated YES mice. B: Diabetes incidence upon pI:C stimulation (six daily injections i.p./100 μg pI:C) in YES mice compared with the STZ-stimulated YES mice (five daily injections i.p./50 mg/kg STZ), corticosterone-stimulated YES mice (six daily injections i.p./2 mg/kg corticosterone), PBS-treated YES mice (six daily injections i.p./100 μL PBS 1×), and pI:C-stimulated depleted YES mice (two daily injection i.p. of 500 μg GK1.5 and 500 μg YTS 169.4 followed by three daily injections i.p. of 100 μg pI:C and 500 μg GK1.5 then three daily injections i.p. of 100 μg pI:C). C: Paraffin pancreas sections stained by hematoxylin-eosin in pI:C-induced diabetic YES mice. Magnification ×10. D: Glucagon and insulin immunofluorescent staining of pancreatic section from pI:C-induced diabetic YES mice and YES control mice. E: Glucagon and CD3 immunofluorescent staining of pancreatic section from pI:C-induced diabetic YES mice and YES control mice. F and G: Glucagon (gray), insulin (red), and TLR 3 (green) staining of pancreatic sections from YES mice and from pI:C-induced diabetic YES mice, respectively. D and E: Magnification ×20. F and G: Magnification ×40. *P ≤ 0.05, ***P ≤ 0.003 by log-rank (Mantel-Cox) test.

Close modal
Figure 6

T-cell responses against hPPI. A: Analysis of CD8+ T-cell responses by IFN-γ-ELIspot assays performed in the presence of individual hPPI peptides (hPPI2–11, hPPI6–14, hPPI15–24, hPPI30–39, hPPI33–42, hPPI34–42, hPPI42–51, hPPI101–109), Nef66–97 as irrelevant peptide, or ConA as positive control for splenocytes from diabetic pI:C-treated YES mice compared with splenocytes from unstimulated YES mice and nondiabetic pI:C-treated YES mice. B: Analysis of CD4+ T-cell responses by proliferative assays performed in the presence of full-length hPPI and individual hPPI peptides (hPPI1–15, hPPI8–23, hPPI16–30, hPPI18–30, hPPI20–35, hPPI25–40, hPPI33–47, hPPI40–55, hPPI46–61, hPPI55–70, hPPI61–76, hPPI70–86, hPPI80–97, and hPPI92–110). Splenocytes from diabetic pI:C-treated YES mice and nondiabetic pI:C-treated YES mice compared with the untreated YES mice were stimulated in vitro for 3 days with individual peptides, and proliferation was evaluated by measuring BrdU incorporation. C: Radar chart representing the percentage of frequency of recognition of hPPI peptides in diabetic pI:C-treated YES mice, hPPI-immunized YES mice, and YES control mice. D: Analysis of CD4+ T-cell response by proliferative assays performed in the presence of full-length hPPI and individual ZnT8166–179 peptide or GAD101–115, GAD126–135, GAD207–220, and GAD536–550. Splenocytes from diabetic pI:C-treated YES mice and nondiabetic pI:C-treated YES mice compared with the untreated YES mice were stimulated in vitro for 3 days with individual peptides, and proliferation was evaluated by measuring BrdU incorporation. Each dot represents an individual mouse. *P ≤ 0.05, **P ≤ 0.01 by Mann-Whitney test. Nb, number.

Figure 6

T-cell responses against hPPI. A: Analysis of CD8+ T-cell responses by IFN-γ-ELIspot assays performed in the presence of individual hPPI peptides (hPPI2–11, hPPI6–14, hPPI15–24, hPPI30–39, hPPI33–42, hPPI34–42, hPPI42–51, hPPI101–109), Nef66–97 as irrelevant peptide, or ConA as positive control for splenocytes from diabetic pI:C-treated YES mice compared with splenocytes from unstimulated YES mice and nondiabetic pI:C-treated YES mice. B: Analysis of CD4+ T-cell responses by proliferative assays performed in the presence of full-length hPPI and individual hPPI peptides (hPPI1–15, hPPI8–23, hPPI16–30, hPPI18–30, hPPI20–35, hPPI25–40, hPPI33–47, hPPI40–55, hPPI46–61, hPPI55–70, hPPI61–76, hPPI70–86, hPPI80–97, and hPPI92–110). Splenocytes from diabetic pI:C-treated YES mice and nondiabetic pI:C-treated YES mice compared with the untreated YES mice were stimulated in vitro for 3 days with individual peptides, and proliferation was evaluated by measuring BrdU incorporation. C: Radar chart representing the percentage of frequency of recognition of hPPI peptides in diabetic pI:C-treated YES mice, hPPI-immunized YES mice, and YES control mice. D: Analysis of CD4+ T-cell response by proliferative assays performed in the presence of full-length hPPI and individual ZnT8166–179 peptide or GAD101–115, GAD126–135, GAD207–220, and GAD536–550. Splenocytes from diabetic pI:C-treated YES mice and nondiabetic pI:C-treated YES mice compared with the untreated YES mice were stimulated in vitro for 3 days with individual peptides, and proliferation was evaluated by measuring BrdU incorporation. Each dot represents an individual mouse. *P ≤ 0.05, **P ≤ 0.01 by Mann-Whitney test. Nb, number.

Close modal
Figure 7

Pancreatic infiltrates analysis. A: Analysis of CD8+ T-cell response by IFN-γ-ELIspot assays performed in the presence of a pool of hPPI peptides (hPPI2–11, hPPI6–14, hPPI15–24, hPPI30–39, hPPI33–42, hPPI34–42, hPPI42–51, hPPI101–109) and Nef66–97 as irrelevant peptide for pancreatic infiltrating cells from diabetic pI:C-treated YES mice (n = 6) compared with the nondiabetic pI:C-treated YES mice (n = 3). Each dot represents an individual mouse. B: Analysis of CD4+ T-cell response by proliferative assays performed in the presence of full-length hPPI and irrelevant peptide Nef66–97. Pancreatic infiltrating cells from diabetic pI:C-treated YES mice (n = 12) and nondiabetic pI:C-treated YES mice (n = 3) were stimulated in vitro for 3 days with hPPI protein, and proliferation was evaluated by measuring BrdU incorporation. Each dot represents an individual mouse. C: Immunophenotyping of pancreatic infiltrating cells from diabetic pI:C-treated YES mice (n = 3) and nondiabetic pI:C-treated YES mice (n = 3) for the frequency of B cells, T cells, CD8+ T cells, CD4+ T cells, regulatory T cells defined as CD4+CD25+Foxp3+ T cells, CD11b+ cells, and DCs defined as CD11C+ and CD11c+CD11b+ cells. D: Immunophenotyping of pancreatic lymph node cells from diabetic pI:C-treated YES mice (●; n = 3), nondiabetic pI:C-treated YES mice (●; n = 3), and untreated YES mice (○; n = 3) for the frequency of B cells, T cells, CD8+ T cells, CD4+ T cells, regulatory T cells defined as CD4+CD25+Foxp3+ T cells, CD11b+ cells, DCs defined as CD11c+ and CD11c+CD11b+ cells, and CD103+ DCs. Nb, number. (A high-quality color representation of this figure is available in the online issue.)

Figure 7

Pancreatic infiltrates analysis. A: Analysis of CD8+ T-cell response by IFN-γ-ELIspot assays performed in the presence of a pool of hPPI peptides (hPPI2–11, hPPI6–14, hPPI15–24, hPPI30–39, hPPI33–42, hPPI34–42, hPPI42–51, hPPI101–109) and Nef66–97 as irrelevant peptide for pancreatic infiltrating cells from diabetic pI:C-treated YES mice (n = 6) compared with the nondiabetic pI:C-treated YES mice (n = 3). Each dot represents an individual mouse. B: Analysis of CD4+ T-cell response by proliferative assays performed in the presence of full-length hPPI and irrelevant peptide Nef66–97. Pancreatic infiltrating cells from diabetic pI:C-treated YES mice (n = 12) and nondiabetic pI:C-treated YES mice (n = 3) were stimulated in vitro for 3 days with hPPI protein, and proliferation was evaluated by measuring BrdU incorporation. Each dot represents an individual mouse. C: Immunophenotyping of pancreatic infiltrating cells from diabetic pI:C-treated YES mice (n = 3) and nondiabetic pI:C-treated YES mice (n = 3) for the frequency of B cells, T cells, CD8+ T cells, CD4+ T cells, regulatory T cells defined as CD4+CD25+Foxp3+ T cells, CD11b+ cells, and DCs defined as CD11C+ and CD11c+CD11b+ cells. D: Immunophenotyping of pancreatic lymph node cells from diabetic pI:C-treated YES mice (●; n = 3), nondiabetic pI:C-treated YES mice (●; n = 3), and untreated YES mice (○; n = 3) for the frequency of B cells, T cells, CD8+ T cells, CD4+ T cells, regulatory T cells defined as CD4+CD25+Foxp3+ T cells, CD11b+ cells, DCs defined as CD11c+ and CD11c+CD11b+ cells, and CD103+ DCs. Nb, number. (A high-quality color representation of this figure is available in the online issue.)

Close modal

The clinical management of T1D faces the lack of fully accurate biomarkers of autoimmunity and of efficient and specific immunotherapies. Major improvements in glucose monitoring and insulin therapies have narrowed the mortality gap within the general population and have shifted the safety line that balances risks and benefits toward the safest immunotherapy approaches. Meanwhile, antigen-specific immunotherapy has a long way to go. The autoantigen dose, delivery route, stage of the disease process at which immunotherapy is applied, and use of peptides rather than full-length autoantigens remain open issues in humans (25). Models that would allow direct translation of experimentally defined peptides in man are lacking. In addition, environmental factors that trigger diabetes on a susceptible genetic background remain elusive. We almost exclusively rely on the NOD and MHC class I humanized NOD mice to address such issues (26,27). The NOD mouse shows a unique phenotype that is unlikely to summarize the heterogeneity of T1D mechanisms. It has been selected for spontaneous diabetes development, precluding its use in strategies to explore the triggering of diabetes by environmental factors on conventional genetic backgrounds.

The YES mouse is a new model that lacks mouse class I, class II, and insulin genes and expresses HLA-A*02:01, HLA-DQ8, and hINS. Parental mice carrying the hINS transgene have normal insulin levels and glucose homeostasis (28). Most differences seen between YES and control mice were minor and likely represent the expected interstrain variability. YES mice remain normoglycemic up to 1 year of age, respond normally to an intraperitoneal glucose challenge, and maintain a normal β-cell mass. The distribution of most immune cell subsets in YES mice is comparable to that of a conventional mouse strain. However, T cells were underrepresented in YES mice. Given an increased percentage of CD3+CD4CD8+ single-positive T cells in the thymus, shifts in peripheral T-cell representation in YES mice is likely to relate with shifted efficiency of human MHC molecules to select T cells. The inverted CD4+/CD8+ T-cell ratio seen in YES and parental HLA-A*02:01/HLA-DQ8 mice suggests a lower efficiency of class II HLA-DQ8 to select CD4+ T cells than of class I HLA-A*02:01 to select CD8+ T cells. Because the expression of the insulin gene controls the selection of insulin-specific T cells in the thymus (4,29), we evaluated the expression of the hINS transgene in thymic cells. YES mice exclusively express the hINS transgene in mTECs, a pattern identical to expression patterns observed in C57BL/6 or NOD mice and in humans. Immunization with hPPI induced neither insulitis nor diabetes in YES mice, indicating that they are tolerant to the hPPI transgene.

The main bias toward T1D development in YES mice is the expression of DQ8. The YES genetic background was closer to the C57BL/6 and CBA than to the NOD strain. YES mice were tolerant to the islets and remained insulitis free in the long-term range, as do parental hINS transgenic mice (10). Upon immunization against hPPI or hPPI peptides, YES mice developed CD8+ T-cell IFN-γ responses to a diverse repertoire of HLA-A*02:01–restricted peptides but remained insulitis free. On the basis of evidence that enterovirus infections are involved in T1D development (30), we addressed whether it was possible to induce diabetes in YES mice. Upon pI:C injections, YES mice developed acute T1D. Diabetes-free mice were refractory to ultimate T1D development upon a second set of pI:C injections or hPPI immunization, suggesting an on/off mechanism in T1D triggering (31,32). Individual responses were observed against a set of hPPI peptides that partially overlaps with peptides that induced CD8+ T-cell responses upon hPPI immunization. Significant responses were seen against leader sequence hPPI6–14 and hPPI15–24 peptides, which we previously reported as driving significant expansions of CD8+ T cells in patients with recent-onset T1D, pointing to the relevance of the YES model to human T1D (16,23,33).

In contrast with class I–restricted epitopes, HLA-DQ8–restricted CD4+ T-cell responses remain ill-defined in man. IFN-γ CD4+ T-cell responses observed after immunization of YES mice against hPPI covered a wide spectrum of peptides scattered along the hPPI sequence, indicating the absence of a prevalent response. Proliferative CD4+ T-cell responses were observed against a more restricted set of peptides overlapping the B-chain and C-peptide sequences. CD4+ T-cells clones obtained from pancreatic infiltrates of a patient with T1D have been characterized as recognizing two HLA-DQ8–restricted C-peptide epitopes that were overlapping with peptide hPPI61–76 in our model (34). Upon induction of T1D by pI:C, significant CD4+ T-cell proliferative responses were observed against hPPI61–76 and against hPPI peptides that were only partially overlapping with responses induced by immunization of YES mice against hPPI. The partial dissociation of IFN-γ and proliferative responses on one side and of responses induced by immunization against hPPI and upon induction of diabetes by pI:C on the other side was not unexpected. Adjuvant-mediated immunization and pI:C indeed involve different pathways in antigen-presenting cells and CD4+ T-cell activation (3537).

Mechanisms inducing T1D through the loss of immune tolerance to β-cells remain elusive. Our data point to the importance of islet-environment interactions through signals carried by pattern recognition receptors (38,39) and TLRs (toll-like receptors) in the induction of T1D through innate immune upregulation (40,41). This is reminiscent of data involving T1D induction by Coxsackie B4 virus (32,42). The IFN induced with the helicase C domain I (IFIH1/MDA5) gene associates with T1D while mediating the early IFN response to viral RNA (43). Using a TLR3 agonist (44), we induced T1D in YES mice along with a T-cell infiltrate. The β-cells from pI:C-treated diabetic mice showed increased TLR3 expression. On the basis of data indicating TLR3 and MDA5 expression in the islets in vitro (24,45), we analyzed TLR3 expression after pI:C treatment. pI:C itself had a direct effect on β-cells in boosting speckled TLR3 expression predominantly in β-cells. Indeed, a key role of plasmacytoid DCs has been reported in the NOD mouse (32,46).

In conclusion, the YES mouse is a new model to characterize HLA-A*02:01– and HLA-DQ8–restricted hPPI epitopes involved in T1D and study mechanisms of T1D induction and heterogeneity in humans. This model will provide a new avenue to evaluate immune tolerance strategies that may directly apply to immunotherapy of T1D in humans.

Acknowledgments. This work was performed within the Département Hospitalo Universitaire Autoimmune and Hormonal Diseases project. The authors acknowledge Sébastien Jacques and the genotyping platform at Cochin Institute, Paris, France. The authors also acknowledge Raphaël Scharfmann for reading the manuscript and Latif Rachdi and Virginie Aiello-Lorenzo for technical assistance in evaluating β-cell mass and PDX1 staining (all from Cochin Institute, Paris, France).

Funding. This work was supported by Agence Nationale de la Recherche grants R11189KK-RPV11189KKA and ANR2010-Biot-00801 and European Foundation for the Study of Diabetes grant 1-2008-106.

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

Author Contributions. S.L. performed experiments, was involved in discussions, and contributed to writing the manuscript. S.G. performed metabolic experiments. A.G. performed DAB staining. F.Let. was involved in discussions and manuscript editing. B.B. addressed the question of cortisol stress. P.N. performed big data analysis. E.P., M.V., and F.Lem. were involved in Affymetrix genotyping array discussion and manuscript editing. C.B. designed experiments, chaired discussions, and wrote the manuscript. C.B. is the guarantor of this work and, as such, had full access to all data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.

1.
Donath
MY
,
Hess
C
,
Palmer
E
.
What is the role of autoimmunity in type 1 diabetes? A clinical perspective
.
Diabetologia
2014
;
57
:
653
655
[PubMed]
2.
Bluestone
JA
,
Herold
K
,
Eisenbarth
G
.
Genetics, pathogenesis and clinical interventions in type 1 diabetes
.
Nature
2010
;
464
:
1293
1300
[PubMed]
3.
Mahil
SK
,
Catapano
M
,
Di Meglio
P
, et al
.
An analysis of IL-36 signature genes and individuals with IL1RL2 knockout mutations validates IL-36 as a psoriasis therapeutic target
.
Sci Transl Med
2017
;
9
:eaan2514
4.
Thébault-Baumont
K
,
Dubois-Laforgue
D
,
Krief
P
, et al
.
Acceleration of type 1 diabetes mellitus in proinsulin 2-deficient NOD mice
.
J Clin Invest
2003
;
111
:
851
857
[PubMed]
5.
Nakayama
M
,
Abiru
N
,
Moriyama
H
, et al
.
Prime role for an insulin epitope in the development of type 1 diabetes in NOD mice
.
Nature
2005
;
435
:
220
223
[PubMed]
6.
Ziegler
AG
,
Nepom
GT
.
Prediction and pathogenesis in type 1 diabetes
.
Immunity
2010
;
32
:
468
478
[PubMed]
7.
Moriyama
H
,
Abiru
N
,
Paronen
J
, et al
.
Evidence for a primary islet autoantigen (preproinsulin 1) for insulitis and diabetes in the nonobese diabetic mouse
.
Proc Natl Acad Sci U S A
2003
;
100
:
10376
10381
[PubMed]
8.
Jones
EY
,
Fugger
L
,
Strominger
JL
,
Siebold
C
.
MHC class II proteins and disease: a structural perspective
.
Nat Rev Immunol
2006
;
6
:
271
282
[PubMed]
9.
Noble
JA
,
Valdes
AM
,
Varney
MD
, et al.;
Type 1 Diabetes Genetics Consortium
.
HLA class I and genetic susceptibility to type 1 diabetes: results from the Type 1 Diabetes Genetics Consortium
.
Diabetes
2010
;
59
:
2972
2979
[PubMed]
10.
Bucchini
D
,
Ripoche
MA
,
Stinnakre
MG
, et al
.
Pancreatic expression of human insulin gene in transgenic mice
.
Proc Natl Acad Sci U S A
1986
;
83
:
2511
2515
[PubMed]
11.
Pascolo
S
,
Bervas
N
,
Ure
JM
,
Smith
AG
,
Lemonnier
FA
,
Pérarnau
B
.
HLA-A2.1-restricted education and cytolytic activity of CD8(+) T lymphocytes from beta2 microglobulin (beta2m) HLA-A2.1 monochain transgenic H-2Db beta2m double knockout mice
.
J Exp Med
1997
;
185
:
2043
2051
[PubMed]
12.
Madsen
L
,
Labrecque
N
,
Engberg
J
, et al
.
Mice lacking all conventional MHC class II genes
.
Proc Natl Acad Sci U S A
1999
;
96
:
10338
10343
[PubMed]
13.
Nabozny
GH
,
Baisch
JM
,
Cheng
S
, et al
.
HLA-DQ8 transgenic mice are highly susceptible to collagen-induced arthritis: a novel model for human polyarthritis
.
J Exp Med
1996
;
183
:
27
37
[PubMed]
14.
Jain
R
,
Gray
DH
.
Isolation of thymic epithelial cells and analysis by flow cytometry
.
Curr Protoc Immunol
2014
;
107
:
3.26.1
3.26.15
15.
Villaseñor
J
,
Besse
W
,
Benoist
C
,
Mathis
D
.
Ectopic expression of peripheral-tissue antigens in the thymic epithelium: probabilistic, monoallelic, misinitiated
.
Proc Natl Acad Sci U S A
2008
;
105
:
15854
15859
[PubMed]
16.
Toma
A
,
Haddouk
S
,
Briand
JP
, et al
.
Recognition of a subregion of human proinsulin by class I-restricted T cells in type 1 diabetic patients
.
Proc Natl Acad Sci U S A
2005
;
102
:
10581
10586
[PubMed]
17.
Rachdi
L
,
Aïello
V
,
Duvillié
B
,
Scharfmann
R
.
L-leucine alters pancreatic β-cell differentiation and function via the mTor signaling pathway
.
Diabetes
2012
;
61
:
409
417
[PubMed]
18.
Kurasawa
K
,
Sakamoto
A
,
Maeda
T
, et al
.
Short-term administration of anti-L3T4 MoAb prevents diabetes in NOD mice
.
Clin Exp Immunol
1993
;
91
:
376
380
[PubMed]
19.
Grcević
D
,
Lee
SK
,
Marusić
A
,
Lorenzo
JA
.
Depletion of CD4 and CD8 T lymphocytes in mice in vivo enhances 1,25-dihydroxyvitamin D3-stimulated osteoclast-like cell formation in vitro by a mechanism that is dependent on prostaglandin synthesis
.
J Immunol
2000
;
165
:
4231
4238
[PubMed]
20.
Fan
Y
,
Rudert
WA
,
Grupillo
M
,
He
J
,
Sisino
G
,
Trucco
M
.
Thymus-specific deletion of insulin induces autoimmune diabetes
.
EMBO J
2009
;
28
:
2812
2824
[PubMed]
21.
Godfrey
DI
,
Kennedy
J
,
Suda
T
,
Zlotnik
A
.
A developmental pathway involving four phenotypically and functionally distinct subsets of CD3-CD4-CD8- triple-negative adult mouse thymocytes defined by CD44 and CD25 expression
.
J Immunol
1993
;
150
:
4244
4252
[PubMed]
22.
Muggeo
M
,
Ginsberg
BH
,
Roth
J
,
Neville
DM
 Jr
.,
De Meyts
P
,
Kahn
CR
.
The insulin receptor in vertebrates is functionally more conserved during evolution than insulin itself
.
Endocrinology
1979
;
104
:
1393
1402
[PubMed]
23.
Luce
S
,
Lemonnier
F
,
Briand
JP
, et al
.
Single insulin-specific CD8+ T cells show characteristic gene expression profiles in human type 1 diabetes
.
Diabetes
2011
;
60
:
3289
3299
[PubMed]
24.
Wen
L
,
Peng
J
,
Li
Z
,
Wong
FS
.
The effect of innate immunity on autoimmune diabetes and the expression of Toll-like receptors on pancreatic islets
.
J Immunol
2004
;
172
:
3173
3180
[PubMed]
25.
Harbige
J
,
Eichmann
M
,
Peakman
M
.
New insights into non-conventional epitopes as T cell targets: the missing link for breaking immune tolerance in autoimmune disease
?
J Autoimmun
2017
;
84
:
12
20
[PubMed]
26.
Marron
MP
,
Graser
RT
,
Chapman
HD
,
Serreze
DV
.
Functional evidence for the mediation of diabetogenic T cell responses by HLA-A2.1 MHC class I molecules through transgenic expression in NOD mice
.
Proc Natl Acad Sci U S A
2002
;
99
:
13753
13758
[PubMed]
27.
Racine
JJ
,
Stewart
I
,
Ratiu
J
, et al
.
Improved murine MHC-deficient HLA transgenic NOD mouse models for type 1 diabetes therapy development
.
Diabetes
2018
;
67
:
923
935
[PubMed]
28.
Lamothe
B
,
Baudry
A
,
Desbois
P
, et al
.
Genetic engineering in mice: impact on insulin signalling and action
.
Biochem J
1998
;
335
:
193
204
[PubMed]
29.
Pugliese
A
,
Brown
D
,
Garza
D
, et al
.
Self-antigen-presenting cells expressing diabetes-associated autoantigens exist in both thymus and peripheral lymphoid organs
[published correction appears in J Clin Invest 2006;116:548].
J Clin Invest
2001
;
107
:
555
564
[PubMed]
30.
Ghazarian
L
,
Diana
J
,
Simoni
Y
,
Beaudoin
L
,
Lehuen
A
.
Prevention or acceleration of type 1 diabetes by viruses
[published correction appears in Cell Mol Life Sci 2013;70:257].
Cell Mol Life Sci
2013
;
70
:
239
255
31.
Serreze
DV
,
Post
CM
,
Chapman
HD
,
Johnson
EA
,
Lu
B
,
Rothman
PB
.
Interferon-gamma receptor signaling is dispensable in the development of autoimmune type 1 diabetes in NOD mice
.
Diabetes
2000
;
49
:
2007
2011
[PubMed]
32.
Ghazarian
L
,
Diana
J
,
Beaudoin
L
, et al
.
Protection against type 1 diabetes upon Coxsackievirus B4 infection and iNKT-cell stimulation: role of suppressive macrophages
.
Diabetes
2013
;
62
:
3785
3796
[PubMed]
33.
Toma
A
,
Laïka
T
,
Haddouk
S
, et al
.
Recognition of human proinsulin leader sequence by class I-restricted T-cells in HLA-A*0201 transgenic mice and in human type 1 diabetes
.
Diabetes
2009
;
58
:
394
402
[PubMed]
34.
Pathiraja
V
,
Kuehlich
JP
,
Campbell
PD
, et al
.
Proinsulin-specific, HLA-DQ8, and HLA-DQ8-transdimer-restricted CD4+ T cells infiltrate islets in type 1 diabetes
.
Diabetes
2015
;
64
:
172
182
[PubMed]
35.
Nakamura
T
,
Moriguchi
R
,
Kogure
K
,
Harashima
H
.
Incorporation of polyinosine-polycytidylic acid enhances cytotoxic T cell activity and antitumor effects by octaarginine-modified liposomes encapsulating antigen, but not by octaarginine-modified antigen complex
.
Int J Pharm
2013
;
441
:
476
481
[PubMed]
36.
Francica
JR
 Jr
.,
Zak
DE
,
Linde
C
, et al
.
Innate transcriptional effects by adjuvants on the magnitude, quality, and durability of HIV envelope responses in NHPs
.
Blood Adv
2017
;
1
:
2329
2342
[PubMed]
37.
Speth
MT
,
Repnik
U
,
Müller
E
, et al
.
Poly(I:C)-encapsulating nanoparticles enhance innate immune responses to the tuberculosis vaccine Bacille Calmette-Guérin (BCG) via synergistic activation of innate immune receptors
.
Mol Pharm
2017
;
14
:
4098
4112
[PubMed]
38.
Hutton
MJ
,
Soukhatcheva
G
,
Johnson
JD
,
Verchere
CB
.
Role of the TLR signaling molecule TRIF in β-cell function and glucose homeostasis
.
Islets
2010
;
2
:
104
111
[PubMed]
39.
Giarratana
N
,
Penna
G
,
Amuchastegui
S
,
Mariani
R
,
Daniel
KC
,
Adorini
L
.
A vitamin D analog down-regulates proinflammatory chemokine production by pancreatic islets inhibiting T cell recruitment and type 1 diabetes development
.
J Immunol
2004
;
173
:
2280
2287
[PubMed]
40.
Alkanani
AK
,
Hara
N
,
Lien
E
, et al
.
Induction of diabetes in the RIP-B7.1 mouse model is critically dependent on TLR3 and MyD88 pathways and is associated with alterations in the intestinal microbiome
.
Diabetes
2014
;
63
:
619
631
[PubMed]
41.
Swiecki
M
,
McCartney
SA
,
Wang
Y
,
Colonna
M
.
TLR7/9 versus TLR3/MDA5 signaling during virus infections and diabetes
.
J Leukoc Biol
2011
;
90
:
691
701
[PubMed]
42.
McCall
KD
,
Thuma
JR
,
Courreges
MC
, et al
.
Toll-like receptor 3 is critical for coxsackievirus B4-induced type 1 diabetes in female NOD mice
.
Endocrinology
2015
;
156
:
453
461
[PubMed]
43.
Nejentsev
S
,
Walker
N
,
Riches
D
,
Egholm
M
,
Todd
JA
.
Rare variants of IFIH1, a gene implicated in antiviral responses, protect against type 1 diabetes
.
Science
2009
;
324
:
387
389
[PubMed]
44.
Moriyama
H
,
Wen
L
,
Abiru
N
, et al
.
Induction and acceleration of insulitis/diabetes in mice with a viral mimic (polyinosinic-polycytidylic acid) and an insulin self-peptide
.
Proc Natl Acad Sci U S A
2002
;
99
:
5539
5544
[PubMed]
45.
Lincez
PJ
,
Shanina
I
,
Horwitz
MS
.
Reduced expression of the MDA5 Gene IFIH1 prevents autoimmune diabetes
.
Diabetes
2015
;
64
:
2184
2193
[PubMed]
46.
Diana
J
,
Brezar
V
,
Beaudoin
L
, et al
.
Viral infection prevents diabetes by inducing regulatory T cells through NKT cell-plasmacytoid dendritic cell interplay
.
J Exp Med
2011
;
208
:
729
745
[PubMed]
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 http://www.diabetesjournals.org/content/license.

Supplementary data