Because regulatory T-cell (Treg) development can be induced by the same agonist self-antigens that induce negative selection, perturbation of apoptosis will affect both negative selection and Treg development. But how the processes of thymocyte deletion versus Treg differentiation bifurcate and their relative importance for tolerance have not been studied in spontaneous organ-specific autoimmune disease. We addressed these questions by removing a critical mediator of thymocyte deletion, BIM, in the NOD mouse model of autoimmune diabetes. Despite substantial defects in the deletion of autoreactive thymocytes, BIM-deficient NOD (NODBim−/−) mice developed less insulitis and were protected from diabetes. BIM deficiency did not impair effector T-cell function; however, NODBim−/− mice had increased numbers of Tregs, including those specific for proinsulin, in the thymus and peripheral lymphoid tissues. Increased levels of Nur77, CD5, GITR, and phosphorylated IκB-α in thymocytes from NODBim−/− mice suggest that autoreactive cells receiving strong T-cell receptor signals that would normally delete them escape apoptosis and are diverted into the Treg pathway. Paradoxically, in the NOD model, reduced thymic deletion ameliorates autoimmune diabetes by increasing Tregs. Thus, modulating apoptosis may be one of the ways to increase antigen-specific Tregs and prevent autoimmune disease.

The immune system has evolved to allow robust responses against a diverse range of pathogens while preserving self-tolerance to prevent immunopathology. Developing self-reactive thymocytes that express T-cell receptors (TCRs) that bind with high affinity to MHC/self-peptide complexes are usually deleted by apoptosis in the thymus, a process termed “deletion” or “recessive tolerance” (1). A complementary mechanism of immune tolerance, termed “dominant tolerance,” involves the generation of FoxP3-expressing regulatory T cells (Tregs) capable of suppressing the activation of autoreactive, conventional T cells that fail to undergo negative selection in the thymus (e.g., because the cognate self-antigen is not expressed and presented in the thymus) (2). The importance of Tregs and thymic deletion in preventing autoimmunity is illustrated in immunodysregulation polyendocrinopathy enteropathy X-linked syndrome and in autoimmune polyglandular syndrome type 1, respectively, both of which result in widespread autoimmune disease, including type 1 diabetes (3,4).

Tregs have considerable therapeutic potential in autoimmune diseases such as type 1 diabetes. Tregs that express TCR specific for self-antigen can home, along with autoreactive effector T cells, to antigen-expressing target peripheral tissues, such as pancreatic islets and their draining lymph nodes. At these sites, they can suppress not only autoreactive effector cells with TCR specificities for the same antigen they recognize but also others, a phenomenon called bystander suppression.

The signaling events that regulate and presumably distinguish between thymic deletion and Treg development remain incompletely defined. Apoptosis is critical for deletion of self-reactive T cells in both the thymus and the peripheral lymphoid tissues and for thymic Treg development. BIM is a proapoptotic BH3-only protein required for deletion of autoreactive thymocytes (5). BIM-deficient mice not only have defects in T-cell deletion (6) but also have increased numbers of FoxP3-expressing Tregs (7). How BIM controls the development of Tregs in BIM-deficient mice and the functional significance of increased Tregs in BIM-deficient mice have not been studied.

Although it has been demonstrated that high-affinity TCR interaction with self-antigen is required to trigger both thymic deletion and Treg differentiation, these studies involved transgenic expression of antigen or peptide administration or used TCR transgenic T cells present at high clonal frequencies (4,810). However, other data suggest that thymic Treg development uses a limited antigenic niche (11,12), implying that models using ubiquitous antigen presentation or TCR transgenic T cells at high clonal frequencies may not reflect normal events. Therefore, it is important to study autoreactive T-cell deletion and Treg differentiation in a disease with a polyclonal T-cell repertoire and natural self-antigens expressed at physiological levels. To this end, we studied the impact of removing a critical mediator of thymocyte apoptosis, BIM, on thymic deletion and Treg differentiation in the NOD mouse, an animal model of type 1 diabetes (13). In this model, mice express natural self-antigens and have a polyclonal T-cell repertoire. The data show that reducing thymic deletion by disabling thymic apoptosis ameliorates autoimmune diabetes by increasing Tregs.

Mice

BIM-deficient C57BL/6 mice (6) were backcrossed onto the NOD/Lt genetic background for >10 generations. NOD.BDC2.5 mice were provided by D. Mathis and C. Benoist (Harvard University). NOD.Rag1−/− and NOD mice expressing GFP under control of the FoxP3 promoter were obtained from The Jackson Laboratory. C57BL/6 and NOD/Lt mice were obtained from the Walter and Eliza Hall Institute animal breeding facility (Kew, VIC, Australia). Diabetes monitoring and insulitis scoring were performed as described previously (14). Mice with two consecutive blood glucose readings >15 mmol/L were considered diabetic. All animal studies were conducted under specific pathogen-free conditions at St. Vincent’s Institute (Melbourne, VIC, Australia) following the guidelines of the institutional animal ethics committee.

Genotyping

The Bim gene (Bcl2l11) lies within the Idd13 diabetes susceptibility locus. To confirm the genetic background of NODBim−/− mice and define the congenic interval encompassing the Bcl2l11 null allele, DNA was isolated from tail biopsy specimens from 10th generation backcrossed mice and genotyped by the Australian Genome Research Facility using the mouse 5K targeted genotyping array run on the Affymetrix GeneChip Scanner 3000 7G MegAllele system. To refine the 129/Sv-derived congenic interval encompassing the Bcl2l11 null allele, additional polymorphic markers (nucleotide repeats and single nucleotide polymorphisms [SNPs]) were identified using the Mouse Genome Informatics and National Center for Biotechnology Information databases. DNA was genotyped by standard PCR and gel electrophoresis methods (selected nucleotide repeats) and by The Centre for Applied Genomics (The Hospital for Sick Children, Toronto, ON, Canada) using the Illumina medium-density linkage panel (1,449 SNPs), which contained SNPs that further refined the congenic interval. NODBim−/− mice were of NOD genotype across the whole genome except for a region on chromosome 2 encompassing the Bcl2l11 locus. The congenic interval is 129-derived between, but not including, D2mit277 (Chr2:123,255,831) and D2mit338 (Chr2:130,655,046), and no B6 alleles were detected within this interval. The 129-congenic interval is unlikely to contribute to diabetes protection because 129 mice are predicted to harbor a susceptibility, not a resistance, allele in this region of chromosome 2 (15).

Antibodies and Flow Cytometry

Antibodies used were anti-CD4-PerCp-Cy5.5 (RM4-5), anti-CD3 (145-2C11), anti-CD5 (clone 53-7.3), and anti-CD24 (clone M1/69) all conjugated to fluorescein isothiocyanate (FITC); anti-CD44-AlexaFluor 700 (1M7), anti-CD69-allophycocyanin (H1.2F3), and anti-62L-allophycocyanin-Cy7 (MEL-14) (all from BD Biosciences, San Jose, CA); anti-CD11c (N418), anti-B220 (RA3-6B2), anti-CD11b (M1/70), and anti-F4/80 (BM8) all conjugated to eFluor 450 (all from BioLegend, San Diego, CA); and anti-CD8a-Pacific Orange (5H10) (Invitrogen, Carlsbad, CA). FITC-conjugated antibodies specific for individual TCR Vβ chains were purchased as a kit from BD Biosciences. Anti-FoxP3 (FJK-16s), anti-Nur77 (12.14) conjugated to phycoerythrin, anti-Helios-FITC (22F6), and anti-GITR-allophycocyanin (DTA-1) were purchased from eBioscience (San Diego, CA). Analysis was performed on a BD LSRFortessa (BD Biosciences, Franklin Lakes, NJ) using FlowJo (Tree Star, Ashland, OR) software. Intracellular staining was performed according to the manufacturer’s specifications using the BD Cytofix/Cytoperm Plus kit (BD Biosciences, San Jose, CA).

MHC Tetramers and Magnetic Bead–Based Cell Enrichment

The phycoerythrin-conjugated I-A (g7) tetramers, insulin B9–23 (HLVERLYLVCGGEG) (16) and control CLIP (AMKRHGLDNYRGYSL), were provided by the National Institutes of Health Tetramer Core Facility. Tetramer staining and magnetic bead–based cell enrichment were performed as described previously (17).

Western Blotting

Whole thymus or sorted thymocyte subsets were homogenized in lysis buffer containing 50 mmol/L Tris-HCl (pH 8), 150 mmol/L NaCl, 0.5% Triton X-100, and protease inhibitor cocktail (Sigma). Proteins (8 μg/lane) were resolved by SDS-PAGE and transferred using standard procedures to nitrocellulose. Monoclonal mouse antibodies to nuclear factor-κB (NF-κB) p65, phosphorylated IκB-α, and β-actin (Santa Cruz Biotechnology, Dallas, TX), and horseradish peroxidase–conjugated anti-mouse Ig antibodies were used.

Cell Proliferation and Treg Suppression Assays

For carboxyfluorescein succinimidyl ester (CFSE) dilution assay, T cells were labeled with CFSE as previously described (18). For Treg suppression assays, sorted CD25GITRCD4+ or CD4+FoxP3CD25 were used as responder T cells, and CD4+GITR+ or CD4+FoxP3+ cells were used as Tregs. CFSE-labeled responder T cells (5 × 104) were cultured for 72 h with γ-irradiated (4,000 rad) splenocytes (5 × 104) and anti-CD3 monoclonal antibody 1 μg/mL (145-2C11; Bio X Cell) in the presence or absence of the indicated ratio of Tregs.

Statistical Analysis

Data are presented as mean ± SD. Data analysis was performed using GraphPad Prism software (GraphPad Software, San Diego, CA), and the Mann-Whitney test was used to assess statistical significance. The log-rank test was used to perform survival curve analysis.

NODBim−/− Mice Have Impaired Deletion of Autoreactive T Cells

NODBim−/− mice had higher percentages of CD4+CD8 and CD8+CD4 thymocytes and a reduction in CD4+CD8+ thymocytes compared with control NOD mice (Fig. 1A, B, E, and G). The total splenocyte number was significantly increased in NODBim−/− mice, whereas the proportions of CD4+ T cells, CD8+ T cells, and B cells were not statistically different from control NOD mice (Fig. 1A, B, E, and G). These findings are similar to Bim−/− mice on other genetic backgrounds (5,6,19) and suggest impaired thymocyte deletion.

Figure 1

Thymic deletion of autoreactive thymocytes is impaired in NODBim−/− mice. AD: Representative FACS plots of cell suspensions from thymus and spleen of 8–12-week-old NODBim−/− and NODBim+/+ mice stained with the indicated surface marker–specific antibodies. Numbers indicate the percentages of live cells (A and B) or the percentages of Vβ3+ cells after gating on the CD4 single-positive subset (C). D: The numbers of insulin B9–23 tetramer-positive CD4+ T cells after magnetic bead–based enrichment and gating on the CD4 single-positive subset. Numbers indicate the total number of live cells in the indicated organ. Plots represent six to nine mice per strain. EH: Pooled data of the absolute number of cells from NODBim−/−, NODBim+/−, and NODBim+/+ mice are mean ± SD. *P < 0.05, **P < 0.005. Ins, insulin; PLO, peripheral lymphoid organ.

Figure 1

Thymic deletion of autoreactive thymocytes is impaired in NODBim−/− mice. AD: Representative FACS plots of cell suspensions from thymus and spleen of 8–12-week-old NODBim−/− and NODBim+/+ mice stained with the indicated surface marker–specific antibodies. Numbers indicate the percentages of live cells (A and B) or the percentages of Vβ3+ cells after gating on the CD4 single-positive subset (C). D: The numbers of insulin B9–23 tetramer-positive CD4+ T cells after magnetic bead–based enrichment and gating on the CD4 single-positive subset. Numbers indicate the total number of live cells in the indicated organ. Plots represent six to nine mice per strain. EH: Pooled data of the absolute number of cells from NODBim−/−, NODBim+/−, and NODBim+/+ mice are mean ± SD. *P < 0.05, **P < 0.005. Ins, insulin; PLO, peripheral lymphoid organ.

Close modal

In NOD mice, T cells bearing the TCR Vβ3 chain represent only 0.4% of the CD4+ T cells because of clonal deletion of most Vβ3+ immature thymocytes by the mammary tumor virus-3 (Mtv-3) superantigen presented by class II MHC (20). T cells bearing the TCR Vβ3 chain were significantly increased in the spleen and thymus of NODBim−/− mice (Fig. 1C and F and Supplementary Fig. 1A), indicating that loss of BIM causes a defect in superantigen-mediated deletion within a polyclonal T-cell repertoire on the NOD background.

We then assayed the numbers of insulin B9–23–specific CD4+ T cells, a population of T cells known to play a central role in the pathogenesis of type 1 diabetes (21). Proinsulin 2 is expressed in the mouse thymus (22,23), and NOD mice deficient in proinsulin 2 develop accelerated diabetes, suggesting that high-affinity insulin-specific T cells are tolerized in the normal NOD thymus (24). Using the method of staining with tetramer followed by enrichment using magnetic beads (17), we detected rare insulin B9–23–specific CD4+ T cells in the peripheral lymphoid organs and thymus of NOD mice. The small number of insulin B9–23–specific CD4+ T cells (16) in the thymus of control NOD mice suggests that deletion of thymocytes with this specificity is stringent but incomplete. This population was significantly increased (P < 0.05) in NODBim−/− mice (Fig. 1D and H). These data demonstrate that BIM deficiency impairs deletion of autoreactive T cells in autoimmune disease–prone NOD mice.

NODBim−/− Mice Have Reduced Insulitis and Are Completely Protected From Diabetes

We analyzed the effect of impaired deletion on the development of autoimmunity in NODBim−/− mice. NODBim−/− mice developed glomerulonephritis as shown by mesangial deposits of immune complexes (Supplementary Fig. 1B) consistent with the systemic lupus erythematosus–like disease observed in BIM-deficient mice on other genetic backgrounds. By contrast, insulitis was significantly reduced in NODBim−/− mice (Fig. 2A and B). Remarkably, NODBim−/− mice were completely protected from diabetes, and even heterozygous NODBim+/− mice were partially protected from this disease (Fig. 2C). This was surprising because we had expected that the defective deletion caused by BIM deficiency would accelerate diabetes.

Figure 2

NODBim−/− mice are protected from diabetes and develop less severe insulitis compared with control NOD mice. A: More than 100 pancreatic islets from each of the indicated strains of mice were evaluated for lymphocytic infiltration at 10–15 weeks of age (n = 6–11 mice per strain). The y-axis represents the mean ± SD of the insulitis score. *P < 0.05. B: Insulitis stages at 10–15 weeks of age (n = 6–11 mice per strain). C: Incidence of diabetes in NODBim−/− (n = 16), NODBim+/− (n = 21), and NODBim+/+ (n = 15) mice. NODBim−/− vs. NODBim+/+P < 0.0001 and NODBim+/− vs. NODBim−/− or NODBim+/+P < 0.05. D: 2 × 107 splenocytes from newly diabetic NOD mice were injected intravenously into 6–8-week-old γ-irradiated (900 rad) NODBim−/− or NOD recipients. Groups were not statistically different.

Figure 2

NODBim−/− mice are protected from diabetes and develop less severe insulitis compared with control NOD mice. A: More than 100 pancreatic islets from each of the indicated strains of mice were evaluated for lymphocytic infiltration at 10–15 weeks of age (n = 6–11 mice per strain). The y-axis represents the mean ± SD of the insulitis score. *P < 0.05. B: Insulitis stages at 10–15 weeks of age (n = 6–11 mice per strain). C: Incidence of diabetes in NODBim−/− (n = 16), NODBim+/− (n = 21), and NODBim+/+ (n = 15) mice. NODBim−/− vs. NODBim+/+P < 0.0001 and NODBim+/− vs. NODBim−/− or NODBim+/+P < 0.05. D: 2 × 107 splenocytes from newly diabetic NOD mice were injected intravenously into 6–8-week-old γ-irradiated (900 rad) NODBim−/− or NOD recipients. Groups were not statistically different.

Close modal

We next investigated whether protection from diabetes in NODBim−/− mice was due to protection of islets from death by apoptosis mediated by effector T cells. We transferred splenocytes from recently diabetic NOD mice into γ-irradiated NODBim−/− and control NOD mice (Fig. 2D). Both NOD and NODBim−/− recipients developed diabetes at similar rates. The transfer of activated diabetogenic T cells bypasses T-cell priming and activation in the pancreatic lymph node (PLN). To test whether these were affected by loss of BIM, we transferred CFSE-labeled IGRP-specific naive T cells from NOD 8.3 mice into NOD and NODBim−/− mice. We and others have previously used CD8+ T cells from NOD 8.3 to assess the priming of autoreactive T cells in the PLN (18,25). Transferred CFSE-labeled naive IGRP-specific transgenic cells proliferated similarly in PLNs of NOD and NODBim−/− mice (Supplementary Fig. 1C and D), indicating that protection does not derive from impaired priming of autoreactive cells in NODBim−/− mice. These experiments collectively indicate that the protection observed in NODBim−/− mice was not due to β-cell resistance to autoimmune destruction.

It has been reported that BIM deficiency can protect from autoimmunity by affecting the function of effector T cells in a cell-intrinsic manner (26). We detected insulin B9–23–specific CD4+ T cells in the thymus and IGRP206–214-specific and insulin B15–23–specific CD8+ T cells within the insulitis of NODBim−/− mice (Supplementary Fig. 1E), demonstrating that the protection from diabetes seen in NODBim−/− mice is not due to the absence of autoreactive T cells. Moreover, the effector T cells in NODBim−/− mice were functional in the NOD.BDC2.5 model (as discussed later; see Fig. 5C). These experiments demonstrate that the effector T cells in NODBim−/− mice are functional.

NODBim−/− Mice Have Increased Numbers of Antigen-Specific Tregs

Thymocytes expressing TCRs that recognize self-peptide/MHC complexes with sufficient high affinity can also differentiate into Tregs (27). We examined whether the self-reactive T cells that escape deletion in NODBim−/− mice preferentially differentiate into Tregs. The percentages and absolute numbers of CD4+FoxP3+ T cells in the spleen, thymus, PLNs, and inguinal lymph nodes were substantially increased in NODBim−/− mice compared with control NOD mice (Fig. 3A and E). The proportion of Tregs was also increased in the islet-infiltrating cells of NODBim−/− mice compared with NOD mice (Supplementary Fig. 1F). Because NODBim−/− had less insulitis, we could not compare the absolute number of islet-infiltrating Tregs between NOD and NODBim−/− mice. More than 90% of the CD4+FoxP3+ T cells in both NOD and NODBim−/− mice also expressed the cell surface marker GITR (Fig. 3B and Supplementary Fig. 2C). As shown previously in C57BL/6 mice, there were increased CD4+FoxP3+CD25 T cells in NODBim−/− mice compared with wild-type NOD mice (7) (Fig. 3C).

Figure 3

NODBim−/− mice have increased numbers of antigen-specific Tregs. AD: Representative plots showing staining of splenic and thymic CD4 single-positive subsets of T cells from 8–12-week-old NODBim−/− and NODBim+/+ mice (n = 9–13 per group) for expression of FoxP3, GITR, CD25, or Helios. Numbers represent percentages of cells contained within the gate. E: Pooled data of percentages of CD4+FoxP3+ T cells in the indicated organs of 8–12-week-old NODBim−/−, NODBim+/−, and NODBim+/+ mice (mean ± SD of n = 9–13 per group) as determined by intracellular FoxP3 staining and flow cytometry. *P < 0.05, **P < 0.005, ***P < 0.0005. ILN, inguinal lymph node.

Figure 3

NODBim−/− mice have increased numbers of antigen-specific Tregs. AD: Representative plots showing staining of splenic and thymic CD4 single-positive subsets of T cells from 8–12-week-old NODBim−/− and NODBim+/+ mice (n = 9–13 per group) for expression of FoxP3, GITR, CD25, or Helios. Numbers represent percentages of cells contained within the gate. E: Pooled data of percentages of CD4+FoxP3+ T cells in the indicated organs of 8–12-week-old NODBim−/−, NODBim+/−, and NODBim+/+ mice (mean ± SD of n = 9–13 per group) as determined by intracellular FoxP3 staining and flow cytometry. *P < 0.05, **P < 0.005, ***P < 0.0005. ILN, inguinal lymph node.

Close modal

The increase in Tregs was most prominent in the thymus of NODBim−/− mice, suggesting that BIM deficiency promotes an increase in natural Tregs. To further confirm that the excess FoxP3+ cells in NODBim−/− mice are derived from the thymus, we performed expression analysis for the natural Treg-specific marker Helios (28). Indeed, >85% of the Treg population in NODBim−/− mice expressed Helios (Fig. 3D).

Next, we investigated whether thymocytes bearing TCR Vβ3 chains and self-reactive insulin B9–23–specific CD4+ T cells that are normally deleted after encountering self-antigen in NOD mice differentiate preferentially into Tregs in NODBim−/− mice. We examined GFP+ (FoxP3-expressing) insulin B9–23–specific CD4+ T cells and CD4+ T cells bearing TCR Vβ3 chains in transgenic NOD and NODBim−/− mice that express GFP under control of the FoxP3 promoter (NODBim−/−FoxP3-GFP mice) (29). First, we found that FoxP3 (GFP)–expressing T cells bearing TCR Vβ3 and insulin B9–23–specific CD4+ thymocytes were increased in NODBim−/−FoxP3-GFP mice compared with NODFoxP3-GFP mice (Fig. 4A and B). Second, because the majority of FoxP3+ T cells also express the cell surface marker GITR, we also used GITR as a marker of Tregs. We found that GITR-expressing Tregs were enriched in the fraction of T cells bearing TCR Vβ3 chains, suggesting that autoreactive T cells are enriched within the Treg population in NODBim−/− mice (Supplementary Fig. 2A). Incomplete deletion of insulin B9–23–specific CD4+ thymocytes in NOD mice yielded both GITR+ and GITR insulin-specific T cells (Supplementary Fig. 2B). Of note, insulin B9–23–specific CD4+ T cells were significantly increased (P < 0.05) in NODBim−/− mice, and these insulin B9–23–specific CD4+ T cells were enriched for GITR-expressing Tregs (Supplementary Fig. 2B). These results indicate that impaired deletion in NODBim−/− mice diverts most autoreactive T cells into Tregs.

Figure 4

The Tregs in NODBim−/− mice are functional. A: Thymocytes (n = 6) from transgenic NOD, NODBim+/−, or NODBim−/− mice that express GFP under control of the FoxP3 promoter were analyzed by first gating on live DP or CD4 single-positive cells for the proportion of Vβ3 expressing CD4 thymocytes and were further analyzed for the expression of GFP by the gated cells. Numbers indicate the absolute number of cells in the indicated gate in an individual mouse. B: Cells from thymi or peripheral lymphoid organs (PLOs) (pooled spleen and nonpancreatic lymph nodes) (n = 6 in each strain) of transgenic NOD, NODBim+/−, or NODBim−/− mice that express GFP under control of the FoxP3 promoter were enriched for insulin B9–23 tetramer-positive cells and then analyzed by gating on live CD4 single-positive cells for the proportion of CD4+ tetramer-positive cells expressing GFP. Numbers indicate absolute number of cells in the indicated gate in an individual mouse. C: Proliferation of sorted CFSE-labeled CD4+CD25GITR T cells from NOD mice in response to stimulation with CD3 antibody when cultured with sorted CD4+GITR+ T cells from NODBim−/− or NOD mice. D: 2 × 107 splenocytes from newly diabetic NOD mice were transferred either alone (group A, ▲, n = 6) or mixed with 5 × 105 sorted CD4+GITR+ Tregs from either NODBim−/− (group B, ○, n = 6) or NOD (group C, ●, n = 6) mice into 6–8-week-old NODRag1−/− recipient animals. Group B vs. group C, P not significant; group A vs. group B or group C, P < 0.05.

Figure 4

The Tregs in NODBim−/− mice are functional. A: Thymocytes (n = 6) from transgenic NOD, NODBim+/−, or NODBim−/− mice that express GFP under control of the FoxP3 promoter were analyzed by first gating on live DP or CD4 single-positive cells for the proportion of Vβ3 expressing CD4 thymocytes and were further analyzed for the expression of GFP by the gated cells. Numbers indicate the absolute number of cells in the indicated gate in an individual mouse. B: Cells from thymi or peripheral lymphoid organs (PLOs) (pooled spleen and nonpancreatic lymph nodes) (n = 6 in each strain) of transgenic NOD, NODBim+/−, or NODBim−/− mice that express GFP under control of the FoxP3 promoter were enriched for insulin B9–23 tetramer-positive cells and then analyzed by gating on live CD4 single-positive cells for the proportion of CD4+ tetramer-positive cells expressing GFP. Numbers indicate absolute number of cells in the indicated gate in an individual mouse. C: Proliferation of sorted CFSE-labeled CD4+CD25GITR T cells from NOD mice in response to stimulation with CD3 antibody when cultured with sorted CD4+GITR+ T cells from NODBim−/− or NOD mice. D: 2 × 107 splenocytes from newly diabetic NOD mice were transferred either alone (group A, ▲, n = 6) or mixed with 5 × 105 sorted CD4+GITR+ Tregs from either NODBim−/− (group B, ○, n = 6) or NOD (group C, ●, n = 6) mice into 6–8-week-old NODRag1−/− recipient animals. Group B vs. group C, P not significant; group A vs. group B or group C, P < 0.05.

Close modal

Tregs in NODBim−/− Mice Are Functional

To examine whether the protection from diabetes in NODBim−/− mice could be due to increased antigen-specific Tregs, we tested the function of the Tregs. Sorted CD4+GITR+ T cells from NODBim−/− mice or CD4+FoxP3+ T cells from NODBim−/−FoxP3-GFP mice substantially diminished the proliferation of conventional CD4+ T cells in response to stimulation with CD3 agonist antibody in a standard in vitro T-cell suppression assay; their activity was comparable to that of Tregs from control NOD mice or NODFoxP3-GFP mice (Fig. 4C and Supplementary Fig. 2D). We did not observe a significant difference in the suppressive function between CD25hi and CD25lo FoxP3+CD4+ T cells from NODBim−/−FoxP3-GFP mice (Supplementary Fig. 2D). Furthermore, when sorted CD4+GITR+ Tregs from NOD or NODBim−/− mice were cotransferred with diabetogenic splenocytes, both delayed diabetes in NOD.Rag1−/− mice with similar efficacy. These experiments confirm that Tregs in NODBim−/− mice are functional (Fig. 4D).

To examine the function of effector T cells and Tregs in NODBim−/− mice, we used NOD.BDC2.5 mice. Even though sorted BDC2.5 TCR transgenic T cells are highly diabetogenic when transferred into NOD.SCID or NOD.Rag−/− recipients, NOD.BDC2.5 mice spontaneously develop diabetes only at a very low incidence. However, when NOD.BDC2.5 mice are crossed to mice with the Rag mutation and/or FoxP3 Scurfy mutation to eliminate Tregs, 100% of the resulting compound mutant animals develop diabetes rapidly, indicating that Tregs protect NOD.BDC2.5 mice from autoimmune diabetes (30). BIM-deficient NOD.BDC2.5 mice were completely protected from diabetes, and the percentages of CD4+FoxP3+ T cells were increased in these animals compared with NOD.BDC2.5 control mice (Fig. 5A and B). As expected, total spleen cells from NOD.BDC2.5 or BIM-deficient NOD.BDC2.5 mice did not transfer diabetes efficiently when injected into NOD.Rag1−/− mice: Only one of the six recipients receiving NOD.BDC2.5 spleen cells and none of the six recipients receiving BIM-deficient NOD.BDC2.5 spleen cells became diabetic. On the other hand, 5 × 105 sorted CD4+GITR T cells with high BDC2.5 TCR clonotype expression from NOD.BDC2.5 or BIM-deficient NOD.BDC2.5 mice transferred diabetes in all recipients tested (Fig. 5C). These experiments demonstrate that both Tregs and effector T cells in NODBim−/− mice are functional, consistent with the notion that protection from diabetes in NODBim−/− mice is due to increased antigen-specific Tregs.

Figure 5

The effector T cells in NODBim−/− mice are functional. A: Percentages of CD4+FoxP3+ T cells from the thymus or spleen of 8–10-week-old NOD.BDC2.5 and NOD.BDC2.5/Bim−/− mice (n = 6 per strain) were determined by intracellular FoxP3 staining and flow cytometry. B: Incidence of diabetes in NOD.BDC2.5 (n = 16) and NOD.BDC2.5/Bim−/− (n = 10), P < 0.005. C: Incidence of diabetes after 2 × 107 splenocytes from NOD.BDC2.5/Bim−/− (n = 6, ▼, group A) or NOD.BDC2.5 (n = 6, ▲, group B) or 5 × 105 sorted CD4+GITR splenocytes NOD.BDC2.5/Bim−/− (n = 6, ○, group C) or NOD.BDC2.5 (n = 6, ●, group D) mice were injected into 6–8-week-old NODRag1−/− recipient animals. Group A vs. group B, P not significant; group B vs. group C or group D, P < 0.005.

Figure 5

The effector T cells in NODBim−/− mice are functional. A: Percentages of CD4+FoxP3+ T cells from the thymus or spleen of 8–10-week-old NOD.BDC2.5 and NOD.BDC2.5/Bim−/− mice (n = 6 per strain) were determined by intracellular FoxP3 staining and flow cytometry. B: Incidence of diabetes in NOD.BDC2.5 (n = 16) and NOD.BDC2.5/Bim−/− (n = 10), P < 0.005. C: Incidence of diabetes after 2 × 107 splenocytes from NOD.BDC2.5/Bim−/− (n = 6, ▼, group A) or NOD.BDC2.5 (n = 6, ▲, group B) or 5 × 105 sorted CD4+GITR splenocytes NOD.BDC2.5/Bim−/− (n = 6, ○, group C) or NOD.BDC2.5 (n = 6, ●, group D) mice were injected into 6–8-week-old NODRag1−/− recipient animals. Group A vs. group B, P not significant; group B vs. group C or group D, P < 0.005.

Close modal

Increased TCR Stimulation Promotes Treg Development

Thymic deletion of developing T cells requires high-avidity TCR signaling. Of note, during their development in the thymus, Tregs are selected by stronger TCR signals compared with conventional CD4+ T cells (27,31). BIM deficiency may raise the threshold of TCR signaling that developing T cells in the thymus can survive, enabling the maturation of highly self-reactive T cells that would not normally be produced. Consistent with this hypothesis, we detected abnormally increased percentages of double-positive (DP) thymocytes expressing CD69 (an indicator of recent TCR stimulation) in NODBim−/− mice (Fig. 6A), indicating that more DP thymocytes survived after TCR ligation in NODBim−/− mice than in NOD mice. Thymocytes receiving strong TCR signals have been shown to rapidly upregulate CD25 and GITR, and these cells are enriched for Treg precursors (32). We found that the Treg precursors (GITR and CD25 expressing DP thymocytes) were markedly increased in NODBim−/− mice (Fig. 6A), indicating that the antigen-experienced surviving thymocytes in NODBim−/− mice might be diverted to develop into Tregs.

Figure 6

Increased TCR stimulation diverts thymocytes to develop into Tregs. A and B: Representative plots showing staining of thymocytes for FoxP3, GITR, CD5, CD69, and Nur77 in 8–12-week-old NODBim−/− and NODBim+/+ mice (n = 4–6 per group). Numbers represent percentages of cells contained within the gate. C: Representative histograms showing Nur77 expression in the indicated thymocyte population. NODFoxP3-GFP profiles (gray) superimposed on the profiles for NODBim−/−FoxP3-GFP mice (white). Values represent the mean fluorescence intensity (MFI) of n = 5 mice (aged 60–100 days) per group. D: Lysates from whole thymi were examined by Western blot for their content of total IκB-α; phosphorylated (i.e., activated) IκB-α (pIκBα); and, as a loading control, β-actin. Lane 1, C57BL/6Bim−/−; lane 2, C57BL/6; lane 3, NODBim−/−; lane 4, NOD. Pooled data from n = 3 independent experiments. *P < 0.05, **P < 0.005.

Figure 6

Increased TCR stimulation diverts thymocytes to develop into Tregs. A and B: Representative plots showing staining of thymocytes for FoxP3, GITR, CD5, CD69, and Nur77 in 8–12-week-old NODBim−/− and NODBim+/+ mice (n = 4–6 per group). Numbers represent percentages of cells contained within the gate. C: Representative histograms showing Nur77 expression in the indicated thymocyte population. NODFoxP3-GFP profiles (gray) superimposed on the profiles for NODBim−/−FoxP3-GFP mice (white). Values represent the mean fluorescence intensity (MFI) of n = 5 mice (aged 60–100 days) per group. D: Lysates from whole thymi were examined by Western blot for their content of total IκB-α; phosphorylated (i.e., activated) IκB-α (pIκBα); and, as a loading control, β-actin. Lane 1, C57BL/6Bim−/−; lane 2, C57BL/6; lane 3, NODBim−/−; lane 4, NOD. Pooled data from n = 3 independent experiments. *P < 0.05, **P < 0.005.

Close modal

To deduce the TCR signal strength received by NODBim−/− thymocytes, we measured the expression of Nur77 by flow cytometry. The Nr4a family protein Nur77 is required for thymic deletion as well as for FoxP3 expression and Treg differentiation in thymocytes following TCR stimulation (33). Nur77 expression levels are directly proportional to the strength of TCR stimulation (31). Thymocytes from NODBim−/− mice had increased expression of Nur77 compared with their counterparts from control NOD mice (Fig. 6B and C and Supplementary Fig. 3A). This is consistent with the notion that BIM deficiency allows escape of thymocytes bearing high-affinity self-antigen–specific TCRs. It has been reported that increased TCR signaling is a requirement for natural killer T cells, a population that has been shown to be defective in NOD mice. We did not see increased numbers of natural killer T cells in NODBim−/− mice compared with NOD mice (Supplementary Fig. 3C).

Following TCR stimulation, activation of the NF-κB pathway is critical for induction of FoxP3 expression and Treg differentiation (34,35). Thymocytes from BIM-deficient mice displayed augmented levels of phosphorylated IκB-α compared with their counterparts from NOD mice (Fig. 6D and Supplementary Fig. 3B), consistent with the notion that the cells surviving deletion are diverted to Tregs through NF-κB activation.

We studied the impact of BIM deficiency on thymic deletion, Treg development and function, and spontaneous autoimmunity in NOD mice. BIM deficiency increased autoreactive T cells in NOD mice by impairing thymocyte deletion. Unexpectedly, these mice were completely protected from diabetes, although the insulitis observed indicated that some autoimmunity remained. Thymocytes from NODBim−/− mice that expressed an autoreactive TCR survived strong TCR signaling and showed preferential differentiation into Tregs. These cells expressed high levels of Nur77, a transcriptional regulator that plays a key role in Treg development by translating strong TCR signals induced by self-antigens into the FoxP3 developmental program. They also had increased activation of NF-κB, a transcriptional program that is essential for Treg development.

Deficiency of another proapoptotic BH3-only protein, PUMA, in addition to loss of BIM, was reported to further impair deletion (19). In contrast to Bim−/− mice, Bim−/−Puma−/− mice on a C57BL/6 background spontaneously develop organ-specific autoimmune pathology, with leukocyte infiltration in the salivary gland, liver, lung, and pancreas and serum autoantibodies to these tissues. However, despite the almost complete block observed in thymocyte deletion, the autoimmunity in C57BL/6.Bim−/−Puma−/− mice is not fatal. The present data, in conjunction with the observation that Bim−/−Puma−/− mice also have more Tregs than Bim−/− mice (7), suggest that increased Tregs substantially restrain autoimmune disease in these settings. The current study shows that this increase in Tregs involves the diversion of thymocytes with autoreactive TCRs and reveals for the first time to our knowledge that this process can prevent autoimmune disease.

We found that diabetes protection is complete when Treg number is further increased by BIM deficiency in NOD.BDC2.5 mice and that the intermediate Treg increase and diabetes protection in NODBim+/− mice suggest that diabetes protection in NOD mice is due to expansion of Tregs. Additionally, it is noteworthy that the phenotype of NODBim−/− is similar to other mouse models in which protection from diabetes is predominantly due to Tregs, such as in the NOD.BDC2.5 mice and in NOD mice deficient in tumor necrosis factor receptor 1 (30,36). In these models, like ours, some insulitis develops, indicating the initiation of autoimmune responses. However, Tregs prevent the progression of autoimmune attack by acting in the draining PLNs or directly in the islets, thereby protecting from diabetes (37,38).

FoxP3+-expressing Tregs in NODBim−/− mice expressed lower levels of CD25 than those from control NOD mice. This has also been observed in BIM-deficient mice on a C57BL/6 background (7). Developing Treg precursors in the thymus upregulate CD25 and GITR as early as the DP (CD4+CD8+) stage (31,39). It was hypothesized that DP precursors that upregulate CD25 have a survival advantage due to increased affinity for interleukin 2. Transforming growth factor-β might also be protective to Tregs experiencing strong TCR signals from self-MHC and, thus, may be another key signal in the discrimination between clonal deletion and clonal diversion (40). BIM-deficient thymocytes are intrinsically excellent survivors, even in the absence of upregulation of CD25, and these cells are diverted to become Tregs after they encounter self-antigens in the thymus. Therefore, thymocytes that encounter high-affinity antigens are deleted unless they are saved either by interleukin 2 and transforming growth factor-β or, in this case, by inhibition of apoptosis due to loss of BIM. Consistent with this notion, thymic FoxP3+ Tregs in transgenic mice that express constitutively active IκB-α with a resultant increased NF-κB activity (41) did not express CD25 in the thymus because of the prosurvival function of NF-κB. Potentially, loss of BIM could also promote Treg survival in the periphery and be a strategy to enhance Treg therapy.

Immunological tolerance is maintained in normal individuals by a delicate balance between effector T cells and Tregs. In NOD mice, a combination of several defects in tolerance mechanisms, including failure to eliminate autoreactive T cells, results in diabetes. Several studies have proposed that this is due to either resistance to clonal deletion, perhaps as a result of a partial defect in BIM, or increased competition for positively selecting niches in the thymus (4248). The present study shows that complete BIM deficiency in NOD mice substantially impairs deletion of diabetogenic autoreactive, insulin-specific T cells but that the health of the host is preserved because autoreactive T cells that survive deletion differentiate into antigen-specific Tregs and thereby maintain immune tolerance. BIM promotes Tregs from thymic generation to peripheral survival of Tregs. Thus, modulating apoptosis may be one of the ways to increase generation and survival of antigen-specific Tregs to prevent autoimmune disease and transplant rejection.

Acknowledgments. The authors thank L. Mackin (St. Vincent's Institute, Melbourne, Australia) for help with genetic analysis; A. Irvin, S. Fynch, L. Elkerbout, N. Sanders, D. Novembre-Cycon, R. Branch, and A. Gomes (all from St. Vincent's Institute, Melbourne, Australia) for help with mice and cytometry; D. Mathis (Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA) for the NOD.BDC2.5 mice; P. Bouillet (The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia) for Bim−/− mice; O. Kanagawa (Akashi City Hospital, Akashi, Japan) for the BDC2.5 clonotypic antibody; P. Santamaria (Julia McFarlane Diabetes Research Centre, Snyder Institute for Chronic Diseases, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada) for the NOD.8.3 mice; J. Lin (University of Melbourne, Melbourne, Australia) for help with MHC class I tetramer production; and the National Institutes of Health Tetramer Core Facility for MHC class II tetramers.

Funding. This work was supported by grants and fellowships from the National Health and Medical Research Council of Australia, JDRF, the Leukemia and Lymphoma Society of America, and the Operational Infrastructure Support Scheme of the Government of Victoria.

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

Author Contributions. B.K. contributed to designing and performing the experiments and to the writing of the manuscript. J.C., G.J., P.T., T.C., C.S., E.N.G., K.L.G., J.A.W., and Y.Z. performed experiments. T.C.B., D.G., and A.S. contributed to the writing of the manuscript. J.A. contributed to designing and performing the experiments. H.E.T. and T.W.H.K. supervised the study and contributed to the writing of the manuscript. B.K. and T.W.H.K. 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.

1.
Starr
TK
,
Jameson
SC
,
Hogquist
KA
.
Positive and negative selection of T cells
.
Annu Rev Immunol
2003
;
21
:
139
176
[PubMed]
2.
Wing
K
,
Sakaguchi
S
.
Regulatory T cells exert checks and balances on self tolerance and autoimmunity
.
Nat Immunol
2010
;
11
:
7
13
[PubMed]
3.
Zheng
Y
,
Rudensky
AY
.
Foxp3 in control of the regulatory T cell lineage
.
Nat Immunol
2007
;
8
:
457
462
[PubMed]
4.
Jordan
MS
,
Boesteanu
A
,
Reed
AJ
, et al
.
Thymic selection of CD4+CD25+ regulatory T cells induced by an agonist self-peptide
.
Nat Immunol
2001
;
2
:
301
306
[PubMed]
5.
Bouillet
P
,
Purton
JF
,
Godfrey
DI
, et al
.
BH3-only Bcl-2 family member Bim is required for apoptosis of autoreactive thymocytes
.
Nature
2002
;
415
:
922
926
[PubMed]
6.
Bouillet
P
,
Metcalf
D
,
Huang
DC
, et al
.
Proapoptotic Bcl-2 relative Bim required for certain apoptotic responses, leukocyte homeostasis, and to preclude autoimmunity
.
Science
1999
;
286
:
1735
1738
[PubMed]
7.
Zhan
Y
,
Zhang
Y
,
Gray
D
, et al
.
Defects in the Bcl-2-regulated apoptotic pathway lead to preferential increase of CD25 low Foxp3+ anergic CD4+ T cells
.
J Immunol
2011
;
187
:
1566
1577
[PubMed]
8.
Feuerer
M
,
Jiang
W
,
Holler
PD
, et al
.
Enhanced thymic selection of FoxP3+ regulatory T cells in the NOD mouse model of autoimmune diabetes
.
Proc Natl Acad Sci U S A
2007
;
104
:
18181
18186
[PubMed]
9.
Apostolou
I
,
Sarukhan
A
,
Klein
L
,
von Boehmer
H
.
Origin of regulatory T cells with known specificity for antigen
.
Nat Immunol
2002
;
3
:
756
763
[PubMed]
10.
Atibalentja
DF
,
Byersdorfer
CA
,
Unanue
ER
.
Thymus-blood protein interactions are highly effective in negative selection and regulatory T cell induction
.
J Immunol
2009
;
183
:
7909
7918
[PubMed]
11.
Bautista
JL
,
Lio
CW
,
Lathrop
SK
, et al
.
Intraclonal competition limits the fate determination of regulatory T cells in the thymus
.
Nat Immunol
2009
;
10
:
610
617
[PubMed]
12.
Leung
MW
,
Shen
S
,
Lafaille
JJ
.
TCR-dependent differentiation of thymic Foxp3+ cells is limited to small clonal sizes
.
J Exp Med
2009
;
206
:
2121
2130
[PubMed]
13.
Anderson
MS
,
Bluestone
JA
.
The NOD mouse: a model of immune dysregulation
.
Annu Rev Immunol
2005
;
23
:
447
485
[PubMed]
14.
Krishnamurthy
B
,
Chee
J
,
Jhala
G
, et al
.
Complete diabetes protection despite delayed thymic tolerance in NOD8.3 TCR transgenic mice due to antigen-induced extrathymic deletion of T cells
.
Diabetes
2012
;
61
:
425
435
[PubMed]
15.
Leiter
EH
,
Reifsnyder
PC
,
Wallace
R
,
Li
R
,
King
B
,
Churchill
GC
.
NOD × 129.H2(g7) backcross delineates 129S1/SvImJ-derived genomic regions modulating type 1 diabetes development in mice
.
Diabetes
2009
;
58
:
1700
1703
[PubMed]
16.
Stadinski
BD
,
Zhang
L
,
Crawford
F
,
Marrack
P
,
Eisenbarth
GS
,
Kappler
JW
.
Diabetogenic T cells recognize insulin bound to IAg7 in an unexpected, weakly binding register
.
Proc Natl Acad Sci U S A
2010
;
107
:
10978
10983
[PubMed]
17.
Moon
JJ
,
Chu
HH
,
Hataye
J
, et al
.
Tracking epitope-specific T cells
.
Nat Protoc
2009
;
4
:
565
581
[PubMed]
18.
Krishnamurthy
B
,
Dudek
NL
,
McKenzie
MD
, et al
.
Responses against islet antigens in NOD mice are prevented by tolerance to proinsulin but not IGRP
.
J Clin Invest
2006
;
116
:
3258
3265
[PubMed]
19.
Gray
DH
,
Kupresanin
F
,
Berzins
SP
, et al
.
The BH3-only proteins Bim and Puma cooperate to impose deletional tolerance of organ-specific antigens
.
Immunity
2012
;
37
:
451
462
[PubMed]
20.
McDuffie
M
,
Schweiger
D
,
Reitz
B
,
Ostrowska
A
,
Knight
AM
,
Dyson
PJ
.
I-E-independent deletion of V beta 17a+ T cells by Mtv-3 from the nonobese diabetic mouse
.
J Immunol
1992
;
148
:
2097
2102
[PubMed]
21.
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]
22.
Deltour
L
,
Leduque
P
,
Blume
N
, et al
.
Differential expression of the two nonallelic proinsulin genes in the developing mouse embryo
.
Proc Natl Acad Sci U S A
1993
;
90
:
527
531
[PubMed]
23.
Derbinski
J
,
Schulte
A
,
Kyewski
B
,
Klein
L
.
Promiscuous gene expression in medullary thymic epithelial cells mirrors the peripheral self
.
Nat Immunol
2001
;
2
:
1032
1039
[PubMed]
24.
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]
25.
Yamanouchi
J
,
Verdaguer
J
,
Han
B
,
Amrani
A
,
Serra
P
,
Santamaria
P
.
Cross-priming of diabetogenic T cells dissociated from CTL-induced shedding of beta cell autoantigens
.
J Immunol
2003
;
171
:
6900
6909
[PubMed]
26.
Ludwinski
MW
,
Sun
J
,
Hilliard
B
, et al
.
Critical roles of Bim in T cell activation and T cell-mediated autoimmune inflammation in mice
.
J Clin Invest
2009
;
119
:
1706
1713
[PubMed]
27.
Hsieh
CS
,
Lee
HM
,
Lio
CW
.
Selection of regulatory T cells in the thymus
.
Nat Rev Immunol
2012
;
12
:
157
167
[PubMed]
28.
Thornton
AM
,
Korty
PE
,
Tran
DQ
, et al
.
Expression of Helios, an Ikaros transcription factor family member, differentiates thymic-derived from peripherally induced Foxp3+ T regulatory cells
.
J Immunol
2010
;
184
:
3433
3441
[PubMed]
29.
Zhou
X
,
Jeker
LT
,
Fife
BT
, et al
.
Selective miRNA disruption in T reg cells leads to uncontrolled autoimmunity
.
J Exp Med
2008
;
205
:
1983
1991
[PubMed]
30.
Chen
Z
,
Herman
AE
,
Matos
M
,
Mathis
D
,
Benoist
C
.
Where CD4+CD25+ T reg cells impinge on autoimmune diabetes
.
J Exp Med
2005
;
202
:
1387
1397
[PubMed]
31.
Moran
AE
,
Holzapfel
KL
,
Xing
Y
, et al
.
T cell receptor signal strength in Treg and iNKT cell development demonstrated by a novel fluorescent reporter mouse
.
J Exp Med
2011
;
208
:
1279
1289
[PubMed]
32.
Lio
CW
,
Hsieh
CS
.
A two-step process for thymic regulatory T cell development
.
Immunity
2008
;
28
:
100
111
[PubMed]
33.
Sekiya
T
,
Kashiwagi
I
,
Yoshida
R
, et al
.
Nr4a receptors are essential for thymic regulatory T cell development and immune homeostasis
.
Nat Immunol
2013
;
14
:
230
237
[PubMed]
34.
Long
M
,
Park
SG
,
Strickland
I
,
Hayden
MS
,
Ghosh
S
.
Nuclear factor-kappaB modulates regulatory T cell development by directly regulating expression of Foxp3 transcription factor
.
Immunity
2009
;
31
:
921
931
[PubMed]
35.
Isomura
I
,
Palmer
S
,
Grumont
RJ
, et al
.
c-Rel is required for the development of thymic Foxp3+ CD4 regulatory T cells [published correction appears in J Exp Med 2010;207:899]
.
J Exp Med
2009
;
206
:
3001
3014
[PubMed]
36.
Chee
J
,
Angstetra
E
,
Mariana
L
, et al
.
TNF receptor 1 deficiency increases regulatory T cell function in nonobese diabetic mice
.
J Immunol
2011
;
187
:
1702
1712
[PubMed]
37.
Tang
Q
,
Adams
JY
,
Tooley
AJ
, et al
.
Visualizing regulatory T cell control of autoimmune responses in nonobese diabetic mice
.
Nat Immunol
2006
;
7
:
83
92
[PubMed]
38.
Feuerer
M
,
Shen
Y
,
Littman
DR
,
Benoist
C
,
Mathis
D
.
How punctual ablation of regulatory T cells unleashes an autoimmune lesion within the pancreatic islets
.
Immunity
2009
;
31
:
654
664
[PubMed]
39.
Cabarrocas
J
,
Cassan
C
,
Magnusson
F
, et al
.
Foxp3+ CD25+ regulatory T cells specific for a neo-self-antigen develop at the double-positive thymic stage
.
Proc Natl Acad Sci U S A
2006
;
103
:
8453
8458
[PubMed]
40.
Ouyang
W
,
Beckett
O
,
Ma
Q
,
Li
MO
.
Transforming growth factor-beta signaling curbs thymic negative selection promoting regulatory T cell development
.
Immunity
2010
;
32
:
642
653
[PubMed]
41.
Hettmann
T
,
Leiden
JM
.
NF-kappa B is required for the positive selection of CD8+ thymocytes
.
J Immunol
2000
;
165
:
5004
5010
[PubMed]
42.
Choisy-Rossi
CM
,
Holl
TM
,
Pierce
MA
,
Chapman
HD
,
Serreze
DV
.
Enhanced pathogenicity of diabetogenic T cells escaping a non-MHC gene-controlled near death experience
.
J Immunol
2004
;
173
:
3791
3800
[PubMed]
43.
Zucchelli
S
,
Holler
P
,
Yamagata
T
,
Roy
M
,
Benoist
C
,
Mathis
D
.
Defective central tolerance induction in NOD mice: genomics and genetics
.
Immunity
2005
;
22
:
385
396
[PubMed]
44.
Serreze
DV
,
Choisy-Rossi
CM
,
Grier
AE
, et al
.
Through regulation of TCR expression levels, an Idd7 region gene(s) interactively contributes to the impaired thymic deletion of autoreactive diabetogenic CD8+ T cells in nonobese diabetic mice
.
J Immunol
2008
;
180
:
3250
3259
[PubMed]
45.
Lesage
S
,
Hartley
SB
,
Akkaraju
S
,
Wilson
J
,
Townsend
M
,
Goodnow
CC
.
Failure to censor forbidden clones of CD4 T cells in autoimmune diabetes
.
J Exp Med
2002
;
196
:
1175
1188
[PubMed]
46.
Liston
A
,
Lesage
S
,
Gray
DH
, et al
.
Generalized resistance to thymic deletion in the NOD mouse; a polygenic trait characterized by defective induction of Bim
.
Immunity
2004
;
21
:
817
830
[PubMed]
47.
Mingueneau
M
,
Jiang
W
,
Feuerer
M
,
Mathis
D
,
Benoist
C
.
Thymic negative selection is functional in NOD mice
.
J Exp Med
2012
;
209
:
623
637
[PubMed]
48.
Holler
PD
,
Yamagata
T
,
Jiang
W
,
Feuerer
M
,
Benoist
C
,
Mathis
D
.
The same genomic region conditions clonal deletion and clonal deviation to the CD8alphaalpha and regulatory T cell lineages in NOD versus C57BL/6 mice
.
Proc Natl Acad Sci U S A
2007
;
104
:
7187
7192
[PubMed]

Supplementary data