CD137 (TNFRSF9) is an activation-inducible T-cell costimulatory molecule and a member of the tumor necrosis factor (TNF) receptor superfamily. Cd137 is also a candidate gene (in the Idd9.3 interval) for autoimmune diabetes in NOD mice. Here, we demonstrate that anti-CD137 treatment protects NOD mice from diabetes. Anti-CD137–treated mice are not protected from insulitis and still harbor pathogenic T-cells, as demonstrated by transfer studies. Transfer of CD4+, but not CD8+, cells from anti-CD137–treated pre-diabetic NOD mice into NOD-scid mice delayed diabetes onset. Anti-CD137 treatment significantly increased the number of CD4+CD25+ cells, which demonstrated intracellular Foxp3 expression and in vitro suppressive activity. The CD4+CD25+ cell subset from anti-CD137–treated mice transferred complete protection from diabetes, whereas the CD4+CD25 cell subset offered no significant protection. Anti-CD137 treatment of NOD-scid recipients of diabetic spleen cells, however, hastened the onset of disease, showing that the effect of anti-CD137 treatment depends on the balance of pathogenic and protective cells. These results support a critical role for CD137 acting in the early phase of autoimmune diabetes to enhance regulatory cell production. Disease-associated CD137 alleles are likely ineffectual at stimulating a regulatory T-cell population sufficient to prevent disease.

The NOD mouse is a well-established immunogenetic model of spontaneous type 1 diabetes (1). NOD mice spontaneously develop insulitis (infiltration of the pancreatic islet cells) starting at 3 weeks of age and develop diabetes around 20 weeks. The NOD mouse model exemplifies the modern approach to characterizing a genetically complex autoimmune disease. The first genome scan of NOD mice demonstrated that multiple recessive loci, termed “insulin-dependent diabetes” (Idd) loci, were linked to diabetes resistance or susceptibility (2). At present, at least 20 genes are thought to play a role in this complex, multigenic process (1). The strongest genetic contribution is from the major histocompatibility complex (MHC) class II molecule, I-Ag7; however, even two copies of I-Ag7 are insufficient to mediate diabetes when placed on a nonautoimmune non-MHC background (3). Therefore, although substitution of other MHC class II molecules can prevent diabetes (indicating that I-Ag7 is necessary for disease), it is not both necessary and sufficient; many non-MHC Idd loci contribute critically to the disease process.

To establish the identity and function of non-MHC Idd loci, Idd congenic mice are bred to contain B6/B10 resistance (Idd) intervals on a predominant NOD (disease-related) genetic background. These congenic mice show decreased susceptibility to diabetes, confirming that the gene or genes whose B6/B10 alleles were resistant to disease were “captured” in the congenic intervals (1). The congenic genetic intervals are initially quite large, but a “congenic mapping” approach has led to the identification of several “candidate” genes in the intervals (49). One such candidate gene, in the Idd9.3 locus, is Cd137 (6). Idd9.3 has been mapped to a 1.2-mb interval with 15 genes, including Cd137 (10). Cannons et al. (10) recently demonstrated that the NOD allele of CD137, compared with the B10 allele, had decreased T-cell signaling; NOD T-cells stimulated with a CD137L produced less interleukin (IL)-2 than NOD.Idd9.3 congenic T-cells.

CD137 is an inducible member of the TNF receptor superfamily expressed on activated T-cells (11). It is upregulated on T-cells 48 h after activation in vitro and functions as a T-cell costimulatory molecule (12,13). The CD137 ligand is expressed on activated antigen-presenting cells (APCs), especially at the sites of inflammation in vivo (1417). CD137 stimulation augments T-cell proliferation and can function as a T-cell costimulatory molecule in the absence of CD28 (13,18). Anti-CD137 antibodies have elucidated the function of CD137 in vivo, causing expansion of both CD4 and CD8 T-cell subsets (12,19). T-cells from CD137−/− or CD137L−/− mice have decreased memory responses to antigen recall; CD137L−/− mice adoptively transferred with antigen-specific CD8 TCR TG cells showed decreased late-stage T-cell survival (20). These results combined with the expression kinetics of CD137 suggest a costimulatory role acting later in T-cell activation and critically in the activation of memory T-cells. Anti-CD137 antibodies have demonstrated activity in several murine autoimmune models. Anti-CD137 blocked development of collagen arthritis, accompanied by suppression of anti-collagen antibodies and decreased CD4+ recall responses to type-2 collagen (17,21). In a murine lupus model, anti-CD137 reversed established disease and dramatically reduced autoantibody production (22). Anti-CD137 also prevented experimental autoimmune encephalomyelitis (EAE); notably, however, mice induced to get EAE by adoptive transfer of pathogenic T-cell clones were not protected by anti-CD137 (23). Overall, these results demonstrate a powerful effect of CD137 on T-cells and on the progression of autoimmune disease states.

Recently, CD4+CD25+ T-cells have emerged as essential regulatory cells controlling autoimmunity and antitumor immunity in human and animal models (24). In the setting of type 1 diabetes in NOD mice, quantitative and qualitative deficiencies of CD4+CD25+ T regulatory (Treg) cells have been reported (25,26), and transfer of CD4+CD25+ cells suppressed the onset of diabetes (27). Although the precise mechanisms employed by Treg cells remain unclear, costimulatory signaling is clearly important (25). Two studies reported that CD137 was upregulated in CD4+CD25+ Tregs (27,28), although conflicting results on the role of CD137 in Tregs were reported using CD137−/− mice (29,30).

In this study, we demonstrate that treatment of NOD mice with anti-CD137 antibody prevented autoimmune diabetes but did not eliminate pancreatic insulitis. Pathogenic cells, in fact, persisted in protected NOD mice, as shown by transfer studies. CD4+ cells isolated from anti-CD137–treated mice ameliorated adoptive transfer of diabetes into NOD-scid mice, whereas control-treated CD4+ cells did not. Using transfer of anti-CD137–treated or control pre-diabetic splenocytes into NOD-scid, we show that anti-CD137 significantly increased the number of CD4+CD25+ cells in treated mice, and transfer of these CD4+CD25+ cells completely prevented diabetes in NOD-scid recipients, whereas the CD4+CD25 cell subset offered no significant protection. Treatment of NOD-scid mice with anti-CD137 after transfer of diabetic CD4+ and CD8+ cells not only did not prevent disease, but also hastened its onset, demonstrating that CD137 expression on autoreactive memory cells can mediate destructive effects. These results implicate a role for the Idd9.3 candidate gene product, CD137, in type 1 diabetes pathogenesis and suggest that the balance of CD137 stimulation between regulatory and pathogenic T-cells could represent a therapeutic target.

NOD and NOD-scid colonies were maintained under specific pathogen-free conditions in our animal facilities. Mice were handled in accordance with the institutional animal care guidelines of the University of Pittsburgh School of Medicine. Urinary glucose analysis was performed using Tes-tape (Shionogi, Osaka, Japan) once a week. Plasma glucose levels were determined when glycosuria was detected, and mice with a blood glucose level above 300 mg/dl were considered diabetic. Antibodies against mouse CD3, CD4, CD8, CD25, CD28, CD49b, and T-cell β-receptor were purchased from BD Bioscience. Anti-Foxp3 antibody was purchased from eBioscience. Anti-mouse CD137 antibody (3H3) was produced by R.S. Mittler as previously described (22).

Histology.

The pancreas of NOD mice was removed at the indicated times, fixed in 10% formaldehyde, and embedded in paraffin. Thin sections at six levels, 150 μm apart, were cut for staining with hematoxylin-eosin to evaluate islet-infiltrating cells by light microscopy. At least 35 islets from each recipient mouse were observed and scored using the following criteria: grade 0, islets free of insulitis; grade 1, peri-insulitis; grade 2, intra-insulitis with mononuclear cell infiltration of <50% of the area of each islet; and grade 3, intra-insulitis with mononuclear cell infiltration of >50% of the area of each islet.

Preparation, sorting, and stimulation of splenocytes and adoptive transfer.

The spleen from each mouse was removed aseptically and minced. After lysing erythrocytes, the cells were washed three times with PBS. To sort CD4+, CD4+CD25+, CD4+DX5, or CD8+ cells, splenocytes were prepared by magnetic separation using a magnetic sorting system (Miltenyi Biotec, Auburn, CA) according to the manufacturer’s instructions. The purities of sorted cells were >90%. In other experiments, purified CD4 populations were obtained by sorting using flow cytometry. To stimulate sorted splenocytes, cells were suspended in RPMI 1640 supplemented with 10% heat-inactivated fetal bovine serum (Gibco-BRL) and penicillin/streptomycin (Gibco-BRL). The CD4+ cells (1 × 106) resuspended in 2 ml culture medium were transferred to each well of a 24-well anti-CD3 antibody precoated plate with or without soluble anti-CD28 antibody (1 μg/ml). The cells were cultured for 72 h at 37°C in a humidified 5% CO2 incubator, and the supernatant was collected at the end of culture.

For suppression experiments, CD4+CD25+ cells, 5 × 104 of effector or suppressor cells, or both were seeded to 96-well plate with soluble anti-CD3 antibodies and irradiated splenocytes as APCs.

Flow cytometry.

Cells were incubated with Fc blocker (Pharmingen) and stained with labeled antibodies for 25 min at 4°C. Samples were analyzed on an FACS Caliber (Becton Dickinson, Miami, FL). Intracellular cytokine staining of Foxp3 was performed according to the manufacturer’s instructions.

Cytokine measurement by enzyme-linked immunosorbent assay.

Cytokines (interferon-γ [IFN-γ], IL-4, and IL-10) were measured by enzyme-linked immunosorbent assay (ELISA) as previously described (31). Briefly, flat-bottom 96-well plates were coated with anti–IFN-γ, anti–IL-4, or anti–IL-10 antibodies (Pharmingen), and supernatants were added. After incubation, biotinylated anti-cytokine antibodies and then europium-avidin solution (Perkin Elmer, Turku, Finland) were added. After adding enhancement solution (Perkin Elmer), plates were measured with an ELISA reader (Perkin Elmer).

Statistical analysis.

Results are presented as means ± SE. Differences between the two groups were analyzed using Mann-Whitney U test for nonparametric unpaired observations. Log-rank tests were used to compare the incidence of diabetes between two groups using JMP IN software (SAS Institute) and Graph-pad (Graph-pad Software).

Anti-CD137 prevents diabetes but does not prevent insulitis nor eliminate pathogenic cells.

To test the effect of anti-CD137 on the course of type 1 diabetes in NOD mice, we treated 6-week-old female NOD mice with a total of three 200-μg doses of anti-CD137 antibody (clone 3H3) (22) administered over a 9-week interval every 3rd week. Control mice received PBS alone; mice were followed for glycosuria. The administration of anti-CD137 antibody significantly suppressed type 1 diabetes (Fig. 1; P = 0.0005).

It has previously been published that when placed on the NOD background, the Idd9.3 region (containing the B10 allele of Cd137) decreased diabetes incidence but did not result in decreased insulitis (6). We tested whether anti-CD137 treatment ameliorated insulitis; as shown in Fig. 2A, anti-CD137 did not suppress pancreatic islet infiltration (“insulitis”) in mice protected from diabetes. To further evaluate the progression of insulitis in treated mice, NOD mice were treated at 6 and 9 weeks of age with 200 μg anti-CD137 antibody or with PBS alone, and at 10 weeks of age the pancreas was scored for insulitis. As shown in Fig. 2B, the insulitis score between treated and control NOD mice was no different. Therefore we conclude that anti-CD137 treatment prevents type 1 diabetes but not by preventing insulitis.

The presence of insulitis in the treated mice raised the question of whether anti-CD137 directly or indirectly affected pathogenic T-cells, for example, by inducing deletion or regulation of autoreactive T-cells. We addressed this possibility by transferring 20 × 106 splenocytes from anti-CD137–treated nondiabetic NOD mice, aged over 200 days, into NOD-scid recipients. Although the donor mice were protected from diabetes, seven of nine NOD-scid recipients developed type 1 diabetes, indicating that the donors retained pathogenic T-cells (Fig. 2C). This result demonstrated that pathogenic T-cells were not destroyed or inactivated in anti-CD137–treated NOD mice.

CD4+ cells from mice recently treated with anti-CD137 antibody can ameliorate adoptive transfer of diabetes.

The above experiments raised the question: why did splenocytes from aged, but nondiabetic, treated mice transfer disease? We reasoned that, because some anti-CD137–treated mice eventually develop diabetes (Fig. 1), the regulatory effect likely decreased over time. As a result, when splenocytes from aged treated mice were transferred into NOD-scid, homeostatic expansion of autoreactive cells in the NOD-scid lymphopenic environment (32) might overwhelm any remaining regulatory effect. The hypothesis resulting from this scenario is that regulatory cells from recently treated mice should be able to ameliorate disease in the NOD-scid transfer model. To test this hypothesis we used a pre-diabetic NOD transfer system, as previously described (33), to investigate the protective cell populations in the pre-diabetic mice. CD4+ cells from anti-CD137–treated 10-week-old pre-diabetic NOD mice, but not from PBS-treated age-matched controls, significantly delayed disease transfer into NOD-scid recipients (Fig. 3; P = 0.0016). (Note the donor population used in this model is pre-diabetic; as shown in Fig. 3, diabetic splenocytes transfer disease more rapidly.) Therefore freshly recruited CD4+ regulatory cells can counteract the disease process even in the NOD-scid transfer model. Notably, however, in the NOD-scid transfer model, the protective effect of anti-CD137 antibody treatment was less than in “normal” NOD mice (Fig. 1), likely as a result of rapid homeostatic effector expansion in the NOD-scid environment. It has previously been reported that anti-CD137 treatment can induce CD8+ regulatory cells as a protective mechanism (17). To test the hypothesis that regulatory CD8+ cells could have suppressor function in treated NOD mice, CD8+ cells from either anti-CD137–treated or PBS-treated control NOD mice were transferred to NOD-scid recipients with diabetic CD4+ cells. CD8+ cells from treated NOD mice, however, did not have protective ability, indicating that CD8+ regulatory cells are unlikely to participate in anti-CD137–induced diabetes protection in NOD mice (data not shown). These results are in agreement with data of Foell et al. (22), which showed that anti-CD137–mediated reversal of systemic lupus erythematosus (SLE) was independent of CD8+ T-cells.

Anti-CD137 treatment alters the cytokine phenotype of CD4+ cells and increases the proportion of IL-10–producing DX5+ T-cells; Dx5+ cells do not prevent diabetes.

The co-transfer experiment shown above encouraged us to evaluate the functional properties of CD4+ cells induced by anti-CD137 antibody treatment. As shown in Fig. 4, sorted CD4+ splenocytes from anti-CD137–treated mice produced significantly more IL-4 and IL-10 and less IFN-γ than CD4+ cells from untreated mice.

To determine the source of IL-4/IL-10–producing cells, we examined Dx5+ NKT cells, because it has been recently shown that CD137−/− mice have decreased numbers of CD3+/Dx5+ NK T-cells, and it is well established that NKT cells produce substantial quantities of IL-4 and IL-10 (34,35). We hypothesized that one mechanism of anti-CD137–mediated protection occurred through its effects on NK T-cell numbers and function. To test this hypothesis, we isolated Dx5+ cells from anti-CD137–treated and untreated NOD mice (at 10 weeks of age as above) and examined the numbers of positive cells. The number of Dx5+ T-cells increased in anti-CD137–treated NOD spleen and peripheral lymph node (P = 0.0045 and 0.0012, respectively; Fig. 5A).

Next, we asked to what extent the change in cytokine production in the CD4+ population was due to Dx5+ cells. Stimulated CD4+ cells from anti-CD137–treated spleens produced more IL-4 and IL-10 than CD4+ cells from PBS-treated mice (Fig. 5B, solid bars), and removing DX5+ cells from the CD4+ population eliminated the enhanced level of cytokine production; the CD4+DX5 cells from treated mice had the same production of IL-4 and IL-10 as untreated CD4+ cells (Fig. 5B, hatched bars).

Finally, to test whether Dx5+ NKT cells had a protective effect in vivo, we used two approaches. First, CD4+Dx5+ cells were purified using sorting by flow cytometry. NOD-scid recipients then received either CD4+DX5 populations alone or CD4+DX5 cells plus CD4+Dx5+ cells from anti-CD137–treated pre-diabetic mice, along with diabetic CD8+ cells. As shown in Fig. 5C, there was no significant benefit in diabetes prevention obtained by adding CD4+Dx5+ to CD4+Dx5 cells, compared with CD4+Dx5 cells alone. In the second approach, NOD-scid mice received either whole CD4+ (including Dx5+) cells from untreated pre-diabetic NOD mice or CD4+DX5 cells, along with diabetic CD8+ cells. Again, as shown in Fig. 5D, there was no significant difference in diabetes onset between the two groups, indicating no treatment effect of depleting Dx5+ cells in the transfer system. Both CD4+Dx5+ and CD4+Dx5 transfers from anti-CD137–treated mice delayed diabetes compared with CD4+ or CD4+Dx5 populations from untreated mice (Fig. 5C compared with D, P = 0.0023), but neither Dx5+ nor Dx5 fractions from untreated mice delayed diabetes onset (Fig. 5D; compare with PBS-treated CD4 cells in Fig. 3). This result showed that there was no protective effect of the CD4+Dx5+ population in the NOD-scid transfer system (although it remains possible that a protective effect of Dx5 cells exists and is not shown by our experiments) but confirmed that there must be at least one protective CD4+ cell population in the anti-CD137–treated CD4+Dx5 cell population.

Anti-CD137 treatment increases the CD4+CD25+ Treg cell population; anti-CD137–treated CD4+CD25+ cells prevent adoptive transfer of diabetes; anti-CD137 binds specifically to CD4+CD25+ cells in vivo.

The transfer of type 1 diabetes from splenocytes of anti-CD137–treated nondiabetic mice and the protective capability of anti-CD137–treated CD4+ subsets indicated that anti-CD137 treatment did not destroy/inactivate pathogenic CD4+ T-cells but acted on regulatory cells. The presence of a protective effect in the treated CD4+Dx5 subset led us to investigate other regulatory CD4 subsets. It has been well established that CD4+CD25+ regulatory cells can modulate the development of type 1 diabetes in the presence of pathogenic T-cells. Quantitative and qualitative defects of CD4+CD25+ Treg cells have been reported in NOD mice, and CD4+CD25+ Tregs can prevent type 1 diabetes in NOD mice (25,26). Therefore, we investigated the quantitative and qualitative phenotype of CD4+CD25+ Treg cells in anti-CD137–treated NOD mice.

As shown in Fig. 6A, we found that naïve CD4+CD25+ cells in NOD mice upregulate CD137 compared with CD25 cells, supporting two previous reports, which indicated that CD4+CD25+ cells upregulated CD137 compared with CD4+CD25 cells (27,28). The upregulation of CD137 on CD25+ cells suggested a mechanism whereby CD137 antibody treatment could preferentially affect CD4+CD25+ cells. As shown in Fig. 6B, anti-CD137 antibody treatment increased the number of CD4+CD25+ T-cells in NOD mice.

To confirm that increased CD4+CD25+ T-cells were regulatory cells and not polyclonally activated T-cells upregulating CD25, intracellular Foxp3 expression was analyzed. CD25+ cells in anti-CD137–treated mice were Foxp3+ (Fig. 6C), indicating that the administration of anti-CD137 antibody induced Foxp3+CD4+CD25+ Treg cells associated with diabetes protection. To prove the regulatory function of CD4+CD25+ Treg cells in vitro, we performed suppression studies, which showed that CD4+CD25+ cells from anti-CD137–treated NOD mice had suppressive activity on CD25 cells from both naïve and anti-CD137–treated mice (Fig. 6D). These findings indicated that CD137 antibody–induced CD4+CD25+ cells have the Treg cell phenotype.

To directly prove that treatment-induced CD4+CD25+ cells have protective ability, we transferred CD4+CD25+ or CD4+CD25 cells from anti-CD137–treated mice, along with diabetic CD8+ cells, to NOD-scid recipients. As shown in Fig. 6E, all NOD-scid recipients transferred with CD4+CD25+ cells were completely protected from diabetes compared with those that received CD4+CD25 cells (P < 0.0001). Moreover, mice receiving anti-CD137–treated CD4+ cells from which CD25+ cells were depleted (CD4+CD25) showed no significant difference in disease onset compared with CD4+ T-cells transferred from NOD mice treated with PBS alone (Fig. 6E, P = 0.91), which strongly supports the hypothesis that anti-CD137 therapy acts on CD4+CD25+ cells. Interestingly, we found that some of the mice receiving CD25 cells developed inflammatory bowel disease (IBD) (manifested by wasting and rectal prolapse) or neuropathy, consistent with previous reports (data not shown; 36,37). Of mice receiving the CD25 cells, five of nine developed diabetes, whereas four of nine developed either IBD or neuropathy. The finding that diabetes and neuropathy/IBD were mutually exclusive is consistent with the report of Setoguchi et al. (36), who had similar results.

To test whether anti-CD137 had a preferential cell target in vivo, we harvested splenocytes from NOD mice treated twice with 3H3, 1 week after the final treatment. We stained for CD4 and CD25 and used the second-step (anti-IgG2a-bio) antibody specific for 3H3, followed by strep-avidin phycoeythrin. As shown in Fig. 6F, 3H3 was bound only to CD4+CD25+ cells, not CD4+CD25 cells in vivo, consistent with a mechanism of action directed to this Treg subset. There was not significant in vivo binding of 3H3 to CD8 or Dx5+ cell subsets.

Anti-CD137 treatment of diabetic splenocytes can hasten disease onset in a transfer system.

Finally, we tested directly whether anti-CD137 antibody could act on pathogenic CD4+ cells by using a different cell transfer system, in which CD4+ and CD8+ cells were sorted from diabetic NOD splenocytes and transferred into NOD-scid recipients. At the time of transfer, recipient NOD-scid mice were treated with a single dose of 200 μg anti-CD137 or PBS. Treatment of NOD-scid recipients with anti-CD137 at the time of adoptive transfer did not prevent diabetes compared with PBS-treated control NOD-scid recipients and slightly but significantly increased the onset of diabetes (Fig. 7). This result confirms that the protective effect of anti-CD137 did not act by inactivating pathogenic CD4+ and CD8+ T-cells and, in fact, shows that activating CD137 on autoreactive T-cells could enhance disease onset. This result agrees with the data of Sun et al. (23), who showed that although anti-CD137 therapy can prevent EAE, it could not prevent EAE after adoptive transfer of a pathogenic T-cell population. The lack of effect of anti-CD137 antibody at time of transfer into NOD-scid is likely due to an imbalance of regulatory cells compared with pathogenic cells in diabetic NOD spleen populations.

We have demonstrated that treatment specifically directed to the Idd9.3 candidate gene product, CD137, can prevent diabetes in NOD mice and showed that anti-CD137 therapy increased the number of CD4+Dx5+ and CD4+CD25+ Treg cells. By using transfer models into NOD-scid mice, we showed that the effect of anti-CD137 treatment is not directly on the CD4+ pathogenic autoreactive T-cell population (because these cells persist in treated mice and can transfer disease) but is on Treg cells that control the pathogenicity of the autoreactive cells.

Anti-CD137 therapy has previously been demonstrated to prevent murine SLE, EAE, and collagen arthritis (17,2123). Ours is the first report, to our knowledge, to show that it prevents autoimmune diabetes. The only previous report of the effect of CD137 on the diabetes disease process is that overexpression of a Cd137 transgene increased the incidence of diabetes (38). Although our results indicate a protective effect of CD137 stimulation in type 1 diabetes of NOD mice, our results combined with the results of Sytwu et al. (38) suggest that there can be too much of a good thing. CD137 is a member of the TNF receptor superfamily; in this regard, it is useful to recall that TNF-α had dual effects in type 1 diabetes: anti–TNF-α treatment at 9–10 weeks prevented diabetes, whereas TNF-α given to neonates increased diabetes severity (3941). Our results show that anti-CD137 is most effective at preventing diabetes when given early in the disease process. Once a cadre of autoreactive T-cells is fully developed, anti-CD137 cannot prevent disease and may even worsen it (as shown by the earlier onset of diabetes when anti-CD137 is given at the time of transfer of NOD CD4+ diabetic cells into NOD-scid; Fig. 7). Moreover, splenocytes taken from aged anti-CD137 mice can transfer diabetes, suggesting a finite regulatory capacity of the anti-CD137 induced Treg cells. Because autoimmune, activated pathogenic CD4+ T-cells upregulate CD137, it is possible also that once the autoimmune repertoire has expanded, anti-CD137 activates the autoreactive cells, opposing the protective effect of CD4+CD25+ Treg cells. In this regard, it is noteworthy that we find that anti-CD137 treatment of lymphocytic chonomeningitis virus–infected mice leads to sustained high-level production of TNF-α (Mittler lab, unpublished data).

Nevertheless, our results are important for several reasons. Cd137 is a strong candidate gene for Idd9.3; i.e., it is highly likely that CD137 has a significant role in the pathogenesis of spontaneous type 1 diabetes in NOD mice. Lyons et al. (6) showed that although the Idd9.3 region containing the B10 allele of Cd137 decreased diabetes incidence on an NOD background, it did not prevent insulitis, consistent with our result. Moreover, these authors showed that the insulitic lesions in the Idd9 congenic upregulated IL-4, consistent with our demonstration that anti-CD137–treated CD4+ cells upregulate IL-4 production (6). Cannons et al. (10) have recently followed up this work by demonstrating that T-cells from NOD mice congenic for the B10 allele of Cd137 proliferate more vigorously to anti-CD3 plus anti-CD137 stimulation than NOD cells, i.e., that the NOD Cd137 allele may have relatively defective signaling capability. The authors speculate that decreased signaling through NOD CD137 could result in defective regulatory cell production. Our results are consistent with this hypothesis, because we show that agonistic stimulation through CD137 can prevent diabetes in NOD mice by the induction of a regulatory cell population. Although our results do not prove that Cd137 is the Idd9.3 gene, they are entirely consistent with this conclusion and supportive of prior studies defining the role of CD137 in type 1 diabetes.

Our results show that anti-CD137 mediates protection from diabetes via effects on CD4+CD25+ Treg cells. It has previously been shown that NOD CD4+CD25+ Treg cells numbers and functional capacity are decreased and that boosting NOD CD4+CD25+ Treg cell function can prevent diabetes (25,26). What we show for the first time is that NOD CD4+CD25+ Treg cells function may be linked to the Idd9.3 candidate gene, Cd137. We show that CD4+CD25+ Treg cells upregulate CD137 compared with the CD4+CD25 subset and that transfer of the CD25+ subset completely prevented diabetes, whereas the CD4+CD25 cell subset offered no significant protection. These findings are consistent with previous reports showing that CD4+CD25+ Treg cells upregulate CD137 (27,28) and with the increase of CD4+CD25+ cells in anti-CD137–treated mice in various models, although regulatory function was not proved in the latter reports (21,23). The effects of anti-CD137 antibody on CD4+CD25+ Treg cells are still controversial. 4-1BB ligand induces proliferation of CD4+CD25+ Treg cells and maintains their suppressor function (29), in contrast to no observable effect of anti-CD137 antibody on CD4+CD25+ Treg cells in vitro (30). This study also reported that regulatory cells from CD137−/− mice could control CD45RBhigh T-cell–induced colitis, implying that CD4+CD25+ Treg cells may exist independent of CD137 signaling (30). Moreover, in the setting of experimental autoimmune thyroiditis, the administration of anti-CD137 antibody breaks tolerance and worsens the disease (42). The double-edged effect of CD137 signaling likely depends on the balance of pathogenic and protective subsets at the time of treatment in any given model system, as we showed in Fig. 7. None-theless, our findings of specific binding of anti-CD137 antibody only to CD4+CD25+ cells in vivo plus our demonstration that these cells mediate the protective effect of the CD4 population in anti-CD137–treated mice strongly support the hypothesis that the CD4+CD25+CD137+ Treg cell subset is a critical protective subset in type 1 diabetes.

We confirmed the observation recently published by Sakaguchi and colleagues (36,37) that CD25-depleted CD4 cells transfer several autoimmune diseases. In particular, we also confirmed their observation that the transfer of diabetes versus another organ-specific autoimmune syndrome, such as IBD or peripheral neuropathy, is mutually exclusive in this system (36). The mechanism for this striking organ specificity and exclusivity of autoimmune attack remains unknown. However, we have recently published a similar effect using genetic manipulation, in which we showed that modification of the NOD background by addition of Idd loci from B6/B10 on chromosomes 3 and 4 completely prevented diabetes while producing an autoimmune biliary disease (43,44). The remarkable “switching” of the tissue focus of autoimmune attack via environmental (CD25 depletion) or genetic (introgression of congenic regions) mechanisms is as yet unexplained but is clearly of great importance to understanding the pathogenesis of autoimmunity and type 1 diabetes.

In conclusion, we, for the first time, report that anti-CD137 antibody therapy suppresses type 1 diabetes in NOD mice and that this effect is dependent on induction of Treg cells. Treatment must be initiated before the development of a cadre of autoreactive T-cells, suggesting competing effects on protective and pathogenic cell subsets in type 1 diabetes. These results implicate a role for the Idd9.3 candidate gene, Cd137, in type 1 diabetes pathogenesis and identify CD137 as a potential therapeutic target.

FIG. 1.

Anti-CD137 antibody prevents type 1 diabetes in NOD mice. Female NOD mice aged 42–44 days were treated three times with 200 μg anti-CD137 antibody (○; n = 16) or 200 μg PBS (•; n = 16) (see arrows). The onset of diabetes was detected by checking for glucosuria every week and confirmed when the blood glucose levels were >300 mg/dl. *P = 0.0005 vs. control by log-rank test.

FIG. 1.

Anti-CD137 antibody prevents type 1 diabetes in NOD mice. Female NOD mice aged 42–44 days were treated three times with 200 μg anti-CD137 antibody (○; n = 16) or 200 μg PBS (•; n = 16) (see arrows). The onset of diabetes was detected by checking for glucosuria every week and confirmed when the blood glucose levels were >300 mg/dl. *P = 0.0005 vs. control by log-rank test.

Close modal
FIG. 2.

Anti-CD137 does not prevent insulitis nor delete pathogenic T-cells. A: The pancreases of anti-CD137–treated, diabetes-free female NOD mice aged 207–254 days were studied by hematoxylin-eosin staining. One representative of eight mice is shown. B: Quantification of insulitis in anti-CD137 antibody–treated female NOD mice at 10 weeks of age. Female NOD mice aged 46–55 days were treated twice with 200 μg anti-CD137 antibody (n = 8) or 200 μg PBS (n = 6) and scored for insulitis 1 week after the second injection. C: Splenocytes from anti-CD137–treated nondiabetic mice can transfer diabetes into NOD-scid. Spleens were harvested from three anti-CD137–treated NOD mice aged over 200 days and transferred into a total of nine NOD-scid mice.

FIG. 2.

Anti-CD137 does not prevent insulitis nor delete pathogenic T-cells. A: The pancreases of anti-CD137–treated, diabetes-free female NOD mice aged 207–254 days were studied by hematoxylin-eosin staining. One representative of eight mice is shown. B: Quantification of insulitis in anti-CD137 antibody–treated female NOD mice at 10 weeks of age. Female NOD mice aged 46–55 days were treated twice with 200 μg anti-CD137 antibody (n = 8) or 200 μg PBS (n = 6) and scored for insulitis 1 week after the second injection. C: Splenocytes from anti-CD137–treated nondiabetic mice can transfer diabetes into NOD-scid. Spleens were harvested from three anti-CD137–treated NOD mice aged over 200 days and transferred into a total of nine NOD-scid mice.

Close modal
FIG. 3.

CD4+ splenocytes from anti-CD137–treated mice ameliorate transfer of diabetes into NOD-scid mice. Female NOD-scid mice received (0.5–2) × 106 diabetic CD8 cells plus 2 × 106 CD4+ splenocytes from pre-diabetic female NOD mice, age 74 days, treated by either CD137 or PBS as above (see transfer scheme). The sum of three independent experiments is shown (anti-CD137, ▴, n = 9; PBS, ♦, n = 8), P = 0.0016 by log-rank test. A group receiving CD4+ and CD8+ cells from diabetic NOD donors is shown for comparison of diabetes onset between diabetic and pre-diabetic donors.

FIG. 3.

CD4+ splenocytes from anti-CD137–treated mice ameliorate transfer of diabetes into NOD-scid mice. Female NOD-scid mice received (0.5–2) × 106 diabetic CD8 cells plus 2 × 106 CD4+ splenocytes from pre-diabetic female NOD mice, age 74 days, treated by either CD137 or PBS as above (see transfer scheme). The sum of three independent experiments is shown (anti-CD137, ▴, n = 9; PBS, ♦, n = 8), P = 0.0016 by log-rank test. A group receiving CD4+ and CD8+ cells from diabetic NOD donors is shown for comparison of diabetes onset between diabetic and pre-diabetic donors.

Close modal
FIG. 4.

Anti-CD137 treatment increases IL-4 and IL-10 production and decreases IFN-γ production. Female NOD mice aged 49–55 days were treated with 200 μg anti-CD137 antibody (n = 7) or 200 μg PBS (n = 5) every 3 weeks and studied 1 week after the second CD137 treatment. CD4+ splenocytes were purified by magnetic sorting and stimulated with plate-bound anti-CD3 antibody for 3 days. The supernatants were collected and IFN-γ (i), IL-4 (ii), and IL-10 (iii) concentrations were measured by ELISA. The europium counts for each sample are shown. i, P = 0.0074; ii, P = 0.0074; and iii, P = 0.04 by Mann-Whitney U test.

FIG. 4.

Anti-CD137 treatment increases IL-4 and IL-10 production and decreases IFN-γ production. Female NOD mice aged 49–55 days were treated with 200 μg anti-CD137 antibody (n = 7) or 200 μg PBS (n = 5) every 3 weeks and studied 1 week after the second CD137 treatment. CD4+ splenocytes were purified by magnetic sorting and stimulated with plate-bound anti-CD3 antibody for 3 days. The supernatants were collected and IFN-γ (i), IL-4 (ii), and IL-10 (iii) concentrations were measured by ELISA. The europium counts for each sample are shown. i, P = 0.0074; ii, P = 0.0074; and iii, P = 0.04 by Mann-Whitney U test.

Close modal
FIG. 5.

Anti-CD137 treatment increases the population of cytokine-producing TCR+Dx5+ cells, but anti-CD137–treated CD4+Dx5+ cells do not ameliorate diabetes onset. A: Anti-CD137 treatment significantly increases the numbers of peripheral TCR+DX5+ cells. Female NOD mice aged 44–55 days were treated twice with 200 μg anti-CD137 antibody or PBS at a 3-week interval and studied 1 week after the second injection. Spleen and peripheral lymph nodes were analyzed by fluorescence-activated cell sorting. P = 0.0045 and 0.0012 for spleen and lymph node, respectively, by Mann-Whitney U test. Anti-CD137 (n = 8). B: Increased IL-4 and IL-10 in anti-CD137–treated mice is due to the CD4+Dx5+ (▪) cell subset. Female NOD mice aged 44 days were treated as above and studied at 1 week after the second CD137 treatment. CD4+ and CD4+DX5 (□) splenocytes were purified by magnetic sorting and stimulated with plate-bound anti-CD3 antibody and soluble anti-CD28 antibody for 3 days. Supernatants were analyzed by ELISA for IFN-γ, IL-4, and IL-10 concentrations. One representative of four experiments is shown. C: CD4+Dx5+ cells from anti-CD137–treated NOD mice do not ameliorate disease onset compared with CD4+Dx5 cells. CD4+Dx5+ and CD4+Dx5 cell fractions were sorted by flow cytometry from anti-CD137–treated NOD mice, and recipient NOD-scid mice received either CD4+Dx5 cells alone (solid triangles, n = 8) or CD4+Dx5+ cells combined with CD4+Dx5 cells (solid squares, n = 7), along with diabetic CD8+ cells. Two separate experiments were performed; the Dx5+ cells offered no significant additional protective effect. D: Depletion of Dx5+ cells from naïve pre-diabetic NOD CD4+ cells does not enhance diabetes. Whole CD4+ (2 × 106; inverted triangles) or DX5-depleted CD4+ splenocytes (open squares) were purified by magnetic beads from 63- to 65-day-old female untreated NOD mice, and transferred into female NOD-scid mice with CD8+ splenocytes (2 × 106) from diabetic female NOD mice (n = 7 each group, two separate experiments).

FIG. 5.

Anti-CD137 treatment increases the population of cytokine-producing TCR+Dx5+ cells, but anti-CD137–treated CD4+Dx5+ cells do not ameliorate diabetes onset. A: Anti-CD137 treatment significantly increases the numbers of peripheral TCR+DX5+ cells. Female NOD mice aged 44–55 days were treated twice with 200 μg anti-CD137 antibody or PBS at a 3-week interval and studied 1 week after the second injection. Spleen and peripheral lymph nodes were analyzed by fluorescence-activated cell sorting. P = 0.0045 and 0.0012 for spleen and lymph node, respectively, by Mann-Whitney U test. Anti-CD137 (n = 8). B: Increased IL-4 and IL-10 in anti-CD137–treated mice is due to the CD4+Dx5+ (▪) cell subset. Female NOD mice aged 44 days were treated as above and studied at 1 week after the second CD137 treatment. CD4+ and CD4+DX5 (□) splenocytes were purified by magnetic sorting and stimulated with plate-bound anti-CD3 antibody and soluble anti-CD28 antibody for 3 days. Supernatants were analyzed by ELISA for IFN-γ, IL-4, and IL-10 concentrations. One representative of four experiments is shown. C: CD4+Dx5+ cells from anti-CD137–treated NOD mice do not ameliorate disease onset compared with CD4+Dx5 cells. CD4+Dx5+ and CD4+Dx5 cell fractions were sorted by flow cytometry from anti-CD137–treated NOD mice, and recipient NOD-scid mice received either CD4+Dx5 cells alone (solid triangles, n = 8) or CD4+Dx5+ cells combined with CD4+Dx5 cells (solid squares, n = 7), along with diabetic CD8+ cells. Two separate experiments were performed; the Dx5+ cells offered no significant additional protective effect. D: Depletion of Dx5+ cells from naïve pre-diabetic NOD CD4+ cells does not enhance diabetes. Whole CD4+ (2 × 106; inverted triangles) or DX5-depleted CD4+ splenocytes (open squares) were purified by magnetic beads from 63- to 65-day-old female untreated NOD mice, and transferred into female NOD-scid mice with CD8+ splenocytes (2 × 106) from diabetic female NOD mice (n = 7 each group, two separate experiments).

Close modal
FIG. 6.

CD4+CD25+ cells express CD137; treatment with anti-CD137 boosts CD25+ cell numbers; and anti-CD137–treated CD25+ cells prevent diabetes. A: Naïve NOD CD4+CD25+ cells express increased CD137 compared with CD25 cells. Splenocytes from 50- to 75-day-old NOD mice were analyzed for CD137 expression on CD4+CD25+ and CD4+CD25 cells. One representative of 11 NOD mice is shown. B: Anti-CD137 treatment increases the numbers of CD4+CD25+ cells. Female NOD mice 5–8 weeks of age were treated with 200 μg anti-CD137 antibody (3H3) twice at 3-week intervals, and splenocytes were studied 1 week after the second treatment. The ratio of CD4+CD25+ cells to CD4+ cells is shown. C: Anti-CD137–treated CD25+ cells express Foxp3. Female NOD mice aged 43 days were treated with 200 μg anti-CD137 antibody (3H3) or PBS twice with 3-week intervals and studied at 1 week after the second treatment. Surface expression of CD4 and CD25 and intracellular Foxp3 expression were analyzed. Foxp3 (red) and isotype control (green) are shown. D: Anti-CD137–treated CD25+ cells show suppressive function in vitro. Female NOD mice aged 43 days were treated with 200 μg anti-CD137 antibody (3H3) or PBS twice with 3-week intervals and studied at 1 week after second treatment. CD25+ (5 × 104) or CD25 (5 × 104) cells (from either treated or naïve NOD mice) or both were cultured with irradiated splenocytes and anti-CD3 antibody for 4 days. [3H]thymidine was added during the final 18 h of culture. E: CD25+ cells from anti-CD137–treated mice protect against diabetes. Female NOD mice aged 38–49 days were treated with 200 μg anti-CD137 antibody (3H3) twice at 3-week intervals; at 10 weeks, CD25+ and CD25 populations were sorted, and CD25+ (4–6 × 105) or CD25 (4–6 × 105) splenocytes were transferred to female NOD-scid mice with CD8+ splenocytes from female diabetic NOD mice. Results from two separate experiments are shown; the PBS-treated pre-diabetic CD4+ group is the same as shown in Fig. 3. F: Anti-CD137 antibody specifically binds CD4+CD25+ cells in vivo. Female NOD mice were treated with 200 mg anti-CD137 antibody (3H3) or PBS twice at 3-week intervals and studied at 1 week after the second treatment. Splenocytes were stained with CD4, CD25 (or CD8/CD44 or CD4/Dx5), and anti-IgG2a-bio antibodies followed by strep-avidin phycoeythrin or step-avidin APC. Only CD4+CD25+ cells showed binding by anti-CD137. One representative of five experiments.

FIG. 6.

CD4+CD25+ cells express CD137; treatment with anti-CD137 boosts CD25+ cell numbers; and anti-CD137–treated CD25+ cells prevent diabetes. A: Naïve NOD CD4+CD25+ cells express increased CD137 compared with CD25 cells. Splenocytes from 50- to 75-day-old NOD mice were analyzed for CD137 expression on CD4+CD25+ and CD4+CD25 cells. One representative of 11 NOD mice is shown. B: Anti-CD137 treatment increases the numbers of CD4+CD25+ cells. Female NOD mice 5–8 weeks of age were treated with 200 μg anti-CD137 antibody (3H3) twice at 3-week intervals, and splenocytes were studied 1 week after the second treatment. The ratio of CD4+CD25+ cells to CD4+ cells is shown. C: Anti-CD137–treated CD25+ cells express Foxp3. Female NOD mice aged 43 days were treated with 200 μg anti-CD137 antibody (3H3) or PBS twice with 3-week intervals and studied at 1 week after the second treatment. Surface expression of CD4 and CD25 and intracellular Foxp3 expression were analyzed. Foxp3 (red) and isotype control (green) are shown. D: Anti-CD137–treated CD25+ cells show suppressive function in vitro. Female NOD mice aged 43 days were treated with 200 μg anti-CD137 antibody (3H3) or PBS twice with 3-week intervals and studied at 1 week after second treatment. CD25+ (5 × 104) or CD25 (5 × 104) cells (from either treated or naïve NOD mice) or both were cultured with irradiated splenocytes and anti-CD3 antibody for 4 days. [3H]thymidine was added during the final 18 h of culture. E: CD25+ cells from anti-CD137–treated mice protect against diabetes. Female NOD mice aged 38–49 days were treated with 200 μg anti-CD137 antibody (3H3) twice at 3-week intervals; at 10 weeks, CD25+ and CD25 populations were sorted, and CD25+ (4–6 × 105) or CD25 (4–6 × 105) splenocytes were transferred to female NOD-scid mice with CD8+ splenocytes from female diabetic NOD mice. Results from two separate experiments are shown; the PBS-treated pre-diabetic CD4+ group is the same as shown in Fig. 3. F: Anti-CD137 antibody specifically binds CD4+CD25+ cells in vivo. Female NOD mice were treated with 200 mg anti-CD137 antibody (3H3) or PBS twice at 3-week intervals and studied at 1 week after the second treatment. Splenocytes were stained with CD4, CD25 (or CD8/CD44 or CD4/Dx5), and anti-IgG2a-bio antibodies followed by strep-avidin phycoeythrin or step-avidin APC. Only CD4+CD25+ cells showed binding by anti-CD137. One representative of five experiments.

Close modal
FIG. 7.

Anti-CD137 treatment after transfer of diabetic splenocytes hastens onset of diabetes. Diabetic splenocytes were purified into CD4+ and CD8+ subsets and transferred into NOD-scid recipients. One day after transfer, the NOD-scid recipients were treated with either PBS (n = 5) or anti-CD137 (n = 6); from two separate experiments. Anti-CD137 treatment after transfer of diabetic splenocytes hastens disease onset (P = 0.016). Note: the PBS-treated group here is the same as shown in Fig. 3.

FIG. 7.

Anti-CD137 treatment after transfer of diabetic splenocytes hastens onset of diabetes. Diabetic splenocytes were purified into CD4+ and CD8+ subsets and transferred into NOD-scid recipients. One day after transfer, the NOD-scid recipients were treated with either PBS (n = 5) or anti-CD137 (n = 6); from two separate experiments. Anti-CD137 treatment after transfer of diabetic splenocytes hastens disease onset (P = 0.016). Note: the PBS-treated group here is the same as shown in Fig. 3.

Close modal

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

W.M.R. has received National Institutes of Health Grant RFA-A102-006.

1.
Wicker LS, Todd JA, Peterson LB: Genetic control of autoimmune diabetes in the NOD mouse.
Annu Rev Immunol
13
:
179
–200,
1995
2.
Todd JA, Aitman TJ, Cornall RJ, Ghosh S, Hall JR, Hearne CM, Knight AM, Love JM, McAleer MA, Prins JB, et al.: Genetic analysis of autoimmune type 1 diabetes mellitus in mice.
Nature
351
:
542
–547,
1991
3.
Yui MA, Muralidharan K, Moreno-Altamirano B, Perrin G, Chestnut K, Wakeland EK: Production of congenic mouse strains carrying NOD-derived diabetogenic genetic intervals: an approach for the genetic dissection of complex traits.
Mamm Genome
7
:
331
–334,
1996
4.
Hill NJ, Lyons PA, Armitage N, Todd JA, Wicker LS, Peterson LB: NOD Idd5 locus controls insulitis and diabetes and overlaps the orthologous CTLA4/IDDM12 and NRAMP1 loci in humans.
Diabetes
49
:
1744
–1747,
2000
5.
Wicker LS, Chamberlain G, Hunter K, Rainbow D, Howlett S, Tiffen P, Clark J, Gonzalez-Munoz A, Cumiskey AM, Rosa RL, Howson JM, Smink LJ, Kingsnorth A, Lyons PA, Gregory S, Rogers J, Todd JA, Peterson LB: Fine mapping, gene content, comparative sequencing, and expression analyses support Ctla4 and Nramp1 as candidates for Idd5.1 and Idd5.2 in the nonobese diabetic mouse.
J Immunol
173
:
164
–173,
2004
6.
Lyons PA, Hancock WW, Denny P, Lord CJ, Hill NJ, Armitage N, Siegmund T, Todd JA, Phillips MS, Hess JF, Chen SL, Fischer PA, Peterson LB, Wicker LS: The NOD Idd9 genetic interval influences the pathogenicity of insulitis and contains molecular variants of Cd30, Tnfr2, and Cd137.
Immunity
13
:
107
–115,
2000
7.
Lyons PA, Armitage N, Argentina F, Denny P, Hill NJ, Lord CJ, Wilusz MB, Peterson LB, Wicker LS, Todd JA: Congenic mapping of the type 1 diabetes locus, Idd3, to a 780-kb region of mouse chromosome 3: identification of a candidate segment of ancestral DNA by haplotype mapping.
Genome Res
10
:
446
–453,
2000
8.
Ueda H, Howson JM, Esposito L, Heward J, Snook H, Chamberlain G, Rainbow DB, Hunter KM, Smith AN, Di Genova G, Herr MH, Dahlman I, Payne F, Smyth D, Lowe C, Twells RC, Howlett S, Healy B, Nutland S, Rance HE, Everett V, Smink LJ, Lam AC, Cordell HJ, Walker NM, Bordin C, Hulme J, Motzo C, Cucca F, Hess JF, Metzker ML, Rogers J, Gregory S, Allahabadia A, Nithiyananthan R, Tuomilehto-Wolf E, Tuomilehto J, Bingley P, Gillespie KM, Undlien DE, Ronningen KS, Guja C, Ionescu-Tirgoviste C, Savage DA, Maxwell AP, Carson DJ, Patterson CC, Franklyn JA, Clayton DG, Peterson LB, Wicker LS, Todd JA, Gough SC: Association of the T-cell regulatory gene CTLA4 with susceptibility to autoimmune disease.
Nature
423
:
506
–511,
2003
9.
Vijayakrishnan L, Slavik JM, Illes Z, Greenwald RJ, Rainbow D, Greve B, Peterson LB, Hafler DA, Freeman GJ, Sharpe AH, Wicker LS, Kuchroo VK: An autoimmune disease-associated CTLA-4 splice variant lacking the B7 binding domain signals negatively in T cells.
Immunity
20
:
563
–575,
2004
10.
Cannons JL, Chamberlain G, Howson J, Smink LJ, Todd JA, Peterson LB, Wicker LS, Watts TH: Genetic and functional association of the immune signaling molecule 4-1BB (CD137/TNFRSF9) with type 1 diabetes.
J Autoimmun
25
:
13
–20,
2005
11.
Watts TH: TNF/TNFR family members in costimulation of T cell responses.
Annu Rev Immunol
23
:
23
–68,
2005
12.
Cannons JL, Lau P, Ghumman B, DeBenedette MA, Yagita H, Okumura K, Watts TH: 4-1BB ligand induces cell division, sustains survival, and enhances effector function of CD4 and CD8 T cells with similar efficacy.
J Immunol
167
:
1313
–1324,
2001
13.
Vinay DS, Kwon BS: Role of 4-1BB in immune responses.
Semin Immunol
10
:
481
–489,
1998
14.
Futagawa T, Akiba H, Kodama T, Takeda K, Hosoda Y, Yagita H, Okumura K: Expression and function of 4-1BB and 4-1BB ligand on murine dendritic cells.
Int Immunol
14
:
275
–286,
2002
15.
Laderach D, Wesa A, Galy A: 4-1BB-ligand is regulated on human dendritic cells and induces the production of IL-12.
Cell Immunol
226
:
37
–44,
2003
16.
Seko Y, Takahashi N, Oshima H, Shimozato O, Akiba H, Takeda K, Kobata T, Yagita H, Okumura K, Azuma M, Nagai R: Expression of tumour necrosis factor (TNF) ligand superfamily co-stimulatory molecules CD30L, CD27L, OX40L, and 4-1BBL in murine hearts with acute myocarditis caused by Coxsackievirus B3.
J Pathol
195
:
593
–603,
2001
17.
Seo SK, Choi JH, Kim YH, Kang WJ, Park HY, Suh JH, Choi BK, Vinay DS, Kwon BS: 4-1BB-mediated immunotherapy of rheumatoid arthritis.
Nat Med
10
:
1088
–1094,
2004
18.
Saoulli K, Lee SY, Cannons JL, Yeh WC, Santana A, Goldstein MD, Bangia N, DeBenedette MA, Mak TW, Choi Y, Watts TH: CD28-independent, TRAF2-dependent costimulation of resting T cells by 4-1BB ligand.
J Exp Med
187
:
1849
–1862,
1998
19.
Hurtado JC, Kim YJ, Kwon BS: Signals through 4-1BB are costimulatory to previously activated splenic T cells and inhibit activation-induced cell death.
J Immunol
158
:
2600
–2609,
1997
20.
Dawicki W, Watts TH: Expression and function of 4-1BB during CD4 versus CD8 T cell responses in vivo.
Eur J Immunol
34
:
743
–751,
2004
21.
Foell JL, Diez-Mendiondo BI, Diez OH, Holzer U, Ruck P, Bapat AS, Hoffmann MK, Mittler RS, Dannecker GE: Engagement of the CD137 (4-1BB) costimulatory molecule inhibits and reverses the autoimmune process in collagen-induced arthritis and establishes lasting disease resistance.
Immunology
113
:
89
–98,
2004
22.
Foell J, Strahotin S, O’Neil SP, McCausland MM, Suwyn C, Haber M, Chander PN, Bapat AS, Yan XJ, Chiorazzi N, Hoffmann MK, Mittler RS: CD137 costimulatory T cell receptor engagement reverses acute disease in lupus-prone NZB x NZW F1 mice.
J Clin Invest
111
:
1505
–1518,
2003
23.
Sun Y, Lin X, Chen HM, Wu Q, Subudhi SK, Chen L, Fu YX: Administration of agonistic anti-4-1BB monoclonal antibody leads to the amelioration of experimental autoimmune encephalomyelitis.
J Immunol
168
:
1457
–1465,
2002
24.
Sakaguchi S: Naturally arising CD4+ regulatory T cells for immunologic self-tolerance and negative control of immune responses.
Annu Rev Immunol
22
:
531
–562,
2004
25.
Salomon B, Lenschow DJ, Rhee L, Ashourian N, Singh B, Sharpe A, Bluestone JA: B7/CD28 costimulation is essential for the homeostasis of the CD4+CD25+ immunoregulatory T cells that control autoimmune diabetes.
Immunity
12
:
431
–440,
2000
26.
Wu AJ, Hua H, Munson SH, McDevitt HO: Tumor necrosis factor-alpha regulation of CD4+CD25+ T cell levels in NOD mice.
Proc Natl Acad Sci U S A
99
:
12287
–12292,
2002
27.
Gavin MA, Clarke SR, Negrou E, Gallegos A, Rudensky A: Homeostasis and anergy of CD4(+)CD25(+) suppressor T cells in vivo.
Nat Immunol
3
:
33
–41,
2002
28.
Zheng G, Wang B, Chen A: The 4-1BB costimulation augments the proliferation of CD4+CD25+ regulatory T cells.
J Immunol
173
:
2428
–2434,
2004
29.
Choi BK, Bae JS, Choi EM, Kang WJ, Sakaguchi S, Vinay DS, Kwon BS: 4-1BB-dependent inhibition of immunosuppression by activated CD4+CD25+ T cells.
J Leukoc Biol
75
:
785
–791,
2004
30.
Maerten P, Kwon BS, Shen C, De Hertogh G, Cadot P, Bullens DM, Overbergh L, Mathieu C, Van Assche G, Geboes K, Rutgeerts P, Ceuppens JL: Involvement of 4-1BB (CD137)-4-1BBligand interaction in the modulation of CD4 T cell-mediated inflammatory colitis.
Clin Exp Immunol
143
:
228
–236,
2006
31.
Koarada S, Wu Y, Olshansky G, Ridgway WM: Increased nonobese diabetic Th 
1
:
Th2
(IFN-gamma:IL-4) ratio is CD4+ T cell intrinsic and independent of APC genetic background.
J Immunol
169
:
6580
–6587,
2002
32.
King C, Ilic A, Koelsch K, Sarvetnick N: Homeostatic expansion of T cells during immune insufficiency generates autoimmunity.
Cell
117
:
265
–277,
2004
33.
Irie J, Shimada A, Oikawa Y, Shigihara T, Saruta T: N-acetyl-cysteine accelerates transfer of diabetes into non-obese diabetic scid mice.
Diabetologia
47
:
1803
–1809,
2004
34.
Vinay DS, Choi BK, Bae JS, Kim WY, Gebhardt BM, Kwon BS: CD137-deficient mice have reduced NK/NKT cell numbers and function, are resistant to lipopolysaccharide-induced shock syndromes, and have lower IL-4 responses.
J Immunol
173
:
4218
–4229,
2004
35.
Yang Y, Ueno A, Bao M, Wang Z, Im JS, Porcelli S, Yoon JW: Control of NKT cell differentiation by tissue-specific microenvironments.
J Immunol
171
:
5913
–5920,
2003
36.
Setoguchi R, Hori S, Takahashi T, Sakaguchi S: Homeostatic maintenance of natural Foxp3(+) CD25(+) CD4(+) regulatory T cells by interleukin (IL)-2 and induction of autoimmune disease by IL-2 neutralization.
J Exp Med
201
:
723
–735,
2005
37.
Ono M, Shimizu J, Miyachi Y, Sakaguchi S: Control of autoimmune myocarditis and multiorgan inflammation by glucocorticoid-induced TNF receptor family-related protein(high), Foxp3-expressing CD25+ and CD25− regulatory T cells.
J Immunol
176
:
4748
–4756,
2006
38.
Sytwu HK, Lin WD, Roffler SR, Hung JT, Sung HS, Wang CH, Cheng TL, Tsou SC, Hsi SC, Shen KL: Anti-4-1BB-based immunotherapy for autoimmune diabetes: lessons from a transgenic non-obese diabetic (NOD) model.
J Autoimmun
21
:
247
–254,
2003
39.
Jacob CO, Aiso S, Michie SA, McDevitt HO, Acha-Orbea H: Prevention of diabetes in nonobese diabetic mice by tumor necrosis factor (TNF): similarities between TNF-alpha and interleukin 1.
Proc Natl Acad Sci U S A
87
:
968
–972,
1990
40.
Cope A, Ettinger R, McDevitt H: The role of TNF alpha and related cytokines in the development and function of the autoreactive T-cell repertoire.
Res Immunol
148
:
307
–312,
1997
41.
Green EA, Eynon EE, Flavell RA: Local expression of TNFalpha in neonatal NOD mice promotes diabetes by enhancing presentation of islet antigens.
Immunity
9
:
733
–743,
1998
42.
Morris GP, Chen L, Kong YC: CD137 signaling interferes with activation and function of CD4+CD25+ regulatory T cells in induced tolerance to experimental autoimmune thyroiditis.
Cell Immunol
226
:
20
–29,
2003
43.
Koarada S, Wu Y, Fertig N, Sass DA, Nalesnik M, Todd JA, Lyons PA, Fenyk-Melody J, Rainbow DB, Wicker LS, Peterson LB, Ridgway WM: Genetic control of autoimmunity: protection from diabetes, but spontaneous autoimmune biliary disease in a nonobese diabetic congenic strain.
J Immunol
173
:
2315
–2323,
2004
44.
Irie J, Wu Y, Wicker LS, Rainbow D, Nalesnik MA, Hirsch R, Peterson LB, Leung PS, Cheng C, Mackay IR, Gershwin ME, Ridgway WM: NOD.c3c4 congenic mice develop autoimmune biliary disease that serologically and pathogenetically models human primary biliary cirrhosis.
J Exp Med
203
:
1209
–1219,
2006