In nonobese diabetic (NOD) mice, a deficiency in the number and function of invariant natural killer T-cells (iNKT cells) contributes to the onset of type 1 diabetes. The activation of CD1d-restricted iNKT cells by α-galactosylceramide (α-GalCer) corrects these deficiencies and protects against spontaneous and recurrent type 1 diabetes. Although interleukin (IL)-4 and IL-10 have been implicated in α-GalCer–induced protection from type 1 diabetes, a precise role for these cytokines in iNKT cell regulation of susceptibility to type 1 diabetes has not been identified. Here we use NOD.IL-4–/– and NOD.IL-10–/– knockout mice to further evaluate the roles of IL-4 and IL-10 in α-GalCer–induced protection from type 1 diabetes. We found that IL-4 but not IL-10 expression mediates protection against spontaneous type 1 diabetes, recurrent type 1 diabetes, and prolonged syngeneic islet graft function. Increased transforming growth factor-β gene expression in pancreatic lymph nodes may be involved in α-GalCer–mediated protection in NOD.IL-10–/– knockout mice. Unlike the requirement of IL-7 and IL-15 to maintain iNKT cell homeostasis, IL-4 and IL-10 are not required for α-GalCer–induced iNKT cell expansion and/or survival. Our data identify an important role for IL-4 in the protection against type 1 diabetes by activated iNKT cells, and these findings have important implications for cytokine-based therapy of type 1 diabetes and islet transplantation.

Autoimmune type 1 diabetes spontaneously develops in nonobese diabetic (NOD) mice and is characterized by T-cell infiltration (insulitis) followed by a progressive destruction of pancreatic islet β-cells. Current evidence suggests that immune dysregulation leading to defective regulatory T-cell function elicits type 1 diabetes (1,2). In NOD mice, effector CD4+ T-cells of type 1 diabetes are Th1 cells, and regulatory CD4+ T-cells may include Th2 cells (24). The deficiency in interleukin (IL)-4 production by T-cells mediates immune dysregulation and was proposed to be a causal factor of type 1 diabetes in NOD mice (2,5). Murine IL-4 restores normal NOD T-cell proliferative responsiveness in vitro (1,2,5), and anti-CD28 and IL-4 treatments protect NOD mice from type 1 diabetes (1,2,6). Despite this and other evidence for a role of IL-4 secreting Th2 cells in protection against type 1 diabetes (79), a “protective” role for Th2 cells has not been demonstrated (3). Other regulatory T-cell subsets, including CD4+CD25+ T-cells and invariant natural killer T-cells (iNKT cells) also mediate protection from type 1 diabetes (1014).

iNKT cells are identified by the coexpression of an invariant T-cell receptor (TCR) and various natural killer (NK) cell-related surface markers, including NK1.1, high levels of cytokine (IL-4, interferon [IFN]-γ) production upon activation, and the recognition of glycolipid molecules bound to the nonclassic major histocompatibility complex (MHC) class I molecule CD1d (13,14). TCR-α chains expressed by iNKT cells consist of Vα14-Jα18 gene segments and exhibit a strong bias for Vβ8.2 followed by Vβ2 and Vβ7 in mice. Although the natural ligands recognized by iNKT cells remain unknown, the synthetic glycosphingolipid α-galactosylceramide (α-GalCer) isolated from Agelas mauritanius marine sponges can activate iNKT cells. Upon activation with α-GalCer, iNKT cells can regulate susceptibility to autoimmune disease and tumor surveillance (13,14).

We and others (1318) have shown that deficiencies in iNKT cell number and function mediate the development of type 1 diabetes in NOD mice and that α-GalCer–induced iNKT cell activation corrects these deficiencies and reduces the incidence of spontaneous and recurrent type 1 diabetes in these mice. The latter protection from type 1 diabetes is associated with a polarized Th2 milieu marked by elevated IL-4 and IL-10 and reduced IFN-γ levels in the spleen and pancreas as well as increased IL-10R transcription in the spleen (15,16). Neutralization of IL-10 activity by an anti–IL-10R mAb treatment in vivo abrogates the protective effect of α-GalCer in cyclophosphamide (CY)-induced type 1 diabetes (15). Administration of anti–IL-4 and anti–IL-10 mAbs blocks protection from type 1 diabetes afforded by adoptive transfer of TCR+CD4CD8 thymocytes containing iNKT cells (19). While these observations suggest that protection from type 1 diabetes by α-GalCer is mediated by IL-10 and/or IL-4, the mechanism(s) of iNKT cell regulation of susceptibility to type 1 diabetes remains unknown (14).

Here we use NOD.IL-4–/– and NOD.IL-10–/– mice to show that IL-4 but not IL-10 mediates protection against spontaneous type 1 diabetes, recurrent type 1 diabetes, and prolonged syngeneic islet graft function by α-GalCer–activated iNKT cells. Thus, we have identified an important role for IL-4 in the protection against spontaneous and recurrent type 1 diabetes by α-GalCer–activated CD1d-restricted iNKT cells.

NOD/Del and NOD.Scid mice were bred in a specific pathogen-free barrier facility at The Robarts Research Institute (London, ON, Canada). Insulitis and type 1 diabetes are detectable by 4–6 weeks and 4–6 months of age, respectively, in our colony of female NOD mice. The incidence of type 1 diabetes in this colony is 25–30% at 15 weeks of age and >80% by 25 weeks. NOD.IL-4–/– and NOD.IL-10–/– mice were obtained from The Jackson Laboratory (Bar Harbor, ME), and their knockout phenotypes were confirmed by PCR typing (20).

Assessment of diabetes.

Mice were monitored for type 1 diabetes by measurement of blood glucose levels (BGLs) twice weekly using a Glucometer (Bayer, Toronto, ON). NOD mice that displayed BGL >11.1 mmol/l on two consecutive readings were indicative of the onset of type 1 diabetes.

CY-induced diabetes.

Pre-diabetic (8–9 weeks old) female NOD and NOD.IL10–/– mice were injected intraperitoneally with CY (200 mg/kg) (Sigma, St. Louis, MO) on day 0 and day 10. Diabetes onset was monitored every other day for 30 days postinjection.

Adoptive cell transfer.

NOD and NOD.IL10–/– mice (12–13 weeks old) were treated with α-GalCer or vehicle on days 0, 2, 4, 6, and 8. Spleens were collected on day 10, and spleen single-cell suspensions were prepared. Splenocytes (1 × 107 cells) pooled from each group (n = 5 mice) were transferred intraperitoneally to 5- to 6-week-old NOD.Scid recipient mice (15).

Detection of iNKT cells by flow cytometry.

NOD mice (9–10 weeks old) were treated with α-GalCer or vehicle (kindly supplied by Kirin Brewery Co., Gunma, Japan) on days 0, 2, 4, 6, and 8, as described (15). Tissues were collected on day 10, and spleen and pancreatic lymph node (PLN) single-cell suspensions were prepared. Cells (1 × 106) were treated (15 min, 4°C) with an anti-FcγR mAb (Clone 2.4G2, BD Pharmingen, San Diego, CA) and then stained (20 min, 23°C) with an anti–TCR-β mAb (Clone H57-597, BD Pharmingen) and empty or α-GalCer–loaded CD1 tetramers (kind gift from Dr. M. Kronenberg, La Jolla Institute for Allergy and Immunology, San Diego, CA). Flow cytometry analyses were conducted using a FACSCalibur and CELLQuest software (BD Biosciences, San Jose, CA). The specificity of staining in all tissues was verified by examining tissues from NOD.CD1d−/− mice.

Treatment of mice with α-GalCer.

For protection against spontaneous type 1 diabetes, 10-week-old NOD, NOD.IL-4–/–, and NOD.IL-10–/– mice were treated with α-GalCer (5 μg/dose) every other day for five doses and then boosted at 13–14 weeks of age. For protection against CY-induced diabetes in NOD and NOD.IL-10–/– mice, α-GalCer (5 μg/dose) or vehicle was injected intraperitoneally on days 0, 2, 4, 6, and 8 after CY challenge. For islet transplantation, islet graft recipients were treated with α-GalCer (5 μg/dose) on days −1, 2, 7, 14, and 21, as reported (15).

Islet transplantation.

Pancreatic islets were isolated from 3- to 4-week-old male NOD mice by collagenase digestion and discontinuous density gradient purification. After overnight culture, islets (n = 400) were transplanted to the renal subcapsular space of newly (<7 days) diabetic (spontaneous or CY-induced) female NOD mice (15).

cDNA array analysis.

Total RNA was extracted from PLNs of α-GalCer–or vehicle (5 μg/dose)-treated NOD, NOD.IL-4–/–, and NOD.IL-10–/– mice using a RNeasy mini kit (Qiagen). Aliquots of RNA (2 μg) were used to analyze gene expression using a mouse inflammatory cytokine and receptors GEArray (SuperArray), as described (21). Relative amounts of mRNA transcripts were quantitated using a Molecular Imager System and Molecular Analyst imaging software (BioRad, Hercules, CA) and were estimated by comparing their signal intensities with the signal derived from GAPDH and β-actin. At least a twofold change in signal intensity from two separate experiments was considered significant, as described (21).

Statistical analysis.

Statistical analyses were performed using the Student’s t test (for analyses of NKT cell frequency) and the log rank test (for type 1 diabetes incidence and islet transplantation). P values ≤0.05 were considered to be statistically significant.

α-GalCer protection against spontaneous type 1 diabetes is mediated by IL-4.

Modulation of iNKT cells by α-GalCer reduces the incidence of spontaneous type 1 diabetes in NOD mice even when administered after the onset of invasive insulitis (15,16). We therefore investigated whether α-GalCer treatment initiated at 10 weeks of age protects against spontaneous type 1 diabetes in NOD.IL-4–/– and NOD.IL-10–/– mice. Consistent with our previous report (15), α-GalCer induced protection against type 1 diabetes in NOD mice compared with vehicle-treated mice (P < 0.05), as the incidence of spontaneous type 1 diabetes was reduced from 80 to 42% at 32 weeks of age (Fig. 1A). However, the incidence of type 1 diabetes in α-GalCer–treated NOD.IL-4–/–mice was not significantly reduced compared with vehicle-treated NOD.IL-4–/–mice (85 vs. 65%, P > 0.05). α-GalCer–treated NOD.IL-10–/– mice did not develop type 1 diabetes during 15–25 weeks of age, when colitis is evident in these mice (our unpublished observations). These data suggest that prevention of spontaneous type 1 diabetes by α-GalCer is mediated by IL-4 and not IL-10.

α-GalCer protects against CY-induced type 1 diabetes in NOD.IL-10–/− mice.

Previously, we reported that α-GalCer–mediated protection against CY-induced type 1 diabetes is associated with the ability of splenocytes from CY-challenged and α-GalCer–treated female NOD mice to secrete more IL-4 and IL-10 and less IFN-γ than splenocytes from vehicle-treated mice upon in vitro restimulation with α-GalCer (15). To further evaluate the significance of increased IL-10 levels induced upon iNKT cell activation, as NOD.IL-10–/– mice spontaneously develop colitis starting at 12–15 weeks of age, we investigated whether NOD mice deficient in IL-10 expression are protected from CY-induced type 1 diabetes upon treatment with α-GalCer. Consistent with our previous study, Fig. 2A shows that α-GalCer treatment protects from CY-induced type 1 diabetes in NOD mice, with the incidence of type 1 diabetes being 35% in α-GalCer–treated NOD mice and 75% in vehicle-treated NOD mice (P < 0.01), respectively. NOD.IL-10–/– mice were also protected against CY-induced type 1 diabetes (P < 0.01) (Fig. 2B), and the level of protection exceeded that detected in NOD wild-type (Fig. 2A) mice (P < 0.01). α-GalCer treatment did not protect from type 1 diabetes in NOD.CD1d–/– mice, which are deficient in iNKT cells (Fig. 2C). An adoptive transfer model of type 1 diabetes was also used to test the role of IL-10 in the development of type 1 diabetes upon iNKT cell activation. Similar to our previous report that spleen cells from α-GalCer–treated NOD mice have a diminished capacity to transfer type 1 diabetes to NOD.Scid mice (15), spleen cells from NOD.IL-10–/– mice harvested 2 weeks after α-GalCer treatment also demonstrated a reduced capacity to transfer type 1 diabetes (Figs. 3A and B). Thus, in contrast to the requirement for IL-4 in GalCer-mediated protection against type 1 diabetes, IL-10 does not appear to be required for this protection.

α-GalCer–induced prolongation of syngeneic islet graft depends on the activity of IL-4 but not IL-10.

Prolongation of pancreaticoduodenal graft survival in nonrecurrent spontaneous diabetic BB rats is associated with the proliferation of donor-derived iNKT cells in hepatic and splenic tissues and higher serum levels of IL-4 (22). The acceptance of xenogeneic islet grafts in mice is also dependent on iNKT cells (23). iNKT cells therefore appear to enhance transplanted islet syngeneic graft and xenograft survival.

Previously, we showed that α-GalCer treatment prolongs graft function significantly in newly diagnosed diabetic NOD recipients of syngeneic islet transplants (15). Since a recurrent autoimmune response in islet graft recipients may be prevented by treatment with IL-4 and IL-10 (24), we determined whether IL-4 and/or IL-10 mediates the ability of α-GalCer–activated iNKT cells to prolong syngeneic islet graft survival and prevent recurrent type 1 diabetes. While islet grafts failed in 90% of control vehicle-treated NOD spontaneous diabetic recipients by day 8 posttransplantation, grafts in 90% of α-GalCer–treated NOD mice were still functional at this time, as evidenced by the maintenance of euglycemia (Fig. 4A). In contrast, treatment of newly diagnosed NOD.IL-4–/– diabetic mice treated with α-GalCer did not prolong graft function in islet transplant recipients compared with vehicle-treated NOD.IL-4–/– diabetic mice (Fig. 4B). Moreover, whereas 100% of islet grafts failed by day 12 posttransplantation in all vehicle-treated NOD and NOD.IL-4–/– mice and α-GalCer–treated NOD.IL-4–/– mice, islet grafts survived as long as 25 days in ∼40% of α-GalCer–treated NOD mice (Fig. 4A). However, the lack of IL-4 expression does not accelerate graft rejection, and syngeneic islet grafts survive in vehicle-treated NOD.IL-4−/− mice for a period similar to that in vehicle-treated NOD mice. Since NOD.IL-10–/– mice spontaneously develop colitis starting at 12–15 weeks of age, we used CY-induced diabetic mice as islet graft recipients. All islet grafts in α-GalCer–treated CY-induced NOD.IL-10–/– diabetic mice retained function at 20–30 days posttransplant, but these grafts failed within 10 days posttransplant in all vehicle-treated NOD.IL-10–/– mice (Fig. 4C). Some α-GalCer–treated NOD.IL-10–/– mice with functional islet grafts had to be sacrificed due to the development of colitis. Removal of grafts in NOD and NOD.IL-10–/– mice with long surviving grafts resulted in a prompt return to a hyperglycemic state (Figs. 5A and B), which indicated that the maintenance of euglycemia in α-GalCer–treated diabetic recipient mice is islet graft dependent. Thus, α-GalCer–prolonged syngeneic islet graft function occurs in an IL-4–dependent and IL-10–independent manner.

α-GalCer–treated NOD.IL-4–/– and NOD.IL-10–/– mice differ in their cytokine gene expression profile compared with wild-type NOD mice.

To further determine how IL-4 and not IL-10 mediates α-GalCer–mediated protection from type 1 diabetes, we conducted cDNA array analyses of the gene expression of inflammatory cytokines in the PLNs of NOD.IL-4−/− and NOD.IL-10–/– mice. Compared with wild-type NOD mice, vehicle-treated NOD.IL-4−/− mice show an increased expression of IL-10Rβ, CXCR5, CCR6, CCR7, TARC, BLC, MIG, and SDF-1 and decreased expression of CXCR-4 (Table 2). In contrast, vehicle-treated NOD.IL-10–/– mice show an increased expression of IL-2, CXCR5, CCR7, Scya6, BLC, MIG, SDF-1, and tumor necrosis factor (TNF)-R1 and a decreased expression of IL-11 and CXCR4 compared with NOD mice. Interestingly, transforming growth factor (TGF)-β gene expression was found to be elevated in vehicle-treated NOD.IL-10–/– mice compared with NOD and NOD.IL-4−/− mice. In general, the gene expression profiles in α-GalCer–treated NOD.IL-4−/− and -NOD.IL-10–/– mice were very similar to those from vehicle-treated control mice, with the exception that IL-11 gene expression was decreased in NOD.IL-4−/− mice (Table 2). NOD.IL-10–/– mice displayed increased IL-6R, MDC, MCP-2, and TGF-β expression when compared with NOD mice (Table 2). Given that IL-11 and TGF-β elicit protection against type 1 diabetes (2527) and that SDF-1/CXCR4 interaction and LT-β mediate the development of type 1 diabetes (28,29), our data suggest that the α-GalCer–mediated protection seen in NOD.IL-10–/– mice may be due to the increased expression of TGF-β. Elevated TGF-β expression may compensate for deficient IL-10 expression and thus decrease the incidence of type 1 diabetes. On the other hand, decreased expression of IL-11 and increased expression of SDF-1/CXCR4 and LT-β may be associated with deficient IL-4 expression and contribute to the requirement of IL-4 in α-GalCer–mediated protection from type 1 diabetes.

IL-4 and IL-10 are not required for the expansion or survival of iNKT cells.

To determine whether IL-4 or IL-10 is required for α-GalCer to promote the expansion and/or survival of iNKT cells in secondary lymphoid organs, we examined the frequency of iNKT cells in the spleen and PLNs of α-GalCer–treated and –untreated NOD.IL-4–/– and NOD.IL-10–/– mice. iNKT cells were tracked by double-staining with an anti-TCRβ mAb and CD1d tetramers loaded with α-GalCer (15). The frequencies of iNKT cells in the spleen and PLNs of wild-type NOD and all three knockout NOD mouse strains were increased significantly after 8 days of α-GalCer treatment compared with the frequencies observed in vehicle-treated mice (P < 0.01). However, no significant differences were observed in the frequencies of iNKT cells in the spleens of the α-GalCer–treated wild-type and cytokine knockout NOD mice (Table 1, experiment 1). Whereas α-GalCer stimulated about a twofold increase in the frequency of iNKT cells in the PLN of NOD.IL-4–/– mice relative to that in NOD.IL-10–/– and wild-type NOD mice, the frequencies observed in NOD and NOD.IL-10–/– mice were similar (P > 0.05). No significant differences were detected in the spleen and PLNs of vehicle-treated mice (Table 1, experiment 1). Similar frequencies of iNKT cells were also found in the spleen and PLNs of NOD and NOD.IL-10–/– mice at 30 days after CY and α-GalCer treatment (Table 1, experiment 2). Thus, unlike the requirement of IL-7 and IL-15 to maintain iNKT cell homeostasis (30), IL-4 and IL-10 do not appear to be required for α-GalCer–induced iNKT cell expansion and/or survival.

This study provides direct evidence for the requirement of IL-4 but not IL-10 in the protection against spontaneous type 1 diabetes and prolongation of syngeneic islet graft function by α-GalCer–activated iNKT cells. In the case of IL-10, this observation is consistent with the preliminary results of a recent report that IL-10 may not be necessary for iNKT-mediated protection against type 1 diabetes induced by transfer of diabetogenic BDC2.5 T-cells to Vα14Cα–/– NOD mice (31).

Curiously, a deficiency of Vα14+ iNKT cells in CD1d-deficient NOD mice is causally linked to the induction of type 1 diabetes, and yet this Vα14+ iNKT cell deficiency does not alter the Th1 and Th2 cytokine profiles in these mice (14). Moreover, an accelerated accumulation of CCR4+ diabetogenic T-cells in pancreatic islets is associated with Vα14+ iNKT cell dysfunction, indicating that Vα14+ iNKT cells may have another important role in the prevention of autoaggressive T-cell recruitment to sites of inflammation (32). It is noteworthy that mature myeloid CD8α dendritic cells (DCs) accumulate in the PLNs only after treatment with α-GalCer, and that this accumulation protects against type 1 diabetes. Transfer of these myeloid DCs into NOD mice significantly protects them against type 1 diabetes (17). Therefore, an immunoregulatory role for Vα14+ iNKT cells in the recruitment of tolerogenic myeloid DCs (CD8α myeloid DC) to PLN, rather than a Th1/Th2 cytokine imbalance, is suggested (14,17).

Unexpectedly, our findings appear to differ from our previous report that neutralization of IL-10 activity by an anti–IL-10R antibody in vivo abrogates the protective effect of α-GalCer in CY-induced type 1 diabetes (15). One explanation for this difference may be that the timing of anti–IL-10R administration or production of IL-10 may differ during the development of type 1 diabetes, as reflected by an altered biological activity of IL-10 in vivo. Another explanation is that IL-10 can exhibit seemingly paradoxical effects on susceptibility to type 1 diabetes in NOD mice. Systemic treatment of young NOD mice with IL-10 protects against type 1 diabetes (33,34). In contrast, anti–IL-10 mAb treatment of young NOD mice reduces the severity of insulitis (35). Whereas BALB/c mice that express an IL-10 transgene in their islet β-cells do not develop type 1 diabetes, NOD.IL-10 mice that express an IL-10 transgene in islet β-cells develop type 1 diabetes at an accelerated rate (36,37). IL-10 contributes early to the pathology of type 1 diabetes via a CD8+ T-cell–dependent pathway, as anti-CD8 antibody–mediated depletion of CD8+ T-cells inhibits the onset of type 1 diabetes without attenuating the severity of insulitis (38). In addition, IL-10 can also block the regulatory activity in vitro of a CD3+CD4CD8 T-cell subset (39), amplify CD4+ T-cell production of the islet β-cell destructive IFN-γ and TNF-α cytokines (40), exacerbate graft rejection (4143), and mediate the development of certain autoimmune lymphoproliferative disorders (37,44,45). These sets of findings raise the possibilities that IL-10 may inhibit and/or delete activated regulatory T-cells, such as iNKT cells, and that a lack of IL-10 expression in NOD.IL-10−/− mice may enhance the level of protection from type 1 diabetes induced by iNKT cell activation. Further experimentation is required to test these possibilities.

In addition, our cDNA array data indicate that the increased level of TGF-β gene expression in the PLNs of α-GalCer–treated NOD.IL-10–/– mice compared with α-GalCer–or vehicle-treated NOD and NOD.IL-4–/– mice may be responsible for the protection observed in these mice. Consistent with our findings in IL-10–deficient NOD mice, Sturlan et al. (46) observed increased plasma levels of TGF-β in IL-10–deficient mice when studying colitis. Recently, a role for TGF-β in NKT cell function was described, as Gansuvd et al. (47) demonstrated that rhesus NKT cells can secrete large amounts of TGF-β and are in a semianergic state that leads to polarization toward a Th3 regulatory cell phenotype with regulatory/suppressive function in vitro. It is interesting that we observed a greater increase in TGF-β gene expression in IL-10–deficient NOD mice relative to NOD mice after α-GalCer–induced iNKT activation, which suggests that activated iNKT cells might overexpress TGF-β in the absence of IL-10 expression. This may be explained by the observation that IL-10 and TGF-β tend to be cosecreted at sites of inflammation. A study by Kitani et al. (48) using intranasal administration of a TGF-β–encoding plasmid showed that while TGF-β rapidly induces IL-10 production, IL-10 does not induce the secretion of TGF-β, suggesting that these two cytokines might be coordinately regulated. Thus, in α-GalCer–treated NOD.IL-10–/– mice, an increase in TGF-β expression in the PLNs may either compensate for the absence of expression of IL-10 or may be the result of a lack of negative regulation induced by the cytokine. In contrast, in α-GalCer–treated NOD.IL-4–/– mice, a diminished level of IL-11 expression is detectable and no compensatory effect of TGF-β in the PLNs is apparent. This may explain why α-GalCer–treated NOD.IL-4–/– mice are not protected from type 1 diabetes and further supports the notion that α-GalCer–mediated protection is IL-4 dependent.

A recent report demonstrated that CXCR5/BLC and CCR7/MDC interactions are crucial factors in the development of autoimmunity (49,50). We found that expression of CXCR5, BLC, CCR7, and MDC are elevated in vehicle-treated NOD.IL-4–/– and NOD.IL-10–/– mice compared with vehicle-treated NOD mice. Nonetheless, we did not observe any accelerated onset of spontaneous type 1 diabetes in these mice. On the other hand, complete protection was obtained in α-GalCer–treated NOD.IL-10–/– mice. Thus, it is possible that increased CXCR5/BLC and CCR7/MDC interactions do not mediate the lack of protection from type 1 diabetes in α-GalCer–treated NOD.IL-4–/– mice.

Our observations also differ from the result that α-GalCer is unable to protect B6.IL-4–/– and B6.IL-10–/– mice against experimental autoimmune encephalomyelitis (51). Therefore, the mechanism of iNKT cell–dependent protection from different autoimmune diseases may depend on the genetic background in which a given disease develops.

In new onset diabetic NOD mice, the grafting of syngeneic islets in the renal subcapsular area restores euglycemia. However, due to a recurrent autoimmune response, the graft is rejected within 10 days of transplantation and hyperglycemia returns. Renal subcapsular islet grafts in NOD mice are infiltrated predominantly by Th1 cells, and various types of immunomodulation (e.g., treatment with IL-4, CFA, or insulin) elicit a Th1- to Th2-type shift and suppress recurrent type 1 diabetes (2). Here we show that α-GalCer–prolonged syngeneic islet graft function occurs in an IL-4–dependent and IL-10–independent manner, similar to the ability of IL-4 but not IL-10 to mediate α-GalCer–induced protection against spontaneous type 1 diabetes. Thus, due to these differential cytokine requirements, activated iNKT cells may operate via different mechanisms to restore tolerance during the development of spontaneous and recurrent type 1 diabetes.

FIG. 1.

Protection against spontaneous type 1 diabetes by α-GalCer is mediated by IL-4. Female NOD (A) and NOD.IL-4–/– (B) mice were treated every other day with α-GalCer (•, n = 16) or vehicle (▪, n = 15) (5 μg/mouse) starting at 10 weeks of age for 2 weeks and received a booster of α-GalCer (5 μg/mouse) at 14 weeks of age. Mice were screened for glycemia twice weekly starting at 14 weeks of age. The difference observed in the incidence of type 1 diabetes at 32 weeks of age is significant between α-GalCer–and vehicle-treated NOD mice (P < 0.05) but not between α-GalCer–and vehicle-treated NOD.IL-4–/– mice (P > 0.05).

FIG. 1.

Protection against spontaneous type 1 diabetes by α-GalCer is mediated by IL-4. Female NOD (A) and NOD.IL-4–/– (B) mice were treated every other day with α-GalCer (•, n = 16) or vehicle (▪, n = 15) (5 μg/mouse) starting at 10 weeks of age for 2 weeks and received a booster of α-GalCer (5 μg/mouse) at 14 weeks of age. Mice were screened for glycemia twice weekly starting at 14 weeks of age. The difference observed in the incidence of type 1 diabetes at 32 weeks of age is significant between α-GalCer–and vehicle-treated NOD mice (P < 0.05) but not between α-GalCer–and vehicle-treated NOD.IL-4–/– mice (P > 0.05).

Close modal
FIG. 2.

Protection against CY-induced diabetes by α-GalCer is not mediated by IL-10. Female NOD (A), NOD.IL-10–/– (B), and NOD.CD1d–/– (C) mice (8–9 weeks old, n = 8–15) were challenged with CY (200 mg/kg) at day 0 and day 10 and received α-GalCer (•) or vehicle (▪) (5 μg · mouse−1 · dose−1) intraperitoneally at days 0, 2, 4, 6, and 8 after CY challenge. Mice were screened for glycemia every other day starting at day 15, and mice with BGLs of >11.1 mmol/l on two consecutive readings were considered diabetic. Data from one of three representative and reproducible experiments are shown.

FIG. 2.

Protection against CY-induced diabetes by α-GalCer is not mediated by IL-10. Female NOD (A), NOD.IL-10–/– (B), and NOD.CD1d–/– (C) mice (8–9 weeks old, n = 8–15) were challenged with CY (200 mg/kg) at day 0 and day 10 and received α-GalCer (•) or vehicle (▪) (5 μg · mouse−1 · dose−1) intraperitoneally at days 0, 2, 4, 6, and 8 after CY challenge. Mice were screened for glycemia every other day starting at day 15, and mice with BGLs of >11.1 mmol/l on two consecutive readings were considered diabetic. Data from one of three representative and reproducible experiments are shown.

Close modal
FIG. 3.

Spleen cells from α-GalCer–treated NOD and NOD.IL-10–/– mice do not transfer type 1 diabetes. Splenocytes (107 cells) from individual α-GalCer–treated (•, n = 8) and vehicle-treated (▪, n = 8) NOD (A) and NOD.IL-10–/– (B) mice (11–12 weeks old) were collected 2 weeks after the last injection of α-GalCer and transferred into NOD.Scid recipients (n = 8–10). Mice were screened for glycemia twice weekly starting at 5 weeks post transfer. Data are from one of two representative and reproducible experiments.

FIG. 3.

Spleen cells from α-GalCer–treated NOD and NOD.IL-10–/– mice do not transfer type 1 diabetes. Splenocytes (107 cells) from individual α-GalCer–treated (•, n = 8) and vehicle-treated (▪, n = 8) NOD (A) and NOD.IL-10–/– (B) mice (11–12 weeks old) were collected 2 weeks after the last injection of α-GalCer and transferred into NOD.Scid recipients (n = 8–10). Mice were screened for glycemia twice weekly starting at 5 weeks post transfer. Data are from one of two representative and reproducible experiments.

Close modal
FIG. 4.

Prolongation of syngeneic islet graft function by α-GalCer treatment requires IL-4 but not IL-10 expression. A and B: Prolongation of islet graft survival conferred by α-GalCer is dependent on IL-4 expression. Newly diagnosed spontaneous diabetic female NOD (A, n = 10) and NOD.IL-4–/– (B, n = 5–6) mice were transplanted under the kidney capsule with ∼400 syngeneic islets from 3- to 4-week-old male NOD donors. Graft recipients were treated with 5 μg/dose of vehicle or α-GalCer on days –1, 2, 7, 14, and 21. Recipients were monitored every other day for their BGLs. C: Prolongation of islet graft induced by α-GalCer treatment is not dependent on IL-10 expression. Newly diagnosed CY-induced diabetic female NOD.IL-10–/– mice were transplanted with islets, treated with α-GalCer (n = 6) or vehicle (n = 4), and monitored for their BGLs as described in A and B above.

FIG. 4.

Prolongation of syngeneic islet graft function by α-GalCer treatment requires IL-4 but not IL-10 expression. A and B: Prolongation of islet graft survival conferred by α-GalCer is dependent on IL-4 expression. Newly diagnosed spontaneous diabetic female NOD (A, n = 10) and NOD.IL-4–/– (B, n = 5–6) mice were transplanted under the kidney capsule with ∼400 syngeneic islets from 3- to 4-week-old male NOD donors. Graft recipients were treated with 5 μg/dose of vehicle or α-GalCer on days –1, 2, 7, 14, and 21. Recipients were monitored every other day for their BGLs. C: Prolongation of islet graft induced by α-GalCer treatment is not dependent on IL-10 expression. Newly diagnosed CY-induced diabetic female NOD.IL-10–/– mice were transplanted with islets, treated with α-GalCer (n = 6) or vehicle (n = 4), and monitored for their BGLs as described in A and B above.

Close modal
FIG. 5.

Maintenance of euglycemia in α-GalCer–treated diabetic recipient mice is islet graft dependent. Removal of grafts in α-GalCer–treated NOD (A) and NOD.IL-10–/– (B) mice with long surviving grafts resulted in a prompt return to a hyperglycemic state. Unilateral nephrectomy to remove the grafts from mice treated with α-GalCer (see Figs. 2A and C) is denoted by narrows.

FIG. 5.

Maintenance of euglycemia in α-GalCer–treated diabetic recipient mice is islet graft dependent. Removal of grafts in α-GalCer–treated NOD (A) and NOD.IL-10–/– (B) mice with long surviving grafts resulted in a prompt return to a hyperglycemic state. Unilateral nephrectomy to remove the grafts from mice treated with α-GalCer (see Figs. 2A and C) is denoted by narrows.

Close modal
TABLE 1

Frequency of iNKT cells in the spleen and PLNs

StrainSpleen
PLN
α-GalCerVehicleα-GalCerVehicle
Experiment 1*     
 NOD.IL-4−/− 1.3 ± 0.3 0.7 ± 0.2 2.8 ± 0.4 0.7 ± 0.2 
 NOD.IL-10−/− 1.4 ± 0.2 0.6 ± 0.2 1.5 ± 0.2 0.6 ± 0.1 
 NOD 1.4 ± 0.4 0.6 ± 0.1 1.5 ± 0.2 0.7 ± 0.1 
Experiment 2     
 NOD.IL-10−/− 1.2 ± 0.3 0.7 ± 0.2 1.3 ± 0.2 0.4 ± 0.2 
 NOD 1.3 ± 0.3 0.6 ± 0.2 1.4 ± 0.2 0.5 ± 0.1 
StrainSpleen
PLN
α-GalCerVehicleα-GalCerVehicle
Experiment 1*     
 NOD.IL-4−/− 1.3 ± 0.3 0.7 ± 0.2 2.8 ± 0.4 0.7 ± 0.2 
 NOD.IL-10−/− 1.4 ± 0.2 0.6 ± 0.2 1.5 ± 0.2 0.6 ± 0.1 
 NOD 1.4 ± 0.4 0.6 ± 0.1 1.5 ± 0.2 0.7 ± 0.1 
Experiment 2     
 NOD.IL-10−/− 1.2 ± 0.3 0.7 ± 0.2 1.3 ± 0.2 0.4 ± 0.2 
 NOD 1.3 ± 0.3 0.6 ± 0.2 1.4 ± 0.2 0.5 ± 0.1 

Data are means ± SD.

*

Female NOD, NOD.IL-4−/−, and NOD.IL-10−/− mice (9–10 weeks old, n = 5–7) were treated with vehicle or α-GalCer (5 μg/dose) on days 0, 2, 4, 6, and 8. On day 10, spleen and PLNs were collected, and iNKT cells were stained with α-GalCer–loaded CD1d tetramers and anti–TCR-β.

P < 0.01 vs. NOD and NOD.IL-10−/− mice.

Female NOD and NOD.IL-10−/− mice (8–9 weeks old) were challenged with CY (200 mg/kg) on day 0 and day 10 and received α-GalCer or vehicle (5 μg·mouse−1·dose−1 i.p.) on days 0, 2, 4, 6, and 8 after CY challenge. On day 30, spleen and PLNs were collected from nondiabetic mice and double-stained with an anti–TCR-β antibody and CD1d tetramers loaded with α-GalCer.

TABLE 2

Gene expression in the PLN of α-GalCer–or vehicle-treated NOD.IL-4−/−, NOD.IL-10−/−, and NOD mice

Mice treated with α-GalCer
Mice treated with vehicle
IL-4−/−/NODIL-10−/−/NODIL-10−/−/IL-4−/−IL-4−/−/NODIL-10−/−/NODIL-10−/−/IL-4−/−
IL-2 4.8 2.2 −2.2 NS 
IL-6R NS 55 17 NS NS NS 
IL-10Rβ NS −3.5 12 NS 10 
IL-11 −3 NS 2.5 NS −4 −6 
IL-15Rα NS NS 2.5 NS NS 3.5 
IL-16 NS NS 2.5 NS NS −4 
CXCR5 40 70 NS 100 30 −3 
CXCR4 NS NS −2.8 −5 NS 
CCR6 NS 20 10 NS NS 
CCR7 20 20 NS 2.5 −2.5 
Scya5 NS NS −3 
Scya6 81 100 NS 775 300 −2.5 
TARC NS NS −5 
MDC NS 20 10 NS NS NS 
MCP-2 NS 25 14 NS NS NS 
BLC 23 NS 
IP-9 NS NS NS NS 
MIG NS 10 
SDF-1 NS 
TNFR1 3.5 NS NS NS 
LT-β NS NS NS NS NS 
TGFβ1 NS 2.9 NS 2.1 2.5 
Mice treated with α-GalCer
Mice treated with vehicle
IL-4−/−/NODIL-10−/−/NODIL-10−/−/IL-4−/−IL-4−/−/NODIL-10−/−/NODIL-10−/−/IL-4−/−
IL-2 4.8 2.2 −2.2 NS 
IL-6R NS 55 17 NS NS NS 
IL-10Rβ NS −3.5 12 NS 10 
IL-11 −3 NS 2.5 NS −4 −6 
IL-15Rα NS NS 2.5 NS NS 3.5 
IL-16 NS NS 2.5 NS NS −4 
CXCR5 40 70 NS 100 30 −3 
CXCR4 NS NS −2.8 −5 NS 
CCR6 NS 20 10 NS NS 
CCR7 20 20 NS 2.5 −2.5 
Scya5 NS NS −3 
Scya6 81 100 NS 775 300 −2.5 
TARC NS NS −5 
MDC NS 20 10 NS NS NS 
MCP-2 NS 25 14 NS NS NS 
BLC 23 NS 
IP-9 NS NS NS NS 
MIG NS 10 
SDF-1 NS 
TNFR1 3.5 NS NS NS 
LT-β NS NS NS NS NS 
TGFβ1 NS 2.9 NS 2.1 2.5 

Total RNA was extracted from the PLNs of α-GalCer–or vehicle-treated mice and was used to analyze gene expression by GEArray. A greater than twofold change in both two separate experiments was considered significant. The average expression value was shown. NS, less than a twofold change.

Q.-S.M. and D.L. contributed equally to this work.

Q.-S.M. is currently affiliated with the Center for Biotechnology and Genomic Medicine, Departments of Pathology and Medicine, Medical College of Georgia, Augusta, Georgia.

This work was supported by grants (to T.L.D.) from the Juvenile Diabetes Research Foundation, Ontario, Research and Development Challenge Fund, Canadian Institutes of Health Research, and London Health Sciences Center Multi-Organ Transplant Program. T.L.D. is the Sheldon H. Weinstein Scientist in Diabetes at the University of Western Ontario.

We thank Dr. M. Kronenberg for kindly providing empty and α-GalCer–loaded CD1 tetramers, Kirin Brewery Co. for kindly supplying α-GalCer or vehicle, and all members of our laboratory for their advice and encouragement.

1
Bergerot I, Arreaza G, Cameron M, Chou H, Delovitch TL: Role of T-cell anergy and suppression in susceptibility to IDDM.
Res Immunol
148
:
348
–358,
1997
2
Delovitch TL, Singh B: The nonobese diabetic mouse as a model of autoimmune diabetes: immune dysregulation gets the NOD.
Immunity
7
:
727
–738,
1997
3
Katz JD, Benoist C, Mathis D: T helper cell subsets in insulin-dependent diabetes.
Science
268
:
1185
–1188,
1995
4
Rabinovitch A: Immunoregulatory and cytokine imbalances in the pathogenesis of IDDM: therapeutic intervention by immunostimulation?
Diabetes
43
:
613
–621,
1994
5
Cameron MJ, Arreaza GA, Delovitch TL: Cytokine- and costimulation-mediated therapy of IDDM.
Crit Rev Immunol
17
:
537
–544,
1997
6
Cameron MJ, Arreaza GA, Zucker P, Chensue SW, Strieter RM, Chakrabarti S, Delovitch TL: IL-4 prevents insulitis and insulin-dependent diabetes mellitus in nonobese diabetic mice by potentiation of regulatory T helper-2 cell function.
J Immunol
159
:
4686
–4692,
1997
7
Serreze DV: Autoimmune diabetes results from genetic defects manifest by antigen presenting cells.
FASEB J
7
:
1092
–1096,
1993
8
Bergerot I, Arreaza GA, Cameron MJ, Burdick MD, Strieter RM, Chensue SW, Chakrabarti S, Delovitch TL: Insulin B-chain reactive CD4+ regulatory T-cells induced by oral insulin treatment protect from type 1 diabetes by blocking the cytokine secretion and pancreatic infiltration of diabetogenic effector T-cells.
Diabetes
48
:
1720
–1729,
1999
9
Homann D, Holz A, Bot A, Coon B, Wolfe T, Petersen J, Dyrberg TP, Grusby MJ, von Herrath MG: Autoreactive CD4+ T cells protect from autoimmune diabetes via bystander suppression using the IL-4/Stat6 pathway.
Immunity
11
:
463
–472,
1999
10
Kukreja A, Cost G, Marker J, Zhang C, Sun Z, Lin-Su K, Ten S, Sanz M, Exley M, Wilson B, Porcelli S, Maclaren N: Multiple immuno-regulatory defects in type-1 diabetes.
J Clin Invest
109
:
131
–140,
2002
11
Bach, JF: Regulatory T cells under scrutiny.
Nat Rev Immunol
3
:
189
–198,
2003
12
Bluestone JA, Abbas AK: Natural versus adaptive regulatory T cells.
Nat Rev Immunol
3
:
253
–257,
2003
13
Sharif S, Arreaza GA, Zucker P, Mi QS, Delovitch TL: Regulation of autoimmune disease by natural killer T cells.
J Mol Med
80
:
290
–300,
2002
14
Wilson SB, Delovitch TL: Janus-like role of regulatory iNKT cells in autoimmune disease and tumour immunity.
Nat Rev Immunol
3
:
211
–222,
2003
15
Sharif S, Arreaza GA, Zucker P, Mi QS, Sondhi J, Naidenko OV, Kronenberg M, Koezuka Y, Delovitch TL, Gombert JM, Leite-De-Moraes M, Gouarin C, Zhu R, Hameg A, Nakayama T, Taniguchi M, Lepault F, Lehuen A, Bach JF, Herbelin A: Activation of natural killer T cells by alpha-galactosylceramide treatment prevents the onset and recurrence of autoimmune type 1 diabetes.
Nat Med
7
:
1057
–1062,
2001
16
Hong S, Wilson MT, Serizawa I, Wu L, Singh N, Naidenko OV, Miura T, Haba T, Scherer DC, Wei J, Kronenberg M, Koezuka Y, Van Kaer L: The natural killer T-cell ligand alpha-galactosylceramide prevents autoimmune diabetes in non-obese diabetic mice.
Nat Med
7
:
1052
–1056,
2001
17
Naumov YN, Bahjat KS, Gausling R, Abraham R, Exley MA, Koezuka Y, Balk SB, Strominger JL, Clare-Salzer M, Wilson SB: Activation of CD1d-restricted T cells protects NOD mice from developing diabetes by regulating dendritic cell subsets.
Proc Natl Acad Sci U S A
98
:
13838
–13843,
2001
18
Wang B, Geng YB, Wang CR: CD1-restricted NK T cells protect nonobese diabetic mice from developing diabetes.
J Exp Med
194
:
313
–320,
2001
19
Hammond KJ, Poulton LD, Palmisano LJ, Silveira PA, Godfrey DI, Baxter AG: alpha/beta-T cell receptor (TCR)+CD4-CD8- (NKT) thymocytes prevent insulin-dependent diabetes mellitus in nonobese diabetic (NOD)/Lt mice by the influence of interleukin (IL)-4 and/or IL-10.
J Exp Med
187
:
1047
–1056,
1998
20
Serreze DV, Chapman HD, Post CM, Johnson EA, Suarez-Pinzon WL, Rabinovitch A: Th1 to Th2 cytokine shifts in nonobese diabetic mice: sometimes an outcome, rather than the cause, of diabetes resistance elicited by immunostimulation.
J Immunol
166
:
1352
–1359,
2001
21
Mi QS, Ly D, Lamhamedi-Cherradi SE, Salojin KV, Zhou L, Grattan M, Meagher C, Zucker P, Chen YH, Nagle J, Taub D, Delovitch TL: Blockade of tumor necrosis factor-related apoptosis-inducing ligand exacerbates type 1 diabetes in NOD mice.
Diabetes
52
:
1967
–1975,
2003
22
Tori M, Ito T, Kitagawa-Sakakida S, Nozawa M, Matsuda H, Shirakura R: Importance of donor-derived lymphocytes in the protection of pancreaticoduodenal or islet grafts from recurrent autoimmunity: a role for RT6+NKR-P1+ T cells.
Transplantation
70
:
32
–38,
2000
23
Ikehara Y, Yasunami Y, Kodama S, Maki T, Nakano M, Nakayama T, Taniguchi M, Ikeda S: CD4(+) Valpha14 natural killer T cells are essential for acceptance of rat islet xenografts in mice.
J Clin Invest
105
:
1761
–1767,
2000
24
Rabinovitch A, Suarez-Pinzon WL, Sorensen O, Rajotte RV, Power RF: Combination therapy with cyclosporine and interleukin-4 or interleukin-10 prolongs survival of synergeneic pancreatic islet grafts in nonobese diabetic mice: islet graft survival does not correlate with mRNA levels of type 1 or type 2 cytokines, or transforming growth factor-beta in the islet grafts.
Transplantation
64
:
1525
–1531,
1997
25
Nicoletti F, Zaccone P, Conget I, Gomis R, Moller C, Meroni PL, Bendtzen K, Trepicchio W, Sandler S: Early prophylaxis with recombinant human interleukin-11 prevents spontaneous diabetes in NOD mice.
Diabetes
48
:
2333
–2339,
1999
26
Green EA, Gorelik L, McGregor CM, Tran EH, Flavell RA: CD4+CD25+ T regulatory cells control anti-islet CD8+ T cells through TGF-beta-TGF-beta receptor interactions in type 1 diabetes.
Proc Natl Acad Sci U S A
100
:
10878
–10883,
2003
27
Belghith M, Bluestone JA, Barriot S, Megret J, Bach JF, Chatenoud L: TGF-beta-dependent mechanisms mediate restoration of self-tolerance induced by antibodies to CD3 in overt autoimmune diabetes.
Nat Med
9
:
1202
–1208,
2003
28
Matin K, Salam MA, Akhter J, Hanada N, Senpuku H: Role of stromal-cell derived factor-1 in the development of autoimmune diseases in non-obese diabetic mice.
Immunology
107
:
222
–232,
2002
29
Bachmann MF: Blocking lymphotoxin: a potential therapy for diabetes?
Trends Immunol
22
:
420
,
2001
30
Matsuda JL, Gapin L, Sidobre S, Kieper WC, Tan JT, Ceredig R, Surh CD, Kronenberg M: Homeostasis of V alpha 14i NKT cells.
Nat Immunol
3
:
966
–974,
2002
31
Beaudoin L, Laloux V, Novak J, Lucas B, Lehuen A: NKT cells inhibit the onset of diabetes by impairing the development of pathogenic T cells specific for pancreatic beta cells.
Immunity
17
:
725
–736,
2002
32
Shi FD, Flodstrom M, Balasa B, Kim SH, Van Gunst K, Strominger JL, Wilson SB, Sarvetnick N: Germ line deletion of the CD1 locus exacerbates diabetes in the NOD mouse.
Proc Natl Acad Sci U S A
98
:
6777
–6782,
2001
33
Pennline KJ, Roque-Gaffney E, Monahan M: Recombinant human IL-10 prevents the onset of diabetes in the nonobese diabetic mouse.
Clin Immunol Immunopathol
71
:
169
–175,
1994
34
Zheng XX, Steele AW, Hancock WW, Stevens AC, Nickerson PW, Roy-Chaudhury P, Tian Y, Strom TB: A noncytolytic IL-10/Fc fusion protein prevents diabetes, blocks autoimmunity, and promotes suppressor phenomena in NOD mice.
J Immunol
158
:
4507
–4513,
1997
35
Lee MS, Mueller R, Wicker LS, Peterson LB, Sarvetnick N: IL-10 is necessary and sufficient for autoimmune diabetes in conjunction with NOD MHC homozygosity.
J Exp Med
183
:
2663
–2668,
1996
36
Wogensen L, Huang X, Sarvetnick N: Leukocyte extravasation into the pancreatic tissue in transgenic mice expressing interleukin 10 in the islets of Langerhans.
J Exp Med
178
:
175
–185,
1993
37
Balasa B, Van Gunst K, Jung N, Balakrishna D, Santamaria P, Hanafusa T, Itoh N, Sarvetnick N: Islet-specific expression of IL-10 promotes diabetes in nonobese diabetic mice independent of Fas, perforin, TNF receptor-1, and TNF receptor-2 molecules.
J Immunol
165
:
2841
–2849,
2000
38
Balasa B, Davies JD, Lee J, Good A, Yeung BT, Sarvetnick N: IL-10 impacts autoimmune diabetes via a CD8+ T cell pathway circumventing the requirement for CD4+ T and B lymphocytes.
J Immunol
161
:
4420
–4427,
1998
39
Zhang ZX, Stanford WL, Zhang L: Ly-6A is critical for the function of double negative regulatory T cells.
Eur J Immunol
32
:
1584
–1592,
2002
40
Moore KW, de Waal MR, Coffman RL, O’Garra A: Interleukin-10 and the interleukin-10 receptor.
Annu Rev Immunol
19
:
683
–765,
2001
41
Qian S, Li W, Li Y, Fu F, Lu L, Fung JJ, Thomson AW: Systemic administration of cellular interleukin-10 can exacerbate cardiac allograft rejection in mice.
Transplantation
62
:
1709
–1714,
1996
42
Li W, Fu F, Lu L, Narula SK, Fung JJ, Thomson AW, Qian S: Systemic administration of anti-interleukin-10 antibody prolongs organ allograft survival in normal and presensitized recipients.
Transplantation
66
:
1587
–1596,
1998
43
Furukawa Y, Becker G, Stinn JL, Shimizu K, Libby P, Mitchell RN: Interleukin-10 (IL-10) augments allograft arterial disease: paradoxical effects of IL-10 in vivo.
Am J Pathol
155
:
1929
–1939,
1999
44
Yanagi K, Haneji N, Hamano H, Takahashi M, Higashiyama H, Hayashi Y: In vivo role of IL-10 and IL-12 during development of Sjogren’s syndrome in MRL/lpr mice.
Cell Immunol
168
:
243
–250,
1996
45
Watanabe N, Ikuta K, Nisitani S, Chiba T, Honjo T: Activation and differentiation of autoreactive B-1 cells by interleukin 10 induce autoimmune hemolytic anemia in Fas-deficient antierythrocyte immunoglobulin transgenic mice.
J Exp Med
196
:
141
–146,
2002
46
Sturlan S, Oberhuber G, Beinhauer BG, Tichy B, Kappel S, Wang J, Rogy MA: Interleukin-10-deficient mice and inflammatory bowel disease associated cancer development.
Carcinogenesis
22
:
665
–671,
2001
47
Gansuvd B, Hubbard WJ, Hutchings A, Thomas FT, Goodwin J, Wilson SB, Exley MA, Thomas JM: Phenotypic and functional characterization of long-term cultured rhesus macaque spleen-derived NKT cells.
J Immunol
171
:
2904
–2911,
2003
48
Kitani A, Fuss I, Nakamura K, Kumaki F, Usui T, Strober W: Transforming growth factor (TGF)-{beta}1-producing regulatory T cells induce Smad-mediated interleukin 10 secretion that facilitates coordinated immunoregulatory activity and amelioration of TGF-{beta}1-mediated fibrosis.
J Exp Med
198
:
1179
–1188,
2003
49
Muller G, Hopken UE, Lipp M: The impact of CCR7 and CXCR5 on lymphoid organ development and systemic immunity.
Immunol Rev
195
:
117
–135,
2003
50
Luther SA, Lopez T, Bai W, Hanahan D, Cyster JG: BLC expression in pancreatic islets causes B cell recruitment and lymphotoxin-dependent lymphoid neogenesis.
Immunity
12
:
471
–481,
2000
51
Singh AK, Wilson MT, Hong S, Olivares-Villagomez D, Du C, Stanic AK, Joyce S, Sriram S, Koezuka Y, Van Kaer L: Natural killer T cell activation protects mice against experimental autoimmune encephalomyelitis.
J Exp Med
194
:
1801
–1811,
2001