Islet allograft survival limits the long-term success of islet transplantation as a potential curative therapy for type 1 diabetes. A number of factors compromise islet survival, including recurrent diabetes. We investigated whether CD39, an ectonucleotidase that promotes the generation of extracellular adenosine, would mitigate diabetes in the T cell–mediated multiple low-dose streptozotocin (MLDS) model. Mice null for CD39 (CD39KO), wild-type mice (WT), and mice overexpressing CD39 (CD39TG) were subjected to MLDS. Adoptive transfer experiments were performed to delineate the efficacy of tissue-restricted overexpression of CD39. The role of adenosine signaling was examined using mutant mice and pharmacological inhibition. The susceptibility to MLDS-induced diabetes was influenced by the level of expression of CD39. CD39KO mice developed diabetes more rapidly and with higher frequency than WT mice. In contrast, CD39TG mice were protected. CD39 overexpression conferred protection through the activation of adenosine 2A receptor and adenosine 2B receptor. Adoptive transfer experiments indicated that tissue-restricted overexpression of CD39 conferred robust protection, suggesting that this may be a useful strategy to protect islet grafts from T cell–mediated injury.

Pancreas or islet transplantation is a potentially curative therapy for patients with type 1 diabetes (T1D), eliminating the need for exogenous insulin and the inherent risks of life-threatening hyperglycemic and hypoglycemic episodes. Unlike the complex surgery of whole pancreas transplantation, isolated islets are delivered percutaneously into the portal circulation. However such transplanted islets face several challenges. The first is an immediate response, termed the instant blood-mediated inflammatory reaction (IBMIR), involving platelet binding, activation of the complement and coagulation cascades, and infiltration of islets by monocytes and neutrophils. Further loss of islets occurs within a day of transplantation, mediated by activated hepatic natural killer cells and neutrophils (1). Islets surviving these innate immune processes are then subject to the alloimmune response and recurrent autoimmunity.

Recipients of transplanted islets are patients with T1D and as such are already primed for islet autoimmunity. Recurrent autoimmune disease has been demonstrated in animal models and in humans after either islet or pancreatic transplantation (2) and is intertwined with the alloimmune response. There is some evidence that individuals who are particularly prone to autoimmunity are more likely to experience rapid allograft rejection (3). Further, an increase or re-emergence of islet autoantibodies has been observed despite immunosuppression (4,5), irrespective of complete human leukocyte antigen match (6) or mismatch (7).

In animal models, recurrent autoimmune disease potentiates the immune response, resulting in earlier graft rejection after intraportal islet transplantation (8). Indeed, strategies that have been demonstrated to prolong islet allograft survival in chemically induced (high-dose streptozotocin) diabetic recipients have failed to do so in autoimmune NOD mice (9). These data suggest that recurrent autoimmunity can destroy islet allografts even in the absence of an alloimmune response. Although the mechanism of recurrent autoimmunity is not fully defined, T cells are involved primarily through major histocompatibility complex class II recognition (10).

We are interested in the role of purinergic catabolism and adenosine signaling in immune and thrombotic responses to islet transplantation. CD39 is a membrane-bound enzyme that hydrolyzes extracellular purinergic nucleotides, including the platelet agonist ADP. We have generated mice overexpressing CD39 (CD39TG) from the H-2Kb promoter, which directs expression to all nucleated cells. Ubiquitous expression has been demonstrated on circulating cells by flow cytometry, and throughout the tissues (including on β-cells) by immunohistochemistry (11,12). We have previously shown that the overexpression of CD39 on murine islets attenuated thrombosis when the islets were mixed with human blood (11), and this approach has been heralded as a potential anti-IBMIR strategy (13). The objective of this study was to examine the impact of CD39 overexpression on diabetes induction using the multiple low-dose streptozotocin (MLDS) model, as a prelude to investigations using CD39TG islets in transplantation where recurrent diabetes may compromise graft survival. Although it shares fewer features with human T1D than the NOD model, we used the MLDS model because it is more convenient to investigate the effect of various genetic modifications and its validity as a model of T cell–mediated diabetes has recently been confirmed (14).

The overexpression of CD39 on the islet surface does not impact glucose homeostasis (11) and thus such islets would be potentially suitable for transplantation. In addition to preventing IBMIR, the overexpression of CD39 on islets may confer additional downstream benefits for islet grafts attenuating T cell–mediated islet graft loss from recurrent autoimmunity and allograft rejection. The end product of CD39’s catalytic activity is AMP, and overexpression of CD39 increases the level of AMP available for conversion to adenosine (12) by the ecto-enzyme CD73 (15). Extracellular adenosine binds to four specific receptors (A1R, A2AR, A2BR, and A3R), promoting a range of effects that are predominantly anti-inflammatory (16). Studies using CD39KO mice have identified a role for ectonucleotidase expression in the regulation of organ-protective adenosine receptor signaling (17). Consistent with this, mice overexpressing CD39 are protected from renal ischemia-reperfusion injury (IRI) by an A2AR-dependent mechanism (18). Kidney isografts from these mice performed significantly better after prolonged cold storage than control isografts, indicating that organ-restricted overexpression of CD39 was sufficient for this protective effect (18). Adenosine receptor signaling influences the development of diabetes in nontransplant settings in the NOD and MLDS models. Activation of adenosine receptors by the nonspecific agonist 5′-N-ethylcarboxamidoadenosine (NECA) ameliorated diabetes in both models (19). In the MLDS model, A1R and A3R agonists had a modest protective effect compared with NECA, whereas the A2AR agonist had no effect. The A2BR antagonist reversed the effect of NECA, suggesting that A2BR signaling played the predominant role in regulating MLDS-induced diabetes (19).

In this study, we hypothesized that the level of CD39 expression may affect susceptibility to T cell–dependent diabetes by influencing the rate of proinflammatory extracellular nucleotide catabolism and subsequent generation of anti-inflammatory adenosine. CD39TG and CD39KO were examined for their response to MLDS and compared with wild-type (WT) controls. To examine the utility of tissue-restricted expression of CD39, which is of direct relevance to clinical transplantation, adoptive transfer of bone marrow (BM) between selected lines was performed prior to MLDS treatment. Finally, we examined the importance of adenosine receptor signaling by using mutant mice and pharmacological inhibition of the adenosine receptors.

Mice.

Male C57BL/6 CD39TG (12), CD39KO (20), A2AR null (A2ARKO) (21), and WT mice were maintained in the BioResources Centre (St. Vincent’s Hospital) and fed ad libitum with commercial rodent diet. All animal experiments were approved by the St. Vincent’s Hospital Animal Ethics Committee.

Induction of diabetes.

Mice were injected intraperitoneally with 50 mg streptozotocin (Sigma-Aldrich, Sydney, NSW, Australia) per kilogram body weight (BW) daily for 5 days (22). Nonfasting blood glucose levels (BGLs) in tail vein blood were measured using an AccuChek glucometer (Roche, Hawthorn, VIC, Australia). Two consecutive nonfasting BGL readings between 15 and 20 mmol/L or ≥20 mmol/L were considered hyperglycemic and diabetic, respectively.

Isolation of pancreatic islets.

Pancreata were perfused with ice-cold Hank’s buffered saline solution with 0.45 mg/mL collagenase P (Roche), incubated at 37°C for 15 min, and passed through a 500-μm nylon mesh. Digested pancreata were washed with RPMI. Islets separated by density gradient centrifugation in histopaque-RPMI (Life Technologies, Mulgrave, VIC, Australia) were picked manually.

Adoptive transfer experiments.

Male mice (4–5 weeks old) were subjected to 5 Gy of irradiation and received 5 × 106 BM cells intravenously from age-matched mice the next day. Mice received MLDS treatment 5 weeks later.

Immunohistochemistry.

Paraffin-embedded pancreas sections (4 μm) were stained with hematoxylin and eosin (H&E). After antigen retrieval, sections were stained with polyclonal guinea pig anti-insulin antibody (Dako, Campbellfield, VIC, Australia) and rabbit anti–guinea pig-horseradish peroxidase (Dako) or monoclonal rabbit anti-CD3+ antibody (Abcam, Cambridge, U.K.) and biotinylated donkey anti-rabbit antibody (GE Healthcare, Rydalmere, NSW, Australia). Sections were then developed using 3,3′-diaminobenzidine. TUNEL staining was performed with the Klenow FragEL DNA fragmentation detection kit (Merck Serono, Frenchs Forest, NSW, Australia).

Flow cytometry.

Peripheral blood leukocytes were incubated with fluorescein isothiocyanate–conjugated anti-human CD39 (Ancell, Australia) and analyzed using a FACSCalibur flow cytometer and CellQuest software.

In vitro cytokine treatment.

Hand-picked islets (n = 100) were cultured for 72 h with combinations of mouse interleukin-1β (IL-1β) (100 units/mL), mouse tumor necrosis factor-α (1,000 units/mL), and mouse interferon-γ (1,000 units/mL). After cytokine treatment, islets were incubated for 1 h at 37°C with 0.5 mg/mL 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide. After addition of DMSO, the concentration of formazan produced by viable cells was measured at an absorbance of 550 nm. Results were calculated as percentage viability versus untreated control wells.

KT3 treatment.

Mice were administered intraperitoneally with a 0.75-mg dose of the T cell–depleting anti-CD3 monoclonal antibody (KT3) (23) at days −2 and 0, followed by MLDS from day 0 to 4. T-cell numbers were assessed at days −3, 15, and 30.

Pharmacological agents.

A2BR inhibitor (A2BRi) PSB1115 (0.5 mg/kg BW b.i.d.; Tocris Bioscience, Bristol, U.K.), A1R inhibitor PSB-36 (0.5 mg/kg BW b.i.d.; Sigma-Aldrich), and A1R agonist CCPA (0.1 mg/kg BW daily; Sigma-Aldrich) were administered intraperitoneally.

Reverse transcription quantitative real-time PCR.

Quantitative real-time PCR (qRT-PCR) was performed on an ABI 7500 Fast Real-Time PCR System (Life Technologies). Islet RNA was isolated using the PureLink RNA Mini Kit (Life Technologies). Complementary DNA was generated by reverse transcription using primer-probe sets (IL-1β, Mm01336189_m1; IL-6, Mm00446190_m1; intercellular adhesion molecule-1 (ICAM-1), Mm00516023_m1; inducible nitric oxide synthase (iNOS), Mm00440502_m1; CD3γ, Mm00438095_m1; A1R, Mm01308023_m1; A2AR, Mm00802075_m1; A2BR, Mm01285229_s1; A3R, Mm00802076_m1) and TaqMan universal PCR Mastermix (Life Technologies). Gene expression was analyzed against the reference gene 18S (24). Relative expression was calculated as 2−∆Ct (where ∆ denotes the change in the threshold cycle [Ct]), and fold change (as calculated by 2−∆∆Ct) was determined against the WT untreated islets.

Statistical analysis.

Results are expressed as mean ± SEM. The data were analyzed by Student t test and one-way ANOVA test.

The level of CD39 expression modulates susceptibility to MLDS-induced diabetes.

After MLDS treatment, the mean BGL of WT mice rose steadily after day 6, reaching hyperglycemic levels (15.8 ± 1.2 mmol/L) by day 12 and progressing to diabetes (21.2 ± 2.4 mmol/L) by day 42. CD39KO mice exhibited a heightened susceptibility to MLDS, with a faster and more pronounced rise in BGL. In contrast, CD39TG mice were significantly protected (Fig. 1A). Incidence of diabetes was 71% for WT, 100% for CD39KO, and 14% for CD39TG mice (Fig. 1B).

FIG. 1.

Expression levels of CD39 are inversely related to the susceptibility to MLDS-induced diabetes. Nonfasting BGLs (mean ± SEM) (A) and diabetes incidence (B) in CD39KO (▲, n = 6), WT (■, n = 7), and CD39TG (●, n = 6) mice after MLDS. WT mice treated with citrate buffer (X, n = 2). ***P < 0.001 vs. MLDS-treated WT mice. Diabetes incidence (C) of MLDS-treated chimeric CD39KOWTBM (▲, n = 4) and WTWTBM (□, n = 6) mice. ns, not significant.

FIG. 1.

Expression levels of CD39 are inversely related to the susceptibility to MLDS-induced diabetes. Nonfasting BGLs (mean ± SEM) (A) and diabetes incidence (B) in CD39KO (▲, n = 6), WT (■, n = 7), and CD39TG (●, n = 6) mice after MLDS. WT mice treated with citrate buffer (X, n = 2). ***P < 0.001 vs. MLDS-treated WT mice. Diabetes incidence (C) of MLDS-treated chimeric CD39KOWTBM (▲, n = 4) and WTWTBM (□, n = 6) mice. ns, not significant.

Close modal

CD39KO mice have both hepatic insulin resistance, which portends to glucose intolerance (25), and hyperproliferative T cells secondary to dysfunctional regulatory T cells (26). To investigate whether these features contributed to the heightened MLDS response, CD39KO mice were sublethally irradiated and adoptively transferred with WT BM (CD39KOWTBM) prior to treatment with MLDS. Diabetes incidence in CD39KOWTBM was similar to that of WTWTBM mice (Fig. 1C), suggesting that in this model, the dysfunctional T-cell phenotype due to CD39 deficiency rather than hepatic insulin resistance was responsible for the increased susceptibility to diabetes.

To confirm the role of T cells in this model, WT mice were depleted of T cells prior to MLDS. These mice showed a significant reduction in the incidence of diabetes (Supplementary Fig. 1). In a separate experiment, MLDS-treated mice were killed at the onset of diabetes for histological and immunohistochemical analysis of the pancreas. For WT and CD39KO mice, this was performed when BGL first exceeded 20 mmol/L (days 24 and 10, respectively); for CD39TG mice, which never reached this level, the pancreas was harvested on day 30. Although the architecture of islets from MLDS-treated WT mice was largely preserved (Fig. 2A, i), a reduction in the intensity of insulin staining (Fig. 2A, ii) and a CD3+ T-cell infiltrate were observed (Fig. 2A, iii). Consistent with the more severe diabetes in MLDS-treated CD39KO mice, islets from these mice showed disrupted morphology (Fig. 2A, iv), weaker insulin staining (Fig. 2A, v), and a heavier cellular infiltrate (Fig. 2A, vi). In contrast, the architecture of CD39TG islets was preserved (Fig. 2A, vii) and they stained strongly and uniformly for insulin (Fig. 2A, viii), with minimal CD3+ T-cell infiltration (Fig. 2A, ix). The mean insulitis scores were 47, 33, and 20% for the CD39KO, WT, and CD39TG animals, respectively.

FIG. 2.

Expression of CD39 reduces T-cell infiltration and proinflammatory cytokine gene expression. Pancreata were harvested from WT and CD39KO mice when mean BGL exceeded 20 mmol/L (days 24 and 10, respectively), and from CD39TG mice at day 30, and were stained with H&E (A, i, iv, and vii) and insulin (ii, v, and viii). Presence of T cells was determined by CD3+ immunohistochemistry (A, iii, vi, and ix). Arrows indicate presence of leukocytes. RNA from islets isolated from untreated (UT) or MLDS-treated (MLDS) mice was analyzed by qRT-PCR. Expression of CD3γ relative to 18S (B). Fold change of IL-1β (C), IL-6 (D), ICAM-1 (E), and iNOS (F). *P < 0.05. ND, not detected; ns, not significant.

FIG. 2.

Expression of CD39 reduces T-cell infiltration and proinflammatory cytokine gene expression. Pancreata were harvested from WT and CD39KO mice when mean BGL exceeded 20 mmol/L (days 24 and 10, respectively), and from CD39TG mice at day 30, and were stained with H&E (A, i, iv, and vii) and insulin (ii, v, and viii). Presence of T cells was determined by CD3+ immunohistochemistry (A, iii, vi, and ix). Arrows indicate presence of leukocytes. RNA from islets isolated from untreated (UT) or MLDS-treated (MLDS) mice was analyzed by qRT-PCR. Expression of CD3γ relative to 18S (B). Fold change of IL-1β (C), IL-6 (D), ICAM-1 (E), and iNOS (F). *P < 0.05. ND, not detected; ns, not significant.

Close modal

CD39 overexpression prevents islet T-cell infiltration and inflammatory gene expression after MLDS.

Cytokines released from T cells are critical in the progression of T1D through the inhibition of β-cell function and promotion of apoptosis. Islets were isolated from untreated or MLDS-treated WT and CD39TG mice 10–12 days after MLDS treatment, the time of maximum insulitis in this model (27), and analyzed by qRT-PCR. Expression of the specific T-cell marker CD3γ was detected only in islets from treated WT mice (Fig. 2B), consistent with the T-cell staining pattern (Fig. 2A, iii). Expression of the proinflammatory cytokines IL-1β and IL-6, the adhesion molecule ICAM-1, and the cytokine-inducible enzyme iNOS was significantly upregulated in WT islets after MLDS but was not significantly changed by MLDS in CD39TG islets (Fig. 2C–F).

Overexpression of CD39 on tissues is sufficient to protect against MLDS.

CD39TG mice have an enhanced capacity to generate adenosine (12), and isolated islets from these mice have increased nucleoside triphosphate diphosphohydrolase (NTPDase) catalytic activity (11). CD39TG mice also have a selective partial CD4+ T-cell lymphopenia, and the residual T cells are hypoproliferative (28). Adoptive transfer experiments were used to investigate whether the reduced susceptibility of CD39TG mice was due to the CD4+ T-cell lymphopenia or to the effects of CD39 expression on tissues. Transfer of WT BM into sublethally irradiated CD39TG mice produced chimeric mice with CD39 overexpression restricted to the tissues (CD39TGWTBM). Successful reconstitution was verified by flow cytometric analysis of peripheral blood leukocytes (Supplementary Fig. 2). WTWTBM chimeric mice were used as controls. The reconstitution of CD39TG BM in irradiated WT (WTCD39TGBM) or CD39TG recipient mice (CD39TGCD39TGBM) was incomplete (data not shown; 28), and these mice were not further examined.

CD39TGWTBM chimeric mice were resistant to MLDS (Fig. 3A), and their islets exhibited normal morphology and insulin staining 30 days after MLDS treatment (Fig. 3B). Protection did not appear to be due to an inherent resistance of the islets to proinflammatory cytokines, because islets isolated from CD39TG mice were equally susceptible to proinflammatory cytokines in vitro as islets from WT mice (Fig. 3C). We speculate that in vivo, increased CD39 activity in the tissues enhances adenosine generation, tempering the host’s immune response and subsequent T-cell infiltration of islets. To investigate this possibility, CD39TG mice were adoptively transferred with BM from CD39KO or A2ARKO mice, both of which have hyperproliferative T cells (26), and treated with MLDS. Chimeric CD39TGCD39KOBM and CD39TGA2ARKOBM mice remained resistant to MLDS (Fig. 3D and E).

FIG. 3.

Overexpression of CD39 on tissues mediates protection in MLDS diabetes. A: Diabetes incidence of WTWTBM (□, n = 6) and CD39TGWTBM (●, n = 6) mice. ***P < 0.001. B: H&E (i) and insulin staining (ii) of islets from CD39TGWTBM mice 30 days after MLDS. C: Viability of WT (black columns) and CD39TG (white columns) islets after exposure to cytokines as indicated. Means of three independent experiments ± SEM. ns, not significant. D: Diabetes incidence of CD39TGCD39KOBM (●, n = 8) and CD39KOCD39KOBM (△, n = 8) mice. ***P < 0.001. E: Diabetes incidence of CD39TGA2ARKOBM (●, n = 5) and A2ARKOA2ARKOBM (◇, n = 4) mice. ***P < 0.001.

FIG. 3.

Overexpression of CD39 on tissues mediates protection in MLDS diabetes. A: Diabetes incidence of WTWTBM (□, n = 6) and CD39TGWTBM (●, n = 6) mice. ***P < 0.001. B: H&E (i) and insulin staining (ii) of islets from CD39TGWTBM mice 30 days after MLDS. C: Viability of WT (black columns) and CD39TG (white columns) islets after exposure to cytokines as indicated. Means of three independent experiments ± SEM. ns, not significant. D: Diabetes incidence of CD39TGCD39KOBM (●, n = 8) and CD39KOCD39KOBM (△, n = 8) mice. ***P < 0.001. E: Diabetes incidence of CD39TGA2ARKOBM (●, n = 5) and A2ARKOA2ARKOBM (◇, n = 4) mice. ***P < 0.001.

Close modal

The protective effect of CD39 overexpression is mediated by A2 receptor signaling.

Adenosine signals via four receptors, the expression of which has been examined in whole pancreas (19). Given that the A2AR is involved in β-cell regeneration (29), we sought to define the expression of these receptors at the transcript level in isolated islets (eliminating acinar and vascular tissue) from untreated and MLDS-treated mice. Basal expression of A1R (Fig. 4A) and A2AR (Fig. 4B) was similar in CD39TG and WT islets, but expression of A2BR was significantly higher in CD39TG (Fig. 4C). MLDS treatment did not significantly affect expression of A1R or A2AR in either mouse line (Fig. 4A and B). The A2BR was upregulated by MLDS in WT but not CD39TG islets (Fig. 4C). No A3R expression was detected on islets from either line, irrespective of treatment.

FIG. 4.

Expression of adenosine receptors in isolated islets. RNA from islets isolated at day 10 from untreated (UT) or MLDS-treated (MLDS) mice was analyzed by qRT-PCR for relative expression of A1R (A), A2AR (B), and A2BR (C). Data are expressed as means of four independent experiments ± SEM. *P < 0.05. ns, not significant.

FIG. 4.

Expression of adenosine receptors in isolated islets. RNA from islets isolated at day 10 from untreated (UT) or MLDS-treated (MLDS) mice was analyzed by qRT-PCR for relative expression of A1R (A), A2AR (B), and A2BR (C). Data are expressed as means of four independent experiments ± SEM. *P < 0.05. ns, not significant.

Close modal

Antagonism or agonism of the A1R did not alter the susceptibility of the WT mice to MLDS-induced diabetes (Supplementary Fig. 3); however, deletion of A2AR increased susceptibility. Given the hyperproliferative T-cell response of A2ARKO mice (26), A2ARKO mice were adoptively transferred with WT BM, generating chimeric A2ARKOWTBM mice that were treated with MLDS. The reverse experiment (A2ARKO BM into WT mice: WTA2ARKOBM) was also performed. Deletion of the A2AR on either the tissues or the circulating cells conferred increased susceptibility to MLDS-induced diabetes (Fig. 5B).

FIG. 5.

Protection by overexpression of CD39 is mediated by A2 receptor signaling. A: Diabetes incidence of A2ARKO (♦, n = 4), WT (■, n = 4), CD39TG (●, n = 4), and A2ARKO/CD39TG (fx5.1, n = 7) mice. **P < 0.01 vs. WT; ***P < 0.001 vs. WT; ns, not significant vs. WT. B: Diabetes incidence of A2ARKOWTBM (♦, n = 5), WTA2ARKOBM (■, n = 8), A2ARKOA2ARKOBM (◇, n = 4), and WTWTBM (□, n = 4) mice. *P < 0.05, WTA2ARKOBM vs. WTWTBM; **P < 0.01, A2ARKOWTBM vs. WTWTBM. C: Hyperglycemia incidence of WT + saline (■, n = 8), CD39TG + saline (●, n = 8), WT + A2BRi (□, n = 8), and CD39TG + A2BRi (○, n = 8) mice. *P < 0.05, WT + A2BRi vs. WT + saline (days 6–12) (i); **P < 0.01, CD39TG + A2BRi vs. CD39TG + saline (ii). ns, not significant. D: Diabetes incidence of WT + saline (■, n = 8) and WT + A2BRi (□, n = 8) mice. *P < 0.05. E: Diabetes incidence of A2ARKO/CD39TG + saline (fx5.2, n = 4) and A2ARKO/CD39TG + A2BRi (fx5.1, n = 9) mice. ns, not significant.

FIG. 5.

Protection by overexpression of CD39 is mediated by A2 receptor signaling. A: Diabetes incidence of A2ARKO (♦, n = 4), WT (■, n = 4), CD39TG (●, n = 4), and A2ARKO/CD39TG (fx5.1, n = 7) mice. **P < 0.01 vs. WT; ***P < 0.001 vs. WT; ns, not significant vs. WT. B: Diabetes incidence of A2ARKOWTBM (♦, n = 5), WTA2ARKOBM (■, n = 8), A2ARKOA2ARKOBM (◇, n = 4), and WTWTBM (□, n = 4) mice. *P < 0.05, WTA2ARKOBM vs. WTWTBM; **P < 0.01, A2ARKOWTBM vs. WTWTBM. C: Hyperglycemia incidence of WT + saline (■, n = 8), CD39TG + saline (●, n = 8), WT + A2BRi (□, n = 8), and CD39TG + A2BRi (○, n = 8) mice. *P < 0.05, WT + A2BRi vs. WT + saline (days 6–12) (i); **P < 0.01, CD39TG + A2BRi vs. CD39TG + saline (ii). ns, not significant. D: Diabetes incidence of WT + saline (■, n = 8) and WT + A2BRi (□, n = 8) mice. *P < 0.05. E: Diabetes incidence of A2ARKO/CD39TG + saline (fx5.2, n = 4) and A2ARKO/CD39TG + A2BRi (fx5.1, n = 9) mice. ns, not significant.

Close modal

A2ARKO mice were bred with CD39TG mice to generate A2ARKO/CD39TG mice (Supplementary Fig. 4). A2ARKO/CD39TG mice were not protected from MLDS-induced diabetes (Fig. 5A), implicating a role for A2AR signaling in the protection mediated by CD39 overexpression. However, the response to MLDS was less severe than in A2ARKO mice (Fig. 5A), suggesting that signaling via a second adenosine receptor may also be involved. To address this, WT and CD39TG mice were treated with A2BRi, which led to a greater incidence of hyperglycemia (Fig. 5C) and diabetes (Fig. 5D). CD39TG mice treated with A2BRi rapidly became hyperglycemic with kinetics similar to that of WT mice (Fig. 5C), although they did not progress to diabetes, providing evidence for an independent role for the A2BR in early CD39-mediated protection. Finally, to examine the effect of inhibiting both A2AR and A2BR signaling, A2ARKO/CD39TG mice were treated with A2BRi. Blockade of the A2BR did not further augment the incidence of diabetes in these mice (Fig. 5E).

The major challenge for islet transplantation as a clinical therapy is to improve the long-term function of the transplanted islets. Currently, the minority of islet recipients remain insulin independent, reflecting islet loss over time (30). Significant factors contributing to this progressive loss include IBMIR, recurrent autoimmunity, and the alloimmune response. We have previously reported that overexpression of CD39, a key antithrombotic and anti-inflammatory enzyme, protected mouse islets from IBMIR in vitro (11). In this study, we used the MLDS model to investigate the role of purinergic catabolism and signaling in the regulation of islet inflammation and injury in vivo. Although MLDS is not a perfect model of autoimmunity associated with islet transplantation or T1D, it shares a dependence on the action of T cells in its pathogenesis.

The overexpression of CD39 on β-cells does not perturb islet function (11). Fasting insulin levels in CD39TG mice were comparable to WT mice (data not shown), suggesting normal insulin sensitivity. Reduced insulin sensitivity has been reported acutely after treatment of WT mice with NECA, a nonspecific adenosine agonist (31). We have previously shown that basal levels of adenosine in CD39TG mice are comparable to that of WT but are increased after a proinflammatory insult (12). Although adenosine concentrations were not measured in the current study, the protection of CD39 overexpression was mitigated by A2R inhibition or deletion, implicating a role for adenosine signaling. It is possible that insulin sensitivity was altered after MLDS; however, persistent normoglycemia in CD39TG mice indicates an adequate β-cell response.

The susceptibility of mice to MLDS was strikingly influenced by the level of expression of CD39. Mice lacking CD39 became diabetic faster and with a higher overall incidence than WT mice, whereas mice overexpressing CD39 were significantly protected. The rapid onset of diabetes in MLDS-treated CD39KO mice was likely due to defective regulatory T-cell function (26) rather than impaired glucose tolerance, as reconstitution with WT BM reduced the incidence of diabetes to that of WT mice. In the case of CD39TG mice, chimeric mice reconstituted with a WT immune system retained protection from MLDS, suggesting that tissue-generated adenosine was responsible for reduced susceptibility. In vitro data indicated that CD39TG islets were not inherently resistant to proinflammatory cytokines.

T1D is characterized by the expression of proinflammatory cytokines by islet-invading mononuclear cells (32). Unlike high-dose streptozotocin treatment, which is directly toxic to β-cells, MLDS promotes leukocytic infiltration of islets that peaks at 10–12 days (27), comprised principally of CD3+ T cells. After activation, these cells secrete cytokines such as IL-1β, which contribute to β-cell dysfunction and death (33). Further, cytokine-induced expression of iNOS by the β-cell itself is a key factor in islet cell death. The increase in inflammatory cytokine expression in islets from MLDS-treated WT mice likely reflects T-cell infiltration, which was evident histologically and associated with a reduction of insulin staining (Fig. 2). Islets from MLDS-treated CD39TG mice showed minimal leukocytic infiltration, no upregulation of inflammatory cytokines, and preserved insulin content. Despite a relatively high level of apoptosis in the islets (not shown), CD39TG mice maintained near-normal glycemic control, which may reflect enhanced β-cell regenerative capacity due to increased pancreatic NTPDase activity (29,34). Adenosine signaling has recently been implicated in β-cell–specific regeneration. In a zebrafish model, the nonselective agonist NECA did not protect against β-cell death but promoted β-cell regeneration by increasing proliferation through A2AR-dependent mechanisms (29). Further, NECA treatment did not significantly increase the number of β-cells in normal development but reduced BGL by 30% and increased β-cell mass eightfold in mice treated with 150 mg/kg streptozotocin for 2 days (29).

Ectonucleotidase expression has been defined within the mouse, rat, and human pancreas (3537). NTPDase1/CD39 is localized to blood vessels and acinar tissue, NTPDase2 to capillaries and connective tissue surrounding islets and acini, and NTPDase3 exclusively to islet cells. NTPDase8 was not detected. CD73 completes the enzymatic cascade for extracellular adenosine generation (38) and has been detected on rat, but not human or mouse, islets (35). CD73 expression is, however, well documented both on leukocytes (39,40) and the vasculature (41). This is similar to rat liver, where CD73 and CD39, and other NTPDases, are expressed by different but adjacent cells in distinct compartments and differentially regulate adenosine generation and signaling (38). Adenosine is a potent systemic anti-inflammatory molecule and inhibits the proliferation of CD4+ T cells in a dose-dependent fashion through the A2AR (26). The A2AR is expressed on regulatory T cells (42) together with CD39 (26) and CD73 (43). Further, A2AR activation on regulatory T cells has been shown to mitigate renal IRI through increased expression of the membrane protein programmed death-1 (42).

The adenosine receptor repertoire has previously been defined at the transcript level within the whole pancreas of CD-1 mice (19). All four adenosine receptors were expressed at comparable levels at baseline, with increased A1R expression after MLDS treatment (19). We analyzed purified islets, thus eliminating the potentially confounding contribution of exocrine tissue in the whole pancreas. The A3R was not detected and was not investigated further. Although the A1R was expressed on islets, the effect of pharmacological inhibition or activation of A1R on MLDS-induced diabetes was minimal, suggesting a redundant role in this model. This is in contrast to data demonstrating a modest protective effect of A1R activation in the MLDS model (19). However, the effect of the nonspecific agonist NECA in that study was significantly greater than the specific A1R agonist CCPA, suggesting that A1R was not the predominant receptor involved in the protection observed (19).

A2AR transcripts were detected in islets under basal conditions, with little change after MLDS. The A2AR has well-defined effects on T-cell proliferation (26). In keeping with this, deletion of the A2AR on circulating cells conferred heightened susceptibility to MLDS-induced diabetes (Fig. 5). Additionally, tissue-specific deletion of A2AR increased susceptibility (Fig. 5), implicating a previously unrecognized role for A2AR signaling on the tissues in this model of diabetes. In mice overexpressing CD39, the role of the A2AR on circulating cells was redundant as CD39TGA2ARKOBM chimeric mice were fully protected (Fig. 3E). It is possible that the level of adenosine generated is so high that it now signals via different adenosine receptors on T cells or indeed via a different mechanism involving other cell types.

The A2BR is recognized as a low-affinity adenosine receptor with activation occurring only in pathological conditions when the adenosine concentration is substantially increased (44). Expression of A2BR in islets was upregulated after MLDS treatment, and antagonism of A2BR hastened the onset and overall incidence of diabetes (Fig. 5). We speculate that the release of ATP secondary to cellular injury parallels cellular infiltration. CD39 expressed on tissues, together with CD73 expressed on intraislet capillaries or infiltrating cells, sequentially converts ATP to adenosine, which activates A2BR to limit islet infiltration and destruction. The greater capacity of CD39TG mice to generate adenosine, and higher basal expression of the A2BR in these mice, putatively provides protection against early MLDS-induced injury.

The mechanism underpinning the protective effect of CD39TG expression includes signaling through both the A2AR and A2BR. Blockade or deletion of either receptor mitigated the protective effect of CD39 overexpression. Involvement of more than one adenosine receptor parallels the effects of adenosine in renal IRI, where A2AR signaling predominates on circulating CD4+ T cells (45) and macrophages (46), and A2BR signaling within the renal parenchyma is also important (47). It is intriguing that the treatment of A2ARKO/CD39TG mice with an A2BR antagonist did not further exacerbate the diabetic response. Recent speculation surrounds the requirement for cooperation between the A2AR and the A2BR for each receptor to be fully functional (48), which may account for the lack of exaggerated response in the A2ARKO/CD39TG mice with A2BRi.

Islets that survive the transplant process and recurrent autoimmunity then face destruction by the alloimmune response. A2AR agonists (49) and adenosine (50) prevent the early loss of islets posttransplantation, and adenosine, together with low-dose immunosuppression, prevents islet allograft rejection (50). The overexpression of CD39 is protective in this T cell–mediated model of diabetes by signaling through A2AR and A2BR, which combined with its anti-IBMIR (11,13) and putative immunosuppressive effects, makes CD39 overexpression an attractive strategy for prolonging islet survival after intraportal transplantation.

This work is supported by grants from the St. Vincent’s Hospital Research Endowment Fund (to J.S.J.C.) and the Juvenile Diabetes Research Foundation (ITP 4-2006-1025 to K.M.D.).

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

J.S.J.C. performed most of the experiments and wrote the manuscript. J.L.M. assisted in islet isolation and provided intellectual input. H.E.T. and A.J.F.d’A. provided intellectual input and reviewed the manuscript. S.F., L.E., and L.M.-S. performed pancreatic perfusion and removal. P.H. scored the insulitis in the islets. S.C.R. provided intellectual input and reviewed the manuscript. J.-F.C. provided the A2ARKO mice. P.J.C. provided intellectual input, wrote and reviewed the manuscript, and supervised experiments. K.M.D. conceived experimental plans, wrote and reviewed the manuscript, and supervised experiments. K.M.D. is the guarantor of this work and, as such, had full access to all the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.

The authors thank the BioResources Centre (St. Vincent’s Hospital) for all aspects of mouse care.

1.
Yasunami
Y
,
Kojo
S
,
Kitamura
H
, et al
.
Valpha14 NK T cell-triggered IFN-gamma production by Gr-1+CD11b+ cells mediates early graft loss of syngeneic transplanted islets
.
J Exp Med
2005
;
202
:
913
918
[PubMed]
2.
Azzi
J
,
Geara
AS
,
El-Sayegh
S
,
Abdi
R
.
Immunological aspects of pancreatic islet cell transplantation
.
Expert Rev Clin Immunol
2010
;
6
:
111
124
[PubMed]
3.
Petruzzo
P
,
Andreelli
F
,
McGregor
B
, et al
.
Evidence of recurrent type I diabetes following HLA-mismatched pancreas transplantation
.
Diabetes Metab
2000
;
26
:
215
218
[PubMed]
4.
Bosi
E
,
Braghi
S
,
Maffi
P
, et al
.
Autoantibody response to islet transplantation in type 1 diabetes
.
Diabetes
2001
;
50
:
2464
2471
[PubMed]
5.
Vantyghem
MC
,
Fajardy
I
,
Pigny
P
, et al
.
Kinetics of diabetes-associated autoantibodies after sequential intraportal islet allograft associated with kidney transplantation in type 1 diabetes
.
Diabetes Metab
2003
;
29
:
595
601
[PubMed]
6.
Sutherland
DE
,
Goetz
FC
,
Sibley
RK
.
Recurrence of disease in pancreas transplants
.
Diabetes
1989
;
38
(
Suppl 1
):
85
87
[PubMed]
7.
Tydén
G
,
Reinholt
FP
,
Sundkvist
G
,
Bolinder
J
.
Recurrence of autoimmune diabetes mellitus in recipients of cadaveric pancreatic grafts
.
N Engl J Med
1996
;
335
:
860
863
[PubMed]
8.
Okitsu
T
,
Bartlett
ST
,
Hadley
GA
,
Drachenberg
CB
,
Farney
AC
.
Recurrent autoimmunity accelerates destruction of minor and major histoincompatible islet grafts in nonobese diabetic (NOD) mice
.
Am J Transplant
2001
;
1
:
138
145
[PubMed]
9.
Markees
TG
,
Serreze
DV
,
Phillips
NE
, et al
.
NOD mice have a generalized defect in their response to transplantation tolerance induction
.
Diabetes
1999
;
48
:
967
974
[PubMed]
10.
Makhlouf
L
,
Kishimoto
K
,
Smith
RN
, et al
.
The role of autoimmunity in islet allograft destruction: major histocompatibility complex class II matching is necessary for autoimmune destruction of allogeneic islet transplants after T-cell costimulatory blockade
.
Diabetes
2002
;
51
:
3202
3210
[PubMed]
11.
Dwyer
KM
,
Mysore
TB
,
Crikis
S
, et al
.
The transgenic expression of human CD39 on murine islets inhibits clotting of human blood
.
Transplantation
2006
;
82
:
428
432
[PubMed]
12.
Dwyer
KM
,
Robson
SC
,
Nandurkar
HH
, et al
.
Thromboregulatory manifestations in human CD39 transgenic mice and the implications for thrombotic disease and transplantation
.
J Clin Invest
2004
;
113
:
1440
1446
[PubMed]
13.
Inverardi
L
,
Ricordi
C
.
Therapeutic approaches to counteract immediate blood-mediated inflammatory reaction in islet transplantation
.
Transplantation
2006
;
82
:
312
313
[PubMed]
14.
Lin
M
,
Yin
N
,
Murphy
B
, et al
.
Immune cell-derived c3 is required for autoimmune diabetes induced by multiple low doses of streptozotocin
.
Diabetes
2010
;
59
:
2247
2252
[PubMed]
15.
Zimmermann
H
.
Nucleotides and cd39: principal modulatory players in hemostasis and thrombosis
.
Nat Med
1999
;
5
:
987
988
[PubMed]
16.
Haskó
G
,
Linden
J
,
Cronstein
B
,
Pacher
P
.
Adenosine receptors: therapeutic aspects for inflammatory and immune diseases
.
Nat Rev Drug Discov
2008
;
7
:
759
770
[PubMed]
17.
Yegutkin
GG
.
Nucleotide- and nucleoside-converting ectoenzymes: Important modulators of purinergic signalling cascade
.
Biochim Biophys Acta
2008
;
1783
:
673
694
[PubMed]
18.
Crikis
S
,
Lu
B
,
Murray-Segal
LM
, et al
.
Transgenic overexpression of CD39 protects against renal ischemia-reperfusion and transplant vascular injury
.
Am J Transplant
2010
;
10
:
2586
2595
[PubMed]
19.
Németh
ZH
,
Bleich
D
,
Csóka
B
, et al
.
Adenosine receptor activation ameliorates type 1 diabetes
.
FASEB J
2007
;
21
:
2379
2388
[PubMed]
20.
Enjyoji
K
,
Sévigny
J
,
Lin
Y
, et al
.
Targeted disruption of cd39/ATP diphosphohydrolase results in disordered hemostasis and thromboregulation
.
Nat Med
1999
;
5
:
1010
1017
[PubMed]
21.
Chen
JF
,
Huang
Z
,
Ma
J
, et al
.
A(2A) adenosine receptor deficiency attenuates brain injury induced by transient focal ischemia in mice
.
J Neurosci
1999
;
19
:
9192
9200
[PubMed]
22.
Leiter
EH
.
Multiple low-dose streptozotocin-induced hyperglycemia and insulitis in C57BL mice: influence of inbred background, sex, and thymus
.
Proc Natl Acad Sci USA
1982
;
79
:
630
634
[PubMed]
23.
Mottram
PL
,
Han
WR
,
Murray-Segal
LJ
,
Mandel
TE
,
Pietersz
GA
,
McKenzie
IF
.
Idarubicin-anti-CD3: a new immunoconjugate that induces alloantigen-specific tolerance in mice
.
Transplantation
1997
;
64
:
684
690
[PubMed]
24.
Bustin
SA
,
Benes
V
,
Garson
JA
, et al
.
The MIQE guidelines: minimum information for publication of quantitative real-time PCR experiments
.
Clin Chem
2009
;
55
:
611
622
[PubMed]
25.
Enjyoji
K
,
Kotani
K
,
Thukral
C
, et al
.
Deletion of cd39/entpd1 results in hepatic insulin resistance
.
Diabetes
2008
;
57
:
2311
2320
[PubMed]
26.
Deaglio
S
,
Dwyer
KM
,
Gao
W
, et al
.
Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression
.
J Exp Med
2007
;
204
:
1257
1265
[PubMed]
27.
McEvoy
RC
,
Andersson
J
,
Sandler
S
,
Hellerström
C
.
Multiple low-dose streptozotocin-induced diabetes in the mouse. Evidence for stimulation of a cytotoxic cellular immune response against an insulin-producing beta cell line
.
J Clin Invest
1984
;
74
:
715
722
[PubMed]
28.
Pommey, S, Lu, B, McRae, J, et al. 2012. Liver grafts from CD39-overexpressing mice are protected from ischemia reperfusion injury due to reduced numbers of resident CD4(+) T cells. Hepatology. 24 July 2012 [Epub ahead of print]
29.
Andersson
O
,
Adams
BA
,
Yoo
D
, et al
.
Adenosine signaling promotes regeneration of pancreatic β cells in vivo
.
Cell Metab
2012
;
15
:
885
894
[PubMed]
30.
Ryan
EA
,
Paty
BW
,
Senior
PA
, et al
.
Five-year follow-up after clinical islet transplantation
.
Diabetes
2005
;
54
:
2060
2069
[PubMed]
31.
Figler
RA
,
Wang
G
,
Srinivasan
S
, et al
.
Links between insulin resistance, adenosine A2B receptors, and inflammatory markers in mice and humans
.
Diabetes
2011
;
60
:
669
679
[PubMed]
32.
Cnop
M
,
Welsh
N
,
Jonas
JC
,
Jörns
A
,
Lenzen
S
,
Eizirik
DL
.
Mechanisms of pancreatic beta-cell death in type 1 and type 2 diabetes: many differences, few similarities
.
Diabetes
2005
;
54
(
Suppl. 2
):
S97
S107
[PubMed]
33.
Kutlu
B
,
Naamane
N
,
Berthou
L
,
Eizirik
DL
.
New approaches for in silico identification of cytokine-modified beta cell gene networks
.
Ann N Y Acad Sci
2004
;
1037
:
41
58
[PubMed]
34.
Annes
JP
,
Ryu
JH
,
Lam
K
, et al
.
Adenosine kinase inhibition selectively promotes rodent and porcine islet β-cell replication
.
Proc Natl Acad Sci USA
2012
;
109
:
3915
3920
[PubMed]
35.
Lavoie
EG
,
Fausther
M
,
Kauffenstein
G
, et al
.
Identification of the ectonucleotidases expressed in mouse, rat, and human Langerhans islets: potential role of NTPDase3 in insulin secretion
.
Am J Physiol Endocrinol Metab
2010
;
299
:
E647
E656
[PubMed]
36.
Künzli
BM
,
Berberat
PO
,
Giese
T
, et al
.
Upregulation of CD39/NTPDases and P2 receptors in human pancreatic disease
.
Am J Physiol Gastrointest Liver Physiol
2007
;
292
:
G223
G230
[PubMed]
37.
Kukulski
F
,
Lévesque
SA
,
Sévigny
J
.
Impact of ectoenzymes on p2 and p1 receptor signaling
.
Adv Pharmacol
2011
;
61
:
263
299
[PubMed]
38.
Fausther
M
,
Lecka
J
,
Soliman
E
, et al
.
Coexpression of ecto-5′-nucleotidase/CD73 with specific NTPDases differentially regulates adenosine formation in the rat liver
.
Am J Physiol Gastrointest Liver Physiol
2012
;
302
:
G447
G459
[PubMed]
39.
Salmi
M
,
Jalkanen
S
.
Cell-surface enzymes in control of leukocyte trafficking
.
Nat Rev Immunol
2005
;
5
:
760
771
[PubMed]
40.
Ålgars
A
,
Karikoski
M
,
Yegutkin
GG
, et al
.
Different role of CD73 in leukocyte trafficking via blood and lymph vessels
.
Blood
2011
;
117
:
4387
4393
[PubMed]
41.
Colgan
SP
,
Eltzschig
HK
,
Eckle
T
,
Thompson
LF
.
Physiological roles for ecto-5′-nucleotidase (CD73)
.
Purinergic Signal
2006
;
2
:
351
360
[PubMed]
42.
Kinsey
GR
,
Huang
L
,
Jaworska
K
, et al
.
Autocrine adenosine signaling promotes regulatory T cell-mediated renal protection
.
J Am Soc Nephrol
2012
;
23
:
1528
1537
[PubMed]
43.
Kobie
JJ
,
Shah
PR
,
Yang
L
,
Rebhahn
JA
,
Fowell
DJ
,
Mosmann
TR
.
T regulatory and primed uncommitted CD4 T cells express CD73, which suppresses effector CD4 T cells by converting 5′-adenosine monophosphate to adenosine
.
J Immunol
2006
;
177
:
6780
6786
[PubMed]
44.
Fredholm
BB
,
Abbracchio
MP
,
Burnstock
G
, et al
.
Nomenclature and classification of purinoceptors
.
Pharmacol Rev
1994
;
46
:
143
156
[PubMed]
45.
Day
YJ
,
Huang
L
,
Ye
H
,
Li
L
,
Linden
J
,
Okusa
MD
.
Renal ischemia-reperfusion injury and adenosine 2A receptor-mediated tissue protection: the role of CD4+ T cells and IFN-gamma
.
J Immunol
2006
;
176
:
3108
3114
[PubMed]
46.
Day
YJ
,
Huang
L
,
Ye
H
,
Linden
J
,
Okusa
MD
.
Renal ischemia-reperfusion injury and adenosine 2A receptor-mediated tissue protection: role of macrophages
.
Am J Physiol Renal Physiol
2005
;
288
:
F722
F731
[PubMed]
47.
Grenz
A
,
Zhang
H
,
Hermes
M
, et al
.
Contribution of E-NTPDase1 (CD39) to renal protection from ischemia-reperfusion injury
.
FASEB J
2007
;
21
:
2863
2873
[PubMed]
48.
Moriyama
K
,
Sitkovsky
MV
.
Adenosine A2A receptor is involved in cell surface expression of A2B receptor
.
J Biol Chem
2010
;
285
:
39271
39288
[PubMed]
49.
Chhabra
P
,
Wang
K
,
Zeng
Q
, et al
.
Adenosine A(2A) agonist administration improves islet transplant outcome: evidence for the role of innate immunity in islet graft rejection
.
Cell Transplant
2010
;
19
:
597
612
[PubMed]
50.
Nitta
T
,
Itoh
T
,
Matsuoka
N
, et al
.
Prevention of early loss of transplanted islets in the liver of mice by adenosine
.
Transplantation
2009
;
88
:
49
56
[PubMed]
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. See http://creativecommons.org/licenses/by-nc-nd/3.0/ for details.

Supplementary data