Cytokines, particularly interleukin (IL)-1β, have been postulated to cause β-cell destruction in type 1 diabetes. We tested the efficacy of an IL-1 cytokine trap in counteraction of suppressive and toxic effects after exposure of rat pancreatic islets in vitro to IL-1β. The IL-1 cytokine trap used herein comprised extracellular domains of the IL-1 receptor accessory protein and the human IL-1 receptor 1 arranged inline and fused to the Fc portion of human IgG1. Groups of isolated rat pancreatic islets were maintained in medium culture with or without IL-1β (150 pmol/l) for 48 h in the absence or presence of the IL-1 trap at 1-, 10-, or 100-fold excess the molar concentration of the cytokine. IL-1β alone induced a strong inhibition of insulin secretion and glucose oxidation rate and a marked increase in medium nitrite accumulation as an indicator of nitric oxide generation. When the IL-1 trap was used at a ratio 10:1 or 100:1, a complete protection against these effects were observed. Moreover, the IL-1 trap (100:1) blocked the increased islet cell death seen in islets treated with a combination of IL-1β + tumor necrosis factor-α + interferon-γ as well as functional suppression induced by the cytokine combination. In conclusion, we show that addition of an IL-1 trap can protect rat pancreatic islets in vitro against noxious effects induced by IL-1β. Exploring the IL-1 trap in relevant animal models of type 1 diabetes represents an interesting future intervention strategy.

Despite extensive research efforts, the pathogenesis of type 1 diabetes is not fully understood. The prevailing view is that an immune reaction selectively destroys the insulin-producing β-cells in the pancreas (13). It has been proposed that viruses, specific toxins, autoreactive antibodies, macrophages, T-cells, cytokines, nitric oxide (NO), and free oxygen radicals, alone or in combination, mediate the destruction of β-cells. Among cytokines, the proinflammatory cytokines interleukin (IL)-1β, tumor necrosis factor (TNF)-α, and interferon (IFN)-γ are of particular interest (46). This notion is based on studies showing that exposure in vitro to these cytokines causes impaired β-cell function and eventually lethal cell damage of rodent and human pancreatic islet β-cells (79). Furthermore, several animal studies have shown that interference with the action or production of these cytokines can affect the progression of type 1 diabetes in animal models (1014).

There are several mechanisms described by which the activity of IL-1 may be reduced, e.g., the IL-1 receptor antagonist (IL-1Ra), anti–IL-1β monoclonal antibodies, IL-1 receptor type 1 antibodies, IL-1 receptor accessory protein (IL-1RAcP) antibodies, soluble forms of IL-1RAcP, inhibitors of IL-1β converting enzyme, i.e., caspase-1, and soluble IL-1 receptors (1517). Some of these principles have also been attempted in alleviating the symptoms of inflammatory and/or immune-mediated diseases in humans in which cytokines, especially IL-1β, have been implicated to possess a key role.

Recently, a new type of high-affinity blocker of cytokine action has been developed—so-called cytokine traps (18). Cytokine traps are molecules consisting of fusions between the Fc region of IgG1 and extracellular domains of cytokine receptor components involved in cytokine binding as schematically shown in (18). Besides effectively blocking cytokine action in vitro in various assays, such molecules could also counteract evolution in vivo of immune reactions in murine models of arthritis and asthma.

Successful therapies, without essential side effects, in preventing type 1 diabetes in humans are lacking. In the present investigation, we aimed to study the efficacy of an IL-1 cytokine trap to counteract suppressive and cytotoxic effects after exposure of isolated adult rat pancreatic islets in vitro to IL-1β, which could form a basis for testing the cytokine trap in relevant animal models of type 1 diabetes. The IL-1 cytokine trap used herein comprised extracellular domains of the IL-1 receptor accessory protein (IL-1RAcP) and the human IL-1 receptor 1 arranged inline and fused to the Fc portion of human IgG1 (18). Our findings show that the cytokine trap could completely block the IL-1β actions studied.

Animals, islet isolation, tissue culture, and cytokine exposure.

Pancreatic islets from male Sprague-Dawley rats, bred in a local colony (BK Universal, Sollentuna, Sweden), were isolated by a collagenase digestion procedure (19). Groups of 150–200 islets were kept free-floating in culture medium RPMI-1640 (Sigma, St. Louis, MO) supplemented with 10% (vol/vol) FCS (Sigma), benzylpenicillin (100 units/ml), and streptomycin (0.1 mg/ml) for 5–7 days before use (19).

Islets in groups of ∼50 were subsequently transferred to new nonattachment culture dishes containing 2.25 ml RPMI-1640 + 0.25 ml FCS. Human IL-1 trap (18) at different concentrations was added, and after 30 min, human IL-1β (2.5 ng/ml corresponding to 25 units/ml and 150 pmol/l; PeproTech, London, U.K.) or a cytokine combination was added (25 units/ml IL-1β + 1,000 units/ml tumor necrosis factor [TNF]-α [human, PeproTech] + 1,000 units/ml interferon [IFN]-γ [rat; R&D Systems, Abingdon, U.K.]). The islets were subsequently maintained in culture for 48 h before tested. The human IL-1 trap was provided by Dr. Margaret Karow and colleagues (Regeneron Pharmaceuticals, Tarrytown, NY). The molecular weight of IL-1β is 17.0 kDa, and the molecular weight of the cytokine trap is 202 kDa. The study comprised the experimental groups as follows: control = no trap and no cytokine; TL = 150 pmol/l IL-1 trap (1× IL-1β concentration); TM = 1,500 pmol/l IL-1 trap (10 × IL-1β concentration); TH = 15,000 pmol/l IL-1 trap (100 × IL-1β concentration); TC = 150,000 pmol/l Fc protein (1,000 × IL-1β concentration); IL = IL-1β (150 pmol/l); ILTL = IL-1β (150 pmol/l) + IL-1 trap (150 pmol/l, i.e., 1 × IL-1β concentration); ILTM = IL-1β (150 pmol/l) + IL-1 trap (1,500 pmol/l, i.e., 10 × IL-1β concentration); ILTH = IL-1β (150 pmol/l) + IL-1 trap (15,000 pmol/l i.e., 100 × IL-1β concentration); ILTC = IL-1β (150 pmol/l) + Fc protein (150,000 pmol/l, i.e., 1,000 × IL-1β concentration). Cytokine mixture = IL-1β (150 pmol/l) + TNF-α (1,000 units/ml) + IFN-γ (1,000 units/ml). Cytokine mixture + TH = [IL-1β (150 pmol/l) + TNF-α (1,000 units/ml) + IFN-γ (1,000 units/ml)] + IL-1 trap (15,000 pmol/l, i.e., 100 × IL-1β concentration). Cyt. mix. + TC = [IL-1β (150 pmol/l) + TNF-α (1,000 units/ml) + IFN-γ (1,000 units/ml)] + Fc protein (150,000 pmol/l, i.e., 1,000 × IL-1β).

Medium insulin accumulation, glucose-stimulated insulin release, and glucose oxidation rate.

After the 48-h culture, a sample of the culture medium was collected. Then duplicate groups of 10 islets each were incubated during 1 h at 1.7 mmol/l glucose and during a 2nd hour at 16.7 mmol/l glucose (19). The insulin concentrations of the media were measured using a rat insulin enzyme-linked immunosorbent assay according to the manufacturer’s instructions (Mercodia, Uppsala, Sweden). In triplicate groups of 10 islets, the islet glucose oxidation rate was subsequently measured as previously described (20).

Nitrite determination.

To aliquots of the culture medium (250 μl), 100 μl of a reagent solution containing 10 ml 100% acetic acid, 300 μl 10% sulfanilamide (dissolved in acetone), and 30 μl naphtylendiamine (dissolved in redistilled H2O) was added. The reaction was carried out at 20°C for 15 min, and the absorbance was measured at 540 nm in a Labsystems iEMS spectrophotometer against a nitrite standard curve.

Assessment of cell viability by flow cytometry.

Islets in groups of 50 were cultured without or with the cytokine mixture and IL-1 trap or Fc control protein for 48 h. Then the islets were incubated with 10 μg/ml propidium iodide for 15 min and subsequently dissociated with 0.05% trypsin/Hanks’ balanced salt solution. Changes in light scattering and fluorescence emission were determined by flow cytometry with a Becton Dickinson FACSCalibur equipped with CellQuest software (Becton Dickinson, San Jose, CA). A total of 5,000–10,000 cells per sample were analyzed by exciting the cells at 488 nm and examining fluorescence at 650 nm. Propidium iodide stains both apoptotic and necrotic cells, the latter with high intensity. As a cell shrinks or loses volume, forward-scattered light decreases. Untreated cells were gated on a forward scatter versus propidium iodide fluorescence dot plot, and the same gates were then applied to cytokine-treated cells for calculation of the fractions of apoptotic and/or necrotic cells (referred to as dead islet cells).

Statistical analysis.

From the glucose-stimulated insulin release and glucose oxidation rate experiments, a mean was calculated from each duplicate or triplicate group of islets and then considered as one separate observation. Furthermore, every observation represented different islet donors. Data are expressed as means ± SE, and groups of data were compared, using one-way repeated-measures ANOVA or Student’s t test. Statistical analysis was performed using SigmaStat (SPSS, Chicago, IL).

Addition of 150 pmol/l IL-1β to rat pancreatic islets for 48 h caused a pronounced reduction in the accumulation of insulin in the culture medium compared with control islets not exposed to the cytokine (Fig. 1; IL). This was not affected by the lowest concentration of the cytokine trap (ILTL), while it was fully prevented when the cytokine trap was added at a 10-fold (ILTM) and 100-fold (ILTH) molar excess of the cytokine trap in relation to the IL-1β concentration. As a protein control we added an Fc control protein at a concentration 1,000-fold higher than the cytokine trap, but this failed to counteract the IL-1β action (ILTC). In some initial experiments, we also tested the cytokine trap at a 1,000-fold excess, and thus a full protection against IL-1β was observed. Because we then also observed a similar protection already with 10- and 100-fold excess of cytokine trap, the 1,000-fold concentration of the trap was abandoned in the subsequent experiments; however, the TC group was nevertheless maintained at a 1,000-fold concentration. Addition of the cytokine trap (TL, TM, and TH) and the Fc control protein (TC) alone did not affect the medium insulin accumulation.

After culture for 48 h, the islets were tested in short-term experiments. When the insulin secretory rate was studied at low glucose (1.7 mmol/l), there were no significant differences among the groups, although there was a trend toward a decline in the IL and ILTC groups (Fig. 2A). On the other hand, at high glucose (16.7 mmol/l), glucose-stimulated insulin release was markedly depressed after adding IL-1β (Fig. 2B). The cytokine trap prevented this decline at all three tested concentrations, although the protection in the ILTL group was not complete. Treatment of islets in culture with the cytokine trap or the Fc control protein only did not affect the insulin release at low or high glucose.

As a test on islet glucose metabolism, the glucose oxidation rate after exposure to IL-1β and the cytokine trap was assessed (Fig. 3). The cytokine caused ∼50% reduction in the glucose oxidation rate, and this was also seen in the groups ILTL and ILTC. Again, the inhibitory action of IL-1β was counteracted by the higher concentrations of the cytokine trap.

Next, nitrite accumulation in the culture medium was determined as an indicator if NO formed (Fig. 4). IL-1β induced a strong increase in medium nitrite, and this was also the case in the groups ILTL and ILTC. The elevated nitrite level was prevented by higher concentrations of the cytokine trap. In the control groups, there was no change in the basal nitrite accumulation.

We then aimed to investigate if the IL-1 trap could also affect cytokine-induced islet cell death by fluorescence-activated cell sorter analysis after propidium iodide staining. To elicit a marked islet cell death, we used a cytokine combination (IL-1β + TNF-α + IFN-γ), since IL-1β treatment alone for 48 h does not cause much islet cytotoxicity. A representative experiment is shown in Fig. 5. Non–cytokine-exposed control cells (black area) indicates essentially viable cells (Fig. 5A), whereas cells exposed to the cytokine combination show a distribution with an increased amount of dead cells (white area). In Fig. 5B, the islets have been exposed to the cytokine mixture + IL-1 trap (100-fold × IL-1β concentration), and these cells showed a distribution nearly identical as that seen for control cells, whereas in Fig. 5C, it is shown that the Fc control failed to prevent cytokine-induced cell death. The data from three independent experiments are summarized in Fig. 6, showing the protective effect of the IL-1 trap against the cytotoxicity induced by the cytokine combination.

Finally we tested if the IL-1 trap also could affect functional effects induced by the cytokine mixture under the same experimental conditions as above (Table 1). The cytokine mixture induced a marked nitrite accumulation, and this was counteracted, although not completely, by the IL-1 trap. The glucose oxidation rate and medium insulin accumulation was strongly suppressed by the cytokine mixture, and this was fully prevented by the IL-1 trap. The insulin release at low glucose was elevated after exposure to the cytokine mixture and inhibited at high glucose. The IL-1 trap protected against these effects, although there was a trend that the insulin secretion at high glucose was not fully restored (P = 0.15). The TC control protein could not influence the functional effects induced by the cytokine mixture.

Proinflammatory cytokines and in particular IL-1β have been postulated to be involved in pancreatic β-cell destruction leading to type 1 diabetes (49). A large body of experimental data have shown that prolonged in vitro exposure of rodent pancreatic β-cells to elevated concentrations of IL-1β can cause both inhibition of β-cell function and cell death. Regarding isolated human islet cells, a combination of cytokines, e.g., IL-1β + TNF-α + IFN-γ, is required to obtain a similar effect. In the present study, we explored in vitro the efficacy of a multicomponent IL-1 trap in influencing IL-1β–induced suppression of rat pancreatic islet function as well as islet cytotoxicity induced by a cytokine combination. We found that addition of the IL-1 trap exerted a complete protection against IL-1β–induced inhibition of islet metabolism (glucose oxidation rate) and glucose-stimulated insulin secretion. This was most likely achieved by a prevention of IL-1β–mediated increase in NO generation, since elevated NO concentrations can be most harmful to rat islet function (2123). However, in studies using NO synthase inhibitors and islets derived from inducible NO synthase–deficient mice, it has been shown that IL-1β also exerts suppressive actions that are NO independent (2427). In the present study, it seems as if also the latter NO-independent suppressive actions by IL-1β were counteracted by the IL-1 trap.

Previously, we examined the IL-1Ra in a study of rat pancreatic islets with a design similar to the present investigation (28). After 48 h of coincubation, a 1,000-fold molar excess of IL-1Ra was required to completely block the effects of IL-1 on β-cell function. Based on measurements of islets DNA contents, IL-1Ra could also prevent IL-1β–induced cell loss. In subsequent studies, we further demonstrated that continuous infusion of IL-1Ra using osmotic minipumps could protect against hyperglycemia in two murine type 1 diabetes models, namely multiple low-dose streptozotocin diabetes (29) and recurrence of disease in NOD mice (10). We also attempted a gene delivery system for IL-1Ra in the recurrence of disease model, but the protective effect was only marginal, probably due to the relatively transient expression of IL-1Ra (29). The IL-1 trap not only has a superior ability to inhibit IL-1β in ex vivo assays, in humans, it also has superior pharmacokinetics, with an in vivo half-life of 5–9 days (30), when compared with IL-1Ra, which has a reported in vivo half-life of 5–6 h (31). Thus, the IL-1 trap may require less frequent dosing and/or exhibit improved in vivo efficacy than IL-1Ra.

In line with the findings using IL-1Ra (32), we observed a protective effect of the IL-1 trap when it was added at a 10-fold or higher concentration compared with IL-1β. It might be expected that a 1:1 ratio would show more of a protective action, compared with IL-1β addition alone, than it actually does. However, it is possible that a small fraction of free IL-1β in the in vitro system still exists at the 1:1 ratio, which can cause a suppressive islet response. Indeed, we have seen that a 10- to 20-fold lower concentration of IL-1β than used herein, i.e., 7.5–15 pmol/l, lead to an inhibition of the glucose oxidation rate and an elevated medium nitrite accumulation by rat pancreatic islets (33); thus, even if 95% of the IL-1β was inhibited, there would still be a signal from the remaining free 5%. Consistent with this, previous results using an MRC5 bioassay, in which the IL-1 trap has an IC50 of 2 pmol/l and IL-1Ra of 73 pmol/l, indicate that when the IL-1 trap is present at 3 pmol/l, a 1:0.75 ratio with the 4 pmol/l IL-1β in the assay, ∼25% of bioactivity remained, whereas at a 1:4 ratio, 95% of bioactivity was inhibited (15). This is in contrast to IL-1Ra, which in this bioassay required a 1,000-fold excess over IL-1β to block 95% of the bioactivity. As a control protein for the IL-1 trap in the islet experiments, we used the Fc protein of human IgG1 (experimental group TC) at 1,000-fold molar excess compared with IL-1β. This protein failed to counteract any of the IL-1β–induced effects examined, suggesting that the protective actions on islet function and viability by the IL-1 trap is due to it specific inhibition of IL-1β and not due to the addition of a nonspecific protein or via cytokine binding to the Fc part of the IL-1 trap.

The IL-1 trap could also block islet cytotoxicity in response to a combination of IL-1β + TNF-α + IFN-γ. It has been speculated that the signals and/or intracellular events leading to suppressed islet cell function can be separated from the mechanisms leading to cytokine-mediated cell killing (34,35). If so, however, addition of the IL-1 trap prevented both functional suppression and cell death. This is in agreement with the view that the IL-1 trap would block the binding of IL-1β to the islet cells. Moreover, it suggests that blockage of IL-1β alone can be sufficient to counteract cell killing induced by a cytokine combination. It is unclear by which mechanism(s) the IL-1 trap reduced the cell death induced by the cytokine combination. A possibility is that a reduced NO generation has contributed to this effect. The fact that the IL-1 trap tended not to fully restore a reduction in glucose-stimulated insulin release in response to the cytokine mixture may be because some elevation of NO production has taken place under this condition. Indeed, it has been reported that the combination of TNF-α and IFN-γ can induce nitrite production and a decrease in insulin secretion from rat islets (26).

In conclusion, the present investigation shows that addition of an IL-1 trap could completely prevent rat pancreatic islets in vitro against noxious effects induced by IL-1β. Because the cytokine has been suggested to have a key role in β-cell degeneration in type 1 diabetes, testing the IL-1 trap in relevant animal models represents an interesting future therapeutic alternative.

FIG. 1.

Medium insulin accumulation after culture of rat pancreatic islets for 48 h with or without IL-1β (150 pmol/l) in the absence or presence of an IL-1 trap. The designated experimental groups are as follows: control = no trap and no cytokine; TL = IL-1 trap (1 × IL-1 β concentration); TM = IL-1 trap (10 × IL-1 β concentration); TH = IL-1 trap (100 × IL-1 β concentration); TC = Fc protein (1,000 × IL-1 β concentration); IL = IL-1β; ILTL = IL-1 β + IL-1 trap (1 × IL-1 β concentration); ILTM = IL-1 β + IL-1 trap (10 × IL-1 β concentration); ILTH = IL-1 β + IL-1 trap (100 × IL-1 β concentration); and ILTC = IL-1 β + Fc protein (1,000 × IL-1 β concentration). Data are means ± SE for six to eight observations. *P < 0.05 or less vs. control, using one-way repeated-measures ANOVA, with subsequent all pairwise comparison procedures by the Bonferroni t test. #P ≤ 0.05 vs. IL.

FIG. 1.

Medium insulin accumulation after culture of rat pancreatic islets for 48 h with or without IL-1β (150 pmol/l) in the absence or presence of an IL-1 trap. The designated experimental groups are as follows: control = no trap and no cytokine; TL = IL-1 trap (1 × IL-1 β concentration); TM = IL-1 trap (10 × IL-1 β concentration); TH = IL-1 trap (100 × IL-1 β concentration); TC = Fc protein (1,000 × IL-1 β concentration); IL = IL-1β; ILTL = IL-1 β + IL-1 trap (1 × IL-1 β concentration); ILTM = IL-1 β + IL-1 trap (10 × IL-1 β concentration); ILTH = IL-1 β + IL-1 trap (100 × IL-1 β concentration); and ILTC = IL-1 β + Fc protein (1,000 × IL-1 β concentration). Data are means ± SE for six to eight observations. *P < 0.05 or less vs. control, using one-way repeated-measures ANOVA, with subsequent all pairwise comparison procedures by the Bonferroni t test. #P ≤ 0.05 vs. IL.

Close modal
FIG. 2.

Short-term insulin secretion at 1.7 mmol/l (A) and 16.7 mmol/l (B) of rat pancreatic islets treated as described in Fig. 1. Data are means ± SE for four to five observations. *P < 0.05 or less vs. control; #P < 0.05 or less vs. IL, using one-way repeated-measures ANOVA, with subsequent all pairwise comparison procedures by the Bonferroni t test. See Fig. 1 legend for explanation of abbreviations.

FIG. 2.

Short-term insulin secretion at 1.7 mmol/l (A) and 16.7 mmol/l (B) of rat pancreatic islets treated as described in Fig. 1. Data are means ± SE for four to five observations. *P < 0.05 or less vs. control; #P < 0.05 or less vs. IL, using one-way repeated-measures ANOVA, with subsequent all pairwise comparison procedures by the Bonferroni t test. See Fig. 1 legend for explanation of abbreviations.

Close modal
FIG. 3.

Islet glucose oxidation rate at 16.7 mmol/l of rat pancreatic islets treated as described in Fig. 1. Data are means ± SE for five observations. *P < 0.05 or less vs. control, using one-way repeated-measures ANOVA, with subsequent all pairwise comparison procedures by the Bonferroni t test. See Fig. 1 legend for explanation of abbreviations.

FIG. 3.

Islet glucose oxidation rate at 16.7 mmol/l of rat pancreatic islets treated as described in Fig. 1. Data are means ± SE for five observations. *P < 0.05 or less vs. control, using one-way repeated-measures ANOVA, with subsequent all pairwise comparison procedures by the Bonferroni t test. See Fig. 1 legend for explanation of abbreviations.

Close modal
FIG. 4.

Medium nitrite accumulation from rat pancreatic islets treated as described in Fig. 1. Data are means ± SE for six observations. *P < 0.05 or less vs. control; #P < 0.05 or less vs. IL, using one-way repeated-measures ANOVA, with subsequent all pairwise comparison procedures by the Bonferroni t test. See Fig. 1 legend for explanation of abbreviations.

FIG. 4.

Medium nitrite accumulation from rat pancreatic islets treated as described in Fig. 1. Data are means ± SE for six observations. *P < 0.05 or less vs. control; #P < 0.05 or less vs. IL, using one-way repeated-measures ANOVA, with subsequent all pairwise comparison procedures by the Bonferroni t test. See Fig. 1 legend for explanation of abbreviations.

Close modal
FIG. 5.

Representative FACS cell distributions after propidium iodide staining following islet exposure for 48 h to no cytokines (control) or a cytokine mixture composed of IL-1β (150 pmol/l) + TNF-α (1,000 units/ml) + IFN-γ (1,000 units/ml). A: Control cells (black area) and cells exposed to the cytokine mixture (white area). B: Control cells (black area) and cells exposed to the cytokine mixture + IL-1 trap (100 × IL-1β concentration = TH) (white area). C: Control cells (black area) and cells exposed to the cytokine mixture + Fc protein (1,000 × IL-1β concentration = TC) (white area). FL3-H denotes the intensity of fluorescence.

FIG. 5.

Representative FACS cell distributions after propidium iodide staining following islet exposure for 48 h to no cytokines (control) or a cytokine mixture composed of IL-1β (150 pmol/l) + TNF-α (1,000 units/ml) + IFN-γ (1,000 units/ml). A: Control cells (black area) and cells exposed to the cytokine mixture (white area). B: Control cells (black area) and cells exposed to the cytokine mixture + IL-1 trap (100 × IL-1β concentration = TH) (white area). C: Control cells (black area) and cells exposed to the cytokine mixture + Fc protein (1,000 × IL-1β concentration = TC) (white area). FL3-H denotes the intensity of fluorescence.

Close modal
FIG. 6.

Fraction of dead islet cells after treatments as described in Fig. 5. Data are means ± SE for three experiments per group. *P < 0.05 or less vs. control, using the Student’s t test.

FIG. 6.

Fraction of dead islet cells after treatments as described in Fig. 5. Data are means ± SE for three experiments per group. *P < 0.05 or less vs. control, using the Student’s t test.

Close modal
TABLE 1

Effects of a 48-h exposure to a cytokine mixture and the IL-1 trap on islet functions

ControlCytokine mixtureCytokine mixture + THCytokine mixture + TC
Functions     
    Medium insulin accumulation (ng/10 islets × h) 12 ± 2.5 4.4 ± 0.29* 19 ± 2.0 3.7 ± 0.73* 
    >Insulin release (ng/10 islets × h)     
         1.7 mmol/l 4.5 ± 0.46 9.3 ± 0.84* 5.3 ± 0.58 10 ± 1.1* 
        16.7 mmol/l 55 ± 3.5 13 ± 2.9* 37 ± 4.1 17 ± 6.0* 
Glucose oxidation rate (pmol/10 islets × 90 min) 319 ± 32 47.5 ± 17* 325 ± 29 41.3 ± 4.6* 
Medium nitrite accumulation (pmol/10 islets × h) 0.11 ± 0.11 3.2 ± 0.11* 1.2 ± 0.053* 3.1 ± 0.18* 
ControlCytokine mixtureCytokine mixture + THCytokine mixture + TC
Functions     
    Medium insulin accumulation (ng/10 islets × h) 12 ± 2.5 4.4 ± 0.29* 19 ± 2.0 3.7 ± 0.73* 
    >Insulin release (ng/10 islets × h)     
         1.7 mmol/l 4.5 ± 0.46 9.3 ± 0.84* 5.3 ± 0.58 10 ± 1.1* 
        16.7 mmol/l 55 ± 3.5 13 ± 2.9* 37 ± 4.1 17 ± 6.0* 
Glucose oxidation rate (pmol/10 islets × 90 min) 319 ± 32 47.5 ± 17* 325 ± 29 41.3 ± 4.6* 
Medium nitrite accumulation (pmol/10 islets × h) 0.11 ± 0.11 3.2 ± 0.11* 1.2 ± 0.053* 3.1 ± 0.18* 

Data are means ± SE for four observations. Cytokine mixture denotes IL-1β (150 pmol/l) + TNF-α (1,000 units/ml) + IFN-γ (1,000 units/ml); TH denotes IL-1 trap (100 × IL-1β concentration); and TC denotes Fc protein (1,000 × IL-1β concentration).

*

P < 0.05 vs. control;

P < 0.05 vs. Cytokine mixture, using one-way repeated-measures ANOVA, with subsequent all pairwise comparison procedures by the Bonferroni t test.

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

This study was supported by grants from the Swedish Research Council (72X-8273), the Novo Nordic Fund, the Swedish Diabetes Association, the European Foundation for the Study of Diabetes, and the Family Ernfors Fund.

We appreciate Dr. Margaret Karow and colleagues (Regeneron Pharmaceuticals) for providing the IL-1 trap and for valuable discussions and comments on the manuscript. We thank Astrid Nordin and Ing-Britt Hallgren for excellent technical assistance.

1
Jasinski JM, Eisenbarth GS: Hypothesis for the pathogenesis of type 1A diabetes.
Drugs Today (Barc)
41
:
141
–149,
2005
2
Gianani R, Eisenbarth GS: The stages of type 1A diabetes: 2005.
Immunol Rev
204
:
232
–249,
2005
3
Atkinson MA: ADA Outstanding Scientific Achievement Lecture 2004: Thirty years of investigating the autoimmune basis for type 1 diabetes: why can’t we prevent or reverse this disease?
Diabetes
54
:
1253
–1263,
2005
4
Nerup J, Mandrupoulsen T, Helqvist S, Andersen HU, Pociot F, Reimers JI, Cuartero BG, Karlsen AE, Bierre U, Lorenzen T: On the pathogenesis of IDDM.
Diabetologia
37
:
S82
–S89,
1994
5
Mandrup-Poulsen T: The role of interleukin-1 in the pathogenesis of IDDM.
Diabetologia
39
:
1005
–1029,
1996
6
Rabinovitch A, Suarez-Pinzon WL: Role of cytokines in the pathogenesis of autoimmune diabetes mellitus.
Rev Endocr Metab Disord
4
:
291
–299,
2003
7
Sandler S, Eizirik DL, Svensson C, Strandell E, Welsh M, Welsh N: Biochemical and molecular actions of interleukin-1 on pancreatic β-cells.
Autoimmunity
10
:
241
–253,
1991
8
Eizirik DL, Mandrup-Poulsen T: A choice of death: the signal-transduction of immune-mediated β-cell apoptosis.
Diabetologia
44
:
2115
–2133,
2001
(erratum in Diabetologia 45:936, 2002)
9
Eizirik DL, Darville MI: β-Cell apoptosis and defense mechanisms: lessons from type 1 diabetes.
Diabetes
50 (Suppl. 1)
:
S64
–S69,
2001
10
Sandberg JO, Eizirik DL, Sandler S: IL-1 receptor antagonist inhibits recurrence of disease after syngeneic pancreatic islet transplantation to spontaneously diabetic non-obese diabetic (NOD) mice.
Clin Exp Immunol
108
:
314
–317,
1997
11
Flodstrom M, Tyrberg B, Eizirik DL, Sandler S: Reduced sensitivity of inducible nitric oxide synthase-deficient mice to multiple low-dose streptozotocin-induced diabetes.
Diabetes
48
:
706
–713,
1999
12
Holstad M, Sandler S: A transcriptional inhibitor of TNF-α prevents diabetes induced by multiple low-dose streptozotocin injections in mice.
J Autoimmun
16
:
441
–447,
2001
13
Flodström-Tullberg M, Yadav D, Hägerkvist R, Tsai D, Secrest P, Stotland A, Sarvetnick N: Target cell expression of suppressor of cytokine signaling-1 prevents diabetes in the NOD mouse.
Diabetes
52
:
2696
–2700,
2003
14
Yang ZD, Chen M, Ellett JD, Fialkow LB, Carter JD, Nadler JL: The novel anti-inflammatory agent lisofylline prevents autoimmune diabetic recurrence after islet transplantation.
Transplantation
77
:
55
–60,
2004
15
Dinarello CA: Therapeutic strategies to reduce IL-1 activity in treating local and systemic inflammation.
Curr Opin Pharmacol
4
:
378
–385,
2004
16
Dinarello CA: Blocking IL-1 in systemic inflammation.
J Exp Med
201
:
1355
–1359,
2005
17
Dinarello CA: The many worlds of reducing interleukin-1.
Arthritis Rheum
52
:
1960
–1967,
2005
18
Economides AN, Carpenter LR, Rudge JS, Wong V, Koehler-Stec EM, Hartnett C, Pyles EA, Xu XB, Daly TJ, Young MR, Fandl JP, Lee F, Carver S, McNnay J, Bailey K, Ramakanth S, Hutabarat R, Huang TT, Radziejewski C, Yancopoulos GD, Stahl N: Cytokine traps: multi-component, high-affinity blockers of cytokine action.
Nat Med
9
:
47
–52,
2003
19
Sandler S, Andersson A, Hellerström C: Inhibitory effects of interleukin-1 on insulin-secretion, insulin-biosynthesis, and oxidative-metabolism of isolated rat pancreatic-islets.
Endocrinology
121
:
1424
–1431,
1987
20
Andersson A, Sandler S: Viability tests of cryopreserved endocrine pancreatic cells.
Cryobiology
20
:
161
–168,
1983
21
Eizirik DL, Delaney CA, Green MHL, Cunningham, JM, Thorpe JR, Pipeleers DG, Hellerstrom C, Green IC: Nitric oxide donors decrease the function and survival of human pancreatic islets.
Mol Cell Endocrinol
118
:
71
–83,
1996
22
Delaney CA, Cunningham JM, Green MHL, Green IC: Nitric oxide rather than superoxide or peroxynitrite inhibits insulin secretion and causes DNA damage in HIT-T15 cells.
Adv Exp Med Biol
426
:
335
–339,
1997
23
Rothe H, Kolb H: Strategies of protection from nitric oxide toxicity in islet inflammation.
J Mol Med
77
:
40
–44,
1999
24
Andersen HU, Jørgensen KH, Egeberg J, Mandrupoulsen T, Nerup J: Nicotinamide prevents interleukin-1 effects on accumulated insulin release and nitric-oxide production in rat islets of Langerhans.
Diabetes
43
:
770
–777,
1994
25
Eizirik DL, Sandler S, Welsh N, Cetkovicvrlje M, Nieman A, Geller DA, Pipeleers DG, Bendtzen K, Hellerstrom C: Cytokines suppress human islet function irrespective of their effects on nitric-oxide generation.
J Clin Invest
93
:
1968
–1974,
1994
26
Dunger A, Cunningham JM, Delaney CA, Lowe JE, Green MHL, Bone AJ, Green IC: Tumor necrosis factor-α and interferon-γ inhibit insulin secretion and cause DNA damage in unweaned-rat islets: extent of nitric oxide involvement.
Diabetes
45
:
183
–189,
1996
27
Andersson AK, Flodstrom M, Sandler S: Cytokine-induced inhibition of insulin release from mouse pancreatic β-cells deficient in inducible nitric oxide synthase.
Biochem Biophys Res Commun
281
:
396
–403,
2001
28
Eizirik DL, Tracey DE, Bendtzen K, Sandler S: An interleukin-1 receptor antagonist protein protects insulin-producing β-cells against suppressive effects of interleukin-1-β.
Diabetologia
34
:
445
–448,
1991
29
Saldeen J, Sandler S, Bendtzen K, Welsh N: Liposome-mediated transfer of IL-1 receptor antagonist gene to dispersed islet cells does not prevent recurrence of disease in syngeneically transplanted NOD mice.
Cytokine
12
:
405
–408,
2000
30
Guler HP, Caldwell J, Littlejohn T, McIlwain H, Offenberg H, Stahl N: A phase 1, single dose escalation study of IL-1 trap in patients with rheumatoid arthritis.
Arthritis Rheum
44
:
S370
,
2001
31
Yang BB, Baughman S, Sullivan JT: Pharmacokinetics of anakinra in subjects with different levels of renal function.
Clin Pharmacol Ther
74
:
85
–94,
2003
32
Sandberg JO, Andersson A, Eizirik DL, Sandler S: Interleukin-1 receptor antagonist prevents low-dose streptozotocin-induced diabetes in mice.
Biochem Biophys Res Commun
202
:
543
–548,
1994
33
King A, Andersson A, Sandler S: Cytokine-induced functional suppression of microencapsulated rat pancreatic islets in vitro.
Transplantation
70
:
380
–383,
2000
34
Ling ZD, Intveld PA, Pipeleers DG: Interaction of interleukin-1 with islet β-cells: distinction between indirect, aspecific cytotoxicity and direct, specific functional suppression.
Diabetes
42
:
56
–65,
1993
35
Eizirik DL, Sandler S, Palmer JP: Repair of pancreatic β-cells: a relevant phenomenon in early IDDM.
Diabetes
42
:
1383
–1391,
1993