Heme oxygenase-1 (HO-1) is an enzyme with potent immunoregulatory capacity. To evaluate the effect of HO-1 on autoimmune diabetes, female NOD mice at 9 weeks of age received a single intravenous injection of a recombinant adeno-associated virus bearing HO-1 gene (AAV-HO-1; 0.5 × 1010-2.5 × 1010 viruses/mouse). In a dose-dependent manner, HO-1 transduction reduced destructive insulitis and the incidence of overt diabetes examined over a 15-week period. HO-1–mediated protection was associated with a lower type 1 T-helper cell (Th1)–mediated response. Adaptive transfer experiments in NOD.scid mice demonstrated that splenocytes isolated from AAV-HO-1–treated mice were less diabetogenic. Flow cytometry analysis revealed no significant difference in the percentages of CD4+CD25+ regulatory T-cells between saline-treated and AAV-HO-1–treated groups. However, the CD11c+ major histocompatibility complex II+ dendritic cell population was much lower in the AAV-HO-1–treated group. A similar protective effect against diabetes was observed in NOD mice subjected to carbon monoxide (CO) gas (250 ppm CO for 2 h, twice per week). These data suggest that HO-1 slows the progression to overt diabetes in pre-diabetic NOD mice by downregulating the phenotypic maturity of dendritic cells and Th1 effector function. CO appears to mediate at least partly the beneficial effect of HO-1 in this disease setting.

Type 1 diabetes is a chronic autoimmune disorder characterized by the progressive destruction of pancreatic insulin-producing β-cells (1). Studies on NOD mice, which spontaneously develop type 1 diabetes with features similar to the human disease, have demonstrated that the disease is initiated by infiltration of antigen-presenting cells (APCs), particularly dendritic cells, into pancreatic islets followed by recruitment of T- and B-cells (2,3). In these mice, insulitis appears around 3–4 weeks of age and is well established by 10 weeks without clinical symptoms. The progression to overt diabetes occurs in 80% of female mice by 30 weeks of age. Evidence suggests that β-cell destruction is mediated primarily by a skewed type 1 T helper cell (Th1)–mediated response and the production of proinflammatory cytokines (4). How the biased Th1 phenotype and the progression to destructive insulitis are regulated over the course of type 1 diabetes development is not fully understood. Nevertheless, it is well documented that full activation of naive T-cells requires presentation of antigen in the context of the major histocompatibility complex (MHC) II complex and costimulatory signals from mature dendritic cells (57). Dendritic cells from NOD mice have been shown to exhibit enhanced APC function and abnormally high costimulatory and Th1-inducing abilities (810). Furthermore, increasing evidence suggests that the autoimmune process is caused by the failure of immunosuppressive mechanisms (1113). A naturally occurring CD4+CD25+ regulatory T (Treg) cell population with suppressive function has been shown to control the progression from nondestructive insulitis to aggressive islet destruction in NOD mice (14). The CD4+CD25+ Treg cell population declines during autoimmune diabetes (14,15). In view of the complexity of type 1 diabetes etiology, the identification of endogenous molecules that modulate Th1 induction or immunosuppressive function may facilitate the design of effective therapies for preventing or delaying disease onset or progression.

Heme oxygenase-1 (HO-1) is a stress-response enzyme that catalyzes the degradation of heme to free iron, carbon monoxide (CO), and biliverdin in mammalian cells (16,17). Over the past few years, accumulated evidence has supported an immunoregulatory function for HO-1. It has been shown that HO-1 overexpression prevents graft rejection in organ transplantation (18). HO-1 and CO exhibit potent antiproliferative effects on T-cells, and HO-1 promotes activation-induced cell death of alloreactive T-cells (1922). A study on HO-1–deficient mice revealed that the absence of HO-1 correlates with a Th1-weighted shift in cytokine responses, suggesting a pivotal role for HO-1 in regulating the immune response (23). Furthermore, HO-1 is constitutively expressed in human CD4+CD25+ cells and is involved in immune suppression mediated by Foxp3, a master transcription factor regulating CD4+CD25+ cell development (24,25). A recent study revealed that HO-1 is expressed in immature dendritic cells, and HO-1 expression decreases with dendritic cell maturation (26). Induction of HO-1 expression inhibits dendritic cell maturation. These findings support the functional importance of HO-1 in immune regulation.

The aforementioned properties of HO-1 provoked our interest in investigating the impact of HO-1 on the development of type 1 diabetes in NOD mice. To determine the effect of HO-1 on the progression to overt diabetes in pre-diabetic mice with insulitis, we performed systemic gene transfer of HO-1 mediated by adeno-associated virus (AAV) in female NOD mice at 9 weeks of age. The effect of HO-1 gene transduction on the onset of diabetes was then assessed over a 15-week period. Our results clearly showed that systemic HO-1 expression suppressed the progression of insulitis and delayed the onset of hyperglycemia.

Construction of recombinant AAV vector.

We used the AAV helper-free system from Stratagene (La Jolla, CA) for vector construction. Mouse HO-1 cDNA was obtained by RT-PCR of RNA isolated from heme-treated murine J774 cells and subcloned into the pCMV-MCS plasmid. Recombinant AAV vector was then prepared according to the manufacturer's instructions. The large-scale production and purification of AAV virus were conducted using heparin affinity chromatography as described by Clark et al. (27). The recombinant AAV particles were quantified by real-time PCR using a PE-Applied Biosystems Prism 7700 sequence detector (Foster City, CA) and fluorescent probe sets. The primers and probe used for quantification of AAV-HO-1 were as follows: HO-1 5′ primer, 5′-AGGTGATGCTGACAGAGGAACAC-3′; HO-1 3′ primer, 5′-TAGCAGGCCTCTGACGAAGTG-3′; HO-1 probe, 5′-6-carboxy-fluorescein-AAGACCAGAGTCCCTCACAGATGGCG-6-carboxy-tetramethyl-rhodamine-3′.

Animals.

Female NOD and NOD.scid mice were purchased from The Jackson Laboratories (Bar Harbor, ME) and kept in specific pathogen-free conditions. Female NOD mice at 9 weeks of age were subjected to retro-orbital plexus injection of saline or the indicated amounts of AAV-HO-1 in 50 μl saline. Some NOD mice at 8–9 weeks of age received two intraperitoneal injections of cyclophosphamide (200 mg/kg body wt) with a 1-week interval to facilitate the onset of diabetes. To test the effect of CO, animals were exposed to 250 ppm CO gas as described previously (28) for 2 h twice per week. Animals were monitored twice per week for hyperglycemia as defined by two consecutive nonfasting blood glucose levels >240 mg/dl. Mice were killed at 24 weeks of age. All animal procedures were approved by the institutional animal care and use committee of Academia Sinica.

Splenocyte studies.

Splenocytes from all mice killed at 24 weeks of age were isolated and cultured at 5 × 105 cells/well in 200 μl RPMI 1640 containing 10% fetal bovine serum in a 96-well, flat-bottom plate. After incubation with or without concanavalin A (ConA; 5 μg/ml per well) for 48 h, media were collected for the measurement of cytokine production. For flow cytometric experiments, splenocytes (1 ×106 cells/ml) were preincubated with purified rat anti-mouse CD16/CD32 (eBioscience) to block Fc receptor binding, followed by staining with fluorescein isothiocyanate (FITC)-conjugated anti-CD4 antibody (BD Biosciences), phycoethrin-conjugated anti-CD25 antibody (BD Biosciences), APC-conjugated anti-CD11c antibody (BD Biosciences), or FITC-conjugated anti–I-Ag7 MHC II antibody (BD Biosciences) as indicated. To examine intracellular Foxp3 expression, splenocytes were immunostained with an APC–anti-mouse Foxp3 staining set (eBiosciences) according to the manufacturer's instruction. Flow cytometry data were acquired and analyzed using a FACS Calibur (Becton Dickinson) and Cell Quest software (version 3.3; Becton Dickinson).

Adoptive transfer.

Freshly isolated splenocytes from 24-week-old saline-treated or AAV-HO-1–treated (2.5 × 1010 genome particles) NOD mice were injected at 1 × 107 cells/mouse into 5-week-old female NOD.scid mice via the retro-orbital plexus. Hyperglycemia was then assessed twice per week.

Cytokine measurements.

Cytokines interleukin (IL)-2, IL-4, IL-10, and γ-interferon (IFN-γ) were measured by ELISA kits (R&D Systems).

Histopathology.

The pancreas was removed, fixed in Bouin's solution (Sigma) for 16 h, embedded in paraffin, and sectioned at 5 μm. Sections were hematoxylin/eosin stained and evaluated on a blinded basis by two individuals using scoring criteria described by others (29): stage 0, normal islet; stage 1, peri-insulitis; stage 2, insulitis in <50% of the islet; and stage 3, insulitis in >50% of the islet. At least 20 islets from each pancreas were examined.

Immunohistochemistry.

Sections were deparaffinized, rehydrated, and pretreated with target retrieval solution (Dako) at 95°C for 30 min. Endogenous peroxidase was blocked in a solution of 3% H2O2 for 10 min at room temperature. After incubation with 5% bovine serum albumin in PBS at 37°C for 30 min, sections were incubated with rabbit polyclonal anti–HO-1 (1:100; Stressgen) at 37°C for 1 h, followed by three washes with PBS containing 0.1% Tween-20 (PBST). Sections were then incubated with horseradish peroxidase–conjugated secondary antibody (1:200) for another 1 h at 37°C. After three washes, antigen was visualized after incubation with diaminobenzidine/H2O2 followed by counterstain with hematoxylin reagent. Negative control was performed by using rabbit normal IgG as the primary antibody.

Western blot analysis.

Tissues were homogenized in lysis buffer containing 25 mmol/l Tris-HCl, pH 7.9, 1% Triton X-100, 10 mmol/l EDTA, 1 mmol/l sodium vanadate, 1 mmol/l phenylmethylsulfonyl fluoride (PMSF), and 1 μg/ml leupeptin. After centrifugation at 14,000 rpm for 30 min, the supernatant was removed, and protein concentration was determined by Bio-Rad protein assay. An equal amount of protein (50 μg) from each sample was separated by SDS-PAGE and transblotted onto nitrocellulose membranes. After blocking for 12 h with 5% nonfat milk in PBST, membranes were incubated with rabbit anti–HO-1 antibody (Stressgen), rabbit anti–HO-2 antibody (Santa Cruz), or rabbit anti–glyceraldehyde-3-phosphate dehydrogenase antibody (Santa Cruz) for 1 h at 37°C. After three washes with PBST, membranes were incubated with horseradish peroxidase–conjugated secondary antibody for 1 h. Antigen was then detected using the ECL system.

HO activity measurement.

Tissues were homogenized in ice-cold 0.1 mol/l potassium phosphate buffer, pH 7.4, containing 1 mmol/l EDTA, and 0.5 mmol/l PMSF and centrifuged at 13,000 rpm at 4°C for 10 min. Supernatant proteins (1 mg) were then incubated in dark with 200 μl of reaction mixture containing mouse liver cytosol (1 mg protein), 50 μmol/l hemin, 1 mmol/l NADPH, 2 mmol/l glucose-6-phosphate, and 0.2 unit glucose-6-phosphate dehydrogenase in 0.1 mol/l potassium phosphate buffer, pH 7.4, at 37°C for 1 h. Bilirubin was then extracted with 1 ml chloroform and measured by the absorbance difference between 464 and 530 nm with an extinction coefficient of 40 mmol/l per centimeter.

Statistical analysis.

Data were presented as mean ± SD and analyzed using the Mann-Whitney-Wilcoxon test. Hyperglycemia frequencies and survival rates were analyzed by the Mantel-Haenszel χ2 test. P < 0.05 was considered statistically significant.

Effect of AAV-mediated systemic HO-1 transduction on type 1 diabetes in NOD mice.

The expression levels of HO-1 and HO-2 and the net HO activity in NOD mice were first examined. Western blot analysis revealed no significant difference in the expression levels of HO-1 or HO-2 in lung, pancreas, and liver between young (10 weeks) and old (24 weeks) mice (Fig. 1A and B). However, the HO activities measured in indicated tissues appeared to be lower in old NOD mice compared with young counterparts (Fig. 1C). To evaluate the potential effect of HO-1 on type 1 diabetes development, we constructed a recombinant AAV vector carrying mouse HO-1 gene for the in vivo gene transfer in NOD mice. AAV-mediated transgene expression was first confirmed in HT1080 cells (data not shown). To assess the effect of HO-1 on type 1 diabetes, 9-week-old female NOD mice received saline or various doses of AAV-HO-1 via retro-orbital plexus injection. The incidence of hyperglycemia in each group was examined for 15 weeks. As shown in Fig. 2A, 10% of the saline-treated control mice developed hyperglycemia in the following 1–3 weeks. In contrast, the onset of diabetes was substantially delayed in mice receiving AAV-HO-1. The salutary effect of AAV-HO-1 was dose dependent, and 85% of mice receiving the highest dose of AAV-HO-1 (2.5 × 1010 virus particles/mouse) remained normoglycemic at the time of killing (P < 0.001 vs. control group). Histological assessment of pancreatic sections from the animals revealed that the extent of insulitis was inversely correlated with the dose of AAV-HO-1 received (Fig. 2C). In contrast to the control group in which only ∼15% of the examined islets were devoid of lymphocyte infiltration, >50% of the examined islets in the group treated with the highest dose of AAV-HO-1 were normal (P < 0.01 vs. control group). To confirm that this protective effect resulted from sustained transgene expression, Western blot analysis was performed to examine HO-1 expression in various tissues 15 weeks after gene delivery. As shown in Fig. 3A and B, the levels of HO-1 protein in liver and spleen, which express high basal levels of HO-1, were not significantly enhanced by AAV-HO-1 administration. In contrast, HO-1 expression in lung, heart, and pancreas was significantly higher in mice receiving AAV-HO-1 (2.5 × 1010 virus particles/mouse) compared with control counterparts. Likewise, HO activities determined in these tissues were also higher in AAV-HO-1–treated mice (Fig. 3C). When immunostaining experiment was performed with pancreatic sections, it was noted that HO-1 was primarily expressed in β-cells of islets and was much prominent in mice receiving AAV-HO-1 (Fig. 3D). Together, these results support an inverse association between HO-1 expression and the onset of autoimmune diabetes in NOD mice.

Effect of HO-1 transduction on diabetogenic properties of T-cells.

To examine whether HO-1 expression affects the balance of Th1/Th2 responses, we measured Th1 and Th2 cytokines in serum samples and in media from ConA-activated splenocytes isolated from surviving animals 15 weeks after saline or AAV-HO-1 (2.5 ×1010 particles) administration. As shown in Fig. 4A, significant decreases in serum levels of IL-2 and IFN-γ were observed in mice receiving AAV-HO-1. However, the levels of IL-4 and IL-10 were not significantly different between the saline- and AAV-HO-1–treated groups. Likewise, the production of IL-2 and IFN-γ, but not of IL-4 and IL-10, by activated splenocytes was substantially lower in AAV-HO-1–treated mice (Fig. 4B). To further confirm that HO-1 overexpression reduced the diabetogenicity of lymphocytes, an adoptive transfer experiment was performed in female NOD.scid mice. Splenocytes isolated from 24-week-old NOD mice with or without AAV-HO-1 treatment for 15 weeks were transferred into 5-week-old NOD.scid mice. Results showed that 9 of 10 mice receiving splenocytes from control donors became diabetic by 4 weeks after transfer, whereas the onset of diabetes was significantly delayed in recipients receiving splenocytes from AAV-HO-1–treated donors (P < 0.05) (Fig. 4C).

Effect of HO-1 transduction on immunoregulatory cells.

To determine whether systemic HO-1 overexpression affects the level of CD4+CD25+ Treg cells, splenocytes isolated from 24-week-old mice pretreated without or with AAV-HO-1 (2.5 × 1010 particles) for 15 weeks were subjected to flow cytometry analysis. The percentage of CD4+CD25+ Treg cells in total splenocytes of the AAV-HO-1–treated group (2.60 ± 0.12%, n = 3) was not significantly different from that of the saline-treated group (2.73 ± 0.28%, n = 5) (Fig. 5A). When the frequencies of Foxp3 expression were examined, again there was no significant difference in the percentage of CD4+Foxp3+ cells between saline and AAV-HO-1–treated groups (3.74 ± 0.35 vs. 3.57 ± 0.11%). We next examined the population of CD11c+ dendritic cells, which are crucial for the activation of autoreactive T-cells. The percentage of CD11c+ splenocytes was similar between the control and AAV-HO-1–treated groups (0.70 ± 0.14 vs. 0.74 ± 0.26%). However, when the expression level of MHC II, an index of dendritic cell maturation, in this cell population was analyzed, it was found that the percentage of CD11c+MHC II+ cells was reduced by 35% in the AAV-HO-1–treated group compared with the saline-treated control (Fig. 5B and C).

Effect of CO exposure on type 1 diabetes.

To determine whether CO, a by-product of heme degradation, has a role in HO-1–mediated type 1 diabetes protection, we exposed 9-week-old NOD mice to a low dose of CO gas (250 ppm for 2 h) twice per week. As shown in Fig. 6A, CO treatment for a 15-week period did not significantly affect the expression levels of HO-1 in tissues examined. However, the incidence of diabetes in the CO-treated group was significantly reduced compared with that of the control group (P < 0.001) (Fig. 6B). Consistently, insulitis was less severe in CO-treated animals (Fig. 6C). In contrast to control mice, in which only ∼12% of the examined islets were normal, ∼40% of examined islets were normal in CO-treated mice (P < 0.01 vs. control group). To examine whether CO treatment also affects the Th1/Th2 response, cytokine expression in ConA-activated splenocytes isolated from control and CO-treated mice was assessed. As shown in Fig. 7, the levels of IL-2 and IFN-γ, but not of IL-4 and IL-10, were significantly lower in the CO-treated group compared with the control group. To further explore whether CO exerts protective effect on mice developing overt diabetes, NOD mice at 8–9 weeks of age received two intraperitoneal injections of cyclophosphamide (200 mg/kg body wt) to accelerate the onset of diabetes. Fifty percent of mice became diabetic within 3 weeks after the first injection of cyclophosphamide (data not shown). Mice with diabetes onset were either left untreated or treated with CO gas as described above beginning at day 3 after the disease onset (day 0) twice per week. It was found that CO treatment did not affect the hyperglycemia of diabetic mice (data not shown). However, CO significantly prolonged the survival of these animals (Fig. 8). In contrast to the untreated group in which 50% of the animals died at the first week after diabetes onset, all CO-treated animals were still alive at 6 weeks after the disease onset (P < 0.05 vs. control group).

Decreased HO-1 expression has been shown in human patients with diabetes (30,31). Studies on experimental diabetes also revealed lower levels of HO-1 expression and HO activity in streptozotocin-induced diabetic rats compared with nondiabetic controls (32,33). In the present study, we did not observe significant changes in HO-1 and HO-2 protein expression along with aging process in NOD mice. However, a significant reduction in net HO activity was noticed in old NOD mice. This finding appears to be similar to a recent report showing that hyperglycermia did not affect HO-1 and HO-2 expression but caused a decrease in HO-1 activity (33). It is conceivable that development of hyperglycermia in old NOD mice may have an impact on HO activity. Earlier studies by others have demonstrated that HO-1 overexpression can attenuate vascular endothelial damage and prevent cardiac ischemia/reperfusion injury in experimental diabetes (32,3436). Furthermore, overexpression of HO-1 in islets provided protection from apoptosis and increased allograft survival (3740). These findings support the multiple protective roles of HO-1 in diabetes. However, whether HO-1 has an immunoregulatory function in the course of type 1 diabetes development remains to be established. To address this issue, in the present study, we performed in vivo HO-1 gene transfer in NOD mice using AAV vector that has been shown to mediate sustained and stable transgene expression in animals. Our results demonstrated that a single intravenous injection of AAV-HO-1 in pre-diabetic NOD mice significantly attenuated the progression of destructive insulitis and the onset of diabetes. The protection conferred by AAV-HO-1 appears to be correlated with the systemic induction of HO-1 in several tissues, including the pancreas. Immunostaining of pancreatic sections revealed that HO-1 was predominantly expressed in β-cells. Because we did not specifically investigate whether pancreatic HO-1 overexpression can prevent the autoimmune destruction of β-cells in situ, this possibility cannot be completely ruled out in the present experimental setting. Nevertheless, we provide evidence showing that systemic HO-1 gene transduction resulted in suppression of the Th1 cell response. AAV-HO-1 administration significantly reduced the levels of the Th1-type cytokines IL-2 and IFN-γ in circulation and in activated splenocytes, without affecting the levels of the Th2-type cytokines IL-4 and IL-10. An influence of HO-1 on the Th1/Th2 cytokine profile has also been reported in a study of liver allografts after HO-1 gene transfer (41). A recent study on HO-1–deficient mice revealed a profound Th1 response after T-cell stimulation (23). The preferential suppression of Th1 effector cell function by HO-1 suggests that HO-1 has a beneficial effect in various diseases involving the Th1-mediated immune response. Because destructive insulitis is closely associated with Th1 cell activation and Th1-type cytokine production, it is apparent that HO-1 suppresses type 1 diabetes in NOD mice by influencing the diabetogenicity of T-cells. Adoptive transfer of splenocytes from AAV-HO-1–treated mice demonstrated late onset of diabetes in NOD.scid recipients, confirming the immunosuppressive effect of HO-1 on autoreactive T-cells.

Over the past few years, considerable evidence has supported the vital role of CD4+CD25+ Treg cells in maintaining peripheral tolerance (1113). A decline in the CD4+CD25+ Treg cell population was noted in NOD mice (14,15). Transfer of polyclonal CD4+CD25+ Treg cells prevented diabetes in NOD mice (42). Furthermore, systemic overexpresssion of IL-10 in NOD mice increased the CD4+CD25+ Treg population and ameliorated diabetes (43). Recently, HO-1 has been implicated in the suppressive capacity of CD4+CD25+ Treg cells (44). To examine whether HO-1–mediated type 1 diabetes protection in NOD mice involves regulation of CD4+CD25+ Treg cells, we analyzed the percentage of CD4+CD25+ Treg cells in splenocytes. Our results showed that the AAV-HO-1–treated group did not show a higher proportion of CD4+CD25+ Treg cells in splenocytes, indicating that HO-1–induced suppression of diabetogenic T-cells is not achieved by elevating the Treg population. Because the present study did not assess the suppressive activity of CD4+CD25+ Treg cells, it is unclear whether HO-1 affected CD4+CD25+ Treg function in our experimental setting. Further study is required to clarify this issue. Along with Treg cells, dendritic cells play an integral role in controlling autoimmunity and self tolerance. Recently, it has been demonstrated that immature dendritic cells induce peripheral tolerance by either increasing Treg cells or inducing T-cell anergy (45). Dendritic cells in NOD mice have abnormally high immunostimulatory and Th1-inducing abilities (810). Downregulating the phenotypic maturation of dendritic cells suppresses the immune response and induces peripheral tolerance in NOD mice (46). A recent study showed that HO-1 expression inhibits dendritic cell maturation and proinflammatory and allogeneic immune responses while preserving IL-10 production (26). It is of great interest to determine whether dendritic cell maturation is affected by HO-1 in NOD mice. Flow cytometry analysis revealed that the total number of CD11c+ splenocytes was not altered by AAV-HO-1 administration in NOD mice. However, the percentage of CD11c+ cells expressing I-Ag7 MHC II was reduced by 35% in AAV-HO-1–treated mice compared with the control group. This finding supports the idea that HO-1 overexpression in NOD mice suppressed the functional maturation of dendritic cells, which in turn limited Th1 induction and disease progression.

HO-1 is an endoplasmic reticulum–anchored protein, and its protective functions are primarily mediated through its reaction by-products (16,17). Recently, studies have shown that both CO and bilirubin can prevent endothelial cell sloughing and prolong the survival of allogeneic islets in type 1 diabetes, likely through different mechanisms (33,47,48). In the present study, we specifically assessed the potential role of CO in HO-1–mediated type 1 diabetes protection in NOD mice. Animals were subjected to a 2-h exposure of low-dose CO twice per week for a period of 15 weeks. We admit that this treatment may not exactly resemble the same situation in AAV-HO-1–treated mice in which CO was constantly produced by HO-1 in situ. Nevertheless, it was interesting to note that the pre-diabetic mice subjected to the periodic CO treatment had a significantly lower incidence of diabetes compared with the untreated control mice. Similar to what we observed in the AAV-HO-1–treated mice, Th1 cytokines produced by activated splenocytes were markedly reduced in the CO-treated group. Previous studies on various disease settings in animals have revealed multiple cellular mechanisms underlying the protections conferred by CO inhalation. For example, CO has been shown to activate heme-containing soluble guanylyl cyclase to increase cellular cGMP level or modulate p38 kinase-mediated signaling transduction (17). Recently, there are reports showing that CO treatment can protect animals against liver failure and chronic colitis through induction of HO-1 expression (49,50). We did not observe changes in the level of HO-1 protein expression in NOD mice after CO treatment, suggesting that the beneficial effect of CO observed in present experimental setting is unlikely mediated through upregulating HO-1 expression. Nevertheless, to completely rule out the implication of HO-1, additional experiments with the condition of HO-1 activity inhibition or HO-1 deficiency in NOD mice will be needed. The detailed mechanism responsible for the protective effect of exogenous CO treatment on type 1 diabetes remains to be determined. Further experiment demonstrated that the periodic CO treatment also prolonged the survival of NOD mice with chemical-induced overt diabetes. Together, these results suggest a possible role for CO in the protective function of HO-1 in NOD mice.

In conclusion, the present study provides convincing evidence to support an immunosuppressive function of HO-1 during the course of type 1 diabetes. The beneficial effect of HO-1 is most likely through downregulating dendritic cell maturation, thereby limiting T-cell activation and Th1 response. These findings suggest a new therapeutic approach for treating type 1 diabetes by manipulating the expression level of HO-1 with pharmacological agents or gene-based therapy.

FIG. 1.

HO expression in NOD mice. A: HO Western blots in indicated tissues of young (10 weeks) and old (24 weeks) mice. B and C: Quantitative results of Western blots and HO activities in young (▪) and old (□) mice. *P < 0.05 vs. young group.

FIG. 1.

HO expression in NOD mice. A: HO Western blots in indicated tissues of young (10 weeks) and old (24 weeks) mice. B and C: Quantitative results of Western blots and HO activities in young (▪) and old (□) mice. *P < 0.05 vs. young group.

Close modal
FIG. 2.

HO-1 overexpression delayed type 1 diabetes in NOD mice. A: Animals received an administration of saline (•; n = 18) or AAV-HO-1 at doses of 0.5 × 1010 (○; n = 10), 1 × 1010 (▴; n = 10), or 2.5 × 1010 (▵; n = 20) viruses/mouse. The percentage of normoglycemic mice in each group was determined. B: Representative hematoxylin-eosin–stained pancreatic sections showing various stages of insulitis. C: Histogram depicting percentages of normal islets (stage 0, ▪) and islets with insulitis at various stages (stage 1, ; stage 2, ; and stage 3, □) in different groups.

FIG. 2.

HO-1 overexpression delayed type 1 diabetes in NOD mice. A: Animals received an administration of saline (•; n = 18) or AAV-HO-1 at doses of 0.5 × 1010 (○; n = 10), 1 × 1010 (▴; n = 10), or 2.5 × 1010 (▵; n = 20) viruses/mouse. The percentage of normoglycemic mice in each group was determined. B: Representative hematoxylin-eosin–stained pancreatic sections showing various stages of insulitis. C: Histogram depicting percentages of normal islets (stage 0, ▪) and islets with insulitis at various stages (stage 1, ; stage 2, ; and stage 3, □) in different groups.

Close modal
FIG. 3.

HO-1 expression in mice after receiving saline or AAV-HO-1 (2.5 × 1010 viruses/mouse) administration. A: HO-1 Western blots in indicated tissues. B and C: Quantitative results of Western blots and HO activities in saline-treated (▪) and AAV-HO-1–treated (□) groups. *P < 0.05 vs. saline-treated group. D: Representative images showing HO-1 immunostaining in pancreatic sections. Original magnification ×200.

FIG. 3.

HO-1 expression in mice after receiving saline or AAV-HO-1 (2.5 × 1010 viruses/mouse) administration. A: HO-1 Western blots in indicated tissues. B and C: Quantitative results of Western blots and HO activities in saline-treated (▪) and AAV-HO-1–treated (□) groups. *P < 0.05 vs. saline-treated group. D: Representative images showing HO-1 immunostaining in pancreatic sections. Original magnification ×200.

Close modal
FIG. 4.

Effect of HO-1 on T-cell response. A: Serum cytokine levels in mice receiving saline (n = 5) or AAV-HO-1 (n = 9) for 15 weeks. *P < 0.05 vs. saline-treated group. B: Cytokine levels in culture medium of splenocytes from saline-treated (n = 6) and AAV-HO-1–treated (n = 8) groups in the absence (▪) or presence (□) of ConA activation. *P < 0.05 vs. saline-treated group. C: NOD.scid mice received splenocytes from 24-week-old NOD mice previously treated with saline (•) or AAV-HO-1 (○) (n = 10 for each group). Recipients were monitored for diabetes every other day.

FIG. 4.

Effect of HO-1 on T-cell response. A: Serum cytokine levels in mice receiving saline (n = 5) or AAV-HO-1 (n = 9) for 15 weeks. *P < 0.05 vs. saline-treated group. B: Cytokine levels in culture medium of splenocytes from saline-treated (n = 6) and AAV-HO-1–treated (n = 8) groups in the absence (▪) or presence (□) of ConA activation. *P < 0.05 vs. saline-treated group. C: NOD.scid mice received splenocytes from 24-week-old NOD mice previously treated with saline (•) or AAV-HO-1 (○) (n = 10 for each group). Recipients were monitored for diabetes every other day.

Close modal
FIG. 5.

Effect of HO-1 overexpression on splenocytes. Flow cytometry analysis of Treg population (A) and CD11c+ MHC II+ dendritic cell population (B) in spleens of NOD mice treated with saline or AAV-HO-1. C: The percentage of CD11c+ MHC II+ dendritic cells in saline-treated (n = 5) or AAV-HO-1–treated (n = 6) group. *P < 0.05 vs. saline-treated group.

FIG. 5.

Effect of HO-1 overexpression on splenocytes. Flow cytometry analysis of Treg population (A) and CD11c+ MHC II+ dendritic cell population (B) in spleens of NOD mice treated with saline or AAV-HO-1. C: The percentage of CD11c+ MHC II+ dendritic cells in saline-treated (n = 5) or AAV-HO-1–treated (n = 6) group. *P < 0.05 vs. saline-treated group.

Close modal
FIG. 6.

Effect of CO on type 1 diabetes progression. NOD mice were untreated (•; n = 18) or treated (○; n = 20) with CO gas twice per week for 15 weeks. A: Representative HO-1 Western blots. B: The percentage of normoglycemic mice in each group was assessed. C: The severity of insulitis in each group was scored at various stages (stage 0, ▪; stage 1, ; stage 2, ; and stage 3, □).

FIG. 6.

Effect of CO on type 1 diabetes progression. NOD mice were untreated (•; n = 18) or treated (○; n = 20) with CO gas twice per week for 15 weeks. A: Representative HO-1 Western blots. B: The percentage of normoglycemic mice in each group was assessed. C: The severity of insulitis in each group was scored at various stages (stage 0, ▪; stage 1, ; stage 2, ; and stage 3, □).

Close modal
FIG. 7.

Effect of CO on splenocyte function. Splenocytes from control (n = 4) and CO-treated (n = 6) NOD mice were incubated with (□) or without (▪) ConA for 48 h. Cytokine levels in culture medium were determined. *P < 0.05 vs. control group.

FIG. 7.

Effect of CO on splenocyte function. Splenocytes from control (n = 4) and CO-treated (n = 6) NOD mice were incubated with (□) or without (▪) ConA for 48 h. Cytokine levels in culture medium were determined. *P < 0.05 vs. control group.

Close modal
FIG. 8.

Effect of CO on the survival of diabetic mice. Chemical-induced diabetic mice were untreated (•; n = 6) or treated with CO (○; n = 3) starting from day 3 after disease onset. The percentage of survival in each group over a period of 6 weeks was determined.

FIG. 8.

Effect of CO on the survival of diabetic mice. Chemical-induced diabetic mice were untreated (•; n = 6) or treated with CO (○; n = 3) starting from day 3 after disease onset. The percentage of survival in each group over a period of 6 weeks was determined.

Close modal

Published ahead of print at http://diabetes.diabetesjournals.org on 15 February 2007. DOI: 10.2337/db06-0495.

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.

L.-Y.C. has received the Academia Sinica Investigatorship. This work has received support from the Institute of Biomedical Sciences, Academia Sinica.

1.
Tisch R, McDevitt H: Insulin-dependent diabetes mellitus.
Cell
85
:
291
–297,
1996
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.
Andre I, Gonzalez A, Wang B, Katz J, Benoist C, Mathis D: Checkpoints in the progression of autoimmune diseases: lesions from diabetes models.
Proc Natl Acad Sci U S A
93
:
2260
–2263,
1996
4.
Amrani A, Verdaguer J, Serra P, Tafuro S, Tan R, Suntamaria P: Progression of autoimmune diabetes driven by avidity maturation of a T cell population.
Nature
406
:
739
–742,
2000
5.
Banchereau J, Steinman RM: Dendritic cells and the control of immunity.
Nature
392
:
245
–252,
1998
6.
Banchereau J, Briere F, Caux C, Davoust J, Lebecque S, Liu YJ, Pulendran B, Palucka K: Immunobiology of dendritic cells.
Annu Rev Immunol
18
:
767
–811,
2000
7.
Sallusto F, Lanzavecchia A: Mobilizing dendritic cells for tolerance, priming and chronic inflammation.
J Exp Med
189
:
611
–614,
1999
8.
Weaver DJ, Poligone B, Bui T, Abdel-Motal UM, Baldwin AS, Tisch R: Dendritic cells from nonobese diabetic mice exhibit a defect in NF-κB regulation due to a hyperactive IκB kinase.
J Immunol
167
:
1461
–1468,
2001
9.
Poligone B, Weaver DJ, Sen P, Baldwin AS, Tisch R: Elevated NF-κB activation in nonobese diabetic mouse dendritic cells results in enhanced APC function.
J Immunol
168
:
188
–196,
2002
10.
Marleau AM, Singh B: Myeloid dendritic cells in non-obese diabetic mice have elevated costimulatory and T helper-1-inducing abilities.
J Autoimmunity
19
:
23
–35,
2002
11.
Shevoch EM: CD4+CD25+ suppressor T cells: more questions than answers.
Nat Rev Immunol
2
:
389
–400,
2002
12.
Maloy KJ, Powrie F: Regulatory T cells in the control of immune pathology.
Nat Immunol
2
:
816
–822,
2001
13.
Sakaguchi S, Sakaguchi N, Shimizu J, Yamazaki S, Sakihama T, Itoh M, Kuniyasu Y, Nomura T, Toda M, Takahashi T: Immunologic tolerance maintained by CD4+CD25+ regulatory T cells: their common role in controlling autoimmunity, tumor immunity and transplantation tolerance.
Immunol Rev
182
:
18
–32,
2001
14.
Salomon B, Lenschow DJ, Rhee L, Ashourian N, Singh B, Sharpe A, Bluestone JA: B7/CD28 costimulation is essential for the homeostasis of the CD4+CD25+ immunoregulatory T cells that control autoimmune diabetes.
Immunity
12
:
431
–440,
2000
15.
Pop SM, Wong CP, Culton DA, Clarke SH, Tisch R: Single cell analysis shows decreasing Fox P3 and TGF β1 coexpressing CD4+CD25+ regulatory T cells during autoimmune diabetes.
J Exp Med
201
:
1333
–1346,
2005
16.
Otterbein LE, Choi AM: Heme oxygenase: colors of defense against cellular stress.
Am J Physiol Lung Cell Mol Physiol
279
:
L1029
–L1037,
2000
17.
Otterbein LE, Soares MP, Yamashita K, Bach FH: Heme oxygenase-1: unleashing the protective properties of heme.
Trends Immunol
24
:
449
–455,
2003
18.
Kator M, Busuttil RW, Kupiec-Weglinski JW: Heme oxygenase-1 system in organ transplantation.
Transplantation
74
:
905
–912,
2002
19.
Song R, Mahidhara RS, Zhou Z, Hoffman RA, Seol DW, Flavell RA, Billiar TR, Otterbein LE, Choi AM: Carbon monoxide inhibits T lymphocyte proliferation via caspase-dependent pathway.
J Immunol
172
:
1220
–1226,
2004
20.
Pae HO, Choi BM, Oh GS, Lee MS, Ryu DG, Rhew HY, Kim YM, Chung HT: Roles of heme oxygenase-1 in the antiproliferative and antiapoptotic effects of nitric oxide on Jurkat T cells.
Mol Pharmacol
66
:
122
–128,
2004
21.
Pae HO, Oh GS, Choi BM, Chae SC, Kim YM, Chung KR, Chung HT: Carbon monoxide produced by heme oxygenase-1 suppresses T cell proliferation via inhibition of IL-2 production.
J Immunol
172
:
4744
–4751,
2004
22.
McDaid J, Yamashita K, Chora A, Ollinger R, Strom TB, Li XC, Bach FH, Soares MP: Heme oxygenase-1 modulates the allo-immune response by promoting activation-induced cell death of T cells.
FASEB J
19
:
458
–460,
2005
23.
Kapturczak MH, Wasserfall C, Brusko T, Cambell-Thompson M, Ellis TM, Atkinson MA, Agarwal A: Heme oxygenase-1 modulates early inflammatory responses: evidence from the heme oxygenase-1-deficient mouse.
Am J Pathol
165
:
1045
–1053,
2004
24.
Pae HO, Oh GS, Choi BM, Chae SC, Chung HT: Differential expression of heme oxygenase-1 gene in CD25 and CD25+ subsets of human CD4+ T cells.
Biochem Biophys Res Commun
306
:
701
–705,
2003
25.
Choi BM, Pae HO, Jeong YR, Kim YM, Chung HT: Critical role of heme oxygenase-1 in Foxp3-mediated immune suppression.
Biochem Biophys Res Commun
327
:
1066
–1071,
2005
26.
Chauveau C, Remy S, Royer PJ, Hill M, Tanguy-Royer S, Hubert FX, Tesson L, Brion R, Beriou G, Gregoire M, Josien R, Cuturi MC, Anegon I: Heme oxygenase-1 expression inhibits dendritic cell maturation and proinflammatory function but conserves IL-10 expression.
Blood
106
:
1694
–1702,
2005
27.
Auricchio A, Hildinger M, O'Connor E, Gao G-P, Wilson JM: Isolation of highly infectious and pure adeno-associated virus type 2 vectors with a single-step gravity-flow column.
Human Gene Ther
12
:
71
–76,
2001
28.
Lee TS, Chau LY: Heme oxygenase-1 mediates the anti-inflammatory effect of interleukin-10 in mice.
Nat Med
8
:
240
–246,
2002
29.
Arreaza GA, Cameron MJ, Jaramillo A, Gill BM, Hardy D, Laupland KB, Rapoport MJ, Zucker P, Chakrabarti S, Chensue SW, Qin HY, Singh B, Delovitch TL: Neonatal activation of CD28 signaling overcomes T cell anergy and prevents autoimmune diabetes by an IL-4-dependent mechanism.
J Clin Invest
100
:
2243
–2253,
1997
30.
da Silva JL, Stoltz RA, Dunn MW, Abraham NG, Shibahara S: Diminished heme oxygenase-1 mRNA expression in RPE cells from diabetic donors as quantitated by competitive RT/PCR.
Curr Eye Res
16
:
380
–386,
1997
31.
Bruce CR, Carey AL, Hawley JA, Febbraio MA: Intramuscular heat shock protein 72 and heme oxygenase-1 mRNA are reduced in patients with type 2 diabetes: evidence that insulin resistance is associated with a disturbed antioxidant defense mechanism.
Diabetes
52
:
2338
–2345,
2003
32.
Di Filippo C, Marfella R, Cuzzocrea S, Piegari E, Petronella P, Giugliano D, Rossi F, D'Amico M: Hyperglycemia in streptozotocin-induced diabetic rat increases infarct size associated with low levels of myocardial HO-1 during ischemia/reperfusion.
Diabetes
54
:
803
–810,
2005
33.
Rodella L, Lamon BD, Rezzani R, Sangras B, Goodman AI, Falck JR, Abraham NG: Carbon monoxide and biliverdin prevent endothelial cell sloughing in rats with type I diabetes.
Free Radic Biol Med
40
:
2198
–2205,
2006
34.
Quan S, Kaminski PM, Yang L, Morita T, Inaba M, Ikehara S, Goodman AI, Wolin MS, Abraham NG: Heme oxygnase-1 prevents superoxide anion associated endothelial cell sloughing in diabetic rats.
Biochem Biophys Res Commun
315
:
509
–516,
2004
35.
Abraham NG, Rezzani R, Rodella L, Kruger A, Taller D, Li VG, Goodman AI, Kappas A: Overexpression of human heme oxygenase-1 attenuates endothelial cell sloughing in experimental diabetes.
Am J Physiol Heart Circ Physiol
287
:
H2468
–H2477,
2004
36.
Liu X, Wei J, Peng DH, Layne MD, Yet SF: Absence of heme oxygenase-1 exacerbates myocardial ischemia-reperfusion injury in diabetic mice.
Diabetes
54
:
778
–784,
2005
37.
Pileggi A, Molano RD, Berney T, Cattan P, Vizzardelli C, Oliver R, Fraker C, Ricordi C, Pastori RL, Bach FH, Inverardi L: Heme oxygenase-1 induction in islet cells results in protection from apoptosis and improved in vivo function after transplantation.
Diabetes
50
:
1983
–1991,
2001
38.
Tobiasch E, Gunther L, Bach FH: Heme oxygenase-1 protects pancreatic beta cells from apoptosis caused by various stimuli.
J Investig Med
49
:
566
–571,
2001
39.
Ribeiro MM, Klein D, Pileggi A, Molano RD, Fraker C, Ricordi C, Inverardi L, Pastori RL: Heme oxygenase-1 fused to a TAT peptide transduces and protects pancreatic beta-cells.
Biochem Biophys Res Commun
305
:
876
–881,
2003
40.
Tu CF, Kuo CH, Juang JH: Effects of heme oxygenase-1 transgenic islets on transplantation.
Transplant Proc
37
:
3465
–3467
41.
Ke B, Shen XD, Melinek J, Gao F, Ritter T, Volk HD, Busuttil RW, Kupiec-Weglinski JW: Heme oxygenase-1 gene therapy: a novel immunomodulatory approach in liver allograft recipients.
Transplant Proc
33
:
581
–582,
2001
42.
Szanya V, Ermann J, Taylor C, Holness C, Fathman CG: The subpopulation of CD4+CD25+ splenocytes that delays adoptive transfer of diabetes expresses L-selectin and high levels of CCR7.
J Immunol
169
:
2461
–2465,
2002
43.
Goudy KS, Burkhardt BR, Wasserfall C, Song S, Campbell-Thompson ML, Brusko T, Powers MA, Clare-Salzler MJ, Sobel ES, Ellis TM, Flotte TR, Atkinson MA: Systemic overexpression of IL-10 induces CD4+CD25+cell populations in a dose-dependent fashion.
J Immunol
171
:
2210
–2278,
2003
44.
Brusko TM, Wasserfall CH, Agarwal A, Kapturczak MH, Atkinson MA: An integral role for heme oxygenase-1 and carbon monoxide in maintaining peripheral tolerance by regulatory T cells.
J Immunol
174
:
5181
–5186,
2005
45.
Steinman RM, Hawiger D, Nussenzweig MC: Tolerogenic dendritic cells.
Annu Rev Immunol
21
:
685
–711,
2003
46.
Machen J, Harnaha J, Lakomy R, Styche A, Trucco M, Giannoukakis N: Antisense oligonucleotides down-regulating costimulation confer diabetes-preventive properties to nonobese diabetic mouse dendritic cells.
J Immunol
173
:
4331
–4341,
2004
47.
Wang H, Lee SS, Gao W, Czismadia E, McDaid J, Ollinger R, Soares MP, Yamashita K, Bach FH: Donor treatment with carbon monoxide can yield islet allograft survival and tolerance.
Diabetes
54
:
1400
–1406,
2005
48.
Wang H, Lee SS, Dell'Agnello C, Tchipashvili V, D'Avilla J, Czismadia E, Chin BY, Bach FH: Bilirubin can induce tolerance to islet allografts.
Endocrinology
147
:
762
–768,
2006
49.
Zuckerbraun BS, Billiar TR, Otterbein SL, Kim PK, Liu F, Choi AM, Bach FH, Otterbein LE: Carbon monoxide protects against liver failure through nitric-oxide-induced heme oxygenase 1.
J Exp Med
198
:
1707
–1716,
2003
50.
Hegazi RAF, Rao KN, Mayle A, Sepulveda AR, Otterbein LE, Plevy SE: Carbon monoxide ameliorates chronic murine colitis through a heme oxygenase-1 dependent pathway.
J Exp Med
202
:
1703
–1713,
2005