Abstract
Cytotoxic T lymphocytes (CTLs) are thought to be major effectors involved in viral clearance during acute infections, including hepatitis B virus (HBV) infection. A persistent HBV infection is characterized by a lack of or a weak CTL response to HBV, which may be reflective of tolerance to HBV. Efficient induction of HBV-specific CTLs leads to the clearance of HBV in patients with a chronic HBV infection. Previously, we reported that α-galactosylceramide (α-GalCer), a specific natural killer T (NKT) cell agonist, enhanced the induction of HBV surface antigen (HBsAg)-specific CTLs. In the present study, we found that inhibition of indoleamine 2,3-dioxygenase (IDO) activity enhanced the induction of HBsAg-specific CTLs after immunization with HBsAg and α-GalCer. The administration of HBsAg and α-GalCer increased the production of interleukin-2 and interleukin-12b, which are crucial for the induction of HBsAg-specific CTLs. The production of these cytokines was more strongly enhanced in IDO knockout mice compared with wild-type mice. In addition, α-GalCer induced the production of IDO in CD11b+ cells, and these cells inhibited proliferation of HBsAg-specific CTLs. Our results lead to strategies for improving the induction of HBsAg-specific CTLs.
Keywords: 3-dioxygenase, cytotoxic T lymphocyte, hepatitis B virus, indoleamine 2, vaccination
Introduction
Cytotoxic T lymphocytes (CTLs) have been shown to play a critical role in viral clearance in many types of viral infection.1,2 Antigen-specific CTLs are generated during the T cytotoxic type 1 (Tc1) immune response, which is important in the immune response to hepatitis B virus (HBV). However, clearance of, or at least a continuous suppression of, HBV is not observed in all cases of chronic hepatitis, possibly, because of a weak CTL response to the HBV surface antigen (HBsAg). Therefore, devising an effective therapy for chronic hepatitis B requires elucidation of the mechanism underlying the weakness of the CTL response to HBsAg. Previous reports demonstrated that α-galactosylceramide (α-GalCer), a ligand for Vα14+ natural killer T (NKT) cells, strongly induced the generation of antigen-specific CTLs.3,4 Immunization with HBsAg plus α-GalCer enhanced the induction and proliferation of HBsAg-specific CTLs.5 Moreover, immunization with HBsAg plus α-GalCer enabled the induction of HBsAg-specific CTLs even in HBsAg transgenic (HBsAg-Tg) mice, which generate an extremely tolerant cellular and humoral immune response to HBsAg. Hence, we decided to examine the HBsAg-specific CTL response generated in HBsAg-Tg mice following immunization with α-GalCer and HBsAg. This experiment required direct stimulation by HBsAg in vitro, because a sufficient number of HBsAg-specific CTLs may be difficult to induce in HBsAg-Tg mice in vivo.
Indoleamine 2, 3-dioxygenase (IDO) is an enzyme that participates in the catabolism of the essential amino acid l-tryptophan to l-kynurenine and is a potent immunomodulator.6,7 This enzyme is expressed in epithelial cells, macrophages and dendritic cells and is induced by pro-inflammatory cytokines, including type I and type II interferons (IFN).8–10 The binding of cytotoxic T lymphocyte antigen-4 to CD80/CD86 on the surface of dendritic cells also stimulates transcription and activity of IDO.11,12 Our previous study demonstrated that IDO expression was enhanced by treatment of mice with α-GalCer.13 Although the co-administration of HBsAg and α-GalCer enhanced the induction of HBsAg-specific CTLs, the α-GalCer-induced IDO may inhibit the induction of the CTLs to some extent. Hence, in the absence of IDO, immunization with the HBsAg-α-GalCer combination may elicit a strong antigen-specific Tc1 response and numerous HBsAg-specific CTLs.
In this work, we examined the effect of IDO on the induction and proliferation of HBsAg-specific CTLs in mice immunized with α-GalCer and HBsAg. Our results indicate that elimination of IDO activity enhances the antigen-specific Tc1 response and increases the number of HBsAg-specific CTLs in mice immunized with HBsAg plus α-GalCer.
Materials and methods
Mice
Wild-type (WT) male B10.D2 (H-2d) mice (age 8–10 weeks; weight 25–30 g) were obtained from Japan SLC Inc. (Shizuoka, Japan). IDO knockout (KO) mice on a C57BL/6J background were obtained from Jackson Laboratory (Bar Harbor, ME) and backcrossed with B10.D2 (H-2d) mice. All procedures were conducted in accordance with the National Institutes of Health Guide for the Care and Use of Laboratory Animals, and with the guidelines for the care and use of animals established by the Animal Care and Use Committee of Gifu University, Japan.
Cell lines and reagents
The H-2d mastocytoma cell line P815 was obtained from the American Type Culture Collection (Rockville, MD). P815 cells expressing HBV-preS1, 2 and S regions (P815preS1) and the HBsAg-specific CD8+ CTL clone 6C2 were generously provided by Francis V. Chisari.14,15 The HBsAg peptide S28–39 was synthesized at KURABO (Osaka, Japan). α-GalCer was obtained from Funakoshi Co. Ltd. (Tokyo, Japan) and stored as a 200 μg/ml stock solution in vehicle (0.5% weight/volume polysorbate-20). 1-methyl-d-l-tryptophan (1-MT) was purchased from Sigma-Aldrich (St Louis, MO).
ELISPOT assay
The WT and IDO-KO mice were intraperitoneally inoculated with either HBsAg (10 μg/mouse) or HBsAg (10 μg/mouse) plus α-GalCer (2 μg/mouse). Single-cell suspensions were prepared from the whole spleen on day 7 after the inoculation. A total of 2.0 × 105 splenocytes/well were stimulated for 14–16 hr with 0, 0.1 or 1 μg/ml of HBsAg peptide (S28–39) in 96-well MultiScreen filter plates (Millipore, Billerica, MA) pre-coated with a rat anti-IFN-γ monoclonal antibody (mAb; R4-6A2; BD Biosciences, Franklin Lakes, NJ). The plates were washed and then incubated with a biotinylated polyclonal goat anti-IFN-γ antibody (R&D Systems, Minneapolis, MN) and, after that, with streptavidin–alkaline phosphatase. Spots were visualized by the addition of a 5-bromo-4-chloro-3-indolyl phosphatase solution (Sigma-Aldrich) and counted manually under a microscope (× 40 magnification). The number of cytokine-secreting cells was determined by a single blinded observer, and all data were generated by analysing three separate wells per sample.
Detection of HBsAg-specific CTLs by flow cytometry
Immunodominant HBsAg-specific CTLs in mice are known to be restricted by H-2Ld of MHC class I, and the shortest peptide that can show the maximal activity is HBsAg S28–39, sequence IPQSLDSWWTSL.14 The number of HBsAg-specific CTLs was assessed by flow cytometry, as described previously.16 Peptide-loaded recombinant soluble dimeric murine H-2Ld:Ig (mouse IgG1; BD PharMingen, San Diego, CA) was prepared by mixing soluble dimeric H-2Ld:Ig for 48 hr at 4° with a 160-fold molar excess of HBsAg S28–39. The peptide-loaded dimeric immunoglobulins were then incubated with CD8+ T cells isolated from either immunized or re-stimulated splenocytes.16 After incubation for 1 hr at 4°, the cells were stained with an FITC-conjugated anti-mouse CD8a antibody and a phycoerythrin (PE) -conjugated anti-mouse IgG1 antibody (BD PharMingen). The proportion of HBsAg-specific cells was measured by flow cytometry on a FACSCanto II instrument (Becton Dickinson Immunocytometry Systems, Mountain View, CA).
Cytotoxicity assay
The cytolytic activity of HBsAg-specific CTLs was assessed using a fluorescence-based dye, 5-(and 6-)carboxyfluorescein diacetate succinimidyl ester (CFSE) as described previously.17 Target cells (P815 or P815preS1) were labelled with CFSE as follows. The cells were suspended in PBS and diluted to 1 × 106/ml. For sensitive targets, 0.5 μl of CFSE stock solution (5 mm) was added to 1 ml of cell suspension and the mixture was incubated for 4 min at room temperature. For control targets, 0.5 μl of diluted CFSE solution (100 μm) was used for labelling, in a similar fashion. Labelled targets and various numbers of effector cells were added in a final volume of 200 μl to each well of 96-well round-bottom plates and incubated for 6 hr at 37°. After incubation, sensitive target cells were mixed with control target cells in one tube with PBS containing 1% fetal calf serum and 0.1% sodium azide. Mixed cells were washed once, suspended in 4% paraformaldehyde, and then stored at 4° in the dark before flow cytometric analysis, which was performed on a FACSCanto II instrument (Becton Dickinson Immunocytometry Systems). All samples were assayed in duplicate and the mean percentage of specific lysis was calculated as follows: % specific lysis = [(number of sensitive target cells in the control sample – number of sensitive target cells in the test sample)/number of sensitive target cells in the control sample] × 100. The control sample consisted of target cells incubated without added effector cells, whereas the test sample consisted of target cells incubated with added effector cells.
Flow cytometric analysis of splenocytes
Splenocytes were isolated from the immunized mice as described previously.18 Cell viability and cell number were assessed using a trypan blue exclusion assay. For flow cytometry, 2 × 105 splenocytes were stained with labelled antibodies using a standard protocol. The following antibodies were used: PE-Cy7-labelled anti-mouse CD8 mAb, clone 53-6.7 (eBioscience, San Diego, CA); PE-Cy7-labelled anti-mouse CD11b mAb (clone M1/70; eBioscience); VioBlue-labelled anti-mouse CD11c mAb (clone N418; Miltenyi Biotec GmbH, Bergisch Gladbach, Germany), FITC-labelled anti-mouse CD86 mAb (clone 24F; BD Biosciences); FITC-labelled anti-mouse Ly-6G mAb (clone RB6-8C5; BD Biosciences); PE-labelled anti-mouse CD80 mAb (clone BB1; BD Biosciences); PE-labelled anti-mouse Ly-6c mAb (clone AL-21; BD Biosciences); PE-labelled anti-mouse CD40 mAb (clone FKG45.5; Miltenyi Biotec); and PE-labelled anti-mouse Foxp3 mAb (clone FJK-16s; Biosciences). Samples were acquired on a flow cytometer and data analysis was performed using facsdiva software (BD Biosciences).
Real-time reverse transcription-PCR
Total RNA was isolated and transcribed into complementary DNA (cDNA) using an RNeasy Mini Kit (QIAGEN GmbH, Hilden, Germany) and a high-capacity cDNA transcription kit (Applied Biosystems, Foster City, CA). The resulting cDNA was used as a template for real-time PCR along with primer-probe sets for IDO, interleukin-2 (IL-2), IL-4, IL-6, IL-12b and 18S rRNA (TaqMan Gene Expression Assays; Applied Biosystems) and TaqMan universal PCR master mix (Applied Biosystems) according to the manufacturer's instructions (Applied Biosystems). The 18S rRNA was used as an internal control. Real-time PCR was carried out using a Light-Cycler 480 system (Roche Diagnostic Systems, Basel, Switzerland).
Isolation of CD8+ and CD11b+ cells
Splenocytes were sorted into CD8+ or CD11b+ cells using magnetic beads conjugated with an anti-CD8a or anti-CD11b antibody (Miltenyi Biotec GmbH) as described in previous reports.19,20 The magnetically labelled cells were purified using a quadroMACS system (Miltenyi Biotec GmbH).
HBsAg-specific CTL proliferation assays
Isolated CD11b+ and CD11b− cells were co-cultured with an HBsAg-specific CTL clone (6C2) in a CFSE-based proliferation assay.21 The 6C2 cells were labelled with CFSE as previously described.21 The CFSE-labelled cells were co-cultured with the purified CD11b+ or CD11b− cells in 96-well plates at the cell-count ratios of 1 : 2 and 1 : 5 for the 6C2/CD11b+ and the 6C2/CD11b− combinations, respectively, for 5 days at 37° in a 5% CO2 atmosphere. To induce proliferation, the 6C2 cells were stimulated with irradiated P815PreS1 cells.
Statistics
Values were calculated as the mean ± SEM. Differences between experimental and control groups were analysed using the Kruskal–Wallis test followed by Scheffe's F-test. Significance was assumed at P < 0.05.
Results
Induction of the Tc1 immune response and HBsAg-specific CD8+ T cells in WT and IDO-KO mice
Our previous study demonstrated that α-GalCer-activated invariant NKT cells strongly increased the number of HBsAg-specific CTLs after immunization with HBsAg in vivo.5 Therefore, in the present study, we used α-GalCer as an adjuvant to enhance the HBsAg-specific immune response. To determine whether IDO contributes to the development of the HBsAg-specific immune response, WT and IDO-KO mice were immunized with HBsAg and α-GalCer intraperitoneally. As shown in Fig. 1(a), IDO-KO mice mounted strong Tc1 cellular responses against HBsAg, as indicated by the significant expansion of IFN-γ-producing cells in response to ex vivo re-stimulation with HBsAg peptide 28–39. Next, we examined CTL frequencies in the spleen using recombinant soluble fusion protein H-2Ld:Ig, which can be used to stain CD8+ T cells that recognize the H-2Ld-bound HBsAg peptide S28–39 (Fig. 1b,c). α-GalCer enhanced the numbers of WT as well as IDO-KO HBsAg-specific CD8+ T cells. However, the induction of HBsAg-specific CD8+ T cells was enhanced more strongly for the IDO-KO subset. Moreover, we examined the HBsAg-specific lysis caused by CD8+ T cells from immunized WT and IDO-KO mice ex vivo (Fig. 1d). After immunization of mice with the HBsAg-α-GalCer combination, the HBsAg-specific lysis in CD8+ T cells isolated from the IDO-KO mice was significantly enhanced compared with that induced by the WT cells.
Comparison of DC functions between WT and IDO-KO mice
Since α-GalCer can induce proliferation of the antigen-specific T cells via the activation of dendritic cells (CD11c+ cells), we analysed dendritic cells from WT and IDO-KO mice immunized with HBsAg plus α-GalCer.22 α-GalCer can up-regulate the expression of co-stimulatory molecules such as CD40, CD80 and CD86 in CD11c+ cells (Fig. 2). Although the expression of these molecules on dendritic cells was enhanced bythe immunization with the HBsAg–α-GalCer combination, there was no difference between WT and IDO-KO mice.
Regulatory T cells and myeloid derived suppressor cells are stimulated by immunization with HBsAg and α-GalCer
Previous reports demonstrated that IDO was involved in the induction of regulatory T (Treg) cells.23,24 We therefore examined the proliferation of Treg cells in WT and IDO-KO mice after inoculation with HBsAg and α-GalCer. Flow cytometry analysis revealed that the percentage of Treg cells in the spleen of WT and IDO-KO mice increased equally on day 7 after the immunization (Fig. 3a). The number of Treg cells did not decrease in IDO-KO mice after the HBsAg plus α-GalCer immunization in the present model. Myeloid derived suppressor cells (MDSCs) are identified by the expression of IDO, and were counted using flow cytometry. MDSCs are broadly defined as CD11b+ Ly6G+ mononuclear cells. The administration of HBsAg with α-GalCer expanded the MDSC population in WT and IDO-KO mice (Fig. 3b). This increase in MDSC frequency among spleen cells was observed 7 hr after the immunization, and was similar in both WT and IDO-KO mice.
IDO and cytokine production by WT and IDO-KO splenocytes in response to the intraperitoneal injection of HBsAg plus α-GalCer
Next, we examined whether the expression of IDO in the spleen is enhanced after the intraperitoneal administration of the HBsAg–α-GalCer combination. This immunization promptly induced IDO expression in the spleen (Fig. 4a). Previous studies demonstrated that several cytokines play critical roles in the activation and proliferation of antigen-specific CTLs.25,26 Accordingly, we measured mRNA levels for IL-2, IL-4, IL-6, IL-12b and IFN-γ in whole spleens from WT and IDO-KO mice after the co-administration of HBsAg and α-GalCer (Fig. 4b). The expression of IL-2, IL-4, IL-6 and IL-12b in the spleen of WT and IDO-KO mice was enhanced at 7 hr after the co-administration. In IDO-KO mice, however, compared with WT mice, the expression of IL-2 was significantly increased at 24 and 72 hr after the co-administration. Interleukin-12b expression in IDO-KO mice was also increased at 24 hr after the co-immunization with HBsAg and α-GalCer. These data indicate that after the co-administration, the enhancement of IL-2 and IL-12b expression persisted for longer in the spleen of the IDO-KO mice.
Induction and function of IDO in CD11b+ cells after the immunization of mice with HBsAg and α-GalCer
As shown in Fig. 4(a), the expression of IDO mRNA was strongly enhanced at 7 hr after the administration of HBsAg and α-GalCer. Moreover, the number of CD11b+ Ly6G+ cells (MDSCs) in the spleen also increased at 7 hr after the immunization (Fig. 3b). For this reason, we examined the expression of IDO in CD11b+ cells and CD11b− cells after the immunization of mice with HBsAg and α-GalCer. As a result of the immunization, the expression of IDO mRNA in CD11b+ cells significantly increased compared with that in CD11b− cells (Fig. 5a). IDO was predominantly induced in CD11b+ cells from the spleen. Next, we examined the function of CD11b+ cells in HBsAg-specific T-cell proliferation in WT and IDO-KO mice. 6C2 cells are HBsAg-specific CTL clones. We labelled these CTLs with CFSE and stimulated them using irradiated P815PreS1 cells either in the presence or in the absence of CD11b+ cells from WT or IDO-KO mice (Fig. 5c). 6C2 cell proliferation was evaluated by CFSE dilution. For WT mice, the presence of CD11b+ cells inhibited the proliferation of the HBsAg-specific CTLs 6C2 in response to the target cells, after immunization of mice with HBsAg and α-GalCer. In contrast, enhanced cell proliferation was observed when IDO-KO cells were used under similar conditions. In addition, the expression of IL-12b in CD11b+ cells from immunized WT and IDO-KO mice increased compared with that in the CD11b– subset (Fig. 5b). The enhanced IL-12b production by IDO-KO CD11b+ cells may have contributed to the 6C2 cell proliferation in response to the P815PreS1 cells.
Effect of 1-MT on the induction of the HBsAg-specific immune response
1-Methyl-d-tryptophan is a potent inhibitor of IDO, and hence we used this agent to verify our finding. The WT mice were given 1-MT orally at either 0 or 5 mg/ml in drinking water 2 days before the immunization to 7 days after the immunization. ELISPOT analysis revealed that the HBsAg peptide S28–39-specific Tc1 response in WT mice treated with 1-MT was significantly stronger compared with the untreated mice (Fig. 6a). The induction of HBsAg-specific CD8+ T cells was enhanced by treatment with 1-MT (Fig. 6b,c). These data indicate that the inhibition of IDO activity leads to the enhancement of an HBsAg-specific Tc1 response after immunization with HBsAg and α-GalCer.
Discussion
In the present study, we demonstrated that inhibition of IDO activity enhances the HBsAg-specific Tc1 response and induction of CTLs after immunization with HBsAg and α-GalCer. The expression of IL-2 and IL-12b, which are critical cytokines for inducing the antigen-specific Tc1 response, increases in IDO knockout mice after immunization with HBsAg and α-GalCer. Moreover, CD11b+ splenocytes up-regulate their expression of IDO and suppress the proliferation of HBsAg-specific CTLs after immunization of mice with HBsAg and α-GalCer treatment. Hence, the α-GalCer-induced increase of IDO expression reduced the antigen-specific Tc1 immunity. On the other hand, inhibition of IDO expression after the HBsAg plus α-GalCer immunization strongly enhanced the HBsAg-specific cellular immune response.
Previously, we demonstrated that during immunization with HBsAg, α-GalCer works as an adjuvant and enhances the induction of an HBsAg-specific CTL response in vivo.5 It appears that IL-2 and CD40–CD40 ligand interactions are involved in the enhancement of CTL induction caused by α-GalCer through NKT cell activation. On the other hand, α-GalCer enhances the activity of IDO in mice.13 IDO catalyses the conversion of the essential amino acid l-tryptophan to l-kynurenine and has been identified as an enzyme that has powerful immunomodulatory effects. Furthermore, metabolites of the l-kynurenine pathway have been shown to act as immunosuppressive molecules in the tissue microenvironment.27,28 IDO and the l-tryptophan pathway play critical roles in the generation of immune tolerance against foreign antigens in tissue microenvironments. In the present study, IDO expression in splenocytes also increased after HBsAg and α-GalCer administration (Fig. 5a). As shown in Fig. 1(a), the intraperitoneal injection of HBsAg and α-GalCer increased the HBsAg-specific Tc1 immune response, and this Tc1 response was enhanced more strongly in IDO-KO mice compared with WT mice. Moreover, the number of CD8+ T cells that can specifically recognize HBsAg and cause HBsAg-mediated lysis also increased in IDO-KO mice ex vivo (Fig. 1b–d). These results suggest that the absence of IDO activity significantly enhances the HBsAg-specific Tc1 immune response after the immunization with HBsAg and α-GalCer. 1-MT is a competitive inhibitor of the IDO activity. As shown in Fig. 6, the administration of HBsAg, α-GalCer and 1-MT also enhanced both the HBsAg-specific Tc1 response and the induction of HBsAg-specific CD8+ T cells (Fig. 6a–c). However, the HBsAg-specific Tc1 response in the mice treated with HBsAg, α-GalCer and 1-MT was weak compared with that in the IDO-KO mice treated with HBsAg and α-GalCer. A previous report indicated that the l-kynurenine level in IDO-KO mice was lower than that in 1-MT-treated mice during Toxoplasma gondii infection in vivo.29 Our previous study also indicated a difference in proliferation of splenocytes and viral replication between IDO-KO mice and 1-MT-treated mice during LP-BM5 murine leukaemia virus infection.9 We assumed that complete local depletion of L-tryptophan is critical for the immune system, and the oral administration of 1-MT may fail to fully suppress local IDO activity. Several reports have demonstrated that the enhancement of the immune response by α-GalCer involves an up-regulation of co-stimulatory molecules such as CD80, CD86 and CD40.22,30 In the present work, although α-GalCer enhanced the expression of CD40 and CD86 in CD11c+ cells, this effect was similar in WT and IDO-KO mice (Fig. 2). Real-time PCR analysis revealed that the expression of IL-2 and IL-12b was elevated at 24 and 72 hr after the HBsAg plus α-GalCer immunization in IDO-KO mice compared with that in WT mice (Fig. 4b). The expression of IL-2, IL-4, IL-6 and IL-12b increased at 7 hr after administration, and promptly decreased afterwards in WT mice. On the other hand, in IDO-KO mice, the enhancement of IL-2 and IL-12b expression was maintained between 24 and 72 hr after the immunization with HBsAg and α-GalCer. The persistence of the elevated expression of IL-2 and IL-12b in IDO-KO mice may play a role in the enhancement of the HBsAg-specific Tc1 immune response induced by HBsAg and α-GalCer in combination.
IDO is strongly activated by IFN-γ or pro-inflammatory cytokines in macrophages/monocytes. As shown in Fig. 5(a), among splenocytes, the immunization of mice with HBsAg and α-GalCer induced IDO activity almost exclusively in CD11b+ cells. No enhancement of IDO expression was observed in CD11b– cells. In addition, CD11b+ cells from HBsAg-plus-α-GalCer-immunized WT mice reduced the proliferation of HBsAg-specific CTLs when the latter were stimulated with the specific antigen (Fig. 5c). In contrast, CD11b+ cells from HBsAg-plus-α-GalCer-immunized IDO-KO mice facilitated the proliferation of HBsAg-specific CTLs. These results indicate that IDO-deficient CD11b+ cells treated with α-GalCer have a strong ability to enhance proliferation of HBsAg-specific CTLs. The expression of IL-12b in CD11b+ cells was increased at 7 hr after the immunization with HBsAg plus α-GalCer (Fig. 5b). Moreover, the CD11b+ fraction included the CD11b+ CD11c+ cells, and the expression of co-stimulatory molecules on CD11c+ cells increased after the immunization (Fig. 2). Hence, the CD11b+ cells from the IDO-KO mice immunized with HBsAg and α-GalCer may enhance the proliferation of HBsAg-specific CTLs via an increase in IL-12b production and co-stimulatory molecule expression. The intraperitoneal administration of HBsAg and α-GalCer increased the proportion of CD11b+ Ly6G+ cells in the spleen (Fig. 3b). CD11b+ Ly6G+ cells are known as MDSCs and induce the expression of IDO, which inhibits the immune response.31 Hence, the immunization with HBsAg and α-GalCer increases the proportion of CD11b+ Ly6G+ cells in the spleen as well as the number of IDO-producing cells and thereby inhibits the induction of an HBsAg-specific immune response. Although the immunization with HBsAg and α-GalCer also increased the proportion of CD11b+ Ly6G+ cells in IDO-KO mice, these cells cannot up-regulate IDO and inhibit the HBsAg-specific immune response. Several reports demonstrated that IDO stimulates Treg cells and suppresses the immune response.23,24 On the other hand, there was no difference in the percentage of Treg cells between the non-treated group and the 1-MT-treated group in a rheumatoid arthritis mouse model.32 Our previous report also demonstrated that the expression of FoxP3 mRNA in WT and IDO-KO mice was up-regulated after LP-BM5 murine leukaemia virus infection and there was no difference in FoxP3 mRNA expression.9 The inoculation with HBsAg and α-GalCer injection increased the proportion of CD4+ FoxP3+ T cells in the spleen (Fig. 3a), but there was no difference in Treg cell number between WT mice and IDO-KO mice. Therefore, in this study, Treg cells do not appear to be involved in the HBsAg-specific immune response following immunization with HBsAg plus α-GalCer.
A recent study evaluated the effect of IDO on the humoral immune response, and they demonstrated that inhibition of IDO by the administration with 1-MT at the time of vaccination decreased the serum anti-HBs antibody level after HBsAg vaccination.33 Another report showed that 1-MT inhibit the B cells' ability to differentiate into autoantibody-secreting cells and improve autoimmune arthritis.34 These studies indicated that 1-MT inhibits the humoral immune response. The enhancement of HBs-antibody induction is critical to prevent HBV infection. On the other hand, the induction of a cellular immune response to HBV antigen is pivotal to the complete elimination of HBV during chronic infection.35 Enhancement of the induction of HBsAg-specific CTLs may be helpful in developing strategies to clear HBV in patients with chronic hepatitis.
We can conclude that the inhibition of IDO activity significantly enhanced the induction of HBsAg-specific CTLs following immunization with HBsAg and α-GalCer. The addition of α-GalCer enhances IDO activity, which inhibits the activation and proliferation of antigen-specific CTLs. Hence, abrogation of the α-GalCer-mediated enhancement of IDO activity makes the HBsAg-specific Tc1 response much more powerful. These results lead to strategies for improving the induction of HBsAg-specific CTLs.
Acknowledgments
This work was supported by the Grant-in-Aid for Exploratory Research (24659361) from the Ministry of Education, Culture, Sports, Science and Technology of Japan.
Disclosures
The authors have no financial or commercial conflict of interest.
References
- 1.Guidotti LG. The role of cytotoxic T cells and cytokines in the control of hepatitis B virus infection. Vaccine. 2002;20(Suppl 4):A80–2. doi: 10.1016/s0264-410x(02)00392-4. [DOI] [PubMed] [Google Scholar]
- 2.Shan L, Deng K, Shroff NS, et al. Stimulation of HIV-1-specific cytolytic T lymphocytes facilitates elimination of latent viral reservoir after virus reactivation. Immunity. 2012;36:491–501. doi: 10.1016/j.immuni.2012.01.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Ito H, Seishima M. Regulation of the induction and function of cytotoxic T lymphocytes by natural killer T cell. J Biomed Biotechnol. 2010;2010:641757. doi: 10.1155/2010/641757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Kim D, Hung CF, Wu TC, Park YM. DNA vaccine with α-galactosylceramide at prime phase enhances anti-tumor immunity after boosting with antigen-expressing dendritic cells. Vaccine. 2010;28:7297–305. doi: 10.1016/j.vaccine.2010.08.079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Ito H, Ando K, Ishikawa T, et al. Role of Vα14+ NKT cells in the development of hepatitis B virus-specific CTL: activation of Vα14+ NKT cells promotes the breakage of CTL tolerance. Int Immunol. 2008;20:869–79. doi: 10.1093/intimm/dxn046. [DOI] [PubMed] [Google Scholar]
- 6.Munn DH, Shafizadeh E, Attwood JT, Bondarev I, Pashine A, Mellor AL. Inhibition of T cell proliferation by macrophage tryptophan catabolism. J Exp Med. 1999;189:1363–72. doi: 10.1084/jem.189.9.1363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Mellor AL, Munn DH. Tryptophan catabolism and T-cell tolerance: immunosuppression by starvation? Immunol Today. 1999;20:469–73. doi: 10.1016/s0167-5699(99)01520-0. [DOI] [PubMed] [Google Scholar]
- 8.Fujigaki S, Saito K, Sekikawa K, et al. Lipopolysaccharide induction of indoleamine 2,3-dioxygenase is mediated dominantly by an IFN-γ-independent mechanism. Eur J Immunol. 2001;31:2313–8. doi: 10.1002/1521-4141(200108)31:8<2313::aid-immu2313>3.0.co;2-s. [DOI] [PubMed] [Google Scholar]
- 9.Hoshi M, Saito K, Hara A, et al. The absence of IDO upregulates type I IFN production, resulting in suppression of viral replication in the retrovirus-infected mouse. J Immunol. 2010;185:3305–12. doi: 10.4049/jimmunol.0901150. [DOI] [PubMed] [Google Scholar]
- 10.Hoshi M, Matsumoto K, Ito H, et al. l-Tryptophan–kynurenine pathway metabolites regulate type I IFNs of acute viral myocarditis in mice. J Immunol. 2012;188:3980–7. doi: 10.4049/jimmunol.1100997. [DOI] [PubMed] [Google Scholar]
- 11.Fallarino F, Grohmann U, Hwang KW, et al. Modulation of tryptophan catabolism by regulatory T cells. Nat Immunol. 2003;4:1206–12. doi: 10.1038/ni1003. [DOI] [PubMed] [Google Scholar]
- 12.Grohmann U, Fallarino F, Bianchi R, et al. A defect in tryptophan catabolism impairs tolerance in nonobese diabetic mice. J Exp Med. 2003;198:153–60. doi: 10.1084/jem.20030633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Ito H, Hoshi M, Ohtaki H, et al. Ability of IDO to attenuate liver injury in α-galactosylceramide-induced hepatitis model. J Immunol. 2010;185:4554–60. doi: 10.4049/jimmunol.0904173. [DOI] [PubMed] [Google Scholar]
- 14.Ando K, Guidotti LG, Wirth S, et al. Class I-restricted cytotoxic T lymphocytes are directly cytopathic for their target cells in vivo. J Immunol. 1994;152:3245–53. [PubMed] [Google Scholar]
- 15.Ando K, Guidotti LG, Cerny A, Ishikawa T, Chisari FV. CTL access to tissue antigen is restricted in vivo. J Immunol. 1994;153:482–8. [PubMed] [Google Scholar]
- 16.Kasahara S, Ando K, Saito K, et al. Lack of tumor necrosis factor α induces impaired proliferation of hepatitis B virus-specific cytotoxic T lymphocytes. J Virol. 2003;77:2469–76. doi: 10.1128/JVI.77.4.2469-2476.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Nakagawa Y, Watari E, Shimizu M, Takahashi H. One-step simple assay to determine antigen-specific cytotoxic activities by single-color flow cytometry. Biomed Res. 2011;32:159–66. doi: 10.2220/biomedres.32.159. [DOI] [PubMed] [Google Scholar]
- 18.Ito H, Ando K, Nakayama T, et al. Role of Vα14 NKT cells in the development of impaired liver regeneration in vivo. Hepatology. 2003;38:1116–24. doi: 10.1053/jhep.2003.50471. [DOI] [PubMed] [Google Scholar]
- 19.Ito H, Ando K, Ishikawa T, et al. Role of TNF-α produced by nonantigen-specific cells in a fulminant hepatitis mouse model. J Immunol. 2009;182:391–7. doi: 10.4049/jimmunol.182.1.391. [DOI] [PubMed] [Google Scholar]
- 20.Ohtaki H, Ito H, Hoshi M, et al. High susceptibility to lipopolysaccharide-induced lethal shock in encephalomyocarditis virus-infected mice. Sci Rep. 2012;2:367. doi: 10.1038/srep00367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Mandapathil M, Lang S, Gorelik E, Whiteside TL. Isolation of functional human regulatory T cells (Treg) from the peripheral blood based on the CD39 expression. J Immunol Methods. 2009;346:55–63. doi: 10.1016/j.jim.2009.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Joyee AG, Uzonna J, Yang X. Invariant NKT cells preferentially modulate the function of CD8α+ dendritic cell subset in inducing type 1 immunity against infection. J Immunol. 2010;184:2095–106. doi: 10.4049/jimmunol.0901348. [DOI] [PubMed] [Google Scholar]
- 23.Yan Y, Zhang GX, Gran B, et al. IDO upregulates regulatory T cells via tryptophan catabolite and suppresses encephalitogenic T cell responses in experimental autoimmune encephalomyelitis. J Immunol. 2010;185:5953–61. doi: 10.4049/jimmunol.1001628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Matteoli G, Mazzini E, Iliev ID, et al. Gut CD103+ dendritic cells express indoleamine 2,3-dioxygenase which influences T regulatory/T effector cell balance and oral tolerance induction. Gut. 2010;59:595–604. doi: 10.1136/gut.2009.185108. [DOI] [PubMed] [Google Scholar]
- 25.Bachmann MF, Schorle H, Kuhn R, et al. Antiviral immune responses in mice deficient for both interleukin-2 and interleukin-4. J Virol. 1995;69:4842–6. doi: 10.1128/jvi.69.8.4842-4846.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Widmer MB, Grabstein KH. Regulation of cytolytic T-lymphocyte generation by B-cell stimulatory factor. Nature. 1987;326:795–8. doi: 10.1038/326795a0. [DOI] [PubMed] [Google Scholar]
- 27.Terness P, Bauer TM, Rose L, et al. Inhibition of allogeneic T cell proliferation by indoleamine 2,3-dioxygenase-expressing dendritic cells: mediation of suppression by tryptophan metabolites. J Exp Med. 2002;196:447–57. doi: 10.1084/jem.20020052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Fallarino F, Grohmann U, Puccetti P. Indoleamine 2,3-dioxygenase: from catalyst to signaling function. Eur J Immunol. 2012;42:1932–7. doi: 10.1002/eji.201242572. [DOI] [PubMed] [Google Scholar]
- 29.Murakami Y, Hoshi M, Hara A, et al. Inhibition of increased indoleamine 2,3-dioxygenase activity attenuates Toxoplasma gondii replication in the lung during acute infection. Cytokine. 2012;59:245–51. doi: 10.1016/j.cyto.2012.04.022. [DOI] [PubMed] [Google Scholar]
- 30.Taraban VY, Martin S, Attfield KE, et al. Invariant NKT cells promote CD8+ cytotoxic T cell responses by inducing CD70 expression on dendritic cells. J Immunol. 2008;180:4615–20. doi: 10.4049/jimmunol.180.7.4615. [DOI] [PubMed] [Google Scholar]
- 31.Yu J, Du W, Yan F, et al. Myeloid-derived suppressor cells suppress antitumor immune responses through IDO expression and correlate with lymph node metastasis in patients with breast cancer. J Immunol. 2013;190:3783–97. doi: 10.4049/jimmunol.1201449. [DOI] [PubMed] [Google Scholar]
- 32.Scott GN, DuHadaway J, Pigott E, et al. The immunoregulatory enzyme IDO paradoxically drives B cell-mediated autoimmunity. J Immunol. 2009;182:7509–17. doi: 10.4049/jimmunol.0804328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Eleftheriadis T, Sparopoulou T, Antoniadi G, Liakopoulos V, Stefanidis I, Galaktidou G. Suppression of humoral immune response to hepatitis B surface antigen vaccine in BALB/c mice by 1-methyl-tryptophan co-administration. Daru. 2011;19:236–9. [PMC free article] [PubMed] [Google Scholar]
- 34.Pigott E, Mandik-Nayak L. Addition of an indoleamine 2,3,-dioxygenase inhibitor to B cell-depletion therapy blocks autoreactive B cell activation and recurrence of arthritis in K/BxN mice. Arthritis Rheum. 2012;64:2169–78. doi: 10.1002/art.34406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Guidotti LG, Ishikawa T, Hobbs MV, Matzke B, Schreiber R, Chisari FV. Intracellular inactivation of the hepatitis B virus by cytotoxic T lymphocytes. Immunity. 1996;4:25–36. doi: 10.1016/s1074-7613(00)80295-2. [DOI] [PubMed] [Google Scholar]