We previously showed that regulatory T cells (Tregs) from T cell–specific Socs1-deficient mice (Socs1fl/flLck-Cre+ mice) easily convert into Th1- or Th17-like cells (ex-Tregs), which lose Foxp3 expression and suppressive functions in vivo. Because Tregs in Socs1fl/flLck-Cre+ mice are constantly exposed to a large amount of inflammatory cytokines produced by non-Tregs in vivo, in this study we analyzed Treg-specific Socs1-deficient mice (Socs1fl/flFoxp3YFP-Cre mice). These mice developed dermatitis, splenomegaly, and lymphadenopathy that were much milder than those in Socs1fl/flLck-Cre+ mice. A fate mapping study revealed that Socs1 deficiency accelerated the conversion of Tregs to Foxp3IFN-γ+ ex-Tregs in the tumor microenvironment and suppressed tumor growth. When transferred into Rag2−/− mice, Tregs from Socs1fl/flLck-Cre+ mice easily lost Foxp3 expression, whereas those from Socs1fl/flFoxp3YFP-Cre mice maintained Foxp3 expression. Although Tregs from Socs1fl/flLck-Cre+ mice produced IFN-γ after a 3-d culture in response to anti-CD3/CD28 Ab stimulation in vitro, Tregs from Socs1fl/flFoxp3YFP-Cre mice did not. This finding suggested that the inflammatory conditions in Socs1fl/flLck-Cre+ mice modified the born nature of Socs1-deficient Tregs. To investigate this mechanism, Tregs from Socs1fl/flFoxp3YFP-Cre mice were cultured with APCs from Socs1fl/flLck-Cre+ mice. These APCs facilitated STAT4 phosphorylation, IFN-γ production, and loss of Foxp3 expression in Tregs from Socs1fl/flFoxp3YFP-Cre mice in an IL-12–dependent manner. The results indicate that Socs1-deficient Tregs tend to convert into ex-Tregs under the inflammatory conditions in which APCs are highly activated, and that SOCS1 could be a useful target for enhancement of anti-tumor immunity.

It has been shown that the terminally differentiated state of natural regulatory T cells (nTregs) is not defined completely by Foxp3 expression, because Foxp3+ Tregs are a heterogeneous population consisting of a committed Treg lineage and an uncommitted subpopulation with developmental plasticity (1). This uncommitted subset of Tregs has been shown to lose Foxp3 expression and to rapidly convert to effector/helper T cells by transfer into a lymphopenic host (1) or under inflammatory conditions (ex-Tregs) (2). Such ex-Tregs (2) have the potential to trigger autoimmunity because they show effector-memory phenotypes with pathogenic cytokine production, and their TCRs are hypothesized to be autoreactive to self-antigens (35). For example, Komatsu et al. (6) reported that Th17 cells that developed from Foxp3+ T cells play a crucial role in the pathogenesis of autoimmune arthritis. In addition, an uncommitted subset of Tregs lacks hypomethylation of the Treg-specific demethylated region (TSDR), which is responsible for stable Foxp3 expression (79). Even in committed Tregs, Foxp3 has been reported to be reversibly downregulated in a certain condition without losing Treg characteristics (9).

Suppressor of cytokine signaling 1 (SOCS1) inhibits the JAK–STAT pathway (10), and uncontrolled IFN-γ signaling results from a deficiency of SOCS1 (11, 12). SOCS1 is highly expressed in Tregs (13) and plays a role in maintaining suppressive functions of nTregs (13, 14). Recent research has shown that SOCS1 is a direct target of the ubiquitin-conjugating enzyme Ubc13, which is involved in the TCR-stimulated activation of the IKK–NF-κB pathway, and this targeting of SOCS1 prevents their conversion into effector-like T cells (15). We have shown that nTregs from T cell–specific Socs1-deficient mice (Socs1fl/flLck-Cre+ mice) easily convert into ex-Tregs, which lose suppressive functions and Foxp3 expression in vivo with DNA hypermethylation in the conserved noncoding DNA sequence (CNS) 2 (CNS2) of the Foxp3 promoter/enhancer (16). Tregs in Socs1fl/flLck-Cre+ mice are constantly exposed to a large amount of inflammatory cytokines from Socs1-deficient non-Tregs, and the Foxp3 levels in Socs1−/− Tregs with hypomethylated TSDR are restored by deletion of the Ifng gene (16). Thus, unstable expression of Foxp3 in Socs1-deficient Tregs may be dependent on an inflammatory environment.

To evaluate the role of SOCS1 in the stability and suppressive function of Tregs, we analyzed Treg-specific Socs1-deficient mice (Socs1fl/flFoxp3YFP-Cre mice). In this article, we report that the conversion of Tregs to Foxp3IFN-γ+ ex-Tregs in the tumor microenvironment was accelerated by Socs1 deficiency in Tregs, resulting in inhibition of tumor growth. Although Tregs from Socs1fl/flFoxp3YFP-Cre mice maintained suppressive functions and Foxp3 expression when they were transferred into Rag2−/− mice, Tregs from Socs1fl/flFoxp3YFP-Cre mice produced more IFN-γ and showed stronger STAT4 phosphorylation compared with wild-type (WT) Tregs when they were cultured with IL-12–producing APCs from Socs1fl/flLck-Cre+ mice. These results indicate that SOCS1 is one of the essential molecules that maintain Treg stability, particularly under the inflammatory conditions in which APCs are highly activated.

Treg-specific Socs1 conditional knockout mice (Socs1fl/flFoxp3YFP-Cre mice), Socs1+/+Foxp3YFP-Cre mice (littermate control, WT mice), and T cell–specific Socs1 conditional knockout mice (Socs1fl/flLck-Cre+ mice), sex and age matched, were used. Socs1fl/flFoxp3YFP-Cre mice were generated by crossing Socs1fl/fl mice with Foxp3YFP-Cre knock-in mice, which were kindly provided by Dr. Rudensky (17). Socs1fl/flLck-Cre+ mice were reported previously (13, 14) and were crossed with Foxp3-IRES-GFP knock-in mice (18) to mark Foxp3+ cells (16). To fate map Foxp3 in mice, Socs1+/+Foxp3YFP-Cre (WT) mice or Socs1fl/flFoxp3YFP-Cre mice were crossed with R26tdRFP mice, which are knock-in mice carrying a tandem-dimer red fluorescent protein (tdRFP) within the Rosa26 locus (Socs1+/+Foxp3YFP-Cre×R26tdRFP mice or Socs1fl/flFoxp3YFP-Cre×R26tdRFP mice) (19).

Mice were kept under conventional conditions at Keio University (Tokyo, Japan). All experiments involving these mice were approved by and performed according to the guidelines of the Animal Ethics Committee of Keio University.

Total RNA was prepared using the nucleospin RNA XS (MACHEREY-NAGEL). RNA was reverse transcribed to cDNA with random primers (Applied Biosystems) and a high-capacity cDNA reverse transcription kit according to the manufacturer’s protocol (Applied Biosystems). To determine the cellular expression level of each gene, quantitative real-time PCR analysis was performed using a C1000 Thermal Cycler (Bio-Rad). The PCR mixture consisted of 5 μl KAPA SYBR FAST qPCR Kits (Kapa Biosystems), 15 pmol of forward and reverse primers (Socs1, forward: 5′-CGCCAACGGAACTGCTTCTTC-3′; and reverse: 5′-TCAGGTAGTCACGGAGTACC-3′), and cDNA samples in a total volume of 10 μl. Relative RNA abundance was determined based on control–hypoxanthine phosphoribosyltransferase abundance.

Cell surface staining and flow cytometric analysis of CD3, CD4, CD25, CD62L, and CD44 (all Abs from eBioscience) expression were performed as described previously (20). For the isolation of Tregs, CD4+ T cells were positively selected with MACS (Miltenyi), and CD3+CD4+CD25+Foxp3YFP-Cre cells or CD3+CD4+CD25+Foxp3GFP cells were further purified using a FACSAria cell sorter (Becton Dickinson). For isolation of APCs, CD3+ T cells were depleted from splenocytes, using the FACSAria cell sorter (Becton Dickinson). The purity of the sorted populations was invariable (>99%). Intracellular staining of Foxp3, IFN-γ, and T-bet (all Abs from eBioscience) was performed following fixation and permeabilization according to the manufacturer’s instructions. To measure T cell cytokine production, cells were stimulated with PMA (50 ng/ml) and ionomycin (250 ng/ml) in the presence of GolgiPlug (BD Biosciences) for 4 h at 37°C before staining.

B16/F10 melanoma cells (2 × 105 cells) were transplanted into Socs1+/+Foxp3YFP-Cre or Socs1fl/flFoxp3YFP-Cre mice. To observe the conversion of Tregs to Foxp3IFN-γ+ ex-Tregs using a fate mapping system, 5 × 105 Tregs (YFP+RFP+ cells) from Socs1+/+Foxp3YFP-Cre×R26tdRFP or Socs1fl/flFoxp3YFP-Cre×R26tdRFP mice were transferred into WT mice transplanted with 1 × 106 B16/F10 melanoma cells after 2 Gy irradiation. CD3+CD4+YFP+RFP+ cells were isolated as Tregs, using a FACSAria cell sorter. Fifteen days after cell transfer, the mice were sacrificed for experiments, and cells (YFP+RFP+ or YFPRFP+ cells) from the tumors and draining lymph nodes (LNs) were analyzed. Tregs (YFP+RFP+ cells) or ex-Tregs (YFPRFP+ cells) were sorted using the FACSAria cell sorter, and Ifng mRNA levels were quantified using RT-PCR analysis.

Sorted CD4+CD25CD62L+CD44 naive T cells (4 × 105 cells) from WT mice were injected i.v. into Rag2−/− mice in combination with 2 × 105 CD3+CD4+CD25+Foxp3YFP-Cre cells from Socs1+/+Foxp3YFP-Cre or Socs1fl/flFoxp3YFP-Cre mice (8 wk old and sex matched). Mice were observed daily and weighed weekly. Four weeks after cell transfer, the mice were sacrificed for experiments.

Sorted CD3+CD4+CD25+Foxp3YFP-Cre cells (2 × 105 cells) from Socs1+/+Foxp3YFP-Cre or Socs1fl/flFoxp3YFP-Cre mice (8 wk old and sex matched) were injected i.v. into Rag2−/− mice. Mice were observed daily and weighed weekly. Eight weeks after cell transfer, the mice were sacrificed for experiments.

Tregs (CD3+CD4+CD25+Foxp3YFP-Cre cells or CD3+CD4+CD25+Foxp3GFP cells) from WT, Socs1fl/flLck-Cre+, Socs1fl/flFoxp3YFP-Cre (<12 wk), and Socs1fl/flFoxp3YFP-Cre (>12 wk) mice (age and sex matched), at a concentration of 5 × 104 cells per well, were stimulated for 3 d with anti-CD3/anti-CD28 mAbs–coated beads (Dynal) at a 1:1 cell/bead ratio or with the indicated APCs together with IL-2 (20 ng/ml; PeproTech) in 96-well, flat-bottom plates. Where indicated, IL-12 (40 ng/ml; PeproTech), IFN-γ (50 ng/ml; PeproTech), phosphorothioate-modified CpG-ODN (CpG) (1 μM; GeneDesign), or anti–IL-12p40 mAb (10 μg/ml) was added.

Supernatants from cultures of APCs were harvested and pooled. IL-12p40 concentrations were measured using the commercially available Mouse ELISA Ready-SET-Go! Kit (eBioscience). All samples were run in triplicate.

Cell lysates from Tregs (2 × 105 cells) were resolved by NaDodSO4 PAGE (SDS-PAGE) and subjected to Western blot analysis. The membranes were immunoblotted with various Abs, and the bound Abs were visualized with HRP-conjugated Abs against rabbit or mouse IgG (Jackson ImmunoResearch Laboratories), using Chemi-Lumi One Super Western blotting detection reagents (Nacalai Tesque). Abs against phospho-STAT1, phospho-STAT3, and phospho-STAT4 (all from Cell Signaling Technology) and tubulin (Sigma) were used to visualize the corresponding proteins.

Paired Student t test, Mann–Whitney U tests, or Kruskal-Wallis tests with the Bonferroni test were used to determine statistical significance. Error bars show SD.

We previously reported that T cell–specific Socs1 conditional knockout mice (Socs1fl/flLck-Cre+ mice) survived for >6 mo, and all eventually developed dermatitis with age (12, 16). Almost all of these Socs1fl/flLck-Cre+ mice displayed splenomegaly, lymphadenopathy, and abnormal lobulation of the thymus. The autoimmune phenotype of these Socs1fl/flLck-Cre+ mice has been thought to be primarily caused by hyperactivation of effector T cells, because most CD4+ T cells showed effector-memory phenotypes (CD44highCD62Llow) (16) and Ifng−/−Socs1fl/flLck-Cre+ mice did not develop dermatitis (data not shown).

To examine the role of SOCS1 specifically in Tregs, we generated Treg-specific Socs1-deficient mice (Socs1fl/flFoxp3YFP-Cre mice) by crossing Socs1fl/fl mice with Foxp3YFP-Cre knock-in mice (17). We confirmed Socs1 gene deletion in Foxp3YFP-Cre-positive cells, but not in Foxp3YFP-Cre-negative cells. In Socs1+/+Foxp3YFP-Cre (WT) mice, the expression of Socs1 was higher in Tregs than in non-Tregs (Supplemental Fig. 1). Similar to the T cell–specific Socs1-deficient Socs1fl/flLck-Cre+ mice, dermatitis, splenomegaly, and LN swelling were observed in the Treg-specific Socs1-deficient Socs1fl/flFoxp3YFP-Cre mice (Fig. 1A, 1B). However, penetration of these phenotypes was ∼40% in these mice, and dermatitis was milder and later than that observed in the Socs1fl/flLck-Cre+ mice (16). Unlike in the Socs1fl/flLck-Cre+ mice, abnormal lobulation of the thymus was not detected in the Socs1fl/flFoxp3YFP-Cre mice.

FIGURE 1.

Characterization of Socs1fl/flFoxp3YFP-Cre mice. (A) Representative appearance of Socs1fl/flFoxp3YFP-Cre mouse >12 wk old (green arrows indicate dermatitis), and representative macroscopic view of the spleen and LNs of a Socs1+/+Foxp3YFP-Cre (WT) mouse and its age- (>12 wk) and sex-matched Socs1fl/flFoxp3YFP-Cre counterpart. (B) Incidence of dermatitis in Socs1fl/flFoxp3YFP-Cre mice (n = 20, the total number of individual animals analyzed). (C) Foxp3 protein and YFP expression in Tregs among gated CD4+ T cells from the thymus, spleen, and LNs of female Socs1fl/flFoxp3YFP-Cre/+ mice and Socs1+/+Foxp3YFP-Cre/+ littermates. Data are representative of three independent experiments (n = 3, the total number of individual animals analyzed). Percentages and absolute numbers were summarized, and the data are presented as the mean ± SD of three independent experiments. (D) Incidence of dermatitis in Socs1fl/flFoxp3YFP-Cre/+ mice (n = 8, the total number of individual animals analyzed). (E) Representative CD62L (upper) and CD44 (lower) expression on total CD3+CD4+ (left), CD3+CD4+CD25+Foxp3+ (middle), and CD3+CD4+CD25+Foxp3 (right) cells from the LNs of WT and Socs1fl/flFoxp3YFP-Cre mice, as determined using flow cytometry. Data are representative of five independent experiments (n = 5, the total number of individual animals analyzed). (F) Representative T-bet expression on CD3+CD4+CD25+Foxp3+ cells from the LNs of WT Socs1+/+, Socs1fl/flLck-Cre+, Socs1fl/flFoxp3YFP-Cre (<12 wk), and Socs1fl/flFoxp3YFP-Cre (>12 wk) mice, as determined using flow cytometry. Data are representative of three independent experiments (n = 3, the total number of individual animals analyzed). **p < 0.01, Student t test.

FIGURE 1.

Characterization of Socs1fl/flFoxp3YFP-Cre mice. (A) Representative appearance of Socs1fl/flFoxp3YFP-Cre mouse >12 wk old (green arrows indicate dermatitis), and representative macroscopic view of the spleen and LNs of a Socs1+/+Foxp3YFP-Cre (WT) mouse and its age- (>12 wk) and sex-matched Socs1fl/flFoxp3YFP-Cre counterpart. (B) Incidence of dermatitis in Socs1fl/flFoxp3YFP-Cre mice (n = 20, the total number of individual animals analyzed). (C) Foxp3 protein and YFP expression in Tregs among gated CD4+ T cells from the thymus, spleen, and LNs of female Socs1fl/flFoxp3YFP-Cre/+ mice and Socs1+/+Foxp3YFP-Cre/+ littermates. Data are representative of three independent experiments (n = 3, the total number of individual animals analyzed). Percentages and absolute numbers were summarized, and the data are presented as the mean ± SD of three independent experiments. (D) Incidence of dermatitis in Socs1fl/flFoxp3YFP-Cre/+ mice (n = 8, the total number of individual animals analyzed). (E) Representative CD62L (upper) and CD44 (lower) expression on total CD3+CD4+ (left), CD3+CD4+CD25+Foxp3+ (middle), and CD3+CD4+CD25+Foxp3 (right) cells from the LNs of WT and Socs1fl/flFoxp3YFP-Cre mice, as determined using flow cytometry. Data are representative of five independent experiments (n = 5, the total number of individual animals analyzed). (F) Representative T-bet expression on CD3+CD4+CD25+Foxp3+ cells from the LNs of WT Socs1+/+, Socs1fl/flLck-Cre+, Socs1fl/flFoxp3YFP-Cre (<12 wk), and Socs1fl/flFoxp3YFP-Cre (>12 wk) mice, as determined using flow cytometry. Data are representative of three independent experiments (n = 3, the total number of individual animals analyzed). **p < 0.01, Student t test.

Close modal

When we compared the relative populations of Socs1-sufficient and Socs1-deficient Tregs in a single Socs1fl/flFoxp3YFP-Cre/+ female mouse, we observed a strong difference in their ratios and absolute cell numbers in the thymus, spleen, and LNs (Fig. 1C). Because Foxp3 is randomly expressed in each chromosome owing to lyonization of the X chromosome, the YFP+/YFP ratio is 1:1 in Foxp3YFP-Cre/+ female mice. Indeed, the YFP+/YFP ratio in the WT Socs1+/+Foxp3YFP-Cre/+ females was nearly 1:1 in both the thymus and the periphery (the spleen and LNs) (Fig. 1C). However, the YFP+/YFP ratio in the Socs1fl/flFoxp3YFP-Cre/+ females was 5–7:1 in the thymus and periphery. These results indicated that Socs1 deficiency in Tregs provides a great advantage in terms of increasing their population in a cell-autonomous manner, which is consistent with our previous report that showed an expansion of Tregs in Socs1fl/flLck-Cre+ mice (16).

We found that Socs1fl/flFoxp3YFP-Cre/+ female mice also develop splenomegaly, lymphadenopathy, and dermatitis 2.5 mo after birth (Fig. 1D), suggesting a cell-intrinsic role of SOCS1 in Tregs. However, compared with Socs1fl/flFoxp3YFP-Cre female mice (Fig. 1B), the onset of autoimmune symptoms of Socs1fl/flFoxp3YFP-Cre/+ female mice was slower and severity was lower, suggesting that normal Socs1-sufficient Tregs can partly suppress the pathogenicity of Socs1-deficient Tregs. These observations support our hypothesis that pathogenic phenotypes of Socs1-deficient Tregs are obvious only under inflammatory conditions.

Next, we examined whether Socs1 deficiency in Tregs affects naive and effector T cells. The fraction of CD62Llow cells was greatly increased in Tregs from Socs1fl/flFoxp3YFP-Cre mice compared with controls (Fig. 1E, upper middle panel). CD44 levels were not altered between WT and Socs1-deficient Tregs (Fig. 1E, lower middle panel). Interestingly, non-Treg (YFP−) CD4+ T cells from Socs1fl/flFoxp3YFP-Cre mice showed reduced CD62L and higher CD44 expression compared with controls (Fig. 1E, right), suggesting that activated or effector CD4+ T cells increased in Socs1fl/flFoxp3YFP-Cre mice (Fig. 1E, left). In addition, Tregs in the Socs1fl/flLck-Cre+ and in the older Socs1fl/flFoxp3YFP-Cre mice were found to express higher levels of T-bet (Fig. 1F), suggesting that T-bet expression in Tregs correlates with severe inflammation in vivo. These data suggest that Socs1-deficient Tregs have some defects in normal Treg functions, thereby increasing the activation and/or memory status of Socs1-sufficient non-Tregs in Socs1fl/flFoxp3YFP-Cre mice.

To examine whether deletion of Socs1 in Tregs facilitates anti-tumor immunity, 2 × 105 B16/F10 melanoma cells were transplanted into WT or Socs1fl/flFoxp3YFP-Cre mice. The tumor grew more slowly in the Socs1fl/flFoxp3YFP-Cre mice than in the WT mice (Fig. 2A). Next, to investigate the fate of Tregs in vivo, fate mapping mice were generated by crossing Socs1fl/flFoxp3YFP-Cre and R26tdRFP knock-in mice carrying a tdRFP within the Rosa26 locus. Tregs (YFP+RFP+ cells) from fate mapping mice were transferred into WT mice transplanted with 1 × 106 B16/F10 melanoma cells after 2 Gy irradiation. Transfer of WT Tregs (Socs1+/+ Tregs) enhanced tumor growth, whereas transfer of Tregs from Socs1fl/flFoxp3YFP-Cre×R26tdRFP mice (Socs1−/− Tregs) suppressed tumor growth compared with control mice without Treg transfer (Fig. 2B). Transferred Tregs did not cause autoimmune phenotypes in these mice. In the draining LN, the percentage of the ex-Treg fraction (YFP in RFP+) was not different between Socs1−/− Tregs and Socs1+/+ Tregs (Fig. 2C, upper panels, 2D). However, within the tumor, the ex-Treg fraction was significantly increased in Socs1−/− Tregs compared with Socs1+/+ Tregs (Fig. 2C, lower panels, 2D). In addition, Socs1−/− ex-Tregs (YFPRFP+ cells) isolated from the tumors expressed higher levels of Infg than did Socs1+/+ ex-Tregs (Fig. 2E). These data suggest that the environment within the tumor facilitated the pathogenic conversion of Socs1-deficient Tregs into Foxp3IFN-γ+ ex-Tregs.

FIGURE 2.

Inhibition of the growth of B16/F10 melanoma cells in Socs1fl/flFoxp3YFP-Cre mice and accelerated conversion of Socs1-deficient Tregs to IFN-γ–producing ex-Tregs in the tumor. (A) B16/F10 melanoma cells (2 × 105 cells) were transferred into Socs1+/+Foxp3YFP-Cre or Socs1fl/flFoxp3YFP-Cre mice, and tumor growth was assessed (the red circles indicate tumors). Data are presented as the mean ± SD of 10 independent experiments (n = 10, the total number of individual animals analyzed). *p < 0.05, **p < 0.01, Student t test. (B) For the fate mapping system, 5 × 105 Tregs (YFP+RFP+ cells) from Socs1+/+Foxp3YFP-Cre×R26tdRFP or Socs1fl/flFoxp3YFP-Cre×R26tdRFP mice (Socs1+/+ or Socs1−/− Tregs) were transferred into WT mice transplanted with 1 × 106 B16/F10 melanoma cells after 2 Gy irradiation. Tumor growth was assessed in these mice. Data are presented as the mean ± SD of eight independent experiments (n = 8, the total number of individual transferred animals analyzed). Transferred cells (Socs1+/+ or Socs1−/− Tregs) were sorted from 50 animals in each group. *p < 0.05, **p < 0.01, Kruskal–Wallis tests with Bonferroni test. (C and D) At 15 d after cell transfer in (B), mice were sacrificed for experiments, and cells (YFP+RFP+ or YFPRFP+ cells) from the tumors and draining LNs were analyzed. Data are representative of three independent experiments (n = 3, the total number of individual transferred animals analyzed). Transferred cells (Socs1+/+ or Socs1−/− Tregs) were sorted from 20 animals in each group. Percentages of YFP cells in RFP+ cells were summarized, and data are presented as the mean ± SD of three independent experiments. **p < 0.01, Student t test. (E) Transferred Tregs from Socs1+/+Foxp3YFP-Cre×R26tdRFP or Socs1fl/flFoxp3YFP-Cre×R26tdRFP mice were sorted from the tumors after 15 d of transfer as Tregs (YFP+RFP+ cells) or ex-Tregs (YFPRFP+ cells), and Ifn-γ mRNA levels were analyzed using quantitative RT-PCR. Results are presented relative to expression of the control gene Hprt. Data are presented as the mean ± SD of three independent experiments. n = 3, the total number of individual transferred animals analyzed. Transferred cells (Socs1+/+ or Socs1−/− Tregs) were sorted from 20 animals in each group. **p < 0.01, Mann–Whitney U tests.

FIGURE 2.

Inhibition of the growth of B16/F10 melanoma cells in Socs1fl/flFoxp3YFP-Cre mice and accelerated conversion of Socs1-deficient Tregs to IFN-γ–producing ex-Tregs in the tumor. (A) B16/F10 melanoma cells (2 × 105 cells) were transferred into Socs1+/+Foxp3YFP-Cre or Socs1fl/flFoxp3YFP-Cre mice, and tumor growth was assessed (the red circles indicate tumors). Data are presented as the mean ± SD of 10 independent experiments (n = 10, the total number of individual animals analyzed). *p < 0.05, **p < 0.01, Student t test. (B) For the fate mapping system, 5 × 105 Tregs (YFP+RFP+ cells) from Socs1+/+Foxp3YFP-Cre×R26tdRFP or Socs1fl/flFoxp3YFP-Cre×R26tdRFP mice (Socs1+/+ or Socs1−/− Tregs) were transferred into WT mice transplanted with 1 × 106 B16/F10 melanoma cells after 2 Gy irradiation. Tumor growth was assessed in these mice. Data are presented as the mean ± SD of eight independent experiments (n = 8, the total number of individual transferred animals analyzed). Transferred cells (Socs1+/+ or Socs1−/− Tregs) were sorted from 50 animals in each group. *p < 0.05, **p < 0.01, Kruskal–Wallis tests with Bonferroni test. (C and D) At 15 d after cell transfer in (B), mice were sacrificed for experiments, and cells (YFP+RFP+ or YFPRFP+ cells) from the tumors and draining LNs were analyzed. Data are representative of three independent experiments (n = 3, the total number of individual transferred animals analyzed). Transferred cells (Socs1+/+ or Socs1−/− Tregs) were sorted from 20 animals in each group. Percentages of YFP cells in RFP+ cells were summarized, and data are presented as the mean ± SD of three independent experiments. **p < 0.01, Student t test. (E) Transferred Tregs from Socs1+/+Foxp3YFP-Cre×R26tdRFP or Socs1fl/flFoxp3YFP-Cre×R26tdRFP mice were sorted from the tumors after 15 d of transfer as Tregs (YFP+RFP+ cells) or ex-Tregs (YFPRFP+ cells), and Ifn-γ mRNA levels were analyzed using quantitative RT-PCR. Results are presented relative to expression of the control gene Hprt. Data are presented as the mean ± SD of three independent experiments. n = 3, the total number of individual transferred animals analyzed. Transferred cells (Socs1+/+ or Socs1−/− Tregs) were sorted from 20 animals in each group. **p < 0.01, Mann–Whitney U tests.

Close modal

Defective suppressive activity of Socs1-deficient Tregs from Socs1fl/flLck-Cre+ mice in vivo has been reported; thus, Socs1-deficient Tregs did not suppress colitis in Rag2−/− mice transferred with naive T cells (16). In contrast, Tregs from Socs1fl/flFoxp3YFP-Cre mice retained suppressive activity in vivo; Rag2−/− mice transferred with Socs1-deficient Tregs from Socs1fl/flFoxp3YFP-Cre mice with naive T cells did not show severe colitis, although the body weight of these mice was slightly decreased (Fig. 3A). Foxp3 expression in these Socs1−/− Tregs was as stable as that in the Socs1+/+ Tregs after transfer into Rag2−/− mice (94.5% Foxp3+ in WT Tregs versus 88.4% in Socs1-deficient Tregs) (Fig. 3B), whereas more severe loss of Foxp3 was observed when Tregs from Socs1fl/flLck-Cre+ mice were used (16). IFN-γ levels in the mesenteric LNs in Rag2−/− mice cotransferred with naive T cells and Socs1−/− Tregs from Socs1fl/flFoxp3YFP-Cre mice were nearly the same as those in Rag2−/− mice transferred with naive T cells and WT Tregs (data not shown).

FIGURE 3.

Maintenance of suppressive activity and Foxp3 expression of Socs1-deficient Tregs from Socs1fl/flFoxp3YFP-Cre mice (<12 wk) in Rag2−/− mice. (A and B) CD3+CD4+CD25+YFP+ Tregs (2 × 105 cells) from the LNs of Ly5.2 Socs1+/+Foxp3YFP-Cre mice (Socs1+/+ Tregs) or Ly5.2 Socs1fl/flFoxp3YFP-Cre mice (<12 wk) (Socs1−/− Tregs) were cotransferred with 4 × 105 Ly5.1 CD4+CD25CD62L+CD44 naive T cells into Rag2−/− mice. Four weeks later, recipient mice were analyzed. (A) Body weight changes. (B) Flow cytometric analysis of Foxp3 expression on Ly5.2+CD3+CD4+ T cells from the LNs of Rag2−/− mice (percentage of Foxp3 is shown in the bar graph). Data are representative of three independent experiments (n = 3, the total number of individual transferred animals analyzed). Transferred Tregs (Socs1+/+ or Socs1−/− Tregs) were sorted from 15 animals in each group. Transferred naive T cells were sorted from three animals. Data are presented as the mean ± SD of three independent experiments. (C and D) Tregs (2 × 105 cells) from the LNs of Ly5.2 Socs1+/+Foxp3YFP-Cre mice (Socs1+/+ Tregs) or Ly5.2 Socs1fl/flFoxp3YFP-Cre mice (<12 wk) (Socs1−/− Tregs) were transferred into Rag2−/− mice. Eight weeks later, recipient mice were analyzed. (C) Body weight changes over time. (D) Flow cytometric analysis of Foxp3 expression on CD3+CD4+ T cells from the LNs of Rag2−/− mice (percentage of Foxp3 is shown in the bar graph). Data are representative of three independent experiments (n = 3, the total number of individual transferred animals analyzed). Transferred cells (Socs1+/+ or Socs1−/− Tregs) were sorted from 15 animals in each group. Data are presented as the mean ± SD of three independent experiments. Student t test was performed.

FIGURE 3.

Maintenance of suppressive activity and Foxp3 expression of Socs1-deficient Tregs from Socs1fl/flFoxp3YFP-Cre mice (<12 wk) in Rag2−/− mice. (A and B) CD3+CD4+CD25+YFP+ Tregs (2 × 105 cells) from the LNs of Ly5.2 Socs1+/+Foxp3YFP-Cre mice (Socs1+/+ Tregs) or Ly5.2 Socs1fl/flFoxp3YFP-Cre mice (<12 wk) (Socs1−/− Tregs) were cotransferred with 4 × 105 Ly5.1 CD4+CD25CD62L+CD44 naive T cells into Rag2−/− mice. Four weeks later, recipient mice were analyzed. (A) Body weight changes. (B) Flow cytometric analysis of Foxp3 expression on Ly5.2+CD3+CD4+ T cells from the LNs of Rag2−/− mice (percentage of Foxp3 is shown in the bar graph). Data are representative of three independent experiments (n = 3, the total number of individual transferred animals analyzed). Transferred Tregs (Socs1+/+ or Socs1−/− Tregs) were sorted from 15 animals in each group. Transferred naive T cells were sorted from three animals. Data are presented as the mean ± SD of three independent experiments. (C and D) Tregs (2 × 105 cells) from the LNs of Ly5.2 Socs1+/+Foxp3YFP-Cre mice (Socs1+/+ Tregs) or Ly5.2 Socs1fl/flFoxp3YFP-Cre mice (<12 wk) (Socs1−/− Tregs) were transferred into Rag2−/− mice. Eight weeks later, recipient mice were analyzed. (C) Body weight changes over time. (D) Flow cytometric analysis of Foxp3 expression on CD3+CD4+ T cells from the LNs of Rag2−/− mice (percentage of Foxp3 is shown in the bar graph). Data are representative of three independent experiments (n = 3, the total number of individual transferred animals analyzed). Transferred cells (Socs1+/+ or Socs1−/− Tregs) were sorted from 15 animals in each group. Data are presented as the mean ± SD of three independent experiments. Student t test was performed.

Close modal

Next, we examined the fate of Foxp3+ Tregs under lymphopenic conditions. Tregs from the LNs of WT or Socs1fl/flFoxp3YFP-Cre mice were transferred into Rag2−/− mice, and Foxp3 positivity was examined using flow cytometry. We previously reported a similar experiment using Tregs from the LNs of WT or Socs1fl/flLck-Cre+ mice in which, 6 wk after transfer, Rag2−/− mice transferred with Tregs from Socs1fl/flLck-Cre+ mice developed more profound colitis than did those transferred with Tregs from the WT mice, and the Socs1fl/flLck-Cre+ Tregs showed a greater loss of Foxp3 expression than did WT Tregs (16). However, Rag2−/− mice transferred with Tregs from the Socs1-deficient Socs1fl/flFoxp3YFP-Cre mice showed only a slight body weight loss and did not have colitis even when observed 8 wk later (Fig. 3C). In addition, these Socs1-deficient Tregs maintained the same levels of Foxp3 expression as WT Tregs (44.3% Foxp3+ in WT Tregs versus 42.3% in Socs1-deficient Tregs) (Fig. 3D). Thus, in contrast to the results of Tregs from Socs1fl/flLck-Cre+ mice, suppressive functions and Foxp3 expression were maintained with relative stability in SOCS1-deficient Tregs from Socs1fl/flFoxp3YFP-Cre mice under lymphopenic conditions.

When stimulated with anti-CD3/CD28 Abs in vitro, Tregs from Socs1fl/flLck-Cre+ mice (Socs1fl/flLck-Cre+ Tregs) showed a loss of Foxp3 expression and produced IFN-γ similar to that shown in the in vivo experiments in Fig. 3, whereas WT Tregs (Socs1+/+ Tregs) or Tregs from Socs1fl/flFoxp3YFP-Cre mice (Socs1fl/flFoxp3YFP-Cre Tregs) did not (Fig. 4A, upper). The results of stimulation with APCs (CD3+ T cell–depleted spleen cells) from WT mice were the same as those obtained when using CD3/CD28 stimulation (Fig. 4A, middle). However, when these Tregs were cultured with APCs from Socs1fl/flLck-Cre+ mice, the Socs1fl/flFoxp3YFP-Cre Tregs lost Foxp3 expression and produced IFN-γ to a level statistically similar to that produced by the Socs1fl/flLck-Cre+ Tregs (Fig. 4A, lower, 4B). The proportions of Foxp3IFN-γ+ and Foxp3+IFN-γ+ cells in Socs1fl/flFoxp3YFP-Cre Tregs were significantly increased when they were cocultured with APCs from Socs1fl/flLck-Cre+ mice, compared with coculture with anti-CD3/CD28 Abs or APCs from WT mice (Fig. 4B, Supplemental Fig. 2). Thus, when Tregs were cultured with APCs from highly inflammatory Socs1fl/flLck-Cre+ mice in vitro, Socs1fl/flFoxp3YFP-Cre Tregs also converted to ex-Tregs. The amount of IFN-γ production from Socs1-deficient Tregs was dependent on the number of APCs and was independent of the number of Tregs (data not shown).

FIGURE 4.

Production of IFN-γ from Socs1-deficient Tregs from Socs1fl/flFoxp3YFP-Cre mice when cultured with an inflammatory cytokine and APCs. (A) Sorted Tregs were cultured in vitro for 72 h with IL-2 (20 ng/ml) in each condition, and the expression of IFN-γ and Foxp3 was analyzed using flow cytometry. Sorted Tregs (5 × 104 cells per well) from Socs1+/+, Socs1fl/flLck-Cre+, or Socs1fl/flFoxp3YFP-Cre (<12 wk) mice were cultured in vitro with anti-CD3/CD28 beads (upper) or APCs (2 × 105 CD3+ T cell–depleted spleen cells per well) from WT (middle) or Socs1fl/flLck-Cre+ (lower) mice. Data are representative of three independent experiments. Cultured Tregs were sorted from nine animals in each group. Cultured APCs were sorted from three animals. (B) The proportion of the Foxp3IFN-γ+ fraction in the cells in (A) was summarized and statistically processed. Data are presented as the mean ± SD of three independent experiments. (C) The supernatant from a 4-d culture of Socs1+/+ Tregs (5 × 104 cells per well) with Socs1+/+ APCs (2 × 105 cells per well) was used as a WT culture supernatant (WT culture sup), or the supernatant from a 4-d culture of Socs1fl/flLck-Cre+ Tregs (5 × 104 cells per well) with APCs (2 × 105 cells per well) from Socs1fl/flLck-Cre+ mice was used as a Socs1fl/flLck-Cre+ culture supernatant (Socs1fl/flLck-Cre+ culture sup). Sorted Tregs were cultured in vitro for 72 h in each condition, and the expression of IFN-γ and Foxp3 was analyzed using flow cytometry. Left, Tregs (5 × 104 cells per well) from Socs1+/+ or Socs1fl/flFoxp3YFP-Cre mice (<12 wk) were cultured with anti-CD3/CD28 beads and WT culture supernatant or Socs1fl/flLck-Cre+ culture supernatant. Right, Tregs (5 × 104 cells per well) from Socs1+/+ or Socs1fl/flFoxp3YFP-Cre mice (<12 wk) were cultured with APCs (2 × 105 cells per well) from Socs1+/+ mice and WT culture supernatant or Socs1fl/flLck-Cre+ culture supernatant. Data are representative of three independent experiments. Cultured Tregs were sorted from 12 animals in each group. Cultured APCs were sorted from three animals. For the supernatant, Tregs were sorted from 12 animals in each group, and APCs were sorted from three animals in each group. **p < 0.01, Kruskal–Wallis tests with the Bonferroni test.

FIGURE 4.

Production of IFN-γ from Socs1-deficient Tregs from Socs1fl/flFoxp3YFP-Cre mice when cultured with an inflammatory cytokine and APCs. (A) Sorted Tregs were cultured in vitro for 72 h with IL-2 (20 ng/ml) in each condition, and the expression of IFN-γ and Foxp3 was analyzed using flow cytometry. Sorted Tregs (5 × 104 cells per well) from Socs1+/+, Socs1fl/flLck-Cre+, or Socs1fl/flFoxp3YFP-Cre (<12 wk) mice were cultured in vitro with anti-CD3/CD28 beads (upper) or APCs (2 × 105 CD3+ T cell–depleted spleen cells per well) from WT (middle) or Socs1fl/flLck-Cre+ (lower) mice. Data are representative of three independent experiments. Cultured Tregs were sorted from nine animals in each group. Cultured APCs were sorted from three animals. (B) The proportion of the Foxp3IFN-γ+ fraction in the cells in (A) was summarized and statistically processed. Data are presented as the mean ± SD of three independent experiments. (C) The supernatant from a 4-d culture of Socs1+/+ Tregs (5 × 104 cells per well) with Socs1+/+ APCs (2 × 105 cells per well) was used as a WT culture supernatant (WT culture sup), or the supernatant from a 4-d culture of Socs1fl/flLck-Cre+ Tregs (5 × 104 cells per well) with APCs (2 × 105 cells per well) from Socs1fl/flLck-Cre+ mice was used as a Socs1fl/flLck-Cre+ culture supernatant (Socs1fl/flLck-Cre+ culture sup). Sorted Tregs were cultured in vitro for 72 h in each condition, and the expression of IFN-γ and Foxp3 was analyzed using flow cytometry. Left, Tregs (5 × 104 cells per well) from Socs1+/+ or Socs1fl/flFoxp3YFP-Cre mice (<12 wk) were cultured with anti-CD3/CD28 beads and WT culture supernatant or Socs1fl/flLck-Cre+ culture supernatant. Right, Tregs (5 × 104 cells per well) from Socs1+/+ or Socs1fl/flFoxp3YFP-Cre mice (<12 wk) were cultured with APCs (2 × 105 cells per well) from Socs1+/+ mice and WT culture supernatant or Socs1fl/flLck-Cre+ culture supernatant. Data are representative of three independent experiments. Cultured Tregs were sorted from 12 animals in each group. Cultured APCs were sorted from three animals. For the supernatant, Tregs were sorted from 12 animals in each group, and APCs were sorted from three animals in each group. **p < 0.01, Kruskal–Wallis tests with the Bonferroni test.

Close modal

To clarify the factors of APCs from Socs1fl/flLck-Cre+ mice that were important for the conversion of Tregs into ex-Tregs, the culture supernatant from each in vitro culture of Tregs plus APCs was added to the culture of Tregs and anti-CD3/CD28 Abs or APCs (Fig. 4C). Thus, Socs1+/+ Tregs and Socs1fl/flFoxp3YFP-Cre Tregs were cultured in the presence of WT culture supernatant (i.e., supernatant from a 4-d culture of Socs1+/+ Tregs with Socs1+/+ APCs) or Socs1fl/flLck-Cre+ culture supernatant (i.e., supernatant from a 4-d culture of Socs1fl/flLck-Cre+ Tregs with APCs from Socs1fl/flLck-Cre+ mice). Both Socs1+/+ Tregs and Socs1fl/flFoxp3YFP-Cre Tregs efficiently converted to Foxp3IFN-γ+ cells when they were cultured in the presence of the Socs1fl/flLck-Cre+ culture supernatant (Fig. 4C, lower panels). Furthermore, Socs1fl/flFoxp3YFP-Cre Tregs produced the highest levels of IFN-γ from the Foxp3 fraction when they were cocultured with Socs1+/+ APCs in the presence of the Socs1fl/flLck-Cre+ culture supernatant (Fig. 4C, lower right). The proportions of the Foxp3IFN-γ+ fraction and the Foxp3+IFN-γ+ fraction are summarized in Supplemental Fig. 3. There was no significant difference between Socs1+/+ Tregs and Socs1fl/flFoxp3YFP-Cre Tregs in the production of IFN-γ in the Foxp3+ fraction. These results suggest that the Socs1fl/flLck-Cre+ culture supernatant contains some soluble factors that convert Socs1fl/flFoxp3YFP-Cre Tregs into pathogenic ex-Tregs.

The production of IFN-γ from Socs1+/+ or Socs1fl/flFoxp3YFP-Cre Tregs cultured with APCs from Socs1fl/flLck-Cre+ mice was completely inhibited by the anti–IL-12p40 Ab (Fig. 5A), but not by the anti–IFN-γ Ab (data not shown). IFN-γ production from the Foxp3 fraction was observed in Socs1fl/flFoxp3YFP-Cre Tregs, but not in Socs1+/+ Tregs cocultured with Socs1+/+ APCs in the presence of IL-12 (Fig. 5B, Supplemental Fig. 4). To further characterize the role of APCs in the conversion of Socs1-deficient Tregs to Foxp3IFN-γ+ ex-Tregs, Tregs were cocultured with Socs1+/+ APCs that were stimulated with IFN-γ or the CpG oligonucleotide. APCs activated with IFN-γ induced Foxp3IFN-γ+ and Foxp3+IFN-γ+ cells from Socs1fl/flFoxp3YFP-Cre Tregs (Fig. 5C, upper); however, APCs activated with the CpG oligonucleotide induced more IFN-γ+ cells than did APCs activated with IFN-γ (Fig. 5C, lower). APCs from Socs1fl/flFoxp3YFP-Cre mice produced significantly more IL-12p40 than did APCs from Socs1+/+ mice in response to the CpG oligonucleotide (Fig. 6A). Thus, APCs from Socs1fl/flFoxp3YFP-Cre mice are already somewhat activated probably because of inflammatory conditions in these mice. Regarding the culture supernatant used in Fig. 4C, IL-12p40 was also highly secreted from APCs from Socs1fl/flLck-Cre+ mice when they were cocultured with Socs1fl/flLck-Cre+ Tregs (Fig. 6B), suggesting that APCs were already highly activated in Socs1-deficient mice and their activation level was similar to that of APCs stimulated with the CpG oligonucleotide.

FIGURE 5.

IL-12 production from APCs for the conversion of Socs1-deficient Tregs into ex-Tregs. Sorted Tregs were cultured in vitro for 72 h with IL-2 (20 ng/ml) in each condition, and the expression of IFN-γ and Foxp3 was analyzed using flow cytometry. (A) Tregs (5 × 104 cells per well) from Socs1+/+ or Socs1fl/flFoxp3YFP-Cre mice (<12 wk) mice were cultured with APCs (2 × 105 cells per well) from Socs1fl/flLck-Cre+ mice in the presence of anti–IL-12p40 Ab or isotype control (10 μg/ml). Data are representative of three independent experiments. (Cultured Tregs were sorted from nine animals in each group. Cultured APCs were sorted from three animals.) Data are presented as the mean ± SD of three independent experiments. (B) Left, Tregs (5 × 104 cells per well) from Socs1+/+ or Socs1fl/flFoxp3YFP-Cre mice (<12 wk) were cultured with anti-CD3/CD28 beads in the presence or absence of IL-12 (40 ng/ml). Right, Tregs (5 × 104 cells per well) from Socs1+/+ or Socs1fl/flFoxp3YFP-Cre mice (<12 wk) were cultured with APCs (2 × 105 cells per well) from Socs1+/+ mice in the presence or absence of IL-12 (40 ng/ml). Data are representative of three independent experiments. Cultured Tregs were sorted from 12 animals in each group. Cultured APCs were sorted from three animals. (C) Tregs (5 × 104 cells per well) from Socs1+/+ or Socs1fl/flFoxp3YFP-Cre mice (<12 wk) mice were cultured with APCs (2 × 105 cells per well) from Socs1+/+ mice in the presence of IFN-γ (50 ng/ml) (upper) or CpG (1 μM) (lower). Data are representative of three independent experiments. Cultured Tregs were sorted from six animals in each group. Cultured APCs were sorted from three animals. Data are presented as the mean ± SD of three independent experiments. *p < 0.05, **p < 0.01, Student t test.

FIGURE 5.

IL-12 production from APCs for the conversion of Socs1-deficient Tregs into ex-Tregs. Sorted Tregs were cultured in vitro for 72 h with IL-2 (20 ng/ml) in each condition, and the expression of IFN-γ and Foxp3 was analyzed using flow cytometry. (A) Tregs (5 × 104 cells per well) from Socs1+/+ or Socs1fl/flFoxp3YFP-Cre mice (<12 wk) mice were cultured with APCs (2 × 105 cells per well) from Socs1fl/flLck-Cre+ mice in the presence of anti–IL-12p40 Ab or isotype control (10 μg/ml). Data are representative of three independent experiments. (Cultured Tregs were sorted from nine animals in each group. Cultured APCs were sorted from three animals.) Data are presented as the mean ± SD of three independent experiments. (B) Left, Tregs (5 × 104 cells per well) from Socs1+/+ or Socs1fl/flFoxp3YFP-Cre mice (<12 wk) were cultured with anti-CD3/CD28 beads in the presence or absence of IL-12 (40 ng/ml). Right, Tregs (5 × 104 cells per well) from Socs1+/+ or Socs1fl/flFoxp3YFP-Cre mice (<12 wk) were cultured with APCs (2 × 105 cells per well) from Socs1+/+ mice in the presence or absence of IL-12 (40 ng/ml). Data are representative of three independent experiments. Cultured Tregs were sorted from 12 animals in each group. Cultured APCs were sorted from three animals. (C) Tregs (5 × 104 cells per well) from Socs1+/+ or Socs1fl/flFoxp3YFP-Cre mice (<12 wk) mice were cultured with APCs (2 × 105 cells per well) from Socs1+/+ mice in the presence of IFN-γ (50 ng/ml) (upper) or CpG (1 μM) (lower). Data are representative of three independent experiments. Cultured Tregs were sorted from six animals in each group. Cultured APCs were sorted from three animals. Data are presented as the mean ± SD of three independent experiments. *p < 0.05, **p < 0.01, Student t test.

Close modal
FIGURE 6.

Increased production of IL-12p40 by APCs affected by Socs1-deficient Tregs and activation of STAT4 in Socs1-deficient Tregs by IL-12 for conversion into ex-Tregs. (A) APCs (2 × 105 cells per well) from Socs1+/+ or Socs1fl/flFoxp3YFP-Cre mice (<12 wk) mice were cultured for 72 h with IFN-γ (50 ng/ml) or CpG (1 μM), following which IL-12p40 levels in the culture supernatant were measured using ELISA. Data are presented as the mean ± SD of three independent experiments. Cultured APCs were sorted from six animals in each group. **p < 0.01, Student t test and Kruskal–Wallis tests with the Bonferroni test. (B) Tregs (5 × 104 cells per well) and APCs (2 × 105 cells per well) from WT mice were cocultured for 4 d (WT). Similarly, Tregs and APCs from Socs1fl/flLck-Cre+ mice were cocultured for 4 d (Socs1fl/flLck-Cre+). IL-12p40 levels in the culture supernatant were measured using ELISA. Data are presented as the mean ± SD of three independent experiments. **p < 0.01, Student t test. Tregs were sorted from 12 animals in each group, and APCs were sorted from three animals in each group. (C) Analysis of the phosphorylation of STAT1, STAT3, and STAT4 in Tregs. Tregs (1 × 105 cells) from WT or Socs1fl/flFoxp3YFP-Cre mice were stimulated with or without IL-12 (40 ng/ml) for 30 min, following which phosphoSTATs were detected by immunoblotting. Tubulin was analyzed as a loading control. Data are representative of three independent experiments. Cultured Tregs were sorted from 12 animals in each group.

FIGURE 6.

Increased production of IL-12p40 by APCs affected by Socs1-deficient Tregs and activation of STAT4 in Socs1-deficient Tregs by IL-12 for conversion into ex-Tregs. (A) APCs (2 × 105 cells per well) from Socs1+/+ or Socs1fl/flFoxp3YFP-Cre mice (<12 wk) mice were cultured for 72 h with IFN-γ (50 ng/ml) or CpG (1 μM), following which IL-12p40 levels in the culture supernatant were measured using ELISA. Data are presented as the mean ± SD of three independent experiments. Cultured APCs were sorted from six animals in each group. **p < 0.01, Student t test and Kruskal–Wallis tests with the Bonferroni test. (B) Tregs (5 × 104 cells per well) and APCs (2 × 105 cells per well) from WT mice were cocultured for 4 d (WT). Similarly, Tregs and APCs from Socs1fl/flLck-Cre+ mice were cocultured for 4 d (Socs1fl/flLck-Cre+). IL-12p40 levels in the culture supernatant were measured using ELISA. Data are presented as the mean ± SD of three independent experiments. **p < 0.01, Student t test. Tregs were sorted from 12 animals in each group, and APCs were sorted from three animals in each group. (C) Analysis of the phosphorylation of STAT1, STAT3, and STAT4 in Tregs. Tregs (1 × 105 cells) from WT or Socs1fl/flFoxp3YFP-Cre mice were stimulated with or without IL-12 (40 ng/ml) for 30 min, following which phosphoSTATs were detected by immunoblotting. Tubulin was analyzed as a loading control. Data are representative of three independent experiments. Cultured Tregs were sorted from 12 animals in each group.

Close modal

Finally, to examine whether Socs1-deficient Tregs are more sensitive to stimulation with IL-12 than are WT Tregs, we measured STAT phosphorylation in response to IL-12. As shown in Fig. 6C, much stronger tyrosine phosphorylation of STAT4 was observed in Socs1-deficient Tregs from Socs1fl/flFoxp3YFP-Cre mice than in Socs1-sufficient Tregs after 30 min of IL-12 stimulation (Fig. 6C). The activation status of STAT1 and STAT3 was not affected by IL-12. These data suggest that hyperactivation of STAT4 by Socs1 deficiency is responsible for the high levels of IFN-γ production from Socs1-deficient Tregs.

In the current study, we demonstrated that Tregs from Socs1fl/flFoxp3YFP-Cre mice maintained Foxp3 expression when they were transferred into Rag2−/− mice, unlike Tregs from Socs1fl/flLck-Cre+ mice. This finding is in agreement with a previous report that Foxp3 expression of Socs1-deficient Tregs from Socs1fl/flFoxp3YFP-Cre mice was maintained in the presence of naive and effector T cells (14). The reasons why Foxp3 expression in Socs1−/− Tregs from Socs1fl/flFoxp3YFP-Cre mice is more stable than that in Socs1−/− Tregs from Socs1fl/flLck-Cre+ mice are that 1) the environment around Tregs in Socs1fl/flLck-Cre+ mice include inflammatory conditions generated by Socs1-deficient non-Tregs; and 2) SOCS1 in Socs1fl/flLck-Cre+ mice is deleted in the Treg progenitors, whereas SOCS1 in Socs1fl/flFoxp3YFP-Cre mice is removed after Treg maturation. However, when Tregs from Socs1fl/flFoxp3YFP-Cre mice were cultured with IL-12–producing APCs from Socs1fl/flLck-Cre+ mice, STAT4 was activated in these Tregs, and, similar to Tregs from Socs1fl/flLck-Cre+ mice, they lost Foxp3 expression, which was accompanied by the production of inflammatory cytokines. CpG-stimulated APCs or, in particular, IL-12–stimulated APCs, caused Socs1-deficient Tregs from Socs1fl/flFoxp3YFP-Cre mice to produce IFN-γ from the Foxp3 fraction. IFN-γ–producing Foxp3+ cells were reported to be detected in the peripheral mononuclear cells of multiple sclerosis patients (21). These ex-Tregs develop an effector-memory phenotype, produce pathogenic cytokines, and cause autoimmunity. It is of note that they also inhibit tumor progression.

The Foxp3 promoter contains a newly identified Nr4a transcription factor binding site, which is critical for nTreg development in the thymus (22, 23), in addition to transcription factor NFAT and AP-1 binding sites, as well as a TATA and a CAAT box (24). Three highly conserved CNSs, CNS1, CNS2, and CNS3, were also identified. The Treg-specific CpG hypomethylation of the CNS2 TSDR has been reported as indispensable for the development of Foxp3+ T cells as nTregs (25), for lineage stability, and for suppressive activity (26, 27). In addition to epigenetic modification, a recent study has indicated that the coexpression of Foxp3 with at least one of the transcription factors GATA-1, IRF4, Lef1, Ikzf4, or Satb1 is responsible for some of the Treg phenotype gene expression (28). In addition, biochemical and mass-spectrometric analyses have shown that Foxp3 forms complexes with several cofactors, such as GATA3, NFAT, or Runx1/Cbfβ (2932). These protein–protein interactions between Foxp3 and other factors influence Foxp3 stability, although it is not clear how these Foxp3 binding proteins affect Treg stability (33). To date, various factors have been reported to regulate Foxp3 expression, including Smad2/3 (34), Runx1 (30, 35), STAT5 (36), c-Rel (37), TRAF6 (38), PTEN (39), and SOCS1 (16), although they are not bound to Foxp3 directly. The Smad2/3-deficient Treg phenotypes were similar to those observed in Socs1-deficient Tregs (34).

In a previous study, we showed that the CNS2 TSDR of freshly isolated Socs1-deficient Tregs was fully demethylated, even in Socs1fl/flLck-Cre+ mice under inflammatory conditions, indicating that they are committed Foxp3+ Tregs (16). It has been shown that purified Tregs are very stable, even under inflammatory conditions (40), and that uncommitted, not fully demethylated Foxp3+ Tregs become ex-Tregs (9). In contrast, consistent with our results, Tregs with a fully TSDR were found to lose Foxp3 expression in an experimental autoimmune encephalomyelitis model (41). These ex-Tregs with an Ag-specific TCR appeared only when they were adoptively transferred into mice with ongoing experimental autoimmune encephalomyelitis. We also found that Tregs from Socs1fl/flFoxp3YFP-Cre mice lost Foxp3 expression that was accompanied by activated STAT4 when they were cultured with APCs from Socs1fl/flLck-Cre+ mice. In support of these results, almost all previous studies related to Treg plasticity have been studied under lymphopenic or inflammatory conditions (1, 2, 9, 16, 4248). Although we must exclude the experimental technological limitation of methylation sequencing read depth by the Sanger sequencing method from our results, committed Socs1-deficient or STAT4-activated Tregs were determined to convert to ex-Tregs under inflammatory conditions. Additional experiments are needed to examine how SOCS1 or STAT4 is associated with the Treg fate of lineage commitment status. It has recently been reported that a TCR-mediated IKK–NF-κB signaling pathway including Ubc13 maintains the suppressive activity of Tregs by controlling the expression of SOCS1 (15), and the necessity of TCR signaling for Treg-suppressive function was shown to be independent of Foxp3 expression and IL-2 receptor signaling (49). STAT4 has been reported to be a molecule that is crucial for the onset or pathogenesis of lupus (50, 51), and it should be clarified whether STAT4 expression is involved in Treg development or Treg-suppressive functions in lupus.

It has also been shown that committed Tregs are memory Tregs that can re-express Foxp3 and maintain their suppressive function, even if they transiently downregulate Foxp3 expression (9). CNS2 contains binding sites for the transcription factors CREB (8) and STAT5 (52), whose engagement with the TSDR is considered necessary for Foxp3 reinduction in addition to TCR and/or IL-2 signaling (9). However, in our B16/F10 melanoma model, Socs1-deficient ex-Tregs effectively inhibited tumor growth. Using a fate mapping system, we determined that Socs1-deficient Tregs in the tumor lost Foxp3 expression compared with Socs1-sufficient (WT) Tregs. Both Socs1-deficient and Socs1-sufficient Tregs maintained Foxp3 expression in the draining LN. Moreover, in the tumor, it was found that IFN-γ was produced from the Foxp3 population in the Socs1-deficient Tregs. It is assumed that these ex-Tregs were retained to produce tumor-suppressive cytokines. For plasticity of Socs1-deficient Tregs, IL-12 produced by APCs was needed to downregulate Foxp3 expression in conjunction with activation of STAT4. IL-6, which is an inflammatory cytokine produced by APCs, has also been reported to downregulate Foxp3 expression (46, 47, 53). Understanding how SOCS1 is associated with the induction and/or maintenance of the conversion of Tregs to Foxp3IFN-γ+ ex-Tregs is fundamental to the management of immunological settings for autoimmunity or cancer.

Our studies demonstrated that SOCS1 is indispensable for the suppression of Tregs in vivo, which is consistent with previously reported results from T cell–specific SOCS1-deficient mice (16). Excessive activation of STAT4 in Tregs under the inflammatory conditions in which APCs are highly activated is kept in check by SOCS1 to prevent the deviation of activated Tregs into Th1-like cells. Our findings also raise the intriguing possibility that the upregulation of SOCS1 in Tregs at appropriate levels reinforces Treg functions because SOCS1 may protect Tregs from the harmful effects of inflammatory cytokines that accelerate Treg plasticity. These findings may improve Treg therapy for autoimmune diseases and organ transplantation. Furthermore, the knockdown of SOCS1 in Tregs may be helpful in providing stronger anti-tumor immunity by reprogramming Tregs into effector cells.

We thank T. Takimoto, T. Kobayashi, S. Hidano, and N. Shiino for providing technical assistance and Y. Ushijima for manuscript preparation.

This work was supported by special Grants-in-Aid from the Ministry of Education, Culture, Sports, Science and Technology of Japan, the Takeda Science Foundation, SENSHIN Medical Research Foundation, Takeshi Nagao Intractable Diseases Research Fund, and Japan Rheumatism Foundation (to R.T.) and by special Grants-in-Aid from the Ministry of Education, Culture, Sports, Science and Technology of Japan and the Japan Society for the Promotion of Science (to A.Y.).

The online version of this article contains supplemental material.

Abbreviations used in this article:

CNS

conserved noncoding DNA sequence

LN

lymph node

nTreg

natural regulatory T cell

SOCS1

suppressor of cytokine signaling 1

tdRFP

tandem-dimer red fluorescent protein

Treg

regulatory T cell

TSDR

Treg-specific demethylated region

WT

wild-type.

1
Komatsu
,
N.
,
M. E.
Mariotti-Ferrandiz
,
Y.
Wang
,
B.
Malissen
,
H.
Waldmann
,
S.
Hori
.
2009
.
Heterogeneity of natural Foxp3+ T cells: a committed regulatory T-cell lineage and an uncommitted minor population retaining plasticity.
Proc. Natl. Acad. Sci. USA
106
:
1903
1908
.
2
Zhou
,
X.
,
S. L.
Bailey-Bucktrout
,
L. T.
Jeker
,
C.
Penaranda
,
M.
Martínez-Llordella
,
M.
Ashby
,
M.
Nakayama
,
W.
Rosenthal
,
J. A.
Bluestone
.
2009
.
Instability of the transcription factor Foxp3 leads to the generation of pathogenic memory T cells in vivo.
Nat. Immunol.
10
:
1000
1007
.
3
Hsieh
,
C. S.
,
Y.
Zheng
,
Y.
Liang
,
J. D.
Fontenot
,
A. Y.
Rudensky
.
2006
.
An intersection between the self-reactive regulatory and nonregulatory T cell receptor repertoires.
Nat. Immunol.
7
:
401
410
.
4
Stritesky
,
G. L.
,
S. C.
Jameson
,
K. A.
Hogquist
.
2012
.
Selection of self-reactive T cells in the thymus.
Annu. Rev. Immunol.
30
:
95
114
.
5
Ziegler
,
S. F.
2006
.
FOXP3: of mice and men.
Annu. Rev. Immunol.
24
:
209
226
.
6
Komatsu
,
N.
,
K.
Okamoto
,
S.
Sawa
,
T.
Nakashima
,
M.
Oh-hora
,
T.
Kodama
,
S.
Tanaka
,
J. A.
Bluestone
,
H.
Takayanagi
.
2014
.
Pathogenic conversion of Foxp3+ T cells into TH17 cells in autoimmune arthritis.
Nat. Med.
20
:
62
68
.
7
Floess
,
S.
,
J.
Freyer
,
C.
Siewert
,
U.
Baron
,
S.
Olek
,
J.
Polansky
,
K.
Schlawe
,
H. D.
Chang
,
T.
Bopp
,
E.
Schmitt
, et al
.
2007
.
Epigenetic control of the foxp3 locus in regulatory T cells.
PLoS Biol.
5
:
e38
.
8
Kim
,
H. P.
,
W. J.
Leonard
.
2007
.
CREB/ATF-dependent T cell receptor-induced FoxP3 gene expression: a role for DNA methylation.
J. Exp. Med.
204
:
1543
1551
.
9
Miyao
,
T.
,
S.
Floess
,
R.
Setoguchi
,
H.
Luche
,
H. J.
Fehling
,
H.
Waldmann
,
J.
Huehn
,
S.
Hori
.
2012
.
Plasticity of Foxp3(+) T cells reflects promiscuous Foxp3 expression in conventional T cells but not reprogramming of regulatory T cells.
Immunity
36
:
262
275
.
10
Zhou
,
H.
,
R.
Miki
,
M.
Eeva
,
F. M.
Fike
,
D.
Seligson
,
L.
Yang
,
A.
Yoshimura
,
M. A.
Teitell
,
C. A.
Jamieson
,
N. A.
Cacalano
.
2007
.
Reciprocal regulation of SOCS 1 and SOCS3 enhances resistance to ionizing radiation in glioblastoma multiforme.
Clin. Cancer Res.
13
:
2344
2353
.
11
Hanada
,
T.
,
T.
Kobayashi
,
T.
Chinen
,
K.
Saeki
,
H.
Takaki
,
K.
Koga
,
Y.
Minoda
,
T.
Sanada
,
T.
Yoshioka
,
H.
Mimata
, et al
.
2006
.
IFNgamma-dependent, spontaneous development of colorectal carcinomas in SOCS1-deficient mice.
J. Exp. Med.
203
:
1391
1397
.
12
Tanaka
,
K.
,
K.
Ichiyama
,
M.
Hashimoto
,
H.
Yoshida
,
T.
Takimoto
,
G.
Takaesu
,
T.
Torisu
,
T.
Hanada
,
H.
Yasukawa
,
S.
Fukuyama
, et al
.
2008
.
Loss of suppressor of cytokine signaling 1 in helper T cells leads to defective Th17 differentiation by enhancing antagonistic effects of IFN-gamma on STAT3 and Smads.
J. Immunol.
180
:
3746
3756
.
13
Lu
,
L.-F.
,
T.-H.
Thai
,
D. P.
Calado
,
A.
Chaudhry
,
M.
Kubo
,
K.
Tanaka
,
G. B.
Loeb
,
H.
Lee
,
A.
Yoshimura
,
K.
Rajewsky
,
A. Y.
Rudensky
.
2009
.
Foxp3-dependent microRNA155 confers competitive fitness to regulatory T cells by targeting SOCS1 protein.
Immunity
30
:
80
91
.
14
Lu
,
L. F.
,
M. P.
Boldin
,
A.
Chaudhry
,
L. L.
Lin
,
K. D.
Taganov
,
T.
Hanada
,
A.
Yoshimura
,
D.
Baltimore
,
A. Y.
Rudensky
.
2010
.
Function of miR-146a in controlling Treg cell-mediated regulation of Th1 responses.
Cell
142
:
914
929
.
15
Chang
,
J.-H.
,
Y.
Xiao
,
H.
Hu
,
J.
Jin
,
J.
Yu
,
X.
Zhou
,
X.
Wu
,
H. M.
Johnson
,
S.
Akira
,
M.
Pasparakis
, et al
.
2012
.
Ubc13 maintains the suppressive function of regulatory T cells and prevents their conversion into effector-like T cells.
Nat. Immunol.
13
:
481
490
.
16
Takahashi
,
R.
,
S.
Nishimoto
,
G.
Muto
,
T.
Sekiya
,
T.
Tamiya
,
A.
Kimura
,
R.
Morita
,
M.
Asakawa
,
T.
Chinen
,
A.
Yoshimura
.
2011
.
SOCS1 is essential for regulatory T cell functions by preventing loss of Foxp3 expression as well as IFN-gamma and IL-17A production.
J. Exp. Med.
208
:
2055
2067
.
17
Rubtsov
,
Y. P.
,
J. P.
Rasmussen
,
E. Y.
Chi
,
J.
Fontenot
,
L.
Castelli
,
X.
Ye
,
P.
Treuting
,
L.
Siewe
,
A.
Roers
,
W. R.
Henderson
Jr.
, et al
.
2008
.
Regulatory T cell-derived interleukin-10 limits inflammation at environmental interfaces.
Immunity
28
:
546
558
.
18
Wang
,
Y.
,
A.
Kissenpfennig
,
M.
Mingueneau
,
S.
Richelme
,
P.
Perrin
,
S.
Chevrier
,
C.
Genton
,
B.
Lucas
,
J.
DiSanto
,
H.
Acha-Orbea
, et al
.
2008
.
Th2 lymphoproliferative disorder of LatY136F mutant mice unfolds independently of TCR-MHC engagement and is insensitive to the action of Foxp3+ regulatory T cells.
J. Immunol.
180
:
1565
1575
.
19
Luche
,
H.
,
O.
Weber
,
T.
Nageswara Rao
,
C.
Blum
,
H. J.
Fehling
.
2007
.
Faithful activation of an extra-bright red fluorescent protein in “knock-in” Cre-reporter mice ideally suited for lineage tracing studies.
Eur. J. Immunol.
37
:
43
53
.
20
Fontenot
,
J. D.
,
J. P.
Rasmussen
,
L. M.
Williams
,
J. L.
Dooley
,
A. G.
Farr
,
A. Y.
Rudensky
.
2005
.
Regulatory T cell lineage specification by the forkhead transcription factor foxp3.
Immunity
22
:
329
341
.
21
Dominguez-Villar
,
M.
,
C. M.
Baecher-Allan
,
D. A.
Hafler
.
2011
.
Identification of T helper type 1-like, Foxp3+ regulatory T cells in human autoimmune disease.
Nat. Med.
17
:
673
675
.
22
Sekiya
,
T.
,
I.
Kashiwagi
,
N.
Inoue
,
R.
Morita
,
S.
Hori
,
H.
Waldmann
,
A. Y.
Rudensky
,
H.
Ichinose
,
D.
Metzger
,
P.
Chambon
,
A.
Yoshimura
.
2011
.
The nuclear orphan receptor Nr4a2 induces Foxp3 and regulates differentiation of CD4+ T cells.
Nat. Commun.
2
:
269
.
23
Sekiya
,
T.
,
I.
Kashiwagi
,
R.
Yoshida
,
T.
Fukaya
,
R.
Morita
,
A.
Kimura
,
H.
Ichinose
,
D.
Metzger
,
P.
Chambon
,
A.
Yoshimura
.
2013
.
Nr4a receptors are essential for thymic regulatory T cell development and immune homeostasis.
Nat. Immunol.
14
:
230
237
.
24
Mantel
,
P.-Y.
,
N.
Ouaked
,
B.
Rückert
,
C.
Karagiannidis
,
R.
Welz
,
K.
Blaser
,
C. B.
Schmidt-Weber
.
2006
.
Molecular mechanisms underlying FOXP3 induction in human T cells.
J. Immunol.
176
:
3593
3602
.
25
Ohkura
,
N.
,
M.
Hamaguchi
,
H.
Morikawa
,
K.
Sugimura
,
A.
Tanaka
,
Y.
Ito
,
M.
Osaki
,
Y.
Tanaka
,
R.
Yamashita
,
N.
Nakano
, et al
.
2012
.
T cell receptor stimulation-induced epigenetic changes and Foxp3 expression are independent and complementary events required for Treg cell development.
Immunity
37
:
785
799
.
26
Ohkura
,
N.
,
Y.
Kitagawa
,
S.
Sakaguchi
.
2013
.
Development and maintenance of regulatory T cells.
Immunity
38
:
414
423
.
27
Zheng
,
Y.
,
S.
Josefowicz
,
A.
Chaudhry
,
X. P.
Peng
,
K.
Forbush
,
A. Y.
Rudensky
.
2010
.
Role of conserved non-coding DNA elements in the Foxp3 gene in regulatory T-cell fate.
Nature
463
:
808
812
.
28
Fu
,
W.
,
A.
Ergun
,
T.
Lu
,
J. A.
Hill
,
S.
Haxhinasto
,
M. S.
Fassett
,
R.
Gazit
,
S.
Adoro
,
L.
Glimcher
,
S.
Chan
, et al
.
2012
.
A multiply redundant genetic switch ‘locks in’ the transcriptional signature of regulatory T cells.
Nat. Immunol.
13
:
972
980
.
29
Rudra
,
D.
,
P.
deRoos
,
A.
Chaudhry
,
R. E.
Niec
,
A.
Arvey
,
R. M.
Samstein
,
C.
Leslie
,
S. A.
Shaffer
,
D. R.
Goodlett
,
A. Y.
Rudensky
.
2012
.
Transcription factor Foxp3 and its protein partners form a complex regulatory network.
Nat. Immunol.
13
:
1010
1019
.
30
Kitoh
,
A.
,
M.
Ono
,
Y.
Naoe
,
N.
Ohkura
,
T.
Yamaguchi
,
H.
Yaguchi
,
I.
Kitabayashi
,
T.
Tsukada
,
T.
Nomura
,
Y.
Miyachi
, et al
.
2009
.
Indispensable role of the Runx1-Cbfbeta transcription complex for in vivo-suppressive function of FoxP3+ regulatory T cells.
Immunity
31
:
609
620
.
31
Ono
,
M.
,
H.
Yaguchi
,
N.
Ohkura
,
I.
Kitabayashi
,
Y.
Nagamura
,
T.
Nomura
,
Y.
Miyachi
,
T.
Tsukada
,
S.
Sakaguchi
.
2007
.
Foxp3 controls regulatory T-cell function by interacting with AML1/Runx1.
Nature
446
:
685
689
.
32
Wu
,
Y.
,
M.
Borde
,
V.
Heissmeyer
,
M.
Feuerer
,
A. D.
Lapan
,
J. C.
Stroud
,
D. L.
Bates
,
L.
Guo
,
A.
Han
,
S. F.
Ziegler
, et al
.
2006
.
FOXP3 controls regulatory T cell function through cooperation with NFAT.
Cell
126
:
375
387
.
33
Huehn
,
J.
,
J. K.
Polansky
,
A.
Hamann
.
2009
.
Epigenetic control of FOXP3 expression: the key to a stable regulatory T-cell lineage?
Nat. Rev. Immunol.
9
:
83
89
.
34
Takimoto
,
T.
,
Y.
Wakabayashi
,
T.
Sekiya
,
N.
Inoue
,
R.
Morita
,
K.
Ichiyama
,
R.
Takahashi
,
M.
Asakawa
,
G.
Muto
,
T.
Mori
, et al
.
2010
.
Smad2 and Smad3 are redundantly essential for the TGF-beta-mediated regulation of regulatory T plasticity and Th1 development.
J. Immunol.
185
:
842
855
.
35
Rudra
,
D.
,
T.
Egawa
,
M. M.
Chong
,
P.
Treuting
,
D. R.
Littman
,
A. Y.
Rudensky
.
2009
.
Runx-CBFbeta complexes control expression of the transcription factor Foxp3 in regulatory T cells.
Nat. Immunol.
10
:
1170
1177
.
36
Yao
,
Z.
,
Y.
Kanno
,
M.
Kerenyi
,
G.
Stephens
,
L.
Durant
,
W. T.
Watford
,
A.
Laurence
,
G. W.
Robinson
,
E. M.
Shevach
,
R.
Moriggl
, et al
.
2007
.
Nonredundant roles for Stat5a/b in directly regulating Foxp3.
Blood
109
:
4368
4375
.
37
Hori
,
S.
2010
.
c-Rel: a pioneer in directing regulatory T-cell lineage commitment?
Eur. J. Immunol.
40
:
664
667
.
38
Muto
,
G.
,
H.
Kotani
,
T.
Kondo
,
R.
Morita
,
S.
Tsuruta
,
T.
Kobayashi
,
H.
Luche
,
H. J.
Fehling
,
M.
Walsh
,
Y.
Choi
,
A.
Yoshimura
.
2013
.
TRAF6 is essential for maintenance of regulatory T cells that suppress Th2 type autoimmunity.
PLoS One
8
:
e74639
.
39
Huynh
,
A.
,
M.
DuPage
,
B.
Priyadharshini
,
P. T.
Sage
,
J.
Quiros
,
C. M.
Borges
,
N.
Townamchai
,
V. A.
Gerriets
,
J. C.
Rathmell
,
A. H.
Sharpe
, et al
.
2015
.
Control of PI(3) kinase in Treg cells maintains homeostasis and lineage stability.
Nat. Immunol.
16
:
188
196
.
40
Rubtsov
,
Y. P.
,
R. E.
Niec
,
S.
Josefowicz
,
L.
Li
,
J.
Darce
,
D.
Mathis
,
C.
Benoist
,
A. Y.
Rudensky
.
2010
.
Stability of the regulatory T cell lineage in vivo.
Science
329
:
1667
1671
.
41
Bailey-Bucktrout
,
S. L.
,
M.
Martinez-Llordella
,
X.
Zhou
,
B.
Anthony
,
W.
Rosenthal
,
H.
Luche
,
H. J.
Fehling
,
J. A.
Bluestone
.
2013
.
Self-antigen-driven activation induces instability of regulatory T cells during an inflammatory autoimmune response.
Immunity
39
:
949
962
.
42
Addey
,
C.
,
M.
White
,
L.
Dou
,
D.
Coe
,
J.
Dyson
,
J. G.
Chai
.
2011
.
Functional plasticity of antigen-specific regulatory T cells in context of tumor.
J. Immunol.
186
:
4557
4564
.
43
Oldenhove
,
G.
,
N.
Bouladoux
,
E. A.
Wohlfert
,
J. A.
Hall
,
D.
Chou
,
L.
Dos Santos
,
S.
O’Brien
,
R.
Blank
,
E.
Lamb
,
S.
Natarajan
, et al
.
2009
.
Decrease of Foxp3+ Treg cell number and acquisition of effector cell phenotype during lethal infection.
Immunity
31
:
772
786
.
44
Tang
,
Q.
,
J. Y.
Adams
,
C.
Penaranda
,
K.
Melli
,
E.
Piaggio
,
E.
Sgouroudis
,
C. A.
Piccirillo
,
B. L.
Salomon
,
J. A.
Bluestone
.
2008
.
Central role of defective interleukin-2 production in the triggering of islet autoimmune destruction.
Immunity
28
:
687
697
.
45
Wan
,
Y. Y.
,
R. A.
Flavell
.
2007
.
Regulatory T-cell functions are subverted and converted owing to attenuated Foxp3 expression.
Nature
445
:
766
770
.
46
Xu
,
L.
,
A.
Kitani
,
I.
Fuss
,
W.
Strober
.
2007
.
Cutting edge: regulatory T cells induce CD4+CD25-Foxp3- T cells or are self-induced to become Th17 cells in the absence of exogenous TGF-beta.
J. Immunol.
178
:
6725
6729
.
47
Yang
,
X. O.
,
R.
Nurieva
,
G. J.
Martinez
,
H. S.
Kang
,
Y.
Chung
,
B. P.
Pappu
,
B.
Shah
,
S. H.
Chang
,
K. S.
Schluns
,
S. S.
Watowich
, et al
.
2008
.
Molecular antagonism and plasticity of regulatory and inflammatory T cell programs.
Immunity
29
:
44
56
.
48
Yurchenko
,
E.
,
M. T.
Shio
,
T. C.
Huang
,
M.
Da Silva Martins
,
M.
Szyf
,
M. K.
Levings
,
M.
Olivier
,
C. A.
Piccirillo
.
2012
.
Inflammation-driven reprogramming of CD4+ Foxp3+ regulatory T cells into pathogenic Th1/Th17 T effectors is abrogated by mTOR inhibition in vivo.
PLoS One
7
:
e35572
.
49
Levine
,
A. G.
,
A.
Arvey
,
W.
Jin
,
A. Y.
Rudensky
.
2014
.
Continuous requirement for the TCR in regulatory T cell function.
Nat. Immunol.
15
:
1070
1078
.
50
Deng
,
Y.
,
B. P.
Tsao
.
2010
.
Genetic susceptibility to systemic lupus erythematosus in the genomic era.
Nat. Rev. Rheumatol.
6
:
683
692
.
51
Sestak
,
A. L.
,
B. G.
Fürnrohr
,
J. B.
Harley
,
J. T.
Merrill
,
B.
Namjou
.
2011
.
The genetics of systemic lupus erythematosus and implications for targeted therapy.
Ann. Rheum. Dis.
70
(
Suppl. 1
):
i37
i43
.
52
Burchill
,
M. A.
,
J.
Yang
,
C.
Vogtenhuber
,
B. R.
Blazar
,
M. A.
Farrar
.
2007
.
IL-2 receptor beta-dependent STAT5 activation is required for the development of Foxp3+ regulatory T cells.
J. Immunol.
178
:
280
290
.
53
Tsuji
,
M.
,
N.
Komatsu
,
S.
Kawamoto
,
K.
Suzuki
,
O.
Kanagawa
,
T.
Honjo
,
S.
Hori
,
S.
Fagarasan
.
2009
.
Preferential generation of follicular B helper T cells from Foxp3+ T cells in gut Peyer’s patches.
Science
323
:
1488
1492
.

The authors have no financial conflicts of interest.

Supplementary data