Introduction

The exposure to environmental stimuli causes mucosal surfaces to develop local mechanisms to maintain homeostasis at steady-state conditions, all-the-while mounting strong immune responses when required. CD4+Foxp3+ regulatory T (TREG) cells are critical mediators of peripheral immune self-tolerance and modulators of immune responses directed towards a spectrum of self and non-self antigens.1,2 Foxp3 is a lineage-specifying transcription factor essential for the development and function of TREG cells. Induction and sustained expression of Foxp3 defines the core transcriptional programme of TREG cells, establishing their phenotype and suppressive function, as well as regulating key cell-intrinsic, homeostatic processes, including cytokine signalling pathways.3 Inheritable mutations of the foxp3 gene result in the onset of a severe autoimmune disease in scurfy mice and the immunodysregulation polyendocrinopathy enteropathy X-linked syndrome (IPEX) in humans.4,5

In addition to secondary lymphoid organs, Foxp3+ TREG cells are also abundant in non-lymphoid tissues, particularly mucosal surfaces, where they contribute to maintain immune homeostasis.6,7 Local inflammatory signals can force CD4+ T cells to undergo significant functional plasticity in order to acquire specialised effector functions in situ, and adapt to the evolving nature of immune responses, particularly during infections.8 Growing evidence indicates that Foxp3+ TREG cells can also acquire tissue-specific adaptations that promote their local homeostasis and function.9,10 We and others have previously shown that the stability of Foxp3 expression and TREG cell function is a dynamic process dictated by the inflammatory environment. Settings of lymphopenia, chronic inflammation, or infections favour the lineage and functional plasticity of Foxp3+ TREG cells by downregulating Foxp3 expression, compromising suppressive function, and reprogramming them into IFNγ- or IL17-producing-like exFoxp3+ T cells which, in turn, promote potent effector immune responses in the host.11 Recently, studies have shown that Foxp3+ TREG cells can also co-express lineage-defining transcription factors of their target TEFF cells such as T-bet, RORγT or GATA3, allowing TREG cells to localise and suppress the associated CD4+ T-helper (Th) cell immune responses.12,13,14 For instance, TREG-mediated suppression of Th17 responses is dependent on CCR6, a characteristic chemokine receptor of Th17 cells, and STAT3, a signal transducer and activation of transcription downstream of IL-6 and IL-23.15 Moreover, TREG cells from the lamina propria adapt to the local microbial environment of the gut by expressing the Th17-associated transcription factor RORγT, a feature that confers suppressive function during gut-specific immune responses.16 Similarly, CXCR3+T-bet+ TREG cells suppress Th1 cell responses,17,18 while GATA3+ TREG cells were identified in the lung and gut of healthy mice19 and accumulated more readily during Th2-mediated inflammation.20 Thus, inflammatory cues can modulate the epigenetic and transcriptional landscape of Foxp3+ TREG cells, forcing them to specialise their functions for a context-dependent adaptation to evolving immune responses.

By applying an unbiased screen to uncover the mechanistic events leading to a downregulation of Foxp3 expression in TREG cells, we identify a unique role of the IL-1 family of cytokines in modulating the reprogramming potential of TREG cells during inflammation. We show that cell surface expression of the IL-33R (ST2, Il1rl1) identifies a subset of functionally stable Helios+Foxp3+ TREG cells that are resistant to plasticity and loss of suppressive function, whereas IL1R1 (Il1r1) expression identifies RORγT+ Helios TREG cells that acquire a Th17 cell phenotype and fail to suppress. While a Th17 polarizing milieu induces IL-1R1 expression on TREG cells, IL-33 favours the stability of the Helios+ST2+ TREG cell phenotype in similar conditions. Importantly, ST2+ TREG cells accumulate early in the lungs upon infection with influenza virus or the fungal pathogen Cryptococcus neoformans, and segregates suppressive from inflammatory TREG cells throughout the course of infection. Abrogation of IL-33, but not IL-1 signalling, in TREG cells compromises their suppressive function and fuels inflammatory responses in vivo. Importantly, lack of IL-1 signalling alters the dynamics of ST2+ and RORγT+ Foxp3+ TREG populations in the lung, and augments susceptibility to fungal infection. Overall, we show that IL-1 and IL-33 exert opposite roles in controlling the adaptation and functional specialisation of Foxp3+ TREG cells at mucosal surfaces.

Results

Differential IL-33R (ST2) and IL1R1 expression distinguishes functionally stable from unstable TREG cells

To delineate the mechanistic basis of functional plasticity of TREG cells in vivo, we exploited a Foxp3+ TREG cell transfer model in lymphopenic hosts that we previously described.11 Specifically, Foxp3+ TREG (GFP+) cells from Ly5.1+ Foxp3GFPki reporter mice were FACS-purified and transferred into TCRβ-/- Ly5.2+ recipient mice. After 21 days, TREG cells that preserved Foxp3 expression (GFP+, stable TREG [sTREG]) and TREG cells that lost Foxp3 expression (GFP-, unstable TREG, termed exTREG cells) were isolated based on GFP expression from donor Ly5.1+ CD4+ splenocytes, and subjected to transcriptional microarray analysis. Donor Ly5.1+CD4+GFP- TEFF cells were also FACS-isolated at day 21 from mice that received only TEFF (Ly5.1+CD4+GFP) cells, thus devoid of any Foxp3-expressing cells at day 0 (Fig. 1a). Freshly sorted CD4+ GFP and GFP+ T cells from Foxp3GFPki reporter mice were also included in the microarray analysis as control. Hierarchical cluster analysis of the top 654 genes (p < 0.05) identified between TREG, exTREG and activated TEFF cells reveals a closer relationship between exTREG cells and activated TEFF cells compared to stable TREG cells (Fig. 1b). We observed that the majority of genes that are significantly modulated upon transfer (>1 log2 fold change) are not shared between sTREG and exTREG cells (Fig. 1c). We observed that exTREG cells down-regulated some of the major TREG signature genes (Ctl4, IL-10 and Nrp1) and up-regulated genes associated with Th1 and Th17 T cells (Tbx21 and  il23r; Fig. 1d). Interestingly, the mRNA levels of IL-33 receptor (Il1rl1, IL-33R, ST2) were significantly higher in TREG cells that maintained Foxp3 (sTREG) compared to exTREG and TEFF cells. In contrast, mRNA expression of IL-1 receptor (Il1r1) was significantly reduced in stable TREG cells when comparing with exTREG (Fig. 1e). Analysis of cell surface-bound ST2 expression by flow cytometry confirmed findings of increased mRNA levels, demonstrating that a subset of sTREG cells readily expressed ST2 after transfer (Fig. 1f). In order to further confirm these results, we transferred total CD4+ T cells from the fate-mapping Foxp3GFP-Cre X Rosa26-lox-stop-lox tdTomato mice21 into TCRβ Ly5.1+ mice (Fig. 1g, h; S1). Again, we observed a difference in the surface expression of ST2 between stable (tdTomato+GFP+) vs. exTREG cells (tdTomato+ GFP) in the spleen (Fig. 1g, h; S1). We also observed a tendency towards higher expression of the IL1R1 in the exTREG when compared to sTREG cells (S1). These results illustrate that differential expression of ST2 and IL1R1 segregates distinct subsets of TREG cells with divergent effector outcomes.

Fig. 1
figure 1

Identification of ST2 as a marker of functionally stable TREG cells. a CD4+GFP+Ly5.1+ T cells from Foxp3GFPki C57BL/6 mice were FACS-sorted and adoptively transferred into TCRβ−/− mice. The cells were harvested 21 days post-transfer based on GFP expression and microarray analysis (see Methods) was performed (splenocytes of TCRβ−/− mice were isolated and pooled before sorting the distinct populations; Two separate T cell transfers were performed, the cells isolated and processed individually for microarray analysis). The top 654 genes that varied (p < 0.05 cut-off) between the groups are shown. b Transcriptional signatures of stable TREG (sTREG), exTREG and TEFF cells isolated at day 21 post transfer. c Venn diagram showing the distribution of differentially expressed genes (cut-off of >1 log2 fold change) between exTREG and sTREG relative to TREG (CD4+GFP+) from day 0. d Highest ranking (>2 fold change) genes in exTREG and sTREG at day 21 relative to TREG cells from day 0. e Pearson correlation of log2 fold change gene expression directly comparing sTREG cells and exTREG cells identifying the differential expression of Il1r1 (blue) and Il1rl1 (red). Dotted lines show ±1 log2 fold change. f Representative flow cytometry profile of surface expression of IL1RL1 (IL-33R, ST2) (top) and IL1R1 (bottom) in CD4+CD45.1+ T cells in the spleen at day 21 after transfer (N = 4); Controls are fluorescence minus one (FMO). g, h CD4+ T cells from Foxp3GFP-CRE Rosa26RFP fate-tracking mice were sorted and adoptively transferred (i.p.) into a TCRβ Ly5.1+ mouse, and examined 10 days post-transfer (N = 4). g (Left) Representative plots showing frequencies of GFP+RFP+ (Foxp3+; sTREG) and GFP-RFP+ (exTREG) cells in CD4+CD45.2+ (right panel). Cut-off population of GFPhi and GFPlo T cells. h Representative histograms of expression levels of ST2 (left panel) and IL1R1 (right panel) between GFPhi (red) and GFPlo (blue) CD4+CD45.2+ RFP+ T cells

ST2 expression delineates stable TREG cells from IFNγ- and IL-17A-producing TREG cells during viral infection

We hypothesised that ST2+ TREG cells increase in an inflammatory microenvironment that harbours elevated levels of IL-33, an alarmin induced by infectious pathogens particularly at mucosal surfaces.22 To test this hypothesis, we examined the functional dynamics of ST2+ TREG cells in the context of a type-1 immune-driven respiratory viral infection in vivo. Infection with Influenza A H1N1/PR8 induces pulmonary expression and release of IL-33 early during the course of disease.23,24 Infected mice demonstrated an increase in pulmonary CD4+ T cells that coincided with the peak of weight loss (Fig. 2a). ST2+ TREG cell numbers in the lungs were found to increase starting by day 4 and to peak at day 10 post-infection (Fig. 2b–d). Interestingly, the increase in ST2+ TREG cells during the course of infection was seen only in lungs of infected mice (Fig. 2e), suggesting that IL-33 acts locally in the lung mucosal environment. Since Foxp3+ TREG cells homing to inflammatory sites have the potential to adopt an inflammatory phenotype by co-expressing RORγT as well as IL-17A or IFNγ,11,25 we determined whether ST2 expression by TREG cells correlated with expression of these inflammatory phenotypes. We observed that at peak infection the production of IFNγ and IL-17A originated exclusively from pulmonary ST2 TREG and ST2 TEFF cells (Fig. 2f–h). Similarly, the proportions and absolute cell numbers of IFNγ and IL-17A secreting Foxp3+ TREG cells increased during peak infection and were restricted to the ST2 cell subset (Fig. 2g, h). These data show that IL-33-responsive (ST2+) TREG cells do not secrete inflammatory cytokines and represent a stable population of TREG cells during influenza virus infection.

Fig. 2
figure 2

The ST2 receptor segregates stable TREG cells from pro-inflammatory IFNγ and IL-17A-producing TREG cells in vivo. a CD4+ T cells peak in the lung by day 4 post-infection with Influenza A PR8/H1N1. C57BL/6 mice were infected with 20 pfu of PR8/H1N1 intra-nasally and necropsy was performed at days 0, 4, 10 and 17 post-infection. Representative of 4 distinct experiments. b ST2 expression (red quadrant) is selectively increased in CD4+Foxp3+ T cells during the course of an Influenza A infection. (N = 5). c, d TREG cells and ST2+ TREG cells accumulate in the lung and peak at day 10 after infection. (One way ANOVA. ***p < 0.001; **p < 0.01). e The increase in ST2+ TREG cells at day 10 post-infection is conscribed to the lung. f Representative flow cytometry plots of Foxp3+ (left) and Foxp3 (right) CD4+ T cells in the lungs of infected mice at day 10 post-infection. g, h ST2 (blue), but not ST2+ (red), TREG cells produce IFNγ or IL17-A at day 10 post-infection. Student t-test. *p < 0.05; **p < 0.01; ***p < 0.001

IL-1 inhibits ST2 expression on Helios+ TREG cells

To further detail the nature and origin of ST2+ TREG cells, we assessed their phenotype in secondary lymphoid organs (pLN) as well as mucosal tissues (colon) of naïve C57BL/6 mice. We observed a significant frequency of ST2+ TREG cells in the colonic lamina propria that is not observed in pLN (Fig. 3a). At steady state, ST2+ TREG cells possess a memory profile (S2), a result consistent with a study by Schiering et al.19 Spleen-derived ST2+ TREG cells consistently expressed high levels of Nrp1, Helios and TIGIT, a phenotype reflecting thymic origin (Fig. 3b). Furthermore, thymic-derived CD4SP Foxp3-GFP+ T cells upregulate the ST2 receptor,26 and also maintain high levels of Helios expression (S3A-B). We confirmed that IL-33 was sufficient to induce the expression of ST2 and the transcription factor GATA3 in TREG cells (S3C).19,26 Moreover, this induction in ST2 expression correlated with the accumulation of Helios+ TREG cells in IL-33 treated cultures (Fig. 3c, d), and an increase in CD25 and Foxp3 expression in ST2+ TREG cells (Fig. 3e, f). As IL-33 enhanced the proliferative capacity of TREG cells,27 but did not affect the suppressive ability of TREG cells in vitro (S4), we then hypothesised that the specific accumulation of Helios+ TREG cells could lead to an overall increase in a suppressive environment at the mucosa, as Helios was recently associated with increased suppressive function in TREG cells.28 Interestingly, upon transfer of GFP+ (Foxp3+) T cells in TCRβ−/− C57BL/6 recipient mice, stable Helios+ TREG cells expressed ST2 (Fig. 3g, green panel), whereas Helios TREG cells showed increased IL1R1 expression (Fig. 3g, orange panel). Analysis of ST2 expression on de novo generated GFP+ (pTREG) cells from mice that received only GFP T cells revealed that pTREG cells, in contrast to TREG cells, fail to upregulate the ST2 receptor (Fig. 3h). This was further confirmed in vitro, as TGFβ generated iTREG cells did not upregulate ST2 in the presence of IL-33 (S5).

Fig. 3
figure 3

IL-1 inhibits ST2 expression on Helios+ TREG cells. a Representative flow cytometry profile of ST2 expression in TREG cells from pLN and colon of naïve C57BL/6 mice (N = 5 mice). b Expression levels of Neuropilin 1, Helios and TIGIT by ST2+ (red) and ST2 (blue) TREG cells from the spleen of naïve C57BL/6 mice were analysed by flow cytometry. Light grey histograms represent staining with isotype control. Representative of 3 mice, repeated in 3 separate experiments. c, d CD4+ T cells from spleens of WT and ST2−/−BALB/c mice were FACS-isolated and activated by plate-bound α-CD3 and α-CD28 in the presence of IL-2 (50 U/ml) and/or IL-33 (10 ng/ml) for 72 h. Expression levels of Foxp3, ST2 and Helios were analysed by flow cytometry, and d frequencies of Foxp3+Helios+ TREG cells were determined. One-way ANOVA; *p < 0.05. Triplicates. Representative of three distinct experiments. e, f Mean fluorescence intensity of CD25 and Foxp3 expression of ST2+ (red) and ST2 (blue) TREG cells in (c). Student t-test. ***p < 0.001. (triplicates; N = 5). g ST2+ TREG cells segregate with Helios expression in the TREG adoptive transfer model. CD4+GFP+ T cells were transferred as described in Fig. 1a and analysed at day 21. Expression levels of ST2 and IL1R1 in Helios+Foxp3+ (green), HeliosFoxp3+ (blue) and HeliosFoxp3 (red). (N = 4 per group; Representative of 3 experiments). h Representative flow cytometry plots of ST2 and GFP (Foxp3) expression by the transferred GFPneg and GFPpos (Ly5.1+ T cells) in the spleen at day 21. i Peripherally -induced TREG cells (pTREG) do not induce ST2 expression after transfer. Student t-test. ***p < 0.001. (N = 4 per group; Representative of 3 experiments). j–l IL1R1−/− mice show increased frequencies of ST2+ TREG cells in the lung at the steady state. j Representative flow cytometric analysis of Helios vs. ST2 expression in the lungs of naïve (WT), IL1R1−/− or ST2−/− BALB/c mice. k Frequencies of ST2+ TREG cells in the lung. l Mean fluorescence intensities (MFI) of ST2 expression among ST2+ TREG cells. One-way ANOVA; Tukey correction. ***p < 0.001. (N = 3–6 per group; Representative of 3 separate experiments). m Representative flow cytometry plots of the expression of ST2 and IL1R1 on CD4+Foxp3+ TREG cells isolated from the lungs of BALB/c mice, and (N) Mean Fluorescence intensity (MFI) of IL1R1 on ST2+ (red) and ST2 (blue) TREG cells. Paired Student’s t-test; ***p < 0.01. Representative of three distinct experiments (N = 5 mice per experiment)

As IL1R1 was preferentially expressed in unstable TREG cells with reprogramming potential, we further assessed ST2 expression in T cells from mice lacking IL1R1 (IL1R1−/−; Fig. 3j–l). Importantly, we observed an increase in the frequencies of ST2+ Helios+ TREG cells at the steady-state in the lungs of IL1R1−/− mice when compared to WT mice (Fig. 3j, k). Moreover, we show that ST2+ TREG cells from IL1R1−/− mice expressed higher levels (MFI) of ST2 than their counterparts from WT mice (Fig. 3l) suggesting that IL-1 inhibits the expression of ST2 in TREG cells. Finally, we show that ST2 and IL1R1 are differentially expressed among TREG cells at the resting state in the lung of BALB/c mice (Fig. 3m, n). Overall, these results illustrate that subsets of TREG cells possess the ability to respond to either IL-1 or IL-33 at mucosal sites, and that IL-1 signalling regulates ST2 expression in TREG cells.

Absence of IL-33-, but not IL-1-, signalling converts Helios+TREG cells to a pro-inflammatory phenotype that fuels intestinal inflammation

To determine whether IL-1 or IL-33 signalling affected the stability and suppressive function of TREG cells in vivo, we exploited the CD4+ TEFF cell-induced model of intestinal inflammation (colitis). Specifically, we transferred TEFF cells alone (Thy1.1+, CD90.1+) or in the presence of WT, ST2−/− or IL1R1−/− TREG cells (Thy1.2+, CD90.2+) into SCID/beige mice and monitored the dynamics of TREG/TEFF responses relative to colitis development (Fig. 4a). In contrast to mice receiving TEFF cells alone (Thy1.1+), all mice receiving TREG cells did not display significant weight loss by day 28 post-transfer (Fig. 4b). SCID/beige mice that received ST2−/− TREG cells displayed a higher colitis score than the ones with WT TREG, which confirmed the results from Schiering and colleagues.19 Strikingly, we show that mice that received IL1R1−/− TREG effectively prevented colitis onset by day 28 (Fig. 4c). Concomitantly, mice co-transferred with ST2−/− TREG cells showed significantly increased numbers of CD11b+Ly6G+ neutrophils in the colon compared to mice receiving WT or IL1R1−/− TREG cells (Fig. 4d). In addition, IL1R1−/−, but not ST2−/−, TREG fully inhibited the accumulation of IL17A-producing TEFF cells in the colon highlighting the lack of suppressive ability of TREG cells in the colon when IL-33, but not IL-1, signalling is abrogated in TREG cells (Fig. 4e). Moreover, mice co-transferred with IL1R1−/− TREG cells show enhanced frequencies of Foxp3+ TREG cells in the colon and mesLN, but not in the spleen, compared to mice that received WT or ST2−/− Foxp3+ TREG cells (Fig. 4f; S6). IL1R1−/− TREG cells expressed higher levels of ST2, and this coincided with increased Helios expression in the colon (Fig. 4g, h). Consistently, mice that received IL1R1−/− TREG cells also showed increased numbers of ST2+ TREG cells and reduced numbers of RORγT+IL-17A+TREG cells, compared to mice that received WT or ST2−/− TREG cells (Fig. 4i, j). Strikingly, a greater number of IL-17A producing TREG cells that lost Foxp3 expression (exTREG) were found in mice that received ST2−/− TREG cells in contrast of recipients of WT or IL1R1−/− TREG cells (Fig. 4k). Importantly, the increased numbers of colonic neutrophils positively correlated with the increased frequencies of RORγT+IL17A+ TREG cells in all groups (Fig. 4l). Finally, we assessed if we could prevent TREG cell reprogramming in vivo, as determined by the production of IL17A and expression of RORγT in TREG cells by IL-33 supplementation. To this end, as described in Fig. 1, we adoptively transferred 2 × 105 Ly5.1+CD4+GFP+ (Foxp3+) TREG cells i.v. into TCRβ−/− mice, and injected rIL-33 i.p. (100 ng/mouse) every 48 h. 14 days after the TREG transfer, we observed a significant (p < 0.05) reduction in the frequencies of IL17A+ T cells and an increase in the accumulation of IL17A+ exTREG cells (S7A-B). Concomitantly, we observed a trend towards reduced expression of the transcription factor RORγT in Foxp3+ T cells and a significant increase in ST2 expression (S7C-D). These results highlight that IL-1 and IL-33 exert opposite functions in controlling adaptation and functional specialisation of TREG cells during intestinal inflammation.

Fig. 4
figure 4

Absence of IL-33-, but not IL-1-, signalling converts Helios+ TREG cells to a pro-inflammatory phenotype that fuels intestinal inflammation. a SCID/beige mice received CD4+CD45RBhiCD25Thy1.1+ effector T cells (TEFF, 4 × 105) either alone or with CD4+CD25hi TREG cells (2×105) from WT, ST2−/−, or IL1R1−/− BALB/C mice. Necropsy was done at day 28 post-transfer. Pooled results are shown from three separate experiments. Where applicable, the results were compiled using a common denominator in each experiment (average of the WT). b Mice that received TEFF alone show weight loss by day 28 post-transfer. c Mean colitis score of each mouse (see Methods). Five blinded observers. One-way ANOVA. **p < 0.01. d Relative number of cells (to the average of WT) of CD11b+Ly6G+ cells in the colon of mice, assessed by flow cytometry. e Relative number of cells (to the average of WT) of Th17 cells (CD4+IL-17A+ amongst Thy1.1+ cells) assessed by flow cytometry. One-way ANOVA. ***p < 0.001; **p < 0.01; *p < 0.05. f IL1R1−/− TREG cell show less Foxp3 loss in the colon after transfer. Two-way ANOVA. (Tukey) **p < 0.01. g IL1R1−/− TREG cells co-express Helios and ST2. h ST2-expressing CD90.2+ TREG cells are Helios+. i A higher frequency of IL1R1−/− TREG cells express ST2 compared to WT TREG. Two-way ANOVA. Tukey correction. ***p < 0.001. j CD90.2+ (Thy1.2+) CD4+Foxp3+ TREG cells from ST2−/− donors accumulate as RORγT+ in the colon of the mice (relative to the average number of WT). One-way ANOVA. **p < 0.01. k exTREG cells originating from ST2−/− TREG cells accumulate as IL17A+ T cells in the colon of mice (relative to the average number of WT). l The numbers of Th17-like exTREG cells correlate with increased neutrophil accumulation in the colon of mice. Pearson correlation analysis. Representative of three separate experiments

Th17 polarizing conditions induce IL-1R1 expression on TREG cells and promote their proliferation

Our results show that lack of IL-33 signalling in TREG cells compromises their function, while IL-1R1 deficiency favours TREG cell responses highlighting the inhibitory role of IL-1 for the stability and function of TREG cells. We hypothesised that IL-1R1 expression could promote RORγT expression in TREG cells. We then examined the effects of IL-1β on the phenotype and function of TREG cells in vitro by establishing a mixed culture of Ly5.2+ TREG and Ly5.1+ TEFF cells under neutral or Th17 differentiated conditions (Fig. 5a). Indeed, under Th17 polarizing conditions, Foxp3+ TREG cells readily express RORγT, which is undetectable when cultured in control medium (Fig. 5b). Moreover, RORγT+Foxp3+ T cells expressed higher levels of IL-1R1 than medium-treated TREG and RORγT+ TEFF cells (Fig. 5c, d). Inclusion of IL-1β to the Th17 polarizing conditions was critical for enhanced proliferation and survival of TREG cells in vitro (Fig. 5e, f). Consistently, Anakinra, a potent IL-1R antagonist (Kineret®), significantly blocked the proliferation of TREG cells in Th17 polarizing conditions, suggesting a specific role for IL-1β in the expansion and/or survival of RORγT+ TREG cells (Fig. 5e, f; S8). This feature was not due to an indirect effect of IL-1β on TEFF cells, as replacing WT with IL1-R1−/− TREG in this assay impaired the proliferation of these cells upon IL-1β stimulation (S8C-D). Furthermore, we observed that RORγT expression was highest in Helios TREG cells, which coincided with the expression of IL-1R1 (S8B). Finally, IL-1R1+ TREG cells displayed a diminished suppressive ability on in vitro differentiated Th17 cells in the presence of IL-1β (Fig. 5g), an observation not due to a reduction in the frequency of TREG in this culture (S9). Thus, Th17 polarizing conditions skew the functional adaptation of TREG cells and modulate their responsiveness to IL-1β.

Fig. 5
figure 5

Th17 polarizing conditions induce IL1-R1 expression on TREG cells, favour their proliferation but hinder their suppressive ability. Ly5.2+CD4+GFP+ TREG cells were FACS-sorted from C57BL/6 Foxp3GFPki mice and co-cultured (1:2 ratio) with Ly5.1+CD4+GFP- (TEFF) sorted from similar mice, α-CD3 (1 μg/ml) and irradiated APCs (CD4neg fraction from the Ly5.2+) for 72 h. Representative of five experiments in triplicates. a Representative flow cytometry plot of the gating strategy. b Th17 polarizing conditions [IL-6 (10 ng/ml) + TGFβ (1 ng/ml)], not IL-1β (25 ng/ml), upregulate RORγT in CD4+Foxp3+ T cells in vitro. One way ANOVA. Tukey correction. ***p < 0.001. c RORγT is co-expressed with IL-1R1 on TREG cells in Th17 polarizing conditions. d RORγT+ Foxp3+ T cells express the IL-1R1 receptor upon Th17 polarization to a greater extent than RORγT+ TEFF cells. e IL-1β enhances the proliferation of TREG cells in Th17 polarizing conditions. Ly5.2+CD4+GFP+ T cells were stained with V450 proliferation dye prior to culture. ANAKINRA (Kineret®). TREG in medium (blue), in Th17 polarizing conditions (red), in Th17 + IL-1β (green) or in Th17 +  IL-1β + Anakinra (100 μg/ml) (grey). f CD4+Foxp3+Ly5.2+ T cell counts after 72 h. One way ANOVA, Tukey correction. *p < 0.05; **p < 0.01; ***p < 0.001. g IL-1β signalling enhanced proliferation of TEFF (Ly5.2+) cells even in the presence of TREG cells at a 1:2 ratio. CD4+Foxp3-Ly5.1+ TEFF cells alone (white), in the presence of TREG at 1:2 ratio (blue), in Th17 polarizing conditions (red) or in the presence of IL-1β (green)

IL-33 favours the stability of Helios+ST2+ TREG cells in Th17 polarizing conditions

We showed that ST2+ TREG cells are robust suppressors and refractory to plasticity. We then determined whether ST2+ TREG cells maintain this resistance and their phenotype even in Th17 polarizing conditions. To this end, we TCR-activated ST2+ and ST2- TREG cell subsets in the presence or absence of Th17 differentiation conditions (IL-6, TGFβ and IL-1β), and assessed their ability to lose Foxp3 and Helios expression and acquire RORγT and IL-1R1 expression. We confirmed that IL-33 contributed to the maintenance of Helios and ST2 expression by ST2+ TREG cells (Fig. 6a). Interestingly, even when deprived of IL-33, only a small portion of ST2+ TREG cells downregulates ST2 and Helios expression in the span of 72 h and up-regulated RORγT and IL-1R1 (Fig. 6b). This portion of TREG cells was significantly lower than what we observed in ST2 TREG cells. Since ST2+ TREG could resist the inflammatory signals that lead to TREG instability, we wanted to assess the effect of IL-33 in the total pool of TREG cells in Th17 polarizing conditions. When we added increasing amounts of IL-33 to these conditions, we observed an increase in RORγT IL1R1 ST2+ TREG cells in culture (Fig. 6c). In fact, the IL-33-induced expression of ST2 was not affected by Th17 conditions (Fig. 6d). We observed that the ST2+ TREG cells in the culture resisted IL-1R1 and RORγT expression (Fig. 6e, f). Finally, in order to understand the effect of IL-2 on the differentiation of IL-1R1+ TREG cells, we added IL-2 to Th17 polarizing conditions in the presence of saturating amounts of IL-1β and IL-33 (S10). We observed that IL-2 was sufficient to skew the differentiation of Foxp3+ T cells towards stable ST2+ TREG cells even in increasing amounts of IL-6 (S10 A-B). Interestingly, although ST2+ TREG cells expand in the presence of IL-2 and IL-33, IL1R1+ TREG show an increased competitive expansion as they accumulate faster in the presence of IL-6 (S10C). In fact, the expression of IL-1R1 on TREG cells increased in the presence of IL-6 in a dose-dependent manner (S10D). These conditions reveal the dichotomy between ST2+ and IL1R1+ TREG cells and illustrate how distinct  cytokine signals in situ promote the accumulation of stable vs. unstable TREG cells.

Fig. 6
figure 6

IL-33 favours the stability of Helios+ST2+ TREG cells in Th17  polarizing conditions. a, b ST2+ and ST2 CD4+GFP+ (TREG) were isolated from the spleen of Foxp3GFPki mice and plated in the presence of Ly5.1+CD4+GFP (TEFF) sorted from Ly5.1+Foxp3GFPki mice, soluble α-CD3 (1 μg/ml) and irradiated APCs (CD4neg fraction from the Ly5.2+) at a 1:2 ratio for 72 h. Representative of four distinct experiments in triplicates. a TREG cells resist the loss of ST2 and Helios expression under pro-inflammatory conditions in vitro. ST2+ and ST2- CD4+GFP+ T cells from the spleen of Foxp3GFPki were activated in the presence of IL-6 (10 ng/ml), TGFβ (1 ng/ml) and IL-1β (25 ng/ml) for 72 h. b Loss of Helios expression is associated with an increase in RORγT expression in Foxp3+ TREG cells in vitro. One-way-ANOVA; *p < 0.05; **p < 0.01; ***p < 0.001. c–f CD4+GFP+ TREG cells were isolated from the spleen of Foxp3GFPki mice and co-cultured in the presence of CTV-labelled CD4+GFP TEFF cells (1:2 ratio), α-CD3 (1 μg/ml) and irradiated APCs (CD4neg fraction from the Ly5.2+) for 72 h. c, d IL-33 facilitates the up-regulation of the ST2 receptor on TREG cells in Th17 polarizing conditions. One way ANOVA. Tukey correction. **p < 0.01. e, f ST2+ TREG cells resist the expression of RORγT and IL-1R1. Paired Student’s t-test; *p < 0.05; **p < 0.01; ***p < 0.001

Lack of IL-1 signalling alters the dynamics of ST2+ and RORγT+ Foxp3+ TREG subsets in fungal infection

It is well established that IL-1 and IL-33 mediate distinct cellular responses of various immune cell types, including T cells.29 Notably, IL-1 signalling plays a fundamental role in host defence against fungal diseases,30 whereas IL-33 expression is exploited by the pathogen to evade clearance.31 After observing that IL1R1−/− mice exhibited increased ST2-expressing TREG cells at the steady state (Fig. 3j–l), we hypothesised that absence of IL-1R1 signalling would further favour IL-33 activity in vivo to enhance ST2+ TREG cell responses during infection. To understand the roles of ST2 and IL-1R1 on the dynamics of the TREG cell responses in disease, we infected mice intratracheally with the fungal pathogen Cryptococcus neoformans which induces the production of IL-3331 and IL-1α/β.32,33 When compared to WT mice, IL-1R1−/− mice exhibited reduced fungal clearance in lung and brain (Fig. 7a). Further characterisation of CD4+ T cell populations revealed that lack of IL-1R1 signalling leads to significantly reduced Th17 responses during the course of infection, as evidenced by the expression of the Th17 associated transcription factor RORγT in both Foxp3+ and Foxp3 T cells (S11A). Interestingly, the lack of IL-1R1 signalling significantly increased the proportion and production of ST2 in Foxp3+ TREG cells in the lungs at day 14 of infection (Fig. 7b, c). These differences correlated with a significant increase in the frequencies of CD4+Foxp3+ T cells in the lungs of IL-1R1−/− mice (Fig. 7d) and an increased ratio of TREG (# CD4+Foxp3+) to TEFF (# CD4+Foxp3) cells in the lungs (Figure S11B–D) underlining how the lack of IL-1R1 signalling affects the TREG/TEFF cell balance in the lungs. Importantly, the frequencies of pulmonary ST2+ TREG cells initially increased in both WT and IL-1R1−/− mice but remained high throughout the course of infection in the lungs of IL-1R1−/− mice when compared to WT mice (Fig. 7e). Moreover, IL-1R1−/− mice showed significantly decreased frequencies of RORγT+ TREG cells in the lungs during the course of disease (Fig. 7f). Concomitantly, we observed a decrease in IL-17A-producing TREG cells at day 14 (Fig. 7g). As we previously observed in vitro, RORγT+ TREG cells were largely ST2 and Helios (Fig. 7h; S11E). In fact, ST2+ TREG cells expressed the transcription factor GATA3 (Fig. 7i). Moreover, ST2+ contrary to RORγT+ TREG cells maintained high levels of Helios expression (S11F). As such, GATA3+ and RORγT+ expressing Foxp3+ T cells represent two distinct subpopulations of pulmonary TREG cells during infection (Fig. 7j), and the absence of IL-1R1 significantly shift the populations in the lungs at day 14 of infection in favour of GATA3+Foxp3+ TREG cells (Fig. 7k; S11G). These results reveal the antagonistic roles of IL-33 and IL-1 in modulating the functional specialisation of Foxp3+ TREG cell during lung infection.

Fig. 7
figure 7

Absence of IL-1R1 signalling in vivo skews the TREG cell response in favour of ST2+ TREG cells during the course of disease. a IL-1R1−/− mice show increased fungal burden in the lungs (left) and brains (right) upon intra-tracheal exposure to 104 CFU/mice of Cryptococcus neoformans 52D. (N = 4–5 per group; Representative of 3 distinct experiments). b Representative flow cytometry profiles of ST2 relative to Foxp3 expression in CD4+ T cells at day 14 post-infection (lung). c Higher frequencies of Foxp3+ TREG cells in the IL-1R1−/− (red) compared to WT mice (white). d ST2-expressing TREG cells represent a major population of Foxp3+ T cells in IL-1R1−/− mice (red) throughout the course of infection in opposition to WT mice (white). e ST2+ TREG cells accumulate early in the lungs during infection but decrease in WT (white) compared to IL-1R1−/− mice (red). f IL-1R1−/− mice (red) show decreased frequencies of RORγT+ CD4+Foxp3+ TREG cells in the lungs at the peak of adaptive immunity. g IL-17A production by Foxp3+ T cells originates from ST2 TREG cells in the lungs at day 14 post-infection. h ST2 TREG expressing RORγT represent a significant fraction of TREG cells in the lungs during the course of infection. Two-way ANOVA. Tukey correction. ***p < 0.001. **p < 0.01. *p < 0.05. i ST2 expression on TREG cells correlates with increased GATA3 expression in the lungs at day 14 in both WT (white) and IL-1R1−/− (red). j, k Absence of IL-1R1 influences the balance between GATA3+ and RORγT+ TREG cells in the lungs (day 14) in WT (white) when compared to IL-1R1−/− (red). One-way ANOVA. ***p < 0.001. **p < 0.01. *p < 0.05

Discussion

CD4+Foxp3+ TREG cells, key regulators of innate and adaptive immunity, accumulate locally during the course of various acute and chronic infections, inhibit immune responses and limit pathology.34 In viral infections, TREG cells reduce the efficacy of CD4+ and CD8+ T cell responses in the early stages of viral infections.35 However, Foxp3+ TREG cells also have a protective role after viral clearance by controlling immune-mediated and microbe-associated pathology.36,37 These divergent roles suggest a delicate control in the functional dynamics of Foxp3+ TREG cells for the regulation of immune responses to infections.

An increasing body of evidence reveals that Foxp3+ TREG cells do not have a fixed lineage identity but rather display considerable functional adaptability by altering their epigenetic and transcriptional landscapes to adapt to inflammatory conditions, in turn acquiring specialised roles for efficient modulation of immune responses.38 This functional specialisation was characterized by the loss of Foxp3 expression in TREG cells, the consequential loss of their suppressive phenotype, and reprogramming into inflammatory Th1/Th17-like exFoxp3 TEFF cells in lymphopenia, infections, organ-specific autoimmunity, and in tumour microenvironments.9,11,14,21 Moreover, Foxp3+ TREG cells can also undergo a more subtle form of functional adaptability by transiently co-expressing lineage-specifying transcription factors for a more efficient control of TEFF cells expressing the same transcription factors in inflammatory sites.12,13,14 These cellular outcomes suggest a delicate control in the stability and fate of Foxp3+ TREG cells, allowing them to adjust their suppressive potential, and concurrently differentiate into inflammatory TEFF cells.

By applying an unbiased screen to delineate the mechanistic events leading to the loss of Foxp3 in TREG cells, we identified the differential expression of the cell surface receptor for IL-33 (IL-33R, ST2) or IL-1 (IL-1R1) between stable (Foxp3hi) and exTREG cells, respectively. We make the novel finding that IL-33 and IL-1 play opposing roles in dictating the dynamics and functional specialisation of TREG cells at inflamed sites during viral and fungal infection as well as chronic inflammation.

Distinct types of TEFF cells express the receptors for IL-1 (IL-1R1), IL-18 (IL-18RA) and/or IL-33 (T1/ST2, IL-33 receptor) in order to enhance their proliferation and cytokine production.39 Recent studies reveal the presence of an ST2+ TREG cell subset in mucosal and lymphoid tissues.19,26,40 IL-33 and IL-1α/β are released locally by many non-hematopoietic and hematopoietic cells at external and internal barrier surfaces upon cell stress where they function as endogenous alarmins.22 Interestingly, IL-33 was recently shown to favour the expansion of TREG cells in the gut as ST2−/− TREG cells show diminished functions in the same tissue environment.19 However, the role of IL-1 for TREG cell function was largely unknown. This was probably due to the requirement for IL-1R1+ TREG cells to differentiate into RORγT+Foxp3+ dual expressers prior to IL-1R1 signalling.

We confirm here that IL-33-mediated effects on TREG cells occur in the presence of IL-219, whereas expression of IL-1R1 is induced in the presence of IL-6 and TGFβ, essential polarizing cytokines for Th17 differentiation.41 Interestingly, IL-33-stimulated dendritic cells42 and mast cells43 were shown to produce IL-2 upon IL-33 stimulation, highlighting a positive regulatory loop to support ST2+ TREG cell stimulation. On the other hand, production of IL-1β by dendritic cells is enhanced by IL-6, IL-21 and IL-23, and promotes Th17 differentiation.44 Interestingly, both IL-1β and IL-33 enhanced survival and proliferation of polarized TREG cells within their respective inflammatory milieu in vitro. Previous studies have shown that thymic-derived TREG cells were able to respond to IL-33 and IL-2 in order to upregulate the ST2 receptor.26,45 We also confirm the strong link between ST2 and the expression of the transcription factor Helios and the surface receptors Neuropilin 1 and TIGIT, whose collective expression reflects TREG cells of thymic origin and enhanced TREG stability.28,46,47,48 In contrast, RORγT+ TREG cells show reduced Helios expression both in vivo and in vitro. We did not observe ST2 expression in in vitro or peripherally -induced TREG cell subsets (iTREG and pTREG, respectively), in exTREG cells, nor was ST2 expression prominently seen in Helios TREG cells, in contrast to a recent report,19 although further characterization of ST2+ TREG cells in the gut environment, relative to the lung, is required. Interestingly, Helios expression by tTREG cells was necessary to maintain immune homeostasis in later stages of life.28 Future studies are required to confirm whether Helios plays an important role in stabilising tTREG cell function, and if inflammatory conditions can modulate Helios expression or function.

To characterise the dynamics of ST2+ TREG cells in the context of an immune challenge, we studied their fate in two mouse models of lung infections: an acute model of murine-adapted Influenza A virus H1N1/PR8, and a chronic Cryptococcus neoformans fungal infection. In both models, we observed that ST2+ TREG cells peak early during the course of infection and at timepoints that precede the onset of peak anti-pathogen immune responses. Recently, it was suggested that ST2+ TREG cells play a particularly important role in tissue protection.40 As such, we observed that ST2+ TREG cells resisted production of pro-inflammatory cytokines throughout the course of disease, in contrast to their ST2 counterparts, which display significant plasticity by co-expressing RORγT and develop the potential to secrete inflammatory cytokines like IL-17A. The early accumulation of ST2+ TREG cells during lung injury may reflect an attempt by local mucosal TREG cells to maintain homeostasis in the early events. However, as inflammation progresses, the adaptive immune response switches to an effective inflammatory response, where newly adapted ST2 TREG cells appear. This mechanism is particularly evident during chronic infection with Cryptococcus neoformans, where the pathogen favours IL-33,31 IL-1α and IL-1β32,33 release in the early phase of disease to enhance pathogenicity. In fact, we observed that marked elevation of ST2+ TREG cells lead to an overall increase of the TREG to TEFF ratio and increased virulence and pathogenicity. Thus, IL-33 release observed with pathogenic Cryptococcus neoformans strains may promote the stability of immunosuppressive ST2+ TREG cell pool, thereby counteracting anti-microbial immune responses. On the other hand, recent evidence reveals that excessive IL-33 levels, as evidenced by lung treatment with IL-33, could alter the suppressive function of ST2+ TREG cells,49 a phenomenon we did not observe in our Th1 and Th17-driven infectious models. Thus, other signalling pathways are probably involved in the loss of function of ST2+ TREG cells and will be the focus of further investigation.

In this study, we assessed the effect of IL-1β on the suppressive response of TREG on Th17 cells. We show that IL-1β stimulation almost completely abrogated the suppressive ability of TREG cells. The inability of IL-1R1+ TREG cells to suppress Th17 cells is further substantiated by their increased survival and proliferation in the presence of IL-1, consistent with a study by Ben-Sasson et al. showing that IL-1 enhances Th17 cell proliferation.50 In accordance with these results, during infection with Cryptococcus neoformans, no elevation of RORγΤ and IL-17 expression in ST2 TREG cells was observed in IL-1R1-deficient mice, unlike their wild-type counterparts. In contrast, we found that IL-1R1-deficiency led to marked elevation of GATA3+ ST2+ TREG cells with an overall increase of the TREG to TEFF ratio and increased virulence and pathogenicity. Moreover, we observed that lack of IL-1R1 on TREG cells caused enhanced stability and suppressive ability of the Foxp3+ TREG cell population during the course of T-cell-induced colitis. In agreement with a previous report,19 we observed that ST2−/− TREG cells were inefficient in suppressing T cell-induced colitis. Strikingly, we show that the lack of IL-33R signalling in TREG cells leads to the acquisition of pro-inflammatory characteristics, and promotion of the colitogenic process. Conversely, we also show that IL-1R1−/− TREG cells have increased maintenance of Foxp3 expression in donor TREG cells, and an up-regulation of the IL-33R in this inflammatory setting. In addition, we observed that IL-1R1 is required for the proliferation of co-expressing RORγT+Foxp3+ cells, further highlighting the role of IL-1R1 for the functional adaptability of TREG cell responses. In their 2010 report, Li et al. showed that IL-1β enhanced IL17A production by TREG cells in undifferentiated co-cultures with TEFF cells in vitro,41 an observation not aligned with our conclusions. The reasons for this are unclear but may relate to the reduced TREG cell-mediated regulation of APC function in the absence of IL-1R1 signalling, and consequential induction of IL-17 secretion by TEFF cells in culture supernatants. Nonetheless, these results illustrate that distinct polarizing signals on TREG cells are required for them to acquire the ability to respond to the distinct members of the IL-1 family induced during immune challenge such as IL-1 and IL-33.

Overall, we show that two distinct members of the IL-1 family of cytokines, IL-33 and IL-1, have differential effects on the functional adaptation of Foxp3+ TREG cells at mucosal surfaces during immune challenge. We show that IL-1, but not IL-33, impedes the suppression of TEFF cells by TREG cells in highly inflammatory conditions. In contrast, IL-33 stabilises the TREG cell phenotype and function, all-the-while restricting the potential of Foxp3+ TREG cells to adopt inflammatory features in challenged mucosal sites. These results highlight a mechanism by TREG cells to adapt to the inflammatory conditions throughout the evolving nature of an infection to fine-tune anti-pathogen adaptive immunity and protect the host from pathology. They provide an additional dimension to the role and fate of TREG cells in disease. Further characterisation of the processes that lead to the adaptation of TREG cells will pave the way towards the development of therapies that aim to modulate their response during the course of disease.

Materials and methods

Mice

WT (Ly5.2+) and congenic (Ly5.1+)TCR-β−/− C57BL/6 mice were obtained from Taconic Laboratories while C57BL/6.Foxp3GFP reporter knock-in (Foxp3GFP) mice11 were provided by Alexander Rudensky and bred into the congenic background (Ly5.1+) for more than 10 generations. Inbred BALB/c were purchased from Charles River. Thy1.1+ BALB/c (JAX® CBy.PL(B6)-Thy1a/ScrJ #005443), T1/ST2−/− BALB/c mice51 and IL-1R1−/− BALB/c mice (10-generation backcross from JAX #003018 B6 IL-1R1Tm1Rom1 strain on the BALB/c background) were bred on site. IL-1R1−/− mice were kept as homozygous for breeding purposes. Foxp3GFP-CRE X Rosa26 lox-stop-lox tdTomato fate-tracking mice on C57Bl/6 background were obtained from Jeffrey Bluestone (University of California).21 All mice were used at 10–12 weeks of age. All mice were housed and bred under specific pathogen-free conditions and used according to institutional guidelines at McGill University.

Lymphocyte isolation

Isolation of T cells was performed, as previously described.24 In brief, after CO2 euthanasia, the lungs were perfused with cold PBS through the right ventricle. The collected lungs were then digested in RPMI 1640 with 5% FBS (Wisent) containing collagenase A (0,5 mg/ml) and collagenase D (0,5 mg/ml) in the presence of DNAse I (0.005 µM) (Sigma-Aldrich) for 45 min at 37 °C and then mechanically processed in the same manner as the mediastinal LN, the spleen and the inguinal lymph nodes. The lamina propria T cells (colon) were obtained through the processing of the colon as previously described.11 The cells were then filtered through a cell strainer and kept in complete RPMI medium.

Purification of T cell subsets

For in vivo adoptive transfers, TREG cells from splenocytes of Ly5.1+ Foxp3GFPki C57BL/6 were isolated the following; first CD4+ T cells were enriched using an autoMACS (Miltenyi Biotec), and then CD4+GFP+ T cells (purity >99%) were sorted using a FACSAriaTM (BD Biosciences). ST2+ and ST2 Foxp3+ TREG cells were isolated by FACSAriaTM as CD4+GFP+ ST2+ or ST2 T cells. When working on the BALB/c background, CD4+CD25hi (top 50% of CD25+ cells) and CD4+CD45RBhiCD25 cells were sorted using a FACSAriaTM the day of the transfer.

In vitro assays

FACS-sorted CD4+Foxp3+ TREG cells (GFP+, 50 × 103) expressing ST2 (ST2+) or not (ST2-) were activated in 96-well flat-bottomed (0.2 ml) plates previously coated with α-CD3 (5 μg/ml) and α-CD28 (2 μg/ml), and in the presence of IL-2 (100 U/ml - or otherwise indicated) in RPMI (Wisent) supplemented with 10% FBS at 37 °C for 72 h. For in vitro stimulation assays, IL-33 (10 ng/ml - or otherwise indicated), IL-6 (10 ng/ml - or otherwise indicated), mouse recombinant TGFβ1 (1 ng/ml) and/or IL-1β (25 ng/ml) (R&D biosystems) were added at the start of the culture unless otherwise stated. In some studies, T cell cultures were performed in the presence of Anakinra (Kineret™; Swedish Orphan Biovitrum, Stockholm, Sweden). For suppression and polarisation assays, 5.0 × 104 FACS-sorted CD45.2+CD4+GFP- were plated in 96-well flat-bottomed plates together with 2.0×105 irradiated feeder cells (CD4neg fraction) and activated with soluble α-CD3 (1 μg/mL) in the absence of IL-2. CD45.1+CD4+GFP+ TREG cells were added at a 1:2 or 1:4 ratio (2.5 × 104 or 1.25 × 104). The cells were labelled with either Violet proliferation dye V450 (BD Bioscience) or CellTrace™ Violet (Thermofisher), depending on the experiment.

Adoptive T cell transfer

For the microarray, FACSAriaTM sorted CD4+GFP+ or GFP cells from Ly5.1+ Foxp3GFPki C57BL/6 mice were suspended in PBS and transferred intravenously into Ly5.2+ TCRβ−/− animals (1.0 × 105 cells/mouse). Mice were monitored for dehydration and weight loss. Necropsy was performed at day 21 post-injection.

For the recombinant IL-33 injections, the Ly5.2+ TCRβ−/− mice received 2 × 105/mouse of FACSAria™ sorted CD4+GFP+ TREG cells (Ly5.1 + Foxp3GFPki C57BL/6) intravenously and were injected with 100 μg/mice of recombinant IL-33 (R&D biosystems) intra-peritoneum (I.P.) starting from day of injection (Day 0) every 48 h until the day of necropsy (Day 14).

In the colitis experiments, FACSAriaTM sorted Thy1.1+ CD4+CD45RBhiCD25- T cells (naïve TEFF) were kept separate from CD4+CD25hi cells (TREG) originating from WT, T1/ST2−/− or IL-1R1−/− BALB/c mice. At the time of injection, 4.0 × 105 naïve TEFF and 2.0 × 105 TREG cells were mixed and injected in the lower right quadrant of the peritoneum (I.P.). Weight of mice was monitored daily. CD4+ T cells from Foxp3GFP-CRE X Rosa26 lox-stop-lox tdTomato fate-tracking mice were sorted and adoptively transferred (I.P) into a TCRβ Ly5.1+ mouse and left for homeostatic proliferation for 10 days.

Microarray analysis

Freshly isolated (Day 0) CD4+ GFP+ (fresh TREG) and GFP (fresh TEFF) were isolated from Foxp3GFPki Ly5.1+ mice and transferred into TCRβ−/− Ly5.1+ (see Fig. 1a). At day 21 post-transfer, Ly5.2+CD4+ T cells were isolated and further separated based on GFP signal: (1) GFPhi (TREG), (2) GFPneg (exTREG) or (3) GFPneg (TEFF from GFP transferred mice). Two additional groups of cells were analysed: (4) freshly-sorted TREG (fresh TREG) and (5) freshly sorted TEFF cells (fresh TEFF) from day 0. RNA was isolated using the RNeasy Mini Kit from Qiagen as per the manufacturer’s instructions. Samples were run on a Illumina® MouseWG-6 v2.0 chip which contains 45281 mouse probes and 974 control probes (Illumina® – Genome Québec), and the resulting raw expression data were extracted, annotated, robust spline normalised and background-adjusted using illuminaMousev2.db and bead array packages in R. The top 654 genes that varied (p<0.05 cut-off) were selected and the expression patterns were analysed between the groups. A modified ANOVA for microarray analysis (eBayes function from the Limma package) was used to compare across all conditions for each gene.

Intranasal infection with Influenza A

The mouse-adapted Influenza A virus (IAV) H1N1 strain A/Puerto Rico/8/34 was propagated and titrated by plaque assay on Madin-Darby canine kidney cells, as described.52 Mice were anaesthetised by intramuscular injection with a mixture of 10 mg/kg ketamine (Ayerst Veterinary Laboratories) and 125 mg/kg xylazine (Bayer). For a sublethal infection, 20 plaque-forming units (PFU) of IAV per 20 g body weight were administered intranasally. The mice were monitored daily for clinical score and weight loss.

Intratracheal infection with C. neoformans

C.neoformans 52D (ATCC no. 24067) was grown and prepared as previously described.31 In brief, a single colony of C. neoformans from a Sabouraud dextrose agar (Becton Dickinson) was resuspended in Sabouraud dextrose broth (Becton Dickson) and then grown further in a rotating culture until stationary phase (48 h) at room temperature for in vivo infections. Subsequently, the culture was spun, washed twice with PBS, and resuspended to a desired concentration in PBS. Concentrations of C. neoformans were verified by plating on Sabouraud dextrose agar at 37 °C for 72 h followed by determination of CFU. The mice were anaesthetised with 10 mg/kg ketamine (Ayerst Veterinary Laboratories) and 125 mg/kg xylazine (Bayer) intra-peritoneally. To access the trachea, a vertical 1-cm incision of the skin was made below the jaw. A 22-gauge catheter (Becton Dickson) was inserted into the trachea. In a volume of 50 μl sterile PBS, 2 × 105 CFU/ml C. neoformans was instilled, followed quickly by 50 μl volume of air. The incision was closed using a 9 mm EZ Clip Wound Closing Kit (Stoelting). Mice were monitored daily following surgery.

Colitis score

Mean colitis score of the colon of each mouse was assessed by 5 distinct double-blinded observers following the guidelines from Erben et al.53 (4–5 mice per group; 3 distinct experiments).

Flow cytometry

After lymphocyte isolation, single-cell suspensions were stained with the following fluorescence-conjugated mAbs, purchased from Thermofisher (eBioscience) unless otherwise stated: α-CD4–Alexa700 (GK1.5), α-CD8-V500 (53-6.7) (BD Biosciences), α-ST2-PerCP710 (RMST2-2), α-CD25-PECy7 (PC61) (BD Biosciences), α-Foxp3-FITC or PE (FJK-16s), α-IL-17A-APC (eBio17B7), α-IFNγ-PECy7 (XMG1.2), α- RORγT-PE (AFKJS-9) or α-RORγT BV786 (Q31-378) (BD Bioscience), α-GATA3-Alexa647 (BD Biosciences), α-CD45.1 PE (A20) (PharMingen), α-Helios-Pacific Blue or PE (22F6) (Biolegend), α-CD121a/IL-1R1 (35F5) (BD Biosciences) and CD90.2 Alexa 780 (53-2.1). Non-viable cells were excluded using fixable viability dye eFluor780 or 506 reagent (Thermofisher). Data were acquired using a FACS Fortessa X-20 flow cytometer (BD Biosciences) and analysed using FlowJo version 9 software (TreeStar).

Statistical analysis

For all experiments, the mean and standard deviation are shown, unless otherwise stated. Multiple comparisons were tested using a two-way ANOVA with a Tukey post-test for comparison of all individual means within a figure or One-way ANOVA when required. For single comparisons, N unpaired Student’s t-test was used with the p-value expressed in the figure legend. All statistical analysis was performed with GraphPad Prism version 5 software (GraphPad Software).