Main

Whether Treg cells expressing the TH1-associated transcription factor T-bet represent a stable sub-lineage of cells with unique function or a transient activation state remains unknown. To address this question, we assessed the stability of T-bet expression in Treg cells using a novel Tbx21tdTomato-T2A-creERT2 knock-in allele combined with the R26Y recombination and Foxp3Thy1.1 reporters. The resulting Tbx21RFP-creERT2 mice showed a range of red fluorescent protein (RFP) expression and CreERT2 activity, which faithfully reflected endogenous T-bet protein levels in major lymphocyte subsets (Fig. 1a, Extended Data Fig. 1a, b). RFP+ Treg cells comprised between 30–70% of CD44hiCD62Llo effector Treg cells in lymphoid organs and non-lymphoid tissues; interestingly, intestinal Treg cells exhibited prevalent co-expression of T-bet and RORγt, but not T-bet and GATA3 (Extended Data Fig. 1d–i).

Figure 1: Stable T-bet expression in a subset of peripheral Treg cells.
figure 1

a, Splenic cells in Tbx21RFP-creERT2 mice 3 weeks after tamoxifen gavage on days −2 and 0. Numbers on graph indicate the mean. Data are mean ± s.e.m. b, Schematic of tamoxifen administration to Tbx21RFP-creERT2 mice (top) and flow cytometry (bottom) of splenic CD4 Thy1.1+ and Thy1.1 cells. c, Upper, RFP+ (left axis, squares) and YFP+ (right axis, circles) Treg cells. Lower, percentage of RFP+ of YFP+ Treg cells 3 weeks (white symbols), 3 months (grey symbols), and 7 months (black symbols) after tamoxifen gavage. d, Upper, schematic of tamoxifen treatment and N. brasiliensis infection. Lower, percentage of RFP+ among YFP+ Treg cells in mice challenged with PBS (white circles) and N. brasiliensis (Nb; black circles); (bottom, right) RFP expression in Treg (shaded histograms) or YFP+ Treg (open histograms) cells from spleens of mice challenged with PBS (black) or N. brasiliensis (red). Data are mean. Two-tailed t-test (NS, not significant). All data are representative of 2 experiments, n ≥ 3 mice per group each.

PowerPoint slide

Source data

Three weeks after tamoxifen administration we found—in contrast to a previous report7—that the vast majority of both yellow fluorescent protein (YFP)-labelled Treg and effector CD4 T cells continued to express RFP (Fig. 1b, c, Extended Data Fig. 1j). The percentage of YFP+ cells expressing RFP was similarly high at three and seven months after tamoxifen administration, although percentages of YFP+ cells declined, indicating that continual Treg cell recruitment into the T-bet+ subset balances out cell turnover over time (Fig. 1b, c, Extended Data Fig. 1j). Indicative of intrinsic stability of T-bet+ Treg cells typical of a differentiated cell state, treatment of Tbx21RFP-creERT2 mice with tamoxifen 3 weeks before infection with the helminth Nippostrongylus brasiliensis did not result in loss of RFP expression among YFP+ Treg (or effector CD4) T cells despite robust TH2 activation and cytokine production in the spleens and lungs of infected mice (Fig. 1d, Extended Data Fig. 3a, data not shown).

The presence of small percentages of YFP+RFP cells 3 weeks after gavage (Fig. 1b, c) suggested that some Treg cells might have experienced transient unstable T-bet expression at the time of tamoxifen administration. Such a scenario would reconcile the above result with an earlier study7. Indeed, in Tbx21RFP-creERT2 mice we observed RFPlo Treg cells that lacked the T-bet-dependent expression of chemokine receptor CXCR3, in addition to RFPhiCXCR3+ cells (Extended Data Fig. 2a). The former exhibited slightly lower CD44 and slightly higher CD62L expression than the latter and RNA sequencing (RNA-seq) analysis suggested that CD44hiRFPloCXCR3 Treg cells were differentiation intermediates between CD44hiRFP cells and CD44hiRFPhiCXCR3+ cells (Extended Data Fig. 2b–d). Around 40% of FACS-sorted RFPloCXCR3 (but not RFPhiCXCR3+) Treg cells lost RFP expression following transfer into lymphoreplete hosts, whereas some became RFPhiCXCR3+ (Extended Data Fig. 2e, f). Notably, populations of RFPloCXCR3 and YFP+RFP cells were also observed within the CD4 non-Treg cell population (Extended Data Fig. 2a). Thus, the observed instability of a low level of T-bet expression is not unique to Treg cells but is indicative of the gradual process of peripheral T cell effector differentiation8,9.

In addition to steady state cues, TH1-polarizing infection can drive increases in T-bet+ Treg cells10. To determine whether infection expands T-bet+ Treg cells present at steady state, or rather induces T-bet expression in T-bet cells, we administered tamoxifen to Tbx21RFP-creERT2 mice 3 weeks before challenge with the intracellular bacteria Listeria monocytogenes. Upon L. monocytogenes challenge, RFP+ Treg and effector CD4 T cell subsets increased markedly; however, YFP+ subsets did not (yielding a decreased YFP+/RFP+ ratio) (Fig. 2a, b, Extended Data Fig. 3b). This pattern was indicative of de novo differentiation of T-bet+ cells from T-bet Treg precursors in parallel with differentiation of TH1 cells. Following transfer, both CD44loCD62Lhi RFP and CD44hiRFP Treg cells upregulated RFP in response to L. monocytogenes infection (Extended Data Fig. 3c). Notably, upon L. monocytogenes infection, preformed T-bet+ Treg cells tagged with YFP prior to infection increased expression of T-bet and CXCR3, but not IL-10, an important suppressor molecule11. The latter was demonstrated by fate mapping experiments in Tbx21RFP-creERT2Il10eGFP/WT mice, which revealed no increase in IL-10 (eGFP+) among YFP+ Treg cells, whereas bulk T-bet (RFP+) IL-10+ cells increased around threefold (Extended Data Fig. 3d–g). Similar results were obtained during lymphocytic choriomeningitis virus infection (data not shown).

Figure 2: Stable differentiation of T-bet+ Treg cells in response to L. monocytogenes infection.
figure 2

a, Schematic of experiment shown in b combining tamoxifen (TX) treatment and L. monocytogenes (Lm) infection in Tbx21RFP-creERT2 mice. b, Percentage of RFP+, YFP+, and YFP+/RFP+ ratio in CD4+ Thy1.1+ (left) and Thy1.1 (right) cells in spleens and livers of mice challenged with PBS and L. monocytogenes. c, Schematic of experiments shown in d, e and f; 1° and 2° indicate primary and secondary challenge, respectively. d, Data presented as in b. e, Percentage of RFP+ of YFP+ Treg cells. f, Data presented as in b. Bars, mean. Two-tailed t-test (***P < 0.001, **P < 0.01, and *P < 0.05, respectively; NS, not significant). All data are representative of ≥ 2 experiments, n ≥ 4 mice per group each.

PowerPoint slide

Source data

We next assessed the persistence and recall response of T-bet+ Treg cells induced by L. monocytogenes infection. To preferentially label infection-induced T-bet+ cells, tamoxifen was administered at the peak of the primary L. monocytogenes response (days 7 and 9). Mice were assessed 8 weeks later, at which time the percentage of splenic and liver Treg cells that were RFP+ had returned to roughly pre-infection levels (Fig. 2c, d). Given the turnover rate of T-bet+ cells (Fig. 1c), we reasoned that by day 60 after infection YFP+ cells would be relatively enriched for infection-induced Treg cells compared to the bulk RFP+ cell pool. Reinfection increased bulk RFP+ Treg and effector CD4 T cells and even more prominently increased the corresponding cell subsets tagged with YFP (Fig. 2d, Extended Data Fig. 3h–j). On day 65 after primary infection, more than 90% of YFP+ Treg cells continued to express T-bet, as did uninfected control cells (Fig. 2e). Furthermore, mice infected with L. monocytogenes that were administered tamoxifen on days 37 and 39 after resolution of the primary response and re-infected on day 60 exhibited a parallel increase in bulk RFP+ and YFP+ Treg cell subsets on day 65, suggesting cells that acquired T-bet expression during primary infection remained T-bet-positive and expanded upon reinfection (Fig. 2f). Together, these studies demonstrate that bacterial infection caused de novo differentiation of T-bet Treg cells into stable T-bet+ cells uniquely suited for reactivation under conditions that drove their initial acquisition of T-bet.

Although the stability of T-bet+ Treg cells suggested a particular function presumably imparted by T-bet itself, we found that 12-week-old Foxp3YFP-creTbx21fl/fl mice were clinically indistinguishable from littermate controls, consistent with previous studies7,12,13. Foxp3YFP-creTbx21fl/fl mice did exhibit mild TH1 (but not CD8 T cell) activation, indicating that T-bet expression in Treg cells moderately potentiated suppression of TH1 autoimmunity (Extended Data Fig. 4a). We considered the possibility that T-bet deficiency might not fully impair the function of T-bet+ Treg cells. As Treg cell suppressor function requires continuous expression of the Foxp3 gene14, we ablated Foxp3 in T-bet+ Treg cells using a novel Tbx21tdTomato-T2A-cre allele (Extended Data Fig. 5a). Loss of Foxp3 expression in T-bet+ Treg cells in 8-week-old Tbx21RFP-creFoxp3fl mice resulted in deceased weight gain, lymphadenopathy, T cell activation, and marked immune infiltration in the lung; with age, loss of hair pigmentation and rectal prolapse were evident (Fig. 3a–d, Extended Data Fig. 5).

Figure 3: Foxp3 ablation in T-bet+ Treg cells results in spontaneous TH1 autoimmune disease.
figure 3

a, Body weights of 8–10-week-old Tbx21RFP-creFoxp3WT (grey circles), Tbx21RFP-creFoxp3fl (red circles), Tbx21RFP-creFoxp3WT/WT (blue circles), and Tbx21RFP-creFoxp3fl/WT (white circles) mice. b, Haematoxylin and eosin staining (left) and histology scores (right) of lungs from Tbx21RFP-cre mice combined with indicated Foxp3 alleles, treated or not with antibiotics (ABX). Tbx21RFP-creFoxp3fl mice show moderate perivascular and peribronchiolar inflammation, mild respiratory epithelial hyperplasia and mucus metaplasia with hyalinization (arrows). Pulmonary arterioles are contracted with thickened media, reactive endothelia and marginating leukocytes (arrowheads). Original magnification, 20×. c, d, Lymph node cell numbers (c) and characterization of T cell populations in spleens (d). e, Flow cytometry of splenic cells in Tbx21RFP-creFoxp3WT (left) and Tbx21RFP-creFoxp3fl (right) mice, gated on fixed CD4+ (top) and live CD4+CD25 (bottom) cells. f, Quantification of RFP and RFP+ CD4 T cells, as shown in e (bottom). g, RFP expression (left) and cytokine production (right) in splenic CD8 T cells. h, Cytokine production by splenic CD4+Foxp3 T cells. i, Representative images (left) and insets (right) of spleen sections from Tbx21RFP-cre mice with CD4 (green, top) or CD8 (green, bottom), RFP (red), Foxp3 (blue) and CD44 (grey). Inset, arrowheads indicate CD4+CD44hiRFP+Foxp3 (top) or CD8+CD44hiRFP+ (bottom) cells and arrows indicate CD4+CD44hiRFP+Foxp3+ cells. j, k, Nearest distances between cells as shown in i. Foxp3+ denotes CD4+CD44hiFoxp3+; Foxp3 (j) denotes CD4+CD44hiFoxp3 and CD8+ (k) denotes CD8+CD44hiRFP+. Each circle (j, k) represents the distance between cells on imaged sections from three mice. Data are mean ± s.e.m. Two-tailed t-test (***P < 0.001, **P < 0.01; NS, not significant). All data are representative of several experiments.

PowerPoint slide

Source data

Indicative of TH1-type inflammation, the majority of expanded effector CD4 and CD8 T cells in Tbx21RFP-creFoxp3fl mice expressed RFP (Fig. 3e–g, Extended Data Fig. 5). Additionally, IFNγ and IL-2 but neither IL-4 nor IL-17 production by T cells were increased compared to controls (Fig. 3g, h, Extended Data Fig. 5). Antibiotic treatment did not mitigate autoimmunity in Tbx21RFP-creFoxp3fl mice, excluding microbial antigens as the drivers of TH1 inflammation (Fig. 3b, data not shown). We considered whether induction of a robust non-TH1 immune response in Tbx21RFP-creFoxp3fl mice might reveal a potential function for T-bet+ Treg cells in its control. However, the TH2 response to N. brasiliensis infection was not increased in Tbx21RFP-creFoxp3fl mice compared to control mice, in contrast to the exacerbated TH2 response observed upon pan-Treg-cell depletion during helminth infection15,16 (Extended Data Fig. 6). Notably, whereas CXCR3+ Treg cells were significantly depleted neither total nor effector Treg cell numbers were diminished, and analysis of Tbx21RFP-creR26Y mice confirmed that a significant proportion of effector Treg cells had not undergone Cre-mediated recombination (Fig. 3d, Extended Data Fig. 5c, f). These results suggested that immune activation could not be attributed to non-specific loss of effector Treg cells.

Cells that are likely to represent ex-Treg cells, which have lost Foxp3 expression, but continue to express high CD25, CD39, CTLA4 and GITR levels2,17,18 were readily found in male Tbx21RFP-creFoxp3fl and, to a lesser extent, female Tbx21RFP-creFoxp3fl/WT mice (Fig. 3e, Extended Data Fig. 5g, h). The lack of autoimmunity in Tbx21RFP-creFoxp3fl/WT females—in which only half of T-bet+ Treg cells lose Foxp3 owing to X-inactivation—indicated that ex-Treg cells were efficiently controlled by remaining T-bet+ Treg cells (Fig. 3). Moreover, upon adoptive transfer into T-cell-deficient hosts, ex-Treg cells induced no more pathology and expanded less than CD4 effector T cells (Extended Data Fig. 7a–c). These results indicate that ex-Treg cells were unlikely drivers of (although it is possible that they may play some role in) the observed autoimmunity.

To determine whether punctual ablation of T-bet+ Treg cells would similarly unleash TH1 inflammation, we generated bone marrow chimaeric mice with a 1:1 mix of either CD45.1+ Foxp3WT or Foxp3KO with CD45.2+ Tbx21RFP-cre/WTR26iDTR haematopoietic precursor cells (Fig. 4). In Foxp3KO:Tbx21RFP-cre/WTRosa26iDTR mixed chimaeras, all T-bet+ Treg cells expressed diphtheria toxin receptor (DTR) and were susceptible to diphtheria-toxin-mediated ablation, whereas the rest of the T-bet-expressing cell types and subsets represented a 1:1 mix of DTR-expressing and non-expressing cells. Before treatment with diphtheria toxin, both sets of mixed chimaeras were healthy with similar basal levels of T cell activation (data not shown). Administration of diphtheria toxin over 2 weeks resulted in weight loss, T cell activation, and a selective increase in IFNγ production by CD4 and CD8 T cells in Foxp3KO:Tbx21RFP-cre/WTR26iDTR chimaeric mice ablated of T-bet+ Treg cells compared to Foxp3WT:Tbx21RFP-cre/WTR26iDTR controls (Fig. 4). Treg cell percentages in experimental mice were only very modestly decreased (from 13 ± 0.53 to 11 ± 0.82, P = 0.022) and, as in Tbx21RFP-creFoxp3fl mice, percentages of CD44hiCD62Llo Treg cells were undiminished compared to controls (Fig. 4b, c). This experimental model is not confounded by generation of ex-Treg cells, providing additional evidence that the latter were not the sole drivers of pathology in the absence of T-bet+ Treg cells. Finally, weight loss was not observed in TcrbKO:Tbx21RFP-cre/WTR26iDTR mixed chimaeras, in which T-bet+ Treg and effector T-bet+ TCRαβ+ cells were simultaneously ablated, implicating the latter in driving disease (Extended Data Fig. 7d, e).

Figure 4: Acute ablation of T-bet+ Treg cells results in TH1 immune activation.
figure 4

Bone marrow chimaeric mice were injected with 0.5 μg diphtheria toxin (DT) on day 0, then treated daily with 0.1 μg of diphtheria toxin until day 15. a, Weight loss in the indicated mice. b, Flow cytometry of splenic CD4 (top) and Treg (bottom) cells in the indicated mice. c, Activation status of CD45.1+ and CD45.2+ Treg cell compartments in spleens of indicated mice. d, e, T cell activation (d) and cytokine production (e) in control (white circles) and T-bet-depleted (black circles) chimaeras. Data are mean ± s.e.m. Two-tailed t-test (**P < 0.01, *P < 0.05; NS, not significant). Data are representative of 2 experiments, n ≥ 6 mice per group.

PowerPoint slide

Source data

RNA-seq analysis revealed that 561 genes, including Tbx21, Cxcr3, Gzmb, Ebi3, Fgl2, and Il10, were more highly expressed in CD44hiRFP+ cells compared to CD44hiRFP Treg cells (Extended Data Fig. 2c). Expression of this gene set was increased upon loss of Foxp3 in ex-Treg cells, suggesting that Foxp3 opposes the transcriptional signature of T-bet+ Treg cells to prevent full TH1 differentiation10 (Extended Data Fig. 4b). Notably, the TH1-associated chemokine receptor CCR5 and adhesion molecule β1-integrin (CD29) were expressed in T-bet+ Treg cells independently of T-bet (Extended Data Fig. 4c, d) indicating that some functional redundancy of homing molecules may in part explain the mild phenotype of Foxp3YFP-creTbx21fl/fl mice. Moreover, we found that the TCR repertoires of CD44hiCXCR3(T-bet)+ and CD44hiCXCR3(T-bet) Treg subsets in DO11.10 TCRβ+ Tcra+/− mice were distinct, suggesting that antigenic specificity of T-bet+ Treg cells may also contribute to distinct localization and suppressor capacity, as recent studies revealed TCR-dependent spatial proximity of Treg and IL-2-producing self-reactive T cells19 (Extended Data Fig. 4e).

Therefore, we sought to determine the relative spatial positioning of T-bet+ and T-bet Treg and effector T cells in secondary lymphoid organs of Tbx21RFP-cre mice. Immunofluorescence imaging revealed pronounced preferential proximity of CD44hiT-bet+ versus CD44hiT-bet Treg cells to CD44hiT-bet+ TH1 and CD8 T cells (Fig. 3i–k, Extended Data Fig. 8a–d). In contrast, CD44hiT-bet+ Treg cells were no nearer to T-bet CD4 effectors than were CD44hiT-bet Treg cells (Fig. 3j, Extended Data Fig. 8c). Notably, the CD44hiT-bet Treg cells remaining in Tbx21RFP-creFoxp3fl mice were no nearer to TH1 or CD8 T cells than were CD44hiRFP Treg cells in healthy Tbx21RFP-creFoxp3WT mice (Extended Data Fig. 8e, f). This result suggests that failure of non-T-bet+ Treg cells to approximate TH1 cells may at least in part account for their inability to suppress TH1 inflammation.

Lastly, to complement T-bet+ Treg cell ‘loss-of-function’ experiments we sought to selectively eliminate T-bet Treg cells. We generated a Foxp3fl-DTR allele by inserting a loxP-flanked IRES-DTReGFP DNA sequence into the 3′ UTR of the Foxp3 gene (Extended Data Fig. 9a) and generated Foxp3fl-DTRTbx21RFP-creERT2 mice (Fig. 5a). After 9 days of diphtheria toxin treatment, Treg cells in mice pre-treated with tamoxifen (day −5 and −3) were present in undiminished percentages and were exclusively T-bet+ and CXCR3+ (Fig. 5a–c). Compared to vehicle (oil)-treated mice, tamoxifen-treated Foxp3fl-DTRTbx21RFP-creERT2 mice displayed robustly suppressed CD8 T cell activation and selective suppression of IFNγ production by CD4 and CD8 T cells, but unrestrained TH2 and TH17 cytokine production (Fig. 5d–f). T-bet+ Treg cells similarly suppressed pre-established TH1, but not TH2 or TH17, activation induced by depletion of Treg cells before tamoxifen treatment (Extended Data Fig. 9b–g). Selective TH1 suppression was not simply a feature of activated Treg cells rebounding after depletion, as partial depletion and recovery of Treg cells in Foxp3DTR mice resulted in prominently inhibited TH2 responses (Extended Data Figs 9h–l, 10).

Figure 5: T-bet+ Treg cells suppress TH1 and CD8+ T cells, but not TH2 or TH17 responses.
figure 5

a, Schematic for tamoxifen administration and depletion of non-T-bet-expressing Treg cells in Foxp3fl-DTRTbx21RFP-creERT2 mice. b, Flow cytometry of splenic CD4 T cells in the indicated mice on day 9, as outlined in a. cf, Treg cell percentages (c), and activated (d) and cytokine-producing (e, f) T cells in spleens of tamoxifen-treated Foxp3Thy1.1Tbx21RFP-creERT2 (open circles), oil-treated Foxp3fl-DTRTbx21RFP-creERT2 mice (black circles) and tamoxifen-treated Foxp3fl-DTRTbx21RFP-creERT2 (grey circles) mice. Data are mean ± s.e.m. Two-tailed t-test (***P < 0.001, **P < 0.01, *P < 0.05; NS, not significant). Data are representative of 2 experiments, n ≥ 2 mice per group each.

PowerPoint slide

Source data

Our studies suggest that T-bet expression in Treg cells denotes a differentiated cell state with unique T-bet-dependent and -independent gene expression and TCR specificity, capable of driving potent immunosuppression limited to circumstances of TH1 and CD8 T cell activation. It is possible that GATA3- and RORγt-expressing Treg cells may play analogous roles in suppression of TH2 and TH17 responses. Such division of anti-inflammatory labour among Treg cells, arising at steady state and during infection, may enable focused regulation of specific T helper cell responses without incurring undesired bystander suppression.

Methods

Animals

Tbx21tdTomato-T2A-creERT2 mice were generated by insertion of a targeting construct into the Tbx21 locus by homologous recombination in embryonic stem cells on the C57BL/6 background; the targeting construct was generated by inserting sequence containing exons 2–5 of the Tbx21 gene from BAC RP23-237M14 (BACPAC Resources Center) into a plasmid backbone containing a PGK promoter driving expression of diphtheria toxin A subunit (DTA) followed by BGHpA sequence (modified PL452 plasmid). A SalI restriction enzyme site was simultaneously engineered into the Tbx21 3′ UTR between the stop codon and the polyadenylation site. The Clontech Infusion HD Cloning system was used to generate in the pUC19 plasmid backbone sequence containing (in order from 5′ to 3′) encephalomyocarditis virus IRES; tandem dimer (td) Tomato; T2A self-cleaving peptide from Thosea asigna virus; Cre recombinase fused to the oestrogen receptor ligand binding domain (ER); followed by a frt site-flanked PGK-Neomycin resistance gene (NEO)-BGHpA cassette. The IRES-tdTomato-T2A-CreERT2-frt-NEO-BGHpA-frt sequence was PCR-amplified and inserted into the SalI site in the Tbx21 3′ UTR in the modified PL452 backbone. The resulting plasmid was linearized with the restriction enzyme NotI before electroporation into embryonic stem cells. Tbx21tdTomato-T2A-cre mice were generated similarly, with Cre recombinase containing a nuclear localization sequence replacing the CreERT2 sequence. Tbx21tdTomato-T2A-creERT2 and Tbx21tdTomato-T2A-cre mice were bred to FLPeR mice to excise the NEO cassette and backcrossed to C57BL/6 mice to remove the FLPeR allele.

Foxp3fl-DTR mice were similarly generated by insertion of a targeting construct into the Foxp3 locus by homologous recombination in embryonic stem cells on the C57BL/6 background; the targeting construct was generated by inserting sequence containing exons 8–13 of the Foxp3 gene from a 30.8-kb cosmid containing the complete Foxp3 gene into the plasmid backbone containing a PGK promoter driving expression of diphtheria toxin A subunit (DTA) followed by BGHpA sequence (modified PL452 plasmid). The Clontech Infusion HD Cloning system was used to generate in the pUC19 plasmid backbone sequence containing (in order from 5′ to 3′) a loxP site; encephalomyocarditis virus IRES; diptheria toxin receptor (DTR) enhanced green fluorescent protein (eGFP) fusion protein; a triple SV40 polyA (STOP); a second loxP site; encephalomyocarditis virus IRES; Thy1.1; followed by a frt site-flanked PGK-Neomycin resistance gene (NEO)-BGHpA cassette. The loxP-IRES-DTReGFP-STOP-loxP-IRES-Thy1.1-frt-NEO-BGHpA-frt sequence was PCR-amplified and inserted into the BaeI site in the Foxp3 3′ UTR in the modified PL452 backbone. The resulting plasmid was linearized with the restriction enzyme NotI before electroporation into embryonic stem cells. Foxp3fl-DTR mice were bred to FLPeR mice to excise the NEO cassette and backcrossed to C57BL/6 mice to remove the FLPeR allele.

Foxp3Thy1.1, R26Y, Foxp3fl, Foxp3KO, RorcGFP, Foxp3YFP-cre, IL-10eGFP, and Tbx21fl mice have been previously described2,20,21,22,23,24,25. CD45.1, R26iDTR, and TcrbKO mice were purchased from Jackson Laboratories26. Foxp3Thy1.1Tbx21CreERT2R26Y (called Tbx21CreERT2 mice in the text) mice are homozygous at each locus. Tbx21RFP-creFoxp3WT, Tbx21RFP-creFoxp3fl, Tbx21RFP-creFoxp3WT/WT, and Tbx21RFP-creFoxp3WT/fl mice described in the text are homozygous for the Tbx21 knock-in allele. Foxp3fl-DTRTbx21RFP-creERT2 mice described in the text are homozygous at each locus. Generation and treatments of mice were performed under protocol 08-10-023 approved by the Sloan Kettering Institute (SKI) Institutional Animal Care and Use Committee. All mouse strains were maintained in the SKI animal facility in specific pathogen free (SPF) conditions in accordance with institutional guidelines and ethical regulations.

For tamoxifen administration, 40 mg tamoxifen dissolved in 100 μl ethanol and subsequently in 900 μl olive oil (Sigma-Aldrich) were sonicated 4 × 30 s in a Bioruptor Twin (Diagenode). Mice were orally gavaged with 200 μl tamoxifen emulsion per treatment. For diphtheria toxin (DT) injections, DT (Sigma-Aldrich) was dissolved in PBS and 200 μl of indicated doses (1 μg per mouse unless otherwise indicated) were injected i.p. per mouse. For antibiotic treatment, mice were weaned onto filtered antibiotic-treated water containing ampicillin, kanamycin, vancomycin and metronidazole (0.1% w/v each).

All mice analysed were sex and aged matched (8–12 weeks old) with the exception of some Tbx21RFP-creFoxp3WT and Tbx21RFP-creFoxp3fl mice used for immunofluorescence imaging that were up to 10 months of age (results were similar to in 8–12-week-old mice).

Isolation of cells

For analysis of YFP-labelled CD4 T cells in Tbx21RFP-creERT2 mice, CD4 T cells in spleens and lymph nodes were enriched using the Dynabeads CD4 Positive Isolation Kit (Invitrogen). To isolate lymphocytes from tissues, mice were euthanized and immediately perfused with 20 ml PBS. Small and large intestines were removed, flushed with PBS and Peyer’s patches were removed. Subsequently, 0.5-cm-long fragments of intestines were washed in PBS and incubated in PBS supplemented with 5% fetal calf serum, 1% l-glutamine, 1% penicillin–streptomycin, 10 mM HEPES, 1 mM dithiothreitol, and 1 mM EDTA for 15 min. Samples were washed and incubated in digest solution (RPMI supplemented with 5% fetal calf serum, 1% l-glutamine, 1% penicillin–streptomycin, 10 mM HEPES, 1 mg ml−1 collagenase, and 1 U ml−1 DNase I) for 10 min twice. After filtering through a 100-μm strainer, cells were resuspended in 35% Percoll to eliminate debis. Lymphocytes from livers and lungs were isolated by 50–60 min incubation in digest solution, filtered through 100-μm strainers, and after debris removal in 35% Percoll, purified by centrifugation (1,000g, 7.5 min) over a step-wise 44%/67% Percoll gradient at room temperature.

Nippostrongylus brasiliensis and Listeria monocytogenes infections

N. brasiliensis was maintained by passage in 9–10-week-old male Wistar rats as previously described27. In brief, rats were injected subcutaneously (s.c.) with 7000 L3 N. brasiliensis and stool was collected on days 6–9 after infection. Fecal pellets were mixed with 5 × 8 bone charcoal and incubated on moist filter paper in Petri dishes at 26 °C for 7 days. L3 larvae were recovered from the edge of the filter paper and the perimeter of the plates and extensively washed with PBS to eliminate contaminants before infection. Mice infections were carried out using a 23G needle at a concentration of 500 L3 N. brasiliensis in 200 μl. For L. monocytogenes infections, frozen stocks were thawed, resuspended in Brain-Heart Infusion media, and grown at 37 °C to an OD600 of 0.1. For primary infections, mice were injected via lateral tail vein with 5–10 × 103 colony-forming units (cfu) of L. monocytogenes diluted in 200 μl PBS. For secondary infection, mice were injected via lateral tail vein with 105 cfu of L. monocytogenes in 200 μl PBS.

Treatments of rats were performed under protocol 08-10-023 approved by the Sloan Kettering Institute (SKI) Institutional Animal Care and Use Committee. Rats were maintained in the SKI animal facility in Biosafety Level 2 conditions in accordance with institutional guidelines and ethical regulations.

Cell transfer experiments

For cell transfer experiments, pooled spleens and lymph nodes were enriched for CD4 T cells using the Dynabeads CD4 Positive Isolation Kit. Cells were FACS-sorted on an Aria II cell sorter (BD Bioscience), washed 3 times in PBS, resuspended in 200 μl PBS, and transferred into recipients via retro-orbital injection.

Generation of bone marrow chimaeric mice

Tcrb−/−Tcrd−/− recipient mice were lethally irradiated with 650 Gy. The following day, bone marrow was isolated from femurs of donor mice and depleted of T cells and RBCs via staining with biotinylated anti-Thy1.2 and anti-Ter119 antibodies followed by magnetic bead negative selection. 5 × 106 total T-cell-depleted bone marrow cells were transferred into recipient mice via retro-orbital injection.

Flow cytometric analysis

Cells were stained with LIVE/DEAD Fixable Yellow Dead Cell Stain (Molecular Probes) and the following antibodies purchased from eBioscience, BioLegend, BD Biosciences, Tonbo, or obtained from the NIH tetramer core facility: anti-CD4 (RM4-5, Biolegend 100548), anti-CD8a (5H10, BD Biosciences 564297), anti-TCRβ (H57-597, eBioscience 47-5961-82), PBS-57-loaded mCD1d tetramer (NIH 26181), anti-Thy1.1 (HIS51, eBioscience 17-0900-82), anti-CD44 (IM7, BioLegend 103026), anti-CD62L (MEL-14, eBioscience 25-0621-82), anti-CXCR3 (CXCR3-173, eBioscience 17-1831-173), anti-CD25 (PC61.5, eBioscience 17-0251), anti-CTLA-4 (UC10-4B9, eBioscience 17-1522-82), anti-GITR (DTA-1, eBioscience 48-5874-82), anti-CD39 (24-DMS1, eBioscience 25-0391-82), anti-CD11b (M1/70, Tonbo Bioscience 25-01120U100), anti-SiglecF (E50-2440, BD Pharmingen 562681), anti-CCR5 (HM-CCR5(7A4) (eBioscience 12-1951-82) and C34-3448 (BD Biosciences 559921), anti-CD29 (eBioHMb1-1, eBioscience 48-0291-80), anti-Foxp3 (FJK-16 s, Tonbo Bioscience 35-5773-U100), anti-T-bet (4B10, BioLegend 644816), anti-RORγt (B2D, eBioscience 12-6981-82), anti-Gata-3 (TWAJ, eBioscience 46-9966-41), anti-DsRed (Living Colours DsRed Polyclonal Antibody, Clontech 632496), anti-IFNγ (XMG1.2, eBioscience 48-7311-80), anti-IL-4 (11B11, eBioscience 51-7041-82), anti-IL-17A (17B7, eBioscience 61-7177-82), anti-IL-13 (eBio13A, eBioscience 12-7133-82), anti-IL-5 (BD Pharmingen, 554396), and anti-IL-2 (JES6-5H4, eBioscience 25-7021-82). Flow cytometric analysis was performed using an LSRII flow cytometer (BD Bioscience) and FlowJo software (Tree Star). Intracellular staining was performed using eBioscience Fixation Permeabilization buffers. For cytokine staining lymphocytes were stimulated with soluble anti-CD3 clone 2C11 (5 μg ml−1) and anti-CD28 clone 37.51 (5 μg ml−1) in the presence of 1 μg ml−1 brefeldin A for 5 h at 37 °C, 5% CO2. Unless otherwise stated, CD4 T cells were pre-gated as TCRβ+ PBS-57-CD1d tetramer cells.

RNA-seq analysis

Pooled spleens and lymph nodes were enriched for CD4 T cells using the Dynabeads CD4 Positive Isolation Kit. CD4+Thy1.1+ cells were FACS-sorted on an Aria II cell sorter (BD Bioscience) into four populations (CD62LhiCD44loRFP, CD44hiRFP, CD44hiRFPloCXCR3, and CD44hiRFPhiCXCR3+ cells) and resuspended in Trizol. Three replicates of each cell subset were generated. RNA-sequencing reads were aligned to the reference mouse genome GRCm38 using the Burrows–Wheeler Aligner (BWA)28 and local realignment was performed using the Genome Analysis Toolkit (GATK)29. For each sample, raw count of reads per gene was measured using R, and DESeq2 R package30 was used to perform differential gene expression among different conditions. A cutoff of 0.05 was set on the obtained P values (that were adjusted using Benjamini–Hochberg multiple testing correction) to get the significant genes of each comparison.

TCR sequencing and data analysis

In brief, following isolation of CD4+ T cells from spleens and lymph nodes of DO11.10 TCRβ transgenic Tcra+/−Foxp3DTR mice using the Dynabeads CD4 Positive Isolation Kit (Invitrogen), CD44hiCXCR3 and CD44hiCXCR3+eGFP(Foxp3)+ Treg and eGFP effector CD4 T cells were FACS sorted and stored in Trizol. TCR sequencing and data analysis were performed as previously described31. Pearson’s correlation of clonotype frequencies for the shared TCR clones was used for the generation of the dendrogram.

Microscopy

Confocal imaging was done using standard conditions. In brief, mice were perfused in PLP buffer. Lymph nodes and spleens were excised, fixed for 1 h at room temperature in 4% paraformaldehyde, and dehydrated at 4 °C in sucrose (30% in PBS). Tissues were snap-frozen in OCT compound (Sakura Tissue-Tek). 10 μm tissue sections were cut and fixed with Acetone for 20 min at −20 °C, rehydrated in PBS and blocked with 10% normal donkey serum, in PBS with 0.3% Triton X-100, followed by overnight antibody staining at 4°C in a humidified chamber. After antibody staining nuclei were stained with 5 μM Draq7 (Abcam) for 20 min at room temperature. Sections were imaged in Prolong Diamond mounting media (Life Technologies). All images were acquired using a confocal microscope (LSM880; Carl Zeiss) with a 40× oil immersion objective. Images were processed and analysed using ImageJ software (version 2.0.0-rc-54/1.51h; National Institutes of Health). Nearest neighbour analysis was performed using MATLAB (version R2016b, MathWorks).

Statistical analysis

All statistical analyses (excluding RNA-seq and TCR sequence analyses, described above) were performed using GraphPad Prism 6 software. Differences between individual groups were analysed for statistical significance using the unpaired or paired two-tailed t-test. *P ≤ 0.05; **P ≤ 0.01; ***P ≤ 0.001; NS, not significant. The Kolmogorov–Smirnov test is used to determine the significance between the distributions of signature genes and the rest of expressed genes. One-way ANOVA is used to compare the means of three or more samples. No statistical method was used to predetermine sample size. The number of mice used in each experiment to reach statistical significance was determined on the basis of preliminary data. No animals were excluded from the analyses. No methods of randomization were used to allocate animals into experimental groups. No blinding was used. Data met assumptions of statistical methods used and variance was similar between groups that were statistically compared.

Code availability

The colocalization program (ImageJ software, 2.0.0-rc-54/1.51h, National Institutes of Health) used to find cell positions and the MATLAB program (software R2016b, MathWorks) to calculate nearest cell distance are provided in the Supplementary Information.

Data availability

The RNA-seq data that support the findings of this study have been deposited in the NIH SRA database with the accession code SRP102941.