Skip to main content
Proceedings of the National Academy of Sciences of the United States of America logoLink to Proceedings of the National Academy of Sciences of the United States of America
. 2015 Nov 16;112(48):14942–14947. doi: 10.1073/pnas.1520393112

Imbalanced signal transduction in regulatory T cells expressing the transcription factor FoxP3

Dapeng Yan a,b,1, Julia Farache a,b,2, Michael Mingueneau a,b, Diane Mathis a,b,3, Christophe Benoist a,b,3
PMCID: PMC4672803  PMID: 26627244

Significance

The homeostasis of FoxP3+ Treg cells, essential controllers of immune and autoimmune responses, integrates inputs from the antigen-specific T-cell receptor (TCR) and from trophic cytokines or chemotactic cues. Expanding upon previous indications that Treg cells might be hyporesponsive, we find that TCR triggering induces dampened responses along all tested pathways in Treg compared with conventional CD4+ T cells, whereas their responses to IFN-I, IL6, or IL2 were equal or stronger. This imbalance indicates that Treg cells are less sensitive to direct recognition of novel antigens, for instance of an infectious nature, and more to cytokine-based cues or intercellular communication.

Keywords: signal transduction, immunoregulation

Abstract

FoxP3+ T regulatory (Treg) cells have a fundamental role in immunological tolerance, with transcriptional and functional phenotypes that demarcate them from conventional CD4+ T cells (Tconv). Differences between these two lineages in the signaling downstream of T-cell receptor-triggered activation have been reported, and there are different requirements for some signaling factors. Seeking a comprehensive view, we found that Treg cells have a broadly dampened activation of several pathways and signaling nodes upon TCR-mediated activation, with low phosphorylation of CD3ζ, SLP76, Erk1/2, AKT, or S6 and lower calcium flux. In contrast, STAT phosphorylation triggered by interferons, IL2 or IL6, showed variations between Treg and Tconv in magnitude or choice of preferential STAT activation but no general Treg signaling defect. Much, but not all, of the Treg/Tconv difference in TCR-triggered responses could be attributed to lower responsiveness of antigen-experienced cells with CD44hi or CD62Llo phenotypes, which form a greater proportion of the Treg pool. Candidate regulators were tested, but the Treg/Tconv differential could not be explained by overexpression in Treg cells of the signaling modulator CD5, the coinhibitors PD-1 and CTLA4, or the regulatory phosphatase DUSP4. However, transcriptome profiling in Dusp4-deficient mice showed that DUSP4 enhances the expression of a segment of the canonical Treg transcriptional signature, which partially overlaps with the TCR-dependent Treg gene set. Thus, Treg cells, likely because of their intrinsically higher reactivity to self, tune down TCR signals but seem comparatively more attuned to cytokines or other intercellular signals.


FoxP3+ T regulatory (Treg) cells help maintain lymphoid homeostasis in many immunological contexts: tolerance to self versus autoimmune deviation, foeto-maternal tolerance, allergy, responses to pathogens, and interactions with commensal microbes (14). Their importance is highlighted by the devastating multiorgan inflammation that occurs in FoxP3-deficient scurfy mice or immunodysregulation polyendocrinopathy enteropathy X-linked (IPEX) patients (5), or upon experimental lineage ablation. In addition, FoxP3+ Treg cells partake in extraimmune regulatory activities, for instance by dampening inflammation in visceral adipose tissue or channeling tissue repair after muscle injury (6). Treg cells influence not only other T cells but also cells of the innate immune system such as natural killer or dendritic cells and macrophages. Treg cells differ transcriptionally from their “conventional” CD4+ counterparts (Tconv) with respect to their transcriptomes, and a canonical “Treg signature” of differentially expressed transcripts has been defined, which collectively define and ensure the stability of the Treg phenotype (refs. 1, 3, and references therein).

The somatically rearranged T-cell receptor (TCR) expressed by Treg cells plays an important part in their differentiation and physiology. The TCR repertoire expressed by Treg cells is quite broad and largely, albeit not completely, distinct from that of Tconv cells (79). Treg differentiation requires TCR interactions with MHC-II molecules (ref. 10 and references therein), as engagement of the TCR by agonist ligands during thymic differentiation strongly favors clonal deviation into the FoxP3+ lineage, either by inducing differentiation along the lineage (11) or because FoxP3+ cells are inherently more resistant to clonal deletion (12). Accordingly, the analysis of mice expressing a Nurr77-GFP reporter, whose expression correlates with the strength of cell activation via the TCR, showed that the reporter was generally expressed at a higher level in Treg than in Tconv cells (13). For mature Treg cells, experiments involving genetically interrupted TCR expression or signaling showed that TCR engagement is not required for Treg survival (at least in the short term) and FoxP3 expression but is necessary for suppressor activity and expression of a component of the typical Treg transcriptional signature (1416). Indeed, Treg-mediated suppression requires engagement by a cognate antigen, even if different from the antigen recognized by the T cell being suppressed (17).

Accordingly, there are indications that, although Treg cells express largely the same canonical signaling armamentarium as Tconv cells, they differ in the quantitative balance of these pathways. Integration of results from many gain- or loss-of-function experiments suggests that, overall, signaling along the NF-κB pathways favors Treg differentiation and activity, whereas AKT/mTORC2 signals are inhibitory (reviewed in ref. 3). Several reports also pointed to differences in Treg signaling architecture, such as a different involvement of PKC-Θ (18), DGKζ (19), SHIP (20), or Dlgh1 (21). Finally, there have been reports that signaling intensity downstream of TCR engagement is reduced in Treg relative to Tconv cells (2224). Here, we set out to assess the spread of signaling particularities in Treg cells and track their possible molecular origins.

Results

Dampened Signals Elicited by the TCR in Treg Cells.

To analyze Treg responsiveness in a tightly controlled manner, whole splenocytes were stimulated in vitro by binding of biotinylated anti-CD3 and -CD28 antibodies, followed by cross-linking with streptavidin, while sharply raising the temperature to 37 °C to initiate signal transduction (24). At various times following the addition of streptavidin, cells were fixed, permeabilized, and stained with antibodies to distinguish Treg and Tconv cells coactivated in the same reactions or to probe phosphorylation levels at different TCR-triggered signaling nodes. FoxP3+ Treg cells were distinguished from Foxp3-negative Tconv by staining with anti-FoxP3 antibody (for experiments in inbred C57BL/6 or mutant mice) or with anti-Thy1.1 for experiments in B6.Foxp3.Cd90.1 reporter mice (25). As illustrated in Fig. 1A, and as expected from much prior literature, this cross-linking led to rapid and very synchronous Erk1/2 phosphorylation in Tconv cells, which then waned within the next 10 min. In contrast, Erk activation was much less extensive in Treg cells, as quantitated in Fig. 1B, with a lower proportion of responding cells and a lower mean fluorescence intensity [the minority of Treg cells that did respond also showed lower levels of phospho-Erk (pErk) than Tconv]. The same results were observed in C57BL/6 mice (Fig. S1B). Higher responses in Tconv were reflected quantitatively, over a number of experiments, by a higher induction ratio (Fig. 1C). On the other hand, the pErk induced in Treg cells persisted longer than in Tconv, as visible in Fig. 1B and manifest in low rates of decay in an exponential fit of the pErk postpeak data, probably reflecting reduced operation of negative feedback loops at these lower induction levels (Fig. 1C and Fig. S1A).

Fig. 1.

Fig. 1.

Lower TCR-induced signals in Treg cells. (A) pErk versus CD90.1 (FoxP3 reporter) cytometry profiles at different times after streptavidin cross-linking of CD3 and CD28, gated on CD4+TCRβ+ cells. (B) Quantitation of data from A, as percent pErk+ cells and mean fluorescence intensity (MFI) (A and B representative of seven independent experiments). (C) Compilation across experiments of pErk induction (2 min MFI/baseline MFI ratio) and of the off-rate (exponent of an exponential fit). (D) Representative (of three experiments) pErk responses at 2 min across a titration of anti-CD3 and -CD28 antibodies (Left) or strepavidin (Right). (E) Representative (of four experiments) pErk responses in CD4+ T cells from BDC2.5 TCR transgenic mice stimulated with mimotope peptide in the presence of antigen-presenting cells (APCs). (F) Calcium flux induced in CD4+ T cells by biotinylated anti-CD3/CD28 antibodies (first arrow), streptavidin cross-linking (second arrow), and control ionomycin (third arrow) (representative of two independent experiments). (G) Changes in MFI of pCD3ζ, pSLP76, and pAKT in CD4+ Tconv and Treg cells after streptavidin cross-linking of CD3 and CD28 (representative of three experiments).

Fig. S1.

Fig. S1.

(A) Computing the pErk signal off-rates (Fig. 1C) via an exponential fit. (B) Flow cytometry data for induced pErk at different time points after cross-linking of CD3 and CD28 in Tconv and Treg cells from B6 mice (representative of five independent experiments). (C) Plot and histogram of CD4+ T cells at different time points after BDC2.5 mimotope agonist peptide stimulation from BDC2.5 TCR transgenic mice (representative of four independent experiments).

This lower response in Treg cells was not due to a requirement for a stronger activation signal, as it was observed across a range of doses of anti-CD3/28 or of cross-linking streptavidin (Fig. 1D). To extend these observations, we also assessed Erk1/2 phosphorylation in T cells from BDC2.5 TCR transgenic mice, stimulated with whole splenocytes loaded with mimotope agonist peptide (26). The response was more persistent than that elicited by antibody-mediated cross-linking, yet the same Treg/Tconv difference was present with this more physiological mode of activation (Fig. 1E and Fig. S1C).

We then compared the response in Treg and Tconv cells along different signaling pathways and nodes downstream of the TCR. In agreement with ref. 22, the increase in intracytoplasmic Ca++ was strongly reduced in Treg relative to Tconv (Fig. 1F). Reduced activation was observed in Treg cells for p-CD3ζ (Y142), indicating that lower signaling efficacy was already manifest at the apex of the signaling cascade, and for Treg p-SLP76 (Y128), p-AKT (Ser473), and p-S6 (S235) (Fig. 1G), indicating that dampened signals are found throughout the different signaling branches.

Correlations of Dampened TCR Signals in Treg Cells.

Given this difference in TCR-induced signals in Tconv and Treg cells, we asked whether it might be tied to some of the known phenotypic differences between these lineages. Most immediately, Treg and Tconv express slightly different levels of TCR, and this quantitative difference could potentially account for the different signal intensities. This was not the case, however. When the response to anti-CD3/CD28 stimulation was parsed by analyzing cells binned across matched quantiles of surface TCR (Fig. 2A, Left), Treg cells showed lower pErk than Tconv cells, at all TCR levels (Fig. 2A).

Fig. 2.

Fig. 2.

Phenotypic correlates of dampened TCR signals in Treg cells. (A) pErk responses in Tconv and Treg cells (Left), in bins of cells matched for TCR surface levels (Right); anti-CD3/CD28 cross-linking is as in Fig. 1 and is representative of four independent experiments. (B) pErk induction versus CD44 level, for TCRβ+CD4+ Tconv and Treg cells gated from the CD90.1 reporter. (C) Different naïve/memory states for Treg and Tconv cells were distinguished (Top), and the pErk responses of these gated cells were tracked over time after anti-CD3/-CD28 cross-linking (representative of three independent experiments).

Responses to TCR engagement are known to be less intense in memory than in naïve T cells (27), and this could be an important confounder, as a significant fraction of Treg cells have a memory phenotype (2830). We used the expression of CD44 and CD62L molecules to distinguish between naïve and memory phenotypes during the response to CD3/CD28 cross-linking. As expected, naïve CD44lo Tconv cells responded more than CD44hi Tconv (Fig. 2B, Left), and naïve CD62Lhi Tconv cells responded more than CD62Llo Tconv (Fig. S2). The same pattern was observed for Treg cells, and it was thus clear that a good part of the Treg/Tconv signaling difference could be explained by the higher proportion of CD44hi low responders among the Treg pool. This bias did not provide the entire explanation for the Treg/Tcov signaling difference, however, as Treg cells responded less intensely even in matched ranges of CD44 expression (Fig. 2B). For quantitation, we gated the three main populations defined by CD44 and CD62L. For CD44loCD62Lhi naïve cells that dominated among Tconv or for CD44loCD62Llo that were more frequent among Treg cells (Fig. 2C, Top), responses were less intense in Treg cells than in Tconv (Fig. 2C, Bottom). They were uniformly low for CD44+CD62Llo effector/memory cells. Thus, although the predominant antigen-experienced phenotype of Treg cells explains part of their lower phosphorylation response to TCR engagement, this characteristic does not provide the whole explanation.

Fig. S2.

Fig. S2.

Flow cytometry data for induced pErk at different time points after cross-linking of CD3 and CD28 in Tconv and Treg cells versus CD62L level (representative of three independent experiments).

Sluggish Phosphorylation Cascades Are Not a General Treg Trait.

Thus, Treg cells show a generally dampened response to activation through the TCR. We then asked whether this is a general characteristic of phosphorylation cascades in Treg cells, possibly due to high expression of inhibitory phosphatases, by testing responses to a range of cytokines (Fig. 3). Responses to cytokine stimulation in vitro were assessed by measuring the phosphorylation of different STAT transducers. In the presence of type I IFN (IFNα or IFNβ), Treg cells showed a significantly higher degree of STAT1 phosphorylation than Tconvs at all time points (Fig. 3A). In contrast, Tconv cells showed a stronger pSTAT1 response to IFNγ than Treg cells (Fig. 3A, Right).

Fig. 3.

Fig. 3.

Robust cytokine signaling in Treg cells. (A) pSTAT1 responses in Tconv and Treg cells stimulated with IFN-α (50 U/mL) or IFN-γ (50 ng/mL). (Bottom Right) Combined results from five independent experiments, shown as pSTAT1 fold change (MFI at 15 min/MFI at baseline). (B) Responses in Treg and Tconv to IL6, as pSTAT3 (Left) or pSTAT1 (Right) through different doses of IL6. (C) pSTAT5 response to IL2 (200 U/mL).

For IL6, there was an interesting variation according to the dose and the signaling molecule: At low doses of IL6, Treg and Tconv showed equivalent STAT3 phosphorylation over time, but the higher level and faster appearance of pSTAT3 at higher doses of IL6 (50 ng/mL) were only manifest in Treg cells. In contrast, STAT1 phosphorylation elicited by IL6 (most convincingly at the higher dose) was equivalent in Treg and Tconv, suggesting that the STAT3 divergence at these high doses was not due to receptor saturation (Fig. 3B, Right). These subtle divergences are in line with the complexity of IL6’s impact in T cells (31) and suggest that the location or configuration of the IL6 receptor, or its immediate signaling assembly, must vary between CD4+ lineages (Stat genes are equally expressed in Treg and Tconv cells; www.immgen.org).

Finally, as expected from the characteristic overexpression of CD25 (Il2ra) in Treg cells, Il2 elicited much stronger pSTAT5 activation in Treg than in Tconv (Fig. 3C), in keeping with previous reports (ref. 32 and references therein). These results indicate that there is complex variability in the sensitivity of Treg and Tconv cells to cytokine signals but that there is no overall Treg unresponsiveness, as might have been suggested by the low TCR-induced responses.

Origin of the Dampened TCR Signals in Treg Cells.

We next attempted to track the root of different signaling downstream from the TCR in Tconv and Treg cells. We considered as plausible candidates several inhibitory molecules known to be overexpressed in Treg cells, likely as a form of negative feedback induced by the frequent self-reactivity of their TCR. The first candidate was CD5, a cell-surface protein of the “scavenger receptor cysteine rich” family, with a large cytoplasmic domain that acts as a modulator of antigen receptor signaling in T and B lymphocytes, in part by recruiting negative regulators such as SHP1 (reviewed in ref. 33). The amount of CD5 displayed on the cell surface is proportional to the intensity of signals received through the TCR, and CD5 is robustly expressed on Treg cells (34, 35). To test whether CD5 more specifically inhibits signaling in Treg cells, we analyzed Erk phosphorylation after CD3/CD28 cross-linking across five matched ranges of surface CD5 (Fig. 4A), which revealed interesting cell specificity. For CD8+ T cells, the relation between CD5 and Erk activation was very marked, with quasi-complete absence of pErk in cells with the most CD5. For CD4+ Tconv, in contrast, CD5 seemed to have much less relevance, and strong responses were observed at essentially all levels of CD5. Treg cells were somehow intermediate, with some relation between Erk response and CD5 levels, albeit not as strong as in CD8+ T cells. It was clear from this comparison, however, that the Treg/Tconv differential response could not be explained by CD5, as it was still observed between cells of matched CD5 levels (Fig. 4A).

Fig. 4.

Fig. 4.

Origin seeking of the dampened TCR signals in Treg cells. (A) pERK responses conditioned on different levels of CD5 (Left) in CD8+ T cells and Tconv and Treg cells after anti-CD3/CD28 cross-linking (representative of three independent experiments). (B) Responses to anti-CD3/CD28 cross-linking in CD4+ T cells (Treg cells distinguished from Tconv by anti-FoxP3 staining, y axis) in mixed hematopoietic chimeras generated in Rag1−/− recipients with an equal proportion of WT and Ctla4−/− (Left) or Pdcd1−/− (Right) bone marrow; WT cells were distinguished with the CD45.1 congenic marker (representative of two mice in two independent experiments). (C) Quantitation of the pErk or pCD3ζ MFI in the experiment shown in B.

CTLA-4 and PD-1 are coinhibitory receptors that negatively regulate T-cell activation, if by means that are not completely established (3638). Both are overexpressed in Treg cells. Because their deletion or blockade leads to broad and potentially confounding T-cell activation and lymphoproliferation, we constructed mixed bone marrow chimeras by mixing equal numbers of congenically marked bone marrow from WT (CD45.1) and Ctla4- or Pdcd1-knockout mice (CD45.2) before transfer into irradiated hosts. In the resulting chimeras, the WT component effectively prevents disease. Eight weeks after transplantation, splenocytes were challenged as above. The activation profiles for p-Erk1/2 or p-CD3ζ proved to be essentially superimposable for WT or deficient cells (Fig. 4B), indicating that these coinhibitory receptors do not affect TCR-mediated signals in this system and that their higher expression in Treg cells is unrelated to the lower signaling intensity.

Dusp4 Modulates Specific Facets of Treg Function and Homeostasis.

The last candidate we examined as a signaling regulator in Treg cells was the Y/T dual-acting DUSP4. Its best recognized substrate is pERK (39, 40), although the full breadth of its substrates is not well charted. Dusp4 is strongly overexpressed in Treg cells, which might plausibly account for dampened TCR-induced signals in Treg cells. We thus analyzed responses in cells from Dusp4-deficient mice (41), in which Treg cells are present in normal proportions and which have no autoimmune or immune dysregulation manifestations. Contrary to our hypothesis, ERK phosphorylation dynamics in Dusp4-deficient T cells proved essentially superimposable to those of cocultured WT cells, for both Treg and Tconv (Fig. 5A).

Fig. 5.

Fig. 5.

Treg phenotypes in Dusp4-deficient mice. (A) pERK and pCD3ζ responses to anti-CD3/CD28 cross-linking in Tconv and Treg cells from Dusp4−/− or WT control mice, mixed in the same reaction tube (representative of three independent experiments). (B) Volcano plots (fold change vs. t test P value) comparing gene expression in WT Treg versus Dusp4−/− Treg cells. Treg signature genes are highlighted in red (induced) or blue (repressed). (C) Volcano plot comparing gene expression in TCR+ and TCR-deleted Treg cells [data from Levine et al. (16)], where red highlights are Dusp4-sensitive transcripts in the present data (defined as fold change > 1.5 and P value < 0.01, per B) (D) Frequencies of CD4+CD25+Foxp3+ Treg cells in different organs of mixed hematopoietic chimeras from mixed WT and Dusp4−/− bone marrow. Data are pooled from two to four independent experiments; each dot is an individual mouse. (E) Naive CD4+ T cells from spleens of WT and Dusp4−/− mice were sorted and stimulated with anti-CD3/CD28 beads in the presence of 20 U/mL IL2, with graded concentrations of TGF-β for 5 d (representative of two independent experiments).

However, because the strong bias in Dusp4 expression made it interesting to track its putative targets, we pursued this analysis by generating gene expression profiles of Treg cells purified from these Dusp4 KO mice, in comparison with those of cohoused B6 mice. The profiles were very similar, with a distribution of Treg signature genes that was mostly in the normal range, save for a small subset of transcripts that were underexpressed in Dusp4−/− Treg cells (22 genes, at fold change and t test P value thresholds of 1.5 and 0.05, respectively; Table S1). These included several markers typical of activated or tissue Treg populations, such as Klrg1 or Il1rl1 (which encodes the Il33 receptor). Several groups have analyzed the homeostasis of Treg cells in which expression of the TCR was terminated by Tamoxifen-controlled germ-line engineering in mature Treg cells (15, 16) and identified a set of Treg signature genes whose expression is dependent on the continued presence of the TCR. Interestingly, the set of DUSP4-sensitive genes identified here showed stronger dependence on TCR signals than the average [Fig. 5C; Kolmogorov–Smirnov P = 10−8 and 10−9, respectively, in the data of Levine et al. and Vahl et al. (15, 16)]. This overlap indicated that DUSP4, perhaps paradoxically, positively modulates the transcriptional consequences of TCR signals in Treg cells.

Table S1.

Dusp4-induced and -suppressed genes in Treg cells

Probe set ID Gene symbol WT no. 1 WT no. 2 Dusp4 KO no. 1 Dusp4 KO no. 2 Fold change WT/Dusp4 KO P value Fold change Treg/Tconv
Induced*
 10353450 Gm4956 1,306 1,151 60 65 19.65 0.00062 9.30
 10425321 Apobec3 2,176 1,938 386 363 5.49 0.00149 1.70
 10357986 Rab6b 638 468 104 97 5.44 0.00871 1.50
 10345436 LOC280487 270 279 71 72 3.86 0.00017 0.99
 10459066 Gm4841 302 219 82 69 3.42 0.02095 1.91
 10430649 Cbx7 1,629 1,686 555 559 2.97 0.00026 1.48
 10357488 Cd55 541 514 168 197 2.90 0.00615 0.52
 10565994 Art2b 3,326 3,209 1,291 1,006 2.87 0.01406 1.37
 10430647 D730005E14Rik 507 427 157 174 2.81 0.00909 1.59
 10578361 Asah1 406 421 141 176 2.63 0.01339 1.02
 10366546 Cpm 669 557 284 231 2.39 0.02459 0.94
 10444821 H2-Q8 2,656 2,138 1,002 1,090 2.28 0.01937 1.44
 10363070 Gp49a 475 382 183 200 2.23 0.02090 2.59
 10511258 Fam132a 350 336 150 159 2.22 0.00200 1.42
 10579347 Ifi30 380 371 185 161 2.18 0.00842 0.96
 10491952 Mgst2 340 288 148 154 2.07 0.01353 1.24
 10476939 Gm4979 670 690 338 322 2.06 0.00165 1.35
 10523595 Ptprv 343 276 160 149 2.00 0.02619 1.02
 10511363 Penk 3,368 3,533 1,648 1,825 1.99 0.00665 12.97
 10573430 Gadd45gip1 776 770 461 344 1.94 0.04499 1.21
 10565735 A630091E08Rik 168 190 92 100 1.86 0.01460 4.20
 10411611 Naip5 185 218 112 108 1.83 0.01858 4.77
 10572235 Lpar2 228 195 123 112 1.80 0.02298 1.18
 10547590 Klrg1 1,988 1,719 1,140 933 1.79 0.04234 13.30
 10455954 Gm4951 299 276 163 159 1.78 0.00530 2.36
 10490246 Gm14326 1,019 1,051 563 606 1.77 0.00462 1.34
 10440131 Gpr15 335 318 206 174 1.73 0.02476 1.06
 10565775 Dgat2 121 138 83 68 1.72 0.04539 1.44
 10450682 H2-T23 2,564 2,413 1,523 1,391 1.71 0.01015 1.37
 10363082 Lilrb4 560 507 318 305 1.71 0.00994 2.87
 10515690 Wrn 301 283 176 169 1.69 0.00472 1.08
 10444824 H2-Q6 4,753 4,475 2,664 2,842 1.68 0.00724 1.49
 10574023 Mt2 244 265 144 161 1.67 0.01785 1.49
 10450675 H2-T24 692 648 439 369 1.66 0.03201 0.91
 10573483 Prdx2 1,319 1,338 843 792 1.63 0.00432 1.78
 10572449 Lsm4 1,482 1,491 931 909 1.62 0.00070 1.20
 10519857 Hgf 102 103 63 66 1.59 0.00318 1.89
 10406614 Mtx3 148 165 104 93 1.59 0.02722 0.76
 10507137 Pdzk1ip1 305 283 189 182 1.59 0.00811 1.29
 10534253 Gtf2ird1 212 182 128 120 1.58 0.03121 1.00
 10588283 Szt2 127 105 73 74 1.57 0.04060 0.97
 10355246 Acadl 377 389 247 242 1.57 0.00161 0.98
 10555041 Alg8 409 380 257 247 1.56 0.00903 0.98
 10360373 E030037K03Rik 636 550 401 363 1.55 0.03868 0.71
 10384458 Plek 173 170 116 106 1.54 0.00942 1.10
 10580160 Mri1 393 402 253 263 1.54 0.00250 1.09
 10430626 Npcd 186 200 123 129 1.53 0.01069 1.45
 10579602 B3gnt3 127 150 88 94 1.52 0.04199 1.19
 10356461 Hjurp 488 455 334 290 1.52 0.03421 0.92
 10450694 H2-T22 1,008 947 705 591 1.51 0.04697 1.31
 10356999 Ptpn13 379 415 257 270 1.51 0.01602 2.88
Suppressed
 10572741 Olfr372 112 120 286 418 0.33 0.0296 1.51
 10560719 2210010C17Rik 353 296 656 759 0.46 0.0205 1.17
 10458589 Prelid2 182 164 307 282 0.59 0.0155 1.36
 10458033 Stard4 184 207 343 334 0.58 0.0119 1.08
 10518364 Rps19-ps3 139 151 249 243 0.59 0.0068 0.99
 10450723 H2-T10 493 451 1,134 1,027 0.44 0.0064 1.11
 10362674 Rnu3a 536 581 1,034 1,019 0.54 0.0045 1.43
 10572591 Ocel1 309 290 678 624 0.46 0.0043 1.12
 10367744 Ust 170 153 1,409 1,304 0.12 0.0010 0.33
 10571274 Gsr 372 366 648 655 0.57 0.0003 1.13
 10579070 Zfp868 170 169 304 303 0.56 0.0000 0.90
*

Expression values and fold change of transcripts that are differentially represented in WT versus Dusp4 KO Treg cells (at a fold change > 1.5 and P value < 0.05).

Expression values and fold change of transcripts that are differentially represented in WT versus Dusp4 KO Treg cells (at a fold change < 0.6 and P value < 0.05).

To better understand the role of Dusp4 in Treg homeostasis, we constructed mixed hematopoietic chimeras, mixing congenically marked bone marrow from WT and KO donors. In most lymphoid compartments, the proportion of Treg cells among CD4+ T cells was equivalent (and the ratio of cells from both donors equivalent across Treg and Tconv compartments), with the exception of the colon, in which Dusp4-deficient Treg cells seemed at a competitive disadvantage (Fig. 5E; in unmanipulated Dusp4-knockout mice, this defect was not as apparent). We also assessed the impact of DUSP4 on iTreg generation, with cocultures in which naïve CD4+ T cells of WT and KO origin were activated in the presence of IL2 + TGFβ. Equivalent responses were observed for WT and Dusp4-deficient cells in those conditions (Fig. 5F).

Discussion

We have investigated the differential activation of TCR signaling cascades in Tconv and Treg cells and sought the origin of the difference. The results indicate that, relative to Tconv counterparts, Treg cells have a broadly dampened activation of several pathways and signaling nodes upon TCR-mediated activation. In contrast, Treg cells were very responsive to several cytokines, painting a picture of a cell population more attuned to generic intercellular communication cues than to antigen-driven signals, in line with the relative dispensability of their TCR (15, 16).

The difference in signaling efficacy appears to start from the apex of the signaling chain, manifest at the level of CD3ζ phosphorylation, and radiate from there along several signaling pathways. In a previous study, we showed ineffective activation of activation modules after TCR triggering in Tconv cells from NOD mice, with computational analysis highlighting an amplification of small initial differences as the signal propagates down the signaling cascades (24). Here, the more robust differences were also detected at downstream nodes (pS6, pErk, Ca++ flux), suggesting that, in this respect, Treg cells resemble NOD Tconv cells. One caveat, related to the functional consequences of these observations, is that it is not clearly known how the differences in protein phosphorylation actually translate into transcriptional or other cell activation readouts, whether the signaling dynamics are linear or whether increased proportions of phosphorylated intermediates are immaterial once a threshold has been reached. Indeed, the range and intensity of early transcriptional responses to anti-CD3/CD28 cross-linking are not grossly different in Treg and Tconv cells (42).

Although their repertoire is in part molded by interaction with self, Treg cells also partake strongly in modulating responses to microbial and environmental nonself, as during responses to infection (43). In this context, one might speculate that the signaling imbalance observed here allows initial responses to develop more effectively, as naïve Tconv would be more sensitive to TCR-mediated activation by foreign antigens, which Treg cells would comparatively ignore. Additionally, the continuous activation of Treg cells could be harmful to the host.

Much, albeit not all, of the dampened signaling in Treg cells could be ascribed to their higher proportion of CD44hi effector/memory cells (2830). Signaling cascades are less readily activated via the TCR in such cells (27), and this distinction also applies to Treg cells. The likeliest interpretation is that prior TCR engagement, by cognate antigen for effector/memory cells or by self-antigen for Treg cells, engages one or more of the negative feedback loops that tune TCR signals. We attempted to identify the molecular nature of this putative negative regulator. It seemed unlikely to be THEMIS (44), because it strongly underexpressed in Treg cells. SOCS1 also seemed an unlikely candidate, even though it is required for Foxp3 expression and Treg specification (45), because SOCS1 overactivity would dampen responses to IFN, which was clearly not the case. The coinhibitory receptors PD1 and CTLA4 are overexpressed in Treg cells, and some reports have suggested an effect of CTLA4 on signal transduction (46), but this was not observed in our experimental system, where adventitious effects of generalized inflammation due to the mutations were avoided by the mixed chimera context. Similarly, the inhibitory phosphatase DUSP4, known to dampen Erk and other kinases, could not account for Treg hyporesponsiveness, although this exploration did allow us to define an interesting set of Treg signature genes whose expression was enhanced by DUSP4.

Consistent with the correlation between CD5 expression and TCR signal strength, expression of CD5 is comparatively high in Treg cells (35, 44). A recent study showed that CD5 facilitates the extrathymic differentiation of pTreg cells by blocking mTOR-dependent signals induced by effector-differentiating cytokines that otherwise inhibit Treg cell induction (35). However, our results showed that Treg/Tconv differential response was not due to CD5 expression. On the other hand, we noted an interesting diversity in the relationship between CD5 levels and signaling strength. Most sensitive were CD8+ T cells, whose degree of Erk activation seemed highly dependent on CD5 levels, whereas signals in CD4+ Tconv cells seemed far less dependent. This divergence may denote differential recruitment of CD5 to the signaling synapse in these lineages or the association between CD5 and Lck, perhaps more impactful in CD8+ T cells as CD8 does not associate directly with Lck, as CD4 does.

In conclusion, we have demonstrated that Tconv and Treg cells respond with different balances to TCR and cytokine stimulation, with Treg cells being more attuned to cytokine- than to TCR-driven cues.

Materials and Methods

Mice and Flow Cytometry.

Animal experiments were performed under Harvard Medical School Institutional Animal Care and Use Committee-reviewed protocol 02954. C57BL/6J, B6.CD45.1 congenic, and B6;129-Dusp4tm1Jmol/J mice (stock 23671) were obtained from the Jackson Laboratory. B6.Foxp3-Cd90.1 mice were kindly provided by Alexander Y. Rudensky, Memorial Sloan Kettering Cancer Center, New York (25). Mice were maintained in specific pathogen-free facilities at Harvard Medical School (Institutional Animal Care and Use Committee protocol 02954). Ctla4- and Pdcd1-deficient mice (B6 background) were a kind gift from P. Sage and A. Sharpe, Harvard Medical School. For Treg analysis, single-cell suspensions from lymphoid organs were stained with antibodies against TCR-β, CD4, CD8 (Biolegend), and FoxP3 (eBioscience) and analyzed on BD LSRII, and data analysis was performed with FlowJo software (Tree Star).

TCR Activation and Phospho-Flow Cytometry.

Spleen cell suspensions from Foxp3-Thy1.1 or chimeric mice were stimulated in medium containing 6 μg/mL biotinylated anti-CD3ε and anti-CD28 stimulatory antibodies and incubated for 2 min at 37 °C before the addition of 24 μg/mL streptavidin. At various times after cross-linking, the stimulation was stopped by the addition of paraformaldehyde to 2% (wt/vol) (room temperature for 20 min). For stimulation of BDC2.5 T cells, BDC2.5 mimotope peptide (Peptide 2.0 Inc.) was added to 100 ng/mL PFA. Fixed cell suspensions were permeabilized by adding ice-cold 100% methanol slowly to prechilled cells, while gently vortexing (1 mL per sample). After overnight incubation at –20 °C, cells were washed with staining buffer and stained with antibodies against TCR-β, CD4, FoxP3, p-Erk1/2 (Y204), p-S6 (Ser235/236) (Cell Signaling Technology), p-CD3ζ (Y142), or p-SLP76 (Y128) (BD Biosciences).

For cytokine responses, cells were incubated at 37 °C with recombinant murine INFγ (0.5–50 ng/mL; R&D Biosciences), INFα (50 U/mL; PBL), INFβ (50 U/mL; PBL), IL6 (0.5–50 ng/mL; Biolegend), and IL2 (200 U/mL; Peprotech). After stimulation, cells were immediately fixed with paraformaldhyde (2% final concentration) for 20 min at room temperature, permeabilized with cold 90% methanol (14 h, −20 °C), washed, and stained with phospho-specific antibodies to p-STAT5 (pY694; BD), pSTAT3 (pY705; BD), and p-STAT1 (pY701; BD).

In Vitro Conversion Assay.

Naïve T cells were activated with anti-CD3/CD28–coated beads (Invitrogen) at a concentration of one bead per cell in the presence of 20 U/mL of recombinant human IL2 (Proleukin; Chiron) and graded doses of recombinant TGF-β (PeproTech).

Mixed Bone Marrow Transfers.

Total bone marrow cells (1 × 106) were isolated from WT (CD45.1), Ctla4, or Pdcd1-deficient mice (CD45.2) and depleted of mature T cells by negative selection of Thy1.2+ cells (Miltenyi Biotech). The two populations were mixed at a 1:1 ratio and were i.v.-transferred into irradiated (1,000 rad) RAG-1−/− mice (6–8 wk of age). Four weeks after irradiation, antibiotic treatment was stopped until flow cytometric analysis at 6–8 wk of age.

Gene Expression Analysis.

RNA was prepared (in biological duplicates) as described (47) from WT-Dusp4 chimera Treg cells. For microarray analysis, RNA was labeled and hybridized to GeneChip Mouse Genome M1.0 ST chip arrays. Raw reads were processed with Affymetrix software, and data were normalized using the robust multipoint averaging algorithm in GenePattern software package. Data were analyzed with the “Multiplot” module from GenePattern.

Acknowledgments

We thank K. Hattori, G. Buruzala, and A. Rhoads for help with mice, flow, and microarrays. Interpretation of results benefited from data assembled by the ImmGen consortium. This work was supported by NIH Grant R37-AI051530 and The JPB Foundation.

Footnotes

The authors declare no conflict of interest.

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1520393112/-/DCSupplemental.

References

  • 1.Josefowicz SZ, Lu LF, Rudensky AY. Regulatory T cells: Mechanisms of differentiation and function. Annu Rev Immunol. 2012;30:531–564. doi: 10.1146/annurev.immunol.25.022106.141623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Feuerer M, Hill JA, Mathis D, Benoist C. Foxp3+ regulatory T cells: Differentiation, specification, subphenotypes. Nat Immunol. 2009;10(7):689–695. doi: 10.1038/ni.1760. [DOI] [PubMed] [Google Scholar]
  • 3.Benoist C, Mathis D. In: Immune Tolerance. Mathis D, Rudensky A, editors. Cold Spring Harbor Press; Cold Spring Harbor, NY: 2013. pp. 31–44. [Google Scholar]
  • 4.Li X, Zheng Y. Regulatory T cell identity: Formation and maintenance. Trends Immunol. 2015;36(6):344–353. doi: 10.1016/j.it.2015.04.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Ramsdell F, Ziegler SF. FOXP3 and scurfy: How it all began. Nat Rev Immunol. 2014;14(5):343–349. doi: 10.1038/nri3650. [DOI] [PubMed] [Google Scholar]
  • 6.Burzyn D, Benoist C, Mathis D. Regulatory T cells in nonlymphoid tissues. Nat Immunol. 2013;14(10):1007–1013. [Google Scholar]
  • 7.Hsieh CS, Zheng Y, Liang Y, Fontenot JD, Rudensky AY. An intersection between the self-reactive regulatory and nonregulatory T cell receptor repertoires. Nat Immunol. 2006;7(4):401–410. doi: 10.1038/ni1318. [DOI] [PubMed] [Google Scholar]
  • 8.Pacholczyk R, Ignatowicz H, Kraj P, Ignatowicz L. Origin and T cell receptor diversity of Foxp3+CD4+CD25+ T cells. Immunity. 2006;25(2):249–259. doi: 10.1016/j.immuni.2006.05.016. [DOI] [PubMed] [Google Scholar]
  • 9.Wong J, et al. Adaptation of TCR repertoires to self-peptides in regulatory and nonregulatory CD4+ T cells. J Immunol. 2007;178(11):7032–7041. doi: 10.4049/jimmunol.178.11.7032. [DOI] [PubMed] [Google Scholar]
  • 10.Krajina T, Leithäuser F, Reimann J. MHC class II-independent CD25+ CD4+ CD8alpha beta+ alpha beta T cells attenuate CD4+ T cell-induced transfer colitis. Eur J Immunol. 2004;34(3):705–714. doi: 10.1002/eji.200324463. [DOI] [PubMed] [Google Scholar]
  • 11.Jordan MS, et al. Thymic selection of CD4+CD25+ regulatory T cells induced by an agonist self-peptide. Nat Immunol. 2001;2(4):301–306. doi: 10.1038/86302. [DOI] [PubMed] [Google Scholar]
  • 12.van Santen HM, Benoist C, Mathis D. Number of T reg cells that differentiate does not increase upon encounter of agonist ligand on thymic epithelial cells. J Exp Med. 2004;200(10):1221–1230. doi: 10.1084/jem.20041022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Moran AE, et al. T cell receptor signal strength in Treg and iNKT cell development demonstrated by a novel fluorescent reporter mouse. J Exp Med. 2011;208(6):1279–1289. doi: 10.1084/jem.20110308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Kim JK, et al. Impact of the TCR signal on regulatory T cell homeostasis, function, and trafficking. PLoS One. 2009;4(8):e6580. doi: 10.1371/journal.pone.0006580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Vahl JC, et al. Continuous T cell receptor signals maintain a functional regulatory T cell pool. Immunity. 2014;41(5):722–736. doi: 10.1016/j.immuni.2014.10.012. [DOI] [PubMed] [Google Scholar]
  • 16.Levine AG, Arvey A, Jin W, Rudensky AY. Continuous requirement for the TCR in regulatory T cell function. Nat Immunol. 2014;15(11):1070–1078. doi: 10.1038/ni.3004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Takahashi T, et al. Immunologic self-tolerance maintained by CD25+CD4+ naturally anergic and suppressive T cells: Induction of autoimmune disease by breaking their anergic/suppressive state. Int Immunol. 1998;10(12):1969–1980. doi: 10.1093/intimm/10.12.1969. [DOI] [PubMed] [Google Scholar]
  • 18.Zanin-Zhorov A, et al. Protein kinase C-theta mediates negative feedback on regulatory T cell function. Science. 2010;328(5976):372–376. doi: 10.1126/science.1186068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Joshi RP, et al. The ζ isoform of diacylglycerol kinase plays a predominant role in regulatory T cell development and TCR-mediated ras signaling. Sci Signal. 2013;6(303):ra102. doi: 10.1126/scisignal.2004373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Collazo MM, et al. SHIP limits immunoregulatory capacity in the T-cell compartment. Blood. 2009;113(13):2934–2944. doi: 10.1182/blood-2008-09-181164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Zanin-Zhorov A, et al. Scaffold protein Disc large homolog 1 is required for T-cell receptor-induced activation of regulatory T-cell function. Proc Natl Acad Sci USA. 2012;109(5):1625–1630. doi: 10.1073/pnas.1110120109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Gavin MA, Clarke SR, Negrou E, Gallegos A, Rudensky A. Homeostasis and anergy of CD4(+)CD25(+) suppressor T cells in vivo. Nat Immunol. 2002;3(1):33–41. doi: 10.1038/ni743. [DOI] [PubMed] [Google Scholar]
  • 23.Li L, et al. CD4+CD25+ regulatory T-cell lines from human cord blood have functional and molecular properties of T-cell anergy. Blood. 2005;106(9):3068–3073. doi: 10.1182/blood-2005-04-1531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Mingueneau M, et al. Single-cell mass cytometry of TCR signaling: Amplification of small initial differences results in low ERK activation in NOD mice. Proc Natl Acad Sci USA. 2014;111(46):16466–16471. doi: 10.1073/pnas.1419337111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Liston A, et al. Differentiation of regulatory Foxp3+ T cells in the thymic cortex. Proc Natl Acad Sci USA. 2008;105(33):11903–11908. doi: 10.1073/pnas.0801506105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Stratmann T, et al. Susceptible MHC alleles, not background genes, select an autoimmune T cell reactivity. J Clin Invest. 2003;112(6):902–914. doi: 10.1172/JCI18337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Farber DL, Acuto O, Bottomly K. Differential T cell receptor-mediated signaling in naive and memory CD4 T cells. Eur J Immunol. 1997;27(8):2094–2101. doi: 10.1002/eji.1830270838. [DOI] [PubMed] [Google Scholar]
  • 28.Huehn J, et al. Developmental stage, phenotype, and migration distinguish naive- and effector/memory-like CD4+ regulatory T cells. J Exp Med. 2004;199(3):303–313. doi: 10.1084/jem.20031562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Sakaguchi S. Naturally arising CD4+ regulatory t cells for immunologic self-tolerance and negative control of immune responses. Annu Rev Immunol. 2004;22:531–562. doi: 10.1146/annurev.immunol.21.120601.141122. [DOI] [PubMed] [Google Scholar]
  • 30.Rosenblum MD, et al. Response to self antigen imprints regulatory memory in tissues. Nature. 2011;480(7378):538–542. doi: 10.1038/nature10664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Scheller J, Chalaris A, Schmidt-Arras D, Rose-John S. The pro- and anti-inflammatory properties of the cytokine interleukin-6. Biochim Biophys Acta. 2011;1813(5):878–888. doi: 10.1016/j.bbamcr.2011.01.034. [DOI] [PubMed] [Google Scholar]
  • 32.Yu A, et al. Selective IL-2 responsiveness of regulatory T cells through multiple intrinsic mechanisms supports the use of low-dose IL-2 therapy in type 1 diabetes. Diabetes. 2015;64(6):2172–2183. doi: 10.2337/db14-1322. [DOI] [PubMed] [Google Scholar]
  • 33.Soldevila G, Raman C, Lozano F. The immunomodulatory properties of the CD5 lymphocyte receptor in health and disease. Curr Opin Immunol. 2011;23(3):310–318. doi: 10.1016/j.coi.2011.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Dasu T, et al. CD5 plays an inhibitory role in the suppressive function of murine CD4(+) CD25(+) T(reg) cells. Immunol Lett. 2008;119(1-2):103–113. doi: 10.1016/j.imlet.2008.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Henderson JG, Opejin A, Jones A, Gross C, Hawiger D. CD5 instructs extrathymic regulatory T cell development in response to self and tolerizing antigens. Immunity. 2015;42(3):471–483. doi: 10.1016/j.immuni.2015.02.010. [DOI] [PubMed] [Google Scholar]
  • 36.Rudd CE, Taylor A, Schneider H. CD28 and CTLA-4 coreceptor expression and signal transduction. Immunol Rev. 2009;229(1):12–26. doi: 10.1111/j.1600-065X.2009.00770.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Walker LS, Sansom DM. The emerging role of CTLA4 as a cell-extrinsic regulator of T cell responses. Nat Rev Immunol. 2011;11(12):852–863. doi: 10.1038/nri3108. [DOI] [PubMed] [Google Scholar]
  • 38.Riley JL. PD-1 signaling in primary T cells. Immunol Rev. 2009;229(1):114–125. doi: 10.1111/j.1600-065X.2009.00767.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Prabhakar S, et al. Targeting DUSPs in glioblastomas—Wielding a double-edged sword? Cell Biol Int. 2014;38(2):145–153. doi: 10.1002/cbin.10201. [DOI] [PubMed] [Google Scholar]
  • 40.Keyse SM. Dual-specificity MAP kinase phosphatases (MKPs) and cancer. Cancer Metastasis Rev. 2008;27(2):253–261. doi: 10.1007/s10555-008-9123-1. [DOI] [PubMed] [Google Scholar]
  • 41.Auger-Messier M, et al. Unrestrained p38 MAPK activation in Dusp1/4 double-null mice induces cardiomyopathy. Circ Res. 2013;112(1):48–56. doi: 10.1161/CIRCRESAHA.112.272963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Wakamatsu E, Mathis D, Benoist C. Convergent and divergent effects of costimulatory molecules in conventional and regulatory CD4+ T cells. Proc Natl Acad Sci USA. 2013;110(3):1023–1028. doi: 10.1073/pnas.1220688110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Belkaid Y, Tarbell K. Regulatory T cells in the control of host-microorganism interactions (*) Annu Rev Immunol. 2009;27:551–589. doi: 10.1146/annurev.immunol.021908.132723. [DOI] [PubMed] [Google Scholar]
  • 44.Gascoigne NR, Acuto O. THEMIS: A critical TCR signal regulator for ligand discrimination. Curr Opin Immunol. 2015;33:86–92. doi: 10.1016/j.coi.2015.01.020. [DOI] [PubMed] [Google Scholar]
  • 45.Takahashi R, et al. 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. 2011;208(10):2055–2067. doi: 10.1084/jem.20110428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Lee K-M, et al. Molecular basis of T cell inactivation by CTLA-4. Science. 1998;282(5397):2263–2266. doi: 10.1126/science.282.5397.2263. [DOI] [PubMed] [Google Scholar]
  • 47.Hill JA, et al. Foxp3 transcription-factor-dependent and -independent regulation of the regulatory T cell transcriptional signature. Immunity. 2007;27(5):786–800. doi: 10.1016/j.immuni.2007.09.010. [DOI] [PubMed] [Google Scholar]

Articles from Proceedings of the National Academy of Sciences of the United States of America are provided here courtesy of National Academy of Sciences

RESOURCES