Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Sep 19.
Published in final edited form as: Nat Immunol. 2009 Mar 8;10(4):427–436. doi: 10.1038/ni.1717

CD27 is a thymic determinant of the balance between interferon-γ- and interleukin 17–producing γδ T cell subsets

Julie C Ribot 1,2, Ana deBarros 1,2, Dick John Pang 3, Joana F Neves 3,4, Victor Peperzak 5, Scott J Roberts 6, Michael Girardi 6, Jannie Borst 5, Adrian C Hayday 7, Daniel J Pennington 3, Bruno Silva-Santos 1,2
PMCID: PMC4167721  NIHMSID: NIHMS583743  PMID: 19270712

Abstract

The production of cytokines such as interferon-γ and interleukin 17 by αβ and γδ T cells influences the outcome of immune responses. Here we show that most γδ T lymphocytes expressed the tumor necrosis factor receptor family member CD27 and secreted interferon-γ, whereas interleukin 17 production was restricted to CD27- γδ T cells. In contrast to the apparent plasticity of αβ T cells, the cytokine profiles of these distinct γδ T cell subsets were essentially stable, even during infection. These phenotypes were established during thymic development, when CD27 functions as a regulator of the differentiation of γδ T cells at least in part by inducing expression of the lymphotoxin-β receptor and genes associated with trans-conditioning and interferon-γ production. Thus, the cytokine profiles of peripheral γδ T cells are predetermined mainly by a mechanism involving CD27.


The past three decades have witnessed intense investigation of T cell generation in the mouse thymus. As a result, insight has been gained into how bone marrow–derived progenitors seed the thymus, rearrange their T cell antigen receptor (TCR) loci, become committed to either the αβ or the γδ T cell lineage and proceed through TCR- mediated selection processes that ensure that ‘useful’ T cells are exported to exert immune functions in the periphery1–3. This notwithstanding, consensus has not been reached on the degree to which intrathymic differentiation events influence peripheral T cell function. For example, although it seems clear that events in the thymus promote the generation of Foxp3+ ‘natural’ regulatory T cells, there is debate over which molecular interactions are involved, with the TCR, CD28 and the lymphotoxin-β receptor (LTβR) all being linked to this1. There is likewise no clear sense of the relative influence of thymic determination versus peripheral ‘plasticity’ and functional ‘conversion’. This is true not only for regulatory T cells but also for the differentiation of helper T cells leading to the production of important cytokines, including interferon-γ (IFN-γ), interleukin 4 (IL-4) and IL-17. In this context, it may also be argued that there has been a disproportionate focus on αβ T cells, when it is increasingly acknowledged that γδ T cells have pleiotropic functional potential and, in arenas such as the production of IFN-γ and IL-17, make critical contributions to host immune competence4,5. Indeed, whereas TCRαβ+ IL-17-producing T helper cells (TH-17 cells) are restricted mainly to the gut6, γδ T cells are a chief source of IL-17 in lymphoid organs and peripheral tissues710. Consequently, there are ongoing attempts to harness the functional potential of γδ T cells in the clinic. Functional T cell subsets are apparently generated by the combined actions of transcription factors. Thus, the transcription factors T-bet and eomesodermin, GATA-3 and c-Maf, and RORγt and Runx1 determine T cell production of IFN-γ, IL-4 and IL-17, respectively11,12. However, their actions can be critically modulated by other molecules, including Notch receptors13. Indeed, there is much biological and clinical interest in the identification of cell surface receptors whose engagement might skew T cell differentiation. In this context, it has been reported that the differentiation of γδ T cells toward IFN-γ production and away from IL-17 production is regulated by specific γδ TCR agonists in the thymus14. Possibly consistent with those findings, particular γδ TCRs are associated with distinct cellular functions15. Those observations notwithstanding, the differentiation of γδ T cells is also regulated by other intrathymic molecular interactions. For example, the differentiation of proliferating, IFN-γ- producing γδ T cells requires TCR-independent interactions between early γδ T cell progenitors and CD4+CD8+ (double-positive (DP)) αβ T cell progenitors. These events, collectively called ‘trans-conditioning’, are determined in part by signals received through the LTβR1618.

On the basis of such perspectives, this paper defines discrete intrathymic progenitors of peripheral IFN-γ- and IL-17-producing γδ T cell subsets, distinguished and regulated by the tumor necrosis factor (TNF) receptor ‘superfamily’ member CD27 (A000546), which engages CD70 (A000547), a TNF-related transmembrane glycoprotein19. CD27+ γδ thymocytes expressed LTβR and genes associated with a T helper type 1 (TH1) phenotype, in contrast to CD27γδ thymocytes, which included the progenitors of IL-17-producing γδ cells. Whereas the relative proportions of the two γδ subsets in the thymus and the periphery were biased toward CD27+ IFN-γ producers, IL-17-producing CD27γδ T cell populations rapidly expanded in response to acute infection such as that posed by malaria.

RESULTS

CD27 expression segregates γδ lymphocyte subsets

Attempts have been made to identify markers of distinct γδ T cell subsets, particularly after the realization that γδ cells are key sources of IL-17 and IFN-γ and contribute to host defense and to certain inflammatory and autoimmune diseases14,2022. Although γδ cells have been subcategorized by their expression of CD25 and CD122 into IL-17 and IFN-γ producers, respectively23, as discussed below, this classification is not generally applicable beyond the peritoneal cavity, and there is no evidence that these markers actively regulate the outcome of T cell differentiation. Here we examine the validity of CD27 as a subset marker for γδ T cells, as CD27 has been linked to the functional segregation of subsets of αβ T cells19,24 and natural killer (NK) cells25.

CD27 expression distinguished two γδ cell subsets in the spleen and lymph nodes (axillary, inguinal, branchial and mesenteric) and in the peripheral tissues (lung or gut) of adult C57BL/6 mice. Overall, 10–30% of γδ cells were CD27 (called ‘γδ27–’ here) and 70– 90% were CD27+ (called ‘γδ27+’ here; Fig. 1a). The two subsets had very different surface phenotypes, with γδ27- cells generally being CD44hiCD62Llo and γδ27+ cells having lower expression of CD44 and higher expression of CD62L (Fig. 1b). They also showed conspicuously little interconversion, as the CD27+ and CD27 phenotypes were maintained even after 3–5 d of continuous stimulation with antibody to CD3ε (anti-CD3ε) in vitro or 2 weeks of residence in lymphopenic mice deficient in the product of recom bination-activating gene 2 (RAG-2; Fig. 1c).

Figure 1.

Figure 1

CD27 expression defines distinct subsets of peripheral γδ T cells. (a) Flow cytometry of cells obtained from various tissues (above plots) of adult wild- type C57BL/6 mice, stained for CD3ε, CD27 and TCRγδ and gated on CD3ε+TCRγδ+ cells. a+i(LN), axillary and inguinal lymph nodes; mLN, mesenteric lymph nodes. Data are representative of three to eight independent experiments with three to five mice each. (b) Flow cytometry of splenocytes stained for CD3ε, CD27 and TCRγδ, plus CD62L or CD44 and gated on CD3ε+TCRγδ+CD27+ cells (γδ27+) and CD3ε+TCRγδ+CD27cells (γδ27−). Data are representative of three independent experiments with three to five mice each. (c) Flow cytometry of γδ27+ or γδ27− T cells sorted from pooled spleens and lymph nodes and cultured separately for 3 d with antigen-presenting cells and anti-CD3ε (1 mg/ml; left) or injected separately intravenously into RAG-2- deficient mice and analyzed after 2 weeks (right), then recovered, stained for CD3ε, CD27 and TCRδ and gated on CD3ε+TCRδ+ cells. Data are representative of three independent experiments. (d–f) Flow cytometry analyzing the proliferation of γδ27+ or γδ27− T cells sorted from pooled spleens and lymph nodes, labeled with CFSE and cultured separately with antigen- presenting cells in presence of various doses of anti-CD3e (horizontal axis; d) or injected intravenously into RAG-2-deficient mice separately (e) or together at ratio of 1:1 (f), and assessed on the basis of CFSE-dilution kinetics at day 3. Data are representative of four independent experiments.

These experiments also showed that the proliferation of γδ27+ cells after TCR stimulation in vitro exceeded that of γδ27– cells (Fig. 1d) and that γδ27+ cells had a considerable advantage over γδ27– lymphocytes when these cells were injected together into lymphopenic mice (Figs. 1e,f). Thus, γδ27+ and γδ27– cells constitute distinct and essentially stable subsets of γδ T lymphocytes in naive mice.

Cytokine production segregates with CD27

When comparing the gene expression of splenic γδ27+ and γδ27– cells from naive C57BL/6 mice, we noted considerable polarization toward constitutive expression of Ifnγ and II17, respectively (Fig. 2a). After 4 h of activation in vitro with phorbol 12-myristate 13-acetate (PMA) and ionomycin, γδ27+ cells had typical TH1-like characteristics, including abundant intracellular IFN-γ and TNF but no IL-17; in contrast, γδ27– populations had far fewer cells that expressed IFN-γ and TNF but had a conspicuous population of IL-17-producing cells (Fig. 2b and Supplementary Fig. 1a online). Neither γδ27+ nor γδ27– cells had substantial expression of IL-4, IL-13 or IL-10 in the conditions used, but they had similar cytolytic capacity (Supplementary Fig. 1a,b and data not shown). Independent reports have cited an association of CD122 expression with IFN-γ-producing γδ T cells14,23 and of the scavenger receptor SCART2 with IL-17-secreting γδ T cells26. Consistent with those findings, expression of CD122 and SCART2 segregated with γδ27+ and γδ27– cells, respectively (Fig. 2c,d). However, the subsets did not overlap perfectly. Thus, many γδ27+ cells were CD122, and a substantial percentage of CD122 cells also produced IFN-γ after short-term activation (Supplementary Fig. 1c). Indeed, as most γδ T cells are CD122, over 50% of IFN-γ-producing γδ T cells may have been CD122 whereas less than 5% were CD27 (Fig. 2b,d) Hence, CD27 expression seemed to segregate much more accurately with IFN-γ production by γδ T cells than did CD122. The degree to which SCART2-expressing cells overlap with γδ27– cells cannot be assessed until reagents facilitating the detection of SCART2 by flow cytometry become generally available.

Figure 2.

Figure 2

CD27 expression segregates IFN-γ- versus IL-17-producing γδ cells in naive and malaria-infected mice. (a) Quantitative real-time PCR of the expression of Ifnγ and II17 by γδ27+ and γδ27− T cells sorted from pooled spleens and lymph nodes of naive C57BL/6 mice, normalized to Efa1 expression and presented as the percent of maximum expression of the gene of interest. (b) Intracellular staining for IFN-γ or IL-17 in γδ27+ and γδ27− T cells sorted from pooled spleens and lymph nodes of naive or Plasmodium berghei ANKA–infected C57BL/6 mice (at day 4 after infection), then stimulated for 4 h with PMA and ionomycin. FSC, forward scatter. Numbers adjacent to outlined areas indicate percent IFN-γ+ cells (top row) or IL-17+ cells (bottom row). (c) Quantitative real-time PCR of the expression of II2rb (encoding CD122) and the gene encoding SCART2 (National Center for Biotechnology Information identification number 5830411N06Rik; called ‘Scart2’ here) by γδ27+ and γδ27− T cells, as described in a. (d) Flow cytometry of splenocytes from naive mice, stained for CD3ε, CD27, TCRγδ and CD122 and gated on CD3ε+TCRγδ+ cells. Numbers in quadrants indicate percent cells in each. (e) Absolute number of total γδ T cells (left) and percent CD69+ cells (middle) and CD44hiCD62Llo cells (right) among γδ27+ and γδ27− T cells from splenocytes obtained from naive mice (0) and mice at days 3 and 6 after infection with P. berghei ANKA and stained for CD3ε, CD27, TCRγδ and CD69, plus CD62L or CD44. (f) Absolute number of total γδ cells (left), γδ27+ T cells (middle) and γδ27− T cells (right) among splenocytes obtained from naive or naive mice and mice at day 4 after infection with ‘titrated’ doses of P. berghei ANKA (5 ×104, 5 ×105 or 5 ×106 parasites per mouse; to achieve variable parasitemia) and stained for CD3ε, CD27 and TCRγδ, presented relative to parasitemia (percent P. berghei ANKA in blood). Each symbol represents an individual mouse (n=4 mice/group). Data are representative of three independent experiments (a,c,e; error bars, s.d.; n=3 samples of four to five mice, pooled) or four to five (b), three (d) or two (f) independent experiments.

To analyze how each γδ subset might respond to infection, we used Plasmodium berghei, because in mice and humans, γδ T cells make important contributions to the response to this pathogen and other apicomplexan parasites2730. We evaluated the spleen and peripheral lymphoid organs of C57BL/6 mice infected with P. berghei (strain ANKA), a model of experimental cerebral malaria that leads to death between 6 and 8 d after infection31. The γδ27+ and γδ27– subsets each proliferated after inoculation with a fixed number of P. berghei ANKA– infected erythrocytes, but with different kinetics. The γδ27+ populations expanded more rapidly (by day 3 after infection); however, by day 6 after infection, γδ27– cells had also increased almost fivefold in number and accounted for approximately 40% of cells in the splenic γδ compartment (Fig. 2e). These findings show that whereas γδ27– cells proliferated relatively poorly in response to stimulation in vitro or during homeostatic expansion in vivo (Fig. 1), they were able to proliferate robustly in response to physiological stimuli in vivo. Moreover, as early as day 3 after infection, approximately 60% of γδ27– cells had an activated CD69+ surface phenotype, compared with only about 20% of γδ27+ cells (Fig. 2e). A higher inoculum of P. berghei ANKA–infected erythrocytes resulted in augmented parasitemia (Supplementary Fig. 2a online) and population expansion of spleen and lymph node γδ T cells that was almost exclusively attributable to γδ27– cells, which at high parasitemia represented over 50% of all γδ T cells (Fig. 2f and Supplementary Fig. 2b). Notably, however, both γδ T cell subsets showed strong conservation of the cytokine (IFN-γ versus IL-17) and surface marker (CD44 and CD62L) phenotypes of naive mice (Fig. 2b,e).

Factors that influence γδ cell IL-17 production

By analyzing cytokine production versus dilution of the cytosolic dye CFSE by total γδ T cells, we confirmed that most IL-17 producers, like γδ27– cells, proliferated poorly after activation in vitro, whereas IFN-γ producers, like γδ27+ cells, proliferated robustly (Supplementary Fig. 3a,b online). Similarly, IL-17 was produced by CD44hiCD62Llo γδ T cells but not by CD44loCD62Lhi γδ T cells (Supplementary Fig. 3c,d). Collectively, these data show that the properties of the γδ27– subset fully resemble those of the IL-17-producing subset of the peripheral γδ T cell pool. Therefore, we considered it appropriate to use γδ27– cells to study the requirements for IL-17 expression. Consistent with reports that IL-17 production by γδ T cells does not require stringent conditioning by transforming growth factor-β (TGF-β), IL-6 or other inflammatory cytokines7,23 required for αβ TH-17 differentiation3234, γδ27– cells expressed IL-17 mRNA immediately after isolation and required only 4 h of treatment with PMA and ionomycin to express IL-17 protein (Fig. 3a,b). Neither TCRαβ+CD4+CD27– effector T cells nor γδ27+ cells produced IL-17 mRNA or protein (Fig. 3a,b). Given that elaboration of IL-17 by γδ27– cells occurs in different conditions than those established for αβ TH-17 cells, we also determined if the TH-17-determining transcription factor RORγt was expressed in γδ27– cells. For this we used a mouse in which green fluorescent protein (GFP) is expressed under the control of Rorc (which encodes RORγt transcriptional elements)6,35. By the criterion of GFP expression, all IL-17-producing γδ27– cells expressed Rorc (Fig. 3c). A small percentage of γδ27+ cells weakly expressed Rorc but did not express IL-17, consistent with data above (Figs. 2b and 3b) and with the generally accepted view that RORγt is necessary but not sufficient for IL-17 induction in αβ T cells11. Nonetheless, we investigated whether the constitutive expression of IL-17 in γδ27– cells could be further upregulated by particular stimuli. Indeed, II17 transcription and IL-17 protein production was augmented in 3-day cultures by activation with anti-CD3e plus IL-2 (in the absence of any other cytokines); this augmentation was further enhanced by TGF-β treatment (Fig. 3a,b). Of note, these stimuli also induced IFN-γ production in a substantial fraction of γδ27– cells (although this fraction was always lower than that of the γδ27+ cell subset; Fig. 3b), which demonstrates some degree of functional plasticity of this population, at least in vitro. That observation notwithstanding, IL-17 expression was achieved without the addition of exogenous IL-6 or IL-21.

Figure 3.

Figure 3

Constitutive expression of IL-17 and RORγt by γδ27− cells. (a) Quantitative real-time PCR analysis of II17 expression by CD4+CD25 αβ T cells (4+25) and γδ27+ or γδ27− T cells sorted from pooled spleens and lymph nodes of C57BL/6 mice and assessed without activation (Fresh) or after activation for 4 d with plate-bound anti-CD3ε plus IL-2, with (+) or without (−) TGF-b, presented as described in Figure 2a. Data are representative of three independent experiments (error bars, s.d.; n=3 mice). (b) Intracellular staining of IFN-γ and IL-17 in CD27+ or CD27 γδ and CD4+CD25αβ T cells sorted from pooled spleens and lymph nodes from C57BL/6 mice, assessed without preactivation (Fresh) or after preactivation for 4 d on plate-bound anti- CD3ε and IL-2 (PreAct) with or without TGF-β, followed by stimulation with PMA and ionomycin. Data are representative of four independent experiments. (c) Intracellular staining for IL-17 and GFP in CD4+CD25, γδ27+ and γδ27− T cells isolated from pooled lymph nodes of RORγt-GFP mice. Data are representative of triplicate experiments with five mice each. (d) Intracellular staining for IFN-γ and IL-17 in flow cytometry–sorted γδ27− and γδ27+ splenocytes activated in vitro for 3 d with anti-CD3ε and IL-2 in the presence (+) of various cytokines (above plots). Data are representative of three independent experiments. Numbers in quadrants (b–d) indicate percent cells in each. (e) Staining for surface Vγ1 or Vγ4 on and intracellular IL-17 in γδ27+ and γδ27− cells sorted from pooled spleens and lymph nodes of C57BL/6 mice, preactivated for 4 d on plate-bound anti-CD3ε plus IL-2 and TGF-β, then stimulated with PMA and ionomycin. Data are representative of three independent experiments.

In addition, the finding that IL-17- producing γδ27– cells lacked transcripts for IL-6 or IL-21 (Supplementary Fig. 4 online) excluded the possibility that these cytokines might be provided in an autocrine way. Likewise, in contrast to the need for antibody blockade of IFN-γ for the optimal polarization of CD4+CD2527– αβ TH-17 cells in vitro7,32 (data not shown), γδ27– cells produced IL-17 even in the presence of IFN-γ-producing cells (Fig. 3b). Moreover, when exogenous IL-6, IL-21 and neutralizing anti-IFN-γ were added to cultures alone or in combination, they had essentially no effect on IL-17 production by TGF-β-treated cells; the addition of IL-23 had only a very small effect (Fig. 3d). Notably, in all conditions tested, most γδ27+ cells failed to produce IL-17 (Fig. 3b,d), which reemphasizes the functional distinction between the two γδ T cell subsets. Although it has been proposed that IL-17 production is more common among cells positive for the variable region Vγ4 than among Vγ1+ cells, conflicting data have left this point contentious and unresolved21,22. In our analyses, IL-17-expressing cells accounted for almost 50% of Vγ4+ γδ27– cells and only about 11% of Vγ1+ γδ27– cells (Fig. 3e). This finding supports the hypothesis that there is an association between the TCR and a bias in the functional differentiation of γδ T cells4,14.

Thymic progenitors of γδ27+ and γδ27– cells

Given their essential stability in naive and infected mice, we assessed whether the γδ27+ and γδ27– subsets were developmentally established in the thymus. We found that TCRγδ+ thymocytes had the highest expression of CD27 of all the T cell–committed subsets (Fig. 4a). However, in contrast to the homogeneous expression of CD27 on mature TCRαβ+ thymocytes, approximately 10% of γδ thymocytes conspicuously lacked CD27 (Fig. 4a). As thymocyte precursors at double-negative stages 2 and 3, from which γδ cells mainly derive1,36,37, express CD25, we used staining for CD25 and CD27 to segregate γδ thymocytes into subsets; 5–10% were CD27+CD25+ (called ‘γδ27,25+’ here), whereas the remaining 90–95% cells were negative for CD25, including 10–15% CD27– cells (γδ27–) and 75–80% CD27+ cells (γδ27+; Fig. 4b). This representation of the subsets was completely similar in independent mouse colonies maintained in two different countries. Moreover, each subset was detectable in the thymus from the earliest embryonic stages tested (embryonic days 14–15 (E14–E15), although the γδ27+ subset greatly expanded between E15 and E18, by which time the relative proportions found in the adult were established (Fig. 4c). The finding that peripheral γδ cells were essentially all CD25– (Fig. 4b) underpins the concern that the proposed use of CD25 to identify IL-17-producing γδ T cells may be applicable only to the peritoneal cavity21 (Supplementary Fig. 5 online) and suggests that the thymic γδ27,25+ subset might constitute a progenitor compartment. Consistent with that hypothesis, γδ27,25+ cells were larger in size than were γδ27+ or γδ27– thymocytes (Fig. 4d), had a CD24hiCD25hiCD62LhiCD44hiCD2loCD5lo ‘immature’ phenotype very similar to that of progenitors at double-negative stages 2 and 3 (Fig. 4e and Supplementary Fig. 6 online), had lower surface expression of TCR than did the γδ27+ and γδ27– subsets (data not shown) and had high proliferative turnover (Fig. 4f,g). In contrast, the surface phenotype and proliferative activity of γδ27+ and γδ27– thymocytes closely mimicked those of their respective peripheral counterparts (Figs. 1b and 4e–g). Next we repopulated fetal thymic reaggregation organ cultures with highly purified γδ27,25+ thymocytes. After 7 d in culture, these γδ27,25+ thymocytes gave rise to both γδ27+ and γδ27– cells (Fig. 4h). This observation confirms that γδ27,25+ thymocytes have the potential to generate γδ27+ and γδ27– cells. Another demonstration of the relationship between the thymic γδ27+ and γδ27– subsets and their peripheral counterparts was evident in their constitutive expression of Ifng and II17, respectively (Fig. 4i). Because this expression was present in E18 thymi, it seems implausible to attribute it to the recirculation of activated peripheral γδ T cells back to the thymus. Likewise, γδ27– cells from E18 thymi already expressed Rorc and its TH-17-driving cofactor Runx1 (ref. 11), whereas fetal γδ27+ cells expressed Tbx21, which encodes T-bet, a chief determinant of TH1 differentiation (Fig. 4j). The idea that such functional segregation was already established in γδ27+ and γδ27– thymocytes, respectively, was also demonstrated by their corresponding expression of IFN-g and IL-17 protein (Supplementary Fig. 7a online). Incubation for 3 d with anti-CD3e and IL-2 supplemented with a ‘cocktail’ of IL-6, IL-21 and IL-23 further increased IL-17 production by γδ27– cells but did not induce IL-17 production by γδ27+ thymocytes (Supplementary Fig. 7b). Thus, the functional potential and constraints of γδ27+ and γδ27– cells seem to be ‘imprinted’ as early as in the fetal thymus.

Figure 4.

Figure 4

Both γδ27+ and γδ27– cells originate from common CD27+CD25+ thymic γδ progenitors. (a) Flow cytometry of thymocytes of C57BL/6 mice (n=3) gated on single-positive stage 4 cells (SP4; CD4+CD8) or γδ cells (TCRγδ+CD3ε). Dashed line, isotype control. (b) Flow cytometry of thymocytes (left) and splenocytes (right) from C57BL/6 mice, stained for CD3ε, CD27, CD25 and TCRγδ, gated on CD3ε+TCRγδ+ cells. Numbers in plots indicate percent cells in each area. (c) Proportion of the subsets defined n b among total γδ thymocytes from embryonic (E14–E18), newborn (days 1–9 (D0–D9)) or adult (6-week-old (W6)) C57BL/6 mice. (d) Flow cytometry of the cell size of the subsets defined in b. (e) Staining of thymocytes from newborn mice (day 2) with antibodies specific for various markers (below plots), gated on the subsets defined in b. (f) Incorporation of the thymidine analog BrdU (5-bromodeoxyuridine) by gated subsets (defined in b) of thymocytes obtained from C57BL/6 mice given BrdU daily intraperitoneally and then stained for CD27, CD25, TCRγδ and BrdU. (g) Intracellular staining of thymocyte subsets (defined in b) with 7- amino- actinomycin D (7-AAD), an indicator of DNA content. 27,25+, γδ27,25+; 27+, γδ27+; 27–, γδ27–. (h) Flow cytometry of γδ25,27+ thymocytes after sorting (Post-sort; left) and after the sorted cells were mixed with thymic stromal cell samples from E15 C57BL/6 mice, depleted of thymocytes and incubated for 7 d (RTOC; right). Numbers adjacent to or in outlined areas indicate percent cells in each. (i) Quantitative RT-PCR analysis of the expression of Ifnγ and II17 in various cell subsets, presented as described in Figure 2a. (j) Semiquantitative RT-PCR analysis of the expression of Rorc, Runx1 and Tbx21 in various cell subsets, normalized to Actb expression (encoding b-actin). For Rorc and Runx1, cells were sorted from pooled organs of over four mice; for Tbx21, γδ thymocyte subsets were purified from pooled FTOCs at day 6 (n>5 cultures). 4+8+, CD4+CD8+. Data are representative of three (a,d,e,h,j) or over ten (b) independent experiments or three (c,g,i) or two (f) independent experiments with three mice (f,g,i) or over three mice (c; error bars (c,f,g,i), s.d.).

CD27 regulates γδ T cell functional potential

The number of γδ T cells, TCR Vγ segment use and profiles of developmental markers (such as CD24, CD25, CD44 and CD62L) were similar in the thymus and peripheral lymphoid organs of wild- type and Cd27–/– mice38 (Fig. 5a, Supplementary Fig. 8a,b online and data not shown). However, TCRγδ+ thymocytes from Cd27–/– mice had lower expression of Ifnγ (Supplementary Fig. 8c), and peripheral γδ cells from Cd27–/– mice had lower expression of IFN-γ and TNF mRNA and protein (Fig. 5b and Supplementary Fig. 8d). The most overt difference was that the proportion of IFN-γhi cells in Cd27–/– mice was approximately 65% lower (Fig. 5b). Likewise, CD44loCD62Lhi γδ T cells from Cd27–/– mice (which in wild-type mice would also express CD27) had less IFN-γ production than their wild-type counterparts had (Supplementary Fig. 8e). Thus, CD27 is a regulator as well as a marker of the functional differentiation of γδ T cells. IL-17 expression was essentially unchanged in Cd27–/– mice and, as in wild-type mice, IL-17 expression was limited to the

Figure 5.

Figure 5

CD27 controls the functional potential of γδ T cells. (a) Staining of TCRγδ and CD3 surface proteins on wild-type (WT) C57BL/6 and CD27-deficient (Cd27−/−) thymocytes (left), and absolute number of TCRγδ+CD3+ thymocytes (right). Data are representative of five independent experiments (error bars, s.d.; n=3 mice). (b) Intracellular staining of various cytokines (left margin) in sorted total γδ T cells obtained from pooled spleens and lymph nodes of wild-type and CD27-deficient mice and stimulated with PMA and ionomycin. Right, percent of total γδ T cells. NS, not significant. *, P<0.05 (Student's t-test). Data are representative of four independent experiments with four to five mice each. (c,d) Absolute number of γδ T cells (c) or IFN-γ+ or IFN-γhi γδ T cells (d) among spleen cells of naive or P berghei ANKA–infected wild-type or Cd27−/− mice. D4PI, day 4 after infection. *, P<0.05 (Student's t-test). Data are representative of two independent experiments (error bars, s.d.; n=5 mice).

CD44hiCD62Llo subset in Cd27–/– mice (Supplementary Fig. 8f)

Infection with P. berghei ANKA elicited a similar expansion of total γδ splenocyte populations in wild-type and Cd27–/– mice (Fig. 5c), but the latter had 60% lower absolute numbers of IFN-γ producers and 80% fewer IFN-ghi cells than did wild-type mice (Fig. 5d). Notably, there was a delay in the progression of cerebral malaria, a severe inflammatory syndrome partly attributable to IFN-γ28, in Cd27–/– mice (Supplementary Fig. 9 online).

To investigate possible mechanisms by which CD27 alters the thymic and peripheral phenotypes of γδ T cells, we considered the selective impairment of IFN-γ production in γδ cells that occurs in TCRβ-deficient mice. Like CD27-deficient mice, TCRβ-deficient mice show no overt defect in γδ T cell numbers. In TCRβ-deficient mice, γδ T cell functional alterations have been attributed mainly to the absence in the thymus of trans-conditioning of γδ cell progenitors by TCRβ+ DP thymocytes1,17. Both γδ T subsets defined here were present in wild-type proportions in TCRβ-deficient mice (data not shown), but γδ27+ cells in TCRβ-deficient mice showed impaired expression of IFN-γ and TNF mRNA (Fig. 6a). We did not find this impairment in γδ27+ cells from TCRα-deficient mice, which had wild-type numbers of DP thymocytes; this suggests that the defective γδ T cell function in CD27-deficient mice may at least in part reflect altered trans-conditioning.

Figure 6.

Figure 6

CD27 signals regulate the differentiation of γδ thymocytes. (a) Quantitative RT-PCR analysis of the expression of Ifng and Tnfa in γδ27+ cells sorted from TCRα-deficient mice (a) and TCRβ-deficient mice (b), presented as described in Figure 2a. Data are representative of two independent experiments (error bars, s.d.; n=3 mice). (b) Gene expression and fluorescence intensity of microarray analysis of wild-type and CD27- deficient (KO) γδ T cells. Black, genes with differences in expression of over twofold (WT/KO); gray, all other genes analyzed. P<0.00003 (adjusted) for genes selected as having differences in expression. Data are representative of two independent experiments. (c) Semiquantitative RT-PCR analysis of the expression of various genes (left margin) in total γδ thymocytes from wild-type or CD27-deficient mice. Data are representative of two independent experiments with triplicate samples of cells purified from four to six mice. (d) Quantitative RT-PCR analysis of the expression of Ltbr in total γδ thymocytes sorted from wild-type and CD27-deficient mice, presented as described in Figure 2a. *, P<0.001 (Student's t-test). Data are representative of two independent experiments (n=3 mice). (e) Semiquantitative RT-PCR analysis of the expression of various genes (left margin) in γδ subsets from wild-type thymi. Data are representative of two independent experiments with triplicate samples of cells purified from four to six mice. (f) Quantitative RT-PCR analysis of the expression of Ifng and II17 in γδ T cell subsets sorted at day 7 from FTOCs treated with a soluble recombinant fusion protein of CD70 and immunoglobulin (10 mg/ml; sCD70 +) or control human immunoglobulin G (sCD70 -), presented as described in Figure 2a. Data are representative of two independent experiments (error bars, s.d.; n=3 mice). *, P<0.05 (Student's t-test). (g) Flow cytometry of FTOCs at day 7, treated with medium alone, anti-CD3ε (0.67 mg/ml), anti–keyhole limpet hemocyanin (0.67 mg/ml; isotype control) or cyclosporin A (0.5 mM). Data are representative of two independent experiments with a pool of three E14 thymic lobes each.

To test that possibility, we compared the transcriptomes of wild-type and CD27-deficient γδ T cells with whole-genome microarrays (ArrayExpress accession number E-NCMF-23; Fig. 6b). Conspicuous among CD27-dependent genes was Ltbr, which encodes LTβR, a mediator of trans-conditioning17, as well as four ‘signature’ genes of LTβR signaling in γδ thymocytes: Crem (also called Icer), Nr4a2 (Nurr1), Rgs2 and Rgs1 (Table 1). We confirmed differences in the expression of these genes in wild-type and CD27-deficient γδ thymocytes by RT-PCR; in contrast, II7ra, which encodes a factor critical for γδ thymocyte development1, was not impaired in CD27-deficient γδ thymocytes (Fig. 6c,d).

Table 1. Transcription in wild-type C57BL/6 and CD27-deficient γδ T lymphocytes.

Expression of genes linked to T cell biology, presented as expression in wild-type cells relative to expression in CD27-deficient γδ T cells (‘fold’ change). Gene ID, identification number in the National Center for Biotechnology Information database. Full cDNA microarray data, ArrayExpress accession number E-NCMF-23. P=0.00003 (threshold for microarray data analysis). Data are representative of two independent experiments.

Gene ID Symbol Description Function Expression
21940 Tnfrsf7 CD27 (TNF-RSF7) Activation 21.11
17000 Ltbr Lymphotoxin-β receptor (TNF-RSF3) Cytokine receptor 9.00
12477 Ctla4 CTLA-4 coreceptor Activation 7.01
12916 Crem CREM/ICER Transcription 5.10
18227 Nr4a2 Nuclear orphan receptor Transcription 4.66
19735 Rgs2 Regulator of G protein signaling Signaling 4.17
50778 Rgs1 Regulator of G protein signaling Signaling 3.86
21943 Tnfsf11 RANK ligand (TNF-SF11) Cytokine 3.66
12487 Cd28 CD28 coreceptor Activation 3.36
19734 Rgs16 Regulator of G protein signaling Signaling 3.25
16364 Irf4 Interferon-regulatory factor Transcription 2.30
16478 Jund1 JunD Transcription 2.20
16172 Il17ra IL-17 receptor α-chain Cytokine receptor 0.14

Consistent with the effects of CD27, wild-type mice showed expression of Ltbr and Ltbr-dependent genes in γδ27,25+ and γδ27+ thymocytes but not in γδ27– thymocytes (Fig. 6e). To derive further evidence that CD27 regulates the differentiation of γδ thymo- cytes, we added to E14 fetal thymic organ cultures (FTOCs) a fusion protein of CD70 and immunoglobulin that acts as a CD27 agonist39. After 1 week of culture, the recovered γδ27+ thymocytes showed significant upregulation of Ifng and γδ27– thymocytes showed down-regulation of II17 (Fig. 6f). Thus, embryonic γδ progenitors were effectively skewed toward IFN-γ production simply through the manipulation of CD70-CD27 signals. To assess whether CD27 might be acting together with other determinants of γδ differentiation, we added to parallel organ cultures either anti-CD3e or cyclosporin A; the former enhances and the latter inhibits TCR signaling. Relative to their representation in control cultures, these treatments increased and decreased representation of the γδ27+ subset, respectively, with reciprocal effects on the γδ27– subset (Fig. 6g). We conclude that the developmental preconditioning of discrete peripheral γδ cell subsets may involve some form of ‘collaboration’ between TCR and CD27 signaling in the thymus.

Finally, to determine whether the classification that we have introduced might apply to other unconventional T cells, we examined invariant NKT cells, as it has been reported that IL-17 expression is restricted to the NK1.1 subset of NKT cells and that this restriction is ‘imprinted’ in the thymus40,41. After isolating NKT cells on the basis of their reactivity to tetramers of CD1d and α-galactosylceramide, we found that surface marker staining ‘assigned’ over 80% of NK1.1 cells to the CD27 compartment, whereas over 80% of CD27+ cells were NK1.1+ (Supplementary Fig. 10 online). Thus, it seems that in the NKT compartment, IL-17-producing cells are also found mainly in the CD27 subset.

DISCUSSION

Here we have described mouse γδ cell subsets that had distinct functional capacities and could be defined by CD27 expression. The γδ27+ subset was more abundant and included cells that produced IFN-γ but essentially no IL-17 immediately after isolation and in all activation conditions used. In contrast, the γδ27– subset comprised essentially all IL-17-producing γδ cells. Both subsets actively responded to infection in vivo by proliferating and producing cytokines. Although γδ27– cells may have produced IFN-γ after in vitro activation, very few did so ex vivo, whether from naive or infected mice. Moreover, their thymic counterparts did not produce IFN-γ, which raises the possibility that the IFN-γ and IL-17 ‘double-producing’ cells were an artifact of in vitro activation protocols, even though ‘double production’ of IL-17 and IFN-γ has been described for TCRαβ+ TH-17 cells6,42. Whereas TCRαβ+ TH1 and TH-17 cells can take up to 7 d to differentiate and expand clonally under the influence of specific combinations of cytokines43, the ability of the γδ T cell subsets described here to express their ‘signature’ cytokines directly ex vivo was apparently acquired during their differentiation in the thymus, when the subsets also adopted their characteristic phenotypic markers and other properties that remained essentially stable, even during infection. These observations define a parallel with NKT cells, another ‘innate-like’ lymphocyte lineage that promptly secretes a panoply of cytokines, including IL-17 and IFN-γ40,44. Having found that NK1.1 NKT cell populations, which are known to include ‘spontaneous’ IL-17-producing cells40,41, were mostly CD27, we propose that CD27 may influence the functional differentiation of several lineages of unconventional T cells. The thymic ‘imprinting’ of the CD27+ and CD27– subsets perhaps reflects the fact that γδ T cells (and other unconventional T cells) may need to provide specific effector cytokines very rapidly, to counter microbial growth and dissemination before maturation of the adaptive response. Indeed, myriad analyses have shown that γδ cells are crucial providers of either IFN-γ or IL-17 in diverse physiological settings810,14,2022. This may be particularly true in early life, when there is a precipitous encounter with environmental antigens and when γδ T cells seem precociously mature compared with αβ T cells in both mice35 and humans (D. Gibbons and A.C.H., unpublished observations). Rapid γδ T cell responses may also determine in part the complexion and magnitude of ensuing adaptive responses14,45. Developmental ‘imprinting’ may more strongly influence γδ T cells than αβ T cells, as defined sets of cytokines seem able to skew bulk populations of peripheral αβ T cells toward widely different functional differentiation pathways (such as TH1, TH-17 and regulatory T cells). In contrast, the various conditions we used here failed to skew peripheral γδ27+ cells toward IL-17 production. Such functional ‘rigidity’ and our notable failure to identify any substantial interconversion of γδ27+ and γδ27– cells in vitro or in vivo suggest that these two subsets, although heterogeneous, constitute discrete lineages. Because it regulates γδ T cell functional differentiation, we believe the TNF receptor family member CD27 is a more incisive marker of γδ T cell subsets than others described14,23,26. CD27 controls the expression of LTβR, another TNF receptor family member and a component of γδ T cell trans-conditioning that is associated with IFN- γ-biased differentiation and high in vitro proliferative potential17. Thus, CD27 may induce or sustain expression of LTβR and its ‘downstream’ targets in γδ27+ thymocytes and in their γδ27,25+ progenitors. This would be consistent with the finding that genes induced by trans-conditioning were expressed in early CD4CD8 progenitors and their expression was then selectively retained or lost in more mature cells. The retention of the expression of such genes in γδ27+ cells would confer their ‘signatory’ proliferative, TH1-like func- tional potential, which is impaired in CD27-deficient mice and LTβR- deficient mice17. Because the lymphotoxin that trans-conditions through LTβR is derived at least in part from TCRβ+ DP thymocytes, this model also readily explains the functional impairment of the γδ27+ subset in TCRβ-deficient mice but not in TCRα-deficient mice. It might also explain the notable increase in the size of the thymic γδ27+ subset between E15 and E18, as this period corresponds to the main expansion of TCRβ+ DP thymocyte populations.

Clearly, as the impairment in IFN-γ production by γδ T cells in mice lacking CD27, LTβR or TCRβ is incomplete, other factors probably influence γδ T cell function. Signaling by the γδ TCR, which has been reported to skew γδ T cells toward IFN-γ production and away from IL-17 production14 may be one such factor. Our findings of TCR bias in the γδ27+ and γδ27– subsets and of the influence of TCR signal manipulation on the relative abundance of γδ27+ and γδ27– subsets in FTOC are consistent with that idea. TCR signals in the thymus may upregulate CD27 (ref. 37), which may then act together with TCR signaling (and/or other differentiation factors) by providing a critical competence to γδ T cells (and possibly other unconventional T cells) so they can progress through TH1 differentiation. Although the transcription factor RORγt characterizes all IL-17-producing γδ27– cells, its method of operating would seem different from that in TCRαβ+ TH-17 cells as, consistent with other data7, we found that IL-17 production by γδ T cells required neither ‘collaborative’ signaling from TGF-β, IL-6 and IL-21 receptors nor interference with IFN production. It is likewise notable that microbe-induced IL-17 production in the gut seems to derive from αβ T cells46, whereas IL-17-producing γδ T cells are relatively rare in the gut compared with their presence in other tissues. Indeed, the relative over-representation of TCRαβ+ TH-17 cells in the mouse gut6 may represent a ‘compensation’ for the relative scarcity of gut γδ27– cells. Another contrast in the IL-17 production of αβ and γδ T cells lies in the augmented γδ27– cell production of IL-17 in response to activation by TGF-β and TCR in the presence of IL-2, a combination that upregulates the transcription factor Foxp3 and a bias toward regulatory T cell function in CD4+ αβ T cells7,47. Indeed, it was reproducibly difficult to detect Foxp3 expression in γδ T cells in wild-type mice18 (data not shown). Possibly, a key feature of IL-17 production by γδ cells may be the ability of RORγt to act without interference from Foxp3. Although γδ27– cells were mostly unaffected in CD27-deficient mice, as might be expected, CD27 gain of function in FTOC did result in lower IL-17 production by these cells, which indicates that CD27 may diminish IL-17 production as well as enhance IFN-g production, at least in T cell progenitors. CD27 is expressed on a plethora of lymphoid cells and bone marrow precursors, where it has been assigned many pleiotropic functions38,39,48,49. Despite its expression on mouse and human thymocytes and its responsiveness to pre-TCR and TCR signals37, CD27 seems to be not critical in the development of conventional αβ T cells38 (unpublished data). That is notable given the constitutive expression of CD70 on thymic epithelial cells of mice and humans50. Thus, here we have reported a previously unknown function for CD27 expression on thymic T cell precursors. Future studies should deter- mine which cells direct key CD70-CD27 interactions in the thymus and whether the functional and phenotypic stability of γδ27+ cells requires sustained interactions between CD27 and CD70 in the periphery. The resolution of such issues will provide a better under- standing of the balance of precommitment and the peripheral plasticity of γδ T cell subsets.

METHODS

Mice

All mice used were adults 4–10 weeks of age. Embryos were obtained by the setting up of timed pregnancies. C57BL/6 (B6), B6.TCRα-deficient, B6.TCRβ-deficient and B6.RAG-2-deficient mice were from Jackson Laboratories. B6.CD27-deficient mice have been described38. B6.RORγt-GFP mice35 were a gift from D. Littman. Mice were bred and maintained in the specific pathogen–free animal facilities of the Instituto de Medicina Molecular (Lisbon), Queen Mary London University (London) and Yale University School of Medicine (New Haven). All experiments involving animals were done in compliance with the relevant laws and institutional guidelines and were approved by local ethics committees.

Cell preparation

Thymuses, lymph nodes and spleens were homogenized and washed in RPMI medium containing 10% (vol/vol) FCS. In some cases, thymocyte samples were depleted of CD8+ and CD4+ cells by treatment with anti-CD8 (31.M) and anti-CD4 (RL172; both from J. van Meerwijk) plus complement (HD Supplies), followed by gradient centrifugation in Lympholyte M (Cederlane Laboratories). Lungs were cut into pieces 5 mm2 and were incubated for 2 h at 37 1C with stirring in complete RPMI medium supplemented with collagenase (0.05 mg/ml; Sigma). Supernatants were collected and live cells were isolated on a gradient of Lympholyte M (Cederlane Laboratories).

Monoclonal antibodies

Fluorescein isothiocyanate–labeled anti-Vγ1 (2.11) was a gift from P. Pereira. The following monoclonal antibodies were from BD Pharmingen: fluorescein isothiocyanate–labeled anti-TCRδ (GL3), anti-Vγ4 (UC3-10A6) and anti-CD62L (MEL14); phycoerythrin-labeled anti-TCRδ (GL3), anti-CD25 (PC61), anti-CD44 (IM7), anti-IFN-γ (XMG1.2), and anti-TNF (MP6-XT22); peridinine chlorophyll protein complex–cyanine 5.5–labeled anti-CD4 (RM4-5) and anti-CD8α (53.6.7); allophycocyanin- labeled anti-CD25 (PC61); biotinylated anti-TCRδ (GL3); peridinine chloro- phyll protein complex–cyanine 5.5–conjugated streptavidin; and purified anti-CD3e (145.2C11). The following monoclonal antibodies were from eBiosciences: phycoerythrin-labeled anti-CD27 (LG.7F9), phycoerythrin- indotricarbocyanine– labeled anti-CD3e (145.2C11), fluorescein isothiocya- nate– or allophycocyanin-labeled anti-IL-17 (eBio17B7), and allophycocya- nin-labeled anti-CD27 (LG.7F9). Phycoerythrin-labeled mCD1d tetramer loaded with the glycolipid PBS57 was from the National Institutes of Health Tetramer Core Facility.

Flow cytometry and cell sorting. Information on these procedures is available in the Supplementary Methods online

Cell culture

For preactivation regimens, cells were incubated for 4 d on plate-bound anti-CD3e (10 mg/ml) with IL-2 (100 U/ml; Sigma), with or without TGF-β (5 ng/ml; R&D Systems). Where indicated, IL-6 (20 ng/ml), IL-21 (100 ng/ml), IL-23 (20 ng/ml) or neutralizing anti-IFN−γ (10 mg/ml) was also added to the medium. For analysis of proliferation, flow cytometry–sorted cells were stained by incubation for 12 min at 37 1C with 5 mM CFSE (carboxy- fluorescein diacetate succinimidyl ester; Molecular Probes). Reactions were quenched by washing with ice-cold RPMI medium supplemented with 10% (vol/vol) FCS. CFSE-labeled responder cells (5 ×104) were cultured in 96-well round-bottomed plates in the presence of antigen-presenting cells (1 ×105 erythrocyte-depleted splenocyte samples treated with mitomycin C (Sigma)) and various doses of anti-CD3e. After 3 d, the proliferation of responder cells was assessed by flow cytometry.

FTOC

The medium for FTOC was RPMI-1640 medium with 10% (vol/vol) FCS, 50 mM β-mercaptoethanol, L-glutamine, nonessential amino acids, 10 mM HEPES, pH 7.2, penicillin and streptomycin. C57BL/6 fetal thymic lobes at E15 were cultured for various times in FTOC medium on nucleopore filters (Whatman) and then were analyzed by flow cytometry. Cultures were provided fresh medium every 3 d. For reaggregate thymic organ culture, E15 thymic lobes treated with 1.35 mM 2-deoxyguanosine (Sigma) were digested for 30 min at 37 1C to a single-cell suspension with 0.125% (vol/vol) trypsin (Invitrogen). Reaggregates were formed by the centrifugation of 7 ×105 stromal cells with 1×103 to 10 ×103 sorted γδ25,27+ cells and pipetting of the pellet onto a nucleopore filter in FTOC medium. Cultures were incubated 7–10 d. For CD27 stimulation, a fusion protein of recombinant CD70 and immunoglobulin (10 mg/ml; provided by A. Al-Shamkhani) was added to the cultures.

Adoptive transfer

Sorted cells were stained with CFSE as described above and were injected intravenously into RAG-2-deficient mice (3 ×105 to 5 ×105 cells per mouse). Splenocytes from injected mice were collected after 3 d (for analysis of short-term division and stability of CD27 expression) or 3 weeks (for analysis of long-term stability of CD27 expression).

Malaria infection. Mice were infected as described31

RT-PCR. RNA was extracted with TRIzol reagent (Invitrogen) and was treated with RNase-free RQ1 DNase (Promega). RNA concentrations were measured with a Nanodrop ND 1000 (Nucliber). Superscript II reverse transcriptase was used according to the manufacturer's protocol (Invitrogen) for the production of cDNA in a reaction volume of 20 ml. Primers for quantitative real-time RT- PCR or semiquantitative RT-PCR (Supplementary Table 1 online) were designed with the Primer3 program or with the Universal ProbeLibrary Assay Design Center (Roche Applied Science). For semiquantitative RT-PCR, cDNA was amplified as described18 in a MyCycler thermocycler (BioRad). For quantitative PCR, cDNA was amplified in a reaction volume of 50 ml, with half the volume (25 ml) being SYBR Green PCR master mix (Applied Biosystems), in a Rotor Gene 6000 (Corbett Research). Reactions consisted of 2 min at 50 1C and 10 min at 95 1C, followed by 50 cycles of 15 s at 95 1C and 1 min at 60 1C, followed by 50 s at 95 1C. Expression was normalized to that of Eef1a1 (encoding eukaryotic translation elongation factor 1a1; called ‘Efa1’ here) or Hprt1 (encoding hypoxanthine guanine phosphoribosyl trans- ferase) and is presented as percent of maximum expression.

Supplementary Material

01

ACKNOWLEDGMENTS

We thank L. Gracç a (Instituto de Medicina Molecular), G. Anderson (Institute for Biomedical Research, Medical Research Council), B. Rocha (Hô pital Necker), J. Demengeot (Instituto Gulbenkian de Ciência), M.M. Mota (Instituto de Medicina Molecular) and B. Stockinger (Institute for Biomedical Research, Medical Research Council) for materials and suggestions; D. Littman (New York University) for B6.RORgt-GFP mice; P. Pereira (Institut Pasteur) for fluorescein isothiocyanate–labeled anti-Vg1; J. van Meerwijk (Institut National pour la Sante̛ et la Recherche Me̛ dicale, Toulouse) for anti-CD8 and anti-CD4; A. Al-Shamkhani (University of Southampton School of Medicine) for the fusion protein of CD70 and immunoglobulin; D. Bruno, A. Pamplona, A. Pena, N.G. Sousa, D.V. Correia, M. Ferreira, A.Q. Gomes, J. Coquet, T. Silberzahn, S. Zelenay, M.L. Bergman and M. Monteiro for experimental assistance; A.L. Caetano, P. Hutchinson, M. Soares, R. Gardner, W. Turnbull andG. Warnes for cell sorting; the microarray facility at the Nederlands Kanker Instituut for array development; M. Rebelo and A. Costa for the maintenance of mouse strains; and J. van Meerwijk and P. Romagnoli for critical reading of the manuscript. Supported by the European Molecular Biology Organization (YIP 1440 to B.S.-S.), The Research Advisory Board of St. Bartholomew's and The Royal London Charity (RAB 06/PJ/08 to D.J.Pa. and D.J.Pe.), the Wellcome Trust (A.C.H.), and the Portuguese Ministry of Science (J.C.R., A.d.B. and J.F.N.; and PTDC/BIA-BCM/71663 to B.S.-S.).

Footnotes

AUTHOR CONTRIBUTIONS

Experiments were done by J.C.R. (Figs. 16), A.d.B. (Figs. 2,4,6), D.J.Pa., J.F.N. and D.J.Pe. (Figs. 4,6), V.P. (Fig. 6b and Table 1), S.J.R. and M.G. (Fig. 3c) and B.S.-S. (Fig. 6c,e); J.B. contributed to designing the research and writing the paper; A.C.H. contributed to designing the research and wrote the paper; D.J.Pe. designed the research (Figs. 36) and wrote the paper; and B.S.-S. designed the research (Figs. 16) and wrote the paper.

Microarray. Information on this procedure is available in the Supplementary Methods

Statistical analysis. The statistical significance of differences between popula- tions was assessed with the Student's t-test; P values of less 0.05 than were considered significant.

Accession codes. UCSD-Nature Signaling Gateway (http://www.signaling-gateway.org): A000546 and A000547Silva; ArrayExpress: microarray data, E-NCMF-23.

References

  • 1.Hayday AC, Pennington DJ. Key factors in the organized chaos of early T cell development. Nat. Immunol. 2007;8:137–144. doi: 10.1038/ni1436. [DOI] [PubMed] [Google Scholar]
  • 2.Bhandoola A, von Boehmer H, Petrie HT, Zuniga-Pflucker JC. Commitment and developmental potential of extrathymic and intrathymic T cell precursors: plenty to choose from. Immunity. 2007;26:678–689. doi: 10.1016/j.immuni.2007.05.009. [DOI] [PubMed] [Google Scholar]
  • 3.Laky K, Fleischacker C, Fowlkes BJ. TCR and Notch signaling in CD4 and CD8 T-cell development. Immunol. Rev. 2006;209:274–283. doi: 10.1111/j.0105-2896.2006.00358.x. [DOI] [PubMed] [Google Scholar]
  • 4.Roark CL, Simonian PL, Fontenot AP, Born WK, O'Brien RL. gd T cells: an important source of IL-17. Curr. Opin. Immunol. 2008;20:353–357. doi: 10.1016/j.coi.2008.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Vermijlen D, et al. Distinct cytokine-driven responses of activated blood γδ T cells: insights into unconventional T cell pleiotropy. J. Immunol. 2007;178:4304–4314. doi: 10.4049/jimmunol.178.7.4304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Ivanov II, et al. The orphan nuclear receptor RORγt directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell. 2006;126:1121–1133. doi: 10.1016/j.cell.2006.07.035. [DOI] [PubMed] [Google Scholar]
  • 7.Lochner M, et al. In vivo equilibrium of proinflammatory IL-17+ and regulatory IL-10+ Foxp3+ RORγt+ T cells. J. Exp. Med. 2008;205:1381–1393. doi: 10.1084/jem.20080034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Lockhart E, Green AM, Flynn JL. IL-17 production is dominated by gd T cells rather than CD4 T cells during Mycobacterium tuberculosis infection. J. Immunol. 2006;177:4662–4669. doi: 10.4049/jimmunol.177.7.4662. [DOI] [PubMed] [Google Scholar]
  • 9.Shibata K, Yamada H, Hara H, Kishihara K, Yoshikai Y. Resident Vd1+ γδ T cells control early infiltration of neutrophils after Escherichia coli infection via IL-17 production. J. Immunol. 2007;178:4466–4472. doi: 10.4049/jimmunol.178.7.4466. [DOI] [PubMed] [Google Scholar]
  • 10.Umemura M, et al. IL-17-mediated regulation of innate and acquired immune response against pulmonary Mycobacterium bovis bacille Calmette-Guerin infection. J. Immunol. 2007;178:3786–3796. doi: 10.4049/jimmunol.178.6.3786. [DOI] [PubMed] [Google Scholar]
  • 11.Zhang F, Meng G, Strober W. Interactions among the transcription factors Runx1, RORgt and Foxp3 regulate the differentiation of interleukin 17-producing T cells. Nat. Immunol. 2008;9:1297–1306. doi: 10.1038/ni.1663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Yang Y, Xu J, Niu Y, Bromberg JS, Ding Y. T-bet and Eomesodermin play critical roles in directing T cell differentiation to Th1 versus Th17. J. Immunol. 2008;181:8700–8710. doi: 10.4049/jimmunol.181.12.8700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Amsen D, et al. Instruction of distinct CD4 T helper cell fates by different notch ligands on antigen-presenting cells. Cell. 2004;117:515–526. doi: 10.1016/s0092-8674(04)00451-9. [DOI] [PubMed] [Google Scholar]
  • 14.Jensen KD, et al. Thymic selection determines γδ T cell effector fate: antigen-naïve cells make interleukin-17 and antigen-experienced cells make interferon γ. Immunity. 2008;29:90–100. doi: 10.1016/j.immuni.2008.04.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Born WK, Reardon CL, O'Brien RL. The function of γδ T cells in innate immunity. Curr. Opin. Immunol. 2006;18:31–38. doi: 10.1016/j.coi.2005.11.007. [DOI] [PubMed] [Google Scholar]
  • 16.Pennington DJ, et al. The inter-relatedness and interdependence of mouse T cell receptor γδ+ and ab+ cells. Nat. Immunol. 2003;4:991–998. doi: 10.1038/ni979. [DOI] [PubMed] [Google Scholar]
  • 17.Silva-Santos B, Pennington DJ, Hayday AC. Lymphotoxin-mediated regulation of γδ cell differentiation by αβ T cell progenitors. Science. 2005;307:925–928. doi: 10.1126/science.1103978. [DOI] [PubMed] [Google Scholar]
  • 18.Pennington DJ, et al. Early events in the thymus affect the balance of effector and regulatory T cells. Nature. 2006;444:1073–1077. doi: 10.1038/nature06051. [DOI] [PubMed] [Google Scholar]
  • 19.Borst J, Hendriks J, Xiao Y. CD27 and CD70 in T cell and B cell activation. Curr. Opin. Immunol. 2005;17:275–281. doi: 10.1016/j.coi.2005.04.004. [DOI] [PubMed] [Google Scholar]
  • 20.Gao Y, et al. γδ T cells provide an early source of interferon g in tumor immunity. J. Exp. Med. 2003;198:433–442. doi: 10.1084/jem.20030584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Roark CL, et al. Exacerbation of collagen-induced arthritis by oligoclonal, IL-17-producing γδ T cells. J. Immunol. 2007;179:5576–5583. doi: 10.4049/jimmunol.179.8.5576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Romani L, et al. Defective tryptophan catabolism underlies inflammation in mouse chronic granulomatous disease. Nature. 2008;451:211–215. doi: 10.1038/nature06471. [DOI] [PubMed] [Google Scholar]
  • 23.Shibata K, et al. Identification of CD25+ γδ T cells as fetal thymus-derived naturally occurring IL-17 producers. J. Immunol. 2008;181:5940–5947. doi: 10.4049/jimmunol.181.9.5940. [DOI] [PubMed] [Google Scholar]
  • 24.Ruprecht CR, et al. Coexpression of CD25 and CD27 identifies FoxP3+ regulatory T cells in inflamed synovia. J. Exp. Med. 2005;201:1793–1803. doi: 10.1084/jem.20050085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Vossen MT, et al. CD27 defines phenotypically and functionally different human NK cell subsets. J. Immunol. 2008;180:3739–3745. doi: 10.4049/jimmunol.180.6.3739. [DOI] [PubMed] [Google Scholar]
  • 26.Kisielow J, Kopf M, Karjalainen K. SCART scavenger receptors identify a novel subset of adult γδ T cells. J. Immunol. 2008;181:1710–1716. doi: 10.4049/jimmunol.181.3.1710. [DOI] [PubMed] [Google Scholar]
  • 27.Ho M, Webster HK, Tongtawe P, Pattanapanyasat K, Weidanz WP. Increased γδ T cells in acute Plasmodium falciparum malaria. Immunol. Lett. 1990;25:139–141. doi: 10.1016/0165-2478(90)90105-y. [DOI] [PubMed] [Google Scholar]
  • 28.Yanez DM, Batchelder J, van der Heyde HC, Manning DD, Weidanz WP. γδ T-cell function in pathogenesis of cerebral malaria in mice infected with Plasmodium berghei ANKA. Infect. Immun. 1999;67:446–448. doi: 10.1128/iai.67.1.446-448.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Ramsburg E, Tigelaar R, Craft J, Hayday A. Age-dependent requirement for γδ T cells in the primary but not secondary protective immune response against an intestinal parasite. J. Exp. Med. 2003;198:1403–1414. doi: 10.1084/jem.20030050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.D'Ombrain MC, Hansen DS, Simpson KM, Schofield L. γδ-T cells expressing NK receptors predominate over NK cells and conventional T cells in the innate IFN-γ response to Plasmodium falciparum malaria. Eur. J. Immunol. 2007;37:1864–1873. doi: 10.1002/eji.200636889. [DOI] [PubMed] [Google Scholar]
  • 31.Pamplona A, et al. Heme oxygenase-1 and carbon monoxide suppress the pathogenesis of experimental cerebral malaria. Nat. Med. 2007;13:703–710. doi: 10.1038/nm1586. [DOI] [PubMed] [Google Scholar]
  • 32.Bettelli E, et al. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature. 2006;441:235–238. doi: 10.1038/nature04753. [DOI] [PubMed] [Google Scholar]
  • 33.Veldhoen M, Hocking RJ, Atkins CJ, Locksley RM, Stockinger B. TGFβ in the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17-producing T cells. Immunity. 2006;24:179–189. doi: 10.1016/j.immuni.2006.01.001. [DOI] [PubMed] [Google Scholar]
  • 34.Mangan PR, et al. Transforming growth factor-b induces development of the TH17 lineage. Nature. 2006;441:231–234. doi: 10.1038/nature04754. [DOI] [PubMed] [Google Scholar]
  • 35.Eberl G, Littman DR. Thymic origin of intestinal αβ T cells revealed by fate mapping of RORgt+ cells. Science. 2004;305:248–251. doi: 10.1126/science.1096472. [DOI] [PubMed] [Google Scholar]
  • 36.Ciofani M, Knowles GC, Wiest DL, von Boehmer H, Zuniga-Pflucker JC. Stage-specific and differential notch dependency at the αβ and γδ T lineage bifurcation. Immunity. 2006;25:105–116. doi: 10.1016/j.immuni.2006.05.010. [DOI] [PubMed] [Google Scholar]
  • 37.Taghon T, Yui MA, Pant R, Diamond RA, Rothenberg EV. Developmental and molecular characterization of emerging b- and γδ-selected pre-T cells in the adult mouse thymus. Immunity. 2006;24:53–64. doi: 10.1016/j.immuni.2005.11.012. [DOI] [PubMed] [Google Scholar]
  • 38.Hendriks J, et al. CD27 is required for generation and long-term maintenance of T cell immunity. Nat. Immunol. 2000;1:433–440. doi: 10.1038/80877. [DOI] [PubMed] [Google Scholar]
  • 39.Carr JM, et al. CD27 mediates interleukin-2-independent clonal expansion of the CD8+ T cell without effector differentiation. Proc. Natl. Acad. Sci. USA. 2006;103:19454–19459. doi: 10.1073/pnas.0609706104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Michel ML, et al. Identification of an IL-17-producing NK1.1neg iNKT cell population involved in airway neutrophilia. J. Exp. Med. 2007;204:995–1001. doi: 10.1084/jem.20061551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Michel ML, et al. Critical role of ROR-gt in a new thymic pathway leading to IL-17-producing invariant NKT cell differentiation. Proc. Natl. Acad. Sci. USA. 2008;105:19845–19850. doi: 10.1073/pnas.0806472105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Lee YK, et al. Late developmental plasticity in the T helper 17 lineage. Immunity. 2009;30:92–107. doi: 10.1016/j.immuni.2008.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Weaver CT, Harrington LE, Mangan PR, Gavrieli M, Murphy KM. Th17: an effector CD4 T cell lineage with regulatory T cell ties. Immunity. 2006;24:677–688. doi: 10.1016/j.immuni.2006.06.002. [DOI] [PubMed] [Google Scholar]
  • 44.Bendelac A, Savage PB, Teyton L. The biology of NKT cells. Annu. Rev. Immunol. 2007;25:297–336. doi: 10.1146/annurev.immunol.25.022106.141711. [DOI] [PubMed] [Google Scholar]
  • 45.Zuany-Amorim C, et al. Requirement for γδ T cells in allergic airway inflammation. Science. 1998;280:1265–1267. doi: 10.1126/science.280.5367.1265. [DOI] [PubMed] [Google Scholar]
  • 46.Ivanov II, et al. Specific microbiota direct the differentiation of IL-17-producing T-helper cells in the mucosa of the small intestine. Cell Host Microbe. 2008;4:337–349. doi: 10.1016/j.chom.2008.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Chen W, et al. Conversion of peripheral CD4+CD25– naive T cells to CD4+CD25+ regulatory T cells by TGF-β induction of transcription factor Foxp3. J. Exp. Med. 2003;198:1875–1886. doi: 10.1084/jem.20030152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Hendriks J, Xiao Y, Borst J. CD27 promotes survival of activated T cells and complements CD28 in generation and establishment of the effector T cell pool. J. Exp. Med. 2003;198:1369–1380. doi: 10.1084/jem.20030916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Nolte MA, et al. Immune activation modulates hematopoiesis through interactions between CD27 and CD70. Nat. Immunol. 2005;6:412–418. doi: 10.1038/ni1174. [DOI] [PubMed] [Google Scholar]
  • 50.Tesselaar K, et al. Expression of the murine CD27 ligand CD70 in vitro and in vivo. J. Immunol. 2003;170:33–40. doi: 10.4049/jimmunol.170.1.33. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

01

RESOURCES