Ly6C and Sca-1 (Ly6A/E) are Ly6 family GPI-anchored surface molecules that are differentially expressed by multiple immune populations. Ly6C expression has been used to distinguish short-lived effector CD4+ T cells from memory precursor effector cells, whereas Sca-1 has been used in the identification of CD8+ memory stem cells. This study examines the expression patterns of these molecules and establishes that, in vitro, IL-27, type I IFN, and IFN-γ are potent inducers of Ly6C and Sca-1 in naive mouse CD4+ and CD8+ T cells, whereas TGF-β limits their expression. The induction of Ly6C and Sca-1 by IL-27 and IFN-γ is dependent on STAT1, but not STAT3 or T-bet. In mouse splenocytes, at homeostasis, Ly6C and Sca-1 expression was not restricted to effector cells, but was also found at various levels on naive and memory populations. However, in response to infection with Toxoplasma gondii, pathogen-specific T cells expressed high levels of these molecules and in this context, endogenous IL-27 and IFN-γ were required for the expression of Ly6C but not Sca-1. Together, these findings highlight the TCR-dependent and cytokine-mediated signals that modulate T cell expression of Ly6C and Sca-1 in vitro and in vivo during infection.

The T cell response to infection or immunization involves the generation of minimally differentiated memory cells as well as highly differentiated effector cells (1). Effector T cells produce the cytokines, granzymes, and other molecules necessary for immediate pathogen control, whereas central memory T cells are long-lived and can differentiate into effector cells upon rechallenge (13). The identification of surface molecules that distinguish effector and memory T cell populations has allowed researchers to track the expansion, evolution, and contraction of the T cell response during infection and has provided insights into how these cells operate. For example, central memory cells upregulate CD44 and express CD62L, which keeps them localized to lymphoid organs. Effector cells are also CD44hi but lose expression of CD62L, allowing them to home to sites of inflammation. Although these definitions have proven useful to define naive, effector, and memory T cells, additional markers, including KLRG1, CXCR3, and Ly6C have been used to further subdivide these populations (48). In current models, a subset of highly differentiated, short-lived effector cells, which for CD8+ T cells are often identified by expression of KLRG1 (8), are specialized to control acute infection. These cells produce high levels of cytokines and granzymes and survive poorly upon adoptive transfer to naive hosts. Less-differentiated CD8+ T cells, identified during acute toxoplasmosis as CXCR3+KLRG1, exhibit the longevity, proliferative capacity, and differentiation potential typical of memory cells (4).

Ly6C and Sca-1 (Ly6A/E) are members of a family of 21 Ly6-like proteins in mice, with 20 Ly6 family homologs in humans (9). Expression of Ly6C has been used to identify highly differentiated effector CD4+ T cells (6, 10, 11). Among virus-specific effector CD4+ T cells, Ly6C+ cells produced more cytokines and effector molecules than Ly6C cells (6, 10). Conversely, Sca-1, in combination with the IL-2R β-chain (CD122) and Bcl-2, has been used to identify mature CD8+ T cells with stem-like properties, termed memory stem cells (1214).

The finding that Ly6C is preferentially expressed by CD4+ short-lived effector cells during acute infection suggests that identification of the factors that modulate Ly6C expression could yield insights into the signals that control the development of memory and effector populations. For example, sustained TCR signals have been found to skew T cells toward terminal differentiation and away from memory development (1, 15), whereas multiple cytokines influence the development of effector and memory populations (16). Relevant to this report, type I IFNs, IFN-γ, and IL-27 have previously been implicated in promoting Ly6C+ and Sca-1+ populations in CD4+, CD8+, and regulatory T cells (1721). However, previous studies have not been able to distinguish whether these stimuli induce Ly6C and Sca-1 expression or simply promote the outgrowth of Ly6C+ or Sca-1+ populations. Furthermore, extant reports have not examined how these cytokines intersect with TCR signaling to impact Ly6C and Sca-1 expression and what roles these signals play in modulating the expression of these molecules in vivo. In utilizing sorted Ly6CSca-1 populations, the current study was able to show that TCR stimulation alone induces Sca-1 on CD4+ and CD8+ T cells, but is not sufficient to induce Ly6C on CD4+ T cells. The cytokines IL-27, IFN-γ, and type I IFN were found to broadly promote the expression of Ly6C and Sca-1, whereas TGF-β inhibited this expression. This cytokine-mediated induction of Ly6C and Sca-1 was largely STAT1-dependent and is not dependent on STAT3 or T-bet. A survey of naive and Ag-experienced T cells in a range of differentiation states found that Ly6C expression is not restricted to effector cells and that Sca-1 expression is not limited to naive/memory-like cells. Furthermore, in vivo studies during infection with Toxoplasma gondii identified a role for endogenous IL-27 and IFN-γ in promoting effector cell expression of Ly6C. Together, these studies provide new insights into the signals that modulate the development of Ly6C+ and Sca-1+ T cell populations but indicate caution in the use of these molecules as markers of highly differentiated effector cells (Ly6C) and minimally differentiated memory stem cells (Sca-1).

Six-week-old female C57BL/6 controls were purchased from Taconic. Mice deficient in Il27ra/WSX-1 (C57BL/6 background) were generated as described (22) and were originally provided by Amgen (Thousand Oaks, CA). STAT1−/− mice (129S6/SvEv-Stat1tm1Rds) and 129S6 control mice were purchased from Taconic. Spleens from CD4-Cre × STAT3−/− mice were provided by A. Villarino and J. J. O’Shea (National Institutes of Health). Mice were housed and bred in specific pathogen-free (SPF) facilities in the Department of Pathobiology at the University of Pennsylvania in accordance with institutional guidelines. The Me49 strain of T. gondii was prepared from chronically infected CBA/ca mice and experimental animals were infected i.p. with 20 cysts.

Splenocytes from C57BL/6 mice were obtained by mechanically dissociating the spleen, filtering it through a 40-μm nylon strainer, and lysing RBCs with ammonium-chloride-potassium lysis buffer. T cells were enriched using a Mouse CD3+ T Cell Enrichment Column (MTCC-25; R&D Systems). Cells were then stained with LIVE/DEAD Fixable Aqua Dead Cell Stain (L34957; Thermo Fisher), anti-CD4 (GK1.5, 100447; BioLegend), anti-CD8 (53-6.7, 562283; BD Biosciences), anti-CD44 (IM7, 0441-82; eBioscience), anti-CD62L (MEL-14, 47-0621-82; eBioscience), anti-Ly6C (HK1.4, 45-5932-82; eBioscience), and anti–Sca-1 (D7, 56-5981-82; eBioscience) Abs and were sorted on a FACSAria II flow cytometer (BD Biosciences). Cells were plated in tissue culture–treated round-bottom 96-well plates, 1–2 × 105 per well in 200 μl RPMI 1640 supplemented with 10% FBS, 100 U/ml penicillin, 100 U/ml streptomycin, 1 mM sodium pyruvate, 1× MEM nonessential amino acids (Life Technologies), 55 μM 2-ME. The tissue culture plates were precoated with 1 μg/ml anti-CD3 (145-2C11; Bio X Cell) for 3 h at 37°C and excess anti-CD3 was rinsed off with PBS. Cells were stimulated in the presence of anti-CD28 (37.N.51.1, 1 μg/ml), IL-2 (100 U/ml; Proleukin), anti–IFN-γ (XMG1.2, 1 μg/ml; Bio X Cell) (except when exogenous IFN-γ was tested), and anti–IL-4 (11B11, 1 μg/ml; Bio X Cell). Recombinant IL-27 (Amgen) was used at a concentration of 50 ng/ml, TGF-β (eBioscience) was used at 5 ng/ml, and Universal type I IFN (PBL Assay Science) was used at a concentration of 2000 U/ml. IFN-γ (R&D Systems), IL-6 (eBioscience), IL-12 (eBioscience), TNF-α (eBioscience), IL-10 (eBioscience), and IL-7 (PeproTech) were used at 10 ng/ml. IL-15 (PeproTech) and IL-15Ra-Fc (R&D Systems) were incubated at 37°C for 30 min at a ratio of 2:9. The resulting IL-15 complexes were used at 55 ng/ml (10 ng/ml IL-15, plus 45 ng/ml IL-15Ra).

Cells were stained with the reagents used for cell sorting, described above, as well as Abs specific for CD122 (5H4, 554452; BD Biosciences), CD127 (SB/199, 121105; BioLegend), CD69 (H1-2F3, 12-0691-83; eBioscience), CD25 (PC61, 553866; BD Biosciences), KLRG1 (2F1, 25-5893-82; eBioscience), and CD49d (R1-2, 103617; BioLegend). For analyses postinfection, splenocytes were harvested as detailed above and peritoneal exudate cells were harvested by i.p. lavage with 7 ml PBS. MHC-I monomers loaded with peptide (SVLAFRRL) from the T. gondii protein Tgd-057 were kindly provided by E. J. Wherry (University of Pennsylvania) and tetramerized by incubation with streptavidin-conjugated PE or allophycocyanin. Some experiments used PE- and allophycocyanin-conjugated MHC-I tetramers loaded with the Tgd-057 peptide that were provided by the National Institutes of Health Tetramer Facility. PE- or allophycocyanin-conjugated MHC-II tetramers loaded with the AS15 peptide AVEIHRPVPGTAPPS were also provided by the National Institutes of Health Tetramer Facility.

Cells were collected on an LSRFortessa (BD Biosciences) and analysis was performed with FlowJo (Tree Star). Cells were gated on lymphocytes (by forward scatter [FSC] and side scatter [SSC]), singlets (by FSC width versus FSC height and SSC width versus SSC height), and live cells (by exclusion of Aqua Dead Cell Stain). CD4+ T cells were gated CD4+CD8Foxp3 and CD8+ T cells were gated CD8+CD4.

Statistical significance was determined using GraphPad Prism software, using Student t test. The p values <0.05 were considered significant.

Taking into consideration previous studies that implicated IL-27 in the regulation of Ly6C on regulatory T cells (21) and Sca-1 on CD8+ T cells in vitro and CD4+ T cells in vivo (19, 20), initial experiments were performed to determine the relationship between IL-27 signaling and TCR stimulation in modulating expression of these molecules. Multiple experiments using bulk CD4+ or CD8+ splenocyte cultures showed that IL-27 in combination with TCR potently promotes T cell expression of Ly6C and Sca-1 (Supplemental Fig. 1). Because subpopulations of splenic CD4+ and CD8+ T cells express Ly6C and/or Sca-1, these experiments were repeated with sort-purified Ly6CSca-1 naive (CD44loCD62L+) T cells (>90% purity). Cells were then labeled with CFSE and cultured in the presence or absence of plate-bound anti-CD3 and soluble anti-CD28 stimulation (henceforth referred to as TCR stimulation) with IL-2, IL-27, and neutralizing anti–IFN-γ and anti–IL-4 Abs (Fig. 1A). After 3 d of culture in the absence of TCR stimulation or IL-27, the cells did not express Ly6C. Culture of CD4+ or CD8+ T cells with IL-27 alone induced modest Ly6C expression. TCR stimulation on its own induced robust proliferation (as seen by CFSE dilution), but did not induce expression of Ly6C on naive CD4+ T cells, and induced Ly6C on a small percentage of CD8+ T cells. However, when naive CD4+ and CD8+ T cells were provided TCR stimulation combined with IL-27, there was a synergistic effect on Ly6C expression, which was apparent even during early divisions. It is notable that among CD4+ T cells, TCR stimulation alone did not induce Ly6C expression, even in those cells that had proliferated. These results demonstrate that Ly6C is not a general activation marker on CD4+ T cells, but in these experiments requires TCR activation in the presence of IL-27.

FIGURE 1.

IL-27 induces Ly6C and Sca-1 on CD4+ and CD8+ T cells. Naive Ly6CSca-1 CD4+ or CD8+ T cells were sorted and stained with CFSE before a 3 d culture in the presence or absence of TCR stimulation and/or IL-27. (A and B) Induction of Ly6C (A) or Sca-1 (B) on naive CD4+ or CD8+ T cells. Left, representative flow cytometry plots. Right, bar charts summarize the results of four experiments. Error bars indicate SEM. *p < 0.05, **p < 0.01, ***p < 0.001.

FIGURE 1.

IL-27 induces Ly6C and Sca-1 on CD4+ and CD8+ T cells. Naive Ly6CSca-1 CD4+ or CD8+ T cells were sorted and stained with CFSE before a 3 d culture in the presence or absence of TCR stimulation and/or IL-27. (A and B) Induction of Ly6C (A) or Sca-1 (B) on naive CD4+ or CD8+ T cells. Left, representative flow cytometry plots. Right, bar charts summarize the results of four experiments. Error bars indicate SEM. *p < 0.05, **p < 0.01, ***p < 0.001.

Close modal

When the role of IL-27 and TCR stimulation in the regulation of Sca-1 was examined, each stimulus alone was sufficient to promote high expression of Sca-1 by CD4+ and CD8+ T cells (Fig. 1B). However, TCR stimulation in the presence of IL-27 resulted in further induction of Sca-1 expression, which was upregulated in early divisions and maintained as cells divided. We previously reported that IL-2 downregulates the IL-27 receptor (23), raising the possibility that the exogenous IL-2 used in these cultures might limit the effect of IL-27. However, exogenous IL-2 did not limit expression of Ly6C and Sca-1 by CD4+ T cells and enhanced their expression by CD8+ T cells (Supplemental Fig. 2). These results demonstrate that IL-27 has a major impact on Sca-1 and Ly6C expression, and that Sca-1 is more readily induced by these stimuli than Ly6C.

Given the impact of IL-27 on Ly6C and Sca-1, studies were performed to assess whether other cytokines (IFN-γ, type I IFN, IL-6, IL-12, IL-4, IL-10, IL-7, IL-15, and TNF-α) modulate expression of these molecules. Ly6CSca-1 naive (CD62L+CD44lo) T cells were sort purified as described above and were given TCR stimulation in the presence of the different cytokines for 72 h before being assayed for expression of Ly6C or Sca-1. Across multiple experiments, IFN-γ, type I IFN, and IL-12 induced expression of Ly6C on naive Ly6C CD4+ T cells, although not as robustly as IL-27 (Fig. 2A). For naive CD8+ T cells, both IFN-γ and type I IFN induced expression of Ly6C but type I IFN was consistently the strongest inducer of Ly6C in these experiments (Fig. 2B). Stimulation of naive CD4+ or CD8+ T cells with TCR alone induced high expression of Sca-1, but this was not further upregulated by cytokines (data not shown). IL-4, IL-10, IL-7, IL-15, and TNF-α did not modulate expression of Sca-1 or Ly6C on CD4+ or CD8+ T cells (Fig. 2, bar charts). Although there is variation in the impact of individual cytokines on different T cell populations, when taken together, these data identify two groups of related cytokines (the IFNs and the IL-6 family members) that use similar signaling pathways that promote T cell expression of Ly6C and Sca-1.

FIGURE 2.

Ly6C expression is modulated by multiple cytokines in vitro. Naive Ly6CSca-1 CD4+ or CD8+ T cells were sorted and stained with CFSE before a 3 d culture in the presence of TCR stimulation and type I IFN, IL-6, IL-12, IL-4, IL-10, IL-7, or IL-15. (A and B) Expression of Ly6C by CD4+ (A) and CD8+ (B) T cells. Left, representative plots for select cytokines. Right, bar charts summarize the results of four experiments. Error bars indicate SEM. *p < 0.05, **p < 0.01, ***p < 0.001.

FIGURE 2.

Ly6C expression is modulated by multiple cytokines in vitro. Naive Ly6CSca-1 CD4+ or CD8+ T cells were sorted and stained with CFSE before a 3 d culture in the presence of TCR stimulation and type I IFN, IL-6, IL-12, IL-4, IL-10, IL-7, or IL-15. (A and B) Expression of Ly6C by CD4+ (A) and CD8+ (B) T cells. Left, representative plots for select cytokines. Right, bar charts summarize the results of four experiments. Error bars indicate SEM. *p < 0.05, **p < 0.01, ***p < 0.001.

Close modal

There are several common elements to the signaling pathways used by the cytokines that most robustly induced expression of Ly6C and Sca-1, specifically the ability to activate STAT1 and to upregulate expression of T-bet (2426). We previously reported that Ly6C expression is T-bet–dependent in vivo during toxoplasmosis (27), but culture of T-bet−/− T cells demonstrated that T-bet is not required for IL-27–mediated induction of Ly6C and Sca-1 in vitro (Supplemental Fig. 3A, 3B). These findings agree with an earlier study that found that stimulation with anti-CD3/28 Abs overcomes the need for T-bet in the induction of Ly6C (28). A single experiment using T cells from STAT3fl/fl × CD4-Cre mice suggested that STAT3 is not required for IL-27–mediated expression of Ly6C or Sca-1 on CD4+ or CD8+ T cells (Supplemental Fig. 3C, 3D). In contrast, when naive CD8+ Ly6CSca-1 T cells from STAT1−/− mice were given TCR stimulation in the presence of IL-27 or IFN-γ, the induction of Ly6C was found to be almost entirely STAT1-dependent (Fig. 3A), as was the induction of Sca-1 (Fig. 3B). However, the type I IFN–mediated induction of Ly6C and Sca-1 was not STAT1-dependent in this system (Fig. 3A, 3B). Similar results were seen for CD4+ T cells (Fig. 3C, 3D). These results demonstrate a key role for STAT1 in IL-27– and IFN-γ–mediated induction of Ly6C and Sca-1.

FIGURE 3.

Signaling pathways involved in cytokine-mediated Ly6C and Sca-1 expression in vitro. WT or STAT1−/− splenocytes were enriched for T cells by negative-selection bead enrichment before a 3 d culture in the presence or absence of TCR stimulation and/or IL-27, IFN-γ, or type I IFN. (A and B) Expression of Ly6C (A) or Sca-1 (B) by WT and STAT1−/− CD8+ T cells. (C and D) Expression of Ly6C (C) or Sca-1 (D) by WT and STAT1−/− CD4+ T cells. Left, representative plots. Right, bar charts summarize the results of three experiments. Error bars indicate SEM. *p < 0.05, **p < 0.01, ***p < 0.001.

FIGURE 3.

Signaling pathways involved in cytokine-mediated Ly6C and Sca-1 expression in vitro. WT or STAT1−/− splenocytes were enriched for T cells by negative-selection bead enrichment before a 3 d culture in the presence or absence of TCR stimulation and/or IL-27, IFN-γ, or type I IFN. (A and B) Expression of Ly6C (A) or Sca-1 (B) by WT and STAT1−/− CD8+ T cells. (C and D) Expression of Ly6C (C) or Sca-1 (D) by WT and STAT1−/− CD4+ T cells. Left, representative plots. Right, bar charts summarize the results of three experiments. Error bars indicate SEM. *p < 0.05, **p < 0.01, ***p < 0.001.

Close modal

Although multiple cytokines promote expression of Ly6C and Sca-1, this screening process revealed that TGF-β was a potent inhibitor of the expression of these molecules. The addition of TGF-β reduced IL-27–mediated expression of Ly6C on CD4+ T cells (Fig. 4A, 4C) and CD8+ T cells (Fig. 4B, 4C), which is similar to results from a previous study that used P14 cells in the context of lymphocytic choriomeningitis virus infection (29). Addition of TGF-β also limited proliferation of CD4+ and CD8+ T cells, as illustrated by the reduced dilution of CFSE (Fig. 4A, 4B, 4D, 4E). This was true in the presence and absence of IL-27 and is consistent with the ability of TGF-β to limit the proliferation and differentiation of naive T cells into terminally differentiated effector cells (30). Addition of TGF-β also reduced TCR-mediated induction of Sca-1 on CD4+ T cells (Fig. 4D, 4F) and on CD8+ T cells (Fig. 4E, 4F), but did not affect the expression of Sca-1 in the presence of IL-27. The relatively modest effect that TGF-β has on IL-27–mediated expression of Sca-1 is consistent with the data in earlier figures that Sca-1 is more robustly expressed than Ly6C, and also indicates that the inhibitory effects of TGF-β are most closely associated with reduced TCR signaling.

FIGURE 4.

TGF-β inhibits induction of Ly6C and Sca-1. Naive Ly6CSca-1 CD4+ or CD8+ T cells were sorted and stained with CFSE before a 3 d culture with TCR stimulation in the presence or absence of IL-27 and/or TGF-β. (AC) Expression of Ly6C by CD4+ (A and C) and CD8+ (B and C) T cells after culture. (DF) Expression of Sca-1 by CD4+ (D and F) and CD8+ (E and F) T cells after culture. (A, B, D, and E) Representative plots. (C and F) Bar charts summarize the results of four experiments. Error bars indicate SEM. *p < 0.05, **p < 0.01.

FIGURE 4.

TGF-β inhibits induction of Ly6C and Sca-1. Naive Ly6CSca-1 CD4+ or CD8+ T cells were sorted and stained with CFSE before a 3 d culture with TCR stimulation in the presence or absence of IL-27 and/or TGF-β. (AC) Expression of Ly6C by CD4+ (A and C) and CD8+ (B and C) T cells after culture. (DF) Expression of Sca-1 by CD4+ (D and F) and CD8+ (E and F) T cells after culture. (A, B, D, and E) Representative plots. (C and F) Bar charts summarize the results of four experiments. Error bars indicate SEM. *p < 0.05, **p < 0.01.

Close modal

Ly6C has been used to identify terminally differentiated effector CD4+ T cells during infection (6, 10, 11), but its expression during homeostasis is not well described. To determine which T cell populations express Ly6C under homeostatic conditions, a survey of uninfected SPF mice was conducted. The expression of high levels of CD44 was used to identify Ag-experienced cells (31) and CD62L was used to identify cells that home to lymph nodes, which are primarily naive and central memory cells (32). In this setting, 20% of naive (CD62L+CD44lo) and 10% of memory (CD62L+CD44hi) CD4+ T cells expressed Ly6C and only 10% of CD4+ T cells with an effector phenotype (CD62LCD44hi) were Ly6C+ (Fig. 5A). For CD8+ T cells, 20% of naive (CD44loCD62L+) cells expressed Ly6C, whereas ∼30% of CD44hiCD62L and 90% of CD44hiCD62L+ CD8+ T cells expressed Ly6C. Thus, Ly6C expression was concentrated on the Ag-experienced cells, but further subsetting based on T cell expression of CD25, KLRG1, CD127, CD69, or CD49d indicated that Ly6C expression is not restricted to a particular effector/memory T cell population (data not shown). However, it is notable that in this survey, the majority of CD122+ CD8+ T cells expressed Ly6C, regardless of their expression of CD62L and CD44 (Supplemental Fig. 4). CD122 is the β subunit of the IL-2 and IL-15 receptors and, along with CD44 and Ly6C, is upregulated on T cells undergoing homeostatic proliferation (2, 33, 34).

FIGURE 5.

Expression of Ly6C and Sca-1 by naive, effector, and memory populations. Splenocytes from WT C57BL/6 mice were harvested and analyzed for expression of Ly6C and Sca-1. (A) Expression of Ly6C by CD4+ (top) and CD8+ (bottom) T cell populations. (B) Expression of Sca-1 by CD4+ (top) and CD8+ (bottom) T cell populations. Left, representative plots. Right, bar charts summarize the results of three experiments. Error bars indicate SEM. *p < 0.05, **p < 0.01.

FIGURE 5.

Expression of Ly6C and Sca-1 by naive, effector, and memory populations. Splenocytes from WT C57BL/6 mice were harvested and analyzed for expression of Ly6C and Sca-1. (A) Expression of Ly6C by CD4+ (top) and CD8+ (bottom) T cell populations. (B) Expression of Sca-1 by CD4+ (top) and CD8+ (bottom) T cell populations. Left, representative plots. Right, bar charts summarize the results of three experiments. Error bars indicate SEM. *p < 0.05, **p < 0.01.

Close modal

When a similar survey was performed to assess the expression of Sca-1 on naive, effector, and memory T cells in SPF mice, 20% of naive (CD44loCD62L+) CD4+ T cells expressed Sca-1, whereas 80% of CD44hiCD62L effector CD4+ T cells were Sca-1+ (Fig. 5B). Similarly, minimal Sca-1 expression was seen in CD44loCD62L+ CD8+ T cells, whereas the highest Sca-1 expression by CD8+ T cells (15%) was seen in the CD44hiCD62L population (Fig. 5B). These findings indicate that Sca-1 was enriched on CD44hi CD4+ and CD8+ T cells, but in this survey was not exclusively expressed by any particular effector/memory population examined.

The above survey used established surface markers to distinguish Ag-experienced and naive cells, but in this setting, it is difficult to determine how expression of Ly6C or Sca-1 correlates with previous Ag exposure. To examine an effector population with a well-defined history, mice were infected with T. gondii and the expression of Ly6C and Sca-1 on parasite-specific T cells during acute toxoplasmosis was examined. Mice were infected i.p. with T. gondii and spleens and peritoneal exudate cells were harvested 10 d postinfection. Toxoplasma-specific T cells were identified by staining with parasite-specific MHC-I or MHC-II tetramers in combination with high expression of LFA-1 (35). At day 10 postinfection, the majority of parasite-specific CD4+ and CD8+ T cells in the peritoneum expressed Ly6C, demonstrating that toxoplasmosis promotes Ly6C expression by parasite-specific T cells (Fig. 6A).

FIGURE 6.

Toxoplasmosis upregulates Ly6C and Sca-1 in T cells. WT C57BL/6 mice were infected with 20 T. gondii cysts i.p. Splenocytes were harvested and analyzed at day 9 of infection. T. gondii–specific T cells were identified by MHC-I and MHC-II tetramers. (A and D) Expression of Ly6C (A) or Sca-1 (D) by CD4+ and CD8+ T cells. (B) Expression of CXCR3 and KLRG1 by T. gondii–specific CD4+ and CD8+ T cells. (C and E) Expression of Ly6C (C) or Sca-1 (E) by different populations of T cells, distinguished by expression of CXCR3 and KLRG1. Left, representative plots. Right, bar charts summarize results from one of two experiments, n = 3–4 mice per experiment. Error bars indicate SEM. *p < 0.05, **p < 0.01.

FIGURE 6.

Toxoplasmosis upregulates Ly6C and Sca-1 in T cells. WT C57BL/6 mice were infected with 20 T. gondii cysts i.p. Splenocytes were harvested and analyzed at day 9 of infection. T. gondii–specific T cells were identified by MHC-I and MHC-II tetramers. (A and D) Expression of Ly6C (A) or Sca-1 (D) by CD4+ and CD8+ T cells. (B) Expression of CXCR3 and KLRG1 by T. gondii–specific CD4+ and CD8+ T cells. (C and E) Expression of Ly6C (C) or Sca-1 (E) by different populations of T cells, distinguished by expression of CXCR3 and KLRG1. Left, representative plots. Right, bar charts summarize results from one of two experiments, n = 3–4 mice per experiment. Error bars indicate SEM. *p < 0.05, **p < 0.01.

Close modal

A recent study (4) proposed that during toxoplasmosis, minimally differentiated memory CD8+ T cells that are CXCR3+KLRG1 give rise to an intermediate CXCR3+KLRG1+ population that in turn downregulates CXCR3 when it differentiates into terminally differentiated effector cells. Consistent with this previous report, at day 10 of infection, 20% of splenic parasite-specific CD8+ T cells were CXCR3+KLRG1, 60% were CXCR3+KLRG1+ and <10% were CXCR3KLRG1+. This analysis was extended to CD4+ T cells, in which 20% were CXCR3+KLRG1, 30% were CXCR3+KLRG1+ and 30% were CXCR3KLRG1+ (Fig. 6B). Seventy percent of parasite-specific CD4+ T cells expressed Ly6C (Fig. 6A). When the cells were subsetted by expression of CXCR3 and KLRG1, Ly6C was expressed by 60% of CXCR3+KLRG1, 75% of CXCR3+KLRG1+, and 70% of CXCR3KLRG1+ parasite-specific CD4+ T cells. Similar results were seen for CD8+ T cells, as Ly6C was expressed by 90% of CXCR3+KLRG1, 95% of CXCR3+KLRG1+, and 85% of CXCR3KLRG1+ parasite-specific CD8+ T cells. Therefore, Ly6C expression on these individual subsets was not exclusive to the CXCR3KLRG1+ population for CD4+ or CD8+ T cells, demonstrating that Ly6C and KLRG1 are not interchangeable markers of differentiation (Fig. 6C).

When a similar analysis was performed for Sca-1, 85% of parasite-specific CD4+ and CD8+ T cells expressed Sca-1 (Fig. 6D). When these cells were subsetted by their expression of KLRG1 and CXCR3, ∼90% of the CXCR3+KLRG1 and CXCR3+KLRG1+ populations expressed Sca-1 in both CD4+ and CD8+ T cells (Fig. 6E). Seventy percent of CXCR3KLRG1+ CD4+ T cells and 60% of CXCR3KLRG1+ CD8+ T cells expressed Ly6C, indicating that Sca-1 is present at a lower frequency on more highly differentiated cells. Nonetheless, Sca-1 is more widely expressed during toxoplasmosis than KLRG1 or CXCR3, and does not enable the ready differentiation of distinct Ag-experienced T cell populations in this experimental system.

IL-27 and IFN-γ are key cytokines during toxoplasmosis (36, 37) and are two of the strongest inducers of Ly6C and Sca-1 in vitro. To determine the contribution of IL-27 and IFN-γ signaling to the expression of Ly6C and Sca-1 during infection, wild-type (WT) and IL-27 receptor (Il27ra)-deficient mice were infected with T. gondii. Mice were also treated with an isotype Ab or a neutralizing anti–IFN-γ Ab on days 3 and 6 of infection. When peritoneal tetramer-positive T cells were examined at day 9 of infection, the percentage of parasite-specific CD4+ T cells expressing Ly6C was substantially lower in Il27ra–deficient mice than in WT mice. Ly6C levels were significantly reduced in CD8+ T cells as well, providing evidence that IL-27 promotes the Ly6C+ population in this system (Fig. 7A, 7B). Additionally, neutralizing IFN-γ resulted in less Ly6C expression by CD4+ T cells, but in Il27ra–deficient mice did not result in a complete ablation of Ly6C expression. In contrast, the absence of the IL-27 receptor did not limit the expression of Sca-1 by parasite-specific T cells (Fig. 7C, 7D). Surprisingly, the blockade of IFN-γ increased Sca-1 expression in both WT and Il27ra−/− mice, possibly because the absence of IFN-γ leads to a marked increase in parasite replication and Ag load that might lead to increased T cell activation. Together, these studies establish that IL-27 and IFN-γ are involved in the regulation of Ly6C expression during toxoplasmosis, but in this setting they were not required for maximal Sca-1 expression.

FIGURE 7.

IL-27 and IFN-γ promote expression of Ly6C in vivo during toxoplasmosis. WT or Il27ra−/− mice were infected with 20 T. gondii cysts i.p. and a subset of mice were treated with anti–IFN-γ Ab on days 3 and 6 of infection. Peritoneal cells were harvested 9 d postinfection and T. gondii–specific cells were identified using MHC-I and MHC-II tetramers. Ly6C expression by tetramer+ CD4+ (A) or CD8+ (B) T cells was examined. Sca-1 expression by tetramer+ CD4+ (C) or CD8+ (D) T cells was also examined. Left, representative plots with mean fluorescence intensity (MFI) indicated by vertical italic numbers. Right, bar charts show representative results from one of two experiments, n = 2–4 mice per group, per experiment. Error bars indicate SEM. *p < 0.05, **p < 0.01, ***p < 0.001.

FIGURE 7.

IL-27 and IFN-γ promote expression of Ly6C in vivo during toxoplasmosis. WT or Il27ra−/− mice were infected with 20 T. gondii cysts i.p. and a subset of mice were treated with anti–IFN-γ Ab on days 3 and 6 of infection. Peritoneal cells were harvested 9 d postinfection and T. gondii–specific cells were identified using MHC-I and MHC-II tetramers. Ly6C expression by tetramer+ CD4+ (A) or CD8+ (B) T cells was examined. Sca-1 expression by tetramer+ CD4+ (C) or CD8+ (D) T cells was also examined. Left, representative plots with mean fluorescence intensity (MFI) indicated by vertical italic numbers. Right, bar charts show representative results from one of two experiments, n = 2–4 mice per group, per experiment. Error bars indicate SEM. *p < 0.05, **p < 0.01, ***p < 0.001.

Close modal

Studies to understand the functions of Ly6 molecules have been performed since the 1970s, but questions still remain about their functions and the factors that influence their expression (38). The association of Ly6C expression with short-lived effector T cells has been reported in multiple experimental systems (6, 10) and the combination of Sca-1, CD122, and Bcl-2 can be used to identify CD8+ memory stem cells (1214). A previous study showed that treatment of mice with IL-27 promotes the development of a memory precursor population of tumor Ag-specific CD8+ T cells, characterized by high expression of Bcl-6, SOCS3, and Sca-1 (19). However, the expression patterns of Ly6C and Sca-1 have not been compared and there is a limited appreciation of how cytokine and TCR-mediated signals are integrated to promote their expression. The in vitro experiments performed here used a defined population of naive Ly6C CD4+ and CD8+ T cells to address any concerns that the stimuli used here might preferentially expand a Ly6C+ population. This approach showed quite modest effects of IL-27, the IFNs, or TCR alone on Ly6C expression, but the combination of cytokine plus TCR synergistically promoted the expression of Ly6C by a subset of activated CD4+ and CD8+ T cells. That this activity was STAT1-dependent correlated well with the range of cytokines that could modulate Ly6C, whereas cytokines that predominantly use STAT3, STAT4, or STAT6 had minimal effects. TGF-β was the only signal identified that suppressed the induction of Ly6C and Sca-1, which correlates with its ability to suppress T cell activation and proliferation (39, 40).

Because Ly6C and Sca-1 have been used as markers to identify T cells at different stages of differentiation, it was notable that the expression of these two molecules was upregulated by the same cytokine signals, in the same cells. Sca-1 was potently induced by either TCR stimulation or cytokine signaling and consequently appeared to be more widely expressed than Ly6C. Indeed, a survey to determine if either of these molecules could be associated with different effector or memory populations found that it was difficult to link them to memory-like or terminally differentiated effector cells based on differential expression of KLRG1 and CXCR3. The ability of these cytokines to induce Ly6C and most notably Sca-1 in the absence of TCR stimulation suggests the need for caution in using these molecules alone to identify Ag-experienced populations.

Infection with T. gondii is dominated by the generation of parasite-specific CD4+ and CD8+ T cells that produce IFN-γ, but this is also a system in which endogenous IL-27 is required to limit the inflammatory response (37). Although the loss of either cytokine signal during infection reduces Ly6C expression, it was relevant to note that in the setting of IL-27R deficiency, there are markedly elevated levels of IFN-γ (37), but IFN-γ blockade did not result in a further reduction in the numbers of parasite-specific effectors that expressed Ly6C. The observation that IL-27 was more important in driving Ly6C in CD4+ T cells than in CD8+ T cells during toxoplasmosis is consistent with the larger effect of IFNs in promoting Ly6C in CD8+ T cells seen in vitro. Nevertheless, the in vivo studies presented here indicate that IL-27 and IFN-γ are not redundant in promoting Ly6C expression during toxoplasmosis.

Although the section above focuses on the regulation of Ly6C, the in vitro and in vivo studies identified common pathways that influence expression of Ly6C and Sca-1, but also highlighted some notable differences. Again, the ability to use a defined, naive Sca-1 starting population helped establish the profound impact of TCR stimulation alone or in combination with cytokines on Sca-1 expression. As seen for Ly6C, not every cytokine was a potent inducer of Sca-1 and those that activated STAT1 seemed dominant, although the use of STAT1-deficient cells does indicate the presence of additional pathways that are involved in this process. Potential STAT1-independent pathways relevant to IL-27 and type I IFNs include p38 MAPK and ERK1/2 (24, 25). However, unlike Ly6C, Sca-1 expression was not attenuated by the loss of IL-27 or IFN-γ in in vivo studies. This contrasts with an earlier report using a model of colitis, in which in vivo administration of an adeno-associated virus vector encoding IL-27 was associated with reduced inflammation and increased expression of Sca-1 by CD4+ T cells (20). This contradiction likely reflects the complexity in trying to distinguish a role for endogenous IL-27 or IFN-γ in a systemic infection, in which many signals including TCR and other cytokines readily promote Sca-1 expression, versus a dominant signal provided by overexpression of IL-27.

There have been few studies that have directly addressed the function of Ly6C and Sca-1 in the immune response. Ly6C has been proposed to be involved in T cell homing to secondary lymphoid organs, possibly through an association with LFA-1 (4143), and ImageStream analysis of T. gondii–specific effectors shows colocalization of Ly6C and CD11a (a subunit of LFA-1) on the surface of these cells (27). IL-27 has been proposed to modulate T cell homing through upregulation of ICAM-1 and selectin ligands on conventional CD4+ T cells (21, 44, 45), as well as CXCR3 on regulatory T cells (21). Thus, upregulation of Ly6C may be an additional mechanism by which IL-27 and/or IFNs modulate T cell trafficking. There is also a literature that suggests a regulatory role for Ly6C and Sca-1 in limiting the T cell response. A mutation in the promoter of Ly6C reduces its expression in NOD, NZB/W, and ST mice, which are strains that spontaneously develop autoimmune diseases (46). Moreover, in the context of TCR stimulation, Abs that crosslink Ly6C or Sca-1 on the surface of T cells limit their ability to produce IL-2 and proliferate (4749). Furthermore, in mice genetically engineered to lack Sca-1 expression, T cells exhibit enhanced proliferation in response to TCR stimulation (50). Additional evidence for a regulatory function of Sca-1 is the finding that transgenic overexpression of Sca-1 limits T cell proliferation (48, 49, 51) and suppresses lymphoproliferation and autoimmunity in lpr/lpr mice (51). Together, these findings suggest that Ly6C and/or Sca-1 may have a role in limiting T cell responses. A suppressive function for Ly6C and Sca-1 would complement reports that IL-27 promotes inhibitory pathways including IL-10 and LAG-3 (5258), and that IL-27 and IFN-γ promote expression of PD-L1 (5860). Additional studies are needed to determine whether Ly6C and Sca-1 primarily function to promote T cell activation and migration, or if any of the shared immune regulatory effects of IL-27 and the IFNs are mediated through the induction of Ly6C and/or Sca-1.

We thank the members of the Hunter laboratory for intellectual and experimental contributions to this study. We thank the University of Pennsylvania Flow Cytometry and Cell Sorting Facility for support.

This work was supported by National Institutes of Health Grant T32 5T32AR007442-28 (to J.H.D.), American Society of Nephrology Ben J. Lipps Research Fellowship (to G.M.C.), University of Pennsylvania School of Medicine Measey Senior Research Fellowship (to G.M.C.), National Institutes of Health Grant R01 5R01AI110201-03 (to C.A.H.), and the Commonwealth of Pennsylvania.

The online version of this article contains supplemental material.

Abbreviations used in this article:

FSC

forward scatter

SPF

specific pathogen-free

SSC

side scatter

WT

wild-type.

1
Joshi
,
N. S.
,
S. M.
Kaech
.
2008
.
Effector CD8 T cell development: a balancing act between memory cell potential and terminal differentiation.
J. Immunol.
180
:
1309
1315
.
2
Murali-Krishna
,
K.
,
R.
Ahmed
.
2000
.
Cutting edge: naive T cells masquerading as memory cells.
J. Immunol.
165
:
1733
1737
.
3
Kaech
,
S. M.
,
W.
Cui
.
2012
.
Transcriptional control of effector and memory CD8+ T cell differentiation.
Nat. Rev. Immunol.
12
:
749
761
.
4
Chu
,
H. H.
,
S. W.
Chan
,
J. P.
Gosling
,
N.
Blanchard
,
A.
Tsitsiklis
,
G.
Lythe
,
N.
Shastri
,
C.
Molina-París
,
E. A.
Robey
.
2016
.
Continuous effector CD8(+) T cell production in a controlled persistent infection is sustained by a proliferative intermediate population.
Immunity
45
:
159
171
.
5
Gerlach
,
C.
,
E. A.
Moseman
,
S. M.
Loughhead
,
D.
Alvarez
,
A. J.
Zwijnenburg
,
L.
Waanders
,
R.
Garg
,
J. C.
de la Torre
,
U. H.
von Andrian
.
2016
.
The chemokine receptor CX3CR1 defines three antigen-experienced CD8 T cell subsets with distinct roles in immune surveillance and homeostasis.
Immunity
45
:
1270
1284
.
6
Marshall
,
H. D.
,
A.
Chandele
,
Y. W.
Jung
,
H.
Meng
,
A. C.
Poholek
,
I. A.
Parish
,
R.
Rutishauser
,
W.
Cui
,
S. H.
Kleinstein
,
J.
Craft
,
S. M.
Kaech
.
2011
.
Differential expression of Ly6C and T-bet distinguish effector and memory Th1 CD4(+) cell properties during viral infection.
Immunity
35
:
633
646
.
7
Peters
,
N. C.
,
A. J.
Pagán
,
P. G.
Lawyer
,
T. W.
Hand
,
E.
Henrique Roma
,
L. W.
Stamper
,
A.
Romano
,
D. L.
Sacks
.
2014
.
Chronic parasitic infection maintains high frequencies of short-lived Ly6C+CD4+ effector T cells that are required for protection against re-infection.
PLoS Pathog.
10
:
e1004538
.
8
Hand
,
T. W.
,
M.
Morre
,
S. M.
Kaech
.
2007
.
Expression of IL-7 receptor alpha is necessary but not sufficient for the formation of memory CD8 T cells during viral infection.
Proc. Natl. Acad. Sci. USA
104
:
11730
11735
.
9
Kong
,
H. K.
,
J. H.
Park
.
2012
.
Characterization and function of human Ly-6/uPAR molecules.
BMB Rep.
45
:
595
603
.
10
Hu
,
Z.
,
M. A.
Blackman
,
K. M.
Kaye
,
E. J.
Usherwood
.
2015
.
Functional heterogeneity in the CD4+ T cell response to murine γ-herpesvirus 68.
J. Immunol.
194
:
2746
2756
.
11
Walunas
,
T. L.
,
D. S.
Bruce
,
L.
Dustin
,
D. Y.
Loh
,
J. A.
Bluestone
.
1995
.
Ly-6C is a marker of memory CD8+ T cells.
J. Immunol.
155
:
1873
1883
.
12
Zhang
,
Y.
,
G.
Joe
,
E.
Hexner
,
J.
Zhu
,
S. G.
Emerson
.
2005
.
Host-reactive CD8+ memory stem cells in graft-versus-host disease.
Nat. Med.
11
:
1299
1305
.
13
Gattinoni
,
L.
,
X. S.
Zhong
,
D. C.
Palmer
,
Y.
Ji
,
C. S.
Hinrichs
,
Z.
Yu
,
C.
Wrzesinski
,
A.
Boni
,
L.
Cassard
,
L. M.
Garvin
, et al
.
2009
.
Wnt signaling arrests effector T cell differentiation and generates CD8+ memory stem cells.
Nat. Med.
15
:
808
813
.
14
Gattinoni
,
L.
,
E.
Lugli
,
Y.
Ji
,
Z.
Pos
,
C. M.
Paulos
,
M. F.
Quigley
,
J. R.
Almeida
,
E.
Gostick
,
Z.
Yu
,
C.
Carpenito
, et al
.
2011
.
A human memory T cell subset with stem cell-like properties.
Nat. Med.
17
:
1290
1297
.
15
Lanzavecchia
,
A.
,
F.
Sallusto
.
2002
.
Progressive differentiation and selection of the fittest in the immune response.
Nat. Rev. Immunol.
2
:
982
987
.
16
Obar
,
J. J.
,
E. R.
Jellison
,
B. S.
Sheridan
,
D. A.
Blair
,
Q. M.
Pham
,
J. M.
Zickovich
,
L.
Lefrançois
.
2011
.
Pathogen-induced inflammatory environment controls effector and memory CD8+ T cell differentiation.
J. Immunol.
187
:
4967
4978
.
17
Schlueter
,
A. J.
,
A. M.
Krieg
,
P.
de Vries
,
X.
Li
.
2001
.
Type I interferon is the primary regulator of inducible Ly-6C expression on T cells.
J. Interferon Cytokine Res.
21
:
621
629
.
18
Dumont
,
F. J.
,
R. G.
Palfree
,
L. Z.
Coker
.
1986
.
Phenotypic changes induced by interferon in resting T cells: major enhancement of Ly-6 antigen expression.
J. Immunol.
137
:
201
210
.
19
Liu
,
Z.
,
J. Q.
Liu
,
F.
Talebian
,
L. C.
Wu
,
S.
Li
,
X. F.
Bai
.
2013
.
IL-27 enhances the survival of tumor antigen-specific CD8+ T cells and programs them into IL-10-producing, memory precursor-like effector cells.
Eur. J. Immunol.
43
:
468
479
.
20
Zhu
,
X.
,
Z.
Liu
,
J. Q.
Liu
,
J.
Zhu
,
J.
Zhang
,
J. P.
Davis
,
J.
Chu
,
J.
Yu
,
J.
Zhou
,
M. S.
Li
,
X. F.
Bai
.
2016
.
Systemic delivery of IL-27 by an adeno-associated viral vector inhibits T cell-mediated colitis and induces multiple inhibitory pathways in T cells.
J. Leukoc. Biol.
100
:
403
411
.
21
Hall
,
A. O.
,
D. P.
Beiting
,
C.
Tato
,
B.
John
,
G.
Oldenhove
,
C. G.
Lombana
,
G. H.
Pritchard
,
J. S.
Silver
,
N.
Bouladoux
,
J. S.
Stumhofer
, et al
.
2012
.
The cytokines interleukin 27 and interferon-γ promote distinct Treg cell populations required to limit infection-induced pathology.
Immunity
37
:
511
523
.
22
Yoshida
,
H.
,
S.
Hamano
,
G.
Senaldi
,
T.
Covey
,
R.
Faggioni
,
S.
Mu
,
M.
Xia
,
A. C.
Wakeham
,
H.
Nishina
,
J.
Potter
, et al
.
2001
.
WSX-1 is required for the initiation of Th1 responses and resistance to L. major infection.
Immunity
15
:
569
578
.
23
Villarino
,
A. V.
,
J.
Larkin
III
,
C. J.
Saris
,
A. J.
Caton
,
S.
Lucas
,
T.
Wong
,
F. J.
de Sauvage
,
C. A.
Hunter
.
2005
.
Positive and negative regulation of the IL-27 receptor during lymphoid cell activation.
J. Immunol.
174
:
7684
7691
.
24
Hall
,
A. O.
,
J. S.
Silver
,
C. A.
Hunter
.
2012
.
The immunobiology of IL-27.
Adv. Immunol.
115
:
1
44
.
25
Platanias
,
L. C.
2005
.
Mechanisms of type-I- and type-II-interferon-mediated signalling.
Nat. Rev. Immunol.
5
:
375
386
.
26
Lazarevic
,
V.
,
L. H.
Glimcher
,
G. M.
Lord
.
2013
.
T-bet: a bridge between innate and adaptive immunity.
Nat. Rev. Immunol.
13
:
777
789
.
27
Harms Pritchard
,
G.
,
A. O.
Hall
,
D. A.
Christian
,
S.
Wagage
,
Q.
Fang
,
G.
Muallem
,
B.
John
,
A.
Glatman Zaretsky
,
W. G.
Dunn
,
J.
Perrigoue
, et al
.
2015
.
Diverse roles for T-bet in the effector responses required for resistance to infection.
J. Immunol.
194
:
1131
1140
.
28
Sullivan
,
B. M.
,
A.
Juedes
,
S. J.
Szabo
,
M.
von Herrath
,
L. H.
Glimcher
.
2003
.
Antigen-driven effector CD8 T cell function regulated by T-bet.
Proc. Natl. Acad. Sci. USA
100
:
15818
15823
.
29
Casey
,
K. A.
,
K. A.
Fraser
,
J. M.
Schenkel
,
A.
Moran
,
M. C.
Abt
,
L. K.
Beura
,
P. J.
Lucas
,
D.
Artis
,
E. J.
Wherry
,
K.
Hogquist
, et al
.
2012
.
Antigen-independent differentiation and maintenance of effector-like resident memory T cells in tissues.
J. Immunol.
188
:
4866
4875
.
30
Gorelik
,
L.
,
R. A.
Flavell
.
2002
.
Transforming growth factor-beta in T-cell biology.
Nat. Rev. Immunol.
2
:
46
53
.
31
Budd
,
R. C.
,
J. C.
Cerottini
,
C.
Horvath
,
C.
Bron
,
T.
Pedrazzini
,
R. C.
Howe
,
H. R.
MacDonald
.
1987
.
Distinction of virgin and memory T lymphocytes. Stable acquisition of the Pgp-1 glycoprotein concomitant with antigenic stimulation.
J. Immunol.
138
:
3120
3129
.
32
Lefrançois
,
L.
2006
.
Development, trafficking, and function of memory T-cell subsets.
Immunol. Rev.
211
:
93
103
.
33
Goldrath
,
A. W.
,
L. Y.
Bogatzki
,
M. J.
Bevan
.
2000
.
Naive T cells transiently acquire a memory-like phenotype during homeostasis-driven proliferation.
J. Exp. Med.
192
:
557
564
.
34
Cho
,
B. K.
,
V. P.
Rao
,
Q.
Ge
,
H. N.
Eisen
,
J.
Chen
.
2000
.
Homeostasis-stimulated proliferation drives naive T cells to differentiate directly into memory T cells.
J. Exp. Med.
192
:
549
556
.
35
Dupont
,
C. D.
,
D. A.
Christian
,
E. M.
Selleck
,
M.
Pepper
,
M.
Leney-Greene
,
G.
Harms Pritchard
,
A. A.
Koshy
,
S.
Wagage
,
M. A.
Reuter
,
L. D.
Sibley
, et al
.
2014
.
Parasite fate and involvement of infected cells in the induction of CD4+ and CD8+ T cell responses to Toxoplasma gondii.
PLoS Pathog.
10
:
e1004047
.
36
Suzuki
,
Y.
,
M. A.
Orellana
,
R. D.
Schreiber
,
J. S.
Remington
.
1988
.
Interferon-gamma: the major mediator of resistance against Toxoplasma gondii.
Science
240
:
516
518
.
37
Villarino
,
A.
,
L.
Hibbert
,
L.
Lieberman
,
E.
Wilson
,
T.
Mak
,
H.
Yoshida
,
R. A.
Kastelein
,
C.
Saris
,
C. A.
Hunter
.
2003
.
The IL-27R (WSX-1) is required to suppress T cell hyperactivity during infection.
Immunity
19
:
645
655
.
38
Shevach
,
E. M.
,
P. E.
Korty
.
1989
.
Ly-6: a multigene family in search of a function.
Immunol. Today
10
:
195
200
.
39
Gorelik
,
L.
,
R. A.
Flavell
.
2000
.
Abrogation of TGFbeta signaling in T cells leads to spontaneous T cell differentiation and autoimmune disease.
Immunity
12
:
171
181
.
40
Kehrl
,
J. H.
,
L. M.
Wakefield
,
A. B.
Roberts
,
S.
Jakowlew
,
M.
Alvarez-Mon
,
R.
Derynck
,
M. B.
Sporn
,
A. S.
Fauci
.
1986
.
Production of transforming growth factor beta by human T lymphocytes and its potential role in the regulation of T cell growth.
J. Exp. Med.
163
:
1037
1050
.
41
Jaakkola
,
I.
,
M.
Merinen
,
S.
Jalkanen
,
A.
Hänninen
.
2003
.
Ly6C induces clustering of LFA-1 (CD11a/CD18) and is involved in subtype-specific adhesion of CD8 T cells.
J. Immunol.
170
:
1283
1290
.
42
Hänninen
,
A.
,
I.
Jaakkola
,
M.
Salmi
,
O.
Simell
,
S.
Jalkanen
.
1997
.
Ly-6C regulates endothelial adhesion and homing of CD8(+) T cells by activating integrin-dependent adhesion pathways.
Proc. Natl. Acad. Sci. USA
94
:
6898
6903
.
43
Hänninen
,
A.
,
M.
Maksimow
,
C.
Alam
,
D. J.
Morgan
,
S.
Jalkanen
.
2011
.
Ly6C supports preferential homing of central memory CD8+ T cells into lymph nodes.
Eur. J. Immunol.
41
:
634
644
.
44
Owaki
,
T.
,
M.
Asakawa
,
N.
Morishima
,
K.
Hata
,
F.
Fukai
,
M.
Matsui
,
J.
Mizuguchi
,
T.
Yoshimoto
.
2005
.
A role for IL-27 in early regulation of Th1 differentiation.
J. Immunol.
175
:
2191
2200
.
45
Ebel
,
M. E.
,
O.
Awe
,
M. H.
Kaplan
,
G. S.
Kansas
.
2015
.
Diverse inflammatory cytokines induce selectin ligand expression on murine CD4 T cells via p38α MAPK.
J. Immunol.
194
:
5781
5788
.
46
Philbrick
,
W. M.
,
S. E.
Maher
,
M. M.
Bridgett
,
A. L.
Bothwell
.
1990
.
A recombination event in the 5′ flanking region of the Ly-6C gene correlates with impaired expression in the NOD, NZB and ST strains of mice.
EMBO J.
9
:
2485
2492
.
47
Yamanouchi
,
S.
,
K.
Kuwahara
,
A.
Sakata
,
T.
Ezaki
,
S.
Matsuoka
,
J.
Miyazaki
,
S.
Hirose
,
T.
Tamura
,
H.
Nariuchi
,
N.
Sakaguchi
.
1998
.
A T cell activation antigen, Ly6C, induced on CD4+ Th1 cells mediates an inhibitory signal for secretion of IL-2 and proliferation in peripheral immune responses.
Eur. J. Immunol.
28
:
696
707
.
48
Codias
,
E. K.
,
T. J.
Fleming
,
C. M.
Zacharchuk
,
J. D.
Ashwell
,
T. R.
Malek
.
1992
.
Role of Ly-6A/E and T cell receptor-zeta for IL-2 production. Phosphatidylinositol-anchored Ly-6A/E antagonizes T cell receptor-mediated IL-2 production by a zeta-independent pathway.
J. Immunol.
149
:
1825
1852
.
49
Fleming
,
T. J.
,
T. R.
Malek
.
1994
.
Multiple glycosylphosphatidylinositol-anchored Ly-6 molecules and transmembrane Ly-6E mediate inhibition of IL-2 production.
J. Immunol.
153
:
1955
1962
.
50
Stanford
,
W. L.
,
S.
Haque
,
R.
Alexander
,
X.
Liu
,
A. M.
Latour
,
H. R.
Snodgrass
,
B. H.
Koller
,
P. M.
Flood
.
1997
.
Altered proliferative response by T lymphocytes of Ly-6A (Sca-1) null mice.
J. Exp. Med.
186
:
705
717
.
51
Henderson
,
S. C.
,
M. M.
Kamdar
,
A.
Bamezai
.
2002
.
Ly-6A.2 expression regulates antigen-specific CD4+ T cell proliferation and cytokine production.
J. Immunol.
168
:
118
126
.
52
Awasthi
,
A.
,
Y.
Carrier
,
J. P.
Peron
,
E.
Bettelli
,
M.
Kamanaka
,
R. A.
Flavell
,
V. K.
Kuchroo
,
M.
Oukka
,
H. L.
Weiner
.
2007
.
A dominant function for interleukin 27 in generating interleukin 10-producing anti-inflammatory T cells.
Nat. Immunol.
8
:
1380
1389
.
53
Do
,
J.-s.
,
A.
Visperas
,
Y. O.
Sanogo
,
J. J.
Bechtel
,
N.
Dvorina
,
S.
Kim
,
E.
Jang
,
S. A.
Stohlman
,
B.
Shen
,
R. L.
Fairchild
, et al
.
2016
.
An IL-27/Lag3 axis enhances Foxp3+ regulatory T cell–suppressive function and therapeutic efficacy.
Mucosal Immunol.
9
:
137
145
.
54
Karakhanova
,
S.
,
T.
Bedke
,
A. H.
Enk
,
K.
Mahnke
.
2011
.
IL-27 renders DC immunosuppressive by induction of B7-H1.
J. Leukoc. Biol.
89
:
837
845
.
55
Mascanfroni
,
I. D.
,
A.
Yeste
,
S. M.
Vieira
,
E. J.
Burns
,
B.
Patel
,
I.
Sloma
,
Y.
Wu
,
L.
Mayo
,
R.
Ben-Hamo
,
S.
Efroni
, et al
.
2013
.
IL-27 acts on DCs to suppress the T cell response and autoimmunity by inducing expression of the immunoregulatory molecule CD39.
Nat. Immunol.
14
:
1054
1063
.
56
Moon
,
S. J.
,
J. S.
Park
,
Y. J.
Heo
,
C. M.
Kang
,
E. K.
Kim
,
M. A.
Lim
,
J. G.
Ryu
,
S. J.
Park
,
K. S.
Park
,
Y. C.
Sung
, et al
.
2013
.
In vivo action of IL-27: reciprocal regulation of Th17 and Treg cells in collagen-induced arthritis.
Exp. Mol. Med.
45
:
e46
.
57
Stumhofer
,
J. S.
,
J. S.
Silver
,
A.
Laurence
,
P. M.
Porrett
,
T. H.
Harris
,
L. A.
Turka
,
M.
Ernst
,
C. J.
Saris
,
J. J.
O’Shea
,
C. A.
Hunter
.
2007
.
Interleukins 27 and 6 induce STAT3-mediated T cell production of interleukin 10.
Nat. Immunol.
8
:
1363
1371
.
58
Hirahara
,
K.
,
K.
Ghoreschi
,
X. P.
Yang
,
H.
Takahashi
,
A.
Laurence
,
G.
Vahedi
,
G.
Sciumè
,
A. O.
Hall
,
C. D.
Dupont
,
L. M.
Francisco
, et al
.
2012
.
Interleukin-27 priming of T cells controls IL-17 production in trans via induction of the ligand PD-L1.
Immunity
36
:
1017
1030
.
59
Bellucci
,
R.
,
A.
Martin
,
D.
Bommarito
,
K.
Wang
,
S. H.
Hansen
,
G. J.
Freeman
,
J.
Ritz
.
2015
.
Interferon-γ-induced activation of JAK1 and JAK2 suppresses tumor cell susceptibility to NK cells through upregulation of PD-L1 expression.
OncoImmunology
4
:
e1008824
.
60
Ke
,
Y.
,
D.
Sun
,
G.
Jiang
,
H. J.
Kaplan
,
H.
Shao
.
2010
.
PD-L1(hi) retinal pigment epithelium (RPE) cells elicited by inflammatory cytokines induce regulatory activity in uveitogenic T cells.
J. Leukoc. Biol.
88
:
1241
1249
.

The authors have no financial conflicts of interest.

Supplementary data