CD4+CD25+ regulatory T cell (Treg) entry into secondary lymphoid organs and local expansion is critical for their immunosuppressive function. Long-term application of the sphingosine-1 phosphate receptor agonist FTY720 exerts pleiotropic anti-inflammatory effects, whereas short-term FTY720 boosts antiviral immunity. In this study, we provide evidence that FTY720 potently inhibits Treg proliferation in vitro and in vivo without affecting their viability, phenotype, or in vitro immunosuppression. In contrast, adoptively transferred Treg exposed ex vivo to FTY720 lost their protective effects in murine models of acute glomerulonephritis and acute graft-vs-host disease. On a cellular level, FTY720 inhibits IL-2-induced STAT-5 phosphorylation, paralleled by a loss of FoxP3 expression during Treg expansion in vitro. Notably, loss of in vivo immunosuppression is not due to impaired migration to or localization within secondary lymphoid organs. We could even show a selective trapping of adoptively transferred Treg in inflammatory lymph nodes by FTY720. Finally, Treg isolated from animals systemically exposed to FTY720 also exhibit a significantly impaired proliferative response upon restimulation when compared with Treg isolated from solvent-treated animals. In summary, our data suggest that sphingosine-1 phosphate receptor-mediated signals induced by FTY720 abrogate their in vivo immunosuppressive potential by blocking IL-2 induced expansion, which is indispensable for their in vivo immunosuppressive activity.

Naturally occurring regulatory T cells (Treg)4 have emerged as a unique CD4+ T cell subset mainly characterized by coexpression of high CD25 levels together with the Forkhead transcription factor FoxP3 (1, 2, 3). Treg play a decisive role in the maintenance of immunological tolerance (2). Various target cells such as CD4+ and CD8+ T cells (4, 5, 6) and NK cells (7), as well as NKT cells (8) and dendritic cells (9), and different immunosuppressive mechanisms including TGF-β (10) and CTLA-4 (11) have been described to date. The proliferative potential of Treg is well documented and is assumed to be of critical importance for counter-regulation of an ongoing effector T cell activation, as a rapid expansion of the Treg pool tips the numerical balance in favor of a more immunosuppressive milieu (12). Among various others, IL-2 has been proven to play a critical role for Treg survival and proliferation (13, 14). Accordingly, the expansion of Treg rather than that of effector T cells in cancer trials testing IL-2 as an immunostimulant support this concept (15). Finally, data from murine models further corroborate these clinical observations, as IL-2 is indispensable for peripheral maintenance of naturally occurring Treg (16).

In addition, the immunosuppressive potential of Treg also depends on direct cell-to-cell contact with target cells. Thus, their migration to sites of immune responses has to be tightly regulated. Secondary lymphoid organs (SLO) are considered to be an important environment for Treg-mediated immune regulation (17), as lymph nodes (LN) as well as the spleen are critical for orchestrating T cell-mediated immune responses (18). Obviously, in case of an exaggerated immune activation within target organs, Treg also have to enter end organs to induce local immune regulation. In line with this idea, it has recently been demonstrated in an experimental model of acute graft-vs-host disease (GvHD) that LN-homing, CD62L-expressing Treg are superior to CD62L-negative Treg in terms of disease control (19, 20). In contrast, in a model of chronic GvHD that rather reflects a situation of ongoing T cell activation within damaged end organs, it appeared to be mainly the CD103+ Treg population that was inhibiting GvHD (21). These observations fit perfectly the recent description of various Treg subpopulations characterized by a distinct chemokine and cell surface receptor expression pattern, i.e., CCR7- (22), CCR5- (23), CXCR4- (23), CD62L- (20, 24), and CD103-expressing (21) Treg. In addition to the data derived from the GvHD models, we could recently demonstrate the potent nephroprotective effects of adoptively transferred Treg in an accelerated model of anti-glomerular basement membrane (GBM) glomerulonephritis (GN) (25). In this particular model, Treg do not infiltrate the inflamed end organ but primarily migrate to the SLO, which most likely induces inhibition of tissue-destructive effector T cells. Thus, especially in the early phase of inflammatory processes, efficient LN occupancy as well as a high proliferative potential of Treg is required for efficient immunosuppression.

FTY720 (2-amino-2-(2-[4-octylphenyl]ethyl)-1,3-propanediol hydrochloride) is a sphingosine analog and an immunomodulatory agent derived from the Chinese fungus Iscaria sinclarii. FTY720 has been proven to prolong allograft survival in both rodents and men (26, 27) by acting as a sphingosine-1 phosphate (S1P) agonist inducing a prolonged down-regulation of the cell surface expression of the S1P receptor. The latter enables a blockade of S1P-mediated T cell egress from SLO, which is normally induced by a S1P gradient from blood and lymph fluid to tissue (28). Among its various target cell effects, the trapping of lymphocytes in SLO and the subsequent induction of lymphopenia is thought to be the predominant mechanism that prevents effector T cell exit into the blood stream and subsequent entry into target tissues (29). Interestingly, immunosuppressive function after systemic long-term application of FTY720 has at least in part been attributed to the induction of FoxP3 in naive T cells, thereby inducing T cells with a regulatory phenotype (30). However, this is just one among the plethora of mechanisms induced by FTY720 in vivo, as it has also been shown to modify lymphatic endothelial (28), dendritic (31), and B cell function (32).

We provide in this report first evidence that the S1P receptor agonist FTY720 potently inhibits Treg proliferation in vitro and in vivo by inhibiting IL-2 mediated mitogenic signals. Despite the fact that FTY720 induced the trapping of Treg in inflamed SLO, the immunosuppressive effects of FTY720-exposed and subsequently adoptively transferred Treg were abrogated in two murine models of inflammation.

C57BL/6 and BALB/c mice (6–12 wk of age) were purchased from Harlan Laboratories. For bone marrow transplant experiments, luciferase-expressing transgenic C57BL/6 L2G85 mice (33) were used. All experiments used age and sex-matched control mice and were performed in accordance with protocols approved by the Institutional Animal Care and Use Committee of the Innsbruck Medical University, Innsbruck, Austria (35/9185.81/G-07/19, Austrian Federal Ministry for Education, Science and Culture (BMBWK)-66.011/0087-BrGT/2005).

CFSE and TAMRA were purchased from Molecular Probes. FTY720 was a gift from Dr. V. Brinkmann (Novartis Pharma) and SEW2871 was purchased from Biomol. Anti-CD3 was purchased from BD Biosciences. The annexin V detection kit was from R&D Systems and IL-2, IL-7, and IL-15 were from Peprotech.

Spleens and LN were removed and gently dissociated to single cell suspensions. CD4+CD25+ T cells (Treg) were magnetically separated using a Treg separation kit (Milteny Biotec) achieving purity of >95%. The selected cells were incubated for 1 h at 37°C with 0.5 μg/ml FTY720-treated FTY-Treg) or solvent-treated Treg (solv-Treg) followed by two washing steps before further use. In some experiments, Treg were incubated with increasing FTY720 or SEW2871 concentrations as indicated.

Isolated Treg were exposed to increasing concentrations of FTY720 ranging from 0.5 μg/ml to 4 μg/ml, washed, and taken into culture using anti-CD3 together with 100 U/ml IL-2. Induction of cell death was determined by detection of annexin V and propidium iodide using FACS analysis at the indicated time points.

To determine the regulatory properties of Treg, cocultures of CD4+CD25 T cells together with equal numbers of either FTY720- or solv-Treg were performed in anti-CD3-precoated (5 μg/ml) wells. As control, the respective T cell population was cultured alone. Proliferation was measured on day 5 by [3H]thymidine incorporation in a beta scintillator.

CFSE-labeled (0.5 μM) Treg at 1 × 106/ml were cultured with 100 ng/ml plate-bound anti-CD3 together with 100 U/ml IL-2 for 2–14 days. Stimulation with anti-CD3 plus IL-2 results in cell division with distinct CFSE fluorescence peaks that allows for discrimination between cycled (at least one division) and noncycled cells by FACS analysis.

All FACS Abs (except the anti-FoxP3 mAb) were purchased from BD Biosciences. Phosphospecific flow cytometry was performed strictly according to the manufacturer’s manual (protocol 3; BD Biosciences) after stimulation using plate bound anti-CD3 (5 μg/ml; BD Biosciences) together with one of the following common γ-chain cytokines: IL-2 (100 U/ml), IL-7 (10 ng/ml), or IL-15 (25 ng/ml). S1P specificity of FTY720-mediated effects on STAT-5 phosphorylation was tested by using SEW2871 at concentrations ranging from 100 to 1000 nM instead of FTY720. Flow cytometric analyses were performed on a FACSCalibur (BD Biosciences) and data were analyzed with Cell Quest software (BD Biosciences). FoxP3 was stained by APC-labeled anti-mouse FoxP3 mAb (eBioscience).

Treg (5 × 106) were lysed on ice and lysates were analyzed by electrophoresis on 10% SDS-polyacrylamide gels after being blotted on hydrophobic polyvinylidene difluoride membrane (GE Healthcare). Mouse anti-phosphorylated STAT-5 Ab (clone 8-5-2, dilution 1/1000; Millipore) was added and binding was subsequently detected by HRP-conjugated goat anti-mouse IgG (dilution 1:2000; DakoCytomation). Chemiluminescent reaction was induced using a Lumigen TMA-6 kit (GE Healthcare) and the blots were analyzed by using a ChemiDoc XRS+ molecular imaging system (Bio-Rad). To evaluate expression of total STAT-5 protein, the blots were incubated with 1× mild Ab stripping solution (Chemicon) and with mouse anti-STAT-5 Ab (clone 89/STAT-5, dilution 1/400; BD Biosciences).

To detect IL-2 and IFN-γ in supernatants, FTY720 or solvent-exposed T cells were restimulated with PMA/ionomycin. Cytokines were detected by ELISA (BD Biosciences).

FTY- and solv-Treg suspensions were differentially labeled with CFSE (green) or TAMRA (red) dyes for adoptive transfer strictly according to the manufacturer’s recommendations and subsequently injected i.v. (5 × 106 of each Treg population) to follow their in vivo migration behavior under steady state or inflammatory conditions. The latter were induced by s.c. injection of 0.02 g of nonviable desiccated Mycobacterium tuberculosis H37a (Difco Laboratories) dissolved in IFA (Sigma-Aldrich) into the footpad. Seven days after the induction of inflammation, differentially labeled FTY- and solv-Tregs were adoptively transferred. Peripheral blood, spleen, draining (popliteal), and nondraining (popliteal, contralateral to footpad injection) LN were harvested 4–48 h after Treg transfer and subsequently prepared either as single cell suspension (for FACS) or snap frozen (for preparation of tissue sections). Images were taken with an upright Leica MZ 16 FA stereomicroscope equipped with a spot camera and operated via MetaMorph software (system implemented by Visitron Systems). Original magnification was ×10.

Acute GvHD (aGvHD) was induced as described previously (34). In brief, recipients were exposed to lethal irradiation with 850 cGy on day 0 followed by i.v. injection of 5 × 106 T cell-depleted bone marrow cells after 1 day. To induce aGvHD, 1 × 106 CD4+/CD8+ T cells in the C57BL/6→BALB/c model, respectively, were injected i.v. on day 2. Treg were derived from donors of the same genetic background as those of conventional T cells (Tconv) and injected i.v. on day 0 at the same dosage as the Tconv. Slides of small bowel and large bowel samples collected on day 7 were stained with H&E and scored by a blinded and experienced pathologist (U.V.G.) according to a previously published histopathology scoring system.

BLI was performed as previously described (33). Briefly, mice were injected i.p. with luciferin (10 μg/g body weight). Ten minutes later, mice were imaged using an IVIS200 charge-coupled device imaging system (Xenogen) for 5 min. Imaging data were analyzed and quantified with Living Image software (Xenogen) and IgorProCarbon (WaveMetrics).

An accelerated model of nephrotoxic nephritis was induced as previously described (35). In brief, mice were preimmunized s.c. with 2 mg/ml rabbit IgG (Jackson ImmunoResearch Laboratories) dissolved in IFA and nonviable desiccated Mycobacterium tuberculosis H37a (Difco Laboratories). Two days later, animals received either 3 × 106 solv-Treg or equal numbers of FTY-Treg i.v. As control, mice received 3 × 106 CD4+CD25 T cells. After 3 days, 5 mg of heat-inactivated rabbit anti-mouse GBM antiserum per 20 g of body weight was injected. Twenty-four-hour urine samples were collected in metabolic cages on days −1, 1, 7, and 14 after the induction of GN. Urinary albumin was determined by a double-sandwich ELISA (Abcam), and urinary creatinine was quantified spectrophotometrically using a commercially available kit (Sigma-Aldrich).

Formalin-fixed tissue was embedded in paraffin, cut in 4-μm sections, and stained with periodic acid Schiff (PAS) for histologic analysis. Sections cut from frozen tissue (4–25 μm) were used for LN migration studies as well as for immunoperoxidase staining of macrophages and T cells in the kidney (25) using a rat anti-mouse macrophage Ab (Clone F4/80; Serotec), a rat anti-mouse CD4 mAb (clone YTS191.1; Serotec), and an IgG2a isotype Ab as negative control. Biotin-conjugated goat anti-rat IgG Ab (Jackson ImmunoResearch Laboratories) was used as a secondary Ab, followed by incubation with an avidin-biotin complex and subsequent development and HE counterstaining. T cells were quantified by counting the number of cells in six adjacent high-power fields of the renal cortex and medulla. A semiquantitative scoring system was performed for the quantification of macrophages: 0, 0 to 4 cells stained positive; 1+, 5 to 10 cells; 2+, 10 to 50 cells; 3+, 50–200 cells; and 4+, >200 cells stained positive per low-power field.

FTY720 was administered i.p. in a dose of 1 mg/kg body weight on two consecutive days, whereas control mice received solvent. Mice were sacrificed 36 h after the first i.p. injection for isolation of Treg and CD4+CD25 control cells from spleen and LN suspensions. The isolated cells were cultured on plates precoated with 100 ng/ml anti-CD3 together with 100 U/ml IL-2 for 5 days and proliferation was determined by [3H]thymidine incorporation.

Results represent data from at least three independent biological experiments. After ANOVA, Student’s t test was used. Values of p < 0.05 were considered significant. Figures show means ± SEM. Statistical analysis was performed using GraphPad Prism.

Only little information is available regarding the direct effects of FTY720 on isolated Treg. Thus, we analyzed the influence of the drug on characteristic Treg properties. First, we tested the effects of FTY720 on Treg viability. Using increasing doses (0.5–4 μg/ml) of FTY720, we observed an increased rate of cell death only after 72 h at the highest dose level of 4 μg/ml, which is known to be toxic for various cell types (Fig. 1). We therefore used the concentration of 0.5 μg/ml for our subsequent in vitro and in vivo experiments. We next tested whether the characteristic properties of Treg function are modulated by short-term FTY720 exposure. The suppressive potential of FTY-Treg, as measured by standard in vitro suppression assays, remained unaltered as compared with solv-Treg, even when FTY-Treg were mixed with CD4+CD25 responder T cells in different ratios ranging from 1:1 to 1:10 (Fig. 2,A; ∗, p < 0.05 for FTY- and solv-Treg vs responder cells alone). Moreover, FTY-Treg suppressed the production of IFN-γ and IL-2 by CD4+CD25 T cells as effectively as their solvent-treated counterparts (Fig. 2,B; ∗, p < 0.05). Finally, FTY-Treg showed a comparable cell surface marker expression (CD62L, CD103, CCR7) as well as intracellular FoxP3 levels after short-term FTY720 exposure remained unaltered (Fig. 2 C). Thus, typical functional and phenotypical hallmarks of bona fide Treg are preserved upon short-term exposure to FTY720 in vitro.

FIGURE 1.

Effects of FTY720 on Treg viability. Isolated Treg were exposed to increasing concentrations of FTY720 as indicated and subsequently taken into culture using anti-CD3 together with 100 U/ml IL-2. Cell death was determined by annexin/propidium iodide and FACS analysis at the indicated time points. Percentages of positive cells are given in the quadrants. A representative experiment is shown (n = 3).

FIGURE 1.

Effects of FTY720 on Treg viability. Isolated Treg were exposed to increasing concentrations of FTY720 as indicated and subsequently taken into culture using anti-CD3 together with 100 U/ml IL-2. Cell death was determined by annexin/propidium iodide and FACS analysis at the indicated time points. Percentages of positive cells are given in the quadrants. A representative experiment is shown (n = 3).

Close modal
FIGURE 2.

Treg phenotype and in vitro function is not affected by FTY720. A, FTY- and solv-Treg were mixed with CD4+CD25 responder T cells in different ratios ranging from 1:1 to 1:10. Proliferation was determined in triplicate by [3H]thymidine incorporation and compared with the proliferation of pure populations of CD4+CD25 control T cells (given as 100%) (∗/∗∗, p < 0.05; n = 10 per group). B, FTY- or solv-Treg were seeded in 24-well plates alone or mixed in a 1:1 ratio with CD4+CD25 T cells in a final concentration of 5 × 106 cells/ml and stimulated with PMA and ionomycin for 16 h. IFN-γ and IL-2 were determined in triplicate in supernatants by ELISA and compared with the cytokine production of pure populations of CD4+CD25 control T cells (∗/∗∗, p < 0.05; n = 6). C, CD103, CD62L, and CCR7 surface stainings and intracellular FoxP3 stainings of either FTY- or solv-Treg (a representative example from five independent experiments is shown).

FIGURE 2.

Treg phenotype and in vitro function is not affected by FTY720. A, FTY- and solv-Treg were mixed with CD4+CD25 responder T cells in different ratios ranging from 1:1 to 1:10. Proliferation was determined in triplicate by [3H]thymidine incorporation and compared with the proliferation of pure populations of CD4+CD25 control T cells (given as 100%) (∗/∗∗, p < 0.05; n = 10 per group). B, FTY- or solv-Treg were seeded in 24-well plates alone or mixed in a 1:1 ratio with CD4+CD25 T cells in a final concentration of 5 × 106 cells/ml and stimulated with PMA and ionomycin for 16 h. IFN-γ and IL-2 were determined in triplicate in supernatants by ELISA and compared with the cytokine production of pure populations of CD4+CD25 control T cells (∗/∗∗, p < 0.05; n = 6). C, CD103, CD62L, and CCR7 surface stainings and intracellular FoxP3 stainings of either FTY- or solv-Treg (a representative example from five independent experiments is shown).

Close modal

The critical role of IL-2 for Treg proliferation is well documented (36). Thus, we next focused on in vitro Treg expandability upon stimulation via the TCR together with IL-2. In contrast to solv-Treg, which can be readily expanded, FTY-Treg exhibit a markedly impaired proliferation in vitro (Fig. 3,A; ∗, p < 0.05). These data were corroborated in vivo by measuring CFSE dilution upon adoptive transfer of either CFSE-labeled FTY- or solv-Treg into mice suffering from footpad inflammation. In this model, FTY-Treg also showed a severely reduced expansion in the draining LN when compared with adoptively transferred solv-Treg. As depicted in Fig. 3 B, only 24% of transferred FTY720-treated, CFSE-positive Tregs divided 48 h after i.v. injection in vivo (∗, p < 0.05), whereas 70% of solv-Treg divided at least once during this time period. This effect was solely detectable in draining LN but not in the spleen, where Treg did not proliferate within 48 h.

FIGURE 3.

FTY-Treg show severe proliferation defects in vitro and in vivo. A, In vitro expansion by TCR stimulation with anti-CD3 together with IL-2 is impaired by FTY720 as shown by CFSE dilution of solv- or FTY-Treg at the indicated time points (n = 7 per group; ∗, p < 0.05 vs solv-Treg). B, Forty-eight hours after adoptive transfer of either CFSE-labeled FTY-Treg or CFSE-labeled solv-Treg into mice suffering from a footpad inflammation, CFSE dilution of transferred Treg in SLO was determined by flow cytometry (n = 8 per group; ∗, p < 0.05 vs solv-Treg).

FIGURE 3.

FTY-Treg show severe proliferation defects in vitro and in vivo. A, In vitro expansion by TCR stimulation with anti-CD3 together with IL-2 is impaired by FTY720 as shown by CFSE dilution of solv- or FTY-Treg at the indicated time points (n = 7 per group; ∗, p < 0.05 vs solv-Treg). B, Forty-eight hours after adoptive transfer of either CFSE-labeled FTY-Treg or CFSE-labeled solv-Treg into mice suffering from a footpad inflammation, CFSE dilution of transferred Treg in SLO was determined by flow cytometry (n = 8 per group; ∗, p < 0.05 vs solv-Treg).

Close modal

On the molecular level, we found that FTY720 pre-exposure of Treg dose-dependently impaired IL-2-induced phosphorylation of STAT-5, which is a critical component of the IL-2 signal transduction pathway (Fig. 4, A and B; ∗, p < 0.05). In contrast, solvent pre-exposure did not alter IL-2-induced phosphorylation of STAT-5. Reduced sensitivity to IL-2-induced signals is not mediated by down-regulation of the IL-2 receptor α-chain upon FTY720 exposure (Fig. 4,C). However, in line with the reduced activation of Treg by IL-2 after FTY720 exposure (which is mirrored by inhibition of proliferation), further up-regulation of the mean fluorescence intensity of CD25 throughout the culture period (at 72 and 96h after culture initiation) is prevented by FTY720 (the percentage of CD25-expressing cells remains unchanged in both fractions; data not shown). It is well documented that the IL-2/STAT-5 pathway plays a decisive role for FoxP3 maintenance in Treg (37). Thus, it is not surprising that the maintenance of FoxP3 expression during in vitro culture of initially FTY720-exposed Treg is affected (Fig. 4 D).

FIGURE 4.

FTY720 blocks IL-2 signaling in Treg. A and B, IL-2-induced STAT-5 phosphorylation (p-STAT-5) of Treg exposed to increasing doses of FTY720 was determined at the indicated time points by phosphospecific flow cytometry (n = 8 per group; ∗, p < 0.05 vs solv-Treg) (A) and Western blotting (B). C, CD25 expression upon either FTY720 or solvent exposure of isolated Treg. Data are presented as mean fluorescence intensity throughout a culture period of 96 h under growth-supporting conditions with anti-CD3 and IL-2 (n = 3 per group; ∗, p < 0.05 vs solv-Treg). D, IL-2 induced maintenance of FoxP3 expression of in vitro cultured Treg was determined in FTY- vs solv-Treg at the indicated time points (n = 4 per group; ∗, p < 0.05).

FIGURE 4.

FTY720 blocks IL-2 signaling in Treg. A and B, IL-2-induced STAT-5 phosphorylation (p-STAT-5) of Treg exposed to increasing doses of FTY720 was determined at the indicated time points by phosphospecific flow cytometry (n = 8 per group; ∗, p < 0.05 vs solv-Treg) (A) and Western blotting (B). C, CD25 expression upon either FTY720 or solvent exposure of isolated Treg. Data are presented as mean fluorescence intensity throughout a culture period of 96 h under growth-supporting conditions with anti-CD3 and IL-2 (n = 3 per group; ∗, p < 0.05 vs solv-Treg). D, IL-2 induced maintenance of FoxP3 expression of in vitro cultured Treg was determined in FTY- vs solv-Treg at the indicated time points (n = 4 per group; ∗, p < 0.05).

Close modal

S1P receptor 1 specificity of the FTY720-mediated effects was proven by using increasing doses of the S1P receptor 1 specific agonist SEW2871 instead of FTY720. In line with our previous data, SEW2871 also dose dependently inhibited IL-2 induced STAT-5 phosphorylation (Fig. 5,A). Of note, phosphorylation of STAT-5 induced by other cytokines of the common γ-chain family (i.e., IL-7 and IL-15) was not prevented by FTY720 (Fig. 5 B).

FIGURE 5.

Inhibition of STAT-5 phosphorylation is specific for S1P1 and IL-2. A, IL-2-induced STAT-5 phosphorylation (pStat5) of Treg exposed to increasing doses of the selective S1P receptor 1 agonist SEW2871 was determined at the indicated time points by phosphospecific flow cytometry (n = 3 per group; ∗, p < 0.05 vs solv-Treg). B, STAT-5 phosphorylation of FTY720 pre-exposed Treg subsequently stimulated with the common γ-chain cytokines IL-2, IL-7 and IL-15 (n = 3 per group; ∗, p < 0.05 vs solv-Treg).

FIGURE 5.

Inhibition of STAT-5 phosphorylation is specific for S1P1 and IL-2. A, IL-2-induced STAT-5 phosphorylation (pStat5) of Treg exposed to increasing doses of the selective S1P receptor 1 agonist SEW2871 was determined at the indicated time points by phosphospecific flow cytometry (n = 3 per group; ∗, p < 0.05 vs solv-Treg). B, STAT-5 phosphorylation of FTY720 pre-exposed Treg subsequently stimulated with the common γ-chain cytokines IL-2, IL-7 and IL-15 (n = 3 per group; ∗, p < 0.05 vs solv-Treg).

Close modal

We subsequently tested the consequence of impaired Treg proliferation induced by the S1P receptor agonist FTY720 in adoptive Treg transfer models. First, we used an established model for aGvHD as an alloimmune model in which Treg are capable of reducing lethality (34). In vivo expansion of aGvHD-inducing T cells was monitored by detecting photons via bioluminescence imaging, as the donor effector T cells carry the luciferase transgene. The extent of the emitted light is known to correlate with T cell expansion and aGvHD severity. In line with previous data, cotransfer of solv-Treg almost completely prevented the expansion of luciferase transgenic, allogeneic Tconv (Fig. 6, A and B). In contrast, short-term exposure of Treg to FTY720 before injection markedly reduced their suppressive cues, which is reflected by a massive expansion of luciferase-positive effector T cells comparable to that seen in Tconv-transferred animals (Fig. 6, A and B). This reduced suppressor function also resulted in significantly reduced overall survival of the group that received Tconv along with FTY-Treg compared with the group receiving Tconv together with solv-Treg (Fig. 6,C; ∗, p = 0.001). Histopathology scoring revealed a significantly higher GvHD score in the group that received Tconv together with FTY-Treg as compared with the group receiving Tconv plus solv-Treg (Fig. 6 D; ∗, p < 0.05). Overall, these results demonstrate that pre-exposure of Treg to FTY720 impairs Treg function in vivo.

FIGURE 6.

FTY720 treatment severely impairs Treg suppressor function in a model of acute GvHD. A, Representative BALB/c recipients from the indicated groups (Tconv, CD4/CD8 conventional T cells given on day 2 after bone marrow transplantation (BMT; n = 10); solv-Treg (n = 15); and FTY-Treg (n = 15)) from different time points (days 12, 14, and 16) after BMT are shown. B, Expansion of luciferase transgenic alloreactive T cells as quantified in photons per second per mouse over time. Pooled data from two independent experiments are shown. C, Survival of BALB/c after lethal irradiation with 8.5 Gy and BMT. Data are pooled from three independent experiments (∗, p < 0.05 solv-Treg vs FTY-Treg). D, Seven days after transplantation, mice from the indicated groups were sacrificed and sections of small bowel and large bowel were stained with H & E. Slides were analyzed by a pathologist blinded to the treatment groups for evidence of pathologic damage (∗, p < 0.05 vs solv-Treg). BM, Bone marrow; ctrl, control; Tc, conventional T cell.

FIGURE 6.

FTY720 treatment severely impairs Treg suppressor function in a model of acute GvHD. A, Representative BALB/c recipients from the indicated groups (Tconv, CD4/CD8 conventional T cells given on day 2 after bone marrow transplantation (BMT; n = 10); solv-Treg (n = 15); and FTY-Treg (n = 15)) from different time points (days 12, 14, and 16) after BMT are shown. B, Expansion of luciferase transgenic alloreactive T cells as quantified in photons per second per mouse over time. Pooled data from two independent experiments are shown. C, Survival of BALB/c after lethal irradiation with 8.5 Gy and BMT. Data are pooled from three independent experiments (∗, p < 0.05 solv-Treg vs FTY-Treg). D, Seven days after transplantation, mice from the indicated groups were sacrificed and sections of small bowel and large bowel were stained with H & E. Slides were analyzed by a pathologist blinded to the treatment groups for evidence of pathologic damage (∗, p < 0.05 vs solv-Treg). BM, Bone marrow; ctrl, control; Tc, conventional T cell.

Close modal

Second, anti-GBM nephritis was selected as an autoimmune model to test FTY-Treg in vivo. According to our previous data (25), transfer of solv-Treg into mice suffering from anti-GBM GN potently inhibited the development of renal inflammation as reflected by significantly reduced proteinuria (Fig. 7,A; ∗, p < 0.05). Histological evaluation revealed only occasional PAS-positive deposits, and the majority of the glomeruli displayed no pathological changes (Fig. 7,C). According to the data from the aGvHD model, ex vivo FTY720 treatment of Treg also abrogated their suppressive effect on nephritic inflammation as demonstrated by significantly elevated albumin/creatinine urine levels when compared with animals receiving solv-Treg (Fig. 7,A). Kidneys displayed classical histological signs of GN such as hypercellularity and focal deposition of PAS-positive material (Fig. 7,D), as well as enhanced infiltration of CD4+ T cells and macrophages (Fig. 7,B; ∗, p < 0.05), which was comparable to mice receiving CD4+CD25 control T cells (Fig. 7 E).

FIGURE 7.

FTY720 treatment abrogates Treg suppressor function in a model of anti-GBM nephritis. To test the immunosuppressive potential of FTY-Treg we adoptively transferred either 3 × 106 FTY-Treg or the same number of solv-Treg in a model of anti-GBM nephritis. A, Treg suppressor function was determined by quantification of proteinuria by measurement of the albumin/creatinine ratio in the urine (n = 9 per group; ∗, p < 0.05 vs solv-Treg). B, The infiltration of inflammatory cells was analyzed by immunohistochemistry. Staining for CD4+ T cells (left panel) and macrophages (right panel) was performed 14 days after induction of anti-GBM nephritis in mice injected with solv-Treg (open bars), FTY-Treg (filled bars), or CD4+CD25 control cells (gray bars; n = 9 per group). Interstitial macrophage (MΦ) and CD4+ accumulation was significantly increased in FTY-Treg-injected mice when compared with solv-Treg treated mice (∗, p < 0.05). HPF, High power field. C, Representative PAS stainings from kidney sections (n = 9 per group) showed that in mice receiving solv-Treg the majority of the glomeruli displayed no pathological changes. D, Ex vivo FTY720 treatment of Treg abrogated their suppressive effect on nephritic inflammation as demonstrated by focal deposition of PAS-positive material (arrowheads), which was comparable to that in mice receiving CD4+CD25 control T cells (E).

FIGURE 7.

FTY720 treatment abrogates Treg suppressor function in a model of anti-GBM nephritis. To test the immunosuppressive potential of FTY-Treg we adoptively transferred either 3 × 106 FTY-Treg or the same number of solv-Treg in a model of anti-GBM nephritis. A, Treg suppressor function was determined by quantification of proteinuria by measurement of the albumin/creatinine ratio in the urine (n = 9 per group; ∗, p < 0.05 vs solv-Treg). B, The infiltration of inflammatory cells was analyzed by immunohistochemistry. Staining for CD4+ T cells (left panel) and macrophages (right panel) was performed 14 days after induction of anti-GBM nephritis in mice injected with solv-Treg (open bars), FTY-Treg (filled bars), or CD4+CD25 control cells (gray bars; n = 9 per group). Interstitial macrophage (MΦ) and CD4+ accumulation was significantly increased in FTY-Treg-injected mice when compared with solv-Treg treated mice (∗, p < 0.05). HPF, High power field. C, Representative PAS stainings from kidney sections (n = 9 per group) showed that in mice receiving solv-Treg the majority of the glomeruli displayed no pathological changes. D, Ex vivo FTY720 treatment of Treg abrogated their suppressive effect on nephritic inflammation as demonstrated by focal deposition of PAS-positive material (arrowheads), which was comparable to that in mice receiving CD4+CD25 control T cells (E).

Close modal

To exclude the possibility that additional mechanisms contribute to the observed loss of Treg-mediated immunosuppression in vivo, we next focused on FTY720 effects on Treg migration upon adoptive transfer. Using steady state conditions as well as a well defined model of rapid local inflammation (i.e., footpad injection of mycobacteria together with Freund’s adjuvant), we i.v. coinjected TAMRA-labeled FTY-Treg together with equal numbers of CFSE-labeled solv-Treg (Fig. 8,A). Under steady state conditions, 6–12 h after transfer increased numbers of FTY-Treg were found in the spleen, but not in peripheral LN (Fig. 8,B). Notably, despite their loss of immunosuppression in vivo in the inflammatory models, inflamed LN occupancy of Treg is even increased by ex vivo exposure of Treg to FTY720 (Fig. 8 C; ∗, p < 0.05). No significant increase of FTY-Treg was detectable in the contralateral nondraining LN under inflammatory conditions.

FIGURE 8.

A, FTY720 induces the trapping of adoptively transferred Treg into inflamed LN. B and C, TAMRA-labeled FTY-Treg were coinjected i.v. together with equal numbers of CFSE-labeled solv-Treg into mice under either steady state (B) or inflammatory conditions (C). Six to 12 hours after adoptive transfer, labeled Treg were detected by flow cytometry of spleen, draining popliteal, and nondraining contralateral LN. The numbers of the CFSE-labeled solv-Treg that migrated to the spleen were set as 100%. Data of three independent experiments are shown (n = 9 animals per group; ∗, p < 0.05 vs solv-Treg). D, To identify the ability of FTY-Treg to properly locate to T cell areas in SLO, equal numbers (3 × 106) of differentially labeled FTY-Treg (red, TAMRA) and solv-Treg (green, CFSE) were coinjected i.v. into mice under inflammatory conditions. Six to 12 hours after adoptive transfer, organs were cryopreserved. Labeled Treg were detected by fluorescence microscopy (original magnification, ×10). Representative examples of the spleen (left panel) and LN (right panel) are shown (n = 4 per group).

FIGURE 8.

A, FTY720 induces the trapping of adoptively transferred Treg into inflamed LN. B and C, TAMRA-labeled FTY-Treg were coinjected i.v. together with equal numbers of CFSE-labeled solv-Treg into mice under either steady state (B) or inflammatory conditions (C). Six to 12 hours after adoptive transfer, labeled Treg were detected by flow cytometry of spleen, draining popliteal, and nondraining contralateral LN. The numbers of the CFSE-labeled solv-Treg that migrated to the spleen were set as 100%. Data of three independent experiments are shown (n = 9 animals per group; ∗, p < 0.05 vs solv-Treg). D, To identify the ability of FTY-Treg to properly locate to T cell areas in SLO, equal numbers (3 × 106) of differentially labeled FTY-Treg (red, TAMRA) and solv-Treg (green, CFSE) were coinjected i.v. into mice under inflammatory conditions. Six to 12 hours after adoptive transfer, organs were cryopreserved. Labeled Treg were detected by fluorescence microscopy (original magnification, ×10). Representative examples of the spleen (left panel) and LN (right panel) are shown (n = 4 per group).

Close modal

Next, we analyzed the positioning of FTY720-exposed Treg in T cell areas of SLO upon adoptive transfer, because an altered localization might also interfere with proper Treg function in vivo. Therefore, we again coinjected differentially labeled FTY720- and solvent-exposed Treg i.v. into mice under steady state as well as inflammatory conditions. As shown in Fig. 8 D, TAMRA labeled FTY-Treg (red fluorescence) perfectly colocalized with CFSE-labeled solv-Treg (green fluorescence) in T cell areas of the spleen and LN after i.v. transfer into mice with local footpad inflammation. This finding was corroborated in healthy mice showing similar positioning patterns of FTY- and solv-Treg in SLO under steady state conditions (data not shown).

Finally, we were interested in determining whether short-term (i.e., 36 h) systemic FTY720 administration also affects Treg proliferative potential. In accordance with the defective proliferation of isolated Treg exposed to FTY720 in vitro, restimulation of Treg isolated from either solvent- or FTY720-exposed animals using anti-CD3 together with IL-2 resulted in a ∼40% reduced proliferation of Treg isolated from FTY720-treated animals (Fig. 9).

FIGURE 9.

Short-term systemic FTY720 administration impairs Treg proliferation. FTY720 was injected i.p. at 1 mg/kg body weight on days 0 and 1. Control animals received solvent injections. Thirty-six hours after the first FTY720 injection, Treg and CD4+CD25 control cells were isolated and restimulated by anti-CD3 together with 100 U/ml IL-2. Proliferation was determined by [3H]thymidine incorporation. Proliferation counts of cell populations isolated from solvent-treated animals were set as 100% (n = 12 per group; ∗, < 0.05).

FIGURE 9.

Short-term systemic FTY720 administration impairs Treg proliferation. FTY720 was injected i.p. at 1 mg/kg body weight on days 0 and 1. Control animals received solvent injections. Thirty-six hours after the first FTY720 injection, Treg and CD4+CD25 control cells were isolated and restimulated by anti-CD3 together with 100 U/ml IL-2. Proliferation was determined by [3H]thymidine incorporation. Proliferation counts of cell populations isolated from solvent-treated animals were set as 100% (n = 12 per group; ∗, < 0.05).

Close modal

The sphingosine analog FTY720 is a promising immunomodulatory agent with proven therapeutic efficacy in various experimental models of autoimmunity (26, 27). The drug is currently tested as an immunosuppressive agent in various clinical trials and will soon be approved for treatment of relapsing multiple sclerosis (38). Immunosuppressive action of FTY720 is suggested to be primarily mediated via the trapping of lymphocytes in SLO, thereby preventing effector T cell exit into the blood stream and subsequent target tissue entry (29). Interestingly, immunosuppressive functions after continuous systemic application of FTY720 or exposure of the whole PBMC fraction to the drug have at least in part been attributed to the induction of Treg (30, 39, 40). However, the fact that FTY720 induces a plethora of mechanisms in various target cells (i.e., lymphatic endothelial, dendritic, T, and B cells) (31, 32) limits the interpretation of direct FTY720-induced effects on Treg. The latter have been shown to play a decisive role in the maintenance of tolerance and have been considered as cellular immunosuppressants for the treatment of exaggerated autoimmune and alloimmune responses (41, 42, 43).

We provide in this report for the first time evidence that the S1P receptor agonist FTY720 is a potent inhibitor of Treg proliferation in vitro and in vivo. Treg expand very efficiently when stimulated appropriately in vitro via the TCR together with IL-2 (14). Accordingly, clinical studies using IL-2 as immunostimulant described a preferential expansion of Treg (15), which can be explained by a low IL-2 receptor signaling threshold that supports Treg development and maintenance (44). Pre-exposure of isolated Treg to FTY720 in vitro almost completely prevented Treg proliferation induced by TCR stimulation and IL-2. Previous reports, which have already demonstrated that S1P inhibits the proliferative response of CD4+ T cells (45, 46, 47), support the concept of a T cell inhibitory effect of S1P receptor agonists in addition to their well characterized effects on lymphocyte migration (29). In line with our in vitro observation, we could further show that FTY720 exposure also profoundly impairs Treg expansion in vivo after adoptive transfer under inflammatory conditions (i.e., a model of local LN inflammation). Recent reports have already defined the critical role of IL-2 for Treg maintenance and expansion (44, 48, 49). On the molecular level, FTY720, as well as the S1P receptor 1-specific agonist SEW2871, severely impairs critical components of the IL-2 signaling pathway in Treg (50), as phosphorylation of STAT-5 and the subsequent IL-2-induced maintenance of FoxP3 expression are inhibited. Interestingly, this inhibitory effect appears to be specific for IL-2, as we were not able to detect any inhibitory effect of the compound on STAT-5 phosphorylation induced by other family members of the common γ-chain cytokines, such as IL-7 and IL-15. Inhibition of FoxP3 maintenance during the culture period is most likely due to the fact that STAT-5 binding sites are located in the promoter region of FoxP3 (37, 50), linking IL-2 signaling to the maintenance of FoxP3 expression. It is of particular importance that FTY720 does not simply reduce IL-2R expression on Treg, suggesting that the modulation of IL-2 signals is indeed mediated by the interference of FTY720 with the intracellular IL-2 signaling pathway. Moreover, exposure of Treg to the FTY720 doses we used for our in vitro and in vivo experiments did not affect their in vitro suppressive function (including anergy, suppression of target T cell proliferation, and cytokine production) and did not affect Treg survival in vitro. However, using two different experimental models of inflammation (i.e., aGvHD as an alloimmune model and anti-GBM GN as an autoimmune model), both of which can be inhibited by adoptive Treg transfer (25, 33, 51), we could clearly demonstrate that FTY-Treg loose their immunosuppressive potential in both models in vivo. This observation was particularly surprising to us, because systemic FTY720 was shown to be additive with the application of in vitro expanded Treg for GVHD inhibition (52), and we also provide evidence that FTY720 induces the efficient trapping of adoptively transferred FTY-Treg in SLO. The latter observation actually suggests that the increased LN occupancy of Treg (especially under inflammatory conditions) would tip the balance in favor of a more immunosuppressive milieu. Using a model of localized footpad inflammation, we show that the homing of FTY-Treg occurred preferentially in inflammatory LN and spleen. These data regarding the effects of FTY720 on Treg migration complement a recent report demonstrating that systemic application of FTY720 primarily affects non-Treg T cell populations (39). The authors described an increased number of Treg in blood and spleens but not in LN after systemic FTY720 application (39). In contrast, we did choose an experimental setup that allowed the evaluation of direct FTY720-induced effects on Treg, which revealed that FTY720 obviously modifies Treg migration in vivo. Due to the fact that a blockade of S1P receptors by FTY720 has been shown to destabilize LN architecture with the displacement of marginal zone B cells (32, 53), we next checked for the localization of FTY-Treg in SLO. However, cotransfer studies of differentially labeled FTY720- and solvent-treated Treg proved the proper occupancy of T cell areas in SLO. These findings are of particular interest, because despite the retention of Treg in inflammatory SLO, their in vivo immunosuppressive activity was lost. We know from the GN model that adoptively transferred Treg primarily enter SLO but not the end organ (25). In addition, it is the LN-homing, CD62L-expressing Treg fraction that is most potent in terms of aGvHD inhibition (19, 20), supporting the concept that Treg LN occupancy is critical for the immunosuppressive function of Treg (17). Thus, it is most likely the FTY720-induced inhibition of Treg proliferation that is responsible for the loss of Treg effects in vivo. Finally, we could further corroborate our previous findings by showing that FTY720 also exerts Treg inhibitory effects when applied in a systemic fashion. In line with our in vitro data, Treg isolated from animals exposed to FTY720 for a short term are hyporesponsive to IL-2 plus TCR-induced proliferation. Our data are in line with a very recent report showing that short-term systemic FTY720 treatment enhanced virus-specific CD4+ and CD8+ T cell responses in a chronic lymphocytic choriomeningitis virus infection model (54). This observation was primarily explained by improved T cell priming within SLO induced by transiently increased LN occupancy of T cells by FTY720, but the effect on Treg function in vivo was not addressed in this report. Our data suggest that concomitant inhibition of immunosuppressive Treg might also account for the observed phenomenon. This observation is supported by a very recent report presented by Liu and colleagues (55) demonstrating that S1P delivers via S1P receptor 1 an intrinsic negative signal activating the Akt/mammalian target of rapamycin pathway leading to inhibition of the thymic generation, peripheral maintenance, and suppressive activity of Treg.

In summary, we show for the first time that the S1P receptor agonist FTY720 induces Treg trapping in inflammatory SLO but concomitantly abrogates their in vivo immunosuppressive potential by inhibition of Treg expansion, which is due to the inhibition of IL-2 induced STAT-5 activation. Our data highlight the need of proper Treg proliferation after adoptive Treg transfer to induce an efficient immunosuppressive impact on effector T cells (i.e., achievement of an optimal Treg to autoreactive or alloreactive T cell ratio) and suggest that the S1P receptor agonism induced by pharmacological agents might represent a rational approach for Treg inhibition. Finally, our data suggest that the inhibition of Treg by S1P receptor agonists might represent an innovative adjuvant for cancer immunotherapy (e.g., in combination with dendritic cell vaccines).

FTY720 was generously provided by Dr. Volker Brinkmann of Novartis. We are indebted to Prof. Gottfried Baier for critically reading the manuscript and for helpful comments.

D.W. receives speaker honoraria from Novartis and is a Novartis Advisory Board member.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1

The project was supported by the Tiroler Landeskrankenanstalten and the Medizinischer Forschungsfonds Tirol. M.S. was supported by the Peter Hans Hofschneider Foundation for Experimental Biomedicine.

A.M.W. designed the study, performed research, analyzed data, and wrote the manuscript. K.H. and R.Z. performed research and analyzed data. C.D., L.M., and M.S. performed research. A.R.R. and G.G. provided infrastructure. U.V.G. conducted blind histopathology scoring. D.W. designed the study, analyzed data, and wrote the manuscript.

2

Parts of this project have been presented at the World Immune Regulation Meeting II in Davos, Switzerland, March 16–20, 2008, and at the American Society of Hematology 50th Annual Meeting and Exposition in San Francisco, CA, December 6–9, 2008.

4

Abbreviations used in this paper: Treg, regulatory T cell; GBM, glomerular basement membrane; GN, glomerulonephritis; GvHD, graft-versus-host disease; aGvHD, acute GvHD; FTY-Treg, FTY720-treated Treg; LN, lymph node; PAS, periodic acid Schiff; SLO, secondary lymphoid organ; solv-Treg, solvent-treated Treg; S1P, sphingosine-1 phosphate; Tconv, conventional T cell.

1
Miyara, M., S. Sakaguchi.
2007
. Natural regulatory T cells: mechanisms of suppression.
Trends Mol. Med.
13
:
108
-116.
2
Sakaguchi, S..
2005
. Naturally arising Foxp3-expressing CD25+CD4+ regulatory T cells in immunological tolerance to self and non-self.
Nat. Immunol.
6
:
345
-352.
3
Ziegler, S. F..
2006
. FOXP3: of mice and men.
Annu. Rev. Immunol.
24
:
209
-226.
4
Dieckmann, D., H. Plottner, S. Berchtold, T. Berger, G. Schuler.
2001
. Ex vivo isolation and characterization of CD4+CD25+ T cells with regulatory properties from human blood.
J. Exp. Med.
193
:
1303
-1310.
5
Piccirillo, C. A., E. M. Shevach.
2001
. Cutting edge: control of CD8+ T cell activation by CD4+CD25+ immunoregulatory cells.
J. Immunol.
167
:
1137
-1140.
6
Shevach, E. M., R. S. McHugh, C. A. Piccirillo, A. M. Thornton.
2001
. Control of T-cell activation by CD4+CD25+ suppressor T cells.
Immunol. Rev.
182
:
58
-67.
7
Trzonkowski, P., E. Szmit, J. Mysliwska, A. Dobyszuk, A. Mysliwski.
2004
. CD4+CD25+ T regulatory cells inhibit cytotoxic activity of T CD8+ and NK lymphocytes in the direct cell-to-cell interaction.
Clin. Immunol.
112
:
258
-267.
8
Azuma, T., T. Takahashi, A. Kunisato, T. Kitamura, H. Hirai.
2003
. Human CD4+CD25+ regulatory T cells suppress NKT cell functions.
Cancer Res.
63
:
4516
-4520.
9
Veldhoen, M., H. Moncrieffe, R. J. Hocking, C. J. Atkins, B. Stockinger.
2006
. Modulation of dendritic cell function by naive and regulatory CD4+ T cells.
J. Immunol.
176
:
6202
-6210.
10
Powrie, F., J. Carlino, M. W. Leach, S. Mauze, R. L. Coffman.
1996
. A critical role for transforming growth factor-β but not interleukin 4 in the suppression of T helper type 1-mediated colitis by CD45RBlow CD4+ T cells.
J. Exp. Med.
183
:
2669
-2674.
11
Takahashi, T., T. Tagami, S. Yamazaki, T. Uede, J. Shimizu, N. Sakaguchi, T. W. Mak, S. Sakaguchi.
2000
. Immunologic self-tolerance maintained by CD25+CD4+ regulatory T cells constitutively expressing cytotoxic T lymphocyte-associated antigen 4.
J. Exp. Med.
192
:
303
-310.
12
Taylor, J. J., M. Mohrs, E. J. Pearce.
2006
. Regulatory T cell responses develop in parallel to Th responses and control the magnitude and phenotype of the Th effector population.
J. Immunol.
176
:
5839
-5847.
13
D'Cruz, L. M., L. Klein.
2005
. Development and function of agonist-induced CD25+Foxp3+ regulatory T cells in the absence of interleukin 2 signaling.
Nat. Immunol.
6
:
1152
-1159.
14
Hoffmann, P., R. Eder, L. A. Kunz-Schughart, R. Andreesen, M. Edinger.
2004
. Large-scale in vitro expansion of polyclonal human CD4+CD25high regulatory T cells.
Blood
104
:
895
-903.
15
Ahmadzadeh, M., S. A. Rosenberg.
2006
. IL-2 administration increases CD4+CD25hi Foxp3+ regulatory T cells in cancer patients.
Blood
107
:
2409
-2414.
16
Zorn, E., H. T. Kim, S. J. Lee, B. H. Floyd, D. Litsa, S. Arumugarajah, R. Bellucci, E. P. Alyea, J. H. Antin, R. J. Soiffer, J. Ritz.
2005
. Reduced frequency of FOXP3+ CD4+CD25+ regulatory T cells in patients with chronic graft-versus-host disease.
Blood
106
:
2903
-2911.
17
Ochando, J. C., A. C. Yopp, Y. Yang, A. Garin, Y. Li, P. Boros, J. Llodra, Y. Ding, S. A. Lira, N. R. Krieger, J. S. Bromberg.
2005
. Lymph node occupancy is required for the peripheral development of alloantigen-specific Foxp3+ regulatory T cells.
J. Immunol.
174
:
6993
-7005.
18
van den Eertwegh, A. J., W. J. Boersma, E. Claassen.
1992
. Immunological functions and in vivo cell-cell interactions of T cells in the spleen.
Crit. Rev. Immunol.
11
:
337
-380.
19
Taylor, P. A., A. Panoskaltsis-Mortari, J. M. Swedin, P. J. Lucas, R. E. Gress, B. L. Levine, C. H. June, J. S. Serody, B. R. Blazar.
2004
. L-Selectinhi but not the L-selectinlo CD4+25+ T-regulatory cells are potent inhibitors of GVHD and BM graft rejection.
Blood
104
:
3804
-3812.
20
Ermann, J., P. Hoffmann, M. Edinger, S. Dutt, F. G. Blankenberg, J. P. Higgins, R. S. Negrin, C. G. Fathman, S. Strober.
2005
. Only the CD62L+ subpopulation of CD4+CD25+ regulatory T cells protects from lethal acute GVHD.
Blood
105
:
2220
-2226.
21
Zhao, D., C. Zhang, T. Yi, C. L. Lin, I. Todorov, F. Kandeel, S. Forman, D. Zeng.
2008
. In vivo-activated CD103+CD4+ regulatory T cells ameliorate ongoing chronic graft-versus-host disease.
Blood
112
:
2129
-2138.
22
Menning, A., U. E. Hopken, K. Siegmund, M. Lipp, A. Hamann, J. Huehn.
2007
. Distinctive role of CCR7 in migration and functional activity of naive- and effector/memory-like Treg subsets.
Eur. J. Immunol.
37
:
1575
-1583.
23
Wysocki, C. A., Q. Jiang, A. Panoskaltsis-Mortari, P. A. Taylor, K. P. McKinnon, L. Su, B. R. Blazar, J. S. Serody.
2005
. Critical role for CCR5 in the function of donor CD4+CD25+ regulatory T cells during acute graft-versus-host disease.
Blood
106
:
3300
-3307.
24
Fu, S., A. C. Yopp, X. Mao, D. Chen, N. Zhang, D. Chen, M. Mao, Y. Ding, J. S. Bromberg.
2004
. CD4+CD25+CD62+ T-regulatory cell subset has optimal suppressive and proliferative potential.
Am. J. Transplant.
4
:
65
-78.
25
Wolf, D., K. Hochegger, A. M. Wolf, H. F. Rumpold, G. Gastl, H. Tilg, G. Mayer, E. Gunsilius, A. R. Rosenkranz.
2005
. CD4+CD25+ regulatory T cells inhibit experimental anti-glomerular basement membrane glomerulonephritis in mice.
J. Am. Soc. Nephrol.
16
:
1360
-1370.
26
Budde, K., M. Schutz, P. Glander, H. Peters, J. Waiser, L. Liefeldt, H. H. Neumayer, T. Bohler.
2006
. FTY720 (fingolimod) in renal transplantation.
Clin Transplant.
20
: (Suppl. 17):
17
-24.
27
Chueh, S. C., L. Tian, M. Wang, M. E. Wang, S. M. Stepkowski, B. D. Kahan.
1997
. Induction of tolerance toward rat cardiac allografts by treatment with allochimeric class I MHC antigen and FTY720.
Transplantation
64
:
1407
-1414.
28
Schwab, S. R., J. G. Cyster.
2007
. Finding a way out: lymphocyte egress from lymphoid organs.
Nat. Immunol.
8
:
1295
-1301.
29
Cyster, J. G..
2005
. Chemokines, sphingosine-1-phosphate, and cell migration in secondary lymphoid organs.
Annu. Rev. Immunol.
23
:
127
-159.
30
Daniel, C., N. Sartory, N. Zahn, G. Geisslinger, H. H. Radeke, J. M. Stein.
2007
. FTY720 ameliorates Th1-mediated colitis in mice by directly affecting the functional activity of CD4+CD25+ regulatory T cells.
J. Immunol.
178
:
2458
-2468.
31
Lan, Y. Y., A. De Creus, B. L. Colvin, M. Abe, V. Brinkmann, P. T. Coates, A. W. Thomson.
2005
. The sphingosine-1-phosphate receptor agonist FTY720 modulates dendritic cell trafficking in vivo.
Am. J. Transplant.
5
:
2649
-2659.
32
Vora, K. A., E. Nichols, G. Porter, Y. Cui, C. A. Keohane, R. Hajdu, J. Hale, W. Neway, D. Zaller, S. Mandala.
2005
. Sphingosine 1-phosphate receptor agonist FTY720-phosphate causes marginal zone B cell displacement.
J. Leukocyte Biol.
78
:
471
-480.
33
Zeiser, R., V. H. Nguyen, A. Beilhack, M. Buess, S. Schulz, J. Baker, C. H. Contag, R. S. Negrin.
2006
. Inhibition of CD4+CD25+ regulatory T-cell function by calcineurin-dependent interleukin-2 production.
Blood
108
:
390
-399.
34
Zeiser, R., D. B. Leveson-Gower, E. A. Zambricki, N. Kambham, A. Beilhack, J. Loh, J. Z. Hou, R. S. Negrin.
2008
. Differential impact of mammalian target of rapamycin inhibition on CD4+CD25+Foxp3+ regulatory T cells compared with conventional CD4+ T cells.
Blood
111
:
453
-462.
35
Rosenkranz, A. R., D. L. Mendrick, R. S. Cotran, T. N. Mayadas.
1999
. P-selectin deficiency exacerbates experimental glomerulonephritis: a protective role for endothelial P-selectin in inflammation.
J. Clin. Invest.
103
:
649
-659.
36
Turka, L. A., P. T. Walsh.
2008
. IL-2 signaling and CD4+CD25+Foxp3+ regulatory T cells.
Front Biosci.
13
:
1440
-1446.
37
Yao, Z., Y. Kanno, M. Kerenyi, G. Stephens, L. Durant, W. T. Watford, A. Laurence, G. W. Robinson, E. M. Shevach, R. Moriggl, et al
2007
. Nonredundant roles for STAT-5a/b in directly regulating Foxp3.
Blood
109
:
4368
-4375.
38
Kappos, L., J. Antel, G. Comi, X. Montalban, P. O'Connor, C. H. Polman, T. Haas, A. A. Korn, G. Karlsson, E. W. Radue.
2006
. Oral fingolimod (FTY720) for relapsing multiple sclerosis.
N. Engl. J. Med.
355
:
1124
-1140.
39
Sawicka, E., G. Dubois, G. Jarai, M. Edwards, M. Thomas, A. Nicholls, R. Albert, C. Newson, V. Brinkmann, C. Walker.
2005
. The sphingosine 1-phosphate receptor agonist FTY720 differentially affects the sequestration of CD4+/CD25+ T-regulatory cells and enhances their functional activity.
J. Immunol.
175
:
7973
-7980.
40
Pinschewer, D. D., A. F. Ochsenbein, B. Odermatt, V. Brinkmann, H. Hengartner, R. M. Zinkernagel.
2000
. FTY720 immunosuppression impairs effector T cell peripheral homing without affecting induction, expansion, and memory.
J. Immunol.
164
:
5761
-5770.
41
Masteller, E. L., Q. Tang, J. A. Bluestone.
2006
. Antigen-specific regulatory T cells–ex vivo expansion and therapeutic potential.
Semin. Immunol.
18
:
103
-110.
42
Sakaguchi, S., N. Sakaguchi, M. Asano, M. Itoh, M. Toda.
1995
. Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor α-chains (CD25): breakdown of a single mechanism of self-tolerance causes various autoimmune diseases.
J. Immunol.
155
:
1151
-1164.
43
Sakaguchi, S., M. Ono, R. Setoguchi, H. Yagi, S. Hori, Z. Fehervari, J. Shimizu, T. Takahashi, T. Nomura.
2006
. Foxp3+ CD25+CD4+ natural regulatory T cells in dominant self-tolerance and autoimmune disease.
Immunol. Rev.
212
:
8
-27.
44
Yu, A., L. Zhu, N. H. Altman, T. R. Malek.
2009
. A low interleukin-2 receptor signaling threshold supports the development and homeostasis of T regulatory cells.
Immunity
30
:
204
-217.
45
Dorsam, G., M. H. Graeler, C. Seroogy, Y. Kong, J. K. Voice, E. J. Goetzl.
2003
. Transduction of multiple effects of sphingosine 1-phosphate (S1P) on T cell functions by the S1P1 G protein-coupled receptor.
J. Immunol.
171
:
3500
-3507.
46
Jin, Y., E. Knudsen, L. Wang, Y. Bryceson, B. Damaj, S. Gessani, A. A. Maghazachi.
2003
. Sphingosine 1-phosphate is a novel inhibitor of T-cell proliferation.
Blood
101
:
4909
-4915.
47
Wang, W., M. H. Graeler, E. J. Goetzl.
2005
. Type 4 sphingosine 1-phosphate G protein-coupled receptor (S1P4) transduces S1P effects on T cell proliferation and cytokine secretion without signaling migration.
FASEB J.
19
:
1731
-1733.
48
Almeida, A. R., B. Zaragoza, A. A. Freitas.
2006
. Indexation as a novel mechanism of lymphocyte homeostasis: the number of CD4+CD25+ regulatory T cells is indexed to the number of IL-2-producing cells.
J. Immunol.
177
:
192
-200.
49
Tang, Q., J. Y. Adams, C. Penaranda, K. Melli, E. Piaggio, E. Sgouroudis, C. A. Piccirillo, B. L. Salomon, J. A. Bluestone.
2008
. Central role of defective interleukin-2 production in the triggering of islet autoimmune destruction.
Immunity
28
:
687
-697.
50
Lin, J. X., W. J. Leonard.
2000
. The role of STAT-5a and STAT-5b in signaling by IL-2 family cytokines.
Oncogene
19
:
2566
-2576.
51
Edinger, M., P. Hoffmann, J. Ermann, K. Drago, C. G. Fathman, S. Strober, R. S. Negrin.
2003
. CD4+CD25+ regulatory T cells preserve graft-versus-tumor activity while inhibiting graft-versus-host disease after bone marrow transplantation.
Nat. Med.
9
:
1144
-1150.
52
Taylor, P. A., M. J. Ehrhardt, C. J. Lees, J. Tolar, B. J. Weigel, A. Panoskaltsis-Mortari, J. S. Serody, V. Brinkmann, B. R. Blazar.
2007
. Insights into the mechanism of FTY720 and compatibility with regulatory T cells for the inhibition of graft-versus-host disease (GVHD).
Blood
110
:
3480
-3488.
53
Cinamon, G., M. A. Zachariah, O. M. Lam, F. W. Foss, Jr, J. G. Cyster.
2008
. Follicular shuttling of marginal zone B cells facilitates antigen transport.
Nat. Immunol.
9
:
54
-62.
54
Premenko-Lanier, M., N. B. Moseley, S. T. Pruett, P. A. Romagnoli, J. D. Altman.
2008
. Transient FTY720 treatment promotes immune-mediated clearance of a chronic viral infection.
Nature
454
:
894
-898.
55
Liu, G., S. Burns, G. Huang, K. Boyd, R. L. Proia, R. A. Flavell, H. Chi.
2009
. The receptor S1P1 overrides regulatory T cell-mediated immune suppression through Akt-mTOR.
Nat. Immunol.
10
:
769
-777.