CD25 + Regulatory T Cells and CD4 + CD25 + Equilibrium between ...

2 downloads 8337 Views 3MB Size Report
Receive free email-alerts when new articles cite this article. Sign up at: .... blast; TCD4T, total CD4 T cell; TL, T lymphocyte; Treg, regulatory T cell;. Tresp ...
The Journal of Immunology

A Dual Action of Rheumatoid Arthritis Synovial Fibroblast IL-15 Expression on the Equilibrium between CD4ⴙCD25ⴙ Regulatory T Cells and CD4ⴙCD25ⴚ Responder T Cells1 Marta Benito-Miguel, Yolanda García-Carmona, Alejandro Balsa, Carlos Pe´rez de Ayala, Tatiana Cobo-Iba´n˜ez, Emilio Martín-Mola, and María-Eugenia Miranda-Caru´s2 We previously described that fibroblast-like cells from the synovium of rheumatoid arthritis patients (RASFib) constitutively express intracellular and surface IL-15, which induces activation of cocultured T cells. Our objective was to study the effect of RASFib IL-15 expression on the function of human CD4ⴙCD25ⴙ regulatory T cells (Treg). RASFib, through their constitutive IL-15 expression, were able to induce the proliferation of human Tregs stimulated through their TCR, and at the same time potentiated their suppressive action on the cytokine secretion of CD4ⴙCD25ⴚ responder T cells (Tresp). In parallel, constitutive RASFib IL-15 expression mediated an up-regulated response of Tresp. Subsequently, total CD4ⴙ T cells, containing natural proportions of Treg and Tresp, secreted an increased amount of pathogenic cytokines when cocultured with RASFib despite the presence of proliferating Treg with superior regulatory potency. In summary, RASFib IL-15 exerts a dual action on the equilibrium between Treg and Tresp by potentiating the suppressive effect of Treg while augmenting the proinflammatory action of Tresp; the result is a shift of the Treg/Tresp balance toward a proinflammatory state. This alteration of the Treg/Tresp equilibrium is not observed in the presence of osteoarthritis synovial fibroblasts or dermal fibroblasts, which do not constitutively express surface IL-15. Additionally, Treg with superior suppressive potency were present in the peripheral blood and the synovial fluid of RA patients, but this enhanced immunoregulatory activity was not able to overcome the increased secretion of pathogenic cytokines by RA-Tresp, indicating that rheumatoid arthritis patients demonstrate an altered Treg/Tresp equilibrium in vivo. The Journal of Immunology, 2009, 183: 8268 – 8279.

T

he hallmark of rheumatoid arthritis (RA),3 a systemic autoimmune disorder, is a chronic joint inflammation. RA synovitis is characterized by hyperplasia of synovial fibroblasts (RASFib), which plays a pivotal role in the pathogenesis of the disease (1). RASFib actively participate in the erosion of cartilage and bone (1). Additionally, RASFib contribute to creating a tissue microenvironment that favors the initiation and perpetuation of the immune response (2). Direct interactions of RASFib with T cells are important contributors to the chronic RA inflammation: RASFib prevent apoptotic cell death of cocultured T cells (3, 4), up-regulate T cell cytokine and adhesion molecule expres-

Department of Rheumatology, Hospital Universitario La Paz, Madrid, Spain Received for publication January 5, 2009. Accepted for publication October 12, 2009. 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 This work was supported by Ministerio de Educacio´n Grant SAF 2006-01670 (to M.-E.M.C., M.B.M.) and a Hospital La Paz Research grant (to Y.G.C.). 2 Address correspondence and reprint requests to Dr. M. Eugenia Miranda-Caru´s, Department of Rheumatology, Hospital La Paz, Paseo de la Castellana, 261, 28046 Madrid, Spain. E-mail address: [email protected] 3

Abbreviations used in this paper: RA, rheumatoid arthritis; HCPB, peripheral blood T cells of healthy controls; HCPBTL, peripheral blood T lymphocytes from healthy controls; MFI, mean fluorescence intensity; MTX, methotrexate; RAPB, peripheral blood T cells from untreated early rheumatoid arthritis patients; RAPBTL, peripheral blood T lymphocytes from early rheumatoid arthritis patients; RAPB-R, peripheral blood T cells from early rheumatoid arthritis patients who had achieved disease remission with treatment; RASF, rheumatoid arthritis synovial fluid; RASFib, rheumatoid arthritis synovial fibroblast; RASFTL, synovial fluid T lymphocytes from rheumatoid arthritis patients; SFib, synovial fibroblast; TCD4T, total CD4⫹ T cell; TL, T lymphocyte; Treg, regulatory T cell; Tresp, responder T cell. Copyright © 2009 by The American Association of Immunologists, Inc. 0022-1767/09/$2.00 www.jimmunol.org/cgi/doi/10.4049/jimmunol.0900007

sion (5), and induce proliferation of T cells upon long-term coculture (6). Naturally occurring regulatory CD4⫹CD25⫹ T cells (Treg) are essential for maintaining peripheral tolerance and controlling autoimmunity (7, 8). Treg are suppressive and anergic to TCR stimulation (7, 8). Suppression by Treg results in decreased proliferation, decreased IL-2, and decreased proinflammatory cytokine production of responder CD4⫹CD25⫺ T cells (Tresp) (9, 10). The mechanism responsible for the suppressor effect of Treg is not clear and seems to be mediated by direct cell contact with Tresp (7, 8). A tight control of Treg function is crucial for the immune system. However, the anergy and/or suppressive capacity of Treg can be modified by cytokines (10 –14), by coculture with activated dendritic cells (12–17), and by TLR ligands (18, 19). RASFib are a known source of pathogenic cytokines in RA (1). We previously reported that constitutively expressed IL-15 on the surface of RASFib is biologically active on cocultured T lymphocytes through direct cell contact (5). IL-15, a potent T cell growth factor, has been described to drive the proliferation of human Treg (20, 21), to increase human Treg suppressor potency (21), and to interfere with human Treg function (14); additionally, it has been reported that human Treg cocultured with dermal fibroblasts proliferate in the presence but not in the absence of exogenous recombinant IL-15 (22). Therefore, our objective was to investigate the role of RASFib IL-15 expression on the properties of Treg. Additionally, we were interested in analyzing the characteristics of Treg and Tresp present in the peripheral blood of early RA patients (RAPB) and the synovial fluid of RA patients (RASF). In fact, T cells present in vivo in the synovial fluid of RA patients (RASFTL) are permanently in contact with RASFib lining the

The Journal of Immunology intimal layer of the hyperplastic synovial membrane (23), and they demonstrate phenotypical and functional signs of activation (24). T cells circulating in the peripheral blood of RA patients do not show such clear signs of activation, and controversial data exist regarding their phenotypical and functional features (24). Likewise, additional data are needed to clarify the nature of the altered regulatory capacity of RA Treg (25).

Materials and Methods Patients Synovial membranes were obtained from 10 RA patients undergoing synovectomy or arthroplasty and from 10 osteoarthritis (OA) patients undergoing arthroplasty. Synovial fluid was obtained from 30 patients with established RA who were receiving treatment with oral methotrexate (MTX) and low-dose prednisone. Peripheral blood was obtained from 30 healthy controls and from 30 early RA patients fulfilling at least four American College of Rheumatology criteria (26), who had never received diseasemodifying drugs or corticosteroids and with a disease duration of ⬍6 mo. La Paz University Hospital in Madrid, Spain, has a monographic clinic that takes care of early arthritis patients referred from a wide primary care area. This opportunity facilitated recruitment of untreated early RA patients for the present study. Among early RA patients there were 9 men and 21 women, 23 (76%) of whom tested positive for IgM rheumatoid factor; their ages were 19 – 82 years (mean, 52.72; SD, 17.23; median, 49 years), duration of symptoms at first evaluation was from 2 to 24 wk (mean, 10.35; SD, 6.5; median, 10 wk), and disease activity score 28 (DAS28) (27) at first evaluation was from 4.75 to 7.64 (mean, 5.93; SD, 0.75; median, 5.72). Ten patients with early RA donated blood for a second time, once disease activity had been controlled. These patients were receiving 15 mg of oral MTX weekly, except for one patient who was taking 20 mg per week and one patient who was taking 25 mg per week. Additionally, all 10 patients except for 1 were taking prednisone, 2.5 mg daily. IgM rheumatoid factor was positive at initial evaluation in 8 of these 10 patients; there were 2 men and 8 women, with ages of 40.4 ⫾ 12.54 years (mean ⫾ SD). DAS28 before initiation of treatment was 5.76 ⫾ 0.81 (mean ⫾ SD) and at the time of second blood drawing it was 1.93 ⫾ 0.5. These patients were in remission as defined by a DAS28 score ⬍2.6 (28). The study was approved by the Hospital Ethics Committee.

8269 CD4-PerCP-Cy5, CD25-FITC, and CD127-PE (all from BD Pharmingen). An Alexa Fluor 647-labeled Ab against the intracellular Ag FoxP3 (clone 236A/E; eBioscience) was used to assess purity of the isolated Treg population.

T cell stimulation T cells were stimulated in flat-bottom 96-well plates (105 cells/well) using two different anti-CD3 mAbs in two different settings: a plate-bound antiCD3 IgG subclass mAb (UCTH1; BD Pharmingen) and a soluble anti-CD3 IgE subclass (T3/4.E; Sanquin, formerly CLB). It is known that anti-CD3 Abs, when bound to plastic culture surfaces, facilitate crosslinking that induces T cell proliferation. In contrast, soluble anti-CD3 Abs do not induce T cell proliferation in the absence of APCs or anti-CD28 Abs. A remarkable exception is an IgE anti-CD3 Ab that was developed at the CLB as a switch variant originating from an IgG1 anti-CD3 MoAb (CLBT3/4.E). Soluble CLB-T3/4.E is able to stimulate T cells directly, without the need for a second signal (30), and it allows the comparison of stimulated T cell responses in the absence or presence of RASFib. No accessory cells and no costimulatory Abs were used in an attempt to induce a mild stimulation of T cells and to avoid the use of agents that may interfere with Treg function.

T cell proliferation assays T cell proliferation was assessed by [3H]thymidine incorporation and by CFSE dilution. For thymidine incorporation assays, 18 h before the termination of the cultures, the plates were pulsed with 0.5 ␮Ci/well [3H]thymidine (GE Healthcare). The cells were harvested on paper filters and [3H]thymidine uptake was measured in a liquid scintillation counter. Additionally, and to track cell division by flow cytometry, cells were labeled before initiating culture with CFSE (Molecular Probes/Invitrogen), at a final concentration of 8 ␮M.

Coculture of T cells with RASFib Coculture experiments were performed with confluent fibroblast cultures prepared 48 h before contact. All experimental conditions were performed in triplicate and variation between replicates was ⬍5%. Fibroblasts were seeded in 96-well plates (Corning) at 1 ⫻ 104 cells per well. Forty-eight hours later, CFSE-labeled total CD4⫹ T cells (TCD4), Tresp, or Treg (105 cells/well) were added and the medium was supplemented with antiCD3IgE mAb (0.25 ␮g/ml). Cocultures were maintained for 5 days.

Transwell system Culture of human fibroblasts RASFib and OASFib were obtained by collagenase digestion (type I; Worthington Biochemical) of human synovial tissue obtained at arthroplasty or synovectomy. Dermal fibroblasts were obtained by collagenase digestion of normal skin obtained from punch biopsies of five healthy volunteers. Cells were plated in 75-cm2 flasks (Corning Costar) and grown in DMEM (Lonza) supplemented with 10% FCS (Lonza), 2 mM L-glutamine, 50 U/ml penicillin, and 50 ␮g/ml streptomycin (Lonza). Cells were passaged at 1/2 dilution when reaching 95% confluence, by gentle trypsinization (0.05% trypsin/0.53 mM EDTA; Invitrogen). Fibroblasts were used between the third and fifth passages. At this time, they appear to be a homogeneous population of fibroblast-like cells that stain positive with anti-Thy-1 (CD90) Ab (29) and are negative for the expression of CD1, CD3, CD19, CD14, HLA class II, CD80, and CD86, as determined by flow cytometry and fluorochrome-conjugated mAbs (BD Pharmingen).

Isolation of CD4⫹ T cells and purification of CD4⫹CD25⫺ and CD4⫹CD25⫹ T cell fractions Mononuclear cells were isolated from human blood by density centrifugation over Ficoll-Paque Plus (GE Healthcare). CD4⫹ T cells were subsequently purified by negative selection in an autoMACS (Miltenyi Biotec) using a CD4⫹ T cell isolation kit II from Miltenyi Biotec, containing a cocktail of biotin-conjugated Abs against CD8, CD14, CD16, CD19, CD36, CD56, CD123, TCR␥/␦, and Glycophorin A, followed by anti-biotin microbeads. Isolated CD4⫹ T cells were 98% pure and free of detectable CD14⫹ monocytes, CD56⫹ NK cells, and CD19⫹ B cells. Subsequently, CD4⫹CD25⫹bright T cells were positively selected using a limiting amount of anti-CD25 microbeads (5 ␮l/107 CD4⫹ cells) (Miltenyi Biotec) in an autoMACS. CD4⫹CD25⫺ T cells were subsequently depleted of any contaminating CD25⫹ T cells following a second round of incubation with CD25⫹ microbeads (20 ␮l/107 CD4⫹ T cells). T cell subpopulations of CD4⫹CD25⫹ T cells and CD4⫹CD25⫺ T cells were used immediately after isolation. In separate experiments, the CD4⫹CD25⫺CD127⫹ Tresp and CD4⫹CD25⫹ CD127⫺ Treg populations were isolated from total CD4⫹ T cells by FACS sorting on a BD Vantage SE (BD Biosciences). These cells were stained with

A 0.4-␮m Transwell system (Corning) was used to conduct some coculture experiments. The system consists of two compartments: a top well, with a porous matrix (0.4 ␮m), and a bottom well. This setup allows coculture of two types of cells to grow in the same medium with soluble factors exchanged through the pores, while preventing direct contact between them. RASFib were grown to confluence in the bottom well, and T cells were added either to the same well, allowing contact, or in the top well, avoiding contact.

Assessment of Treg function The function of naturally occurring CD4⫹CD25⫹ Treg present at physiological rates in human peripheral blood was assessed by comparing the proliferation and IFN-␥ and TNF-␣ secretion of anti-CD3-stimulated total CD4⫹ T cells (TCD4T) against CD25⫹-depleted CD4⫹ T cells (Tresp), in the presence or absence of RASFib. The potency of inhibition exerted by Treg was calculated as: [1 ⫺ (proliferation or cytokine secretion of total CD4⫹ T cells/proliferation or cytokine secretion of CD25⫺ T cells)]. In some experiments, and to determine the per cell suppressive potency of Treg, isolated Treg were cocultured with Tresp at different Treg-to-Tresp ratios (1:1, 1:2, 1:5, 1:10, and 1:20); potency of suppression was then calculated as: [1 ⫺ (proliferation or cytokine secretion of Treg ⫹ Tresp coculture/proliferation or cytokine secretion of Tresp)].

Inhibition by neutralizing mAbs Parallel experiments included the addition of IL-15 blocking agents: a neutralizing anti-IL-15 mAb (10 ␮g/ml) (mab247, mouse IgG1; R&D Systems), a recombinant IL-15R␣/human Fc␥1 chimera (100 ng/ml; R&D Systems), and an IL-15 mutant/murine Fc␥2a chimera (10 ␮g/ml; Chimerigen); the latter is a high-affinity receptor site-specific antagonist for IL15R␣ (31). Alternatively, the binding control anti-HLA class I (clone W6/ 32, 10 ␮g/ml; Sigma-Aldrich), an anti-human MHC class II (HLA-DR, -DP, -DQ, 10 ␮g/ml; BD Biosciences), or appropriate isotype controls (murine IgG1, murine IgG2a, or human IgG1; all from R&D Systems) were used. These Abs were incubated with RASFib for 30 min at 37°C; T cells were subsequently added without washing.

8270

EFFECT OF RA SYNOVIAL FIBROBLAST IL-15 EXPRESSION ON Treg

Intracellular cytokine staining, surface staining, and flow cytometry For intracellular FoxP3 staining, T cells were washed with PBS/2% FCS/ 0.01% NaN3, permeabilized for 10 min with FACS permeabilizing solution 2 (BD Pharmingen), washed again, and incubated on ice for 1 h with an Alexa Fluor 647-labeled anti-FoxP3 mAb (clone 236A/E7; eBioscience) or an irrelevant isotype control mAb. After washing once with PBS/2% FCS/0.01% NaN3 and once with PBS, cells were resuspended in 1% paraformaldehyde and analyzed in a FACSCalibur flow cytometer using CellQuest software (BD Biosciences). For staining of surface molecules, the permeabilization step was omitted. Surface IL-15R␣ was detected with anti-IL-15R␣ mAb (R&D Systems), followed by an Alexa Fluor 488-goat anti-mouse Ab (Molecular Probes/Invitrogen). Fluorochrome-conjugated mAbs from BD Pharmingen were used to examine the expression of the phenotypic markers CD3, CD4, CD25, CD127, and CD69. Mean fluorescence intensity (MFI) is calculated as the difference between the MFI of tested cells and the MFI of background staining.

Soluble cytokine detection ELISAs for IFN-␥ and TNF-␣ were performed in cell-free supernatants using kits from BD Pharmingen.

Quantitative RT-PCR Total cellular RNA was isolated using the RNAqueous kit (Ambion/Applied Biosystems) with DNase treatment. For each sample, 1 ␮g of total RNA was subjected to reverse transcription using an Advantage RT for PCR kit (Clontech). Aliquots (1 ␮l) of the reverse transcription products were used for quantitative PCR in the LightCycler PCR and detection system (Roche Molecular Biochemicals) with the FastStart DNA Master SYBR Green I kit (Roche Diagnostics). The PCR reactions were set up in microcapillary tubes in a volume of 20 ␮l. The following sense and antisense primers were used: FoxP3 sense, 5⬘-CAC CTA CGC CAC GCT CATC-3⬘ and antisense, 5⬘-ACT CAG GTT GTG GCG GAT G-3⬘; IL2R␣ (CD25) sense, 5⬘-TCA GTG CGT CCA GGG ATA CAG G-3⬘ and antisense, 5⬘-TCA GGA GGA GGA CGC TGA TCA G-3⬘; IL-7R␣ (CD127) sense, 5⬘-TCT GGA GAA AGT GGC TAT GC-3⬘ and antisense, 5⬘-CCT GGC GGT AAG CTA CAT C-3⬘. IL-15R␣ primers are located in exon 2 (sense) and exon 5 (antisense) to amplify mRNA encoding all IL-15R␣ variants that contain exon 2, and thus, bind IL-15: IL-15R␣ sense, 5⬘-GGA ATT CAT CAC GTG CCC TCC CCC CAT G-3⬘ and antisense, 5⬘-CGG GAT CCT CAA GTG GTG TCG CTG TGG CCC TG-3⬘ (product size, 543/444). Amplification of the PCR products was monitored by measuring SYBR Green I dye fluorescence. As an external standard, the transcript of 18S rRNA was amplified from the same cDNA samples using primers manufactured by Ambion. Each sample was run in triplicate. Results were analyzed with LightCycler version 3.5.3 software (Roche Diagnostics). Quantities of specific mRNA in the sample were measured according to the corresponding gene-specific standard curve.

Statistical analysis Comparison between groups was by a Mann-Whitney U test. Paired samples were compared using a Wilcoxon matched pairs signed rank sum test. When appropriate, Bonferroni correction for multiple comparisons was applied.

Results Phenotype and function of human peripheral blood CD4⫹CD25⫹ Treg and CD4⫹CD25⫺ Tresp Flow cytometry and quantitative RT-PCR demonstrated that CD4⫹CD25⫹ Treg isolated with limiting amounts of magnetic beads expressed high levels of FoxP3 in sharp contrast with isolated CD4⫹CD25⫺ Tresp (Fig. 1, A and B). Additionally, a remarkable CD127 (IL-7R␣) expression was observed on Tresp but not on Treg (Fig. 1, A and B), further confirming the previously described characteristic phenotype of both populations (32, 33). Furthermore, we observed for the first time that IL15R␣ expression is significantly higher on Treg when compared with Tresp (Fig. 1, A and B), suggesting a role for IL-15 on human Treg biology. The gold standard for Treg identification is functional proof of their inhibitory capacity, since all of the as yet available phenotypical markers can be up-regulated upon T cell activation (34).

Therefore, we next sought to examine the function of naturally occurring CD4⫹CD25⫹ Treg present at physiological rates in human peripheral blood. To this end, the proliferation and IFN-␥ and TNF-␣ secretion of anti-CD3-stimulated TCD4T were compared against CD25⫹-depleted CD4⫹ T cells (Tresp) (Fig. 1C). In the presence of plate-bound anti-CD3 IgG (UCHT1 at 0.25 ␮g/ml; BD Pharmingen) the proliferation and cytokine secretion of Tresp was significantly higher when compared with TCD4T (Fig. 1C). This indicates that the physiological proportion of Treg present in human peripheral blood has a significant regulatory activity. Comparable results were obtained when cells were stimulated with soluble anti-CD3 IgE (CLB-T3/4.E, 0.25 ␮g/ml) (Fig. 1C), consistent with previous observations by van Lier et al. (30). As expected, magnetically sorted CD25⫹ Treg did not secrete IFN- ␥ or TNF-␣ and were anergic to TCR stimulation, that is, they did not proliferate when stimulated with anti-CD3 mAbs (Fig. 1C). Additionally, the per cell potency of Treg isolated using limiting amounts of magnetic beads was tested (Fig. 1D). To this end, Treg were cocultured with Tresp at different Treg/Tresp ratios in the presence of a submaximal stimulus (soluble anti-CD3 IgE, CLBT3/4.E, 0.25 ␮g/ml). An optimal suppressive capacity and a doseresponse curve could be observed (Fig. 1D). Preliminary experiments demonstrated that the inhibitory function of Treg that had been isolated by flow cytometry was not significantly different from that of Treg isolated with limiting amounts of magnetic beads. In the presence of RASFib, Treg-mediated suppression of Tresp proliferation is not apparent, while suppression of IFN-␥ and TNF-␣ secretion is enhanced We then sought to establish whether RASFib are able to modify the equilibrium between Treg and Tresp. To this end, CFSE-labeled TCD4⫹ T cells, Tresp, or Treg were cultured in the absence or presence of RASFib. When compared against pure T cell cultures, coculture with RASFib resulted in increased proliferative responses to soluble anti-CD3 IgE mAb for both TCD4T and isolated Tresp (Fig. 2A); in this setting, the proliferation rate of Tresp was not significantly different from TCD4⫹ T cells (Fig. 2A): in other words, the suppressive effect of natural proportions of Treg on Tresp proliferation was not apparent. Remarkably, proliferation of anti-CD3-stimulated Treg was observed in the presence of RASFib (Fig. 2A). We then examined the effect of RASFib on anti-CD3-stimulated IFN-␥ and TNF-␣ secretion. In the presence of RASFib, IFN-␥ and TNF-␣ secretion were significantly increased when compared with pure T cell cultures for both Tresp and TCD4T (Fig. 2B). At the same time, IFN-␥ and TNF-␣ secretion of Tresp remained significantly higher when compared with TCD4⫹ T cells (Fig. 2B), indicating that the suppressive action of Treg on Tresp cytokine secretion is indeed maintained. Furthermore, the calculated potency of inhibition on cytokine secretion was significantly higher in the presence of RASFib (Fig. 2C). Importantly, the above-reported effects were not observed in the presence of OA synovial fibroblasts or dermal fibroblasts. Furthermore, these effects were not apparent when cocultures were established in the presence of 0.4 ␮M Transwell inserts, which do not allow direct contact between RASFib and T cells. Additionally, these effects were not seen when Treg, Tresp, or TCD4T were cultured in the presence of supernatants taken from cocultures of RASFib with Treg, Tresp, or TCD4T. Of note, T cells did not proliferate and did not secrete IFN-␥ or TNF-␣ when stimulated with soluble IgG anti-CD3 mAb (clone UCTH1; BD Pharmingen) in the presence of RASFib, indicating

The Journal of Immunology

8271

FIGURE 1. Phenotypical and functional study of magnetically sorted human peripheral blood CD4⫹CD25⫹ Treg and CD4⫹CD25⫺ Tresp. A, Flow cytometry histograms demonstrate that CD4⫹CD25⫹ T cells isolated using limiting amounts of magnetic beads express FoxP3 and at the same time are negative for CD127 (IL-7R␣) expression. Conversely, isolated CD4⫹CD25⫺ T cells do not express FoxP3 and are positive for CD127. Additionally, expression of IL-15R␣ on CD4⫹CD25⫹ T cells is significantly higher when compared with isolated CD4⫹CD25⫺ T cells. B, Quantitative RT-PCR demonstrated higher FOXP3, CD25, and IL-15R␣ together with lower IL-7R␣ mRNA expression in CD4⫹CD25⫹ T cells when compared with CD4⫹CD25⫺ T cells. ⴱ, p ⬍ 0.05 vs CD4⫹CD25⫺ T cells. C, In the presence of plate-bound anti-CD3 IgG (UCHT1, 0.25 ␮g/ml; BD Pharmingen) the proliferation and cytokine secretion of CD4⫹CD25⫺ T cells (Tresp) at 5 days of culture was significantly higher when compared with total CD4⫹ T cells. This indicates that the physiological proportion of Treg present in human peripheral blood has a significant regulatory activity. Comparable results were obtained when cells were stimulated with soluble anti-CD3 IgE (CLB-T3/4.E, 0.25 ␮g/ml). In contrast, a soluble anti-CD3 IgG mAb (UCTH1) did not induce T cell proliferation. As expected, magnetically sorted CD25⫹ Treg did not secrete IFN-␥ or TNF-␣ and were anergic to TCR stimulation, that is, they did not proliferate when stimulated with anti-CD3 mAbs. The potency of inhibition exerted by Treg on the proliferation and IFN-␥ and TNF-␣ secretion of Tresp was calculated as: [1 ⫺ (proliferation or cytokine secretion of TCD4T/proliferation or cytokine secretion of Tresp)]. D, Per cell potency of Treg isolated using limiting amounts of magnetic beads. Treg were cocultured for 5 days with Tresp at different Treg/Tresp ratios in the presence of a submaximal stimulus (soluble anti-CD3 IgE, CLB-T3/4.E, 0.25 ␮g/ml). Potency was calculated as: [1 ⫺ (proliferation or cytokine secretion of Treg ⫹ Tresp coculture/proliferation or cytokine secretion of Tresp)].

that RASFib are not expressing molecules with functional costimulatory capacity. FoxP3 expression is up-regulated upon T cell activation It has been reported that activated human CD4⫹ Tresp may undergo an up-regulation of FoxP3 expression that is not sufficient to induce Treg activity (35–38). The functional studies we described in the previous section indicate that Treg are not induced de novo in our system since activated Tresp produced high levels of effector cytokines TNF-␣ and IFN-␥ when compared with TCD4T and as opposed to activated Treg, which did not secrete pathogenic cytokines. Nevertheless, we were interested in examining FoxP3 expression in these T cells. Dot plots of CFSE-labeled TCD4T, Tresp, or Treg doublestained with an allophycocyanin-labeled anti-FoxP3 MoAb confirmed that FoxP3 expression is up-regulated upon activation (Fig. 3). This up-regulation was transient, reached a maximum on the

fifth day, and then progressively declined to return to basal expression levels around the ninth day. In the presence of RASFib, up-regulation was observed not only on activated TCD4T and Tresp but also on proliferating Treg (Fig. 3B). Subsequently, the MFI of FoxP3 expression remained higher among proliferating isolated Treg cocultured with RASFib when compared with proliferating Tresp (Fig. 3B), which is consistent with previously reported observations (35). Remarkably, two populations of Foxp3expressing cells were observed among stimulated TCD4T in the presence, but not in the absence, of RASFib: the majority of cells expressed an up-regulated FoxP3 level with an intensity that was comparable to that observed in stimulated Tresp; a minor population (⬃10%) demonstrated a higher Foxp3 expression, similar to that observed in stimulated Treg (Fig. 3B). In contrast, FoxP3 expression was homogeneous among proliferating Tresp (Fig. 3B). These data suggest that the Treg population present at physiological proportions in human peripheral blood proliferates when

8272

EFFECT OF RA SYNOVIAL FIBROBLAST IL-15 EXPRESSION ON Treg

FIGURE 2. Proliferation and cytokine secretion of total CD4⫹ T cells (TCD4T), isolated Tresp (CD4⫹CD25⫺), and isolated Treg (CD4⫹CD25⫹) stimulated with soluble anti-CD3IgE mAb and cultured for 5 days in the absence or presence of RASFib. A, Proliferation rate, as determined by flow cytometry of CFSE-labeled cells. Representative flow cytometry histograms are shown in the top panel. In the bottom bar histogram, each bar represents the arithmetic mean ⫾ SD of 30 subjects. When compared against pure T cell cultures (left panels), coculture with RASFib (right panels) resulted in augmented proliferative responses of both TCD4T and Tresp. In the cocultures, the proliferative rate of Tresp was not significantly different from that of TCD4⫹ T cells. This indicates that the suppressive effect of natural proportions of Treg on Tresp proliferation is not apparent in this setting. Remarkably, proliferation of Treg was observed in the presence of RASFib. ⴱ, p ⬍ 0.05 vs TCD4T (4⫹); †, p ⬍ 0.05 vs Treg (25⫹); ¥, p ⬍ 0.05 vs same cell population in the absence of RASFib. B, IFN-␥ and TNF-␣ secretion as determined by ELISA. In the presence of RASFib, IFN-␥ and TNF-␣ secretion were significantly increased when compared with pure T cell cultures for both Tresp and TCD4T. At the same time, IFN-␥ and TNF-␣ secretion of Tresp remained significantly higher when compared with TCD4⫹ T cells, indicating that the suppressive action of Treg on Tresp cytokine secretion is preserved. ⴱ, p ⬍ 0.05 vs TCD4T (4⫹); †, p ⬍ 0.05 vs Treg (25⫹); ¥, p ⬍ 0.05 vs same cell population in the absence of RASFib. C, Potency of inhibition exerted by Treg on the proliferation and IFN-␥ and TNF-␣ secretion of Tresp, calculated as [1 ⫺ (proliferation or cytokine secretion of TCD4T/proliferation or cytokine secretion of Tresp)]. In the presence of RASFib no inhibition of proliferation was observed; at the same time, the suppressive potency of Treg on Tresp cytokine secretion was significantly increased. ⴱ, p ⬍ 0.05 vs conditions in the absence of RASFib.

TCD4T are stimulated with anti-CD3 in the presence but not in the absence of RASFib. Effect of RASFib on the per cell suppressive action of Treg The increased Treg suppressive potency on cytokine secretion observed in the presence of RASFib may be attributable, at least in part, to increased Treg proportions secondary to Treg proliferation. Terefore, we next sought to examine the effect of RASFib on the per cell suppressive potency of Treg. To this end, TCD4T from six healthy donors were cultured in the presence or absence of RASFib as described above, harvested on the fifth day of culture, and sorted by flow cytometry to isolate CD25⫹/bright cells. The regulatory potency of these CD25⫹/bright cells was tested in suppression assays free of RASFib, using freshly isolated autologous CD4⫹CD25⫺ cells as Tresp and following the protocol described in the preceding sections. Freshly isolated autologous CD4⫹CD25⫹ Treg served as controls. Accordingly, the following autologous combinations were assayed: (1) Treg precultured in the presence of RASFib with freshly isolated Tresp; (2) Treg precultured in the absence of RASFib with freshly isolated Tresp; and (3) freshly isolated Treg with freshly

isolated Tresp. Proliferation was determined by [3H]thymidine uptake, and cytokine secretion was assayed by ELISA. The suppressive potency of Treg that had been precultured in the presence of RASFib was significantly higher when compared with Treg precultured in the absence of RASFib and with freshly isolated Tregs (Fig. 4). The potency of the two latter groups of Treg was comparable (Fig. 4). Remarkably, the increased suppressive potency induced by RASFib was apparent not only on Tresp cytokine secretion but also on Tresp proliferation (Fig. 4). That is, although Treg suppression of Tresp proliferation is not apparent in the presence of RASFib, these Treg remain potent suppressors of proliferation when harvested and re-tested in the absence of RASFib. This phenomenon is comparable to the reported effect of dendritic cells on Treg function (15). As an additional control, CD25bright cells were isolated from CD4⫹CD25⫺ Tresp cultured for 5 days in the presence or absence of RASFib. When tested in suppression assays with freshly isolated autologous CD25⫺ Tresp, these CD25bright T cells did not demonstrate any regulatory capacity, indicating that Treg are not generated de novo in this system.

The Journal of Immunology

FIGURE 3. FoxP3 expression on activated T cells, cultured in the absence or presence of RASFib. Total CD4⫹ T cells (TCD4T), isolated Tresp (CD4⫹CD25⫺), or isolated Treg (CD4⫹CD25⫹) were stimulated with soluble anti-CD3 IgE mAb and cultured for 5 days in the absence (A) or presence of RASFib (B). Dot plots of CFSE-labeled cells double-stained with an allophycocyanin-labeled anti-FoxP3 mAb show an up-regulated FoxP3 expression on activated cells. In the presence of RASFib, up-regulation was seen not only on activated TCD4T and Tresp but also on proliferating Treg cells (B). Subsequently, the MFI of FoxP3 expression remained higher among proliferating isolated Treg cocultured with RASFib, when compared with proliferating Tresp (B). Remarkably, two populations of Foxp3-expressing cells were observed among stimulated TCD4T in the presence (B), but not in the absence (A), of RASFib: most cells expressed an up-regulated FoxP3 level with an intensity that was comparable to that observed in stimulated Tresp; a minor population (⬃10%) demonstrated a higher Foxp3 expression, similar to that observed in stimulated Treg. In contrast, FoxP3 expression was homogeneous among proliferating Tresp in the presence of RASFib (B).

Neutralization of IL-15 significantly attenuates the effect of RASFib on the equilibrium between Treg and Tresp We have previously reported that constitutively expressed IL-15 on the surface of RASFib is biologically active on cocultured T

8273 lymphocytes through direct cell contact (5). Therefore, it was important to investigate the role of IL-15 as a mediator of the observed action of RASFib on Treg function. To this end, a panel of IL-15-neutralizing agents was tested in cocultures of fibroblasts with TCD4T, Treg, or Tresp. The up-regulated responses of TCD4T and of Tresp observed in coculture with RASFib were significantly attenuated by IL-15 blocking agents (Fig. 5, A and B) but not by isotype control or binding control anti-HLA class I (W6/32) or by anti-HLA class II (DR, DP, DQ) Abs. Furthermore, in this setting the proliferation rate of Tresp was significantly higher when compared with TCD4T (Fig. 5, A and B). That is, in coculture with RASFib and in the presence of IL-15-neutralizing agents, the inhibitory effect of Treg on Tresp proliferation was readily observed. Additionally, neutralization of IL-15 abrogated the preproliferative effect of RASFib on Treg (Fig. 5B). In parallel, neutralization of IL-15 reversed the effect of RASFib on TCD4T and Tresp IFN-␥ and TNF-␣ secretion (Fig. 5B) and restored the potency of cytokine inhibition to that observed in the absence of RASFib (Fig. 5C). This indicates that constitutive RASFib IL-15 expression plays an important role in driving the Tresp and Treg proliferation observed in T cell/ RASFib cocultures, and in augmenting the inhibitory potency of Treg on Tresp cytokine secretion. Increased proportions of CD4⫹CD25⫹CD127⫺FoxP3⫹ T cells are found in the peripheral blood of early RA patients and in the synovial fluid of RA patients We were next interested in analyzing the characteristics of Treg and Tresp present in the peripheral blood of early RA (RAPB) and the synovial fluid of RA patients (RASF), and studying their behavior when stimulated through their TCR in the presence or absence of RASFib. Data were compared against Treg and Tresp from the peripheral blood of healthy controls (HCPB). Additionally, Treg and Tresp from early RA patients who achieved disease remission after follow-up were also studied (RAPB-R). When compared with HCPB CD4⫹ T cells (n ⫽ 30), the proportion of CD25⫹CD127⫺ T cells was significantly increased among RAPB CD4⫹ T cells (n ⫽ 30) (mean ⫾ SD, 9.8 ⫾ 2.7 vs 6.4 ⫾ 2.1, p ⬍ 0,05; range, 8.9 –16.3 vs 4.1– 8.6) (Fig. 6A). These RAPB CD4⫹CD25⫹CD127⫺ T cells expressed FoxP3 together with high levels of IL-15R␣, but they did not express the activation marker CD69 (Fig. 6, B and C). There was no difference in the amount of FoxP3 expression between RAPB CD4⫹CD25⫹ CD127⫺ and HCPB CD4⫹CD25⫹CD127⫺ T cells, as determined by flow cytometry (Fig. 6B). Importantly, reexamination of 10

FIGURE 4. Effect of RASFib on the per cell suppressive potency of Treg. Total CD4 T cells were cultured in the presence or absence of RASFib, harvested on the fifth day of culture, and sorted by flow cytometry to isolate CD25⫹/bright cells. The regulatory potency of these CD25⫹/bright cells was tested in suppression assays free of RASFib, using freshly isolated autologous CD4⫹CD25⫺ cells as Tresp, at different Treg/Tresp ratios. Freshly isolated autologous CD4⫹CD25⫹ Treg served as controls. Accordingly, the following autologous combinations were assayed: (1) Treg precultured in the presence of RASFib with freshly isolated Tresp (RASFibTreg/FrIsolTresp); (2) Treg precultured in the absence of RASFib with freshly isolated Tresp (NoRASFibTreg/FrIsolTresp); (3) freshly isolated Treg with freshly isolated Tresp (FrIsolTreg/FrIsolTresp). Shown is the inhibition potency of Treg on Tresp proliferation, IFN-␥ and TNF-␣ secretion at each tested ratio, calculated as in Fig. 1D. Each point represents the mean and SD of six healthy subjects per group. ⴱ, p ⬍ 0.05 vs suppression assays performed with cocultures of freshly isolated Treg with freshly isolated Tresp.

8274

EFFECT OF RA SYNOVIAL FIBROBLAST IL-15 EXPRESSION ON Treg

FIGURE 5. Effect of IL-15 neutralizing agents on RASFib-induced modifications of the Treg/Tresp balance. A, Total CFSE-labeled CD4⫹ T cells or Tresp (CD4⫹CD25⫺) cells were stimulated with anti-CD3IgE mAb and cocultured for 5 days with RASFib, in the presence of plain culture medium, or medium supplemented with IL-15-neutralizing agents, or isotype control agents or binding control mAb (anti-HLA class I). Results are compared with T cells cultured in the absence of RASFib. Representative flow cytometry histograms of CFSE labeled cells are shown. Open histograms represent proliferation of Tresp (CD4⫹CD25⫺) and filled histograms represent proliferation of total CD4⫹ T cells. Numbers in gray represent the percentage of proliferating total CD4⫹ T cells and numbers in black the percentage of proliferating Tresp. B, Total CFSE-labeled CD4⫹ T cells, Tresp (CD4⫹CD25⫺), or Treg (CD4⫹CD25⫹) were stimulated with anti-CD3IgE mAb and cultured for 5 days in the presence of RASFib, with or without IL-15-neutralizing agents or appropriate isotype or binding controls. Results are compared with T cells cultured in the absence of RASFib. Bar histograms show the proliferation rate of each cell population (TCD4T, Tresp, and Treg) as determined by flow cytometry of CFSE-labeled cells, together with IFN-␥ and TNF-␣ secretion, as determined by ELISA. Each bar represents the mean and SD of 30 subjects. ⴱ, p ⬍ 0.05 vs conditions without RASFib. C, Potency of the inhibition exerted by natural proportions of Treg on the proliferation and cytokine secretion of Tresp, calculated as described in Fig. 2. ⴱ, p ⬍ 0.05 vs conditions without RASFib.

early RA patients at 12 mo, once remission of disease activity had been achieved with treatment (methotrexate and low-dose prednisone), demonstrated that the proportion of circulating CD4⫹ CD25⫹CD127⫺ T cells was no longer different from that of healthy controls (Fig. 6A). The proportion of CD25⫹CD127⫺ T cells was even higher among synovial fluid CD4⫹ T cells of RA patients (n ⫽ 30) (20.5 ⫾ 5.4; p ⬍ 0.01 vs RAPB and vs HCPB) (Fig. 6A). Additionally, the expression level of FoxP3 among these cells was significantly higher when compared with HCPB and RAPB CD4⫹CD25⫺CD127⫹ T cells ( p ⬍ 0.05) (Fig. 6B). Interestingly, most of the RASF CD4⫹CD25⫹CD127⫺ T cells (⬎95%) were CD69-negative, indicating that these cells are probably not activated Tresp (Fig. 6C). Treg from the synovial fluid of RA and the peripheral blood of early RA patients demonstrate an increased suppressive capacity Because the gold standard for Treg identification is demonstration of their inhibitory capacity (34), we next examined the inhibitory function of Treg in RA. The results were compared among the above mentioned four groups: HCPB, RAPB,

RAPB-R, and RASF T cells. Inhibition assays were performed following two different approaches: (1) the regulatory function of natural proportions of Treg in RA was inferred by comparing the responses of anti-CD3-stimulated total CD4⫹ vs CD25⫹ depleted CD4⫹ T cells, and (2) additionally, the per cell potency of RATreg was assessed in cocultures of magnetically sorted CD4⫹CD25⫹ Treg with CD4⫹CD25⫺ Tresp, established at different Treg/Tresp ratios. We thought it interesting to first test the suppressive function of the naturally increased proportion of CD4⫹CD25⫹FoxP3⫹ T cells present among RAPB and RASF CD4⫹ T cells. Of note, TCD4T from RAPB and RASF showed a lower proliferative response to anti-CD3 mAbs when compared with TCD4T from HCPB (Fig. 7A). In contrast, RAPB and RASF CD4⫹CD25⫺ Tresp demonstrated higher proliferation rates when compared with HCPB Tresp (Fig. 7A). Subsequently, the calculated inhibitory potency of Treg on Tresp proliferation was significantly higher for both RAPB and RASF cells when compared with HCPB (Fig. 7D). This suggests that the low proliferative responses of RAPB and RASF TCD4T in response to anti-CD3 mAbs are attributable to the superior suppressive potency of RA Treg.

The Journal of Immunology

8275

FIGURE 6. Frequency and phenotype of CD4⫹ CD25⫹CD127⫺ T cells in RA. A, Representative flow cytometry dot plots, gated for CD4, demonstrate the frequency of CD25⫹CD127⫺ CD4⫹ T cells present in HCPB, RAPB, and RASF. Below, a bar histogram shows the frequencies of CD4⫹CD25⫹CD127⫺ T cells and of FoxP3⫹ T cells in HCPB, RAPB, and RASF together with RAPB-R (peripheral blood of RA patients who have achieved remission). Each bar represents the mean and SD of 30 subjects per group. ⴱ, p ⬍ 0.05 vs HCPB; †, p ⬍ 0.05 vs RAPB. B, Bar histograms show the MFI as determined by flow cytometry, of FoxP3 and IL-15R␣ expression on CD4⫹CD25⫹CD127⫺ and CD4⫹CD25⫺CD127⫹ T cells. Each bar represents the mean and SD of 30 subjects per group. ⴱ, p ⬍ 0.05 vs CD4⫹CD25⫺CD127⫹ T cells; †, p ⬍ 0.05 vs HCPB; ¥, p ⬍ 0.05 vs RAPB. C, Representative flow cytometry dot plots show that most of the CD4⫹CD25⫹ T cells present not only in HCPB and RAPB but also in RASF are negative for CD69 expression.

In parallel, IFN-␥ and TNF-␣ secretion were higher for TCD4T and also for Tresp isolated from both RAPB and RASF, when compared with the corresponding populations isolated from HCPB (Fig. 7, B and C). That is, despite their low proliferative response, stimulated RAPB and RASF TCD4T secrete more IFN-␥ and TNF-␣ than do HCPB TCD4T. At the same time, the potency of inhibition attributable to the action of Treg on Tresp cytokine secretion was significantly increased for both RAPB- and RASFderived T cells, when compared with HCPB (Fig. 7D). When early RA patients were reexamined after disease activity had been controlled with treatment (n ⫽ 10), the functional behavior of total and CD25⫹-depleted CD4 T cells together with the calculated potency of Treg suppression were not different from healthy controls (Fig. 7). The observed increased suppressive action of Treg in RA may be related, at least in part, to the higher proportion of CD4⫹ CD25⫹FoxP3⫹ T cells present in the peripheral blood and the synovial fluid of RA patients. Therefore, we next sought to determine the suppressive potency of RA Treg on a per cell basis. To this end, isolated CD4⫹CD25⫹FoxP3⫹ T cells (Treg) were cocultured with CD4⫹CD25⫺ T cells (Tresp) at different Treg-to-Tresp ratios (1:1, 1:2, 1:5, 1:10, and 1:20). At the highest tested ratio (1:1), Treg from all four studied groups were highly suppressive of Tresp proliferation and IFN-␥ and TNF-␣ secretion, and no significant differences were detected among the groups (Fig. 8A). At a 1:2 and 1:5 ratio, RASF Treg were significantly more suppressive than RAPB Treg, HCPB Treg, and RAPB-R Treg, while no differences were observed among the latter three sets of subjects (Fig. 8A). At 1:10 and 1:20 ratios, the suppressive potency of RAPB Treg was significantly higher when compared with HCPB

Treg, as was the suppresive potency of RASF Treg (Fig. 8A). This indicates that not only RASF but also RAPB Treg have an increased suppressive capacity on a per cell basis when compared with HCPB Treg. We can only speculate that in vivo activation of RA Treg is responsible for this increased suppressive capacity. The properties of RAPB-R Treg were not different from HCPB Tregs. Next, isolated RA Treg were cocultured with isolated RA Tresp at a 1:10 Treg-to-Tresp ratio in the presence of RASFib. The effect of RASFib on RAPB and RASF Treg function was similar to that exerted on HCPB Treg properties. In this setting, inhibition of RA Tresp proliferation was not apparent, while inhibition of RA Tresp IFN-␥ and TNF-␣ secretion were maintained with superior potency (Fig. 8B). Neutralization of IL-15 significantly down-modulated the effect of RASFib (Fig. 8B).

Discussion We have herein shown that in the presence of RASFib, the anergic state of human CD4⫹CD25⫹ Treg is abrogated, and their suppressive action on CD4⫹CD25⫺ Tresp proliferation is not apparent; at the same time, Treg inhibition of Tresp proinflammatory cytokine secretion is not only preserved but potentiated. In parallel, RASFib mediate an up-regulated TNF-␣ and IFN-␥ response of CD4⫹ CD25⫺ Tresp. Subsequently, total CD4⫹ T cells, containing natural proportions of Treg and Tresp, secrete an increased amount of pathogenic cytokines when cocultured with RASFib, despite the presence of proliferating Treg with superior regulatory potency. In summary, RASFib exert a dual action on the functional equilibrium between Treg and Tresp, by potentiating the suppressive effect of Treg while augmenting the proinflammatory action of

8276

EFFECT OF RA SYNOVIAL FIBROBLAST IL-15 EXPRESSION ON Treg

FIGURE 7. Estimation of the regulatory function of natural proportions of CD4⫹CD25⫹ Treg in RA. The function of naturally occurring CD4⫹CD25⫹ Treg present at physiological rates in RA peripheral blood was assessed by testing the response of total CD4⫹ against CD25-depleted CD4⫹ T cells. Results were compared among four groups: HCPB, RAPB, RAPB-R, and RASF. Shown are proliferation (as determined by CFSE dilution) (A), IFN-␥ secretion (B), and TNF-␣ secretion (C) (as determined by ELISA) of total CD4⫹ T cells (4⫹), CD25⫹-depleted CD4⫹ T cells (25⫺), and CD4⫹CD25⫹ T cells (25⫹), stimulated for 5 days with an IgE anti-CD3 mAb. Each point represents the mean and SD of 30 subjects per group. In A–C, ⴱ, p ⬍ 0.05 vs total CD4⫹ T cells of same group of patients, †, p ⬍ 0.05 vs HCPB; ¥, p ⬍ 0.05 vs RAPB. In D, the inhibitory potency of CD4⫹CD25⫹ T cells was calculated as [1 ⫺ (proliferation or cytokine secretion of TCD4T/proliferation or cytokine secretion of CD4⫹CD25⫺ T cells)]. ⴱ, p ⬍ 0.05 vs HCPB.

Tresp; the result is a shift of the Treg/Tresp balance toward a proinflammatory state. Experiments with IL-15-neutralizing agents and with Transwell inserts indicate that surface RASFib IL-15 plays a pivotal role in this effect. Of note, this alteration of the Treg/Tresp equilibrium is not observed in the presence of OASFib or dermal fibroblasts, which do not constitutively express surface IL-15 (5). Experiments with neutralizing anti-MHC class I and class II Abs ruled out an implication of Ag presentation in this effect. Consistent with previous reports (35–38), we observed an induced FoxP3 expression in stimulated Tresp that did not seem to be associated with acquisition of regulatory properties. Additionally, proliferating Tregs in the presence of RASFib demonstrated an up-regulated FoxP3 expression together with a superior regulatory potency. This is in agreement with published data indicating that stimulated Treg show an enhanced suppressive capacity, and that their increased FoxP3 expression remains higher when compared with induced FoxP3 levels in stimulated Tresp (35). Importantly, when Treg that had been cocultured with RASFib were harvested and retested in suppression assays free of RASFib, they demonstrated a superior per cell suppressive potency not only on Tresp cytokine secretion but also on Tresp proliferation. Other cell lineages have been described to modify the properties of Tregs in a comparable manner (12, 13, 15–17). Direct interaction with mature, activated bone marrow-derived dendritic cells, but not with splenic dendritic cells expressing equivalent levels of costimulatory molecules, reverts the anergic state of Treg (15, 16) and interferes with Treg inhibition of Tresp proliferation while preserving their suppressive action on Tresp IL-2 secretion (15). However, when Treg that have been cocultured with dendritic cells are harvested and tested in suppression assays free of dendritic

cells, they demonstrate a potent suppressor action on Tresp proliferation (15). Of note, B cells are unable to expand Treg despite their potent APC function (15, 16). In fact, the role of costimulation on the bone marrow-derived dendritic cell-induced proliferation of Treg is controversial; alternatively, proinflammatory cytokines have been presented as strong candidates (12, 13, 17). Additionally, cytokines derived from activated monocytes present in the RA joint have been reported to interfere with Treg function (39). IL-15, initially described as a T cell growth factor (40, 41), acts through a heterotrimeric receptor consisting of a specific highaffinity binding ␣-chain (IL-15R␣) plus the IL-2 receptor signaling subunits ␤- and common ␥-chain (42, 43). Subsequently, IL-2 and IL-15 share several effects. Both stimulate the proliferation of activated CD4⫹ and CD8⫹ T cells, of B cells stimulated through their BCR, and of NK cells (42, 43). Additionally, IL-2 and IL-15 have some divergent effects: IL-2 promotes the elimination of activated T cells (44 – 46), whereas IL-15 suppresses T cell apoptosis (47). In mice, the development and survival of CD4⫹CD25⫹ Tregs is highly dependent on IL-2 but is independent of IL-15 (48). CD4⫹CD25⫹ Treg potently suppress both proliferation and cytokine secretion of CD4⫹CD25⫺ Tresp (7–9). A hallmark of CD4⫹CD25⫹ Tregs is their inability to proliferate themselves when stimulated through the TCR (7–9), because they lack the capacity to transcribe IL-2 (11). Nevertheless, murine CD4⫹ CD25⫹ Treg do readily proliferate when stimulated via their TCR in the presence of the exogenous cytokines IL-2 or IL-4, but not in the presence of IL-6, IL-7, IL-9, IL-10, or IL-15 (48). Treg proliferation triggered by IL-2 or IL-4 interferes with the inhibition of Tresp proliferation (11); interestingly, at the same time, Treg suppression of IL-2 production is not only spared but significantly

The Journal of Immunology

8277

FIGURE 8. Evaluation of the functional capacity of CD4⫹CD25⫹ Treg in RA on a per cell basis. A, Isolated CD4⫹CD25⫹ T cells (Treg) were cocultured for 5 days with CFSE-labeled CD4⫹CD25⫺ T cells (Tresp) at different Treg-to-Tresp ratios (1:1, 1:2, 1:5, 1:10, and 1:20). Shown is the inhibition potency of Treg on Tresp proliferation and IFN-␥ and TNF-␣ secretion at each tested ratio, calculated as in Fig. 1D. Results were compared among HCPB, RAPB, RAPB-R, and RASF T cells. Each bar represents the mean and SD of 30 subjects per group. ⴱ, p ⬍ 0.05 vs HCPB. B, Isolated CD4⫹ CD25⫹FoxP3⫹ T cells (Treg) were cocultured for 5 days with CFSE-labeled CD4⫹CD25⫺ T cells (Tresp) at a 1:10 Treg-to-Tresp ratio in the absence of presence of RASFib, with or without a neutralizing anti-IL-15 mAb. The inhibition potency of Treg on Tresp proliferation and IFN-␥ and TNF-␣ secretion is shown. Results were compared among HCPB, RAPB, RAPB-R, and RASF T cells. Each bar represents the mean and SD of 30 subjects per group. ⴱ, p ⬍ 0.05 vs cells cultured in the absence of RASFib; †, p ⬍ 0.05 vs HCPB.

enhanced (11). In contrast with murine Treg (48), human CD4⫹ CD25⫹ Treg proliferate and acquire an enhanced suppressor capacity when stimulated via their TCR in the presence of exogenous IL-15 (20, 21); in fact, Koenen et al. reported that IL-15 is a superior growth factor for human Treg (21). Additionally, we have herein described for the first time that IL-15R␣ is highly expressed on human Treg, suggesting a role for IL-15 in Treg biology. IL-15 is expressed intracellularly by nonlymphoid cells such as monocyte-macrophages (42, 43), by dendritic cells (42, 43), and also by fibroblasts (43, 49). Additionally, whereas IL-2 acts in a soluble form, IL-15 is expressed on the cell surface and acts through direct cell contact (43). Surface IL-15 is constitutively present and physiologically active on fibroblasts from some locations. IL-15 on fibroblasts from human spleen regulates NK cell differentiation from blood CD34⫹ progenitors (50), and IL-15 on bone marrow fibroblast-like stromal cells contributes to T cell recruitment and expansion in aplastic anemia (51). We previously described that constitutively expressed IL-15 on the surface of RASFib induces T cell activation and cytokine secretion through direct cell contact (5); in contrast, resting OASFib or dermal fibroblasts do not activate T cells, consistent with their lack of constitutive surface IL-15 expression (5). This illustrates how, as described by C. D. Buckley, synovial fibroblasts are key players in favoring the persistence of the inflammatory joint infiltrate (52) and orchestrate the switch from acute resolving to chronic persistent arthritis. At the same time, it stresses the fact that “fibroblasts from different anatomical sites have diverse phenotype and function, secrete distinct patterns of cytokines and chemokines, and

condition the intrinsic susceptibility of the organs to inflammatory insults” (2, 52). In the present study we have extended our previous observations by showing that RASFib IL-15 expression is able to break the anergic state of CD4⫹CD25⫹ Treg and modify the Treg/Tresp equilibrium. This effect is not apparent in the presence of OASFib or dermal fibroblasts that do not express surface IL-15. In agreement with these findings, Clark and Kupper have described that human Treg cocultured with dermal fibroblasts do not proliferate unless exogenous IL-15 is added to the medium (22). Additionally, our data indicate that increased proportions of CD4⫹CD25⫹CD127⫺ Treg with superior suppressive potency are present in the peripheral blood and the synovial fluid of RA patients. However, this enhanced regulatory activity is not able to overcome the increased secretion of proinflammatory mediators released by preactivated CD4⫹CD25⫺ Tresp. That is, RA patients demonstrate an altered Treg/Tresp equilibrium in vivo. It is widely accepted that the frequency and the suppressor potency of CD4⫹CD25⫹ Treg found in the synovial fluid of RA patients are significantly elevated when compared with peripheral blood (25, 53–56), and our results are consistent with previous observations. A question that has been asked by several investigators (25, 53, 54) is why an active joint inflammation persists in RA despite the presence of elevated frequencies of CD4⫹CD25⫹ T cells with an enhanced suppressive capacity. Our experimental results help to shed some light on this matter. In vivo, RASFTL are in contact with the lining layer of the synovial membrane where hyperplastic synovial fibroblasts are abundant (23). The proliferation of the RA synovial membrane leads to the formation of villi,

8278

EFFECT OF RA SYNOVIAL FIBROBLAST IL-15 EXPRESSION ON Treg

and this enormously augments the contact surface between the lining layer of the synovial membrane on one side and the synovial fluid and its components on the opposite side (23). Salmon et al. (4) demonstrated that decreased apoptosis of RA synovial fluid T cells in vivo is due to direct contact with RA synovial fibroblasts within the joint space. Likewise, the data presented herein suggest that in vivo, direct contact of T cells with RASFib may induce an alteration of the Treg/Tresp equilibrium similar to that observed in our ex vivo experiments, and are important contributors to the increased potency of Treg present in RA synovial fluid. In contrast, previous studies on the frequency and on the functional capacity of circulating CD4⫹CD25⫹ Treg in RA have yielded conflicting results (10, 53, 57, 58). In comparison with healthy controls, increased (53) as well as decreased (58) or equivalent (57) PB Treg frequencies have been reported by different authors. Additionally, comparable (58) as well as decreased (10, 57) regulatory activities relative to HCPB Tregs, have been described in RA patients; furthermore, reported data on the nature of the PB Treg dysfunction in RA are conflicting: both an isolated alteration of cytokine suppression (57) and a combined impairment of Tresp proliferation and cytokine suppression have been observed (10). Several factors may account for these divergent results, including criteria and methods chosen to define and isolate Treg, experimental strategies used for assessing Treg function, potency of the stimulating conditions, clinical activity of RA patients, duration of the disease, and treatment of patients with various drugs. In summary, the criteria and the experimental conditions required to analyze the frequency and function of human CD4⫹CD25⫹ Treg have not been standardized, and therefore results may vary among different studies. In fact, phenotypical characterization of human CD4⫹CD25⫹ FoxP3⫹ Treg is difficult to achieve (34). In contrast with murine cells, the line between human CD25⫹ and CD25⫺ CD4⫹ T lmphocytes is not easy to draw since humans demonstrate a smooth gradation of intensity from negative to highly positive CD25⫹ T cells (59). Also, human CD4⫹ T cells do not display an optimal correlation between the expression of CD25 and FoxP3: most of the human CD4⫹CD25high cells are Foxp3⫹, but in addition a variable percentage of human CD25int cells expresses Foxp3 at lower although substantial levels (34). Subsequently, the gold standard for Treg cell characterization is demonstration of their inhibitory action. Importantly, results of suppression assays are not only related to the purity of isolated Treg but also depend on the potency of the stimulus and on the Treg/Tresp ratio examined. Stronger stimuli tend to mask the effect of Treg and require high Treg/ Tresp ratios to detect Treg-mediated suppression. Conversely, less potent stimuli allow the detection of inhibition at lower Treg/Tresp ratios and facilitate the study of Tregs with lower suppressive potencies. The superior function of RASFTreg can be appreciated under less stringent conditions and using stronger stimuli when compared with RAPB or HCPB Treg; this may explain why more consistent results have been published in the literature on this matter. We observed by flow cytometry that an increased frequency of CD4⫹CD25⫹CD127⫺FoxP3⫹ T cells is present in the peripheral blood of early, untreated RA patients when compared with healthy controls, consistent with data reported by Van Amelsfort et al. on established RA patients (53). Using magnetic beads, we carefully isolated a CD25⫹ population that was CD127⫺, FoxP3⫹, and expressed high levels of IL-15R␣. Suppression tests with submaximal stimuli confirmed an optimal regulatory capacity that was apparent for Treg isolated from all three patient groups studied: RAPB, RAPB-R, and RASF. Furthermore, we observed that the

suppressive potency of RAPB Treg was significantly superior to that of HCPB Treg, whereas the suppressive strength of RASF Treg was maximal. Of note, when high Treg/Tresp ratios were used in the presence of submaximal stimuli, differences in suppressor capacity among the tested Treg populations were not appreciated. With decreasing Treg/Tresp ratios, however, differences among HCPB Treg, RAPB Treg, and RASF Treg became apparent. Interestingly, Tregs isolated from the PB of RA patients that achieved remission were functionally comparable with Tregs from the PB of HC, indicating that control of disease activity is associated with a reversal of the in vivo altered Treg/Tresp equilibrium. Discrepancies of our data with a previous report on the frequency and function of PB Treg from early, untreated, RA patients (58) may be related to the technical matters discussed above. In summary, RASFib IL-15 exerts a dual action on the equilibrium between Treg and Tresp, by potentiating the suppressive effect of Treg while augmenting the proinflammatory action of Tresp, and this results in a shift of the Treg/Tresp balance toward a proinflammatory state. Additionally, Treg with superior suppressive potency are present in the peripheral blood and the synovial fluid of RA patients, but this enhanced immunoregulatory activity is not able to overcome the increased secretion of pathogenic cytokines by RA Tresp, indicating that RA patients demonstrate an altered Treg/Tresp equilibrium in vivo that is comparable to that induced in healthy controls upon coculture with RASFib. Therapeutic strategies directed to restoring this altered equilibrium and/or modifying the action of local IL-15 in the joint may be useful for patients with RA.

Disclosures The authors have no financial conflicts of interest.

References 1. Firestein, G. S. 1996. Invasive fibroblast-like synoviocytes in rheumatoid arthritis: passive responders or transformed aggressors? Arthritis Rheum. 39: 1781–1790. 2. Buckley, C. D., D. Pilling, J. M. Lord, A. N. Akbar, D. Scheel-Toeller, and M. Salmon. 2001. Fibroblasts regulate the switch from acute resolving to chronic persistent inflammation. Trends Immunol. 22: 199 –204. 3. Scott, S., F. Pandolfi, and J. T. Kurnick. 1990. Fibroblasts mediate T cell survival: a proposed mechanism for retention of primed T cells. J. Exp. Med. 172: 1873–1876. 4. Salmon, M., D. Scheel Toellner, A. P. Huissoon, D. Pilling, N. Shamsadeen, H. Hyde, A. D. D’Angeac, P. A. Bacon, P. Emery, and A. N. Akbar. 1997. Inhibition of T cell apoptosis in the rheumatoid synovium. J. Clin. Invest. 99: 439 – 446. 5. Miranda-Carus, M. E., A. Balsa, M. Benito-Miguel, C. Perez de Ayala, and E. Martin-Mola. 2004. IL-15 and the initiation of cell contact-dependent synovial fibroblast-T lymphocyte cross-talk in rheumatoid arthritis: effect of methotrexate. J. Immunol. 173: 1463–1476. 6. Vallejo, A. N., H. Yang, P. A. Klimiuk, C. M. Weyand, and J. J. Goronzy. 2003. Synoviocyte-mediated expansion of inflammatory T cells in rheumatoid synovitis is dependent on CD47-thrombospondin 1 interaction. J. Immunol. 171: 1732–1740. 7. Sakaguchi, S. 2004. Naturally arising CD4⫹ regulatory T cells for immunologic self-tolerance and negative control of immune responses. Annu. Rev. Immunol. 22: 531–562. 8. Shevach, E. M. Regulatory/suppressor T cells in health and disease. 2004. Arthritis Rheum. 50: 2721–2724. 9. Thornton, A. M., and E. M. Shevach. 1998. CD4⫹CD25⫹ immunoregulatory T cells suppress polyclonal T cell activation in vitro by inhibiting interleukin 2 production. J. Exp. Med. 188: 287–296. 10. Valencia, X., G. Stephens, R. Goldbach-Mansky, M. Wilson, E. M. Shevach, and P. E. Lipsky. 2006. TNF downmodulates the function of human CD4⫹CD25high T-regulatory cells. Blood 108: 253–2661. 11. Thornton, A. M., E. E. Donovan, C. A. Piccirillo, and E. M. Shevach. 2004. Cutting edge: IL-2 is critically required for the in vitro activation of CD4⫹CD25⫹ T cell suppressor function. J. Immunol. 172: 6519 – 6523. 12. Pasare, C., and R. Medzhitov. 2003. Toll pathway-dependent blockade of CD4⫹CD25⫹ T cell-mediated suppression by dendritic cells. Science 299: 1033–1036. 13. Fehervari, Z., and S. Sakaguchi. 2004. Control of Foxp3⫹CD25⫹CD4⫹ regulatory cell activation and function by dendritic cells. Int. Immunol. 16: 1769 –1780. 14. Ruprecht, C. R., M. Gattorno, F. Ferlito, A. Gregorio, A. Martini, A. Lanzavecchia, and F. Sallusto. 2005. Coexpression of CD25 and CD27 identifies FoxP3⫹ regulatory T cells in inflamed synovia. J. Exp. Med. 201: 1793–1803.

The Journal of Immunology 15. Brinster, C., and E. M. Shevach. 2005. Bone marrow-derived dendritic cells reverse the anergic state of CD4⫹CD25⫹ T cells without reversing their suppressive function. J. Immunol. 175: 7332–7340. 16. Yamazaki, S., T. Iyoda, K. Tarbell, K. Olson, K. Velinzon, K. Inaba, and R. M. Steinman. 2003. Direct expansion of functional CD25⫹CD4⫹ regulatory T cells by antigen-processing dendritic cells. J. Exp. Med. 198: 235–247. 17. Kubo, T., R. D. Hatton, J. Oliver, X. Liu, C. O. Elson, and C. T. Weaver. 2004. Regulatory T cell suppression and anergy are differentially regulated by proinflammatory cytokines produced by TLR-activated dendritic cells. J. Immunol. 173: 7249 –7258. 18. Sutmuller, R. P., M. H. den Brok, M. Kramer, E. J. Bennink, L.W. Toonen, B. J. Kullberg, L. A. Joosten, S. Akira, M. G. Netea, and G. J. Adema. 2006. Toll-like receptor 2 controls expansion and function of regulatory T cells. J. Clin. Invest. 116: 485– 494. 19. Peng, G., Z. Guo, Y. Kiniwa, K. S. Voo, W. Peng, T. Fu, D. Y. Wang, Y. Li, H. Y. Wang, and R. F. Wang. 2005. Toll-like receptor 8-mediated reversal of CD4⫹ regulatory T cell function. Science 309: 1380 –1384. 20. Dieckmann, D., H. Plottner, S. Berchtold, T. Berger, and 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. 21. Koenen, H. J., E. Fasse, and I. Joosten. 2003. IL-15 and cognate antigen successfully expand de novo-induced human antigen-specific regulatory CD4⫹ T cells that require antigen-specific activation for suppression. J. Immunol. 171: 6431– 6441. 22. Clark, R. A., and T. S. Kupper. 2007. IL-15 and dermal fibroblasts induce proliferation of natural regulatory T cells isolated from human skin. Blood 109: 194 –202. 23. Fassbender, H. G. 2002. Joint process in RA. In Pathology and Pathobiology of Rheumatic Diseases, 2nd Ed. Springer, New York, pp. 62–106. 24. Fox, D. A. 1997. The role of T cells in the immunopathogenesis of rheumatoid arthritis: new perspectives. Arthritis Rheum. 40: 598 – 609. 25. Leipe, J., A. Skapenko, P. E. Lipsky, and H. Schulze-Koops. 2005. Regulatory T cells in rheumatoid arthritis. Arthritis Res. Ther. 7: 93–99. 26. Arnett, F. C., S. M. Edworthy, D. A. Bloch, D. J. McShane, J. F. Fries, N. S. Cooper, L. A. Healey, S. R. Kaplan, M. H. Liang, H. S. Luthra, et al. 1988. The American Rheumatism Association 1987 revised criteria for the classification of rheumatoid arthritis. Arthritis Rheum. 31: 315–324. 27. Prevoo, M. L., M. A. van t Hof, H. H. Kuper, M. A. van Leeuwen, L. B. van de Putte, and P. L. van Riel. 1995. Modified disease activity scores that include twenty-eightjoint counts: development and validation in a prospective longitudinal study of patients with rheumatoid arthritis. Arthritis Rheum. 38: 44 – 48. 28. Balsa, A., L. Carmona, I. Gonzalez-Alvaro, M. A. Belmonte, X. Tena, R. Sanmarti, and the EMECAR Study Group. 2004. Value of disease activity score 28 (DAS28) and DAS28-3 compared to American College of Rheumatology-defined remission in rheumatoid arthritis. J. Rheumatol. 31: 40 – 46. 29. Saalbach, A, U. F. Haustein, U. Anderegg. 2000. A ligand of human thy-1 is localized on polymorphonuclear leukocytes and monocytes and mediates the binding to activated thy-1-positive microvascular endothelial cells and fibroblasts. J. Invest. Dermatol. 115: 882– 888. 30. van Lier, R. A., J. H. Boot, A. J. Verhoeven, E. R. de Groot, M. Brouwer, and L. A. Aarden. 1987. Functional studies with anti-CD3 heavy chain isotype switch-variant monoclonal antibodies: accessory cell-independent induction of interleukin 2 responsiveness in T cells by ␧-anti-CD3. J. Immunol. 139: 2873–2879. 31. Kim, Y., W. Maslinski, X. X. Zheng, G. H. Tesch, V. R. Kelley, and T. B. Strom. 1998. Targeting the IL-15 receptor with an antagonist IL-15/Fc protein blocks delayed type hypersensitivity. J. Immunol. 160: 5742–5748. 32. Seddiki, N., B. Santner-Nanan, J. Martinson, J. Zaunders, S. Sasson, A. Landay, M. Solomon, W. Selby, S. I. Alexander, R. Nanan, et al. 2006. Expression of interleukin (IL)-2 and IL-7 receptors discriminates between human regulatory and activated T cells. J. Exp. Med. 203: 1693–1700. 33. Liu, W., A. L. Putnam, Z. Xu-Yu, G. L. Szot, M. R. Lee, S. Zhu, P. A. Gottlieb, P. Kapranov, T. R. Gingeras, B. Fazekas de St Groth, et al. 2006. CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4⫹ Treg cells. J. Exp. Med. 203: 1701–1711. 34. Shevach, E. M. 2006. From vanilla to 28 flavors: multiple varieties of T regulatory cells. Immunity 25: 195–201. 35. Allan, S. E., S. Q. Crome, N. K. Crellin, L. Passerini, T. S. Steiner, R. Bacchetta, M. G. Roncarolo, and M. K. Levings. 2007. Activation-induced FOXP3 in human T effector cells does not suppress proliferation or cytokine production. Int. Immunol. 19: 345–354. 36. Tran, D. Q., H. Ramsey, and E. M. Shevach. 2007. Induction of FOXP3 expression in naive human CD4⫹FOXP3 T cells by T-cell receptor stimulation is transforming growth factor-␤ dependent but does not confer a regulatory phenotype. Blood 110: 2983–2990. 37. Morgan, M. E., J. H. van Bilsen, A. M. Bakker, B. Heemskerk, M. W. Schilham, F. C. Hartgers, B. G. Elferink, L. van der Zanden, R. R. de Vries, T. W. Huizinga,

8279

38.

39.

40.

41.

42. 43. 44.

45. 46. 47.

48.

49.

50.

51.

52.

53.

54.

55.

56.

57.

58.

59.

et al. 2005. Expression of FOXP3 mRNA is not confined to CD4⫹CD25⫹ T regulatory cells in humans. Hum. Immunol. 66: 13–20. Wang, J., A. Ioan-Facsinay, E. I. van der Voort, T. W. Huizinga, and R. E. Toes. 2007. Transient expression of FOXP3 in human activated nonregulatory CD4⫹ T cells. Eur. J. Immunol. 37: 129 –138. van Amelsfort, J. M., J. A. van Roon, M. Noordegraaf, K. M. Jacobs, J. W. Bijlsma, F. P. Lafeber, and L. S. Taams. 2007. Proinflammatory mediatorinduced reversal of CD4⫹CD25⫹ regulatory T cell-mediated suppression in rheumatoid arthritis. Arthritis Rheum. 56: 732–742. Grabstein, K. H., J. Eisenman, K. Shanebeck, C. Rauch, S. Srinivason, V. Fung, C. Beers, J. Richardson, M. A. Schoenborn, M. Ahdieh, et al. 1994. Cloning of a T cell growth factor that interacts with the ␤ chain of the interleukin-2 receptor. Science 264: 965–968. Burton, J. D., R. N. Bamford, C. Peters, A. J. Grant, G. Kurys, C. K. Goldman, J. Brennan, E. Roessler, and T. A. Waldmann. 1994. A lymphokine, provisionally designated interleukin T and produced by a human adult leukemia line, stimulates T-cell proliferation and the induction of lymphokine-activated killer cells. Proc. Natl. Acad. Sci. USA 91: 4935– 4939. Waldmann, T. A. 2006. The biology of interleukin-2 and interleukin-15: implications for cancer therapy and vaccine design. Nat. Rev. Immunol. 6: 595– 601. Fehniger, T. A., and M. A. Caligiuri. 2001. Interleukin 15: biology and relevance to human disease. Blood 97: 14 –32. Ku, C. C., M. Murakami, A. Sakamoto, J. Kappler, and P. Marrack. 2000. Control of homeostasis of CD8⫹ memory T cells by opposing cytokines. Science 288: 675– 678. Lenardo, M. J. 1996. Fas and the art of lymphocyte maintenance. J. Exp. Med. 183: 721–724. Maloy, K. J., and F. Powrie. 2005. Fueling regulation: IL-2 keeps CD4⫹ Treg cells fit. Nat. Immunol. 6: 1071–1072. Bulfone-Paus, S., D. Ungureanu, T. Pohl, G. Lindner, R. Paus, R. Ruckert, H. Krause, and U. Kunzendorf. 1997. Interleukin-15 protects from lethal apoptosis in vivo. Nat. Med. 3: 1124 –1128. Thornton, A. M., C. A. Piccirillo, and E. M. Shevach. 2004. Activation requirements for the induction of CD4⫹CD25⫹ T cell suppressor function. Eur. J. Immunol. 34: 366 –376. Kurowska, M., W. Rudnicka, E. Kontny, I. Janicka, M. Chorazy, J. Kowalczewski, M. Zio´łkowska, S. Ferrari-Lacraz, T. B. Strom, and W. Mas´lin´ski. 2002. Fibroblast-like synoviocytes from rheumatoid arthritis patients express functional IL-15 receptor complex: endogenous IL-15 in autocrine fashion enhances cell proliferation and expression of Bcl-xL and Bcl-2. J. Immunol. 169: 1760 –1767. Briard, D., D. Brouty-Boye´, B. Azzarone, and C. Jasmin. 2002. Fibroblasts from human spleen regulate NK cell differentiation from blood CD34⫹ progenitors via cell surface IL-15. J. Immunol. 168: 4326 – 4332. Wenxin, L., F. Jinxiang, W. Yong, L. Wenxiang, S. Wenbiao, and Z. Xueguang. 2005. Expression of membrane-bound IL-15 by bone marrow fibroblast-like stromal cells in aplastic anemia. Int. Immunol. 17: 429 – 437. Buckley, C. D. 2003. Michael Mason prize essay 2003: Why do leucocytes accumulate within chronically inflamed joints? Rheumatology (Oxford) 42: 1433–1444. van Amelsfort, J. M., K. M. Jacobs, J. W. Bijlsma, F. P. Lafeber, and L. S. Taams. 2004. CD4⫹CD25⫹ regulatory T cells in rheumatoid arthritis: differences in the presence, phenotype, and function between peripheral blood and synovial fluid. Arthritis Rheum. 50: 2775–2785. Cao, D., V. Malmstro¨m, C. Baecher-Allan, D. Hafler, L. Klareskog, and C. Trollmo. 2003. Isolation and functional characterization of regulatory CD25brightCD4⫹ T cells from the target organ of patients with rheumatoid arthritis. Eur. J. Immunol. 33: 215–223. Mo¨tto¨nen, M., J. Heikkinen, L. Mustonen, P. Isoma¨ki, R. Luukkainen, and O. Lassila. 2005. CD4⫹CD25⫹ T cells with the phenotypic and functional characteristics of regulatory T cells are enriched in the synovial fluid of patients with rheumatoid arthritis. Clin. Exp. Immunol. 140: 360 –367. Liu, M. F., C. R. Wang, L. L. Fung, L. H. Lin, and C. N. Tsai. 2005. The presence of cytokine-suppressive CD4⫹CD25⫹ T cells in the peripheral blood and synovial fluid of patients with rheumatoid arthritis. Scand. J. Immunol. 62: 312–317. Ehrenstein, M. R., J. G. Evans, A. Singh, S. Moore, G. Warnes, D. A. Isenberg, C. Mauri. 2004. Compromised function of regulatory T cells in rheumatoid arthritis and reversal by anti-TNF␣ therapy. J. Exp. Med. 200: 277–285. Lawson, C. A., A. K. Brown, V. Bejarano, S. H. Douglas, C. H. Burgoyne, A. S. Greenstein, A. W. Boylston, P. Emery, F. Ponchel, and J. D. Isaacs. 2006. Early rheumatoid arthritis is associated with a deficit in the CD4⫹CD25high regulatory T cell population in peripheral blood. Rheumatology (Oxford) 45: 1210 –1217. Baecher Allan, C., and D. A. Hafler. 2006. Human regulatory T cells and their role in autoimmune disease. Immunol. Rev. 212: 203–216.