Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

TL1A Induces TCR Independent IL-6 and TNF-α Production and Growth of PLZF+ Leukocytes

  • Kirsten Reichwald ,

    Contributed equally to this work with: Kirsten Reichwald, Tina Z. Jørgensen, Peter Tougaard

    kire@sund.ku.dk (KR); sosk@sund.ku.dk (SS)

    Affiliation Laboratory of Immunology, Faculty of Medical and Health Sciences, University of Copenhagen, Frederiksberg C, Denmark

  • Tina Z. Jørgensen ,

    Contributed equally to this work with: Kirsten Reichwald, Tina Z. Jørgensen, Peter Tougaard

    Affiliation Laboratory of Immunology, Faculty of Medical and Health Sciences, University of Copenhagen, Frederiksberg C, Denmark

  • Peter Tougaard ,

    Contributed equally to this work with: Kirsten Reichwald, Tina Z. Jørgensen, Peter Tougaard

    Affiliation Laboratory of Immunology, Faculty of Medical and Health Sciences, University of Copenhagen, Frederiksberg C, Denmark

  • Søren Skov

    kire@sund.ku.dk (KR); sosk@sund.ku.dk (SS)

    Affiliation Laboratory of Immunology, Faculty of Medical and Health Sciences, University of Copenhagen, Frederiksberg C, Denmark

Abstract

An elevated level of the cytokine TL1A is known to be associated with several autoimmune diseases, e.g. rheumatoid arthritis and inflammatory bowel disease. However, the mode of action of TL1A remains elusive. In this study, we investigated the role of TL1A in a pro-inflammatory setting, using human leukocytes purified from healthy donors. We show that TL1A, together with IL-12, IL-15 and IL-18, directly induces the production of IL-6 and TNF-α from leukocytes. Interestingly, TL1A-induced IL-6 was not produced by CD14+ monocytes. We further show that the produced IL-6 is fully functional, as measured by its ability to signal through the IL-6 receptor, and that the induction of IL-6 is independent of TCR stimulation. Furthermore, the transcription factor PLZF was induced in stimulated cells. These results offer a substantial explanation for the role of TL1A, since TNF-α and IL-6 are directly responsible for much of the inflammatory state in many autoimmune diseases. Our study suggests that TL1A is a possible target for the treatment of autoimmune diseases.

Introduction

TL1A (TNF like Ligand 1A, encoded by TNFSF15) is a cytokine initially described as a T cell co-stimulator, signaling through the receptor DR3 [1]. TL1A is increased in several autoimmune diseases, such as rheumatoid arthritis (RA) [2][4], psoriasis [5], inflammatory bowel disease (IBD) [6], [7] and ankylosing spondylitis [1], [8], [9]. Several different studies have implicated a role of TL1A in the induction of Th17 related cytokines, although opinions differ on the exact role of TL1A [10][12]. Knockout of DR3 or TL1A in different autoimmune mouse models ameliorates disease and delays disease onset of both collagen-induced arthritis [13] and experimental autoimmune encephalomyelitis (EAE) [14]. DR3 is essential for the development of allergic lung inflammation [15], but also involved in protection against certain viruses and bacteria, demonstrating its complex role [16], [17].

IL-6 is recognized as a mediator of inflammation, both because of its direct stimulatory effect on B cells, but also because of the pro-inflammatory potential of the IL-6/sIL-6R complex [18]. IL-6, together with TGF-β, directly induces IL-17 production from pathogenic Th17 cells [19], and blocking of IL-6 has efficacy in RA treatment, in particular when anti-TNF treatment is inadequate [20]. Hence, IL-6 is directly involved in inflammation, but it also renders T cells less responsive to Treg suppression via STAT3 activation [21], [22].

Different groups have shown that TL1A has the capability of synergizing with IL-12 and IL-18 in inducing IFN-γ in NK cells and other T cells [23], [24]. In addition, combinations of IL-12, IL-18 and IL-15 are known not only to induce IFN-γ [25], but also “cytokine memory” in NK cells. This cytokine memory is not clearly understood, but involves priming of NK cells by cytokines, resulting in enhanced IFN-γ production upon re-stimulation [26]. Furthermore, IL-15 has been shown to cooperate with TNF in the activation of dendritic cells [27], and to be involved in disturbing the Treg/Th17 balance [28], [29], which is a hallmark of autoimmune pathogenesis [10], [11], [21], [30]. IL-15 is known to augment memory T cell function [31], but also boosts their autoimmune capacity by inducing expression of classical co-receptors on activated CD4 T cells [32] and of NK receptors on CD8 T cells [33]. In Figure 1, we have summarized the effect of TL1A, IL-12, IL-15 and IL-18 on naïve lymphocytes.

thumbnail
Figure 1. An overview of previous studies on the pro-inflammatory cytokines used in our setup.

IL-15 is known to induce the growth of NK cells and memory CD8 T cells, and also causes CD8 T cells to acquire functional NK receptors [25], [56], [57]. IL-12 and IL-18 are known to induce IFN-γ in both NK and NKT cells in synergy with TL1A [23]. IL-12, IL-15 and IL-18 have recently been shown to induce “cytokine memory” [26]. TL1A is also known to be involved in Th17 development [10], [12], but the mechanism remains unidentified.

https://doi.org/10.1371/journal.pone.0085793.g001

Bystander activation and cytokine activation of different lymphocyte subsets is being intensely studied [34][37] due to its clear significance in orchestrating inflammatory responses. Initially, bystander activation of CD8 T cells or, to a lesser degree, CD4 T cells was described [38], [39], but by now cytokine activation is regarded an innate feature of many conventional T cells and innate-like lymphocytes. In fact, many lymphocytes might only need a combination of an appropriate STAT activator and an IL-1 family cytokine to initiate the production of cytokines [36].

PLZF (promyelocytic leukemia zinc-finger) is a transcription factor involved in stem cell maintenance [40]. It is critical for NKT cell and ILC development and directly induces effector functions in memory CD8 T cells. By now, it is recognized as an inducer of innate-like features in many different lymphocytes [41].

In this study, we show that TL1A in combination with IL-12, IL-15 and IL-18 directly induces antigen-independent IL-6 and TNF-α from monocyte-depleted PBMCs. Although we were unable to identify the exact cell type, we show that the IL-6 produced is functional and TL1A-dependent, and that the stimulation increases PLZF.

Materials and Methods

Purification of Lymphocytes

Buffy coats from healthy blood donors were obtained from the Blood Bank at the Copenhagen University Hospital (Denmark), in agreement with the local ethics committee (Region Hovedstaden). PBMCs were purified by Ficoll-Histopaque density gradient centrifugation. For PBLs, 200 µL washed pan-mouse beads (Invitrogen, Cat# 11041) were added to 2×108 PBMCs in 30 mL complete RPMI, and left in the incubator for at least 1 h to allow phagocytic cells to take up beads or adhere to the plastic culture flask. PBLs were collected by removing cells attached to the beads with a magnet; remaining cells were used for subsequent stimulations. For TCR stimulation studies, PBMCs were plastic depleted twice to obtain PBLs. CD14 depletion was done according to manufacturer’s protocol using CD14 beads (Invitrogen, Cat# 11149D). For depletion studies, Dynabeads Pan Mouse IgG beads (Invitrogen, Cat# 11041) was used according to manufacturer’s protocol. Depletion was done using the same antibodies (same clone and manufacturer, but non-labeled) as used for flow cytometry, as listed below. For cytokine stimulation, cells were set up at a density of 106/mL in complete RPMI and the following cytokine concentrations were used, unless stated otherwise in the figure: IL-12 (RnD Systems, Cat# 219-IL): 4 ng/mL, IL-15 (Peprotech, Cat# 200-15): 10 ng/mL, IL-18 (MBL, Cat# B003-5): 40 ng/mL, TL1A (RnD Systems, Cat# 1319-TL): 100 ng/mL, TL1A blocking Ab (RnD Systems, Cat# MAB7441): 1 µg/mL, tocilizumab (RoActemra, Roche): 10 µg/mL. For different TCR stimulations, plastic-depleted PBLs were stimulated with 4 µg/mL CD3 (eBioscience, Cat #14-0037), 25 µL/mL (according to the manufacturer) CD3/CD28 beads (Invitrogen, Cat # 111.32D) or 2.5 µg/mL SEA (Staphylococcal enterotoxin A (Sigma-Aldrich, Cat#S9399)). The supernatants were harvested on day 6, and bead-based ELISA was performed on the samples.

Flow Cytometry

CFSE staining was done using 5 µM CFSE (Invitrogen, Cat# C34554) as described by Parish et al. [42] and stimulated with cytokines for 6 days prior to flow cytometric analysis. Antibodies used: CD8-APC (BD, Cat# 345775), CD3-APC (BD, Cat# 555335), CD4-APC (Biolegend, Cat# 300514), CD56-APC (eBioscience, Cat#17-0569-42), CD16-PE (Biolegend, Cat#302007), HLA-DR-PE (Biolegend, Cat# 307605), NKG2D-APC (RnD Systems, Cat# FAB139A). For intracellular staining, IL-6-PE (eBioscience, Cat#12-7069) and IFN-γ-FITC (BD, Cat# 554700) were used. Isotype controls were IgG1 APC (BD, cat # 555751), rat IgG1-PE (Invitrogen, Cat # R104) and IgG1-FITC (BD, Cat #555748). PBMCs were incubated for 6 h +/− LPS (Sigma-Aldrich, L2654, 1 µg/mL) and cells were stained using the BD Cytofix/Cytoperm Kit (BD, Cat# 554714), Golgistop (BD, Cat# 554724) or Golgiplug (BD, Cat# 555029) according to the manufacturer’s protocol. Samples were run on an Accuri C6 Flow cytometer and data were analyzed using FCS Express v. 3.

Functional Assay

For the functional testing of IL-6, PBMCs from healthy donors were purified as described above, plated at 5×106 in a thin layer, and left to adhere for 1 h. The supernatant was quickly removed by suction, and one of the following were added: complete RPMI containing either (1) nothing (control), (2) recombinant human IL-6 (1, 10 or 50 ng/mL, Peprotech, Cat# 200-06) or (3) the supernatant from CD14-depleted cells incubated with IL-12, IL-15, IL-18 and TL1A as described above, harvested after 6 days (IL-6 concentration: 27 ng/mL). As a control, tocilizumab (RoActemra, Roche, 8 µg/mL) was applied to two samples with either supernatant or rhIL-6 (10 ng/mL). After 30 min, these solutions were quickly removed by suction, and cold RIPA buffer containing 1 mM EDTA and phosphatase/protease inhibitors (ThermoScientific, Cat# 78441) was added. The cells were extracted for western blot as described below.

Western Blot

For the both functional assay and for the whole cell PLZF blot, samples were extracted with ice cold RIPA buffer including phosphatase/protease and 1 mM EDTA. Samples were left on ice for 15 min, centrifuged for 10 min at 15,000×g and the supernatant collected. For the cytoplasmic/nuclear extract, cells were harvested on day 4 and extracted as described by the manufacturer using the Nuclear Extract Kit (Active Motif, Cat # 40010). Samples were run on 4–12% Bis-Tris gels (Novex, Cat# WG1401BX10) using MOPS buffer (Invitrogen, Cat # NP0001) with an antioxidant (Invitrogen, Cat# NP0005). Proteins were blotted onto a nitrocellulose membrane (Invitrogen, Cat# IB3010-01) using iBlot from Invitrogen, program P3. The membrane was blocked using a 2% Blotto solution (Santa Cruz Biotechnology, Cat# SC-2325,), washed in PBS/0.1% Tween (except for the pSTAT3 membrane, for which TBS/0.1% Tween was used) and incubated with shaking overnight at 4°C with the primary antibody. Primary antibodies used were: PLZF (1∶1000, Santa Cruz Biotechnology, Cat# sc-28319), STAT3: (1∶2000, Cell Signaling, Cat# 4904p) and pSTAT3 (1∶2000, Cell Signaling, Cat# 9145p). After washing again, the secondary Ab was added: swine anti-rabbit HRP (1∶1000, Dako, Cat# P0399) or rabbit anti-mouse HRP (1∶1000, Dako, Cat# P0260) and the membrane was incubated with shaking at RT for 1 h. After washing, the SuperSignal West Pico Chemiluminescent Substrate (Thermo Scientific, Cat# 34080) was added onto the membrane and left for 5 min before developing the films (CarestreamKodakBiomax light film, Sigma-Aldrich, Cat# Z373508-50EA). Blots of STAT3 were performed on the same membranes as those used for pSTAT3. Membranes were stripped for 25 min in Restore PLUS Western Blot Stripping Buffer (Thermo Scientific, Cat# 46430), washed and re-blocked before detection as described above.

Measurement of Cytokine Production

Cells from different setups were harvested at different time points (see figures) and centrifuged for 10 min at 1000×g. Supernatants were collected and used for measurement of cytokines either by bead-based ELISA or standard ELISA kits. For several cytokines, bead-based ELISA kits were used: Diacone Diaplex Th1/Th2/inflammation, Nordic Biosite, 880 100 010. For IL-6 only: Diacone Diaplex, Nordic Biosite, 880 030 001. Both bead-based assays were run on the Accuci C6 flow cytometer, and data were analyzed using the Flowcytomix Pro software (eBioscience).

Results

TL1A Induces Proinflammatory Cytokines

TL1A synergizes with IL-12 and IL-18 in the activation of NK, NKT and T cells [23], [24], and IL-15 has a synergic effect on IL-12 or IL-18 [25]. Since TL1A is elevated in several chronic inflammatory diseases [2], [5], [7], [9], we wanted to investigate the range of cytokines regulated by TL1A. Freshly purified PBMCs were incubated with a combination of different cytokines, and the production of cytokines was measured after 6 days of stimulation. We used a multiplex approach, enabling us to measure 10 different cytokines in one sample (IFN-γ, IL-1β, IL-2, IL-4, IL-6, IL-8, IL-10, IL12p70, IL-17A and TNF-α).

As shown in Figure 2a, the cytokine measurements clearly showed a strong induction of IL-6 and TNF-α by TL1A, whereas classical stimulatory cytokines such as IL-2 and IL-4 were not detected (data not shown). Addition of a TL1A blocking Ab completely abolished the production of IL-6 and TNF-α, demonstrating both specificity and integrity. Also, the blocking effect of anti-TL1AAb ruled out the possibility that the TL1A effect was caused by endotoxin or other impurities.

thumbnail
Figure 2. TL1A induces IL-6 and TNF-α.

Freshly purified PBMCs were incubated with IL-12 (2 ng/mL), IL-15 (10 ng/mL), IL-18 (10 ng/mL), TL1A (100 ng/mL) and TL1AAb (1 µg/mL, blocking antibody). Extra IL-15 (2 ng/mL) was added on day 3. (A) After 6 days, supernatants were collected and different cytokines were measured by bead-based ELISA. Error bars represent the SEM of eight measurements. Statistically significant differences are indicated by ***(t-test, P<0.001). Data are representative of three different experiments with cells from three separate donors (B). After 6 days, cells were stained extracellularly for CD3 or CD8 and intracellularly for IFN-γ and analyzed by flow cytometry as described in the Materials and Methods. The upper panels show gating for lymphocytes; the two lower panels show CD3/IFN-γ and CD8/IFN-γ staining. Data are representative of three different experiments with cells from three separate donors.

https://doi.org/10.1371/journal.pone.0085793.g002

We also measured IFN-γ in PBMCs after 6 days of cytokine stimulation (Figure 2b) using intracellular staining. As expected, the combination of the different cytokines along with TL1A resulted in a strong induction of IFN-γ, as has been shown previously [23], [25], [26].

IL-6 is Rapidly Induced in Monocyte-depleted PBL Cultures

Since monocytes and macrophages are known as the main source of IL-6 in the initiation of inflammation [43], we decided to investigate if monocytes produced IL-6 after stimulation with IL-12,IL-15, IL-18 and TL1A.

We incubated both PBMCs and PBLs with different combinations of cytokines and measured IL-6 production after 1, 3 and 6 days. As shown in Figure 3a, neither TL1A nor the combination of IL-12, IL-15 and IL-18 were able to induce any IL-6 on their own. PBLs stimulated with IL-12, IL-15, IL-18 and TL1A produced higher levels of IL-6 than stimulated PBMCs (4400 pg/mL vs. 2300 pg/mL). The combination of IL-12, IL-18 and TL1A also induced IL-6 production in PBLs, as shown in Figure 4, but the levels were several fold higher when IL-15 was also added (800 pg/mL vs. 4400 pg/mL). The data shown in Figure 3b and Figure 3c demonstrate that IL-6 production was already high at day 3, and higher in PBLs than in PBMCs.

thumbnail
Figure 3. IL-6 production is higher in PBLs than in PBMCs.

PBMCs and PBLs were prepared as described in the Materials and Methods, stimulated for 1, 3 or 6-12 (4 ng/mL), IL-15 (10 ng/mL), IL-18 (40 ng/mL), TL1A (100 ng/mL) and TL1AAb (1 µg/mL) and IL-6 was measured in the supernatants. (A) PBMCs and PBLs were stimulated with different combinations of cytokines and IL-6 was measured on day 6. (B) IL-6 production by PBMCs and PBLs stimulated with IL-12, IL-15, IL-18 and TL1A for 1, 3 and 6 days. (C) IL-6 production after 1, 3 and 6 days by PBLs and PBMCs stimulated with different cytokine combinations. Error bars represent the SEM of two measurements. Data are representative of three different experiments with cells from three separate donors.

https://doi.org/10.1371/journal.pone.0085793.g003

thumbnail
Figure 4. Depletion of CD14+ monocytes enhances TL1A-dependent IL-6.

PBMCs, PBLs and CD14-depleted PBMCs were prepared as described in the Materials and Methods, stimulated for 6 days with combinations of IL-12 (4 ng/mL), IL-15 (10 ng/mL), IL-18 (40 ng/mL), TL1A (100 ng/mL), TL1AAb (1 µg/mL) and tocilizumab (RoActemra, 8 µg/mL), and IL-6 was measured in the supernatants. Error bars represent the SEM of two measurements. Data are representative of three different experiments with cells from three separate donors.

https://doi.org/10.1371/journal.pone.0085793.g004

Hence, when comparing stimulated PBLs with PBMCs, monocytes were not the source of IL-6 (Figure 3). To verify this, we stimulated CD14-depleted PBMCs (Figure 4). The results presented in Figure 4 confirmed that IL-6 was in fact not produced by monocytes, as much higher levels were obtained when stimulating CD14-depleted cells. This shows that IL-6 was produced by CD14-depleted leukocytes only by stimulation with a pro-inflammatory cocktail of TL1A, IL-12, IL-15 and IL-18. To confirm that the difference in IL-6 production between PBLs and PBMCs was not due to uptake of IL-6 by monocytes, we added IL-6R blocking tocilizumab, which did not result in any difference in IL-6 production (Figure 4).

TL1A-induced IL-6 is Functional

It is known that IL-6 signaling results in STAT3 phosphorylation, which inhibits the suppression of T cells by Tregs [21]. To validate the function and integrity of the IL-6 produced, we tested the ability of the collected supernatant to phosphorylate STAT3, using rhIL-6 as a positive control. In short, we purified PBMCs from healthy donors and monocytes enriched by plate adherence. We added media with either rhIL-6 or IL-6 from a supernatant of the cytokine stimulated PBLs. After 30 min, the supernatant was removed and cells were harvested for western blot detection of STAT3 and pSTAT3, as shown in Figure 5. To confirm that phosphorylation was indeed mediated by IL-6, the IL-6R antagonist tocilizumab (RoActemra) was added in two separate samples to block signaling from both the supernatant and rhIL-6 (Figure 5). The results clearly demonstrate that the IL-6 produced was functional, and that IL-6 signaling was essential for STAT3 phosphorylation.

thumbnail
Figure 5. TL1A-induced IL-6 phosphorylates STAT3. A functional pSTAT3 was set up to verify the functionality of TL1A-induced IL-6.

In short, purified PBMCs were incubated for 30-6 (1, 10 or 100 ng/mL) or the supernatant from cells incubated with IL-12 (4 ng/mL), IL-15 (10 ng/mL), IL-18 (40 ng/mL), TL1A (100 ng/mL); the supernatant contained 27 ng IL-6/mL (Sup [27]). PBMC proteins were extracted and pSTAT3 and STAT3 were detected by western blotting as described. Additionally, to verify that IL-6 was in fact the cytokine responsible for STAT3 phosphorylation, both the supernatant (27 ng/mL) and rhIL-6 (10 ng/mL) were added along with tocilizumab (IL-6R blocking Ab). Data are representative of three different experiments using cells from three separate donors and supernatants from two different trials.

https://doi.org/10.1371/journal.pone.0085793.g005

The Effect of TL1A is TCR Independent

To investigate if the IL-6 production was affected by TCR stimuli, we set up an experiment using different combinations of IL-12, IL-18 and TL1A +/− TCR stimuli. In short, PBLs were stimulated with cytokines (IL-12, IL-18 and TL1A), TCR stimuli (Staphylococcal enterotoxin A (SEA), CD3 or CD3/CD28) or both. After 6 days of stimulation, the supernatant was collected and IL-6 was measured. As shown in Figure 6, the combination of IL-12, IL-18 and TL1A resulted in the production of IL-6, as expected. TCR stimulation in general did not result in IL-6 production, although SEA did induce low levels of IL-6 by itself.

thumbnail
Figure 6. IL-6 production is TCR independent.

PBLs were TCR stimulated using anti-CD3 (4 µg/ml), CD3-28 beads or SEA (Staphylococcal enterotoxin A, 2.5 µg/ml). The supernatants were harvested on day 6, and bead-based ELISA was performed on the samples. Error bars represent the SEM of two measurements. Data are representative of three different experiments using PBLs from three different donors.

https://doi.org/10.1371/journal.pone.0085793.g006

The induction of IL-6 was still observed after TCR co-stimulation, although there was a tendency that TCR stimulation downregulated IL-6 production. This indicates that the imprinting achieved by the cytokines is not substantially affected by TCR signaling.

Intracellular IL-6 Staining

The most obvious question now was: exactly which cells are IL-6 producers? IL-6 is routinely measured intracellularly in monocytes after stimulation, and we therefore tried to measure IL-6 by intracellular flow cytometry. We could not detect TL1A-induced IL-6 in cultured PBLs or PBMCs by intracellular staining. We tried different time points (ranging from 6 to 72 h after addition of cytokines) and different inhibitors of intracellular transport and lysosomal function. First, we tried both Golgistop (contains monensin, which blocks transport from the Golgi/ER) and Golgiplug (contains brefeldin A, which blocks exit from the ER) +/− PMA/ionomycin, without success. We speculated that this could be due to the cells using different secretory lysomes than those used in the classical ER/Golgi pathway, and so tried two different lysosomal inhibitors: bafilomycin A (inhibits fusion between autophagosomes and lysosomes) and chloroquine (prevents fusion of endosomes and lysosomes). Still, we did not succeed in measuring intracellular IL-6 in leukocytes (data not shown).

To verify our intracellular IL-6 procedure, we set up PBMCs incubated with LPS and Golgistop/plug for 6 h (Figure 7). This clearly showed that the method applied was not per se flawed, as we could readily measure IL-6 produced by stimulated monocytes. The difficulties in measuring TL1A-induced intracellular IL-6 and possible explanations for this are further reflected on in the Discussion.

thumbnail
Figure 7. Intracellular IL-6 staining is possible in monocytes.

Freshly purified PBMCs were incubated for 6+/− LPS (100 ng/mL) and Golgistop or Golgiplug. Cells were stained intracellularly for IL-6, and analyzed by flow cytometry. Upper panels: monocyte gating for control and LPS samples with Golgiplug/stop. Lower panels: % of gated cells positive for IL-6 expression. Data are representative of three different experiments using PBMCs from three separate donors.

https://doi.org/10.1371/journal.pone.0085793.g007

Characterization of Leukocytes Induced by TL1A

To see if TL1A and the cytokine cocktail had an effect on the proliferation of different leukocytes, we stained PBLs with CFSE [42], added cytokines as listed in Figure 8 and observed the phenotype of growing cells after 6 days of stimulation.

thumbnail
Figure 8. TL1A induces the growth of PLZF+ cells.

(A) PBLs were purified from healthy donors, CFSE stained and stimulated for 6 days with combinations of IL-12 (4 ng/mL), IL-15 (10 ng/mL), IL-18 (40 ng/mL), TL1A (100 ng/mL) and TL1AAb (1 µg/mL). Upper panel shows gating on live cells and (%) proliferating cells as seen by less CFSE/cell. Lower panels show staining of different surface molecules. Note that for CD8, both CD8+ (bright+dim) and CD8dim cells are shown (intermediate (%) in grey); see text for elaboration. Data are representative of three different experiments with cells from three separate donors. (B) PBLs were extracted after 6 days of stimulation with the cytokines listed, and PLZF was detected in whole cell extracts or cytoplasmic/nuclear extracts. Data are essentially representative of three different experiments.

https://doi.org/10.1371/journal.pone.0085793.g008

It was evident from the proliferation data (Figure 8a) that TL1A had a strong synergistic effect when added with IL-12, IL-15 and IL-18. Alone, the combination of IL-12, IL-15 and IL-18 resulted in 14% proliferating cells after 6 days, whereas the addition of TL1A increased this to 30%. Interestingly, TL1A had no effect on its own, indicating that DR3 costimulation is essential, but not alone sufficient for proliferation.

The cells proliferating were to a large degree either CD8bright (8%) or CD8dim (11%), and partially CD3+ (13%) and NKG2D+ (13%). Hence, the cells were most likely a mix of CD8 T cells, NKT and NK cells. Almost no CD4 T cells proliferated. Again, the TL1A Ab effectively blocked the proliferative effect of TL1A (Figure 8a).

Along with this, we decided to detect the transcription factor PLZF (promyelocytic leukemia zinc-finger) using western blot. PLZF was initially described as being involved in stem cell maintenance [40], and is critical for NKT cell and ILC development [41], [44]. PLZF is also able to induce effector functions in memory CD8 T cells, and drives IL-17 production in CD8 cells, along with the stimulation of subtypes of γδ T cells and other innate-like lymphocytes without antigen stimulation [45][48]. As shown in Figure 8b, PLZF was clearly induced in cells treated with the combined cytokine stimulation, indicating NKT or other innate-like leukocytes.

To identify the IL-6 producing cell, we also tested different depletions. We targeted the cell surface markers CD4, CD8, CD16/CD56 and HLA-DR, assuming that these would cover classical CD4/CD8 T-cells, mature NK and NK T-cells and B-cells/dendritic cells. We then set up stimulations of the depletions and measured IL-6 production on day 7 and analyzed cells by flow cytometry. From the flow cytometric analysis shown in Figure 9a it became clear that even though the depletions were successful, cytokine stimulation led to differentiation of CD3+HLA-DR+ in the HLA-DR-depleted cells (from 0% to 16%). Furthermore, none of the depletions had targeted the IL-6 producing cell, as observed in Figure 9b, since all depletions displayed equal or higher IL-6 levels after 7 days of stimulation. Both CD4-, HLA-DR- and CD56/CD16-depleted cells produced significantly higher levels of IL-6 than non-depleted PBLs.

thumbnail
Figure 9. IL-6 produced by PBLs depleted for CD4+, CD8+, HLA-DR+ or CD56+/CD16+ cells.

(A) Freshly purified PBLs stained for CD4, CD8, CD3, HLA-DR, CD16, CD56. Depletion controls for PBLs depleted for CD4+, CD8+, HLA-DR+ or CD16+/CD56+ cells. PBLs and depletions were stimulated for 7 days with combinations of IL-12 (4 ng/mL), IL-15 (10 ng/mL), IL-18 (40 ng/mL) and TL1A (100 ng/mL) and stained for CD4, CD8, CD3, HLA-DR, CD16 and CD56 expression. (B) IL-6 production after 7 days by PBLs and depletions stimulated with IL-12, IL-15, IL-18 and TL1A as described above. Error bars represent the SEM of two measurements. Statistically significant differences by t-test: *** = p<0.001, * = p<0.05, n.s. = not significant. Data are representative of two different experiments with cells from two separate donors.

https://doi.org/10.1371/journal.pone.0085793.g009

Discussion

We have shown that TL1A, together with a pro-inflammatory cocktail of IL-12, IL-15 and IL-18, specifically induces TNF-α and IL-6 from CD14-depleted leukocytes. It is particularly interesting that IL-6 induction was much higher in CD14-depleted cells, since this proinflammatory cytokine is traditionally derived from monocytes. TL1A is elevated in the serum and synovial fluid of RA patients [2], [4], in psoriatic skin lesions [5] and in Crohn’s disease [6], [7]. Both IL-6 and TNF-α have well-described effects on both vascular function and stimulation of proinflammatory leukocytes. Hence, the link between TL1A and induced inflammation is apparent.

TL1A alone or the cytokine combination of IL-12, IL-15 and IL-18 did not result in IL-6 production, demonstrating the need for two different signaling events, similar to that observed for cytokine activation of ILCs [36]. Importantly, TL1A with IL-12 and IL-18 alone was sufficient for the induction of IL-6 production. However, IL-15 increased the level by several fold, and was therefore used in most setups. TL1A is known to synergize with IL-12 and IL-18 in IFN-γ production by NK and NKT cells [23], and IL-15 is known to augment IFN-γ production by NK cells when stimulated with IL-12 [26]. Hence, the effect of IL-15 could be to support indirect cytokine activation of a range of innate-like lymphocytes.

We tried several approaches for detecting the IL-6 producing cells, and were initially quite surprised that this was such a difficult task, since IL-6 is easily detected in LPS-activated monocytes. However, no matter the approach, intracellular IL-6 remained undetectable in our setup. There are several possible explanations for this:

  1. It is well-known that staining for intracellular cytokines will only reveal proteins residing in the ER/Golgi. However, TL1A-induced IL-6 might be transported through an alternative pathway, e.g. lysosomes, that are harder for antibodies to access, regardless of permeabilization. However, we tried an assay for this pathway using lysosomal inhibitors before intracellular staining.
  2. IL-6 might not be fully folded intracellularly, especially if the protein is present inside lysosomal compartments.
  3. IL-6 producing cells are fragile and are lost during handling.
  4. IL-6 might be expressed as a different splice variant, only containing some of the epitopes recognized by our antibodies. IL-6 does have smaller splice variants, but most of these have reduced biological activity.

Even though the IL-6 producing cells remained elusive, the produced IL-6 was functional, specifically in its ability to signal through the IL-6R. STAT3 phosphorylation and IL-6 signaling are known to interfere with the delicate balance between Treg and Teff cells [21], [22], a phenomenon often observed in autoimmune disease [22], [49], [50], supporting the pro-inflammatory potential of TL1A.

We mainly observed growth of CD8+/NKG2D+/CD3+/− cells, indicating NK or NKT cells. It was also clear that there was an induction of the transcription factor PLZF. Note that we tried different loading controls such as ERK and GAPDH (data not shown), but since we compared highly activated and proliferating cells with naïve cells, both of these were also increased. PLZF is known as being critical to the development of NKT cells [44]. In a transgenic PLZF mouse, however, T cells in general acquire a more memory-like phenotype, along with a bias towards IL-17 production [48], and PLZF positive cells induce the growth of CD8+ memory cells [47]. Hence, the presence of PLZF+ cells might simply be NKT cells, but in conjunction with the cytokine profile observed, this might also represent the activation of a broad range of innate-like lymphocytes.

The results from our depletion studies demonstrated that differentiation has to be taken into account when evaluating depletion studies. Although we successfully depleted HLA-DR-positive cells prior to cytokine addition, the stimulation caused significant up-regulation of HLA-DR+CD3+ cells due to activation. We were able to show that the IL-6 did not derive from classical CD4+ or CD8+ cells, and not from mature NK/NKT cells or circulating HLA-DR positive cells. Hence, the source of IL-6 might be unknown precursor, making it difficult to remove them prior to stimulation.

Our results clearly demonstrate that TL1A in a pro-inflammatory environment is a potent mediator of cytokine activation. CD8+ T cells, NK cells and NKT cells have all been described as being affected by some of the cytokines applied with regards to growth and IFN-γ production. However, this is the first time that TL1A has been shown to directly induce IL-6 in cells from healthy donor cells.

There is no doubt that IL-6 plays an important role in several autoimmune diseases. Along with the soluble IL-6 receptor (sIL-6R), it has been shown to be involved in different forms of arthritis, inflammatory bowel disease, psoriasis, asthma and colon cancer [22], [51][53]. This knowledge has paved the road for antibodies targeting IL-6R or gp130 (to which the IL-6R binds for signaling) that are now introduced as part of the treatment strategy in many of the above mentioned diseases.

Anti-IL-6R treatment is often successfully applied when anti-TNF has no effect, which is the case for 20–40% of RA patients [20]. This demonstrates that targeting TNF, although being a major leap in RA treatment, might not be the most effective strategy. TL1A was shown in this study to mediate the activation of different cells, resulting in the production of key inflammatory cytokines. One might therefore think of TL1A as an interesting target for RA treatment. Although this may be true, TL1A/DR3 signaling is critical to the viral immune response [16] and to optimally protect against Salmonella [17]. Hence, blocking TL1A might result in similar adverse effects as seen with anti-TNF and anti-IL-6R therapies [54]. Nonetheless, TL1A still represents an interesting target which seems to be upstream of both IL-6 and TNF, as illustrated in Figure 10.

thumbnail
Figure 10. Proposed role of TL1A in the induction of inflammation and Th17 pathogenesis.

IL-15 is known to support the growth of CD8+ T cells and NK cells [56], [58]. The combination of IL-12, IL-15 and IL-18 induces memory-like NK-cells [26]. We show that TL1A, in combination with IL-12 and IL-18 or IL-12, IL-15 and IL-18, specifically induces TCR-independent TNF-α and IL-6. Both are pro-inflammatory cytokines known to be key players in the development and progression of several autoimmune diseases.

https://doi.org/10.1371/journal.pone.0085793.g010

We believe that future treatment strategies for autoimmune diseases such as RA and psoriasis will not only target cytokines, but the balance between regulatory/inflammatory cells in general. For example, helminths are currently being tested in clinical trials treating multiple sclerosis, due to their ability to alter disordered immune regulation [55]. This rather unspecific approach clearly shows us that we do not yet have a sufficient understanding of the regulation and imprinting of the immune system in autoimmune diseases. Nevertheless, the first step towards such an understanding is to move up the chain of events from the classical pro-inflammatory cytokines. Since TL1A directly induces TNF-α and IL-6, blocking it could be part of the solution in new treatment regimens.

Author Contributions

Conceived and designed the experiments: KR PT TZJ SS. Performed the experiments: KR PT TZJ. Analyzed the data: KR PT TZJ SS. Contributed reagents/materials/analysis tools: KR SS. Wrote the paper: KR SS.

References

  1. 1. Migone TS, Zhang J, Luo X, Zhuang L, Chen C, et al.. (2002) TL1A is a TNF-like ligand for DR3 and TR6/DcR3 and functions as a T cell costimulator. Immunity 16: 479–492. S1074761302002832 [pii].
  2. 2. Bamias G, Siakavellas SI, Stamatelopoulos KS, Chryssochoou E, Papamichael C, et al. (2008) Circulating levels of TNF-like cytokine 1A (TL1A) and its decoy receptor 3 (DcR3) in rheumatoid arthritis. Clin Immunol 129: 249–255 S1521-6616(08)00728-6 [pii];10.1016/j.clim.2008.07.014 [doi].
  3. 3. Cassatella MA, Pereira-da-Silva G, Tinazzi I, Facchetti F, Scapini P, et al. (2007) Soluble TNF-like cytokine (TL1A) production by immune complexes stimulated monocytes in rheumatoid arthritis. J Immunol 178: 7325–7333. 178/11/7325 [pii].
  4. 4. Zhang J, Wang X, Fahmi H, Wojcik S, Fikes J, et al. (2009) Role of TL1A in the pathogenesis of rheumatoid arthritis. J Immunol 183: 5350–5357 jimmunol.0802645 [pii];10.4049/jimmunol.0802645 [doi].
  5. 5. Bamias G, Evangelou K, Vergou T, Tsimaratou K, Kaltsa G, et al. (2011) Upregulation and nuclear localization of TNF-like cytokine 1A (TL1A) and its receptors DR3 and DcR3 in psoriatic skin lesions. Exp Dermatol 20: 725–731 10.1111/j.1600-0625.2011.01304.x [doi].
  6. 6. Bamias G, Kaltsa G, Siakavellas SI, Gizis M, Margantinis G, et al. (2012) Differential expression of the TL1A/DcR3 system of TNF/TNFR-like proteins in large vs. small intestinal Crohn's disease. Dig Liver Dis 44: 30–36 S1590-8658(11)00343-4 [pii];10.1016/j.dld.2011.09.002 [doi].
  7. 7. Bamias G, Martin C III, Marini M, Hoang S, Mishina M, et al. (2003) Expression, localization, and functional activity of TL1A, a novel Th1-polarizing cytokine in inflammatory bowel disease. J Immunol 171: 4868–4874.
  8. 8. Konsta M, Bamias G, Tektonidou MG, Christopoulos P, Iliopoulos A, et al.. (2012) Increased levels of soluble TNF-like cytokine 1A in ankylosing spondylitis. Rheumatology (Oxford). kes316 [pii];10.1093/rheumatology/kes316 [doi].
  9. 9. Bayry J (2010) Immunology: TL1A in the inflammatory network in autoimmune diseases. Nat Rev Rheumatol 6: 67–68 nrrheum.2009.263 [pii];10.1038/nrrheum.2009.263 [doi].
  10. 10. Pappu BP, Borodovsky A, Zheng TS, Yang X, Wu P, et al. (2008) TL1A-DR3 interaction regulates Th17 cell function and Th17-mediated autoimmune disease. J Exp Med 205: 1049–1062 jem.20071364 [pii];10.1084/jem.20071364 [doi].
  11. 11. Niu Q, Cai B, Huang ZC, Shi YY, Wang LL (2011) Disturbed Th17/Treg balance in patients with rheumatoid arthritis. Rheumatol Int. 10.1007/s00296-011-1984-x [doi].
  12. 12. Jones GW, Stumhofer JS, Foster T, Twohig JP, Hertzog P, et al. (2011) Naive and activated T cells display differential responsiveness to TL1A that affects Th17 generation, maintenance, and proliferation. FASEB J 25: 409–419 fj.10-166843 [pii];10.1096/fj.10-166843 [doi].
  13. 13. Bull MJ, Williams AS, Mecklenburgh Z, Calder CJ, Twohig JP, et al. (2008) The Death Receptor 3-TNF-like protein 1A pathway drives adverse bone pathology in inflammatory arthritis. J Exp Med 205: 2457–2464 jem.20072378 [pii];10.1084/jem.20072378 [doi].
  14. 14. Meylan F, Davidson TS, Kahle E, Kinder M, Acharya K, et al. (2008) The TNF-family receptor DR3 is essential for diverse T cell-mediated inflammatory diseases. Immunity 29: 79–89 S1074-7613(08)00269-0 [pii];10.1016/j.immuni.2008.04.021 [doi].
  15. 15. Fang L, Adkins B, Deyev V, Podack ER (2008) Essential role of TNF receptor superfamily 25 (TNFRSF25) in the development of allergic lung inflammation. J Exp Med 205: 1037–1048 jem.20072528 [pii];10.1084/jem.20072528 [doi].
  16. 16. Twohig JP, Marsden M, Cuff SM, Ferdinand JR, Gallimore AM, et al. (2012) The death receptor 3/TL1A pathway is essential for efficient development of antiviral CD4+ and CD8+ T-cell immunity. FASEB J 26: 3575–3586 fj.11-200618 [pii];10.1096/fj.11-200618 [doi].
  17. 17. Buchan SL, Taraban VY, Slebioda TJ, James S, Cunningham AF, et al. (2012) Death receptor 3 is essential for generating optimal protective CD4(+) T-cell immunity against Salmonella. Eur J Immunol 42: 580–588 10.1002/eji.201041950 [doi].
  18. 18. Neurath MF, Finotto S (2011) IL-6 signaling in autoimmunity, chronic inflammation and inflammation-associated cancer. Cytokine Growth Factor Rev 22: 83–89 S1359-6101(11)00005-0 [pii];10.1016/j.cytogfr.2011.02.003 [doi].
  19. 19. Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, et al. (2006) Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature 441: 235–238 nature04753 [pii];10.1038/nature04753 [doi].
  20. 20. Emery P, Keystone E, Tony HP, Cantagrel A, van VR, et al. (2008) IL-6 receptor inhibition with tocilizumab improves treatment outcomes in patients with rheumatoid arthritis refractory to anti-tumour necrosis factor biologicals: results from a 24-week multicentre randomised placebo-controlled trial. Ann Rheum Dis 67: 1516–1523 ard.2008.092932 [pii];10.1136/ard.2008.092932 [doi].
  21. 21. Goodman WA, Young AB, McCormick TS, Cooper KD, Levine AD (2011) Stat3 phosphorylation mediates resistance of primary human T cells to regulatory T cell suppression. J Immunol 186: 3336–3345 jimmunol.1001455 [pii];10.4049/jimmunol.1001455 [doi].
  22. 22. Goodman WA, Levine AD, Massari JV, Sugiyama H, McCormick TS, et al. (2009) IL-6 signaling in psoriasis prevents immune suppression by regulatory T cells. J Immunol 183: 3170–3176 jimmunol.0803721 [pii];10.4049/jimmunol.0803721 [doi].
  23. 23. Papadakis KA, Prehn JL, Landers C, Han Q, Luo X, et al. (2004) TL1A synergizes with IL-12 and IL-18 to enhance IFN-gamma production in human T cells and NK cells. J Immunol 172: 7002–7007.
  24. 24. Papadakis KA, Zhu D, Prehn JL, Landers C, Avanesyan A, et al.. (2005) Dominant role for TL1A/DR3 pathway in IL-12 plus IL-18-induced IFN-gamma production by peripheral blood and mucosal CCR9+ T lymphocytes. J Immunol 174: 4985–4990. 174/8/4985 [pii].
  25. 25. Fehniger TA, Shah MH, Turner MJ, VanDeusen JB, Whitman SP, et al. (1999) Differential cytokine and chemokine gene expression by human NK cells following activation with IL-18 or IL-15 in combination with IL-12: implications for the innate immune response. J Immunol 162: 4511–4520.
  26. 26. Romee R, Schneider SE, Leong JW, Chase JM, Keppel CR, et al. (2012) Cytokine activation induces human memory-like NK cells. Blood 120: 4751–4760 blood-2012-04-419283 [pii];10.1182/blood-2012-04-419283 [doi].
  27. 27. Vujanovic L, Szymkowski DE, Alber S, Watkins SC, Vujanovic NL, et al. (2010) Virally infected and matured human dendritic cells activate natural killer cells via cooperative activity of plasma membrane-bound TNF and IL-15. Blood 116: 575–583 blood-2009-08-240325 [pii];10.1182/blood-2009-08-240325 [doi].
  28. 28. Harris KM (2011) Monocytes differentiated with GM-CSF and IL-15 initiate Th17 and Th1 responses that are contact-dependent and mediated by IL-15. J Leukoc Biol 90: 727–734 jlb.0311132 [pii];10.1189/jlb.0311132 [doi].
  29. 29. Ben AM, Belhadj HN, Moes N, Buyse S, Abdeladhim M, et al. (2009) IL-15 renders conventional lymphocytes resistant to suppressive functions of regulatory T cells through activation of the phosphatidylinositol 3-kinase pathway. J Immunol 182: 6763–6770 182/11/6763 [pii];10.4049/jimmunol.0801792 [doi].
  30. 30. Littman DR, Rudensky AY (2010) Th17 and regulatory T cells in mediating and restraining inflammation. Cell 140: 845–858 S0092-8674(10)00174-1 [pii];10.1016/j.cell.2010.02.021 [doi].
  31. 31. Rodrigues L, Nandakumar S, Bonorino C, Rouse BT, Kumaraguru U (2009) IL-21 and IL-15 cytokine DNA augments HSV specific effector and memory CD8+ T cell response. Mol Immunol 46: 1494–1504 S0161-5890(08)00808-0 [pii];10.1016/j.molimm.2008.12.033 [doi].
  32. 32. Skov S, Bonyhadi M, Odum N, Ledbetter JA (2000) IL-2 and IL-15 regulate CD154 expression on activated CD4 T cells. J Immunol 164: 3500–3505. ji_v164n7p3500 [pii].
  33. 33. Correia MP, Costa AV, Uhrberg M, Cardoso EM, Arosa FA (2011) IL-15 induces CD8+ T cells to acquire functional NK receptors capable of modulating cytotoxicity and cytokine secretion. Immunobiology 216: 604–612 S0171-2985(10)00179-8 [pii];10.1016/j.imbio.2010.09.012 [doi].
  34. 34. Gagnon J, Ramanathan S, Leblanc C, Cloutier A, McDonald PP, et al.. (2008) IL-6, in synergy with IL-7 or IL-15, stimulates TCR-independent proliferation and functional differentiation of CD8+ T lymphocytes. J Immunol 180: 7958–7968. 180/12/7958 [pii].
  35. 35. Bezbradica JS, Medzhitov R (2009) Integration of cytokine and heterologous receptor signaling pathways. Nat Immunol 10: 333–339 ni.1713 [pii];10.1038/ni.1713 [doi].
  36. 36. Guo L, Junttila IS, Paul WE (2012) Cytokine-induced cytokine production by conventional and innate lymphoid cells. Trends Immunol 33: 598–606 S1471-4906(12)00128-7 [pii];10.1016/j.it.2012.07.006 [doi].
  37. 37. Spits H, Di Santo JP (2011) The expanding family of innate lymphoid cells: regulators and effectors of immunity and tissue remodeling. Nat Immunol 12: 21–27 ni.1962 [pii];10.1038/ni.1962 [doi].
  38. 38. Tough DF, Borrow P, Sprent J (1996) Induction of bystander T cell proliferation by viruses and type I interferon in vivo. Science 272: 1947–1950.
  39. 39. Boyman O (2010) Bystander activation of CD4+ T cells. Eur J Immunol 40: 936–939 10.1002/eji.201040466 [doi].
  40. 40. Kotaja N, Sassone-Corsi P (2004) Plzf pushes stem cells. Nat Genet 36: 551–553 10.1038/ng0604-551 [doi];ng0604-551 [pii].
  41. 41. Alonzo ES, Sant'Angelo DB (2011) Development of PLZF-expressing innate T cells. Curr Opin Immunol 23: 220–227 S0952-7915(10)00231-1 [pii];10.1016/j.coi.2010.12.016 [doi].
  42. 42. Quah BJ, Warren HS, Parish CR (2007) Monitoring lymphocyte proliferation in vitro and in vivo with the intracellular fluorescent dye carboxyfluorescein diacetate succinimidyl ester. Nat Protoc 2: 2049–2056 nprot.2007.296 [pii];10.1038/nprot.2007.296 [doi].
  43. 43. Akira S, Takeda K (2004) Toll-like receptor signalling. Nat Rev Immunol 4: 499–511 10.1038/nri1391 [doi];nri1391 [pii].
  44. 44. Savage AK, Constantinides MG, Han J, Picard D, Martin E, et al. (2008) The transcription factor PLZF directs the effector program of the NKT cell lineage. Immunity 29: 391–403 S1074-7613(08)00337-3 [pii];10.1016/j.immuni.2008.07.011 [doi].
  45. 45. Kreslavsky T, Savage AK, Hobbs R, Gounari F, Bronson R, et al. (2009) TCR-inducible PLZF transcription factor required for innate phenotype of a subset of gammadelta T cells with restricted TCR diversity. Proc Natl Acad Sci U S A 106: 12453–12458 0903895106 [pii];10.1073/pnas.0903895106 [doi].
  46. 46. Lee YJ, Jeon YK, Kang BH, Chung DH, Park CG, et al. (2010) Generation of PLZF+ CD4+ T cells via MHC class II-dependent thymocyte-thymocyte interaction is a physiological process in humans. J Exp Med 207: 237–246 jem.20091519 [pii];10.1084/jem.20091519 [doi].
  47. 47. Weinreich MA, Odumade OA, Jameson SC, Hogquist KA (2010) T cells expressing the transcription factor PLZF regulate the development of memory-like CD8+ T cells. Nat Immunol 11: 709–716 ni.1898 [pii];10.1038/ni.1898 [doi].
  48. 48. Kovalovsky D, Alonzo ES, Uche OU, Eidson M, Nichols KE, et al. (2010) PLZF induces the spontaneous acquisition of memory/effector functions in T cells independently of NKT cell-related signals. J Immunol 184: 6746–6755 jimmunol.1000776 [pii];10.4049/jimmunol.1000776 [doi].
  49. 49. van Amelsfort JM, Jacobs KM, Bijlsma JW, Lafeber FP, Taams LS (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 10.1002/art.20499 [doi].
  50. 50. Buckner JH (2010) Mechanisms of impaired regulation by CD4(+)CD25(+)FOXP3(+) regulatory T cells in human autoimmune diseases. Nat Rev Immunol 10: 849–859 nri2889 [pii];10.1038/nri2889 [doi].
  51. 51. Jones SA, Horiuchi S, Topley N, Yamamoto N, Fuller GM (2001) The soluble interleukin 6 receptor: mechanisms of production and implications in disease. FASEB J 15: 43–58 10.1096/fj.99-1003rev [doi];15/1/43 [pii].
  52. 52. Rose-John S, Waetzig GH, Scheller J, Grotzinger J, Seegert D (2007) The IL-6/sIL-6R complex as a novel target for therapeutic approaches. Expert Opin Ther Targets 11: 613–624 10.1517/14728222.11.5.613 [doi].
  53. 53. Assier E, Boissier MC, Dayer JM (2010) Interleukin-6: from identification of the cytokine to development of targeted treatments. Joint Bone Spine 77: 532–536 S1297-319X(10)00198-3 [pii];10.1016/j.jbspin.2010.07.007 [doi].
  54. 54. Schiff MH, Burmester GR, Kent JD, Pangan AL, Kupper H, et al. (2006) Safety analyses of adalimumab (HUMIRA) in global clinical trials and US postmarketing surveillance of patients with rheumatoid arthritis. Ann Rheum Dis 65: 889–894 ard.2005.043166 [pii];10.1136/ard.2005.043166 [doi].
  55. 55. Fleming JO (2013) Helminth therapy and multiple sclerosis. Int J Parasitol 43: 259–274 S0020-7519(12)00315-3 [pii];10.1016/j.ijpara.2012.10.025 [doi].
  56. 56. Itsumi M, Yoshikai Y, Yamada H (2009) IL-15 is critical for the maintenance and innate functions of self-specific CD8(+) T cells. Eur J Immunol 39: 1784–1793 10.1002/eji.200839106 [doi].
  57. 57. Niedbala W, Wei X, Liew FY (2002) IL-15 induces type 1 and type 2 CD4+ and CD8+ T cells proliferation but is unable to drive cytokine production in the absence of TCR activation or IL-12/IL-4 stimulation in vitro. Eur J Immunol 32: 341–347 10.1002/1521-4141(200202)32:2<341::AID-IMMU341>3.0.CO;2-X [pii];10.1002/1521-4141(200202)32:2&#60;341::AID-IMMU341&#62;3.0.CO;2-X [doi].
  58. 58. Fehniger TA, Suzuki K, Ponnappan A, VanDeusen JB, Cooper MA, et al. (2001) Fatal leukemia in interleukin 15 transgenic mice follows early expansions in natural killer and memory phenotype CD8+ T cells. J Exp Med 193: 219–231.