Abstract
Blockade of IL-2R with humanized anti-CD25 antibodies, such as daclizumab, inhibits TH2 responses in human T cells. Recent murine studies have shown that IL-2 also plays a significant role in regulating TH2 cell differentiation by activated STAT5. To explore the role of activated STAT5 in the TH2 differentiation of primary human T-cells we studied the mechanisms underlying IL-2 regulation of C-MAF expression. Chromatin immunoprecipitation studies revealed that IL-2 induced STAT5 binding to specific sites in the C-MAF promoter. These sites corresponded to regions enriched for markers of chromatin architectural features in both resting CD4 and differentiated TH2 cells. Unlike IL-6, IL-2 induced C-MAF expression in CD4 T cells with or without prior TCR stimulation. TCR-induced C-MAF expression was significantly inhibited by treatment with daclizumab or a JAK3 inhibitor, R333. Furthermore, IL-2 and IL-6 synergistically induced C-MAF expression in TCR activated T cells, suggesting functional co-operation between these cytokines. Finally, both TCR-induced early IL-4 mRNA expression and IL-4 cytokine expression in differentiated TH2 cells was significantly inhibited by IL-2 receptor blockade. Thus, our findings demonstrate the importance of IL-2 in TH2 differentiation in human T cells and support the notion that IL-2R directed therapies may have utility in the treatment of allergic disorders.
INTRODUCTION
Signal transducers and activators of transcription (STAT) proteins are activated by a variety of cytokines, growth factors and hormones. They comprise an evolutionarily conserved family of seven proteins in the mammalian genome (1). These proteins regulate vital cellular functions such as proliferation, survival and differentiation. The two STAT5 proteins, STAT5a and STAT5b, are activated by members of the γc family of cytokines (IL-2, IL-4, IL-7, IL-9, IL-15 and IL-21), which together regulate lymphoid development, differentiation and survival of various components of the immune system (2). Studies with knockout and transgenic mice have shown that Stat5a/5b are essential for the development and/or homeostatic maintenance of T cells, including CD8, γδ TCR, CD4+CD25+ FoxP3+ Regulatory T cells (Treg), and most importantly for our studies, the selective differentiation of CD4 T helper (TH) cells (3-8).
T cell differentiation involves epigenetic changes in lineage-associated genes by covalent modifications of DNA and histones and the histone variant H2A.Z (9). Transcriptionally active regions of chromatin are generally enriched with several modifications of histones such as mono-, di-, and tri-methylation of H3K4 (8, 10, 11) and H2A.Z (12), which differentially demarcate promoter and enhancer regions or regions of nucleosomal instability, respectively. A number of different STAT proteins, including Stat5 interact with transcriptional regulatory regions and are known to regulate T cell differentiation by enhancing or repressing key genes involved in these processes (13). TH2 differentiation in both mouse and human CD4 T cells is critically dependent on IL-2 (14, 15). Consistently, Stat5a knockout mice show defective TH2 responses and decreased IL-4 production, while a constitutively active Stat5a mutant can restore IL-4 production in IL-2-deficient CD4 T cells and TH2 differentiation in IL-4Rα-deficient CD4 T cells (6, 16). IL-2-activated Stat5 is necessary for increased transcription and cell surface expression of IL-4R in differentiating TH2 cells (17), and for appropriate chromatin remodeling to enhance accessibility of the murine Il-4 locus (16, 18). Additionally, genome–wide analysis of Stat5 DNA binding in fully differentiated murine TH2 cells reveals several probable Stat5 binding sites, suggesting that Stat5 can potentially regulate numerous TH2 associated factors (17).
The c-maf proto-oncogene was the first lineage-specific factor identified for TH2 cells and belongs to the AP1 family of proteins (19). It binds to a MARE (Maf recognition element) site in the Il-4 promoter and directly transactivates Il-4 gene transcription (19). Over-expression of c-maf in murine TH1 clones induces low levels of endogenous IL-4 synthesis, while transgenic mice overexpressing CD4-specific c-maf preferentially develop a TH2 phenotype and have attenuated production of the TH1 cytokine IFN-γ(20). Recent studies have also shown that c-maf is required for the efficient development of murine T follicular Helper (TfH) and TH17 lineages, as well as for the production of IL-10 by TH17 cells (21-23). Thus, c-maf plays essential roles in the differentiation and function of multiple effector T cell lineages. In murine T-cells, c-maf expression is regulated by IL-6-activated Stat3 (24). However, relatively little is known about the regulation of C-MAF transcription during early human T cell activation prior to differentiation.
In this study, we show for the first time that in primary human CD4 T cells, C-MAF expression is regulated by IL-2 receptor (IL-2R) mediated STAT5 activation, independently of TCR signaling. We elucidate upstream regions of the C-MAF gene containing epigenetic modifications corresponding to transcriptional enhancer regions in undifferentiated and fully differentiated TH1 and TH2 cells and reveal that these are stably maintained irrespective of the differentiation state. We show that IL-2 induces in vivo STAT5 binding to GAS (IFNγ-activated sequences) motifs located within and around these regions of stable epigenetic modifications, and characterize their role in regulating IL-2-induced C-MAF transcription. Furthermore, we show that blockade of IL-2R has a profound inhibitory effect on IL-4 production during in vitro TH2 differentiation. These findings indicate that IL-2 plays an important role in the initiation of TH2 differentiation in human T cells and support the notion that IL-2R directed therapies may have utility in the treatment of allergic disorders (15).
MATERIALS AND METHODS
PBMC and CD4 T cell isolation, culture and treatment
PMBCs were isolated from buffy coats from healthy donors (purchased from National Blood Service, Tooting, UK) by density gradient centrifugation using LSM 1077 (PAA laboratories). CD4 T cells were purified from PBMCs by positive selection with human CD4 microbeads (Miltenyi Biotec) according to manufacturer’s instructions. The purity of CD4 T cells ranged from 93% to 96% by FACS analysis using FITC-conjugated αCD3 and PE-conjugated αCD4 mAbs (both from DAKO) on a FACScalibur instrument (Becton Dickinson). Cells were maintained in culture medium (CM) of RPMI 1640 medium (PAA Laboratories, Austria) supplemented with 10% fetal bovine serum (FBS, PAA laboratories), 2mM L-glutamine and penicillin-streptomycin (100 IU/ml and 100μg/ml, both from Sigma-Aldrich, UK). When indicated, CD4 T cells were stimulated by 2 μg/ml PHA-L (EY laboratories, CA) for 72h and rested in CM for ≥16 hours prior to activation with 100U/ml IL-2 (Roche Diagnostics). CD4 T cells were activated via the TCR by stimulation with plate-bound αCD3 (UCHT1, 10 μg/ml) and soluble αCD28 antibody (2 μg/ml; both from Ancell) with or without the JAK3 inhibitor R333 (5μM; a generous gift from Rigel Pharamceuticals, CA). IL-6 (R&D Systems) was used at 10 ng/ml for stimulation experiments. CD4 T cells were inhibited by treatment with anti-human IL-2 antibody (10 μg/ml; Biosource Inc.) or HAT (daclizumab, 10 μg/ml; kindly provided by Hoffmann-La Roche, NJ), respectively.
Chromatin immunoprecipitation (ChIP)-cloning and ChIP-seq
ChIP-cloning experiments were performed using 1.5 × 107 CD4 T cells stimulated with IL-2 for 20 minutes followed by a 10 minute crosslink with formaldehyde (0.37%) at 37 °C, as previously described (25). The sonicated chromatin was immunoprecipitated for an hour using μMACS Protein-A microbeads (Miltenyi Biotec) and anti-Stat5a or anti-Stat5b monoclonal or control mouse IgG antibodies (all from Zymed). Purified chromatin was blunt-end repaired using T4 DNA polymerase (NEB), ligated to linkers, PCR amplified, and cloned into TOPO vector (Invitrogen) and sequenced (Cogenics).
For identification of the sequences enriched in regions associated with specific histone phenotypes (ChIP-seq experiments), ChIP was performed on native chromatin from ex vivo differentiated human TH2 cells (26). Chromatin was prepared by using the protocol of Feil and colleagues with some minor modifications (http://www.epigenome-noe.net/researchtools/protocol.php?protid=2). Briefly, nuclei were treated with micrococcal nuclease (10U/μl, 7 min) and mono and dinucleosomal chromatin was recovered. Chromatin quality was assessed by agarose gel electrophoresis and semi-quantitated using a nanodrop. Chromatin (20μg) was immunoprecipitated with 3μg anti-H3K4me3 (ab8580) or anti-IgG control, and magnetic Protein G beads (Active Motif). Prior to sequencing, the diversity of sequences enriched by ChIP was assessed by comparing qPCR of input DNA with anti-IgG or anti-H3K4me3 immunoprecipitated DNA using multiple probe sets (ChIP-qPCR). ChIP libraries were prepared from amplified DNA (17 cycles) from two biological replicates of TH2 cells according to the manufacturer’s protocol (Illumina). DNA from separate experiments was sequenced on GAII genome analyzers (BRC Core Facility KCL). Detection of regions of enriched H3K4me3 relative to input DNA was performed by using MACS and SICER programs. Output was converted to browser extensible data (BED) files displaying the number of Tags in 200bp windows and visualized within the UCSC browser.
Preparation of cell extracts and Western blotting
Nuclear and total cell extracts were prepared as described previously (27). Samples were resolved on 8% SDS–PAGE gels and transferred to Immobilon PVDF membrane (Millipore). Western blots were performed using pan-STAT5 (Invitrogen), anti-phospho-STAT5Y694 (New England Biolabs), c-maf polyclonal antibody (M-153, Santa Cruz Biotechnology), anti-Lamin-B or anti-tubulin antibodies (both from Invitrogen) and developed using ECL Plus chemiluminescence reagent (GE Healthcare).
RNA Isolation and Quantitative real time PCR
Total RNA was extracted using TRIzol Reagent (Invitrogen). RNA (250 ng) was reverse-transcribed to cDNA using RevertAid-H Minus M-MuLV Reverse Transcriptase (Fermentas) following manufacturer’s instructions. qRT-PCR was performed in triplicate using 2X Precision SYBR Green MasterMix (PrimerDesign Ltd, UK) in a total reaction volume of 20 μl using the ABI Prism 7900 HT Sequence Detection System (Applied Biosystems) although ChIP-qPCR reactions were performed in duplicate. The human PPIA or 18S rRNA genes were used as reference controls for the various experiments. Ct values were exported with the sequence detector SDS 2.3 software (ABI). Gene-specific primers for PPIA, 18S rRNA, C-MAF, CD25 and IL-2 were obtained from PrimerDesign Ltd. UK. The sequences of all other primers are given in Figure S3.
Luciferase Reporter Assay
HEK293T cells were cultured and transfected to reconstitute IL-2R signaling using the calcium phosphate method, as previously described (28). Trimerized GAS1-4 sequences were synthesized (MWG), cloned into pGL4.23 vector (Promega Corp.), and verified by sequencing (Cogenics). Luciferase assays were performed by using the Dual-Glo luciferase Kit (Promega Corp.), according to manufacturer’s instructions. Firefly luciferase activities were standardized to the corresponding Renilla luciferase activities for each sample to control for transfection efficiencies.
TH2 skewing and Intracellular cytokine staining
Fresh cord blood units (CBU) were obtained from the Programa Concordia Banc de Sang i Teixits, Barcelona, Spain, with prior consent and ethical committee approval. CBU were diluted 1:1 with RPMI 1640 (Lonza) supplemented with 0.6% trisodium citrate and 40 nM 2-ME. Cord blood mononuclear cells were separated by density gradient centrifugation using Ficoll-Paque Premium (GE Healthcare, UK). CD4 T cells were isolated by negative selection using a Miltenyi isolation kit. Mean (±s.d.) positivity for CD45RA on isolated CD4 cells was 93% ± 5% (n=3). Cord blood CD4 cells (0.5-1 million) were activated with IL-2 as follows: αCD3 (10 μg/ml), αCD28 (2 μg/ml; both BD) and IL-2 (30U/ml) ± HAT (10 μg/ml). TH2 skewing was performed as previously described (26) 0.5-1 million cord blood CD4 cells stimulated with αCD3 (10 μg/ml), αCD28 (2 μg/ml), IL-2 (30U/ml), IL-4 (12.5ng/ml; NBS Biologics), anti-IFN-γ and anti-IL-10 (both 5μg/ml, BD) ± HAT (10 μg/ml).
Intracellular staining for IL-4, and IFN-γ was carried out according to standard methods. Briefly, cultured cells were restimulated with Phorbol Myristate Acetate (PMA; 50ng/ml) and Ionomycin (1mM, both Sigma) for 4.5h at 37°C in the presence of Brefeldin A (3 μg/ml; eBioscience) in 1ml of CM. Washed cells were fixed with 4% PFA, blocked with 50% FCS (v/v in PBS), permeabilized with 0.1% saponin (w/v in 1% FCS in PBS) and stained for 30 min in the dark at 4°C with monoclonal antibodies against IL-4 (BD) and IFN-γ (Caltag) or appropriate isotype controls (mouse IgG1-PE (BD) and mouse IgG1-APC (Caltag)) at recommended concentrations. Within 4h of staining, data from live cells, based on forward/side scatter profiles, were acquired on a FACSCalibur flow cytometer (CellQuest software; BD) and analyzed with FlowJo (Treestar Inc, Ashland, OR).
Data analysis
Data analysis utilized Microsoft Excel (Microsoft Corporation, USA) and GraphPad Prism v4 (GraphPad Software, Inc.). Statistical analysis was performed by using paired parametric and non-parametric tests as appropriate. p values of < 0.05 were considered to be statistically significant.
RESULTS
IL-2 activated STAT5 binds to the C-MAF locus in vivo in CD4 T cells
IL-2 is one of the earliest cytokines produced by activated T cells and mediates its actions primarily through the activation of STAT5 proteins. A STAT5-chromatin immunoprecipitation assay (ChIP) was performed using chromatin from freshly isolated CD4 T cells to identify in vivo IL-2-activated STAT5 gene targets. The immunoprecipitated chromatin yielded a number of distinct clones based on sequencing. One clone mapped to chromosome 16 at −152,916 to −153,096 upstream of the C-MAF gene, and contained a consensus GAS motif (Figure 1A). Since IL-2 and STAT5 play significant roles in gene regulation during TH2 cell differentiation, we examined if IL-2-induced STAT5 also regulates C-MAF gene expression.
FIGURE 1.
STAT5 binding sites in the 5′-upstream region of C-MAF were located within or adjacent to architectural regions of chromatin in resting and TH2-differentiated CD4 T cells. A) Sequence of the cloned fragment obtained from STAT5-ChIP assay. Bold: a consensus GAS motif. Italics: a putative imperfect GAS motif, positioned at an optimal distance to facilitate tetrameric STAT5 binding. B) STAT5 binding to the cloned sequence. ChIP was performed on untreated or IL-2-stimulated fresh CD4 T cells by using anti-STAT5a or -STAT5b specific antibodies, or a negative control IgG antibody. Input is a sample of total chromatin taken before immunoprecipitation. C) Chromatin architecture of the C-MAF 5′-upstream regulatory region and positions of associated GAS motifs. Custom Track View (hg 16 build) of DHS sites (DHS1-3), histone modifications H3K4me1, H3K4me3, and H2A.Z in resting CD4 T cells, H3K4me3 in TH1 and TH2 cells, and positions of CpG islands in this region of chromosome 16 upstream of the TSS of C-MAF gene. The positions of consensus GAS motifs (GAS1-5) associated with the three DHSs are also indicated.
In vivo binding of STAT5 was assessed in cross-linked chromatin fragments derived from either IL-2-stimulated or untreated freshly isolated CD4 T cells by immunoprecipitation with anti-STAT5 antibodies in a ChIP assay. DNA was isolated from precipitated chromatin and assayed for the presence of the upstream C-MAF sequence by using PCR with specific primers that detect the aforementioned cloned fragment. IL-2 treatment induced binding of STAT5a and STAT5b to this sequence (Figure 1B). As expected, this sequence was not enriched in the IL-2-treated sample by ChIP performed with control IgG (Figure 1B). Thus, there was specific IL-2-induced binding of STAT5 proteins to a sequence upstream of the C-MAF transcriptional start site (TSS). Since this sequence is located at a long distance upstream of the TSS of the C-MAF gene, we next determined if IL-2 induced STAT5 binds to other 5′ upstream regions that were closer to the C-MAF promoter. We postulated that transcriptionally relevant binding sites would be located within cell-type specific DNase1 hypersensitive (DHS) sites (29, 30).
The genome-wide DHS study of CD4 T cells identified three DHS sites (DHS1-3) in the C-MAF 5′-upstream region (Figure 1C) (30). DHS1 encompasses a broad region containing three peaks located between 78,189,000 and 78,194,500 nucleotides (−2388bp upstream of TSS). DHS2 is between 78,232,000 and 78,233,000 (−39,888bp upstream of TSS) and DHS3 is between 78,361,500 and 78,362,500 (−169,388bp upstream of TSS). Analysis of the sequences within and around the three DHS sites revealed the presence of 5 consensus GAS motifs. GAS1 and GAS5 were within the DHS1-3 peaks whereas GAS2, GAS3, and GAS4 were nearby (Figure 1C). Analysis of the sequence of these GAS motifs revealed strong evolutionary conservation of GAS1 and GAS2, suggesting that these may be important regulatory sequences, while GAS3-5 was less well conserved (Figure S1).
To understand the if these GAS motifs coincided with transcriptionally active chromatin during T cell differentiation, we undertook ChIP-seq analysis of fully differentiated TH1 and TH2 cells using an anti-H3K4me3 antibody and compared sites of enrichment with previously published genome-wide CD4 ChIP-seq data for H3K4me1, H3K4me3 and H2AZ enrichment (10). These studies revealed that GAS1, 2, 4 and 5 were located within regions enriched for monomethylated histone H3K4 while GAS 1, 2 and 4 were also coincident with regions enriched for H3K4me3 in both TH1 and TH2 cells (Figure 1C). All of the GAS motifs coincided with regions enriched for binding of the histone H2 variant H2A.Z. Thus, despite being located a long distance upstream of the C-MAF TSS, the distal GAS motifs mapped to regions containing chromatin modifications consistent with potential enhancer function.
The TSS (left) and the 5′-upstream region of the c-maf gene is depicted with the aforementioned sequences and positions of DHS-associated GAS motifs (GAS1-5), and a negative control GAS6, which is a random, consensus GAS motif (Figure 2A). To determine if IL-2 induces in vivo binding of STAT5 to each of the GAS motifs, we performed a ChIP analysis with anti-STAT5a or control antibody on unstimulated or IL-2-stimulated, PHA-activated human CD4 T cells. The immunoprecipitated chromatin was analyzed for the specific enrichment of GAS1-6 motifs by quantitative real-time PCR analysis (qRT-PCR). STAT5a bound to GAS 1-4 in a specific IL-2-inducible manner, but not to GAS5 or the negative control GAS6 (Fig. 2B). The non-specific IgG control did not enrich chromatin with GAS1-6 sequences in IL-2-stimulated cells by ChIP (Fig. 2B). Thus, IL-2 induced STAT5 binding to multiple sites in the C-MAF promoter at varying distances 5′ to the TSS, and these sites coincided with markers of nucleosomal instability and potential enhancer function. We consistently noted that the lowest level of STAT5 binding was to GAS3, suggesting that this GAS motif alone is insufficient for optimum STAT5 binding. Comparison of the GAS3 sequence (TTCTTTGAA) with the optimal STAT5 binding site (TTC(T/C)N(G/A)GAA) reveals an imperfect match and may suggest a need for cooperative tetrameric STAT5 binding to this region. In this regard, we noted that there is an imperfect GAS motif spaced 7 nucleotides away from GAS3 (indicated in italics in Figure 1A), which may facilitate tetrameric STAT5 binding (28, 31).
FIGURE 2.
IL-2-induced STAT5 binding and the regulatory effects of GAS motifs in the C-MAF 5′ upstream region. A) Schematic outline of the chromosomal locations (bp) of GAS1-GAS5 and a negative control GAS motif (GAS6) relative to the C-MAF TSS on the left. B) Enrichment of chromatin from fresh CD4 T cells by ChIP with anti-STAT5a as compared to control IgG. Sequences were analyzed by qRT-PCR for the different GAS motifs indicated in A. Data are expressed as fold enrichment for IL-2-induced STAT5a binding for each site. Results are representative of three independent experiments from three different donors. C) IL-2 induced STAT5-dependent transcriptional activation of GAS1-, GAS2-, and GAS4-driven luciferase constructs in HEK293T cells reconstituted with IL-2Rβ, γc, JAK3 and STAT5b by transfection. Pooled results from three independent experiments are shown. * p<0.05.
To elucidate the STAT5-dependent enhancer activity of these GAS motifs, we assessed the ability of transfected STAT5b to activate constructs containing GAS motifs 1-4 in luciferase reporter assays performed in an IL-2R reconstitution system in HEK293T cells (28). These studies revealed that GAS1, GAS2, and GAS4-driven luciferase reporter constructs increased IL-2-induced, STAT5b-dependent transcriptional activation (Figure 2C). A similar pattern of transactivation of these reporter constructs was observed with STAT5a (data not shown). The GAS3-driven luciferase reporter construct was not transactivated by STAT5b, consistent with poor binding observed at this GAS motif (Figure 2B). Thus, IL-2 induced specific STAT5 binding and transcriptional activation from promoter proximal (GAS1, 2) and distal (GAS 4) GAS motifs within the C-MAF 5′-upstream region.
IL-2 specifically induces C-MAF gene expression
The role of IL-2 in C-MAF gene expression was elucidated by comparing C-MAF RNA levels from freshly isolated and PHA-treated CD4 T cells that were untreated or treated with IL-2 for 0.5-12 hrs. C-MAF mRNA expression in these samples was analyzed by qPCR. IL-2 significantly induced C-MAF mRNA expression in both freshly isolated (Figure 3A) and PHA-activated CD4 T cells (Figure 3B) between 2 to 6 hours after stimulation, and as early as 30 minutes after stimulation. To determine if IL-2 induction of C-MAF gene transcription was associated with a concomitant increase in protein expression, we prepared nuclear extracts from pre-activated CD4 T cells that were stimulated with IL-2 for the indicated times and examined levels of C-MAF, phosphorylated STAT5 (pYSTAT5), total STAT5, and the loading control Lamin-B1 by immunoblot analysis. As shown in Figure 3C, C-MAF protein expression was clearly increased by 6 hours after IL-2 treatment and followed the peak increase in tyrosine phosphorylated STAT5 protein in the nucleus at 30 minutes. Of note, we observed the sustained presence of IL-2-activated STAT5 in the nucleus for the duration of the experiment (24 hours) (Figure 3C). Taken together, C-MAF is induced without TCR stimulation by IL-2 alone in freshly isolated, as well as PHA-activated, CD4 T cells.
FIGURE 3.
C-MAF expression is specifically activated by IL-2. C-MAF transcription from A) freshly isolated or B) PHA- activated CD4 T cells that were stimulated with IL-2 for the indicated times were assessed by qRT-PCR. Expression of PPIA was used as the reference control. C-MAF mRNA levels are presented as fold increase over unstimulated cells. Pooled results from n=3 (A) and n=5 (B) different donors are shown; * p<0.05 relative to time zero. C) C-MAF protein expression was potently induced in pre-activated CD4 T cells by IL-2 treatment. IL-2 treatment activated STAT5 and induced nuclear translocation of phopho-STAT5 at 30 minutes. Note that the 30 min sample is loaded to the left of the zero time point sample on the gel. Nuclear extracts were analyzed by Western blot analysis for c-MAF, STAT5, pY-STAT5 or a loading control, Lamin-B1 expression. D) IL-2 did not induce expression of the genes WWOX or DNCL2B, which flank C-MAF. A schematic outline of the relative chromosomal locations (bp) of WWOX, C-MAF and DNCL2B is shown. qRT-PCR was performed on RNA from untreated or IL-2-treated freshly isolated CD4 T cells, using 18S rRNA expression as the reference gene. Pooled results from 4 independent donors are shown. Expression of WWOX and DNCL2B following IL-2 stimulation was not significantly different than that of control.
As the cloned sequence from the STAT5-ChIP maps to a 5′ region distal to the TSS of the C-MAF gene, we wished to determine if IL-2-induced STAT5 may regulate either of the flanking genes, namely WWOX (approximately 380Kb 3′ of C-MAF) or DNCL2B (approximately 940 Kb 5′ of C-MAF). We therefore performed qPCR analysis to examine the IL-2-induced expression of these two genes in CD4 T cells. Neither WWOX nor DNCL2B were induced by IL-2 stimulation (Figure 3D). Thus, IL-2 induced binding of STAT5 to upstream sequences specifically activates the C-MAF gene.
TCR+CD28-mediated activation of C-MAF transcription is dependent on IL-2R signaling
In the above studies, exogenous IL-2 alone efficiently stimulated C-MAF expression in the absence of TCR stimulation. Thus, we next examined whether TCR activation of C-MAF was dependent on IL-2 signaling by using clinically relevant inhibitors of the IL-2 receptor (HAT) or signaling molecule JAK3.
HAT (daclizumab) binds to the α-chain (CD25) of the high affinity IL-2R, inhibiting binding of IL-2 to the receptor complex and causing efficient blockade of JAK3/STAT5 activation (15). CD4 T cells were isolated from buffy coats and stimulated with αCD3+αCD28 in the presence or absence of HAT for the indicated times. As shown in Figure 4A, C-MAF expression is significantly induced at 17 hours after stimulation and HAT blocked this induction. Thus, the αCD3+αCD28 stimulated early activation of C-MAF is dependent on IL-2/IL-2R signaling.
FIGURE 4.
TCR-induced C-MAF expression is dependent on IL-2 signaling via the JAK3/STAT5 pathway. A) CD4 T cells were stimulated by αCD3+αCD28 treatment in the presence or absence of HAT (10μg/ml), and mRNA was prepared at various time-points after stimulation and analyzed by qRT-PCR for C-MAF gene expression using PPIA as reference control. C-MAF mRNA levels are presented as fold increase over that of unstimulated cells. Results are from 3 independent donors; * p<0.05. B) and C) CD4 T cells were stimulated with αCD3+αCD28 in the presence or absence of the JAK3 inhibitor R333 (5 μM) for the indicated times. B) STAT5 is activated in a JAK3-dependent manner following TCR stimulation. Whole cell extracts were prepared from cells after treatment. The activation of STAT5 (pY-STAT5) was determined by western blot analysis using anti-phosphotyrosine-STAT5 and pan-STAT5 antibodies and as a loading control, anti-tubulin antibody. The lack of pSTAT5 signal at 6 hours was due to the loss of sample during preparation, as indicated by the anti-tubulin western blot. C) Activation of C-MAF transcription by TCR-stimulation requires JAK3 activity. qRT-PCR analysis was performed to detect the mRNA expression of C-MAF, CD25 and IL-2 genes using PPIA as the reference gene. mRNA expression of each of the genes is presented as fold increase over unstimulated cells. The results show pooled data from 4 independent experiments. * p <0.05.
Because IL-2 induction of C-MAF during T cell activation may specifically involve activation of the JAK3/STAT5 pathway or may utilize additional IL-2 dependent pathways, the effects of a potent JAK3 inhibitor R333 (32) during TCR activation was examined. Fresh CD4 T cells were purified and stimulated with αCD3+αCD28 in the presence or absence of R333; and total cell extracts and RNA were prepared for further analysis. The effect of R333 on activation of STAT5 was investigated by western blot analysis of total cell extracts using phospho-STAT5 and pan-STAT5 antibodies. STAT5 activation (pYSTAT5) was detectable as early as 4 hours, increased by 17 hours, and was blocked by R333 (Figure 4B). Thus, the JAK3-specific inhibitor R333 efficiently inhibited STAT5 activation.
RNA samples were analyzed by real-time PCR for C-MAF, IL-2 and CD25 expression. As noted in Figure 4A, αCD3+αCD28 activation of freshly isolated CD4 T cells increased C-MAF transcription at 17 hours after stimulation. JAK3 inhibitor R333 blocked the induction of C-MAF transcription (Figure 4C). The increase in CD25 and IL-2 expression at 17 hours post-treatment, following peak IL-2 expression at 6-8 hours (data not shown) was also significantly reduced by R333 (Figure 4C). Taken together, these data indicated that the induction of C-MAF transcription during T cell activation was dependent on IL-2 signaling and JAK3/STAT5 activation in CD4 T cells.
IL-2 and IL-6 functionally synergize to potently induce C-MAF gene expression
As both IL-2 and IL-6 can independently activate C-MAF expression and subsequently promote TH2 differentiation, we examined if these cytokines functionally cooperate in the regulation of C-MAF. Purified CD4 T cells were stimulated as above with αCD3+αCD28 or in the co-presence of IL-2 and/or IL-6. As expected, C-MAF transcription was induced by αCD3+αCD28 stimulation, and addition of either cytokine alone modestly increased this induction. However, the presence of both IL-2 and IL-6 significantly increased C-MAF transcription in a synergistic manner (Figure 5A). The presence of an inhibitory antibody to IL-2 or CD25 or the JAK3 inhibitor R333 blocked the synergy, indicating the increase was dependent on IL-2/IL-2R signaling.
FIGURE 5.
IL-2 and IL-6 synergized to potently induce transcription of C-MAF, but not GATA3. CD4 T cells were stimulated with αCD3/αCD28 for 17 hours in the presence of the indicated cytokines (IL-2, IL-6) and/or inhibitors. qRT-PCR was performed to assess mRNA expression of A) C-MAF and GATA3 and B) IL-4 genes. Expression of each of the genes is presented as fold increase over unstimulated cells. In A) and B) n=5; * p<0.05.
To understand if this synergistic effect extended to induction of GATA3, the signature transcription factor of TH2 cells, we quantified GATA3 message in purified CD4 T cells stimulated as above with αCD3+αCD28 or in the co-presence of IL-2 and/or IL-6. In contrast to C-MAF, we did not observe any further increase in αCD3+αCD28 induced GATA3 mRNA expression by IL-2 or the combination of IL-2+IL-6 (Figure 5A). Thus, C-MAF transcription is specifically activated by IL-2 signaling during early CD4 T cell activation.
IL-4 expression is dependent on IL-2 signalling during T cell activation
The key TH2 cytokine gene IL-4 is directly regulated by c-maf (19). We wished to clarify the role of IL-2 signaling in IL-4 activation during T cell activation. CD4 T cells were stimulated with αCD3+αCD28 and IL-2 and IL-6 in the presence or absence of the inhibitors anti-IL-2, HAT, or R333. As expected, IL-4 expression is potently induced by the activating stimuli (Figure 5B). However, the addition of anti-IL-2, or HAT, or R333 significantly reduced IL-4 gene expression, indicating that IL-2R signaling is important for activation of IL-4 transcription during T cell activation (Figure 5B).
IL-2 signalling is required for IL-4 production during TH2 cell differentiation
To determine whether IL-2 signaling was required for IL-4 protein expression during in vitro TH2 cell polarization, we purified naïve CD4 T cells from cord blood samples from three donors and performed in vitro differentiation experiments under TH2 polarizing conditions or TCR (αCD3+αCD28) +IL-2 stimulation, in the presence or absence of the IL-2R-blocking HAT antibody. Fourteen days after treatment, cells were analyzed for cytokine production by flow cytometry. A representative plot is depicted in Figure 6A and the pooled results from all three donors are presented in Figure 6B. As expected, αCD3+αCD28 activation of naïve T cells induced the production of both key TH1 (IFN-γ) and TH2 cytokines (IL-4) (Figure 6A) (26). HAT treatment significantly reduced the percentage of IFN-γ and IL-4 expressing cells, consistent with previous studies using PBMC (15). After 14 days under TH2 conditions, CD4 T cells produced predominantly IL-4, not IFN-γ (Figure 6A). In the presence of HAT, IL-4 was significantly inhibited by approximately 90% in all three donors (Figure 6A and B). Furthermore, if exogenous IL-2 was omitted in the skewing experiments, the cells were unable to polarize to IL-4-producing cells (supplementary Figure S2). Thus, these results indicated that IL-2 signaling plays an important role in the in vitro TH2 differentiation of naïve CD4 T cells.
FIGURE 6.
HAT inhibited IL-4 protein expression during in vitro TH2 differentiation of naïve CD4 T cells. A) Cord blood CD4 T cells were stimulated with αCD3/αCD28±HAT or under TH2 skewing conditions ±HAT for 14 days. Representative intracellular staining for IFNγ and IL-4 in cord blood CD4 T cells that were activated in vitro under TH2 skewing conditions (see materials and methods section) is shown from one of three independent donors. B) Cumulative data from 3 experiments showing mean percentage ± s.d. of cells positive for each cytokine. * p<0.05.
DISCUSSION
The pleiotropic actions of IL-2, one of the earliest cytokines produced following T cell activation, are central to the propagation of the ensuing immune response (33). During human T cell activation, functional blockade of the IL-2R with daclizumab blocks STAT5 activation and inhibits TH1 and TH2 cytokine production (15). We thus hypothesized that in humans IL-2/STAT5 may also play a critical role in the differentiation of both these lineages. In fully differentiated murine TH2 cells, target loci for Stat5 binding have recently been identified by genome wide ChIP-seq analysis. These include motifs in the promoters of known key regulators of TH2 differentiation in the mouse, such as il-4r, gata3 and c-maf (17). Because previous studies have revealed differences between TH1/TH2 differentiation in human and mouse, we sought to establish the role of IL-2/STAT5 in human TH2 differentiation (26). In this study, we show that IL-2 signaling plays a significant role in initiating C-MAF and IL-4 transcription during early T cell activation and thus subsequent IL-4 production during differentiation of human TH2-skewed human CD4 T cells.
The function of c-maf as an essential transcription factor required for IL-4-driven TH2 lineage commitment is well established (19, 20). However, the molecular mechanisms that regulate C-MAF expression prior to specific lineage differentiation are not well understood. We show that during early T cell activation IL-2 increases C-MAF gene expression by inducing STAT5 binding to GAS motifs located in the 5′-upstream region. Of note, the STAT5 binding sites are not only located proximal to the promoter but also at a large distance upstream of the TSS, and map within DHSs present in the genome of resting CD4 T cells (30). Moreover these binding sites coincide with regions of enrichment of the histone marks H2A.Z, H3K4me1 and H3K4me3, which indicate structural features such as enhancers and therefore imply regions of active chromatin. Thus, mechanistically, our studies indicated that the C-MAF locus is already in a transcriptionally permissive configuration in resting CD4 T cells. Furthermore, the H3K4me3 enrichment pattern for this locus in resting CD4 T cells (10) showed a similar pattern to that observed in TH1 and TH2 cells from our ChIP-seq analysis. Therefore, in different CD4 T cell lineages, similar regulatory regions appear to be involved in “priming’ the C-MAF gene for cytokine-specific transcriptional activation. c-maf is also important for murine TH17 differentiation, where it’s expression is regulated by IL-6-activated Stat3 (21). Interestingly, a recent genome-wide ChIP-seq study of Stat3 binding sites in differentiated murine TH17 cells revealed that Stat3 binds to a site that corresponds to the GAS1 region identified in our study (34). Of note, the chromatin architecture in this region appears to be very similar between WT and Stat3-/- mice, suggesting that the c-maf chromatin architecture is unaffected by TH17 lineage differentiation. This finding is consistent with our present observation in human CD4 T cells that there are no major changes in the C-MAF 5′-upstream chromatin architecture between resting and TH1 and TH2 differentiated human CD4 T cells, and supports the notion that the C-MAF locus is in an epigenetically active configuration irrespective of differentiation status, and is primed for transcription before specific lineage differentiation.
Additionally, we provide evidence that IL-2 alone can activate C-MAF in freshly isolated T cells in the absence of TCR stimulation, which we have also confirmed in CD4+CD25− T cells (unpublished observations). Moreover, activation of C-MAF occurs early during T cell activation, following increases in IL-2 and CD25 transcription, and is blocked by functional blockade of the IL-2R signaling (Figure 4). Therefore, IL-2 induced STAT5 is necessary for the expression of C-MAF during early T cell activation. This finding is consistent with the observation that vav-deficient mice, which have impaired IL-2 production, also show lower expression of both c-maf and IL-4 and subsequent TH2 development (35, 36). Murine studies have shown that c-maf is also induced by IL-6 via Stat3 activation, leading to increased il-4 expression and subsequent TH2 differentiation (24, 37). However, in contrast to IL-2, IL-6 activation of c-maf is dependent on TCR stimulation. Our data show that IL-2 and IL-6 can synergize to more potently induce human C-MAF gene expression, and that this synergy is significantly inhibited by blocking IL-2 or IL-2R. Thus, both IL-2-activated STAT5 and IL-6-activated STAT3 are important regulators of C-MAF gene expression.
The IL2/IL-2R blockade studies indicate that during T cell activation, IL-2 signaling is not only required for C-MAF expression but also for early expression of IL-4, indicating that early in differentiation IL-2 plays an important role in creating an environment that favors TH2 polarization. We confirmed this functionally by showing that blockade of high affinity IL-2R with daclizumab significantly inhibited in vitro TH2 polarization of naïve CD4 T cells, as evidenced by the significant decrease in IL-4 expression. Furthermore, there was no TH2 differentiation of naïve T cells when IL-2 was omitted in the in vitro polarization experiments (Figure S2). Previously, murine studies have shown that ICOS-ICOSL interactions are important for c-maf and il-2 expression and subsequent TH2 differentiation. These studies showed that ICOS (i) can be induced in murine cells by IL-4, (ii) enhances IL-2 but not IL-4 production following optimal stimulation through CD3 and CD28; and (iii) is only important during sub-optimal CD3/CD28 T cell stimulation or through CD3 + IL-2 alone (38-40). Since our experimental set-up is APC-independent, we have not dissected the contribution of ICOS-ICOSL interactions here in the αCD3+αCD28/IL-2–mediated induction of human C-MAF and IL-4. In this regard, it will be interesting to elucidate whether IL-4 and C-MAF induction by IL-2 is only important during sub-optimal CD4 T cell activation, similar to murine T cells (40).
While c-maf is important for TH2 differentiation, the critical master regulator of this lineage is GATA3. As in previous murine studies by others we did not find any evidence for the activation of human GATA3 transcription by IL-2 or IL-6 at early times after TCR activation. Similarly, previous murine studies showed that neutralization of IL-2 had no effect on gata3 expression (41). However, our data does not exclude the possibility that IL-2 may regulate GATA3 RNA and/or protein levels in differentiated TH2 cells. Hwang and colleagues have reported that c-maf enhances cd25 expression in developing murine TH2 cells (42), which is consistent with our observation that blockade of IL-2R during αCD3+αCD28 stimulation of CD4 T cells reduces CD25 expression. Since our present studies reveal an important role for IL-2 induced STAT5 in the activation of C-MAF, we speculate C-MAF may play a role in the differentiation of peripheral human Treg cells, whose maintenance and functional integrity are dependent on this pathway. We have observed that C-MAF transcription is induced by αCD3+αCD28 and/or IL-2 stimulation in peripheral human Tregs (unpublished observations). While the functional implications of this observation are presently unclear, it is suggestive of a role for C-MAF in Treg biology, and is supported by the finding that the aryl hydrocarbon receptor can interact with c-maf to promote the differentiation of type 1 regulatory cells in mice (43).
In summary, we have identified and characterized the molecular mechanism by which IL-2-induced STAT5 regulates the TH2 lineage-promoting transcription factor C-MAF in a TCR-independent manner. Furthermore, IL-2 synergizes with IL-6 during T cell activation to significantly promote activation of C-MAF. Additionally, we show that the αCD3+αCD28 -induced activation of the critical TH2 cytokine IL-4 is also dependent on IL-2 signaling. Finally, we demonstrate that IL-4 expression during in vitro TH2 differentiation is inhibited by functional IL-2R blockade. These findings help elucidate the function of IL-2/IL-2R in human TH2 differentiation and support the proposition that IL-2R-directed therapies, such as daclizumab, may have utility in the treatment of allergic disorders. Consistent with this suggestion, a recent controlled clinical trial showed improvement of pulmonary function in asthmatic patients treated with daclizumab (44).
Supplementary Material
ACKNOWLEDGEMENTS
We are grateful to Professor A. Madrigal from the Anthony Nolan Research Institute, UK, for kindly providing cord blood samples. We thank Greg Crawford and Michael Erdos for helpful discussions and Katherine Molnar-Kimber for editorial assistance.
Footnotes
This work was funded by grants to Susan John from the Wellcome Trust UK (074656/Z/04/Z), and the MRC UK (G0400197) to Susan John. PL was supported by a grant from MRC UK (G9536930). BA was supported by grant G0500429 from the MRC UK. AR was supported by a Dorothy Hodgkin Postgraduate Award. The authors acknowledge the support of the MRC Centre for Transplantation and financial support from the Department of Health via the National Institute for Health Research (NIHR) comprehensive Biomedical Research Centre award to Guy’s and St. Thomas’ NHS Foundation Trust in partnership with King’s College London and King’s College Hospital NHS Foundation Trust.
REFERENCES
- 1.Levy DE, Darnell JE., Jr. Stats: transcriptional control and biological impact. Nat Rev Mol Cell Biol. 2002;3:651–662. doi: 10.1038/nrm909. [DOI] [PubMed] [Google Scholar]
- 2.Lin JX, Leonard WJ. The role of Stat5a and Stat5b in signaling by IL-2 family cytokines. Oncogene. 2000;19:2566–2576. doi: 10.1038/sj.onc.1203523. [DOI] [PubMed] [Google Scholar]
- 3.Moriggl R, Sexl V, Piekorz R, Topham D, Ihle JN. Stat5 activation is uniquely associated with cytokine signaling in peripheral T cells. Immunity. 1999;11:225–230. doi: 10.1016/s1074-7613(00)80097-7. [DOI] [PubMed] [Google Scholar]
- 4.Moriggl R, Topham DJ, Teglund S, Sexl V, McKay C, Wang D, Hoffmeyer A, van Deursen J, Sangster MY, Bunting KD, Grosveld GC, Ihle JN. Stat5 is required for IL-2-induced cell cycle progression of peripheral T cells. Immunity. 1999;10:249–259. doi: 10.1016/s1074-7613(00)80025-4. [DOI] [PubMed] [Google Scholar]
- 5.Hoelbl A, Kovacic B, Kerenyi MA, Simma O, Warsch W, Cui Y, Beug H, Hennighausen L, Moriggl R, Sexl V. Clarifying the role of Stat5 in lymphoid development and Abelson-induced transformation. Blood. 2006;107:4898–4906. doi: 10.1182/blood-2005-09-3596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Kagami S, Nakajima H, Suto A, Hirose K, Suzuki K, Morita S, Kato I, Saito Y, Kitamura T, Iwamoto I. Stat5a regulates T helper cell differentiation by several distinct mechanisms. Blood. 2001;97:2358–2365. doi: 10.1182/blood.v97.8.2358. [DOI] [PubMed] [Google Scholar]
- 7.Burchill MA, Yang J, Vogtenhuber C, Blazar BR, Farrar MA. IL-2 receptor beta-dependent STAT5 activation is required for the development of Foxp3+ regulatory T cells. J Immunol. 2007;178:280–290. doi: 10.4049/jimmunol.178.1.280. [DOI] [PubMed] [Google Scholar]
- 8.Birney E, Stamatoyannopoulos JA, Dutta A, Guigo R, Gingeras TR, Margulies EH, Weng Z, Snyder M, Dermitzakis ET, Thurman RE, Kuehn MS, Taylor CM, Neph S, Koch CM, Asthana S, Malhotra A, Adzhubei I, Greenbaum JA, Andrews RM, Flicek P, Boyle PJ, Cao H, Carter NP, Clelland GK, Davis S, Day N, Dhami P, Dillon SC, Dorschner MO, Fiegler H, Giresi PG, Goldy J, Hawrylycz M, Haydock A, Humbert R, James KD, Johnson BE, Johnson EM, Frum TT, Rosenzweig ER, Karnani N, Lee K, Lefebvre GC, Navas PA, Neri F, Parker SC, Sabo PJ, Sandstrom R, Shafer A, Vetrie D, Weaver M, Wilcox S, Yu M, Collins FS, Dekker J, Lieb JD, Tullius TD, Crawford GE, Sunyaev S, Noble WS, Dunham I, Denoeud F, Reymond A, Kapranov P, Rozowsky J, Zheng D, Castelo R, Frankish A, Harrow J, Ghosh S, Sandelin A, Hofacker IL, Baertsch R, Keefe D, Dike S, Cheng J, Hirsch HA, Sekinger EA, Lagarde J, Abril JF, Shahab A, Flamm C, Fried C, Hackermuller J, Hertel J, Lindemeyer M, Missal K, Tanzer A, Washietl S, Korbel J, Emanuelsson O, Pedersen JS, Holroyd N, Taylor R, Swarbreck D, Matthews N, Dickson MC, Thomas DJ, Weirauch MT, Gilbert J, Drenkow J, Bell I, Zhao X, Srinivasan KG, Sung WK, Ooi HS, Chiu KP, Foissac S, Alioto T, Brent M, Pachter L, Tress ML, Valencia A, Choo SW, Choo CY, Ucla C, Manzano C, Wyss C, Cheung E, Clark TG, Brown JB, Ganesh M, Patel S, Tammana H, Chrast J, Henrichsen CN, Kai C, Kawai J, Nagalakshmi U, Wu J, Lian Z, Lian J, Newburger P, Zhang X, Bickel P, Mattick JS, Carninci P, Hayashizaki Y, Weissman S, Hubbard T, Myers RM, Rogers J, Stadler PF, Lowe TM, Wei CL, Ruan Y, Struhl K, Gerstein M, Antonarakis SE, Fu Y, Green ED, Karaoz U, Siepel A, Taylor J, Liefer LA, Wetterstrand KA, Good PJ, Feingold EA, Guyer MS, Cooper GM, Asimenos G, Dewey CN, Hou M, Nikolaev S, Montoya-Burgos JI, Loytynoja A, Whelan S, Pardi F, Massingham T, Huang H, Zhang NR, Holmes I, Mullikin JC, Ureta-Vidal A, Paten B, Seringhaus M, Church D, Rosenbloom K, Kent WJ, Stone EA, Batzoglou S, Goldman N, Hardison RC, Haussler D, Miller W, Sidow A, Trinklein ND, Zhang ZD, Barrera L, Stuart R, King DC, Ameur A, Enroth S, Bieda MC, Kim J, Bhinge AA, Jiang N, Liu J, Yao F, Vega VB, Lee CW, Ng P, Yang A, Moqtaderi Z, Zhu Z, Xu X, Squazzo S, Oberley MJ, Inman D, Singer MA, Richmond TA, Munn KJ, Rada-Iglesias A, Wallerman O, Komorowski J, Fowler JC, Couttet P, Bruce AW, Dovey OM, Ellis PD, Langford CF, Nix DA, Euskirchen G, Hartman S, Urban AE, Kraus P, Van Calcar S, Heintzman N, Kim TH, Wang K, Qu C, Hon G, Luna R, Glass CK, Rosenfeld MG, Aldred SF, Cooper SJ, Halees A, Lin JM, Shulha HP, Xu M, Haidar JN, Yu Y, Iyer VR, Green RD, Wadelius C, Farnham PJ, Ren B, Harte RA, Hinrichs AS, Trumbower H, Clawson H, Hillman-Jackson J, Zweig AS, Smith K, Thakkapallayil A, Barber G, Kuhn RM, Karolchik D, Armengol L, Bird CP, de Bakker PI, Kern AD, Lopez-Bigas N, Martin JD, Stranger BE, Woodroffe A, Davydov E, Dimas A, Eyras E, Hallgrimsdottir IB, Huppert J, Zody MC, Abecasis GR, Estivill X, Bouffard GG, Guan X, Hansen NF, Idol JR, Maduro VV, Maskeri B, McDowell JC, Park M, Thomas PJ, Young AC, Blakesley RW, Muzny DM, Sodergren E, Wheeler DA, Worley KC, Jiang H, Weinstock GM, Gibbs RA, Graves T, Fulton R, Mardis ER, Wilson RK, Clamp M, Cuff J, Gnerre S, Jaffe DB, Chang JL, Lindblad-Toh K, Lander ES, Koriabine M, Nefedov M, Osoegawa K, Yoshinaga Y, Zhu B, de Jong PJ. Identification and analysis of functional elements in 1% of the human genome by the ENCODE pilot project. Nature. 2007;447:799–816. doi: 10.1038/nature05874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Northrup DL, Zhao K. Application of ChIP-Seq and Related Techniques to the Study of Immune Function. Immunity. 34:830–842. doi: 10.1016/j.immuni.2011.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Barski A, Cuddapah S, Cui K, Roh TY, Schones DE, Wang Z, Wei G, Chepelev I, Zhao K. High-resolution profiling of histone methylations in the human genome. Cell. 2007;129:823–837. doi: 10.1016/j.cell.2007.05.009. [DOI] [PubMed] [Google Scholar]
- 11.Kim TK, Hemberg M, Gray JM, Costa AM, Bear DM, Wu J, Harmin DA, Laptewicz M, Barbara-Haley K, Kuersten S, Markenscoff-Papadimitriou E, Kuhl D, Bito H, Worley PF, Kreiman G, Greenberg ME. Widespread transcription at neuronal activity-regulated enhancers. Nature. 465:182–187. doi: 10.1038/nature09033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Jin C, Zang C, Wei G, Cui K, Peng W, Zhao K, Felsenfeld G. H3.3/H2A.Z double variant-containing nucleosomes mark ‘nucleosome-free regions’ of active promoters and other regulatory regions. Nat Genet. 2009;41:941–945. doi: 10.1038/ng.409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Adamson AS, Collins K, Laurence A, O’Shea JJ. The Current STATus of lymphocyte signaling: new roles for old players. Curr Opin Immunol. 2009;21:161–166. doi: 10.1016/j.coi.2009.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Ben-Sasson SZ, Le Gros G, Conrad DH, Finkelman FD, Paul WE. IL-4 production by T cells from naive donors. IL-2 is required for IL-4 production. J Immunol. 1990;145:1127–1136. [PubMed] [Google Scholar]
- 15.McDyer JF, Li Z, John S, Yu X, Wu CY, Ragheb JA. IL-2 receptor blockade inhibits late, but not early, IFN-gamma and CD40 ligand expression in human T cells: disruption of both IL-12-dependent and - independent pathways of IFN-gamma production. J Immunol. 2002;169:2736–2746. doi: 10.4049/jimmunol.169.5.2736. [DOI] [PubMed] [Google Scholar]
- 16.Zhu J, Cote-Sierra J, Guo L, Paul WE. Stat5 activation plays a critical role in Th2 differentiation. Immunity. 2003;19:739–748. doi: 10.1016/s1074-7613(03)00292-9. [DOI] [PubMed] [Google Scholar]
- 17.Liao W, Schones DE, Oh J, Cui Y, Cui K, Roh TY, Zhao K, Leonard WJ. Priming for T helper type 2 differentiation by interleukin 2-mediated induction of interleukin 4 receptor alpha-chain expression. Nat Immunol. 2008;9:1288–1296. doi: 10.1038/ni.1656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Cote-Sierra J, Foucras G, Guo L, Chiodetti L, Young HA, Hu-Li J, Zhu J, Paul WE. Interleukin 2 plays a central role in Th2 differentiation. Proc Natl Acad Sci U S A. 2004;101:3880–3885. doi: 10.1073/pnas.0400339101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Ho IC, Hodge MR, Rooney JW, Glimcher LH. The proto-oncogene c-maf is responsible for tissue-specific expression of interleukin-4. Cell. 1996;85:973–983. doi: 10.1016/s0092-8674(00)81299-4. [DOI] [PubMed] [Google Scholar]
- 20.Ho IC, Lo D, Glimcher LH. c-maf promotes T helper cell type 2 (Th2) and attenuates Th1 differentiation by both interleukin 4-dependent and -independent mechanisms. J Exp Med. 1998;188:1859–1866. doi: 10.1084/jem.188.10.1859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Bauquet AT, Jin H, Paterson AM, Mitsdoerffer M, Ho IC, Sharpe AH, Kuchroo VK. The costimulatory molecule ICOS regulates the expression of c-Maf and IL-21 in the development of follicular T helper cells and TH-17 cells. Nat Immunol. 2009;10:167–175. doi: 10.1038/ni.1690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Saraiva M, Christensen JR, Veldhoen M, Murphy TL, Murphy KM, O’Garra A. Interleukin-10 Production by Th1 Cells Requires Interleukin-12-Induced STAT4 Transcription Factor and ERK MAP Kinase Activation by High Antigen Dose. Immunity. 2009 doi: 10.1016/j.immuni.2009.05.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Xu J, Yang Y, Qiu G, Lal G, Wu Z, Levy DE, Ochando JC, Bromberg JS, Ding Y. c-Maf regulates IL-10 expression during Th17 polarization. J Immunol. 2009;182:6226–6236. doi: 10.4049/jimmunol.0900123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Yang Y, Ochando J, Yopp A, Bromberg JS, Ding Y. IL-6 plays a unique role in initiating c-Maf expression during early stage of CD4 T cell activation. J Immunol. 2005;174:2720–2729. doi: 10.4049/jimmunol.174.5.2720. [DOI] [PubMed] [Google Scholar]
- 25.Nelson EA, Walker SR, Alvarez JV, Frank DA. Isolation of unique STAT5 targets by chromatin immunoprecipitation-based gene identification. J Biol Chem. 2004;279:54724–54730. doi: 10.1074/jbc.M408464200. [DOI] [PubMed] [Google Scholar]
- 26.Cousins DJ, Lee TH, Staynov DZ. Cytokine coexpression during human Th1/Th2 cell differentiation: direct evidence for coordinated expression of Th2 cytokines. J Immunol. 2002;169:2498–2506. doi: 10.4049/jimmunol.169.5.2498. [DOI] [PubMed] [Google Scholar]
- 27.Lin JX, Mietz J, Modi WS, John S, Leonard WJ. Cloning of human Stat5B. Reconstitution of interleukin-2-induced Stat5A and Stat5B DNA binding activity in COS-7 cells. J Biol Chem. 1996;271:10738–10744. [PubMed] [Google Scholar]
- 28.John S, Vinkemeier U, Soldaini E, Darnell JE, Jr., Leonard WJ. The significance of tetramerization in promoter recruitment by Stat5. Mol Cell Biol. 1999;19:1910–1918. doi: 10.1128/mcb.19.3.1910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Xi H, Shulha HP, Lin JM, Vales TR, Fu Y, Bodine DM, McKay RD, Chenoweth JG, Tesar PJ, Furey TS, Ren B, Weng Z, Crawford GE. Identification and characterization of cell type-specific and ubiquitous chromatin regulatory structures in the human genome. PLoS Genet. 2007;3:e136. doi: 10.1371/journal.pgen.0030136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Boyle AP, Davis S, Shulha HP, Meltzer P, Margulies EH, Weng Z, Furey TS, Crawford GE. High-resolution mapping and characterization of open chromatin across the genome. Cell. 2008;132:311–322. doi: 10.1016/j.cell.2007.12.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Soldaini E, John S, Moro S, Bollenbacher J, Schindler U, Leonard WJ. DNA binding site selection of dimeric and tetrameric Stat5 proteins reveals a large repertoire of divergent tetrameric Stat5a binding sites. Mol Cell Biol. 2000;20:389–401. doi: 10.1128/mcb.20.1.389-401.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Ghoreschi K, Laurence A, O’Shea JJ. Selectivity and therapeutic inhibition of kinases: to be or not to be? Nat Immunol. 2009;10:356–360. doi: 10.1038/ni.1701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Malek TR, Yu A, Zhu L, Matsutani T, Adeegbe D, Bayer AL. IL-2 family of cytokines in T regulatory cell development and homeostasis. J Clin Immunol. 2008;28:635–639. doi: 10.1007/s10875-008-9235-y. [DOI] [PubMed] [Google Scholar]
- 34.Durant L, Watford WT, Ramos HL, Laurence A, Vahedi G, Wei L, Takahashi H, Sun HW, Kanno Y, Powrie F, O’Shea JJ. Diverse targets of the transcription factor STAT3 contribute to T cell pathogenicity and homeostasis. Immunity. 32:605–615. doi: 10.1016/j.immuni.2010.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Dennehy KM, Elias F, Na SY, Fischer KD, Hunig T, Luhder F. Mitogenic CD28 signals require the exchange factor Vav1 to enhance TCR signaling at the SLP-76-Vav-Itk signalosome. J Immunol. 2007;178:1363–1371. doi: 10.4049/jimmunol.178.3.1363. [DOI] [PubMed] [Google Scholar]
- 36.Tanaka Y, So T, Lebedeva S, Croft M, Altman A. Impaired IL-4 and c-Maf expression and enhanced Th1-cell development in Vav1-deficient mice. Blood. 2005;106:1286–1295. doi: 10.1182/blood-2004-10-4074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Stritesky GL, Muthukrishnan R, Sehra S, Goswami R, Pham D, Travers J, Nguyen ET, Levy DE, Kaplan MH. The Transcription Factor STAT3 Is Required for T Helper 2 Cell Development. Immunity. 34:39–49. doi: 10.1016/j.immuni.2010.12.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Nurieva RI, Duong J, Kishikawa H, Dianzani U, Rojo JM, Ho I, Flavell RA, Dong C. Transcriptional regulation of th2 differentiation by inducible costimulator. Immunity. 2003;18:801–811. doi: 10.1016/s1074-7613(03)00144-4. [DOI] [PubMed] [Google Scholar]
- 39.Yagi J, Arimura Y, Dianzani U, Uede T, Okamoto T, Uchiyama T. Regulatory roles of IL-2 and IL-4 in H4/inducible costimulator expression on activated CD4+ T cells during Th cell development. J Immunol. 2003;171:783–794. doi: 10.4049/jimmunol.171.2.783. [DOI] [PubMed] [Google Scholar]
- 40.Mesturini R, Nicola S, Chiocchetti A, Bernardone IS, Castelli L, Bensi T, Ferretti M, Comi C, Dong C, Rojo JM, Yagi J, Dianzani U. ICOS cooperates with CD28, IL-2, and IFN-gamma and modulates activation of human naive CD4+ T cells. Eur J Immunol. 2006;36:2601–2612. doi: 10.1002/eji.200535571. [DOI] [PubMed] [Google Scholar]
- 41.Yamane H, Zhu J, Paul WE. Independent roles for IL-2 and GATA-3 in stimulating naive CD4+ T cells to generate a Th2-inducing cytokine environment. J Exp Med. 2005;202:793–804. doi: 10.1084/jem.20051304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Hwang ES, White IA, Ho IC. An IL-4-independent and CD25-mediated function of c-maf in promoting the production of Th2 cytokines. Proc Natl Acad Sci U S A. 2002;99:13026–13030. doi: 10.1073/pnas.202474499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Apetoh L, Quintana FJ, Pot C, Joller N, Xiao S, Kumar D, Burns EJ, Sherr DH, Weiner HL, Kuchroo VK. The aryl hydrocarbon receptor interacts with c-Maf to promote the differentiation of type 1 regulatory T cells induced by IL-27. Nat Immunol. 11:854–861. doi: 10.1038/ni.1912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Busse WW, Israel E, Nelson HS, Baker JW, Charous BL, Young DY, Vexler V, Shames RS. Daclizumab improves asthma control in patients with moderate to severe persistent asthma: a randomized, controlled trial. Am J Respir Crit Care Med. 2008;178:1002–1008. doi: 10.1164/rccm.200708-1200OC. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.