Skip to main content
Blood logoLink to Blood
. 2013 Aug 23;122(16):2823–2836. doi: 10.1182/blood-2013-02-481788

Genome-wide DNA methylation analysis identifies hypomethylated genes regulated by FOXP3 in human regulatory T cells

Yuxia Zhang 1,2, Jovana Maksimovic 3, Gaetano Naselli 1, Junyan Qian 4, Michael Chopin 1,2, Marnie E Blewitt 1,2,5, Alicia Oshlack 3,, Leonard C Harrison 1,2
PMCID: PMC3798997  PMID: 23974203

Key Points

  • Human naive CD4+ T cells and resting nTreg are differentially methylated at 127 regions in their genomic DNA.

  • Forkhead-binding motifs are present in promoter-associated differentially methylated regions, inferring broader epigenetic control of Treg.

Abstract

Regulatory T cells (Treg) prevent the emergence of autoimmune disease. Prototypic natural Treg (nTreg) can be reliably identified by demethylation at the Forkhead-box P3 (FOXP3) locus. To explore the methylation landscape of nTreg, we analyzed genome-wide methylation in human naive nTreg (rTreg) and conventional naive CD4+ T cells (Naive). We detected 2315 differentially methylated cytosine–guanosine dinucleotides (CpGs) between these 2 cell types, many of which clustered into 127 regions of differential methylation (RDMs). Activation changed the methylation status of 466 CpGs and 18 RDMs in Naive but did not alter DNA methylation in rTreg. Gene-set testing of the 127 RDMs showed that promoter methylation and gene expression were reciprocally related. RDMs were enriched for putative FOXP3-binding motifs. Moreover, CpGs within known FOXP3-binding regions in the genome were hypomethylated. In support of the view that methylation limits access of FOXP3 to its DNA targets, we showed that increased expression of the immune suppressive receptor T-cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain (TIGIT), which delineated Treg from activated effector T cells, was associated with hypomethylation and FOXP3 binding at the TIGIT locus. Differential methylation analysis provides insight into previously undefined human Treg signature genes and their mode of regulation.

Introduction

Regulatory T cells (Treg) are pivotal in maintaining T-cell homeostasis and preventing autoimmune disease.1 Prototypic natural CD4+ Treg (nTreg) are derived from the thymus and programmed by the transcription factor Forkhead-box P3 (FOXP3),2,3 which is required for both their development and suppressive function. In humans, naive resting nTreg (rTreg) are relatively homogenous and can be isolated based on surface expression of CD45RA and CD25. Activated nTreg (aTreg), derived from rTreg after antigen activation in vivo, mostly comprise the CD25high fraction of memory (CD45RO+) CD4+ T cells.4 However, there is no clear phenotypic distinction in human blood between aTreg and activated CD45RO+ CD25+ conventional T cells (Tconv)4,5 or from antigen-activated naive CD4+ T cells (Naive) that also transiently express FOXP3.5 Thus, markers that better identify nTreg would aid assessment of immune status in health and disease. nTreg have reduced surface expression of the interleukin (IL)-7 receptor alpha chain (CD127)6,7 and express the Ikaros transcription factor family member Helios.8 However, activation of Tconv also leads to downregulation of CD1275 and expression of HELIOS.9 Lack of consensus about the status of nTreg in human disease, for example, in autoimmune diseases such as type 1 diabetes,10 underscores the need for specific and stable nTreg markers.

Demethylation at the FOXP3 locus allows identification of T cells with a stable nTreg phenotype. Demethylation within a highly conserved region, the Treg-specific demethylated region (TSDR; or conserved noncoding sequence 2, CNS2), is restricted to nTreg and is not seen in recently activated Tconv that transiently express FOXP3.11,12 Demethylation allows chromosome relaxation and binding of FOXP3 itself as well as other transcription factors (CREB/ATF, STAT5, ETs-1, Cbf-β-Runx1) in order to maintain transcriptional activity at the FOXP3 locus.13-16 To further explore the epigenetic landscape in shaping nTreg phenotype and function, we performed global DNA methylation profiling to identify differentially methylated cytosine–guanosine dinucleotides (CpGs) between human rTreg and Naive cells before and after activation in vitro. We describe regions of differential methylation (RDMs) that are potentially able to bind FOXP3 and regulate expression of other Treg signature genes, illustrated directly for TIGIT, which is a suppressive receptor expressed by Treg.

Materials and methods

Human blood T-cell isolation, activation, and phenotyping

This study was conducted in accordance with the Declaration of Helsinki. Approval was obtained from the Walter and Eliza Hall Institute’s human research ethics committee. Blood buffy coat samples were donated by the Australian Red Cross Blood Service. CD4+ T cells were flow-sorted into Naive (CD45RA+ CD45RO CD25) and rTreg (CD45RA+ CD45ROCD25+), CellTrace Violet (CTV) dye labeled, activated for 3 days with anti-CD3 (soluble, 100 ng/mL; OKT3) and anti-CD28 (200 ng/mL; CD28.2; BD) antibodies and irradiated autologous CD4+ T-cell–depleted peripheral blood mononuclear cells in IP5 medium (Iscove’s modified Dulbecco’s medium [Gibco] supplemented with 5% pooled human serum). On day 4, fresh IP5 containing recombinant human IL-2 (final of 20 U/mL) was added, and cells were cultured for an additional 2 days. On day 6, proliferated and CD25+ (CTVdim CD25+) cells were sorted.

Suppressor assay

rTreg were added to CTV-labeled Naive (5 × 104) at 1:1 and 1:2 (suppressor:responder) ratios together with autologous antigen-presenting cells, in duplicate. Cells were stimulated with anti-CD3 antibody (soluble, 200 ng/mL) for 4 days and analyzed for CTV dilution. Suppression was calculated as the percent repression of Naive proliferation.

Chromatin immunoprecipitation and quantitative, reverse-transcription polymerase chain reaction

Naive and rTreg were activated with anti-CD3 (plate-bound, 3 μg/mL), anti-CD28 (soluble, 1 μg/mL) antibodies and IL-2 (200 U/mL) for 6–10 days to obtain Act-Naive and Act-rTreg. Induced Treg (iTreg) were differentiated from Naive with the same stimuli plus transforming growth factor-β1 (5 ng/mL). FOXP3 ChIP, 10–20 million cells, were used. Relative FOXP3 enrichment was first normalized to input DNA, and fold change was calculated compared with an intergenic region. Primers are listed in supplemental Table 1 (available on the Blood Web site).

Genomic DNA isolation, bisulfite conversion, and sequencing

Genomic DNA was isolated with a blood and tissue kit (DNeasy; Qiagen), subjected to sodium bisulfite conversion with a DNA methylation kit (EZ; Zymo Research). Converted DNA was used as the template to amplify the fragments for sequencing and methylation Taqman assay.

Reverse-transcription polymerase chain reaction, quantitative polymerase chain reaction, and Taqman quantitative, reverse-transcription polymerase chain reaction

Total RNA was extracted with reagent (TRI; Invitrogen). For reverse transcription, random hexamers (Qiagen) were used. Quantitative polymerase chain reaction (qPCR) was performed in duplicate with 200 ng first-strand cDNA. Hypoxanthine phosphoribosyltransferase-1 was used as a control. miRNAs were detected using Taqman miRNA assays (Ambion), and U6 snRNA was used as a reference gene. Methylated CpGs were detected using bisulfite-converted genomic DNA and 6-carboxy-2′,4,4′,5′,7,7′–hexachlorofluorescein– and 6-carboxy fluorescein (FAM)-labeled probes. Methylation was calculated using the following formula: % methylation = 100/[1 + 2Ct(methylated - unmethylated)].

Illumina 450k arrays

Bisulfite-converted DNA was used to hybridize the Infinium HumanMethylation450 BeadChips as previously described.17 Illumina iScan was used to scan the BeadChips. The raw intensity data (IDAT) were imported into the R (2.15.2; http://cran.r-project.org/), processed using the minfi (1.4.0) Bioconductor package,18 and normalized for technical variations using SWAN.19 Poorly performing probes (P > .01 in 1 or more samples) and probes interrogating single nucleotide polymorphisms on the array were removed from further analysis. This resulted in a final dataset of 481 473 probes.

Differential methylation analysis

The proportion of methylation at each CpG is represented by the β value, defined as the proportion of the methylated signal to the total signal and calculated from the normalized intensity values. Statistical analyses were performed on M values [M = log2 (methylated/unmethylated)].20 For probe-wise differential methylation analysis, a linear model was fitted, taking into account the paired sample within an individual and using the limma (3.14.4) package.21 P values were adjusted to control for the false discovery rate (FDR) using the Benjamini–Hochberg method.22 Significantly differentially methylated probes (DMPs) were defined as those having an adjusted P < .05 and an absolute difference in mean β values |Δβ| ≥ 0.2.

Identification of RDMs

RDMs were identified using the “dmrFind” in the Bioconductor package charm (2.4.0).23 FDRs were calculated using the “qval” function in the charm package, with numiter = 1000 permutations. RDMs were further refined by the exclusion of regions with an absolute average percentage methylation difference across probes of <15%.

Functional annotation

Functional annotation was performed using the online Database for Annotation, Visualization and Integrated Discovery (DAVID), version 6.7,24,25 on the level of “genes” using default parameters.

Motif identification

Promoter-associated RDM sequences were extracted from the human genome (hg19) and submitted to the MEME motif discovery tool.26 The identified motifs were analyzed using the TOMTOM motif comparison tool18 in conjunction with the JASPAR FAM motifs database.27

Gene set testing

Gene sets include FOXP3 targets in mice28,29 and humans30 based on FOXP3 ChIP experiments, genes regulated by FOXP3 in mice31 and in humans,32 and genes encoding mouse cytokines and transcription factors33 (supplemental Table 2). Gene set testing was performed on all RDMs annotated to genes using 1-tailed Fisher exact tests.

ChIP-seq data analysis

Human FOXP3 ChIP-seq data34 were obtained from the Sequence Read Archive, and the quality was assessed using FastQC (0.10.1). The 35bp reads were then trimmed using Trimmomatic (0.30).35 Bowtie (0.12.7) in best mode with default parameters36 was used to map reads to the human genome (hg19). MACS (1.4.2) with default parameters37 was subsequently used to identify FOXP3 binding peaks.

Details of the methods can be found in the supplemental Methods section. All primers and probes are listed in supplemental Table 1. HumanMethylation450 BeadChips raw data were deposited at Gene Expression Omnibus with accession number GSE49667.

Results

CD4+ T-cell subsets: phenotype, function, and DNA methylation

Human rTreg are a relatively homogenous population that, after activation in vitro, resemble the activated nTreg (aTreg) observed in peripheral blood.4 We sorted Naive and rTreg from 3 healthy male blood donors (M28, M29, M30; Figure 1A, top panel) and activated them for 6 days with anti-CD3 and anti-CD28 antibodies, supplemented with IL-2 at day 4. After activation, both populations proliferated and were CD25+ (Figure 1A, middle panel). The majority (>85%) of activated rTreg (Act-rTreg) and some of the activated Naive (about 30%; Act-Naive) expressed FOXP3 (Figure 1B, bottom panel). In an autologous suppression assay, rTreg inhibited Naive proliferation (Figure 1B).

Figure 1.

Figure 1

CD4+ T-cell subsets: phenotype, function, and DNA methylation. (A) Naive and rTreg from male blood donors were sorted (top panel) and activated with soluble anti-CD3 and -28 antibodies (100 and 200 ng/mL, respectively) and autologous irradiated CD4+ cell-depleted peripheral blood mononuclear cells for 4 days, supplemented with 20 U/mL IL-2 for an additional 2 days. Act-Naive and Act-rTreg were sorted as proliferating CTVdim CD25+ cells (middle panel). FOXP3 expression was examined by intracellular staining (bottom panel). (B) Sorted CTVdim CD25+ cells were incubated with Naive CD4+ T cells (5 × 104) at 1:1, 1:2, 1:4, 1:8, and 1:16 ratios with anti-CD3 antibody (100 ng/mL), with or without IL-2 (20 U/mL) for 5 days. (C) Multidimensional scaling (MDS) analysis of the normalized, filtered data. The MDS plot is analogous to a principal components analysis plot. The axes represent the major sources of variation in the data based on the top 1000 genes with the largest standard deviations between samples; dimension 1 represents the largest source of variation, dimension 2 represents the next largest orthogonal source of variation, followed by dimension 3, etc. The left-hand MDS plot shows that the largest source of variation between samples was the difference in baseline methylation between the donors. Examination of further sources of variation reveals that methylation differences between cell types and activation status are also present, as shown in the right-hand MDS plot. Each sample is labeled with the donor ID and colored by cell type.

We prepared DNA from rTreg, Naive, Act-Naive, and Act-rTreg for methylation analysis on Illumina Infinium HumanMethylation450 (450k) arrays. Following quality control and preprocessing (see “Materials and methods” section), multidimensional scaling (MDS) analysis of the normalized, filtered data indicated that the largest source of variation between the samples was between the donors (Figure 1C, left panel). However, a more thorough examination of other sources of variation revealed consistent differences between cell types and activation status (Figure 1C, right panel). Our statistical analysis therefore accounted for interindividual differences when testing for methylation differences between cell types.

Differential DNA methylation between Naive and rTreg

Between Naive and rTreg, 2315 CpGs were significantly differentially methylated (FDR adjusted P < .05, |Δβ| ≥0.2; supplemental Table 3). These represented <0.5% of the total CpG probes (n = 481 473). More than 70% (n = 1681) of the DMPs were associated with genes (n = 1137), while the remaining 634 were not annotated. With respect to genomic context, 33% (n = 747) of the DMPs were in promoter regions, 40% (n = 920) in gene bodies, 4% (n = 86) in 3′ untranslated regions (UTRs) of known genes, and 24% (n = 541) were not assigned to a genomic feature. Compared with the probes on the 450k array, DMPs showed a significant enrichment in the gene body (P < 2.2 × 10−16, Fisher exact test [FET]) and a deficiency in promoter (P < 2.2 × 10−16, FET), while the proportions in the 3′ UTR and unannotated regions were not significantly different from a randomly selected set (Figure 2A). The most significant clusters identified by DAVID functional annotation clustering analysis24,25 of the genes associated with DMPs were terms T-cell activation and differentiation and regulation of cell death (supplemental Table 4).

Figure 2.

Figure 2

Differential DNA methylation between Naive and rTreg. (A) Functional annotations of the 450k array probes (left panel), 2315 DMPs between Naive and rTreg cells (middle panel), and probes associated with 127 RDMs between Naive and rTreg (right panel). (B) Unsupervised hierarchical clustering of the β values of both the significant DMPs (left panel) and RDM probes (right panel) resulted in distinct groupings of the Naive and rTreg samples. Each horizontal row in the heat map represents a CpG probe and each vertical column is a sample. The color indicates the level of methylation at each CpG in each sample, as described by the color key.

Promoter methylation is inversely correlated with gene expression

Neighboring CpGs are often correlated in methylation status,38,39 and genomic regions are more functionally relevant.40-43 Thus, we performed an analysis to identify RDMs, which can be defined as clusters of physically proximal CpGs (within ∼300bp) that are significantly different in their methylation between cell types.44 One hundred and twenty-seven RDMs were identified between Naive and rTreg (supplemental Table 5). The RDMs encompassed 608 individual CpG probes and were associated with 97 genes and 23 unannotated genomic regions (supplemental Table 6). Of the 608 individual probes, 65% (n = 385) were located in promoter regions, 23% (n = 134) in gene bodies, 2% (n = 11) in 3′ UTRs, and 11% (n = 65) were unannotated (Figure 2A, right panel). In contrast to the individual DMPs, the probes located in RDMs were significantly enriched in promoter annotations (P < 2.2 × 10−16, FET) and a corresponding decrease in gene body (P < 1.823 × 10−05, FET) and unannotated regions (P < 1.348 × 10−14, FET), while the proportion in 3′ UTR did not differ significantly. The top clusters from DAVID analysis24,25 were the associated with the terms T-cell activation and differentiation of the genes associated with RDMs. However, in contrast to the DMPs, the next 2 most significant clusters contained terms associated with repressor activity and plasma membrane (supplemental Table 7).

Unsupervised clustering of the β values of both the DMPs and RDM probes resulted in distinct groupings of the Naive and rTreg samples (Figure 2B). In both cases, hypermethylation and hypomethylation were distributed evenly between rTreg and Naive, which is in contrast to the previous report of relative hypomethylation in nTreg obtained using other methods.45

To explore the relationship between methylation changes and gene expression, we correlated the methylation status of promoter-associated RDMs with gene expression data from Schmidl et al.46 We observed a negative correlation between methylation and gene expression (Pearson correlation coefficient, −0.5; P = .0026; supplemental Figure 1), suggesting that changes in promoter methylation are related to gene expression. The 12Mb methylation data from Schmidl et al46 showed minimal (<0.01%) overlap with the 450k array CpGs. However, 5 RDMs intersected with their regions of interest, of which 3 were consistently differentially methylated in both studies (data not shown).

Activation-induced changes in DNA methylation profiles

The ability of the immune system to combat diverse, potentially pathogenic insults and still maintain tolerance to self is reflected by the lineage and functional plasticity of T cells, including Treg. We therefore examined the fidelity of distinct DNA methylation profiles after antigen activation in Naive and rTreg. Act-rTreg and Act-Naive displayed significant differences in 1689 CpG probes (supplemental Table 8), the majority of which overlapped with the differences found between these 2 T-cell subsets before activation (Figure 3A). Thus, the DNA methylation profiles that distinguished Naive and rTreg were largely unaffected by activation.

Figure 3.

Figure 3

Activation-induced changes in DNA methylation profiles. (A) Act-Naive and Act-rTreg displayed significant differences in 1679 CpG probes (green cross), the majority of which overlapped with the differences found between these 2 T-cell subsets before activation (red circle). The distinct DNA methylation profiles between Naive and rTreg are largely unaffected by activation. (B) Activation-induced methylation changes in 466 CpGs in Naive CD4+T cells (blue dots). Of these, 171 overlapped with the methylation patterns of rTreg and 283 were unique to activation. (C) Activation-induced demethylation at the MIR21 locus. (D) Expression of miR-21 in Naive and Treg before and after activation. Data are represented as mean ± standard error of the mean of 3 donors. *P = .05 (Mann–Whitney test, 1-tailed).

Next, we examined the activation-induced DNA methylation differences in Naive and rTreg. In Naive, methylation at 466 CpGs was significantly different after activation (supplemental Table 9). Of these probes, 36.7% (n = 171) acquired methylation patterns that were similar to those of rTreg and 60.7% (n = 283) were unique to activation (Figure 3B). The genes with activation-associated DMPs were enriched for terms related to transcription, DNA binding, and positive regulation of various cellular processes in the most significant cluster, followed by clusters containing terms DNA binding, nucleotide binding, phosphorylation, and kinase activity (supplemental Table 10). Eighteen RDMs were also identified, 13 of which were associated with known genes (KSR1, MIR21, GFI1, CSF2, NCF4, VMP1, HSD17B8, IL13, ITGAX, RPTOR, AIM2, CCL5, CD59) and 5 were unannotated (supplemental Table 11). The genes with activation-associated RDMs were enriched for terms related to cytokine–cytokine receptor interaction and regulation of T-cell differentiation (supplemental Table 12), indicating that changes in DNA methylation are associated with Naive activation and plasticity. Interestingly, activation did not lead to significant DNA methylation changes in rTreg.

The methylation status of activation-associated RDMs may regulate gene expression. Activation of Naive induced significant demethylation at the microRNA MIR21 locus (Figure 3C). Therefore, we examined expression of miR-21 in Naive and rTreg before and after activation. miR-21 was significantly upregulated after activation in Naive. Activation in vitro did not induce significant changes in rTreg, but aTreg isolated directly ex vivo exhibited significantly increased miR-21 expression (Figure 3D).

Identification of Forkhead-binding motifs in RDMs

We were interested to know if putative transcription factor binding sites were present in promoter-associated RDMs. An overrepresented motif was identified using MEME26 (supplemental Figure 2A). When compared with known binding motif models using TOMTOM,18 this motif was identified as a candidate Forkhead-binding motif (supplemental Figure 2B), similar to one previously described by Sadlon et al.30 Therefore, we investigated the relationship between the RDMs and genomic FOXP3 binding sites. We compared the genomic positions of our 127 RDMs with human ChIP-chip30 and ChIP-seq34 data. The ChIP-chip data identified 8305 FOXP3 binding peaks and the ChIP-seq data identified 26 483 peaks (see “Materials and methods” section). Almost 60% of the ChIP-chip peaks were also covered by the ChIP-seq peaks. Creating a union of the ChIP-chip and ChIP-seq peak regions resulted in 28 885 unique FOXP3 binding loci. Using this list, we found a direct overlap with 24 of our RDMs (supplemental Figure 3). Although this number appears small, the combined list of peaks only covers 33 342 (<7%) of the 450k CpGs. Given the number of genomic bases covered by each RDM and ChIP peak, in the context of the whole genome the number of bases covered by both RDMs and ChIP peaks is significantly overrepresented (P < 2.2 × 10−16; Pearson χ2 test), implying a relationship between methylation status and FOXP3 binding in nTreg. Furthermore, we found that RDMs overlapping FOXP3 peaks were significantly more likely to be hypomethylated in nTreg (P = .0051; 1-sided FET). The positions and methylation status of the RDMs, along with evidence for FOXP3 binding, are summarized in Table 1. We subsequently selected and validated by quantitative reverse transcription-PCR (qRT-PCR) the expression of several other hypomethylated genes in Treg (supplemental Figure 2C). Our findings are consistent with the view that demethylation, especially in promoter regions of Treg genes, increases gene expression.

Table 1.

Genomic coordinates of RDMs and putative FOXP3 binding site association

Gene Full name Chr. Start End nTreg Meth. FOXP3 ChIP
HIVEP3 Human immunodeficiency virus type 1 enhancer binding protein 3 chr1 42384310 42384564 Hyper Genomic
TNFRSF9 Tumor necrosis factor receptor superfamily, member 9 chr1 8001027 8001309 Hypo
AIM2 Absent in melanoma 2 chr1 159046773 159047163 Hypo Genomic
PYHIN1 Pyrin and HIN domain family, member 1 chr1 158900088 158900644 Hypo Genomic
NA NA chr1 38941882 38942403 Hyper
RUNX3 Runt-related transcription factor 3 chr1 25291385 25291546 Hyper Genomic
PRKCZ Protein kinase C, ζ chr1 2003864 2004346 Hypo
VPS13D Vacuolar protein sorting 13 homolog D (Saccharomyces cerevisiae) chr1 12538341 12538678 Hyper
TNFRSF4 Tumor necrosis factor receptor superfamily, member 4 chr1 1150085 1150416 Hypo
CD160 CD160 molecule chr1 145715571 145715719 Hypo
PAOX Polyamine oxidase (exo-N4-amino) chr10 135202522 135203200 Hypo
NA NA chr10 8373416 8373659 Hyper Genomic
NET1 Neuroepithelial cell transforming 1 chr10 5488366 5488628 Hyper
ADO 2-aminoethanethiol (cysteamine) dioxygenase chr10 64565642 64565772 Hypo
NA NA chr10 90749920 90750176 Hypo
FBXO18 F-box protein, helicase, 18 chr10 5935953 5936426 Hypo
CXCR5 Chemokine (C-X-C motif) receptor 5 chr11 118754280 118754593 Hypo
CD248 CD248 molecule, endosialin chr11 66084469 66084841 Hyper
LRRC32 Leucine-rich repeat containing 32 chr11 76380921 76381236 Hypo
APLP2 Amyloid β (A4) precursor-like protein 2 chr11 129992297 129992368 Hypo
BCL9L B-cell CLL/lymphoma 9-like chr11 118781728 118781978 Hyper
CRY2 Cryptochrome 2 (photolyase-like) chr11 45868398 45868501 Hypo
NA NA chr11 62621178 62621406 Hypo Genomic
ACER3 Alkaline ceramidase 3 chr11 76571458 76571610 Hyper
DUSP6 Dual-specificity phosphatase 6 chr12 89744471 89744701 Hyper
IKZF4 IKAROS family zinc finger 4 (Eos) chr12 56414442 56414533 Hypo
KRT72 Keratin 72 chr12 52994933 52995295 Hypo
NA NA chr12 54413000 54413384 Hyper
NCOR2 Nuclear receptor corepressor 2 chr12 124990897 124991139 Hyper
C12orf42 Chromosome 12 open reading frame 42 chr12 103695883 103696381 Hyper
LOC338799 Uncharacterized LOC338799 chr12 122235169 122235560 Hypo
NA NA chr12 125258948 125259531 Hypo Genomic
NCOR2 Nuclear receptor corepressor 2 chr12 124941307 124941423 Hypo
SCNN1A Sodium channel, nonvoltage-gated 1 α subunit chr12 6486598 6486709 Hyper
NA NA chr13 34184932 34185311 Hypo Genomic
EDNRB Endothelin receptor type B chr13 78494064 78494272 Hypo
TFDP1 Transcription factor Dp-1 chr13 114287374 114287735 Hyper
NA NA chr13 40746370 40746648 Hyper
TC2N Tandem C2 domains, nuclear chr14 92333765 92334271 Hyper Genomic
BCL11B B-cell CLL/lymphoma 11B (zinc finger protein) chr14 99664107 99664344 Hyper
MMP14 Matrix metallopeptidase 14 (membrane-inserted) chr14 23305780 23305957 Hyper
APBA2 Amyloid β (A4) precursor protein-binding, family A, member 2 chr15 29213626 29213860 Hyper
SMAD3 SMAD family member 3 chr15 67356310 67356942 Hyper
THBS1 Thrombospondin 1 chr15 39871808 39872071 Hypo
LOC91450 Uncharacterized LOC91450 chr15 78286521 78286737 Hypo
NA NA chr15 70767183 70767649 Hyper
ANKRD11 Ankyrin repeat domain 11 chr16 89421700 89422212 Hypo
ITGAL Integrin, α L (antigen CD11A (p180), lymphocyte function-associated antigen 1; α polypeptide) chr16 30485383 30485966 Hypo
NA NA chr16 84552874 84553585 Hyper
ANKRD11 Ankyrin repeat domain 11 chr16 89390609 89390968 Hypo
ACSF3 acyl-CoA synthetase family member 3 chr16 89163343 89163800 Hyper
CA5A Carbonic anhydrase VA, mitochondrial chr16 87958145 87958493 Hypo
MIR21 microRNA 21 chr17 57917974 57918682 Hypo Genomic/qPCR
KSR1 Kinase suppressor of ras 1 chr17 25798741 25799447 Hypo Genomic
KCNAB3 Potassium voltage-gated channel, shaker-related subfamily, β member 3 chr17 7832764 7833237 Hypo
VMP1 Vacuole membrane protein 1 chr17 57915665 57915773 Hypo Genomic
RPTOR Regulatory associated protein of MTOR, complex 1 chr17 78755379 78755604 Hyper
ST6GALNAC1 ST6 (α-N-acetyl-neuraminyl-2,3-β-galactosyl-1,3)-N-acetylgalactosaminide α-2,6-sialyltransferase 1 chr17 74639731 74640078 Hyper
ZPBP2 Zona pellucida binding protein 2 chr17 38023993 38024636 Hypo Genomic
MEOX1 Mesenchyme homeobox 1 chr17 41739130 41739326 Hypo Genomic
SCARF1 Scavenger receptor class F, member 1 chr17 1548881 1549108 Hyper
SEPT9 Septin 9 chr17 75473577 75474070 Hyper
AXIN2 Axin 2 chr17 63534340 63534688 Hyper
SEPT9 Septin 9 chr17 75451779 75452100 Hyper
TNFRSF13B Tumor necrosis factor receptor superfamily, member 13B chr17 16875349 16875596 Hypo
LAMA3 Laminin, α 3 chr18 21452730 21452895 Hyper
TGIF1 TGFB-induced factor homeobox 1 chr18 3447454 3447713 Hyper
SLC1A5 Solute carrier family 1 (neutral amino acid transporter), member 5 chr19 47288039 47288263 Hypo
EMR3 egf-like module containing, mucin-like, hormone receptor-like 3 chr19 14785593 14785849 Hyper
TYROBP TYRO protein tyrosine kinase binding protein chr19 36400049 36400437 Hyper
GNG8 Guanine nucleotide binding protein (G protein), γ 8 chr19 47139338 47139761 Hypo
KCNN4 Potassium intermediate/small conductance calcium-activated channel, subfamily N, member 4 chr19 44285297 44285594 Hypo Genomic
NA NA chr2 43398079 43398271 Hyper
GALNT5 UDP-N-acetyl-α-d-galactosamine:polypeptide N-Acetylgalactosaminyltransferase 5 (GalNAc-T5) chr2 158114069 158114300 Hyper
CASP10 Caspase 10, apoptosis-related cysteine peptidase chr2 202047246 202047754 Hyper Genomic
HDAC4 Histone deacetylase 4 chr2 240196769 240197131 Hypo Genomic
FAM124B Family with sequence similarity 124B chr2 225265963 225266346 Hyper
SP140 SP140 nuclear body protein chr2 231090376 231090745 Hypo Genomic
NA NA chr2 242805838 242806119 Hypo Genomic
SNED1 Sushi, nidogen, and EGF-like domains 1 chr2 241974858 241975140 Hypo
BCL11A B-cell CLL/lymphoma 11A (zinc finger protein) chr2 60763331 60763808 Hypo
CD8A CD8a molecule chr2 87018944 87019192 Hyper
NA NA chr21 46077454 46077731 Hyper Genomic
HORMAD2 HORMA domain containing 2 chr22 30476089 30476525 Hyper
TIGIT T-cell immunoreceptor with Ig and ITIM domains chr3 114012659 114012912 Hypo Genomic/qPCR
TGFBR2 Transforming growth factor, β receptor II (70/80 kDa) chr3 30647378 30647725 Hyper
VGLL4 Vestigial like 4 (Drosophila) chr3 11623526 11624023 Hyper
CD47 CD47 molecule chr3 107810678 107810791 Hyper
NA NA chr3 151985426 151985869 Hyper
PRDM8 PR domain containing 8 chr4 81119178 81119473 Hyper
NA NA chr4 1202509 1202780 Hypo
PRDM8 PR domain containing 8 chr4 81118343 81118794 Hyper
HPGDS Hematopoietic prostaglandin D synthase chr4 95264019 95264373 Hyper
RGS12 Regulator of G-protein signaling 12 chr4 3371236 3371652 Hyper
NA NA chr4 155661691 155661929 Hypo
NDNF Neuron-derived neurotrophic factor chr4 121991345 121991672 Hypo
CUTA cutA divalent cation tolerance homolog (Escherichia coli) chr6 33384179 33384537 Hypo
TNF Tumor necrosis factor chr6 31544694 31545473 Hypo
ZC3H12D Zinc finger CCCH-type containing 12D chr6 149806331 149806732 Hypo Genomic
RPS6KA2 Ribosomal protein S6 kinase, 90 kDa, polypeptide 2 chr6 166908379 166908811 Hypo
FLOT1 Flotillin 1 chr6 30706619 30706810 Hyper
ZBTB12 Zinc finger and BTB domain containing 12 chr6 31867906 31868025 Hyper
NA NA chr6 33386967 33387089 Hypo
C6orf222 Chromosome 6 open reading frame 222 chr6 36304456 36305155 Hypo
NA NA chr6 6588693 6589075 Hyper
HSD17B8 Hydroxysteroid (17-β) dehydrogenase 8 chr6 33173307 33173489 Hypo
TAGAP T-cell activation RhoGTPase activating protein chr6 159462964 159463300 Hypo Genomic
PLAGL1 Pleiomorphic adenoma gene-like 1 chr6 144386457 144386528 Hypo
TAP1 Transporter 1, ATP-binding cassette, subfamily B (MDR/TAP) chr6 32820102 32820249 Hypo Genomic
TAP2 Transporter 2, ATP-binding cassette, subfamily B (MDR/TAP) chr6 32805398 32805570 Hypo
AIF1 Allograft inflammatory factor 1 chr6 31583915 31584223 Hyper
NA NA chr7 149569883 149570128 Hyper
ATXN7L1 Ataxin 7-like 1 chr7 105319437 105319679 Hyper
GIMAP7 GTPase, IMAP family member 7 chr7 150216872 150217081 Hyper
UPP1 Uridine phosphorylase 1 chr7 48129797 48129992 Hypo
NA NA chr7 73241729 73242028 Hyper
WIPI2 WD repeat domain, phosphoinositide interacting 2 chr7 5271480 5271655 Hyper
GNA12 Guanine nucleotide binding protein (G protein) α 12 chr7 2774414 2774654 Hyper
NA NA chr7 157306340 157306592 Hypo
NA NA chr7 4652671 4652842 Hyper
ZHX1 Zinc fingers and homeoboxes 1 chr8 124284176 124284568 Hypo
NA NA chr8 61822740 61823275 Hyper
ANGPT1 Angiopoietin 1 chr8 108510286 108510343 Hyper
PLEC Plectin chr8 145028170 145028257 Hyper
EIF2C2 Eukaryotic translation initiation factor 2C, 2 chr8 141636174 141636798 Hypo
LOC286467 Family with sequence similarity 195, member A pseudogene chrX 130964695 130965143 Hyper
MAOB Monoamine oxidase B chrX 43741826 43741945 Hypo

Genes bound by FOXP3 in FOXP3-ChIP dataset are in boldface.

NA, not available; ITIM, immunoreceptor tyrosine-based inhibitory motif.

FOXP3 binding coincides with hypomethylation in the genome

FOXP3 can function either as a transcriptional activator (eg, to FOXP3 itself) or suppressor (eg, to IL2 and IFNG). To explore how DNA methylation may be associated with different modes of FOXP3 function, we further examined the methylation status of FOXP3 binding sites in the genome using the published human ChIP-chip30 and ChIP-seq34 data. CpG probes within FOXP3–ChIP peaks were significantly hypomethylated compared with the probes not in the peaks (supplemental Figure 4). As a transcriptional activator, FOXP3 binds to its own demethylated TSDR in Treg to maintain its transcription.10 To investigate whether FOXP3 potentially utilizes similar mechanisms to regulate the expression of other genes, we performed gene set tests on the genes associated with RDMs (see “Materials and methods” section). The differentially expressed genes in Treg and the FOXP3 target genes were significantly enriched in the RDMs (supplemental Table 13). Interestingly, genes directly activated by FOXP3 were enriched in the hypomethylated RDMs in Treg. However, genes directly repressed by FOXP3 were not differentially methylated between rTreg and Naive (supplemental Figure 5). Thus, a range of other factors, including the context of DNA methylation (eg, within or outside a CpG island) and cofactors bound to FOXP3, may ultimately determine the transcriptional regulatory function of FOXP3.

Hypomethylation is required for FOXP3 binding

To examine how methylation affects FOXP3 binding, we took advantage of differential DNA methylation between Act-rTreg and Naive-derived Act-Naive and iTreg lineages, which all express FOXP3 (Figure 4A). By ChIP-qPCR, we observed enrichment of FOXP3 at the demethylated TIGIT and MIR21 loci in Act-rTreg, as well as at other hypomethylated loci (FOXP3 TSDR, CTLA4, and IL2Ra) known to bind to FOXP3. In contrast, at the IL7RA (CD127) locus, repressed by FOXP3, enrichment of FOXP3 was marginal, suggesting indirect or weak binding. As a control, no enrichment of FOXP3 binding was observed at intron 1 of AFM (Figure 4B). Overall, our findings indicate that methylation (and its context) limits FOXP3 binding, thereby influencing Treg function.

Figure 4.

Figure 4

Hypomethylation is required for FOXP3 binding. (A) Illustration of subsets of activated T cells (left panel) and expression of FOXP3 (right panel). (B) Enrichment of FOXP3 binding in hypomethylated Treg gene loci upregulated by FOXP3 and at the FOXP3-repressed IL7RA locus. AFM intron 1 was used as a control. Data are represented as mean + standard deviation of 3 donors; paired t test (2-tailed).

Treg-specific demethylation at the T-cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain locus

T-cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain (TIGIT) is a recently discovered receptor that suppresses activation of T cells, dendritic cells, and natural killer (NK) cells.47-49 We identified an overlapping RDM and FOXP3 binding peak in the TIGIT promoter (supplemental Figure 3). Demethylation at the TIGIT locus was one of the most significantly differentially methylated regions that distinguished Naive and rTreg, not altered by activation, and is required for FOXP3 binding (Figures 4B and 5A). To validate our array data and evaluate the potential of using TIGIT as a new Treg marker, we extended DNA methylation across the TIGIT locus in 5 donors by clonal bisulfite sequencing of CpG dinucleotides proximal to the DMPs. As a control, we also analyzed methylation of the FOXP3 promoter and TSDR on the same samples (supplemental Figure 6). The CpG dinucleotides of TIGIT were hypomethylated in rTreg across several regions, most apparent in biF1, 2, and 5 where putative promoter and enhancer regions are located (Figure 5B, left panel). Demethylation of the TSDR in the FOXP3 locus indicated the purity of isolated Naive and rTreg (Figure 5B). In addition, activation and differentiation of Naive into Act-Naive or iTreg cells did not change the methylation status of TIGIT or FOXP3 (Figure 5C), as measured by methylation-sensitive Taqman-qPCR. TIGIT was described as being expressed in human Treg and activated Tconv cells.47 We found that significantly increased TIGIT protein expression occurred only in Act-rTreg cells, which is in contrast to Act-Naive and iTreg (Figure 5D). Thus, TIGIT, unlike CD25 and FOXP3, can distinguish activated nTreg cells from in vitro activated conventional T cells.

Figure 5.

Figure 5

Treg-specific hypomethylation at the TIGIT locus. (A) DMPs and RDM in TIGIT locus. (B) Location of the PCR fragments neighboring DMPs in TIGIT (biF1-5) and FOXP3 loci and clonal bisulfite sequencing results. Ref. seq, reference sequence; Bis. seq, clonal bisulfite sequencing fragments. (Arrows) DMPs identified from 450k array. Data are represented as the mean of 5 donors. (C) Methylation-sensitive Taqman-qPCR showing methylation at TIGIT (RDM) and FOXP3 (TSDR) loci in Act-Naive and iTreg and demethylation in Act-rTreg. Data are represented as mean + standard deviation of 3 donors; paired t test (2-tailed). (D) Expression of TIGIT and CD25 in Act-Naive, iTreg, and Act-rTreg. Data are representative of 10 healthy individuals all showing similar results.

Delineation of human nTreg based on TIGIT expression

Finally, we determined whether TIGIT expression could further delineate human nTreg. CD4+ rTreg, normally identified by the expression of CD45RA and CD25, were divided by TIGIT into 2 compartments. In the memory fraction, the combination of TIGIT and CD25 also improved the gating strategy for isolating aTreg ex vivo (Figure 6A). In an autologous suppression assay (Figure 6B), these Treg subsets further upregulated TIGIT after activation and suppressed Naive proliferation (Figure 6C-D). However, a fraction (∼30%) of TIGITlo rTreg cells did not upregulate TIGIT (Figure 6C, upper panel), and these cells also showed reduced FOXP3 and increased interferon-gamma (IFN-γ) expression (Figure 6E).

Figure 6.

Figure 6

Expression of TIGIT on Treg and the Treg suppressor function. (A) TIGIT delineates human rTreg and aTreg. (B) Naive proliferation at day 4 in the absence of Treg. (C) TIGIT expression on Treg and Naive (top and middle panel) was analyzed at day 4 during a suppressor assay (bottom panel). (D) Suppression by different Treg subsets presented as percent inhibition of Naive proliferation. Data are presented as mean + standard deviation of 3 donors. (E) Naive, TIGIThi rTreg, and TIGITlo rTreg activated in vitro with plate-bound anti-CD3 (3 μg/mL) and soluble anti-CD28 (1 μg/mL) antibodies and IL-2 (200 U/mL) for 3 days. Cells were supplemented with IL-2 at day 4 and analyzed for FOXP3 and IFN-γ production by intracellular staining at day 6. Data are representative of results from 3 individuals.

Discussion

Demethylation of CpGs in the FOXP3 promoter12,50 and TSDR11,51 are stable epigenetic markers for nTreg. This has important implications for assessing T-cell status in humans in health and disease, illustrated by the fact that while expression of CD25 and FOXP3 leads to variable estimates of Treg frequency in people with type 1 diabetes10 or immune dysregulation, polyendocrinopathy, enteropathy, X-linked–like syndrome,52 TSDR demethylation quantitatively defines Treg frequency.52,53 Apart from being a useful lineage mark, demethylation at the FOXP3 locus is required to maintain the suppressor function of Treg. In this regard, FOXP3 binds to the demethylated TSDR to maintain its own transcription.13-16 We reasoned that other differences in DNA methylation across the genome might further elucidate the lineage and function of Treg. We detected 2315 individual DMPs comprising 127 RDMs between human rTreg and Naive cells and showed that DNA methylation in Naive was plastic and could be altered by cell activation. The latter was also recently described for mouse naive CD4+ T cells.54 Nevertheless, Act-Naive and rTreg retained the majority of the DNA methylation profile of their parent cells, and activation did not induce significant changes in DNA methylation in rTreg.

Inspired by the knowledge that FOXP3 binds to its own demethylated TSDR to maintain its expression, we examined whether FOXP3 was involved in regulating the expression of other Treg genes in a similar fashion. We found that CpGs within the genomic FOXP3 binding sites were significantly hypomethylated. Interestingly, genes activated by FOXP3 were enriched in the hypomethylated RDMs. Genes that are repressed by FOXP3 were not differentially methylated between rTreg and Naive. Ohkura et al45 reported that demethylation was “causative” for increased expression of some Treg signature genes, such as Helios and Eos. Other genes such as Il2, Ifng, and Zap70 were repressed by Foxp3 but showed no difference in DNA methylation. Complementary to their finding, we found that FOXP3 does not change DNA methylation at the TIGIT and FOXP3 loci in Act-Naive and iTreg. Instead, demethylation (within or outside CpG islands) allows FOXP3 to access its targets and exert its function. FOXP3 can act as a transcriptional activator and repressor. We propose that FOXP3 uses different mechanisms to regulate gene activation compared with repression. FOXP3 may directly enhance gene expression by binding to demethylated regions that contain a Forkhead-binding motif, as we demonstrated for TIGIT, MIR21, FOXP3, CTLA4, and CD25. In contrast, cofactors (eg, recruited to CpG island) may be required for FOXP3-mediated gene repression. Different modes of FOXP3 target binding and gene expression regulation have also been suggested by Samstein et al.55

Differential methylation could be associated with heterogeneity of Treg function, for which TIGIT may be illustrative. TIGIT is expressed by Treg47 and suppresses a range of immune cells, including T cells, dendritic cells, and NK cells.47-49 We found that hypomethylation at the TIGIT locus characterizes human Treg and that TIGIT expression allows activated Treg to be better distinguished from effector T cells ex vivo or after activation in vitro. It is likely that demethylation and FOXP3 binding regulate TIGIT expression and that TIGIT in turn prevents nTreg from becoming IFN-γ–secreting effector T cells. MIR21 is another example. The MIR21 locus is demethylated in Treg. In Naive CD4+ T cells, active demethylation occurs after T-cell activation. MIR21 overexpression is associated with human autoimmune diseases such as systemic lupus erythematosus, which is also characterized by aberrant DNA methylation of CD4+ T cells.56

In conclusion, it is sobering to consider that despite the broad genomic coverage of the Infinium 450k array (most RefSeq genes, promoters, 5′ UTRs, first exons, gene bodies, and 3′ UTR regions), the vast majority of CpGs in the genome are not probed by the array. Further characterization of individual DMPs and neighboring regions is required to fully appreciate the methylation landscape of human Treg and to identify potential biomarkers for the assessment of T-cell status in human disease.

Supplementary Material

Supplemental Methods, Tables, and Figures

Acknowledgments

This work was supported by grants from the Australian National Health and Medical Research Council (program grant 305500 to L.C.H.) and Walter and Eliza Hall Institute Genomics Funding (to Y.Z.).

Y.Z. was a Juvenile Diabetes Research Foundation Post-doctoral Fellow, L.C.H. was a Senior Principal Research Fellow, A.O. was a Career Development Fellow of the National Health and Medical Research Council, and M.E.B. was a Queen Elizabeth II Fellow of the Australian Research Council. This work was made possible through Victorian State Government Operational Infrastructure Support and Australian National Health and Medical Research Council Research Institute Infrastructure Support Scheme.

Footnotes

The online version of this article contains a data supplement.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

Authorship

Contribution: Y.Z., M.E.B., and L.C.H. designed the experiments; Y.Z., G.N., and J.Q. performed all the experiments; J.M. and A.O. performed all the analysis; Y.Z. and J.M. drafted the manuscript; L.C.H., A.O., and M.E.B. contributed to the rewriting; and all authors contributed to discussions.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Alicia Oshlack, Murdoch Children's Research Institute, Royal Children's Hospital, 50 Flemington Rd, Parkville 3052, Australia; e-mail: alicia.oshlack@mcri.edu.au; and Leonard C. Harrison, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria, 3052, Australia; e-mail: harrison@wehi.edu.au.

References

  • 1.Wing K, Sakaguchi S. Regulatory T cells exert checks and balances on self tolerance and autoimmunity. Nat Immunol. 2010;11(1):7–13. doi: 10.1038/ni.1818. [DOI] [PubMed] [Google Scholar]
  • 2.Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol. 2003;4(4):330–336. doi: 10.1038/ni904. [DOI] [PubMed] [Google Scholar]
  • 3.Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science. 2003;299(5609):1057–1061. doi: 10.1126/science.1079490. [DOI] [PubMed] [Google Scholar]
  • 4.Miyara M, Yoshioka Y, Kitoh A, et al. Functional delineation and differentiation dynamics of human CD4+ T cells expressing the FoxP3 transcription factor. Immunity. 2009;30(6):899–911. doi: 10.1016/j.immuni.2009.03.019. [DOI] [PubMed] [Google Scholar]
  • 5.Sakaguchi S, Miyara M, Costantino CM, Hafler DA. FOXP3+ regulatory T cells in the human immune system. Nat Rev Immunol. 2010;10(7):490–500. doi: 10.1038/nri2785. [DOI] [PubMed] [Google Scholar]
  • 6.Liu W, Putnam AL, Xu-Yu Z, et al. CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4+ T reg cells. J Exp Med. 2006;203(7):1701–1711. doi: 10.1084/jem.20060772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Seddiki N, Santner-Nanan B, Martinson J, et al. Expression of interleukin (IL)-2 and IL-7 receptors discriminates between human regulatory and activated T cells. J Exp Med. 2006;203(7):1693–1700. doi: 10.1084/jem.20060468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Thornton AM, Korty PE, Tran DQ, et al. Expression of Helios, an Ikaros transcription factor family member, differentiates thymic-derived from peripherally induced Foxp3+ T regulatory cells. J Immunol. 2010;184(7):3433–3441. doi: 10.4049/jimmunol.0904028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Akimova T, Beier UH, Wang L, Levine MH, Hancock WW. Helios expression is a marker of T cell activation and proliferation. PLoS ONE. 2011;6(8):e24226. doi: 10.1371/journal.pone.0024226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Zhang Y, Bandala-Sanchez E, Harrison LC. Revisiting regulatory T cells in type 1 diabetes. Curr Opin Endocrinol Diabetes Obes. 2012;19(4):271–278. doi: 10.1097/MED.0b013e328355a2d5. [DOI] [PubMed] [Google Scholar]
  • 11.Baron U, Floess S, Wieczorek G, et al. DNA demethylation in the human FOXP3 locus discriminates regulatory T cells from activated FOXP3(+) conventional T cells. Eur J Immunol. 2007;37(9):2378–2389. doi: 10.1002/eji.200737594. [DOI] [PubMed] [Google Scholar]
  • 12.Floess S, Freyer J, Siewert C, et al. Epigenetic control of the foxp3 locus in regulatory T cells. PLoS Biol. 2007;5(2):e38. doi: 10.1371/journal.pbio.0050038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Zheng Y, Josefowicz S, Chaudhry A, Peng XP, Forbush K, Rudensky AY. Role of conserved non-coding DNA elements in the Foxp3 gene in regulatory T-cell fate. Nature. 2010;463(7282):808–812. doi: 10.1038/nature08750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Polansky JK, Schreiber L, Thelemann C, et al. Methylation matters: binding of Ets-1 to the demethylated Foxp3 gene contributes to the stabilization of Foxp3 expression in regulatory T cells. J Mol Med (Berl) 2010;88(10):1029–1040. doi: 10.1007/s00109-010-0642-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Kim HP, Leonard WJ. CREB/ATF-dependent T cell receptor-induced FoxP3 gene expression: a role for DNA methylation. J Exp Med. 2007;204(7):1543–1551. doi: 10.1084/jem.20070109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.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(1):280–290. doi: 10.4049/jimmunol.178.1.280. [DOI] [PubMed] [Google Scholar]
  • 17.Dedeurwaerder S, Defrance M, Calonne E, Denis H, Sotiriou C, Fuks F. Evaluation of the Infinium Methylation 450K technology. Epigenomics. 2011;3(6):771–784. doi: 10.2217/epi.11.105. [DOI] [PubMed] [Google Scholar]
  • 18.Hansen KDA. M. minfi: Analyze Illumina's 450k methylation arrays. R package version 1.4.0. 2012.
  • 19.Maksimovic J, Gordon L, Oshlack A. SWAN: Subset-quantile within array normalization for illumina infinium HumanMethylation450 BeadChips. Genome Biol. 2012;13(6):R44. doi: 10.1186/gb-2012-13-6-r44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Du P, Zhang X, Huang CC, Jafari N, Kibbe WA, Hou L, Lin SM. Comparison of beta-value and M-value methods for quantifying methylation levels by microarray analysis. BMC Bioinformatics. 2010;11:587. doi: 10.1186/1471-2105-11-587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Smyth GK. Limma: linear models for microarray data. Bioinformatics and Computational Biology Solutions Using R and Bioconductor 2005:397-420.
  • 22.Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. Journal of the Royal Statistical Society, Series B (Methodological) Vol. 57, No. 1. New York, NY: Wiley; 1995:289-300. [Google Scholar]
  • 23.Aryee MJ, Wu Z, Ladd-Acosta C, et al. Accurate genome-scale percentage DNA methylation estimates from microarray data. Biostatistics. 2011;12(2):197–210. doi: 10.1093/biostatistics/kxq055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Huang W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 2009;4(1):44–57. doi: 10.1038/nprot.2008.211. [DOI] [PubMed] [Google Scholar]
  • 25.Huang W, Sherman BT, Lempicki RA. Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 2009;37(1):1–13. doi: 10.1093/nar/gkn923. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Bailey TL, Elkan C. Fitting a mixture model by expectation maximization to discover motifs in biopolymers. Proc Int Conf Intell Syst Mol Biol. 1994;2:28–36. [PubMed] [Google Scholar]
  • 27.Sandelin A, Wasserman WW. Constrained binding site diversity within families of transcription factors enhances pattern discovery bioinformatics. J Mol Biol. 2004;338(2):207–215. doi: 10.1016/j.jmb.2004.02.048. [DOI] [PubMed] [Google Scholar]
  • 28.Zheng Y, Josefowicz SZ, Kas A, Chu TT, Gavin MA, Rudensky AY. Genome-wide analysis of Foxp3 target genes in developing and mature regulatory T cells. Nature. 2007;445(7130):936–940. doi: 10.1038/nature05563. [DOI] [PubMed] [Google Scholar]
  • 29.Marson A, Kretschmer K, Frampton GM, et al. Foxp3 occupancy and regulation of key target genes during T-cell stimulation. Nature. 2007;445(7130):931–935. doi: 10.1038/nature05478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Sadlon TJ, Wilkinson BG, Pederson S, et al. Genome-wide identification of human FOXP3 target genes in natural regulatory T cells. J Immunol. 2010;185(2):1071–1081. doi: 10.4049/jimmunol.1000082. [DOI] [PubMed] [Google Scholar]
  • 31.Hill JA, Feuerer M, Tash K, et al. Foxp3 transcription-factor-dependent and -independent regulation of the regulatory T cell transcriptional signature. Immunity. 2007;27(5):786–800. doi: 10.1016/j.immuni.2007.09.010. [DOI] [PubMed] [Google Scholar]
  • 32.Pfoertner S, Jeron A, Probst-Kepper M, et al. Signatures of human regulatory T cells: an encounter with old friends and new players. Genome Biol. 2006;7(7):R54. doi: 10.1186/gb-2006-7-7-r54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Wei G, Wei L, Zhu J, et al. Global mapping of H3K4me3 and H3K27me3 reveals specificity and plasticity in lineage fate determination of differentiating CD4+ T cells. Immunity. 2009;30(1):155–167. doi: 10.1016/j.immuni.2008.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Birzele F, Fauti T, Stahl H, et al. Next-generation insights into regulatory T cells: expression profiling and FoxP3 occupancy in Human. Nucleic Acids Res. 2011;39(18):7946–7960. doi: 10.1093/nar/gkr444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Bailey TLEC. Fitting a mixture model by expectation maximization to discover motifs in biopolymers. Proceedings of the Second International Conference on Intelligent Systems for Molecular Biology. 1994:28-36. [PubMed] [Google Scholar]
  • 36.Langmead B, Trapnell C, Pop M, Salzberg SL. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 2009;10(3):R25. doi: 10.1186/gb-2009-10-3-r25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Zhang Y, Liu T, Meyer CA, et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol. 2008;9(9):R137. doi: 10.1186/gb-2008-9-9-r137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Eckhardt F, Lewin J, Cortese R, et al. DNA methylation profiling of human chromosomes 6, 20 and 22. Nat Genet. 2006;38(12):1378–1385. doi: 10.1038/ng1909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Irizarry RA, Ladd-Acosta C, Carvalho B, et al. Comprehensive high-throughput arrays for relative methylation (CHARM). Genome Res. 2008;18(5):780–790. doi: 10.1101/gr.7301508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Hansen KD, Timp W, Bravo HC, et al. Increased methylation variation in epigenetic domains across cancer types. Nat Genet. 2011;43(8):768–775. doi: 10.1038/ng.865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Jaenisch R, Bird A. Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nat Genet. 2003;33(Suppl):245–254. doi: 10.1038/ng1089. [DOI] [PubMed] [Google Scholar]
  • 42.Irizarry RA, Ladd-Acosta C, Wen B, et al. The human colon cancer methylome shows similar hypo- and hypermethylation at conserved tissue-specific CpG island shores. Nat Genet. 2009;41(2):178–186. doi: 10.1038/ng.298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Lister R, Pelizzola M, Dowen RH, et al. Human DNA methylomes at base resolution show widespread epigenomic differences. Nature. 2009;462(7271):315–322. doi: 10.1038/nature08514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Jaffe AE, Murakami P, Lee H, et al. Bump hunting to identify differentially methylated regions in epigenetic epidemiology studies. Int J Epidemiol. 2012;41(1):200–209. doi: 10.1093/ije/dyr238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Ohkura N, Hamaguchi M, Morikawa H, et al. T cell receptor stimulation-induced epigenetic changes and Foxp3 expression are independent and complementary events required for Treg cell development. Immunity. 2012;37(5):785–799. doi: 10.1016/j.immuni.2012.09.010. [DOI] [PubMed] [Google Scholar]
  • 46.Schmidl C, Klug M, Boeld TJ, et al. Lineage-specific DNA methylation in T cells correlates with histone methylation and enhancer activity. Genome Res. 2009;19(7):1165–1174. doi: 10.1101/gr.091470.109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Yu X, Harden K, Gonzalez LC, et al. The surface protein TIGIT suppresses T cell activation by promoting the generation of mature immunoregulatory dendritic cells. Nat Immunol. 2009;10(1):48–57. doi: 10.1038/ni.1674. [DOI] [PubMed] [Google Scholar]
  • 48.Joller N, Hafler JP, Brynedal B, et al. Cutting edge: TIGIT has T cell-intrinsic inhibitory functions. J Immunol. 2011;186(3):1338–1342. doi: 10.4049/jimmunol.1003081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Lozano E, Dominguez-Villar M, Kuchroo V, Hafler DA. The TIGIT/CD226 axis regulates human T cell function. J Immunol. 2012;188(8):3869–3875. doi: 10.4049/jimmunol.1103627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Nagar M, Vernitsky H, Cohen Y, et al. Epigenetic inheritance of DNA methylation limits activation-induced expression of FOXP3 in conventional human CD25-CD4+ T cells. Int Immunol. 2008;20(8):1041–1055. doi: 10.1093/intimm/dxn062. [DOI] [PubMed] [Google Scholar]
  • 51.Polansky JK, Kretschmer K, Freyer J, et al. DNA methylation controls Foxp3 gene expression. Eur J Immunol. 2008;38(6):1654–1663. doi: 10.1002/eji.200838105. [DOI] [PubMed] [Google Scholar]
  • 52.Barzaghi F, Passerini L, Gambineri E, et al. Demethylation analysis of the FOXP3 locus shows quantitative defects of regulatory T cells in IPEX-like syndrome. J Autoimmun. 2012;38(1):49–58. doi: 10.1016/j.jaut.2011.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.McClymont SA, Putnam AL, Lee MR, et al. Plasticity of human regulatory T cells in healthy subjects and patients with type 1 diabetes. J Immunol. 2011;186(7):3918–3926. doi: 10.4049/jimmunol.1003099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Li Y, Chen G, Ma L, et al. Plasticity of DNA methylation in mouse T cell activation and differentiation. BMC Mol Biol. 2012;13:16. doi: 10.1186/1471-2199-13-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Samstein RM, Arvey A, Josefowicz SZ, et al. Foxp3 exploits a pre-existent enhancer landscape for regulatory T cell lineage specification. Cell. 2012;151(1):153–166. doi: 10.1016/j.cell.2012.06.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Pan W, Zhu S, Yuan M, et al. MicroRNA-21 and microRNA-148a contribute to DNA hypomethylation in lupus CD4+ T cells by directly and indirectly targeting DNA methyltransferase 1. J Immunol. 2010;184(12):6773–6781. doi: 10.4049/jimmunol.0904060. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplemental Methods, Tables, and Figures

Articles from Blood are provided here courtesy of The American Society of Hematology

RESOURCES