epigenetic control of foxp3 expression: the key to a stable regulatory t-cell lineage?

7
Research carried out over the past 10–20 years has greatly expanded our understand- ing of how T-cell subsets differentiate and acquire a degree of stability that allows them to be considered a distinct T-cell line- age. This is the case for CD4 + regulatory T (T Reg ) cells that express the transcription factor forkhead box P3 (FOXP3), which is essential for the normal development and function of these cells 1–3 . Together with other suppressive immune-cell popula- tions, FOXP3-expressing T Reg cells have an essential role in maintaining homeostasis of the immune system and in preventing the autoimmune reactivity of self-reactive T cells that have escaped negative selection in the thymus. FOXP3 + T Reg cells can differentiate dur- ing T-cell development in the thymus at the stage of CD4 single-positive thymocytes 4 . It has been reported that antigen presentation by either cortical or medullary thymic epi- thelial cells is sufficient for inducing FOXP3 expression in developing thymocytes 5,6 . These thymically derived or natural T Reg cells retain a stable phenotype following export into the periphery. There, they can become activated by specific antigen and acquire some of the phenotypical properties of effec- tor memory T cells, such as the capacity to migrate into inflamed peripheral tissues, while maintaining FOXP3 expression and their suppressive function 7 . Therefore, this T-cell subset has the defining features of a stable T-cell lineage. In addition, there is now compelling evidence indicating that FOXP3 + T Reg cells can also arise from the conversion of naive conventional CD4 + T cells in the periphery (FIG. 1) following recognition of antigen under tolerogenic conditions 7,8 . It can be assumed that such de novo-generated T Reg cells have an important role in acquired tolerance to, for example, food antigens or commensal gut flora 9,10 . However, the extent to which de novo-induced T Reg cells contribute to the peripheral T Reg -cell pool and whether they have a stable phenotype that is comparable to that of natural T Reg cells are issues that have not yet been thoroughly investigated. Antigenic stimulation of conventional CD4 + T cells in vitro in the presence of transforming growth factor-β (TGFβ) leads to the induction of FOXP3 expression and the acquisition of suppressor function 11 . However, this acquired T Reg -cell pheno- type is unstable, as most of these cells lose FOXP3 expression following restimulation with antigen in the absence of exogenous TGFβ (REF. 12). This indicates that TGFβ is not sufficient for imprinting T cells with the permanent expression of FOXP3 that is needed for a stable T Reg -cell phenotype. Moreover, although conventional human T cells transiently upregulate the expression of FOXP3 following antigenic stimula- tion 13–18 , whether this transient FOXP3 + population has suppressive function is cur- rently disputed. Some groups have reported that these cells acquire transient suppres- sive function 13,18 , whereas others have sug- gested that FOXP3 expression in human T cells does not always directly correlate with suppressive capacity 15–17 . Stable FOXP3 expression is clearly a prerequisite for the maintenance of sup- pressive properties in T Reg cells. Given the potential for these cells to be manipulated in a therapeutic context, it is essential that the factors governing the expression of this lineage-specification factor be defined. In this Progress article, we summarize the current knowledge of how FOXP3 expres- sion — and therefore the T Reg -cell pheno- type — is controlled at the molecular level, highlighting mechanisms of transcriptional regulation and the importance of epigenetic modification of the FOXP3 locus. Requirements for FOXP3 expression Although various signals that induce the expression of FOXP3 have been identi- fied, the precise mechanisms by which the expression of this protein is controlled in T Reg cells are not well understood. So far, it has been established that the synergistic action of signals downstream of the T-cell receptor (TCR), co-stimulatory molecules and cytokine receptors is required for the active transcription of FOXP3 (FIG. 2). TCR signalling. TCR signalling pathways contribute to the induction of FOXP3 expres- sion in both natural and de novo-induced T Reg cells. In human T cells, TCR activation has been shown to lead to the binding of the transcription factors nuclear factor of acti- vated T cells (NFAT) and activator protein 1 (AP1) to the FOXP3 promoter 14 . In mouse T cells, TCR activation results in the binding of cyclic-AMP-responsive-element-binding- protein (CREB) and activating transcription factor (ATF) to an intronic enhancer element in the Foxp3 gene 19 . Accordingly, mice that Epigenetic control of FOXP3 expression: the key to a stable regulatory T-cell lineage? Jochen Huehn, Julia K. Polansky and Alf Hamann Abstract | Regulatory T (T Reg ) cells constitute a unique T‑cell lineage that has a crucial role in immunological tolerance. Several years ago, forkhead box P3 (FOXP3) was identified as the transcription factor that was responsible for determining the development and function of these cells. However, the underlying mechanisms that are involved in the regulation of the FOXP3 gene remain unclear and therefore preclude accurate identification and manipulation of T Reg cells. In this Progress article, we summarize recent advances in understanding how FOXP3 expression is controlled and highlight evidence suggesting that epigenetic regulation of the FOXP3 locus contributes to its role as a lineage‑specification factor. PROGRESS NATURE REVIEWS | IMMUNOLOGY VOLUME 9 | FEBRUARY 2009 | 83 FOCUS ON CD4 + T-CELL DIVERSITY © 2009 Macmillan Publishers Limited. All rights reserved

Upload: alf

Post on 21-Jul-2016

213 views

Category:

Documents


0 download

TRANSCRIPT

Research carried out over the past 10–20 years has greatly expanded our understand-ing of how T-cell subsets differentiate and acquire a degree of stability that allows them to be considered a distinct T-cell line-age. This is the case for CD4+ regulatory T (TReg) cells that express the transcription factor forkhead box P3 (FOXP3), which is essential for the normal development and function of these cells1–3. Together with other suppressive immune-cell popula-tions, FOXP3-expressing TReg cells have an essential role in maintaining homeostasis of the immune system and in preventing the autoimmune reactivity of self-reactive T cells that have escaped negative selection in the thymus.

FOXP3+ TReg cells can differentiate dur-ing T-cell development in the thymus at the stage of CD4 single-positive thymocytes4. It has been reported that antigen presentation by either cortical or medullary thymic epi-thelial cells is sufficient for inducing FOXP3 expression in developing thymocytes5,6. These thymically derived or natural TReg cells retain a stable phenotype following export into the periphery. There, they can become activated by specific antigen and acquire some of the phenotypical properties of effec-tor memory T cells, such as the capacity to migrate into inflamed peripheral tissues,

while maintaining FOXP3 expression and their suppressive function7. Therefore, this T-cell subset has the defining features of a stable T-cell lineage.

In addition, there is now compelling evidence indicating that FOXP3+ TReg cells can also arise from the conversion of naive conventional CD4+ T cells in the periphery (FIG. 1) following recognition of antigen under tolerogenic conditions7,8. It can be assumed that such de novo-generated TReg cells have an important role in acquired tolerance to, for example, food antigens or commensal gut flora9,10. However, the extent to which de novo-induced TReg cells contribute to the peripheral TReg-cell pool and whether they have a stable phenotype that is comparable to that of natural TReg cells are issues that have not yet been thoroughly investigated.

Antigenic stimulation of conventional CD4+ T cells in vitro in the presence of transforming growth factor-β (TGFβ) leads to the induction of FOXP3 expression and the acquisition of suppressor function11. However, this acquired TReg-cell pheno-type is unstable, as most of these cells lose FOXP3 expression following restimulation with antigen in the absence of exogenous TGFβ (reF. 12). This indicates that TGFβ is not sufficient for imprinting T cells with

the permanent expression of FOXP3 that is needed for a stable TReg-cell phenotype. Moreover, although conventional human T cells transiently upregulate the expression of FOXP3 following antigenic stimula-tion13–18, whether this transient FOXP3+ population has suppressive function is cur-rently disputed. Some groups have reported that these cells acquire transient suppres-sive function13,18, whereas others have sug-gested that FOXP3 expression in human T cells does not always directly correlate with suppressive capacity15–17.

Stable FOXP3 expression is clearly a prerequisite for the maintenance of sup-pressive properties in TReg cells. Given the potential for these cells to be manipulated in a therapeutic context, it is essential that the factors governing the expression of this lineage-specification factor be defined. In this Progress article, we summarize the current knowledge of how FOXP3 expres-sion — and therefore the TReg-cell pheno-type — is controlled at the molecular level, highlighting mechanisms of transcriptional regulation and the importance of epigenetic modification of the FOXP3 locus.

Requirements for FOXP3 expressionAlthough various signals that induce the expression of FOXP3 have been identi-fied, the precise mechanisms by which the expression of this protein is controlled in TReg cells are not well understood. So far, it has been established that the synergistic action of signals downstream of the T-cell receptor (TCR), co-stimulatory molecules and cytokine receptors is required for the active transcription of FOXP3 (FIG. 2).

TCR signalling. TCR signalling pathways contribute to the induction of FOXP3 expres-sion in both natural and de novo-induced TReg cells. In human T cells, TCR activation has been shown to lead to the binding of the transcription factors nuclear factor of acti-vated T cells (NFAT) and activator protein 1 (AP1) to the FOXP3 promoter14. In mouse T cells, TCR activation results in the binding of cyclic-AMP-responsive-element-binding-protein (CREB) and activating transcription factor (ATF) to an intronic enhancer element in the Foxp3 gene19. Accordingly, mice that

Epigenetic control of FOXP3 expression: the key to a stable regulatory T-cell lineage?Jochen Huehn, Julia K. Polansky and Alf Hamann

Abstract | Regulatory T (TReg

) cells constitute a unique T‑cell lineage that has a crucial role in immunological tolerance. Several years ago, forkhead box P3 (FOXP3) was identified as the transcription factor that was responsible for determining the development and function of these cells. However, the underlying mechanisms that are involved in the regulation of the FOXP3 gene remain unclear and therefore preclude accurate identification and manipulation of T

Reg cells. In this Progress

article, we summarize recent advances in understanding how FOXP3 expression is controlled and highlight evidence suggesting that epigenetic regulation of the FOXP3 locus contributes to its role as a lineage‑specification factor.

PROgReSS

NATuRE REvIEwS | Immunology vOluME 9 | FEBRuARy 2009 | 83

f o c u s o n c d 4 + T- c e l l d i v e r s i T y

© 2009 Macmillan Publishers Limited. All rights reserved

are deficient for protein kinase Cθ (PKCθ) and calcineurin Aβ, which are both involved in NFAT activation, have a marked reduc-tion in the number of TReg cells. In addition, PKCθ was found to stimulate the activity of the FOXP3 promoter20, which suggests that

PKCθ promotes TReg-cell development by enhancing FOXP3 expression through the activation of the Ca2+–calcineurin–NFAT pathway. These data are supported by the finding that high levels of the calcineurin inhibitor cyclosporine block TReg-cell

induction and function in both mice and humans14,21–23. However, low-dose cyclosporine therapy has recently been reported to increase the size of the TReg-cell population in patients with atopic dermati-tis24, which suggests that partial inhibition of the calcineurin pathway might be beneficial for the induction of TReg cells. Partial inhibi-tion of calcineurin activity might mimic suboptimal TCR stimulation, which was found to result in more efficient induction of TReg cells than strong TCR activation both in vitro25 and in vivo8. Moreover, premature termination of TCR signalling and inhibi-tion of phosphoinositide 3-kinase, AKT or mammalian target of rapamycin induced the expression of FOXP3 by mouse T cells26,27. Together, these data indicate that the duration and strength of the TCR signal are crucial determinants of FOXP3 expression, with shorter and weaker TCR stimulation favouring TReg-cell development.

Co-stimulation. In addition to TCR signal-ling, specific co-stimulatory signals are essential for FOXP3 expression in both natural and de novo-induced TReg cells. CD28 stimulation of TCR-activated thymocytes induces the expression of FOXP3 and the initiation of the TReg-cell differentiation pro-gramme28. By contrast, de novo conversion of conventional T cells into TReg cells in the periphery is impaired by co-stimulation29, suggesting that the requirement for co-stimulation for the induction of FOXP3 expression differs between natural and de novo-induced TReg cells.

Cytokine-mediated signals. Specific cytokine-mediated signals are also essential for the expression of FOXP3. The best evi-dence in support of this is the finding that there is a complete lack of TReg cells in mice that are deficient for the common cytokine-receptor γ-chain (γc), which transmits signals that are mediated by interleukin-2 (Il-2) and several other cytokines30. However, it is not clear which of the cytokines that use the γc for signal transduction are involved in FOXP3 expression.

There is substantial evidence sug-gesting that the signalling cascade that is activated following binding of Il-2 to its receptor, which involves Janus kinase 1 (JAK1), JAK3 and signal transducer and activator of transcription 5 (STAT5), has an integral role in inducing FOXP3 expres-sion (FIG. 2). Indeed, Stat5–/– mice have strongly reduced FOXP3 expression and Jak3–/– mice lack FOXP3 expression com-pletely31,32. Moreover, Il-2-induced STATs

Nature Reviews | Immunology

Cortex

Subcapsularzone

Medulla

T-cell precursor

FOXP3+

FOXP3+

Naive TReg cellFOXP3–

Naive T cell

FOXP3–

Periphery In vitro

Thymus

FOXP3+ effector memory TReg cell

Unstable FOXP3+ TReg cell

FOXP3– effector T cell

FOXP3+ de novo-induced TReg cell

Cortical epithelial cell

Medullaryepithelial cell

+ TGFβ (maintenance)+ IL-2 (maintenance and expansion)

+ TGFβ (maintenance)+/– Retinoic acid+ Other factors?

Antigen-induced differentiation

+ TGFβ +/– Retinoic acid

Thymicdendritic cell

γc cytokines+/– TGFβ

Figure 1 | The origin of regulatory T cells in the thymus and the periphery. The expression of the transcription factor forkhead box P3 (FOXP3) is thought to best define a subset of CD4+ regulatory T (T

Reg) cells that have suppressive functions. Natural T

Reg cells differentiate in the thymus from T‑cell

precursors in a process that is not yet completely understood but is known to involve interactions with thymic epithelial cells in the cortex and the medulla. Cytokines that signal through the common cytokine‑receptor γ‑chain (γ

c) subunit (such as interleukin‑2; IL‑2) are also known to participate in the

generation of TReg

cells, whereas the role of transforming growth factor‑β (TgFβ) in this process is still debated. T

Reg cells are thought to constitute a separate T‑cell lineage and to maintain FOXP3 expression

throughout their life cycle in an IL‑2‑ and TgFβ‑dependent manner. Following antigen encounter in the periphery, naive T

Reg cells (similarly to FOXP3– T cells) acquire features of effector memory cells. T

Reg cells

can also be induced de novo from naive peripheral FOXP3– T cells. In vitro, this conversion can be driven by treatment with TgFβ and may be enhanced by the vitamin A metabolite retinoic acid. However, these cells have an unstable phenotype and transient expression of FOXP3. Conversion can also occur in vivo following various tolerogenic protocols, such as oral antigen administration. It has been reported that TgFβ and retinoic acid have a role in T

Reg‑cell conversion in vivo, although other factors are probably

also involved. In addition, the stability of in vivo‑converted TReg

cells is still unclear.

P r o g r e s s

84 | FEBRuARy 2009 | vOluME 9 www.nature.com/reviews/immunol

P r o g r e s s

© 2009 Macmillan Publishers Limited. All rights reserved

bind directly to evolutionarily conserved regions in the FOXP3 locus and induce the expression of this gene32–34. Although Il-2 is crucial for the maintenance of homeo-stasis and competitive fitness of natural TReg cells in the periphery, it is dispensable for the expression of FOXP3 in the thymus30,35, which suggests that other cytokines that signal through γc-containing receptors, such as Il-7 and Il-15, might compensate for Il-2 during the development of natural TReg cells34.

In addition to γc cytokines, TGFβ also has an important role in TReg-cell biology. Although the contribution of TGFβ to the development of TReg cells in the thymus is still controversial36,37, TGFβ seems to have a central role in the maintenance of FOXP3 expression and homeostasis of natu-ral TReg cells36. Recently, it was shown that TGFβ-inducible early gene 1 (TIEG1; also known as KFl10) can bind to the FOXP3 promoter and cooperate with itchy E3 ubiq-uitin protein ligase homologue (ITCH) to induce FOXP3 expression38. In addition, the TGFβ-induced transcription factor moth-ers against decapentaplegic homologue 3 (SMAD3) has been shown to control the activity of a newly identified FOXP3 intronic enhancer element in cooperation with NFAT39 (FIG. 2).

Several factors have been reported to pro-mote TGFβ-dependent de novo induction of FOXP3 expression. Exposure of T cells to the vitamin A metabolite retinoic acid, which is produced by intestinal lamina-propria den-dritic cells and other cell types9,10, increases the expression and phosphorylation of SMAD3 (reF. 40). This, in turn, leads to an increase in TGFβ-induced FOXP3 expres-sion while preventing the differentiation of the inflammatory T helper 17 (TH17)-cell subset9,10,29,40. Importantly, the de novo induc-tion of TReg cells can occur even at high levels of co-stimulation if retinoic acid is present, suggesting that retinoic acid attenuates the inhibitory effect of co-stimulation on the induction of FOXP3 expression29. These data are supported by a recent report41 showing that retinoic acid indirectly enhances the induction of FOXP3 expression by inhibit-ing the production of counter-regulatory cytokines by CD44hi effector memory T cells. Notch-mediated signals also cooper-ate with TGFβ-mediated signals to regulate FOXP3 expression in conventional T cells42 by directly targeting the FOXP3 promoter through mechanisms that depend on recombination-signal-binding protein for immunoglobulin-k J-region (RBPJ) and hairy and enhancer of split 1 (HES1)43.

In addition to the factors that promote TGFβ-dependent de novo induction of FOXP3 expression, there are various mecha-nisms that negatively regulate TReg-cell dif-ferentiation; lineage-specifying factors of the TH1- and TH2-cell subsets are the most prominent of these mechanisms44–46. More specifically, direct binding of the Il-4-induced proteins GATA-binding protein 3 (GATA3) and STAT6, as well as of the interferon-γ-induced protein interferon-regulatory factor 1 (IRF1), to conserved binding sites in the FOXP3 promoter has been reported to repress its transcriptional activity45,47,48, which indicates that recipro-cal developmental pathways can control the generation of effector cells and TReg cells.

Although these findings support the hypothesis that a delicate balance of TCR-mediated, co-stimulatory and cytokine-mediated signals is mandatory for the transcription of the FOXP3 gene, little is known about the mechanisms by which these signals induce FOXP3 expression at the molecular level. How do the molecular signals that are required for the induction of FOXP3 expression in developing thymo-cytes differ from those that are required for the conversion of conventional T cells into TReg cells? And how do constitutive signals, such as TCR–MHC interactions and basal cytokines, maintain FOXP3 expres-sion in natural TReg cells under steady-state conditions?

Nature Reviews | Immunology

CD28TCR

TGFβ

TGFβreceptor

SMAD4

SMAD2 SMAD3

SMAD2 SMAD3

SMAD2 SMAD3

PLCγ

PKCθ

Cytoplasm

Nucleus

Ca2+

Calcineurin

NFAT p38 ERK JNK

AP1 TIEG1NFAT

SP1

↑ cAMPPI3K

AKT

mTORPKA

CREB andATF

CREB andATF

JUNFOS

γc

JAK3

IL-2receptor

IL-2

PP ST

AT5

STAT

5

STAT5

PP ST

AT5

STAT

5

?

FOXP3

Figure 2 | multiple signalling pathways converge for the induction of forkhead box P3 expression. Signals that are triggered following ligand binding to the T‑cell receptor (TCR), CD28, cytokine recep‑tors that contain common cytokine‑receptor γ‑chain (γ

c; here represented by the interleukin‑2 (IL‑2)

receptor) and the transforming growth factor‑β (TgFβ) receptor, and by the initiation of the cyclic AMP pathway together regulate the expression of the transcription factor forkhead box P3 (FOXP3). These events result in the activation of transcription factors that are involved in FOXP3 expression, including cAMP‑responsive‑element‑binding protein (CReB), activating transcription factor (ATF), SP1, nuclear factor of activated T cells (NFAT), activator protein 1 (AP1), TgFβ‑inducible early gene 1 (TIeg1), mothers against decapentaplegic homologue 3 (SMAD3) and signal transducer and activator of transcription 5 (STAT5). However, the contribution of each pathway (either beneficial or inhibitory) to FOXP3 expres‑sion might differ between different types of T cell; for example, CD28 stimulation is important for the thymic development of FOXP3+ regulatory T cells, whereas the activation of the phospho‑inositide 3‑kinase (PI3K)–AKT–mammalian target of rapamycin (mTOR) pathway inhibits FOXP3 expression in peripheral naive T cells by an unknown mechanism. eRK, extracellular‑signal‑regulated kinase; JAK, Janus kinase; JNK, JUN N‑terminal kinase; PKA, protein kinase A; PKCθ, protein kinase Cθ; PLCγ, phospholipase Cγ.

P r o g r e s s

NATuRE REvIEwS | Immunology vOluME 9 | FEBRuARy 2009 | 85

f o c u s o n c d 4 + T- c e l l d i v e r s i T y

© 2009 Macmillan Publishers Limited. All rights reserved

microRNA and TReg cellsIn the past decade, the discovery of micro-RNAs has revealed another level of com-plexity in the mechanisms that regulate gene expression. These small, non-coding RNAs inhibit protein expression either by enhancing the degradation of target mRNA species or by repressing mRNA translation through a process known as RNA interference. microRNAs are involved in the regulation of a broad range of cell-ular processes, including decision making during lineage differentiation49. In the immune system, microRNAs have a crucial role in leukocyte development and in the modulation of immune responses50. This is also true for the TReg-cell subset, in which microRNA-mediated regulation seems to be particularly important.

First, microRNAs are involved in TReg-cell development in the thymus51. In addition, microRNAs are fundamental for TReg-cell function, as TReg-cell-specific dele-tion of the microRNA pathway led to the development of fatal autoimmune diseases, which was caused by a loss of suppressor function in the peripheral TReg-cell pool52–54. Although experimental evidence is still lacking, it is possible that the contribu-tion of microRNA-mediated regulation to TReg-cell function is controlled by FOXP3, which has been shown to bind close to microRNA-encoding intergenic regions55 and to contribute to the distinct microRNA expression profile that is found in TReg cells51. whether, in turn, the expression of FOXP3 is controlled by microRNAs, as recently suggested52, awaits further inves-tigation. Although it is still unclear how microRNAs control TReg-cell development and function, these early studies certainly indicate that this topic will be the subject of further research.

Epigenetic regulation of FOXP3Recently, several groups have observed that epigenetic regulation is crucial for control-ling the expression of the FOXP3 locus. Epigenetic modifications, which can target histones or the DNA directly, affect gene transcription by altering the accessibility of distinct DNA regions to transcription factors and other DNA-binding molecules. Histones can be modified by site-specific acetylation and by methylation, modifications that are essential for determining the overall chroma-tin structure. In addition, CpG motifs in the DNA, which are rare and are often clustered in CpG-rich regions within promoters, can be methylated or demethylated. when the CpG motifs are methylated, they are often associated with chromatin-remodelling factors, such as methyl-DNA-binding pro-teins, which results in the condensation of chromatin. The opposite occurs following the demethylation of CpG motifs, which results in the relaxation of chromatin and an increased accessibility of target sequences, thereby allowing the binding of specific transcription factors. These epigenetic mechanisms of transcriptional control have been of great interest in recent years, as they are believed to imprint the activity state of specific gene loci, such that an environ-mentally induced phenotype might become heritable and be maintained over numer-ous cell divisions. These mechanisms are discussed in more detail in the review by wilson et al. in this issue of Nature Reviews Immunology56. Here, we focus on the epi-genetic modifications of the FOXP3 locus that allow stable FOXP3 expression. In addition, in BOX 1 we highlight how post-translational modifications of the FOXP3 protein and other downstream events influ-ence FOXP3 function and the chromatin remodelling of its target genes.

In mice and humans, distinct regions of the FOXP3 locus have a pattern of DNA methylation and specific histone modifi-cations that differ between TReg cells and conventional T cells. Sequence analyses have revealed three highly conserved non-coding regions in the FOXP3 locus (FIG. 3), all of which have been found to be subject to epi-genetic modifications and to be involved in regulating the transcription of FOXP3.

FOXP3 promoter. The FOXP3 promoter, which is located 6.5 kb upstream of the first coding exon of FOXP3, is a classic TATA- and CAAT-box-containing promoter that is activated in response to TCR signalling through binding of NFAT and AP1 (reF. 14). TReg cells and resting conventional T cells show differences in the epigenetic modifica-tion of the FOXP3 promoter in both mice and humans: the CpG motifs in the FOXP3 promoter are almost completely demethyl-ated in TReg cells, whereas they are weakly methylated in resting conventional T cells19,57. Furthermore, the FOXP3 promoter shows a stronger association with acetylated histones in TReg cells than in conventional T cells14,19, suggesting that the FOXP3 promoter is more accessible in TReg cells. Following in vitro acti-vation of conventional mouse T cells, Foxp3 promoter methylation is increased19, which might further restrict the accessibility of the promoter and prevent the induction of FOXP3 expression in these cells.

TGFβ sensor. The second highly conserved non-coding region in the FOXP3 locus has been identified as a TGFβ-sensitive element that contains binding sites for NFAT and SMADs. The chromatin in this region is also in an accessible state in cells that express FOXP3, as indicated by the increased levels of acetylated histone H4 in this region in both natural and TGFβ-induced TReg cells39. Moreover, TGFβ-induced chromatin remodelling of this region might even affect the accessibility of the upstream FOXP3 promoter, as the level of promoter demethy-lation was found to be slightly increased in TGFβ-treated mouse T cells19. A similar opening of the FOXP3 promoter (that is, a slight increase in demethylation and in the association with acetylated histones) was observed in activated conventional human T cells that transiently express FOXP3 (reFs 14,57). This effect was observed even without the addition of exogenous TGFβ. However, this activation-induced opening of the FOXP3 promoter might have been due to low levels of TGFβ in the culture medium, as neutralization of TGFβ was recently

Box 1 | Cooperation of forkhead box P3 and chromatin-modifying enzymes

In addition to the recently recognized importance of epigenetic mechanisms for the regulation of forkhead box P3 (FOXP3) gene expression, it has been reported that the interaction of the FOXP3 protein with chromatin-remodelling enzymes, such as histone acetyl transferases and histone deacetylases, is important for FOXP3 function69,70. The recruitment of these enzymes leads to the acetylation of the FOXP3 protein (which is a prerequisite for optimal FOXP3 function67,70) and also results in the modification of the loci to which FOXP3 binds69,71, thereby enabling the epigenetic regulation of FOXP3 target genes70,71. Through these cooperative interactions, FOXP3 can act as both a transcriptional activator and repressor: depending on whether the target genes are activated or inhibited by FOXP3, different enzymes are recruited and the appropriate chromatin modifications (either permissive or repressive) are induced69. Extracellular modifiers, such as T-cell receptor stimuli and interleukin-6- or TGFβ-mediated signals also influence the acetylation of FOXP3 and its chromatin-binding efficiency in a histone deacetylase-dependent manner72. Together, these data indicate that epigenetic modifiers govern regulatory T cells in two ways: first, by controlling FOXP3 gene expression (see main text) and second, by facilitating FOXP3 protein function.

P r o g r e s s

86 | FEBRuARy 2009 | vOluME 9 www.nature.com/reviews/immunol

P r o g r e s s

© 2009 Macmillan Publishers Limited. All rights reserved

reported to completely abrogate the activa-tion-induced transient expression of FOXP3 in human T cells16. This finding suggests that a different sensitivity to low levels of TGFβ in mouse and human T cells might account for the differences in activation-induced FOXP3 expression that are observed in the two species.

TReg-cell-specific demethylated region. The most striking differences regarding the methylation pattern at the FOXP3 locus have been observed in a third, highly conserved CpG-rich region. This region was found to be fully demethylated in TReg cells and methylated in conventional T cells12,19,58,59, and in this article is referred to as the TReg-cell-specific demethylated region (TSDR). In addition, acetylated his-tones H3 and H4, and trimethylated lysine 4 in histone H3, were found to accumulate at the TSDR12. The TSDR has enhancer activ-ity that is markedly decreased following methylation60; this is supported by the find-ing that the transcription factor CREB binds to the TSDR when this region is demethyl-ated19. Together, these data suggest a role for DNA methylation in the molecular regulation of FOXP3 expression. However, the methylation state of the TSDR seems to be irrelevant for determining the level of FOXP3 expression, as TGFβ-induced TReg cells express levels of FOXP3 that are comparable to those of natural TReg cells, despite a lack of TSDR demethylation12,60. Evidence is now accumulating that TSDR demethylation does not act as an on/off switch, but instead determines the stability of FOXP3 expression12,59,60, a concept that is consistent with the known role of epigenetic regulation in T-cell lineage decisions61.

So, demethylation of the TSDR corre-sponds with stability of FOXP3 expression (as in natural TReg cells), whereas T cells that express FOXP3 only transiently (TGFβ-induced TReg cells and recently activated conventional human T cells) have a methyl-ated TSDR12,58,60. These data are supported by the finding that drug-mediated DNA demethylation in conventional T cells led to the induction of stable FOXP3 expression and a TReg-cell phenotype19,59,60, and only the fraction of cells that expressed FOXP3 was found to have a demethylated TSDR60. Interestingly, TReg cells that were induced in vivo from conventional CD4+ T cells fol-lowing targeting of antigen to steady-state dendritic cells that were expressing DEC205 also had stable FOXP3 expression and, cor-respondingly, demethylation of the TSDR60. It remains to be determined whether other

protocols that are known to induce tolerance in vivo (such as oral administration of anti-gen and allergen-specific immunotherapy) also induce TReg cells with a stable pheno-type. It is probable that the methylation status of the TSDR, the FOXP3 promoter and potentially additional regulatory regions will be valuable biomarkers for the detection of stably suppressive TReg cells57,58,62. This is especially relevant in humans, as transient expression of FOXP3 is observed in acti-vated conventional human T cells.

Concluding remarksIn view of the potential clinical applications of TReg cells, elucidating the combination of sig-nals that imprint FOXP3 expression and TReg-cell function in vivo remains an important challenge. Future studies should aim to con-firm that the imprinted differentiation state found in natural TReg cells is as fixed as we currently believe it to be, as existing concepts of the heritability of epigenetic patterns have been challenged by recent reports of fluctuat-ing methylation states of dynamic promoters63

Nature Reviews | Immunology

FOXP3 promoter TGFβ sensor (enhancer) TSDR (enhancer and/or stabilizer)

FOXP3– conventional T cells

TGFβ-induced FOXP3+ cells

Stable FOXP3+ TReg cells

a

b

No FOXP3expression

Unstable FOXP3expression

-1 1 2 3 4 5 6 7 8 9 10 11

TCR stimulation,IL-2

TCR stimulation,TGFβ

?

NFAT NFAT

AP1 TIEG1

TIEG1

SP1 STAT5

SMAD3

Stable FOXP3expression

TCR stimulation,IL-2

TCR stimulation,TGFβ

NFAT NFAT

AP1

SP1 STAT5

STAT5SMAD3

CREBand ATF

DNA methylation

Histoneacetylation

CpG motif

Figure 3 | The FOXP3 locus is subject to epigenetic control. a | The forkhead box P3 (FOXP3) gene, which is located on the X chromosome, is highly conserved, as illustrated in the interspecies conserva‑tion plot (obtained from the University of California Santa Cruz genome assembly web site; seg‑ment shown is located between bases 48,994,000 and 49,009,118). b | Three conserved non‑coding regions that undergo epigenetic modifications and are involved in the regulation of FOXP3 transcrip‑tion are highlighted. epigenetic modifications that occur in these three regulatory regions, including histone acetylation and DNA methylation, are depicted for FOXP3– conventional T cells, transforming growth factor‑β (TgFβ)‑induced FOXP3+ T cells and natural T

Reg cells (natural T

Reg cells show a stable

FOXP3+ phenotype). Note that the TgFβ‑sensor region does not contain Cpg motifs. The hypothetical open chromatin conformation that is induced by permissive histone modifications and DNA demeth‑ylation allows the binding of transcription factors to regulatory sites and thereby enables the induction and stabilization of FOXP3 expression. Upstream signalling pathways that affect these regions when activated are depicted (if known). AP1, activator protein 1; ATF, activating transcription factor; IL‑2, interleukin‑2; CReB, cyclic‑AMP‑responsive‑element‑binding protein; NFAT, nuclear factor of acti‑vated T cells; SMAD3, mothers against decapentaplegic homologue 3; STAT5, signal transducer and activator of transcription 5; TCR, T‑cell receptor; TIeg1, TgFβ‑inducible early gene 1.

P r o g r e s s

NATuRE REvIEwS | Immunology vOluME 9 | FEBRuARy 2009 | 87

f o c u s o n c d 4 + T- c e l l d i v e r s i T y

© 2009 Macmillan Publishers Limited. All rights reserved

and by indications that TReg cells can become Il-17-producing cells following exposure to inflammatory stimuli64–66. The conversion of TReg cells to TH17 cells can be induced by dendritic-cell-derived Il-6, which downregu-lates the expression of FOXP3 in a STAT3-dependent manner64,66. However, one study65 reported that, although Il-6 or TGFβ did not affect the generation of Il-17-producing cells, their differentiation was increased by exogenous Il-1β, Il-23 and Il-21, an effect that could be prevented by histone deacety-lase inhibitors. As recent work suggests that chromatin-modifying agents, such as DNA methyltransferase and histone deacetylase inhibitors, can be used to manipulate TReg-cell biology19,59,60,67,68, there is promise that the pro-gramming and reprogramming of TReg cells at the epigenetic level may be an important approach for the development of drugs that target these cells.

Jochen Huehn and Julia K. Polansky are at the

Department of Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany.

Alf Hamann is at the Department of Experimental Rheumatology, Charité University Medicine,

Berlin, Germany.

Correspondence to J.H. e-mail: [email protected]

doi:10.1038/nri2474 Published online 30 December 2008

1. Fontenot, J. D., Gavin, M. A. & Rudensky, A. Y. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nature Immunol. 4, 330–336 (2003).

2. Khattri, R., Cox, T., Yasayko, S. A. & Ramsdell, F. An essential role for Scurfin in CD4+CD25+ T regulatory cells. Nature Immunol. 4, 337–342 (2003).

3. Hori, S., Nomura, T. & Sakaguchi, S. Control of regulatory T cell development by the transcription factor Foxp3. Science. 299, 1057–1061 (2003).

4. Fontenot, J. D., Dooley, J. L., Farr, A. G. & Rudensky, A. Y. Developmental regulation of Foxp3 expression during ontogeny. J. Exp. Med. 202, 901–906 (2005).

5. Liston, A. et al. Differentiation of regulatory Foxp3+ T cells in the thymic cortex. Proc. Natl Acad. Sci. USA 105, 11903–11908 (2008).

6. Aschenbrenner, K. et al. Selection of Foxp3+ regulatory T cells specific for self antigen expressed and presented by Aire+ medullary thymic epithelial cells. Nature Immunol. 8, 351–358 (2007).

7. Siewert, C. et al. Experience-driven development: effector/memory-like aE

+Foxp3+ regulatory T cells originate from both naive T cells and naturally occurring naive-like regulatory T cells. J. Immunol. 180, 146–155 (2008).

8. Kretschmer, K. et al. Inducing and expanding regulatory T cell populations by foreign antigen. Nature Immunol. 12, 1219–1227 (2005).

9. Sun, C. M. et al. Small intestine lamina propria dendritic cells promote de novo generation of Foxp3 T reg cells via retinoic acid. J. Exp. Med. 204, 1775–1785 (2007).

10. Coombes, J. L. et al. A functionally specialized population of mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-β and retinoic acid-dependent mechanism. J. Exp. Med. 204, 1757–1764 (2007).

11. Chen, W. et al. Conversion of peripheral CD4+CD25– naive T cells to CD4+CD25+ regulatory T cells by TGF-β induction of transcription factor Foxp3. J. Exp. Med. 198, 1875–1886 (2003).

12. Floess, S. et al. Epigenetic control of the foxp3 locus in regulatory T cells. PLOS Biol. 5, e38 (2007).

13. Walker, M. R. et al. Induction of FoxP3 and acquisition of T regulatory activity by stimulated human CD4+CD25– T cells. J. Clin. Invest. 112, 1437–1443 (2003).

14. Mantel, P. Y. et al. Molecular mechanisms underlying FOXP3 induction in human T cells. J. Immunol. 176, 3593–3602 (2006).

15. Gavin, M. A. et al. Single-cell analysis of normal and FOXP3-mutant human T cells: FOXP3 expression without regulatory T cell development. Proc. Natl Acad. Sci. USA 103, 6659–6664 (2006).

16. Tran, D. Q., Ramsey, H. & Shevach, E. M. Induction of FOXP3 expression in naive human CD4+FOXP3– T cells by T cell receptor stimulation is TGF-β- dependent but does not confer a regulatory phenotype. Blood 110, 2983–2990 (2007).

17. Allan, S. E. et al. Activation-induced FOXP3 in human T effector cells does not suppress proliferation or cytokine production. Int. Immunol. 19, 345–354 (2007).

18. Pillai, V., Ortega, S. B., Wang, C. K. & Karandikar, N. J. Transient regulatory T-cells: a state attained by all activated human T-cells. Clin. Immunol. 123, 18–29 (2007).

19. Kim, H. P. & Leonard, W. J. CREB/ATF-dependent T cell receptor-induced FoxP3 gene expression: a role for DNA methylation. J. Exp. Med. 204, 1543–1551 (2007).

20. Gupta, S. et al. Differential requirement of PKC-θ in the development and function of natural regulatory T cells. Mol. Immunol. 46, 213–214 (2008).

21. Wang, H. et al. A potential side effect of cyclosporin A: inhibition of CD4+CD25+ regulatory T cells in mice. Transplantation 82, 1484–1492 (2006).

22. Coenen, J. J. et al. Rapamycin, not cyclosporine, permits thymic generation and peripheral preservation of CD4+ CD25+ FoxP3+ T cells. Bone Marrow Transplant. 39, 537–545 (2007).

23. San Segundo, D., Fabrega, E., Lopez-Hoyos, M. & Pons, F. Reduced numbers of blood natural regulatory T cells in stable liver transplant recipients with high levels of calcineurin inhibitors. Transplant. Proc. 39, 2290–2292 (2007).

24. Brandt, C., Pavlovic, V., Worm, M., Radbruch, A. & Baumgrass, R. Low-dose cyclosporine A therapy increases the regulatory T cell population in patients with atopic dermatitis. Allergy (in the press).

25. Kim, J. M. & Rudensky, A. The role of the transcription factor Foxp3 in the development of regulatory T cells. Immunol. Rev. 212, 86–98 (2006).

26. Sauer, S. et al. T cell receptor signaling controls Foxp3 expression via PI3K, Akt, and mTOR. Proc. Natl Acad. Sci. USA 105, 7797–7802 (2008).

27. Haxhinasto, S., Mathis, D. & Benoist, C. The AKT –mTOR axis regulates de novo differentiation of CD4+Foxp3+ cells. J. Exp. Med. 205, 565–574 (2008).

28. Tai, X., Cowan, M., Feigenbaum, L. & Singer, A. CD28 costimulation of developing thymocytes induces Foxp3 expression and regulatory T cell differentiation independently of interleukin 2. Nature Immunol. 6, 152–162 (2005).

29. Benson, M. J., Pino-Lagos, K., Rosemblatt, M. & Noelle, R. J. All-trans retinoic acid mediates enhanced T reg cell growth, differentiation, and gut homing in the face of high levels of co-stimulation. J. Exp. Med. 204, 1765–1774 (2007).

30. Fontenot, J. D., Rasmussen, J. P., Gavin, M. A. & Rudensky, A. Y. A function for interleukin 2 in Foxp3-expressing regulatory T cells. Nature Immunol. 6, 1142–1151 (2005).

31. Mayack, S. R. & Berg, L. J. Cutting Edge: an alternative pathway of CD4+ T cell differentiation is induced following activation in the absence of γ-chain-dependent cytokine signals. J. Immunol. 176, 2059–2063 (2006).

32. Yao, Z. et al. Nonredundant roles for Stat5a/b in directly regulating Foxp3. Blood 109, 4368–4375 (2007).

33. Zorn, E. et al. IL-2 regulates FOXP3 expression in human CD4+CD25+ regulatory T cells through a STAT dependent mechanism and induces the expansion of these cells in vivo. Blood 108, 1571–1579 (2006).

34. Burchill, M. A., Yang, J., Vogtenhuber, C., Blazar, B. R. & Farrar, M. A. IL-2 receptor β-dependent STAT5 activation is required for the development of Foxp3+ regulatory T cells. J. Immunol. 178, 280–290 (2007).

35. D’Cruz, L. M. & Klein, L. Development and function of agonist-induced CD25+Foxp3+ regulatory T cells in

the absence of interleukin 2 signaling. Nature Immunol. 6, 1152–1159 (2005).

36. Marie, J. C., Letterio, J. J., Gavin, M. & Rudensky, A. Y. TGF-β1 maintains suppressor function and Foxp3 expression in CD4+CD25+ regulatory T cells. J. Exp. Med. 201, 1061–1067 (2005).

37. Liu, Y. et al. A critical function for TGF-β signaling in the development of natural CD4+CD25+Foxp3+ regulatory T cells. Nature Immunol. 9, 632–640 (2008).

38. Venuprasad, K. et al. The E3 ubiquitin ligase Itch regulates expression of transcription factor Foxp3 and airway inflammation by enhancing the function of transcription factor TIEG1. Nature Immunol. 9, 245–253 (2008).

39. Tone, Y. et al. Smad3 and NFAT cooperate to induce Foxp3 expression through its enhancer. Nature Immunol. 9, 194–202 (2008).

40. Xiao, S. et al. Retinoic acid increases Foxp3+ regulatory T cells and inhibits development of Th17 cells by enhancing TGF-β-driven Smad3 signaling and inhibiting IL-6 and IL-23 receptor expression. J. Immunol. 181, 2277–2284 (2008).

41. Hill, J. A. et al. Retinoic acid enhances Foxp3 induction indirectly by relieving inhibition from CD4+CD44hi cells. Immunity 29, 758–770 (2008).

42. Samon, J. B. et al. Notch1 and TGFβ1 cooperatively regulate Foxp3 expression and the maintenance of peripheral regulatory T cells. Blood 112, 1813–1821 (2008).

43. Ou-Yang, H. F. et al. Notch signaling regulates the FOXP3 promoter through RBP-J- and Hes1-dependent mechanisms. Mol. Cell. Biochem. 8 Sep 2008 (doi: 10.1007/s11010-008-9912–9914).

44. Wei, J. et al. Antagonistic nature of T helper 1/2 developmental programs in opposing peripheral induction of Foxp3+ regulatory T cells. Proc. Natl Acad. Sci. USA 104, 18169–18174 (2007).

45. Mantel, P. Y. et al. GATA3-driven Th2 responses inhibit TGF-β1-induced FOXP3 expression and the formation of regulatory T cells. PLoS Biol. 5, e329 (2007).

46. Dardalhon, V. et al. IL-4 inhibits TGF-β-induced Foxp3+ T cells and, together with TGF-β, generates IL-9+ IL-10+ Foxp3– effector T cells. Nature Immunol. 9, 1347–1355 (2008).

47. Takaki, H. et al. STAT6 inhibits TGF-β-mediated Foxp3 induction through direct binding to the Foxp3 promoter, which is reverted by retinoic acid receptor. J. Biol. Chem. 283, 14955–14962 (2008).

48. Fragale, A. et al. IFN regulatory factor-1 negatively regulates CD4+ CD25+ regulatory T cell differentiation by repressing Foxp3 expression. J. Immunol. 181, 1673–1682 (2008).

49. Fazi, F. & Nervi, C. MicroRNA: basic mechanisms and transcriptional regulatory networks for cell fate determination. Cardiovasc. Res. 79, 553–561 (2008).

50. Hoefig, K. P. & Heissmeyer, V. MicroRNAs grow up in the immune system. Curr. Opin. Immunol. 20, 281–287 (2008).

51. Cobb, B. S. et al. A role for Dicer in immune regulation. J. Exp. Med. 203, 2519–2527 (2006).

52. Zhou, X. et al. Selective miRNA disruption in T reg cells leads to uncontrolled autoimmunity. J. Exp. Med. 205, 1983–1991 (2008).

53. Chong, M. M., Rasmussen, J. P., Rundensky, A. Y. & Littman, D. R. The RNAseIII enzyme Drosha is critical in T cells for preventing lethal inflammatory disease. J. Exp. Med. 205, 2005–2017 (2008).

54. Liston, A., Lu, L. F., O’Carroll, D., Tarakhovsky, A. & Rudensky, A. Y. Dicer-dependent microRNA pathway safeguards regulatory T cell function. J. Exp. Med. 205, 1993–2004 (2008).

55. Marson, A. et al. Foxp3 occupancy and regulation of key target genes during T-cell stimulation. Nature 445, 931–935 (2007).

56. Wilson, B., Rowell, E. & Sekimata, M. Epigenetic control of T-helper-cell differentiation. Nature Rev. Immunol. 9, 91–105 (2009).

57. Janson, P. C. et al. FOXP3 promoter demethylation reveals the committed Treg population in humans. PLoS ONE 3, e1612 (2008).

58. Baron, U. et al. DNA demethylation in the human FOXP3 locus discriminates regulatory T cells from activated FOXP3+ conventional T cells. Eur. J. Immunol. 37, 2378–2389 (2007).

59. Nagar, M. et al. Epigenetic inheritance of DNA methylation limits activation-induced expression of FOXP3 in conventional human CD25–CD4+ T cells. Int. Immunol. 20, 1041–1055 (2008).

60. Polansky, J. et al. DNA methylation controls Foxp3 gene expression. Eur. J. Immunol. 38, 1654–1663 (2008).

P r o g r e s s

88 | FEBRuARy 2009 | vOluME 9 www.nature.com/reviews/immunol

P r o g r e s s

© 2009 Macmillan Publishers Limited. All rights reserved

61. Ansel, K. M., Lee, D. U. & Rao, A. An epigenetic view of helper T cell differentiation. Nature Immunol. 4, 616–623 (2003).

62. Wieczorek, G. et al. Quantitative DNA methylation analysis of FOXP3 as a new method for counting regulatory T cells in solid tissue and peripheral blood. Cancer Res. 69, 599–608 (2009).

63. Metivier, R. et al. Cyclical DNA methylation of a transcriptionally active promoter. Nature 452, 45–50 (2008).

64. Xu, L., Kitani, A., Fuss, I. & Strober, W. Cutting Edge: regulatory T cells induce CD4+CD25–Foxp3– T cells or are self-induced to become Th17 cells in the absence of exogenous TGF-β. J. Immunol. 178, 6725–6729 (2007).

65. Koenen, H. J. et al. Human CD25highFoxp3pos regulatory T cells differentiate into IL-17-producing cells. Blood 112, 2340–2352 (2008).

66. Radhakrishnan, S. et al. Reprogrammed FoxP3+ T regulatory cells become IL-17+ antigen-specific autoimmune effectors in vitro and in vivo. J. Immunol. 181, 3137–3147 (2008).

67. Tao, R. et al. Deacetylase inhibition promotes the generation and function of regulatory T cells. Nature Med. 13, 1299–1307 (2007).

68. Reilly, C. M. et al. The histone deacetylase inhibitor trichostatin A upregulates regulatory T cells and modulates autoimmunity in NZB/W F1 mice. J. Autoimmun. 31, 123–130 (2008).

69. Chen, C., Rowell, E. A., Thomas, R. M., Hancock, W. W. & Wells, A. D. Transcriptional regulation by Foxp3 is associated with direct promoter occupancy and

modulation of histone acetylation. J. Biol. Chem. 281, 36828–36834 (2006).

70. Li, B. et al. FOXP3 ensembles in T-cell regulation. Immunol. Rev. 212, 99–113 (2006).

71. Kwon, H. K. et al. Foxp3 induces IL-4 gene silencing by affecting nuclear translocation of NFkB and chromatin structure. Mol. Immunol. 45, 3205–3212 (2008).

72. Samanta, A. et al. TGF-β and IL-6 signals modulate chromatin binding and promoter occupancy by acetylated FOXP3. Proc. Natl Acad. Sci. USA 105, 14023–14027 (2008).

AcknowledgementsWe thank A. Rao for helpful discussions and R. Baumgrass, B. Schraven, J. Lindquist and M. Merkenschlager for critical reading of the manuscript.

DATABASESentrez gene: http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=geneAP1 | CReB | FOXP3| IL‑2 | ITCH| JAK1| JAK3| NFAT | PKCθ | SMAD3 | STAT5 | TgFβ | TIeg1

FURTHER INFORMATIONJochen Huehn’s homepage: http://www.helmholtz‑hzi.de/en/research/research_groups/other_research_groups/experimental_immunologyThe university of california santa cruz genome assembly web site: http://genome.ucsc.edu/

All lInks Are AcTIve In The onlIne Pdf

P r o g r e s s

NATuRE REvIEwS | Immunology vOluME 9 | FEBRuARy 2009 | 89

f o c u s o n c d 4 + T- c e l l d i v e r s i T y

© 2009 Macmillan Publishers Limited. All rights reserved