You are on page 1of 9

FOCUS | Review Article

FOCUS | Review Article https://doi.org/10.1038/s41590-018-0120-4

https://doi.org/10.1038/s41590-018-0120-4

Regulatory T cells in autoimmune disease


Margarita Dominguez-Villar1* and David A. Hafler1,2*

In recent years, the understanding of regulatory T cell (Treg cell) biology has expanded considerably. Key observations have
challenged the traditional definition of Treg cells and have provided insight into the underlying mechanisms responsible for the
development of autoimmune diseases, with new therapeutic strategies that improve disease outcome. This Review summarizes
the newer concepts of Treg cell instability, Treg cell plasticity and tissue-specific Treg cells, and their relationship to autoimmunity.
Those three main concepts have changed the understanding of Treg cell biology: how they interact with other immune and non-
immune cells; their functions in specific tissues; and the implications of this for the pathogenesis of autoimmune diseases.

M
ore than 20 years after their ‘re-discovery’, regulatory T cells autoimmune disease but also to inhibit such disease. That led to the
(Treg cells) have emerged as an important component in hypothesis that in the periphery of normal mice there are two popula-
understanding of the immune response to pathogens and tions of CD4+ T cells: one population potentially capable of inducing
the mechanisms of peripheral tolerance that control the development autoimmunity, and a second population of ‘suppressor’ cells, different
of allergies and autoimmune diseases. In mice and humans, Treg cells from helper T cells, that would inhibit autoreactive T cells. Treg cell
are characterized by high expression of the cytokine IL-2 receptor research during those years floundered, in part due to the failure to find
α​-chain (CD25) and expression of the transcription factor Foxp3, specific cell markers that could define this population and the ambi-
which is required for their development, function and stability1–4. In guity observed in their suppressive mechanisms. The identification of
humans, as CD25 is also expressed by activated CD4+ T cells, absence a population of CD4+ T cells responsible for controlling autoreactive
of the IL-7 receptor α​-chain (CD127) is used as a marker complemen- responses13, as well as the search for a cell-surface marker that would
tary to CD25 to more precisely identify human Treg cells5. Moreover, define this population, indicated that a subset of CD25-expressing
numerous surface receptors have been described that are variably CD4+ T cells present in the periphery of healthy mice was responsible
specific for defined Treg cell subsets, indicative of the heterogeneity of for the inhibition of autoimmunity. The transfer of cell suspensions
this population6. Treg cells can be broadly classified into two groups on from the spleen of BALB/c mice depleted of CD25+ cells into athymic
the basis of their developmental origin. Thymic Treg cells (tTreg cells), nude mice induced autoimmunity that affected several organs, and
also known as ‘natural Treg cells’, are generated in the thymus as a sepa- co-transfer of CD25+CD4+ T cells inhibited disease4. In 2001, human
rate lineage at the stage of CD4+ single-positive thymocytes and are Treg cells were identified in the thymus and peripheral blood of healthy
thought to show enrichment for T cell antigen receptors (TCRs) with people as those CD4+ T cells with very high expression of CD25 and
high affinity for self peptides7. Although the details of their suppres- phenotype and function very similar to that of their rodent counter-
sive mechanisms are still not completely understood, and these mech- part14,15. Although there is still no definitive marker for the isolation
anisms are most probably dependent on the microenvironment and of human Treg cells, these studies, together with the discovery of Foxp3
the target population to be controlled, in general these cells perform as the master transcription factor of Treg cells1,3,16, laid the groundwork
their function both by cell-contact mechanisms that involve specific for the beginning of in-depth analysis of Treg cell biology in health and
cell-surface receptors and by the secretion of inhibitory cytokines such disease (Box 1).
as IL-10, TGF-β​and IL-357. Induced Treg cells (iTreg cells) develop from As for Treg cell function in human autoimmune disease, early
conventional CD4+ T cells in the periphery after antigen encounter studies of patients revealed that in various autoimmune disor-
and in the presence of specific factors such as TGF-β​ and IL-28. So ders, there is a defect in either the number or the function of Treg
far there is (are) no definitive protein marker(s) that distinguish(es) cells isolated from the peripheral blood, with those data being
between these two Treg cell populations in vitro or in vivo, although supported by in vivo models of disease (a comprehensive list of
there are important differences in their epigenetic signatures, and par- autoimmune diseases is in ref. 17). Although some of these early
ticularly in the Foxp3 locus, that make iTreg cells intrinsically unstable data were misleading due to the confounding variable of the iden-
in inflammatory and/or stress conditions8,9. tification of human Treg cells based on the positive expression of
From a historical perspective, the initial observations made in the CD25 as the only marker for Treg cell identification, subsequent
1970s that led to the definition of Treg cells in the 1990s are tightly work has clearly demonstrated that most autoimmune diseases,
linked to autoimmune diseases. Seminal work by Nishizuka and such as type 1 diabetes18–20, multiple sclerosis21,22, systemic lupus
Sakakura back in 1969 demonstrated that thymectomy in healthy erythematosus23, myasthenia gravis24, rheumatoid arthritis25 and
neonatal mice led to inflammation and severe organ-specific auto- others17, display defects in either the number or the function of
immune pathology, which indicated the involvement of a thymus- tTreg cells in peripheral blood.
derived population in control of self-tolerance10. That observation Investigations during the past two decades have advanced
was further confirmed in adult rats that underwent thymectomy knowledge of the mechanisms that underlie the development of tTreg
and were subjected to sublethal irradiation11. Most notably, inocula- cells in the thymus, as well as their gene-expression signature, their
tion with CD4+ T cells from healthy syngeneic mice inhibited dis- role in controlling both immune and non-immune cell responses
ease in both systems, and once autoimmunity was established, CD4+ and their effect on immune system–mediated diseases (reviewed in
T cells isolated from these sick mice and adoptively transferred into refs 7,26), and, finally, the identity of the tissues in which they exert
T cell–deficient mice were able to induce disease12. These experiments functions beyond suppression27,28. Moreover, the traditional view
demonstrated that T cells are able not only to serve as inducers of that Treg cells are a terminally differentiated population not capable

Department of Neurology, Yale School of Medicine, New Haven, CN, USA. 2Department of Immunobiology, Yale School of Medicine, New Haven, CN,
1

USA. *e-mail: margarita.dominguez-villar@yale.edu; david.hafler@yale.edu

Nature Immunology | VOL 19 | JULY 2018 | 665–673 | www.nature.com/natureimmunology 665


© 2018 Nature America Inc., part of Springer Nature. All rights reserved.
Review Article
|
Review Article FOCUS
https://doi.org/10.1038/s41590-018-0120-4 NaTuRe Immunology

Box 1 | How are Treg cells studied in autoimmunity? biology1–3. Inactivating mutations in Foxp3 result in the spontane-
ous development of severe autoimmunity with a scurfy phenotype
Studies of Treg cells in autoimmunity have been facilitated by the in mice29 and IPEX syndrome (‘immune dysregulation, polyendo-
existence of mouse models for many autoimmune diseases130. crinopathy, enteropathy, X-linked’) in humans30,31. Foxp3 is neces-
While these models of autoimmune diseases allow fate-mapping sary for the development, maintenance and function of tTreg cells1–3,
and gene-modification experiments that cannot be performed although alone it is not sufficient for full recapitulation of the Treg
in humans, they do not generally reflect the genetic architecture cell phenotype9,32. Beyond Foxp3, the second requirement necessary
that underlies human autoimmune diseases; that is, the models for establishment of the Treg cell functional program is the genera-
often reflect the consequential aspects of disease pathogenesis tion of a specific epigenetic signature acquired during development
but not the underlying mechanisms that cause the activation of and finalized in the periphery9,33. Both have essential roles in main-
autoreactive T cells. taining Treg cell function, and alteration of Foxp3 or epigenetic mod-
Human Treg cell studies are hindered by the lack of a definitive ifications in auto-inflammatory conditions are probably the cause
surface marker that uniquely isolates Treg cells from other of the Treg cell instability and aberrant plasticity observed in several
T cell populations. Thus, much of the early work assessing the autoimmune settings (Fig. 1). Due to the importance of Foxp3 in
frequency of Treg cells in peripheral blood of patients needs to be Treg cell maintenance and the prevention of autoimmunity, regu-
considered with caution, as Treg cells were defined solely by the lation of Foxp3 expression is a matter of active research. Foxp3 is
positive expression of CD25 and the results can be misleading, subjected to two main layers of regulation, transcriptional and post-
as human conventional T cells can also express CD25 after translational, both of which are responsive to positive and negative
being activated. Moreover, human studies are limited to ex vivo regulation by factors in the tissue environment, including cytokines,
characterization or short in vitro experiments and depend on the metabolic mediators and inflammatory factors.
availability of samples from patients with autoimmunity. Foxp3 expression is a tightly regulated process, and while the
Recent advances in high-throughput technologies that mechanisms by which Foxp3 is transcriptionally regulated by tran-
require small numbers of cells will considerably affect research scription factors during tTreg cell development and in mature Treg
into human Treg cell autoimmunity, allowing the analysis of rare cells have been clearly established (reviewed in ref. 34), data have
Treg cell populations from small tissue samples, etc. Currently shown that in rodents, Foxp3 transcription is also controlled epige-
available technologies are able to determine the epigenetic and netically. The Foxp3 locus contains several conserved non-coding
transcriptional signature at the single-cell level. The combination sequences (CNSs) that are critical for the initiation and maintenance
of these datasets and systems-biology approaches in the analysis of Foxp3 transcription35. Among the three CNSs described so far,
of serial samples obtained from people at different time points, only CNS2 has been demonstrated to prevent autoimmunity. It is a
before and after therapeutic intervention and from different TCR-responsive enhancer with binding sites for Runx1–CBFβ​ tran-
anatomical locations is delivering an enormous amount of scription-factor complexes that is important in maintaining Foxp3’s
information about the response of patients to treatment, disease stability. CNS2-deficient mice develop spontaneous autoimmunity,
course and mechanistic insights. which emphasizes the importance of CNS2 in the stability and func-
tion of Treg cells36,37. Furthermore, CNS2 contains a conserved CpG
island (TSDR) that is hypomethylated specifically in Treg cells; thus,
of secreting pro-inflammatory cytokines and whose only function it is transcriptionally active in tTreg cells but is hypermethylated in
is to suppress T cell responses has been challenged by recent data. naive or effector T cells38,39. This region has been widely used to dis-
Specifically, Treg cells possess some degree of plasticity and instabil- tinguish true tTreg cells from conventional T cells transiently upregu-
ity, although in-depth understanding of the molecular mechanisms lating Foxp3 expression and iTreg cells38,39.
that drive these two states and the relationship between them in dis- Three main post-translational modifications have been
ease settings remains incomplete. described for Foxp3 protein: acetylation, phosphorylation and
This Review will provide an update on recent discoveries in Treg ubiquitination. These modifications affect the stability and DNA-
cell biology in the context of autoimmune disease. From our per- binding capacity of Foxp3 and thus modulate Treg cell function
spective, three main observations have changed the understand- and the development of autoimmunity. The acetylation of spe-
ing of what Treg cells are, how they function in peripheral lymphoid cific lysine residues by lysine acetyltransferases globally stabilizes
organs and non-immune tissues, how they relate to other immune Foxp3 expression and promotes Treg cell function by favoring the
and non-immune cells, and how their phenotype and function can binding of Foxp3 to its transcriptional targets40 and allowing it to
be modulated, with clear consequences for the development of avoid proteasomal degradation40. Several acetylases and deacety-
new therapeutic strategies for various autoimmune diseases. These lases, such as TIP6041 and p30040,42, have been shown to interact
observations are as follows: 1) the instability of Treg cells and their with Foxp3 and control its acetylation. Other post-translational
acquisition of an effector phenotype after losing Foxp3 expression processes include phosphorylation at serine and threonine resi-
under inflammatory conditions; 2) the plasticity of the phenotype dues by several kinases, including PIM-1, PIM-2 and CDK243–46,
of Treg cells, with their acquisition of effector-like properties while and ubiquitination at lysine residues, which targets Foxp3 for
maintaining Foxp3 expression; and 3) the discovery of tissue-spe- proteasomal degradation47. Inflammatory stimuli result in prote-
cific Treg cells, which demonstrates again that Treg cells, like other asome-dependent degradation of Foxp3 mediated by the ubiqui-
immune cells, are influenced by their environment. These obser- tinase Stub1, which binds to Foxp3 and promotes its Lys48-linked
vations have arisen at a time during which the development of ubiquitination48. Overexpression of Stub1 abolishes the suppres-
high-throughput genomic, epigenetic and proteomic technologies sive ability of Treg cells in vitro and in vivo and confers onto Treg
is allowing the analysis of rare cell populations at a single-cell level, cells a phenotype with similarities to that of the TH1 subset of
which will undoubtedly improve knowledge of basic Treg cell biology helper T cells; this raises the question of whether the effector
in general and of human Treg cell biology in particular. T cell–like Treg cell phenotype in this setting is an intermediate
stage en route to instability and loss of Foxp3 protein expression.
Foxp3 as a master regulator of Treg cell phenotype and In contrast, the deubiquitinating enzyme USP7 has high expres-
function sion in Treg cells and is associated with Foxp3 in the nucleus,
The discovery of Foxp3 as the master regulator of Treg cell devel- where it regulates the turnover of Foxp3. Under inflammatory
opment and function was critical for the understanding of Treg cell conditions, this enzyme is downregulated, which facilitates the

666 Nature Immunology | VOL 19 | JULY 2018 | 665–673 | www.nature.com/natureimmunology

© 2018 Nature America Inc., part of Springer Nature. All rights reserved.
FOCUS | Review
| Article
NaTuRe Immunology FOCUShttps://doi.org/10.1038/s41590-018-0120-4
Review Article

Health Autoimmunity

Treg cell instability (ex-Foxp3)

Foxp3 Global Observed in:


Transcriptional regulation protein epigenetic
Epigenetic regulation Environmental expression signature ‘ex-Foxp3’ cell
Post-translational modifications triggers NOD model
miRNAs Autoimmune EAE model
Others? inflammation Treg cell

Expression of pro-inflammatory cytokines


Downregulation of Foxp3
Pathogenic
Resolution of
Foxp3 Global inflammation
protein epigenetic
expression signature

Treg cell ? ?

Resolution of
inflammation Observed in:

TH1-like Treg cells

Treg cell plasticity Type 1 diabetes


Multiple sclerosis
HACs and HDACs
Autoimmune Foxp3 Global Sjögren syndrome
DNA methylation
inflammation protein epigenetic Autoimmune hepatitis
Histone modifications
expression signature Atherosclerosis
Others?
TH2-like Treg cells
Treg cell
Allergy and asthma
Systemic sclerosis

TH17-like Treg cells


Increased expression of helper T cell TFs
Expression of pro-inflammatory cytokines Psoriasis
Maintained Foxp3 expression Rheumatoid arthritis
Dysfunctional

Fig. 1 | The Treg cell functional program in health and autoimmunity. The Treg cell functional program depends on the following two main axes, both of
which are regulated by various factors (gray boxes) and are maintained under healthy conditions: Foxp3 expression, and a global epigenetic signature. In
genetically susceptible people, environmental triggers induce inflammation, and a proportion of Treg cells can lose Foxp3 expression and become unstable
(Treg cell instability; ex-Foxp3) or can maintain Foxp3 expression but alter their global epigenetic signature (Treg cell plasticity), secrete pro-inflammatory
cytokines and display reduced function. Treg cell instability leads to the so-called ‘ex-Foxp3 cells’, which have been observed in mouse models of diabetes
(NOD) and multiple sclerosis (EAE). Treg cell plasticity favors the acquisition of TH1-like, TH2-like or TH17-like properties by the Treg cells; each of these
outcomes have been observed in various autoimmune diseases in mice and humans. While the mechanistic connection between Treg cell instability and Treg
cell plasticity is not known, plastic Treg cells can become ‘normal’ Treg cells after resolution of inflammation. It remains to be determined whether this is also
the case for Treg cell instability. miRNA, microRNA; HACs, histone acetylases; HDACs, histone deacetylases; TFs, transcription factors.

degradation of Foxp340. Furthermore, conditional deletion of Foxp3 works in concert with other transcription factors and
USP7 in Treg cells leads to lethal autoimmunity with a decreased proteins, forming multiprotein complexes that determine the tran-
number of Treg cells in the periphery that display an aberrant TH1- scriptional signature and effector functions of Treg cells. Hundreds
like phenotype in vitro and in vivo49. Targeting USP7 decreases of protein partners have been described, including the transcrip-
recruitment of the acetyltransferase TIP60 to the CNS2 region tion factors Gata-3, NFAT, Runx1, Eos and others54. Global analysis
of Foxp3. TIP60 promotes acetylation-dependent dimerization of the Foxp3-interaction network suggests a model in which Foxp3
of Foxp350,51, and in the absence of TIP60, lethal autoimmunity and its binding partners form multi-protein complexes that bind to
develops52. Other factors have also been shown to control Foxp3 pre-existing DNA enhancers55 and regulate transcription positively
expression, such as HIF-1α​, which is induced by stimulation with or negatively depending on the interacting proteins recruited56. As
IL-6 and the TCR and inhibits Foxp3 through ubiquitination53. mentioned above, Foxp3 also associates with proteins that mediate
Both ubiquitination and acetylation target lysine residues, so epigenetic modifications, such as TIP60, Sirtuin 1 or HDAC741,57,
they might compete in the regulation of Foxp3 expression. In fact, which alter the acetylation state of partner loci and Foxp3 itself,
hyperacetylation of Foxp3 prevents its polyubiquitination and pro- with consequences for the binding of transcription factors and his-
teasomal degradation, which increases its stability40. Such data sug- tone modifications. In this context, disruption of Foxp3’s interac-
gest that post-translational modifications of Foxp3 have a crucial tions with specific proteins diminishes Treg cell function and leads
role in modulating the plasticity or instability of Treg cells, which to autoimmune responses due to increased polyubiquitination of
adds another layer of complexity to regulation of the Treg cell func- Foxp358,59. Other evidence supporting the proposal of the impor-
tional program, with potential consequences for the development tance of Foxp3-interacting partners in Treg cell function includes the
of autoimmunity. observation that different FOXP3 mutations result in a wide range

Nature Immunology | VOL 19 | JULY 2018 | 665–673 | www.nature.com/natureimmunology 667


© 2018 Nature America Inc., part of Springer Nature. All rights reserved.
Review Article
|
Review Article FOCUS
https://doi.org/10.1038/s41590-018-0120-4 NaTuRe Immunology

of IPEX disease severity, reflective of the relative importance of the nervous system at the preclinical and peak stages of EAE that
residues affected in the integrity of the Foxp3 protein and the pro- decreases during EAE resolution. These ex-Foxp3 Treg cells express
tein partners that form the DNA-binding complexes. For example, IFN-γ​and are able to transfer EAE74. It remains to be determined
the most common mutation in IPEX, p.A384T, which disrupts the whether the decrease in ex-Foxp3 cells during disease resolution is
sequence specificity of Foxp3’s DNA binding and alters Foxp3’s due to re-acquisition of Foxp3 expression by ex-Foxp3 cells.
interactions with specific target genes31, inhibits Treg cell function but Another genetic fate-mapping mouse model has provided
preserves the ability of Treg cells to repress inflammatory cytokine evidence that the majority of mature tTreg cells in the spleen and
production, due in part to a specific inhibition of Foxp3’s interaction lymph nodes are relatively stable under homeostatic conditions66.
with TIP6060. Furthermore, experiments with mice have shown that This model, based on inducible labeling of Foxp3+ cells upon treat-
this mutation specifically in Treg cells perturbs the binding of Foxp3 ment with tamoxifen, marks all those cells that expressed Foxp3
to specific target genes, including Batf, which is partly responsible at the moment of tamoxifen administration, and, in contrast to
for the induction of a unique pattern of tissue-restricted inflamma- models with continuous labeling67, prevents the detection of cells
tion in certain non-lymphoid tissues due to defective function of transiently expressing Foxp3. Although Foxp3 expression is stable
these Treg cells61. Additionally, a Foxp3 reporter mouse that expresses under homeostatic conditions, depriving cells of growth factor or
a fusion of Foxp3 with enhanced green fluorescent protein (EGFP) blocking the IL-2 receptor, which induces autoimmunity75, results
at its amino terminus (EGFP-Foxp3), which disrupts the interac- in substantially decreased Foxp3 expression per cell in mature tTreg
tion of Foxp3 with many cofactors, including TIP60, p300 and Eos, cells, and a small population loses Foxp3 expression completely.
does not develop apparent autoimmunity, but its Treg cells display However, they do not produce pro-inflammatory cytokines66, sug-
alterations in function in vivo, with autoimmune-prone mice of the gestive of some degree of instability under specific environmental
non-obese diabetic (NOD) strain developing diabetes faster than settings. This apparent discrepancy might arise from the different
their wild-type counterparts do58. Interestingly, these same Treg cells fate-mapping mouse models used and the type of labeling of Foxp3
are potent suppressors of antibody-mediated arthritis due to ‘pref- cells, which could lead to the labeling of uncommitted Treg cells67
erential’ interaction of EGFP-Foxp3 with the transcription factor or the absence of labeling of ex-Foxp3 cells that appeared before
IRF462. These findings not only demonstrate that certain cofactors tamoxifen administration66. The discordance in results could also
are crucial for Treg cell–mediated function but also suggest that Treg depend on the inflammatory stimuli used to assess Foxp3’s stabil-
cells might be ‘tuned’ to control particular types of inflammation ity. Regardless, in both fate-mapping models, there is a small pop-
by modulating the constituents of Foxp3 protein complexes under ulation of tTreg cells that lose Foxp3 expression, and those Treg cells
specific environmental conditions. Furthermore, we are tempted that remain ‘stable’ display diminished Foxp3 expression at the
to speculate that the variety of functions that Treg cells perform in single-cell level66. As decreased levels of Foxp3 in Treg cells isolated
different tissue environments could be accompanied by the forma- from inflammatory sites have been observed in mouse models of
tion of specific multi-protein complexes with tissue-specific pro- autoimmunity63,76 and in patients with autoimmune diseases24,77–81,
teins that would act cooperatively with Foxp3 in performing Treg cell further work with these models is warranted in order to identify
functions in specific environments. the mechanisms and consequences of long-term decreases in Treg
cell Foxp3 expression. Further data have confirmed the obser-
Treg cell instability vation that most mature tTreg cells are stable under steady-state
Both the expression of Foxp3 and its stability have crucial roles conditions, in a new fate-mapping mouse model in which Foxp3
in the maintenance of Treg cell function63. Thus, conditional dele- lacks CNS1, but these cells become unstable when stimulated in
tion of a Foxp3 allele in mature Treg cells results in effector T cells vitro and in vivo in a model of EAE, losing Foxp3 expression and
that are capable of causing inflammatory tissue lesions64. Although acquiring TH1 cell– and TFH cell–like features73. While epigenetic
the instability of Foxp3 in iTreg cells has been widely observed and changes such as re-methylation of the CNS2 region could account
is intrinsic to their developmental origin8,9, tTreg cells have been for the loss of Foxp3 expression in these settings67,73, the molecu-
investigated to determine how the instability of Foxp3 expression lar mechanisms responsible for the decrease in Foxp3 protein
under basal or inflammatory conditions in specific tissues affects and the potential contribution of post-translational modifica-
the development and resolution of autoimmunity65–67. Loss of Foxp3 tions of Foxp3 protein on the generation of ex-Foxp3 cells remain
expression by tTreg cells has been observed in vitro68,69, in the adop- to be explored.
tive transfer of cells into lymphopenic hosts70, in infectious settings71 Finally, deletion of specific Foxp3 partners can also precipitate the
and in graft-versus-host disease72. A fate-mapping mouse model appearance of ex-Foxp3 cells. For example, Treg cell–specific deletion
in which a yellow fluorescent protein reporter marks all cells that of the chaperone GP96 on the NOD background leads to lethal auto-
at any time expressed Foxp3 in both homeostatic conditions and immunity due to defective suppressive abilities of Treg cells in models
autoimmune inflammatory conditions has been generated. In this of diabetes and colitis. In this system, Treg cells progressively lose
model, a small proportion of apparently stable Treg cells lost Foxp3 Foxp3 expression and gain IFN-γ​secretion, although they maintain
expression and acquired an effector-memory phenotype with dif- their specific TSDR-demethylation pattern59. Mice with Treg cell–
ferent levels of secretion of the pro-inflammatory cytokines IFN-γ​ specific deletion of the transcription factor Helios develop systemic
and IL-1767. These ‘ex-Foxp3 cells’ were able to induce autoimmu- autoimmune pathology characterized by increased germinal-center
nity in an adoptive transfer model on the NOD background and formation, lymphocytic infiltration into non-lymphoid organs and
consisted of a mixed population, on the basis of their level of TSDR glomerulonephritis82. Although Helios does not form protein com-
demethylation, which would suggest that not all ex-Foxp3 cells in plexes with Foxp3 or bind to the Foxp3 locus82, Helios-deficient Treg
this model were once de facto tTreg cells. Subsequent data have sug- cells have increased expression of IFN-γ​and IL-17 and are unstable,
gested that this fate-mapping mouse model was detecting a propor- with decreased expression of Foxp3 and a tendency to completely
tion of Foxp3+ tTreg cells that were either transiently upregulating lose Foxp3 expression82,83. Treg cells deficient in another transcrip-
Foxp3 or were not fully committed to the tTreg cell lineage73. Further tion factor, Eos, exhibit increased expression of IL-2 and IFN-γ​
data in support of the proposal of tTreg cell instability have shown along with reduced suppressive ability, while forced overexpression
that Foxp3 expression is lost in tTreg cells specific for an epitope of of Eos in Treg cells prevents Treg cell instability, even in inflamma-
myelin oligodendrocyte glycoprotein (MOG; amino acids 38–49) tory environments. EosloFoxp3+ tTreg cells are detectable in vivo and
during the development of experimental autoimmune encephalitis have regulatory function, with the ability to acquire helper T cell–
(EAE), with an increased frequency of ex-Foxp3 cells in the central like effector characteristics while maintaining Foxp3 expression;

668 Nature Immunology | VOL 19 | JULY 2018 | 665–673 | www.nature.com/natureimmunology

© 2018 Nature America Inc., part of Springer Nature. All rights reserved.
FOCUS | Review
| Article
NaTuRe Immunology FOCUShttps://doi.org/10.1038/s41590-018-0120-4
Review Article
however, such cells exhibit specific changes in their global DNA- scurfy mice94, with Treg cells displaying a TH1-like Treg cell phenotype
methylation pattern84. and being unable to prevent disease in a colitis model. IFN-γ​ seems
to be involved in the defective function of Treg cells, as Foxo1–/–Ifng–/–
Treg cell plasticity mice partially recover from the wasting syndrome associated with
Plasticity is a property inherent to most if not all immune cells, this model94.
which allows them to adapt their phenotype and function to the Transcriptomic analysis of IFN-γ​+ TH1-like Treg cells at the popu-
changing environment and extracellular ‘danger’ signals. Thus, lation level91 or single-cell level90 has demonstrated that they exhibit
it is not surprising that Treg cells possess some degree of plasticity. lower expression of genes encoding immunosuppressive molecules
Treg cells have the ability to acquire features specific to the type of than that of Treg cells and have altered expression of costimulatory
immune response they control, which is driven mostly by ‘mas- molecules, migratory properties and specific signaling pathways. It
ter’ transcription factors and is regulated by environmental sig- is unknown whether TH1-like Treg cells have a role in disease patho-
nals. Thus, Treg cells acquire expression of the transcription factor genesis and/or protection in specific tissues. TH1-like Treg cells have
T-bet to restrain type 1 inflammation during infection85,86, and they been observed among MOG-specific Treg cells infiltrating the cen-
utilize the transcription factors IRF4 and STAT3 to inhibit TH2 tral nervous system of mice during EAE development. These Treg
cell responses87 and TH17 cell responses88, respectively. While this cells are not able to suppress the infiltration of the central nervous
modality of plasticity seems to be advantageous for the host and system by MOG-specific effector T cells or prevent disease onset;
beneficial to the outcome of the immune response, aberrant plastic- they secrete IFN-γ​at the onset and peak of disease but at a reduced
ity of Treg cells is also observed in several autoimmune diseases, with frequency during the recovery phase102. In contrast, T-bet+Foxp3+
Treg cells expressing pro-inflammatory cytokines, acquiring helper cells in pancreatic tissue seem to be protective in a mouse model of
T cell–like phenotypes and displaying diminished function in most type 1 diabetes103. Finally, while most work on TH1-like Treg cells has
cases but maintaining Foxp3 expression21,83,89–95. Paradoxically, these suggested that IL-12 and/or type 1 cytokines induce the phenotype,
helper T cell–like Treg cells utilize the same transcription factors used the fact that PI(3)K–Akt, a major pathway that integrates diverse
by Treg cells to inhibit specific types of immune responses. Therefore, environmental signals into cell function, is involved in their genera-
the secretion of IFN-γ​by TH1-like Treg cells requires T-bet expres- tion suggests that other environmental cues might have the ability
sion21,91–93, while IL-6-driven TH17-like Treg cells require STAT3 for to induce Treg cell plasticity, as it has been observed, for example,
the secretion of IL-1772, and IL-4-driven TH2-like Treg cells upregu- with increased concentrations of dietary salt or NaCl104.
late IRF4 and Gata-395,96. In most cases, helper T cell–like Treg cells
have a demethylated TSDR in the Foxp3 locus even though they TH17-like Treg cells
share effector features, which suggests that their phenotype might A small proportion of human peripheral Treg cells produce IL-17 in
be reversible21,90,93. They display alterations in their epigenetic sig- healthy people and upregulate the gene encoding the transcription
nature characteristic of those of Treg cells9,32,34,97, which might be the factor RORγ​t (RORC) (TH17-like Treg cells) ex vivo105 while conserv-
underlying mechanism that allows the secretion of pro-inflamma- ing their suppressive ability105,106. Given the well-established devel-
tory cytokines. Current efforts are focused on understanding the opmental relationship between Treg cells and TH17 cells, it remains
signaling pathways that drive this plasticity in specific autoimmune to be determined whether TH17-like Treg cells are a transient stage in
diseases, to harness this flexibility to the treatment of human dis- the de-differentiation of tTreg cells into TH17 cells, as has been sug-
ease92 and the role of Treg cell plasticity in autoimmune disease– gested69,107. In support of that proposal, the conversion of Foxp3+ Treg
related tissues. cells into TH17 cells has a crucial role in the pathogenesis of autoim-
mune arthritis in a collagen-induced arthritis mouse model. This
TH1-like Treg cells conversion is driven by IL-1β​108 and IL-6, and Foxp3+IL-17+ Treg cells
Perhaps the best-characterized tTreg cell plasticity event is the acqui- are observed in the synovium of subjects with active rheumatoid
sition of TH1 cell–like features. In mouse models and patients with arthritis109. Moreover, the conversion of Treg cells into TH17 cells
autoimmune diseases, such as type 1 diabetes93, multiple sclero- has also been reported in the CD18hypo PL/J mouse model of
sis21,91, autoimmune hepatitis98 and Sjogren syndrome99, there is psoriasis108, and TH17-like Treg cells have been observed in skin tissue
an increased frequency of IFN-γ​+Foxp3+ tTreg cells in the periph- of psoriatic patients89.
ery that display lower suppressive ability than that of Treg cells from Perhaps the tissue in which TH17-like Treg cells have been best
healthy age-matched subjects. TH1-like Treg cells upregulate the tran- identified is the gastrointestinal tract. The lamina propria seems
scription factor T-bet and other TH1 cell markers, such as CCR5 and to be enriched for iTreg cells with high expression of the TH17 cell–
CXCR3. Furthermore, in the apolipoprotein E–deficient (Apoe–/–) defining transcription factor RORγ​t. These TH17-like Treg cells seem
mouse model of atherosclerosis, which shares various pathogenic to have a beneficial function, as their absence exacerbates pathogen-
similarities with autoimmune disorders, there is accumulation of esis in several models of mucosal autoimmunity110,111. Furthermore,
IFN-γ​-producing TH1-like Treg cells in the aorta that display altered studies have suggested that Foxp3+RORγ​t+ Treg cells control glomer-
suppressive ability in vitro and in vivo90. Studies of an in vitro model ulonephritis112, and a lack of TH17-like Treg cells results in increased
in which TH1-like Treg cells are generated through the use of IL-12 mortality and organ pathology associated with systemic lupus ery-
have demonstrated that TH1-like Treg cells possess an activated PI(3) thematosus113. In addition to the importance of IL-1β​and IL-6 in
K and Akt kinase pathway and the transcription factor FoxO, which promoting the secretion of IL-17 by Treg cells114,115, other environ-
is partly responsible for their secretion of IFN-γ​and decreased sup- mental factors, including indoleamine 2,3-dioxygenase116, ligation
pressive capacity91. Interestingly, Treg cells isolated from patients of the pattern-recognition receptor TLR2117 and certain infec-
with relapsing–remitting multiple sclerosis also display an activated tions118, have been shown to modulate the conversion of TH17-like
PI(3)K–Akt–FoxO pathway ex vivo, and their suppressive ability is Treg cells either indirectly or directly.
corrected by blockade of the PI(3)K pathway94. In vivo, activation of
PI(3)K–Akt by Treg cell–specific deletion of the PI(3)K phosphatase TH2-like Treg cells
PTEN elicits a type 1 autoimmune disorder, with Treg cells downreg- Patients with systemic sclerosis display an increased frequency
ulating their expression of CD25 and Foxp3 and displaying reduced of TH2-like Treg cells in the skin but not in the peripheral blood,
functionality100,101. Moreover, FoxO itself has been linked to the reg- characterized by the secretion of IL-4 and IL-13 and upregulation
ulation of Treg cell plasticity. Mice with Treg cell–specific deletion of of Gata-3 and IRF-4. Peripheral Treg cells from these patients have
FoxO succumb to a lethal autoimmunity similar to that observed in high expression of ST2, the receptor for the alarmin IL-33. The

Nature Immunology | VOL 19 | JULY 2018 | 665–673 | www.nature.com/natureimmunology 669


© 2018 Nature America Inc., part of Springer Nature. All rights reserved.
Review Article
|
Review Article FOCUS
https://doi.org/10.1038/s41590-018-0120-4 NaTuRe Immunology

skin shows enrichment for this cytokine, which suggests that it Box 2 | Treg cell–specific therapies for autoimmune diseases
might have a role in the reprogramming of Treg cells into a TH2-like
phenotype95. TH2-like Treg cells have also been observed in mutant One of the many therapeutic strategies being investigated in the
(Il4raF709) mice susceptible to allergy and in Treg cells from the autoimmunity field is the restoration of peripheral tolerance, and
peripheral blood of food-allergic patients. TH2-like Treg cells secrete one avenue that has been explored is the development of Treg cell–
IL-4 and/or IL-13 and upregulate the transcription factors IRF4 and based therapeutic strategies. Interesting results have been ob-
Gata-396. Moreover, mice with Treg cell–restricted deletion of the tained with immunotherapies based on the adoptive transfer of
ubiquitin ligase Itch show autoimmune features, and Treg cells are polyclonally expanded Treg cells into patients with autoimmune
not able to control type 2 inflammation. This defect is associated diseases such as type 1 diabetes, cutaneous lupus erythematosus
with the acquisition of a TH2-like phenotype by the Treg cells and or Crohn’s disease131–134. Furthermore, with the development of
with increased expression of Gata-3, activation of STAT6 and secre- technologies to engineer epitope-specific TCRs, such as chimeric
tion of IL-4119. antigen receptors135, future approaches will potentially achieve
Many questions remain to be answered about Treg cell plasticity. an optimal number of Treg cells with TCRs specific for disease-
Does Treg cell plasticity reflect initial heterogeneity of the Treg cell relevant antigens for adoptive transfer into patients with auto-
population, with only a specific subset of Treg cells being able to immunity in those cases in which the autoantigens are known.
acquire effector-like properties65? Do helper T cell–like Treg cells rep- Although it is still too early to know the long-term consequences
resent a transient stage on the path to becoming ex-Foxp3 cells? Can of these therapies, the first results are encouraging and hold
Treg cell plasticity be harnessed for the design of new therapeutic promise for the adoption of these technologies for other autoim-
strategies aimed at modulating Treg cell function in diverse disease mune diseases.
settings? What is the role of helper T cell–like Treg cells in autoim- Another potential therapeutic approach to the restoration of
mune tissues during disease development and progression? peripheral tolerance is the targeting of specific molecules and/or
pathways that are dysfunctional in the existing pool of Treg cells
Tissue-resident Treg cells in patients. For this approach, it will be important to determine
One of the main recent advances in the Treg cell field has been the the physiological relevance and molecular mechanisms driving
discovery that Treg cells populate specific tissues in the body during the plasticity and instability of Treg cells in the patients, as well as
physiological and stress conditions. The characterization of these to improve knowledge of how the dysfunction of tissue-specific
populations and their mechanisms of action will have important Treg cells affect the development of certain autoimmune diseases.
implications for understanding the development, maintenance and In this context, there are many questions still unresolved (as
resolution of autoimmunity in specific organs. Tissue-resident Treg discussed in the main text and Box 3) that will undoubtedly
cells actively perform non-immunological functions and work at improve the design of Treg cell–specific therapeutic options for
maintaining tissue homeostasis and wound repair, roles that could patients with autoimmune disease.
be important for tissue homeostasis in autoimmunity settings.
Studies have begun to define the specific phenotype of tissue-resi-
dent Treg cells in the muscles, skin, lungs, gastrointestinal tract, liver obesity-associated fat-tissue inflammation123. Given that obesity is
and adipose tissue, among other locations (reviewed in ref. 28). In an established risk factor in autoimmune disease, it will be impor-
most cases, these Treg cells seem to be of thymic origin120–122, with tant to define the features and functions of VAT Treg cells in the
considerable oligoclonality of their TCR repertoire, which indicates development of autoimmunity.
that particular antigens in the tissue might be responsible for the Similarly, lung and muscle Treg cells express amphiregulin, the
accumulation of Treg cells in specific niches120,122,123. They are char- ligand for epidermal growth factor receptor, to promote tissue
acterized by the expression of tissue-specific transcription factors repair. Muscle Treg cells accumulate in acutely injured skeletal mus-
and mediators that drive the function of other cells in that tissue, in cle in mouse models of muscle injury and muscular dystrophy, in
support of the notion that the microenvironment exerts important which they control muscle inflammation after injury and promote
effects on the phenotype of Treg cells in a given location. The molec- tissue repair by acting on immune and non-immune cells27,120. Data
ular mechanisms by which tissue-resident Treg cells acquire their in a zebrafish model further support the proposal of a dual role
tissue-specific program have only begun to be explored. Epigenetic for Treg cells in the immunoregulation and tissue regeneration
analysis of mouse Treg cells isolated from various tissues and com- observed in mice. In zebrafish, Treg cell–like cells126 rapidly migrate
pared with Treg cells from the lymph nodes has demonstrated that to damaged organs in models of spinal-cord, heart and retina regen-
tissue Treg cells undergo extensive epigenetic reprograming that is eration, and their function is dependent on the secretion of organ-
globally tissue specific, but that there is a common tissue Treg cell specific regenerative factors. Treg cell–specific ablation inhibits
population characterized by expression of the activation marker organ regeneration127.
KLRG1 and ST2124. Treg cells populate healthy skin and help maintain homeostasis.
The best-studied tissue-specific Treg cell population is the one In healthy people and rodents, skin Treg cells possess an effector–
that is resident in visceral adipose tissue (VAT). VAT Treg cells serve memory phenotype, with expression of Treg cell–specific surface
important roles in the defense against associated metabolic disor- markers and greater proliferative capacity ex vivo than that of their
ders. In mice, lean fat tissue is populated by T cells with a TH2 cell blood counterparts. They express IL-17 and IL-10 and display a
phenotype and shows enrichment for Treg cells that maintain the demethylated TSDR, which would suggest that they are tTreg cells122.
predominance of resident anti-inflammatory macrophages. Treg Interestingly, Treg cells reside in close apposition to hair follicles,
cells accumulate over time and acquire a TH2-like phenotype, with where they contribute to the activation of hair-follicle stem cells by
expression of Gata-3, BATF, IRF4, CCR4 and IL-10121, as well as of promoting their proliferation and differentiation through the Notch
PPARγ​, a transcription factor that controls adipocyte differentiation signaling pathway. Ablation of Treg cells completely abolishes the
and mediates the accumulation, phenotype and function of VAT Treg ability of the hair follicle to regenerate, which indicates that Treg cells
cells121,125. Treg cell–specific deletion of PPARγ​results in the specific are a key component of the skin stem-cell niche128.
loss of VAT Treg cells, which demonstrates the important role of this The maintenance of local tissue homeostasis is of particu-
transcription factor in the development and/or maintenance of lar importance in the intestinal mucosa, where the immune sys-
VAT Treg cells121. The frequency of VAT Treg cells diminishes with age tem must be able to effectively discriminate between pathogens
in obese mice, which suggests that they have a role in modulating and dietary factors or commensal flora. Accordingly, deregulated

670 Nature Immunology | VOL 19 | JULY 2018 | 665–673 | www.nature.com/natureimmunology

© 2018 Nature America Inc., part of Springer Nature. All rights reserved.
FOCUS | Review
| Article
NaTuRe Immunology FOCUShttps://doi.org/10.1038/s41590-018-0120-4
Review Article
Box 3 | Unanswered questions autoimmune pathologies but also in other diseases such as cancer.
As for tissue-resident Treg cells, although research has begun to elu-
• What markers can be used to distinguish tTreg cells from iTreg cidate the mechanisms and factors that control their function in tis-
cells? sues, many questions remain to be answered, mostly those related
• What are the molecular mechanisms and environmental to the mechanisms that tissue Treg cells employ to maintain tissue
triggers responsible for the generation of ex-Foxp3 cells in integrity in autoimmune diseases and the characterization of tissue-
autoimmunity? resident Treg cells in humans (Box 2). Answers to these questions
• What are the details of the molecular mechanisms and envi- will undoubtedly improve the design of Treg cell–specific therapeutic
ronmental cues that drive the development of TH1-, TH22- options for patients with autoimmune diseases.
and TH17-like Treg cell phenotypes in ‘plastic’ Treg cells in
specific autoimmune settings? Received: 4 December 2017; Accepted: 29 January 2018;
• What is the contribution of the Treg cell epigenetic signature Published online: 20 June 2018
to Treg cell instability and plasticity in autoimmunity?
• What are the epigenetic and phenotypic signatures of human References
Treg cells in specific tissues, and how do these signatures con- 1. Fontenot, J. D., Gavin, M. A. & Rudensky, A. Y. Foxp3 programs the
development and function of CD4+CD25+ regulatory T cells. Nat. Immunol.
tribute to disease pathology? 4, 330–336 (2003).
• Does Treg cell plasticity reflect the initial heterogeneity of the 2. Hori, S., Nomura, T. & Sakaguchi, S. Control of regulatory T cell
Treg cell population, with only a specific subset of Treg cells development by the transcription factor Foxp3. Science 299,
being able to acquire effector-like properties? 1057–1061 (2003).
3. Khattri, R., Cox, T., Yasayko, S. A. & Ramsdell, F. An essential role for
• What is the role of Treg cell plasticity and instability in auto-
Scurfin in CD4+CD25+ T regulatory cells. Nat. Immunol. 4,
immune tissues during the development, progression and 337–342 (2003).
resolution of disease? 4. Sakaguchi, S., Sakaguchi, N., Asano, M., Itoh, M. & Toda, M. Immunologic
• What are the mechanisms that Treg cells utilize to maintain self-tolerance maintained by activated T cells expressing IL-2 receptor
tissue integrity, and what is the involvement of tissue-specific alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance
causes various autoimmune diseases. J. Immunol. 155, 1151–1164 (1995).
Treg cells in tissue damage in autoimmunity? 5. Liu, W. et al. CD127 expression inversely correlates with FoxP3 and
suppressive function of human CD4+ T reg cells. J. Exp. Med. 203,
1701–1711 (2006).
6. Sakaguchi, S., Miyara, M., Costantino, C. M. & Hafler, D. A. FOXP3+
immune responses to luminal flora in genetically susceptible peo- regulatory T cells in the human immune system. Nat. Rev. Immunol. 10,
490–500 (2010).
ple are generally recognized as key factors in the pathogenesis of 7. Josefowicz, S. Z., Lu, L. F. & Rudensky, A. Y. Regulatory T cells:
inflammatory bowel disease. While the role of commensal flora in mechanisms of differentiation and function. Annu. Rev. Immunol. 30,
inducing Treg cells has been well characterized, the dynamics and 531–564 (2012).
characterization of Treg cells in the mucosal tissue has only begun 8. Kanamori, M., Nakatsukasa, H., Okada, M., Lu, Q. & Yoshimura, A.
to be appreciated. Thus, the lamina propria seems to show enrich- Induced regulatory T cells: their development, stability, and applications.
Trends Immunol. 37, 803–811 (2016).
ment for Treg cells with high expression of RORγ​t110,129, and although 9. Ohkura, N. et al. T cell receptor stimulation-induced epigenetic changes
most of these are not tTreg cells, they seem to have a positive function and Foxp3 expression are independent and complementary events required
in maintaining tissue homeostasis, and their absence exacerbates for Treg cell development. Immunity 37, 785–799 (2012).
pathogenesis in several models of mucosal autoimmunity. 10. Nishizuka, Y. & Sakakura, T. Thymus and reproduction: sex-linked
How do Treg cells maintain tissue integrity, and what is the dysgenesia of the gonad after neonatal thymectomy in mice. Science 166,
753–755 (1969).
involvement of tissue-specific Treg cells in autoimmune disease? 11. Penhale, W. J., Farmer, A., McKenna, R. P. & Irvine, W. J. Spontaneous
One could speculate that defects in specific tissue-resident Treg cells thyroiditis in thymectomized and irradiated Wistar rats. Clin. Exp.
would be involved in the development, maintenance or resolution Immunol. 15, 225–236 (1973).
of autoimmune disorders that affect that specific tissue; for example, 12. Sakaguchi, S., Takahashi, T. & Nishizuka, Y. Study on cellular events in
is there a specific role for muscle-resident Treg cells in the pathology postthymectomy autoimmune oophoritis in mice. I. Requirement of Lyt-1
effector cells for oocytes damage after adoptive transfer. J. Exp. Med. 156,
of myasthenia gravis, or for VAT Treg cells in autoimmune disorders 1565–1576 (1982).
in which obesity is an important risk factor, or for skin Treg cells in 13. Sakaguchi, S., Fukuma, K., Kuribayashi, K. & Masuda, T. Organ-specific
psoriasis? Finally, while most research performed so far has studied autoimmune diseases induced in mice by elimination of T cell subset. I.
mice, and although there are intrinsic difficulties with translating Evidence for the active participation of T cells in natural self-tolerance;
deficit of a T cell subset as a possible cause of autoimmune disease.
these observations to humans, it will be crucial to determine the
J. Exp. Med. 161, 72–87 (1985).
phenotype and function of tissue-specific Treg cells in healthy people 14. Baecher-Allan, C., Brown, J. A., Freeman, G. J. & Hafler, D. A.
and in patients with autoimmune diseases to understand the role CD4+CD25high regulatory cells in human peripheral blood. J. Immunol. 167,
of tissue-resident Treg cells in the development and maintenance of 1245–1253 (2001).
human autoimmunity. 15. Stephens, L. A., Mottet, C., Mason, D. & Powrie, F. Human CD4+CD25+
thymocytes and peripheral T cells have immune suppressive activity in
vitro. Eur. J. Immunol. 31, 1247–1254 (2001).
Conclusions 16. Gavin, M. A. et al. Foxp3-dependent programme of regulatory T-cell
Intense research in the Treg cell field has improved knowledge of differentiation. Nature 445, 771–775 (2007).
their biology and has provided evidence of the existence of instabil- 17. Miyara, M. et al. Human FoxP3+ regulatory T cells in systemic autoimmune
ity and plasticity within the Treg cell compartment. Accumulating diseases. Autoimmun. Rev. 10, 744–755 (2011).
18. Brusko, T. M., Wasserfall, C. H., Clare-Salzler, M. J., Schatz, D. A. &
data in recent years have shown the presence of ex-Foxp3 cells Atkinson, M. A. Functional defects and the influence of age on the
and helper T cell–like Treg cells in various autoimmune patholo- frequency of CD4+ CD25+ T-cells in type 1 diabetes. Diabetes 54,
gies. Current efforts are focused on determining the molecular 1407–1414 (2005).
mechanisms responsible for the induction of each of these func- 19. Haseda, F., Imagawa, A., Murase-Mishiba, Y., Terasaki, J. & Hanafusa, T.
tional states in the periphery and autoimmune organs, as well as the CD4+ CD45RA− FoxP3high activated regulatory T cells are functionally
impaired and related to residual insulin-secreting capacity in patients with
environmental triggers that drive them. The ultimate goal of these type 1 diabetes. Clin. Exp. Immunol. 173, 207–216 (2013).
studies is to harness the mechanisms that generate the instability 20. Lindley, S. et al. Defective suppressor function in CD4+CD25+ T-cells from
and plasticity of Treg cells to modulate Treg cell function not only in patients with type 1 diabetes. Diabetes 54, 92–99 (2005).

Nature Immunology | VOL 19 | JULY 2018 | 665–673 | www.nature.com/natureimmunology 671


© 2018 Nature America Inc., part of Springer Nature. All rights reserved.
Review Article
|
Review Article FOCUS
https://doi.org/10.1038/s41590-018-0120-4 NaTuRe Immunology
21. Dominguez-Villar, M., Baecher-Allan, C. M. & Hafler, D. A. Identification 50. Song, X. et al. Structural and biological features of FOXP3 dimerization
of T helper type 1-like, Foxp3+​regulatory T cells in human autoimmune relevant to regulatory T cell function. Cell Reports 1, 665–675 (2012).
disease. Nat. Med 17, 673–675 (2011). 51. Xiao, Y. et al. Dynamic interactions between TIP60 and p300 regulate
22. Viglietta, V., Baecher-Allan, C., Weiner, H. L. & Hafler, D. A. Loss of FOXP3 function through a structural switch defined by a single lysine on
functional suppression by CD4+CD25+ regulatory T cells in patients with TIP60. Cell Rep. 7, 1471–1480 (2014).
multiple sclerosis. J. Exp. Med. 199, 971–979 (2004). 52. van Loosdregt, J. et al. Stabilization of the transcription factor Foxp3 by the
23. Bonelli, M. et al. Quantitative and qualitative deficiencies of regulatory deubiquitinase USP7 increases Treg-cell-suppressive capacity. Immunity 39,
T cells in patients with systemic lupus erythematosus (SLE). Int. Immunol. 259–271 (2013).
20, 861–868 (2008). 53. Dang, E. V. et al. Control of TH17/Treg balance by hypoxia-inducible factor
24. Thiruppathi, M. et al. Functional defect in regulatory T cells in myasthenia 1. Cell 146, 772–784 (2011).
gravis. Ann. NY Acad. Sci. 1274, 68–76 (2012). 54. Rudra, D. et al. Transcription factor Foxp3 and its protein partners form
25. van Roon, J. A., Hartgring, S. A., van der Wurff-Jacobs, K. M., Bijlsma, J. a complex regulatory network. Nat. Immunol. 13, 1010–1019 (2012).
W. & Lafeber, F. P. Numbers of CD25+Foxp3+​T cells that lack the IL-7 55. Samstein, R. M. et al. Foxp3 exploits a pre-existent enhancer landscape for
receptor are increased intra-articularly and have impaired suppressive regulatory T cell lineage specification. Cell 151, 153–166 (2012).
function in RA patients. Rheumatology (Oxford) 49, 2084–2089 (2010). 56. Kwon, H. K., Chen, H. M., Mathis, D. & Benoist, C. Different molecular
26. Ohkura, N., Kitagawa, Y. & Sakaguchi, S. Development and maintenance of complexes that mediate transcriptional induction and repression by FoxP3.
Nat. Immunol. 18, 1238–1248 (2017).
regulatory T cells. Immunity 38, 414–423 (2013).
57. Tao, R. et al. Deacetylase inhibition promotes the generation and function
27. Arpaia, N. et al. A Distinct function of regulatory T cells in tissue
of regulatory T cells. Nat. Med. 13, 1299–1307 (2007).
protection. Cell 162, 1078–1089 (2015).
58. Bettini, M. L. et al. Loss of epigenetic modification driven by the Foxp3
28. Panduro, M., Benoist, C. & Mathis, D. Tissue Tregs. Annu. Rev. Immunol.
transcription factor leads to regulatory T cell insufficiency. Immunity 36,
34, 609–633 (2016).
717–730 (2012).
29. Brunkow, M. E. et al. Disruption of a new forkhead/winged-helix protein, 59. Zhang, Y. et al. GP96 is a GARP chaperone and controls regulatory T cell
scurfin, results in the fatal lymphoproliferative disorder of the scurfy functions. J. Clin. Invest. 125, 859–869 (2015).
mouse. Nat. Genet. 27, 68–73 (2001). 60. Bin Dhuban, K. et al. Suppression by human FOXP3+ regulatory T cells
30. Bennett, C. L. et al. The immune dysregulation, polyendocrinopathy, requires FOXP3-TIP60 interactions. Sci. Immunol. 2, eaai9297 (2017).
enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. 61. Hayatsu, N. et al. Analyses of a mutant Foxp3 allele reveal BATF as a
Nat. Genet. 27, 20–21 (2001). critical transcription factor in the differentiation and accumulation of tissue
31. Wildin, R. S. et al. X-linked neonatal diabetes mellitus, enteropathy and regulatory T cells. Immunity 47, 268–283 e269 (2017).
endocrinopathy syndrome is the human equivalent of mouse scurfy. Nat. 62. Darce, J. et al. An N-terminal mutation of the Foxp3 transcription factor
Genet. 27, 18–20 (2001). alleviates arthritis but exacerbates diabetes. Immunity 36, 731–741 (2012).
32. Hill, J. A. et al. Foxp3 transcription-factor-dependent and -independent 63. Wan, Y. Y. & Flavell, R. A. Regulatory T-cell functions are subverted and
regulation of the regulatory T cell transcriptional signature. Immunity 27, converted owing to attenuated Foxp3 expression. Nature 445,
786–800 (2007). 766–770 (2007).
33. Morikawa, H. & Sakaguchi, S. Genetic and epigenetic basis of Treg cell 64. Williams, L. M. & Rudensky, A. Y. Maintenance of the Foxp3-dependent
development and function: from a FoxP3-centered view to an epigenome- developmental program in mature regulatory T cells requires continued
defined view of natural Treg cells. Immunol. Rev. 259, 192–205 (2014). expression of Foxp3. Nat. Immunol. 8, 277–284 (2007).
34. Huehn, J. & Beyer, M. Epigenetic and transcriptional control of Foxp3+ 65. Komatsu, N. et al. Heterogeneity of natural Foxp3+ T cells: a committed
regulatory T cells. Semin. Immunol. 27, 10–18 (2015). regulatory T-cell lineage and an uncommitted minor population retaining
35. Zheng, Y. et al. Role of conserved non-coding DNA elements in the Foxp3 plasticity. Proc. Natl Acad. Sci. USA 106, 1903–1908 (2009).
gene in regulatory T-cell fate. Nature 463, 808–812 (2010). 66. Rubtsov, Y. P. et al. Stability of the regulatory T cell lineage in vivo. Science
36. Feng, Y. et al. Control of the inheritance of regulatory T cell identity by a 329, 1667–1671 (2010).
cis element in the Foxp3 locus. Cell 158, 749–763 (2014). 67. Zhou, X. et al. Instability of the transcription factor Foxp3 leads to the
37. Li, X., Liang, Y., LeBlanc, M., Benner, C. & Zheng, Y. Function of generation of pathogenic memory T cells in vivo. Nat. Immunol. 10,
a Foxp3 cis-element in protecting regulatory T cell identity. Cell 158, 1000–1007 (2009).
734–748 (2014). 68. Hoffmann, P. et al. Loss of FOXP3 expression in natural human
38. Baron, U. et al. DNA demethylation in the human FOXP3 locus CD4+CD25+ regulatory T cells upon repetitive in vitro stimulation.
discriminates regulatory T cells from activated FOXP3(+​) conventional Eur. J. Immunol. 39, 1088–1097 (2009).
T cells. Eur. J. Immunol. 37, 2378–2389 (2007). 69. Koenen, H. J. et al. Human CD25highFoxp3pos regulatory T cells differentiate
39. Floess, S. et al. Epigenetic control of the foxp3 locus in regulatory T cells. into IL-17-producing cells. Blood 112, 2340–2352 (2008).
PLoS Biol. 5, e38 (2007). 70. Duarte, J. H., Zelenay, S., Bergman, M. L., Martins, A. C. & Demengeot, J.
40. van Loosdregt, J. et al. Regulation of Treg functionality by acetylation- Natural Treg cells spontaneously differentiate into pathogenic helper cells in
mediated Foxp3 protein stabilization. Blood 115, 965–974 (2010). lymphopenic conditions. Eur. J. Immunol. 39, 948–955 (2009).
41. Li, B. et al. FOXP3 interactions with histone acetyltransferase and class II 71. Oldenhove, G. et al. Decrease of Foxp3+​Treg cell number and acquisition
of effector cell phenotype during lethal infection. Immunity 31,
histone deacetylases are required for repression. Proc. Natl Acad. Sci. USA
772–786 (2009).
104, 4571–4576 (2007).
72. Laurence, A. et al. STAT3 transcription factor promotes instability of nTreg
42. Beier, U. H., Akimova, T., Liu, Y., Wang, L. & Hancock, W. W. Histone/
cells and limits generation of iTreg cells during acute murine graft-versus-
protein deacetylases control Foxp3 expression and the heat shock response
host disease. Immunity 37, 209–222 (2012).
of T-regulatory cells. Curr. Opin. Immunol. 23, 670–678 (2011).
73. Zhang, Z. et al. Activation and functional specialization of regulatory
43. Deng, G. et al. Pim-2 kinase influences regulatory T cell function and T cells lead to the generation of Foxp3 instability. J. Immunol. 198,
stability by mediating Foxp3 protein N-terminal phosphorylation. 2612–2625 (2017).
J. Biol. Chem. 290, 20211–20220 (2015). 74. Bailey-Bucktrout, S. L. et al. Self-antigen-driven activation induces
44. Li, Z. et al. PIM1 kinase phosphorylates the human transcription factor instability of regulatory T cells during an inflammatory autoimmune
FOXP3 at serine 422 to negatively regulate its activity under inflammation. response. Immunity 39, 949–962 (2013).
J. Biol. Chem. 289, 26872–26881 (2014). 75. Setoguchi, R., Hori, S., Takahashi, T. & Sakaguchi, S. Homeostatic
45. Morawski, P. A., Mehra, P., Chen, C., Bhatti, T. & Wells, A. D. Foxp3 maintenance of natural Foxp3+ CD25+ CD4+ regulatory T cells by
protein stability is regulated by cyclin-dependent kinase 2. J. Biol. Chem. interleukin (IL)-2 and induction of autoimmune disease by IL-2
288, 24494–24502 (2013). neutralization. J. Exp. Med. 201, 723–735 (2005).
46. Nie, H. et al. Phosphorylation of FOXP3 controls regulatory T cell 76. Tang, Q. et al. Central role of defective interleukin-2 production in the
function and is inhibited by TNF-α​in rheumatoid arthritis. Nat. Med 19, triggering of islet autoimmune destruction. Immunity 28,
322–328 (2013). 687–697 (2008).
47. Barbi, J., Pardoll, D. M. & Pan, F. Ubiquitin-dependent regulation of Foxp3 77. Balandina, A., Lécart, S., Dartevelle, P., Saoudi, A. & Berrih-Aknin, S.
and Treg function. Immunol. Rev. 266, 27–45 (2015). Functional defect of regulatory CD4+CD25+ T cells in the thymus of
48. Chen, Z. et al. The ubiquitin ligase Stub1 negatively modulates regulatory patients with autoimmune myasthenia gravis. Blood 105, 735–741 (2005).
T cell suppressive activity by promoting degradation of the transcription 78. Huan, J. et al. Decreased FOXP3 levels in multiple sclerosis patients.
factor Foxp3. Immunity 39, 272–285 (2013). J. Neurosci. Res. 81, 45–52 (2005).
49. Wang, L. et al. Ubiquitin-specific protease-7 inhibition impairs Tip60- 79. Long, S. A. et al. Defects in IL-2R signaling contribute to diminished
dependent Foxp3+ T-regulatory cell function and promotes antitumor maintenance of FOXP3 expression in CD4+CD25+ regulatory T-cells of type 1
immunity. EBioMedicine 13, 99–112 (2016). diabetic subjects. Diabetes 59, 407–415 (2010).

672 Nature Immunology | VOL 19 | JULY 2018 | 665–673 | www.nature.com/natureimmunology

© 2018 Nature America Inc., part of Springer Nature. All rights reserved.
FOCUS | Review
| Article
NaTuRe Immunology FOCUShttps://doi.org/10.1038/s41590-018-0120-4
Review Article
80. Zhang, B., Zhang, X., Tang, F., Zhu, L. & Liu, Y. Reduction of forkhead box 110. Sefik, E. et al. Mucosal immunology. Individual intestinal symbionts
P3 levels in CD4+CD25high T cells in patients with new-onset systemic lupus induce a distinct population of RORγ​+ regulatory T cells. Science 349,
erythematosus. Clin. Exp. Immunol. 153, 182–187 (2008). 993–997 (2015).
81. Moes, N. et al. Reduced expression of FOXP3 and regulatory T-cell 111. Yang, B. H. et al. Foxp3+ T cells expressing RORγ​t represent a stable
function in severe forms of early-onset autoimmune enteropathy. regulatory T-cell effector lineage with enhanced suppressive capacity during
Gastroenterology 139, 770–778 (2010). intestinal inflammation. Mucosal Immunol 9, 444–457 (2016).
82. Kim, H. J. et al. Stable inhibitory activity of regulatory T cells requires the 112. Kluger, M. A. et al. Stat3 programs Th17-specific regulatory T cells to
transcription factor Helios. Science 350, 334–339 (2015). control GN. J. Am. Soc. Nephrol. 25, 1291–1302 (2014).
83. Nakagawa, H. et al. Instability of Helios-deficient Tregs is associated with 113. Kluger, M. A. et al. Treg17 cells are programmed by Stat3 to suppress Th17
conversion to a T-effector phenotype and enhanced antitumor immunity. responses in systemic lupus. Kidney Int. 89, 158–166 (2016).
Proc. Natl Acad. Sci. USA 113, 6248–6253 (2016). 114. Kitani, A. & Xu, L. Regulatory T cells and the induction of IL-17.
84. Sharma, M. D. et al. An inherently bifunctional subset of Foxp3+ T helper Mucosal Immunol 1, S43–S46 (2008).
cells is controlled by the transcription factor eos. Immunity 38, 115. Xu, L., Kitani, A., Fuss, I. & Strober, W. Cutting edge: regulatory T cells
998–1012 (2013). induce CD4+CD25–Foxp3– T cells or are self-induced to become Th17 cells
85. Koch, M. A. et al. The transcription factor T-bet controls regulatory T cell in the absence of exogenous TGF-β​. J. Immunol. 178, 6725–6729 (2007).
homeostasis and function during type 1 inflammation. Nat. Immunol. 10, 116. Sharma, M. D. et al. Indoleamine 2,3-dioxygenase controls conversion of
595–602 (2009). Foxp3+ Tregs to TH17-like cells in tumor-draining lymph nodes. Blood 113,
86. Levine, A. G. et al. Stability and function of regulatory T cells expressing 6102–6111 (2009).
the transcription factor T-bet. Nature 546, 421–425 (2017). 117. Nyirenda, M. H. et al. TLR2 stimulation drives human naive and effector
87. Zheng, Y. et al. Regulatory T-cell suppressor program co-opts transcription regulatory T cells into a Th17-like phenotype with reduced suppressive
factor IRF4 to control TH2 responses. Nature 458, 351–356 (2009). function. J. Immunol. 187, 2278–2290 (2011).
88. Chaudhry, A. et al. CD4+ regulatory T cells control TH17 responses in a 118. Smith, A. A. et al. Characterization of Th17-like Tregs during late stages of
Stat3-dependent manner. Science 326, 986–991 (2009). infection with B. pertussis in mice. Possible immunomodulation by type I
89. Bovenschen, H. J. et al. Foxp3+​regulatory T cells of psoriasis patients interferon. J. Immunol. 196, 196.8 (2016).
easily differentiate into IL-17A-producing cells and are found in lesional 119. Jin, H. S., Park, Y., Elly, C. & Liu, Y. C. Itch expression by Treg cells
skin. J. Invest. Dermatol. 131, 1853–1860 (2011). controls Th2 inflammatory responses. J. Clin. Invest. 123, 4923–4934 (2013).
90. Butcher, M. J. et al. Atherosclerosis-driven Treg plasticity results in 120. Burzyn, D. et al. A special population of regulatory T cells potentiates
formation of a dysfunctional subset of plastic IFNγ​+ Th1/Tregs. Circ. Res. muscle repair. Cell 155, 1282–1295 (2013).
119, 1190–1203 (2016). 121. Cipolletta, D. et al. PPAR-γ​is a major driver of the accumulation and
91. Kitz, A. et al. AKT isoforms modulate Th1-like Treg generation and phenotype of adipose tissue Treg cells. Nature 486, 549–553 (2012).
function in human autoimmune disease. EMBO Rep. 17, 1169–1183 (2016). 122. Sanchez Rodriguez, R. et al. Memory regulatory T cells reside in human
92. Kitz, A. & Dominguez-Villar, M. Molecular mechanisms underlying skin. J. Clin. Invest. 124, 1027–1036 (2014).
Th1-like Treg generation and function. Cell. Mol. Life Sci. 74, 123. Feuerer, M. et al. Lean, but not obese, fat is enriched for a unique
4059–4075 (2017). population of regulatory T cells that affect metabolic parameters. Nat. Med.
93. McClymont, S. A. et al. Plasticity of human regulatory T cells in healthy 15, 930–939 (2009).
subjects and patients with type 1 diabetes. J. Immunol. 186, 124. Delacher, M. et al. Genome-wide DNA-methylation landscape defines
3918–3926 (2011). specialization of regulatory T cells in tissues. Nat. Immunol. 18,
94. Ouyang, W. et al. Novel Foxo1-dependent transcriptional programs control 1160–1172 (2017).
Treg cell function. Nature 491, 554–559 (2012). 125. Hamaguchi, M. & Sakaguchi, S. Regulatory T cells expressing PPAR-γ​
95. MacDonald, K. G. et al. Regulatory T cells produce profibrotic cytokines in control inflammation in obesity. Cell Metab. 16, 4–6 (2012).
the skin of patients with systemic sclerosis. J. Allergy Clin. Immunol. 135, 126. Kasheta, M. et al. Identification and characterization of Treg-like cells in
e9 946–e949 (2015). zebrafish. J. Exp. Med. 214, 3519–3530 (2017).
96. Noval Rivas, M. et al. Regulatory T cell reprogramming toward a 127. Hui, S. P. et al. Zebrafish regulatory T cells mediate organ-specific
Th2-cell-like lineage impairs oral tolerance and promotes food allergy. regenerative programs. Dev. Cell 43, 659–672 (2017).
Immunity 42, 512–523 (2015). 128. Ali, N. et al. Regulatory T cells in skin facilitate epithelial stem cell
97. Wei, G. et al. Global mapping of H3K4me3 and H3K27me3 reveals differentiation. Cell 169, 1119–1129 (2017).
specificity and plasticity in lineage fate determination of differentiating 129. Ohnmacht, C. et al. Mucosal immunology. The microbiota regulates type 2
CD4+ T cells. Immunity 30, 155–167 (2009). immunity through RORγ​t+ T cells. Science 349, 989–993 (2015).
98. Arterbery, A. S. et al. Production of proinflammatory cytokines by 130. Yu, X., Huang, Q. & Petersen, F. History and milestones of mouse models
monocytes in liver-transplanted recipients with de novo autoimmune of autoimmune diseases. Curr. Pharm. Des. 21, 2308–2319 (2015).
hepatitis is enhanced and induces TH1-like regulatory T cells. J. Immunol. 131. Bluestone, J. A. et al. Type 1 diabetes immunotherapy using polyclonal
196, 4040–4051 (2016). regulatory T cells. Sci. Transl. Med. 7, 315ra189 (2015).
99. Yamada, A. et al. Impaired expansion of regulatory T cells in a neonatal 132. Desreumaux, P. et al. Safety and efficacy of antigen-specific regulatory T-cell
thymectomy-induced autoimmune mouse model. Am. J. Pathol. 185, therapy for patients with refractory Crohn's disease. Gastroenterology 143,
2886–2897 (2015). 1207–1217 (2012).
100. Huynh, A. et al. Control of PI(3) kinase in Treg cells maintains homeostasis 133. Marek-Trzonkowska, N. et al. Therapy of type 1 diabetes with
and lineage stability. Nat. Immunol. 16, 188–196 (2015). CD4+CD25highCD127– regulatory T cells prolongs survival of pancreatic
101. Shrestha, S. et al. Treg cells require the phosphatase PTEN to restrain TH1 islets - results of one year follow-up. Clin. Immunol. 153,
and TFH cell responses. Nat. Immunol. 16, 178–187 (2015). 23–30 (2014).
102. Korn, T. et al. Myelin-specific regulatory T cells accumulate in the CNS but 134. Marek-Trzonkowska, N. et al. Administration of CD4+CD25highCD127–
fail to control autoimmune inflammation. Nat. Med 13, 423–431 (2007). regulatory T cells preserves β​-cell function in type 1 diabetes in children.
103. Tan, T. G., Mathis, D. & Benoist, C. Singular role for T-BET+CXCR3+ Diabetes Care 35, 1817–1820 (2012).
regulatory T cells in protection from autoimmune diabetes. Proc. Natl 135. Dawson, N. A. J. & Levings, M. K. Antigen-specific regulatory T cells: are
Acad. Sci. USA 113, 14103–14108 (2016). police CARs the answer? Transl. Res. 187, 53–58 (2017).
104. Hernandez, A. L. et al. Sodium chloride inhibits the suppressive function of
FOXP3+ regulatory T cells. J. Clin. Invest 125, 4212–4222 (2015). Acknowledgements
105. Ayyoub, M. et al. Human memory FOXP3+ Tregs secrete IL-17 ex vivo and We thank members of the Hafler and Dominguez-Villar laboratories for critical reading
constitutively express the TH17 lineage-specific transcription factor RORγ​t. of the manuscript.
Proc. Natl Acad. Sci. USA 106, 8635–8640 (2009).
106. Beriou, G. et al. IL-17-producing human peripheral regulatory T cells retain
suppressive function. Blood 113, 4240–4249 (2009). Competing interests
107. Radhakrishnan, S. et al. Reprogrammed FoxP3+ T regulatory cells become The authors declare no competing interests.
IL-17+ antigen-specific autoimmune effectors in vitro and in vivo.
J. Immunol. 181, 3137–3147 (2008). Additional information
108. Singh, K. et al. Reduced CD18 levels drive regulatory T cell conversion into Reprints and permissions information is available at www.nature.com/reprints.
Th17 cells in the CD18hypo PL/J mouse model of psoriasis. J. Immunol.
190, 2544–2553 (2013). Correspondence should be addressed to M.D. or D.A.H.
109. Komatsu, N. et al. Pathogenic conversion of Foxp3+ T cells into TH17 cells Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in
in autoimmune arthritis. Nat. Med. 20, 62–68 (2014). published maps and institutional affiliations.

Nature Immunology | VOL 19 | JULY 2018 | 665–673 | www.nature.com/natureimmunology 673


© 2018 Nature America Inc., part of Springer Nature. All rights reserved.

You might also like