You are on page 1of 14

Review

Empowering Regulatory
T Cells in Autoimmunity
Isaac R. Kasper,1,* Sokratis A. Apostolidis,1 Amir Sharabi,1,2
and George C. Tsokos1,*
Regulatory T cells (Tregs) are capable of dampening immune-mediated inflam-
Trends
mation and avert the destructive effects of uncontrolled inflammation. Distinct
After differentiation in either the thymus
molecules and pathways, including various transcription factors, phosphatases, or at peripheral sites, Tregs are capable
and kinases, impact the ability of Tregs to function as negative regulators of the of losing forkhead box P3 (FoxP3)
expression and producing proinflam-
immune response, and are presumably amenable to therapeutic manipulation. matory cytokines that potentially contri-
Here, we discuss recently identified molecular networks and the therapeutic bute to autoimmunity and inflammation.
potential for treating autoimmune diseases.
There are many epigenetic and tran-
scriptional programs driven by the T
Cellular Requirements of Tregs cell receptor and interleukin 2 to pro-
As early as the late 1960s, it was recognized that neonatal thymectomy of mice led to tect the expression of Foxp3 in an
inflammatory setting. Many new poten-
autoimmunity [1]. During the 1970s, interleukin 2 (IL-2) was discovered as an important driver
tial therapies to treat autoimmune or
of the immune response [2], yet, during the early 1990s, it was shown that IL-2 and IL-2 receptor inflammatory conditions are aimed at
(IL-2R) deficiency in mice led to autoimmune pathology [3,4], pointing to a paradox that needed maintaining epigenetic stability.
to be resolved. In 1995, Sakaguchi et al. showed that CD4+ CD25high T-cells were functionally
The PI3K-Akt-mammalian target of
Tregs (Box 1) [5]. From then on, Tregs received intense attention, both in terms of their ability to
rapamycin (mTOR) pathway is differen-
control inflammation and autoimmune responses, as well as to hamper the ability of the immune tially modulated in Tregs compared
system to control the growth of tumor cells [6] (Box 2). with effector T cells and represents a
promising therapeutic target to func-
tionally empower Tregs to suppress
Recent work has uncovered many key molecular networks required to develop and sustain Treg
inflammation.
function. Here, we discuss critical molecules, including the kinases/phosphatases mammalian
target of rapamycin (mTOR), protein phosphatase 2A (PP2A), and PTEN, and the transcription New and important insights have been
factors Foxo1, Bach2, and Helios, and the essential cellular processes of autophagy and obtained regarding the requirement for
phosphatases and kinases in integrat-
ubiquitination, and review signaling pathways that either promote or repress the Treg program ing cellular and metabolic signals within
(Box 3). Together, the diverse cellular requirements of Tregs present new therapeutic targets to Tregs during autoimmune diseases.
treat autoimmune diseases and cancer.

Tregs and Immunosuppression


Interest in Tregs was intensified after forkhead box P3 (FoxP3) was defined as a master
transcription factor linked to their function along with high expression of surface CD25. Neither
CD25 nor FoxP3 are perfect markers identifying Tregs because CD25 can be readily expressed 1
Division of Rheumatology, Beth Israel
in antigen or mitogen-stimulated T cells and effector T cells can express FoxP3 transiently in Deaconess Medical Center, Harvard
Medical School, Boston, MA 02215,
USA
Box 1. Thymically Derived and Peripherally Induced Tregs 2
Department of Clinical Immunology,
Tregs can develop in the thymus and peripheral tissues. Peripheral Tregs (iTregs) differentiate from naïve CD4+ T cells in Sackler Faculty of Medicine, Tel-Aviv
peripheral tissues following activation from antigen encounter in the presence of TGF-b, and in the absence of University, Tel-Aviv, 69978, Israel
inflammatory cytokines, such as IL-6, IFN-g, and IL-4 [111]. Thymically derived Tregs (nTregs) require T cell receptor
(TCR) signaling via self-peptide and MHC class II recognition [6]. Both of these distinct populations require stable
expression of Foxp3, because germline deletion of Foxp3 leads to Treg deficiency and lethal autoimmunity [10,11,112]. It *Correspondence:
appears that Foxp3 controls Treg lineage development through its expression in thymic epithelial cells, but does not ikasper@bidmc.harvard.edu
shape the T cell repertoire within the thymus [26,113]. Tregs also require IL-2 and TGF-b, which act to stabilize the (I.R. Kasper) and
expression of Foxp3 [114]. gtsokos@bidmc.harvard.edu
(G.C. Tsokos).

784 Trends in Molecular Medicine, September 2016, Vol. 22, No. 9 http://dx.doi.org/10.1016/j.molmed.2016.07.003
© 2016 Elsevier Ltd. All rights reserved.
Box 2. Treg Heterogeneity and Nonsuppressive Treg Functions Glossary
Treg Heterogeneity CNS (2 and 3): conserved
Tregs are a heterogeneous group of cells and, in addition to FoxP3, they express transcription factors associated with noncoding DNA sequences at the
different T helper (Th) subsets, which they specifically suppress. For instance, Tregs expressing Tbet, IRF4, and Stat3 foxp3 locus that stabilize Tregs by
dampen the Th1, Th2, and Th17 responses, respectively [115–117]. Interestingly, lack of GATA-3, which is a central either inducing (CNS3) or maintaining
transcription factor in Th2 cells, has been shown to reduce the expression of Foxp3 [118]. These GATA-3 deficient Tregs (CNS2) Foxp3 expression.
were found to acquire a Th17 phenotype, ultimately leading to the development of a spontaneous inflammatory disease in Conventional T cells: T
mice [118]. lymphocytes that express a TCR as
well as a CD4 or CD8 coreceptor.
Ex-Tregs: Tregs that have lost
Nonsuppressive Treg Functions
Foxp3 expression and their
There are also well-defined populations of tissue-resident Tregs vital to tissue homeostasis; Tregs that express suppressive function, and that
peroxisome proliferator-activated receptor gamma (PPARg) have been implicated in metabolic homeostasis in visceral produce proinflammatory cytokines.
adipose tissue [119]. Another study indicated that Tregs express amphiregulin and have been reported to contribute to IPEX: an X-linked familial
preventing severe lung injury after Influenza viral infection in mice. Notably, an absence of amphiregulin was not found to autoimmune syndrome caused by
affect the suppressor function of these Tregs [120]. mutations in the FOXP3 locus;
clinically characterized by immune
dysregulation, polyendocrinopathy,
and enteropathy.
Pyrazolopyrimidine derivatives:
Box 3. Autophagy compounds that promote Foxo1
Autophagy is a process targeting intracellular components for lysosomal degradation (e.g., proteins, etc.) and recycling nuclear translocation and
them while maintaining homeostasis during metabolic or infectious stress. The process has emerged as a critical simultaneously inhibit P13 K, thus
homeostatic mechanism in T cells, directing proliferation and differentiation. In Tregs, autophagy is more active compared diminishing the upstream and
with conventional T cells and the process supports their survival. When autophagy genes are selectively deleted, mice downstream effects of mTORC2
develop inflammatory disorders with a contracted Treg compartment [121,122]. Autophagy-deficient Tregs have signaling.
demonstrated increased apoptosis and decreased Foxp3 expression, as well as increased levels of mTORC1 activity Scurfy mice: mouse strain
and glycolysis [121,122]. One possibility is that autophagy degrades lipids to fatty acids, which can then fuel fatty acid developing a spontaneous fatal
oxidation, which is required for Treg metabolism. lymphoproliferative autoimmune
disease due to a missense mutation
within the foxp3 gene, resulting in a
lack of CD4+Foxp3+ Tregs.
humans and mice [7,8]. In 1959, a spontaneous systemic autoinflammatory phenotype in mice
was described and coined the ‘scurfy mouse’ (see Glossary) [9]. Subsequently, the Rudensky
[10] and Sakaguchi [11] labs reported that the scurfy mutation in male mice resided within the
foxp3 gene and, furthermore, that this phenotype was linked to Foxp3 deficiency. In humans, an
X-linked familial autoimmune syndrome of undetermined origin was also linked to FoxP3
deficiency [12]. The syndrome is characterized by immune dysregulation, polyendocrinopathy,
and enteropathy, and was given the acronym ‘IPEX’ [12,13]. Sequencing of the foxp3 gene in
many patients with IPEX revealed functionally detrimental mutations within the gene itself at the
30 untranslated region (UTR) [14]. These studies established the critical role of FoxP3 in the
disease.

Observations in scurfy mice and patients with IPEX demonstrated that Foxp3-expressing Tregs
had the capacity to negatively regulate the immune response. In their absence, an exaggerated
immune response ensued that eventually led to the development of autoimmune disease. Since
the initial observations in scurfy mice and patients with IPEX, aberrant Treg function has been
linked to many autoimmune conditions, such as connective tissue diseases (e.g., systemic lupus
erythematosus; SLE), endocrinopathies (e.g., Type 1 diabetes mellitus), inflammatory bowel
disease (e.g., Crohn's disease), and central nervous system autoimmune diseases (e.g., multiple
sclerosis; MS). In summary, the crucial role of Tregs in the regulation of the immune response has
been clearly established.

Differentiation of Tregs Requires TCR Engagement and Foxp3 Expression in


the Presence of IL-2 and TGF-b
Tregs express high levels of IL-2R/ (CD25), and their proper development and survival requires
the presence of IL-2 [15,16]. However, Tregs do not produce IL-2 [16] and, therefore, depend on
other sources for this cytokine. IL-2Rs, driven by the proximal lck promoter, have been shown to

Trends in Molecular Medicine, September 2016, Vol. 22, No. 9 785


restore the thymic generation of Tregs and to rescue IL-2R-deficient mice from lethal autoim-
munity [17]. IL-2 has been demonstrated to upregulate Foxp3 expression in thymic Treg cell
precursors. Additionally, overexpression of STAT5 (an IL-2-induced transcription factor) using
STAT5 transgenic mice dramatically increased the number of Tregs compared with wild-type
mice [18]. Therefore, it is reasonable to propose that increasing STAT5 by exogenous IL-2
represents a means to empower Tregs. Indeed, Tregs utilize the IL-2–IL-2R–Jak1/3–STAT5
pathway more efficiently than do conventional T cells, and low-dose IL-2 treatment has met
with clinical success for the treatment of graft-versus-host disease (GVHD), hepatitis C virus
(HCV)-related vasculitis and SLE (see below) [19,20].

Treg development also depends on TGF-b signaling, because TGF-b receptor deficiency has
been shown to impede the generation of Tregs [21]. This was evident in mouse studies in which
conditional deletion of TGF-b interrupted the development of thymically derived Tregs (nTregs) at
postnatal days 3–5 but not at postnatal day 7 (by which time nTregs had become highly sensitive
to the presence of IL-2) [21]. As Tregs develop in the thymus, they undergo positive and negative
selection following exposure to self-antigen and affinity of the T cell receptor (TCR) to these
antigens will determine T cell fate (reviewed in [22]). In this regard, TGF-b also facilitates nTreg
development by inhibiting apoptosis during negative selection in the thymus. Mechanistically,
this is due to TGF-b elevating the expression of the antiapoptotic gene BCL2. Indeed, mice
deficient in IL-2R and the TGF-b receptor fail to develop Tregs in the thymus [23].

The conserved noncoding DNA sequence (CNS) elements at the foxp3 locus have a role in the
stability of Tregs. Following T cell activation (e.g., after signals 1 and 2), CNS3 is accessed and
associated with c-Rel, and this binding is thought to enable the opening of the foxp3 locus [24].
Thymic and, to a lesser extent, peripheral CD4+Foxp3+ cells were significantly lost in mice
lacking CNS3 but without affecting the intensity of Foxp3 per cell [25]. Later, it was shown that
CNS3 might increase the probability of Foxp3 induction in response to TCR stimulation and
expand the TCR repertoire of Tregs [26].

Role of foxp3 in Maintaining Treg Lineage Stability


Foxp3 in Tregs
Mouse studies have shown that sustained expression of Foxp3 is an important factor in Treg
lineage stability. Foxp3 preserves the suppressive function of Tregs and opposes the transition
of Tregs into an effector phenotype. In cell fate-mapping experiments, most Tregs that assumed
a proinflammatory effector phenotype had lost Foxp3 expression [27–29]. Foxp3-negative ‘ex-
Tregs’ expressed low levels of CD25, GITR, and CTLA4, all produced IFN-g and IL-17 and, thus,
could not suppress conventional T cells in vitro [7,30]. These studies clearly demonstrate that,
without Foxp3, Tregs are more likely to lose their suppressive function and possibly assume an
inflammatory phenotype.

Phenotypic plasticity allows Tregs to adapt to the diverse and changing environmental cues
from various tissues. In the setting of inflammation or lymphopenia, the finding that these ‘ex-
Tregs’ promote inflammation provoked controversy and raised concerns regarding the validity
and safety of ongoing clinical trials that transfer Tregs adoptively to treat autoimmune
diseases [31].

TCR and IL-2 Signaling Stabilize Foxp3 Expression


There are several ways by which Tregs maintain Foxp3 expression and function. The intronic
enhancer at the Foxp3 locus, termed ‘CNS20 , has binding sites for the transcription factors NFAT
and STAT5. Binding of NFAT and STA5 results in CNS2 being the downstream target of both
TCR and IL-2 signaling (Figure 1) [32]. In fact, studies have shown that CNS2 is crucial for the
maintenance of Foxp3 expression in the presence of either proinflammatory cytokines or limited

786 Trends in Molecular Medicine, September 2016, Vol. 22, No. 9


TCR Nrp1–SEMA4a NOTCH
IL-2R
CD28

PI3K
STAT5 PTEN
P P
NFAT AKT

mTORC1 mTORC2

NF-κB

NFAT
P P
STAT5

FOXO1
FOXP3 RBPJ
CNS2
Proliferaon
NF-κB

Figure 1. Regulatory T Cell (Treg)-Specific Pathways Affecting Forkhead Box P3 (FoxP3) Expression. Murine
studies have revealed that T Cell Receptor (TCR) and interleukin (IL)-2 lead to NFAT and STAT5b binding to the CNS2 locus,
stabilizing Foxp3 expression. TCR engagement and IL-2 trigger PI3K–Akt–mammalian target of rapamycin (mTOR)
signaling, which is negatively regulated by PTEN. Negative regulation of the PI3K–Akt–mTOR axis stabilizes Foxp3
expression. Neuropilin 1–semaphorin 4A (Nrp1–SEMA4a) inhibits PI3 K and promotes PTEN, stabilizing Foxp3 expression.
Depicted are two pathways of Notch signaling; the noncanonical promotes mTORC2 signaling, tagging FOXO1 and
preventing nuclear translocation, thereby destabilizing Foxp3 expression; the canonical pathway facilitates NF-kB nuclear
translocation resulting in cellular proliferation.

levels of IL-2. This highlights an important mechanism by which Treg stability is maintained in the
setting of inflammation, when there is increased IL-2 signaling [33,34].

Epigenetic Traits that Maintain Foxp3 Expression


Foxp3 expression is maintained by multiple epigenetic traits, including DNA methylation and
histone modifications. The CNS2 site is hypomethylated in Tregs compared with conventional T
cells, where this region is fully methylated (closed conformation and transcriptionally inactive)
[35,36]. Importantly, one way in which IL-2 signaling enhances the transcription of Foxp3 is by
maintaining hypomethylation at CNS2 [35]. This hypomethylation physically depends on DNA
methyltransferase 1 (Dnmt1) and Tet methylcytosine dioxygenase 2 (Tet2) [37,38].

Conditional deletion of Dnmt1 has been shown to alter the expression of several hundred
proinflammatory and other genes in Tregs, exemplifying the importance of Dnmt1 for
maintenance of the core gene program underlying Treg development and function
[37,38]. IL-2, along with epigenetic changes in the foxp3 locus, was found to be connected
through Tet2, since downregulation of Tet2 destabilized the expression of Foxp3 in IL-2-
deficient Treg cells. In fact, Tet2 led to the hypomethylation and stability of Foxp3 regardless
of the presence of IL-2. These studies highlight how methylation of the Foxp3 locus can
determine cell fate [37,38].

Trends in Molecular Medicine, September 2016, Vol. 22, No. 9 787


Foxp3 binds to many histone-modifying proteins that promote epigenetic stability. This heter-
ogenous group includes the histone deacetylases (HDACs), p300, TIP60, and Enhancer of zeste
homolog 2 (Ezh2). Although some of these proteins interact directly with Foxp3, many of them
function to repress effector molecules that are upregulated during inflammation and capable of
destabilizing Tregs [39,40].

Moreover, Tregs have been shown to express elevated levels of the deubiquitinase USP7, which
is attached to Foxp3 in the nucleus. In a murine adoptive transfer-induced colitis model, it was
shown that the USP7–Foxp3 association resulted in a decrease in proteasome-mediated
degradation of Foxp3 [41]. By contrast, in a proinflammatory milieu, Foxp3 was reported to
be polyubiquitinated and degraded, thus resulting in elimination of Tregs. This process was
shown to occur in an Hsp70-dependent manner, following the interaction of Foxp3 with the E3
ubiquitin ligase Stub1 [42]. Cbl-b is another E3 ubiquitin ligase that may also affect the
development Tregs and, together with Stub1, leads to Foxp3 ubiquitination and degradation
in the proteasome in mice lacking CD28 [43]. Interestingly, treatment with a proteasome inhibitor
could overcome Foxp3 degradation in these mice and enable the proper thymic development of
Tregs [43].

The lineage stability and function of Tregs is enforced by sustained Foxp3 expression. Stable
Foxp3 expression is accomplished by multiple epigenetic and nonepigenetic mechanisms,
which together represent the coordinated genetic landscape required to sustain Treg function.

Known Molecular Requirements for Treg Sustenance and Function


Transcription Factors
Foxo1 (another member of the Forkhead box family) has been shown to be essential in
maintaining the suppressive function of mature Tregs. When Foxo1 is deleted specifically in
Tregs, mice develop a systemic inflammatory condition resembling the scurfy mouse. Foxo1
acts as a transcriptional enhancer during Treg development [44]. However, it can also prohibit
the production of IFN-g. In wild-type Tregs, Foxo1 binds directly to the Ifng locus, inhibiting IFN-g
production. Deletion of Foxo1 in Tregs allows production of aberrantly high levels of IFN-g, which
in turn impair Treg suppressor function [45]. Under inflammatory conditions, TCR signaling in all
T cells activates Akt, which exports Foxo1 from the nucleus [45]. Wild-type Tregs may have
diminished IFN-g production in an inflammation setting because they express relatively greater
amounts of Foxo1 than do conventional T cells [45]. Therefore, Foxo1 expression in Tregs may
protect these cells from assuming an inflammatory phenotype in a proinflammatory environment.

Bach2 is a transcription factor that regulates immune activation. In the context of CD4+ T cell
activation in the periphery, Bach2 has been shown to repress genes associated with the
polarization and differentiation of helper T cells and to simultaneously promote the peripheral
induction of Tregs [46]. Mice lacking Bach2 develop a wasting disease that is due to both a
severe reduction in Tregs and nonrepressed effector CD4+T cell differentiation [46]. Although the
stability of Foxp3 in existing Tregs and suppression of T cell proliferation were not shown to be
affected in the absence of Bach2, these Tregs were unable to suppress colitis in a lymphopenic
host upon Treg adoptive transfer [46]. In another study, mice harboring Bach2-deficient T cells
presented preferentially reduced survival of Helios-negative Tregs, associated with reduced Bcl-
2 and Mcl-1 levels, but elevated Bim:Bcl-2 ratios [47]. Due to these imbalances, these mice
presented with a fatal Th2-type chronic inflammatory lung disease.

Helios, a member of the Ikaros transcription factor family, was characterized as a marker of
thymically derived Tregs [48]. Recently, Helios was specifically deleted in Tregs by generation of
Foxp3cre/Heliosfl/fl mice [49,50]. These mice developed systemic autoimmune pathology char-
acterized by increased germinal center formation, lymphocytic infiltration into nonlymphoid

788 Trends in Molecular Medicine, September 2016, Vol. 22, No. 9


organs, and glomerulonephritis [49,50]. Helios-deficient Tregs lost their capacity to suppress
effector functions, which they gained instead. They exhibited diminished Foxp3 expression and
increased IFN-g and IL-17 production. Mechanistically, aberrant Treg function was linked to
decreased activation of the STAT5 signaling pathway [49]. Other groups have also reported that
the expression of Helios serves to enhance Treg functions [51]. Taken together, these studies
are indicative of the vital role of Helios in maintaining Treg stability and function.

Kinases and Phosphatases


mTOR is an intracellular serine threonine kinase that is central to T cell function. In T cells, mTOR
integrates cell signaling pathways and metabolism, which enables cell type-specific responses.
Upon T cell activation, TCR, CD28, and IL-2 activate PI3 K via receptor tyrosine kinases. PI3 K,
via AKT, promotes mTOR signaling. T effector cells and Tregs depend on mTOR signaling. In T
effector cells, increased mTOR signaling has been shown to enhance the inflammatory envi-
ronment [52]. By contrast, Tregs must restrain mTOR to preserve their suppressive function [52].

Increased mTOR activity is associated with proinflammatory effector function by skewing T cell
development towards the Th1 and Th17 lineages, while simultaneously contracting the Treg
population [53]. Indeed, when mTOR was deleted in CD4+ T cells, all conventional CD4+ T cells
assumed a Treg phenotype [54]. These phenotypic changes occurred even in the presence of
Th1, Th2, and Th17 polarizing environments in vitro. Despite these polarizing conditions, T cells
lacking mTOR expressed Foxp3 and TGF-b.

T cell metabolism is also regulated by mTOR in a cell lineage-specific manner. For instance,
when T effector cells are activated, they depend on glycolysis to support their nutrient needs.
When stimulated, mTOR upregulates genes associated with the pentose phosphate pathway
and the Glut 1 transporter, thereby facilitating glucose entry into cells and glycolysis [55,56]. By
contrast, to maintain functional capacity in Tregs, mTOR appears to be required not for
glycolysis but rather for lipogenesis [55,56].

Recently, it was established that the distinct multiprotein complex of mTOR mTORC1 was
essential for Treg function. When mTORC1 was conditionally knocked out in Tregs, mice
developed a severe lymphoproliferative autoimmune condition [57]. This was a perplexing
and unexpected result because the Akt–mTORC1 axis was known to antagonize de novo
Treg differentiation. Another unexpected result was that wild-type Tregs had higher basal levels
of mTORC1 activity compared with conventional T cells. Mechanistically, TCR and IL-2 signaling
increased mTORC1 activity in wild-type Tregs via the PI3k–Akt axis. Furthermore, this increase in
mTORC1 was necessary for Tregs to meet their metabolic demands by promoting cholesterol
and lipid metabolism, specifically the mevalonate pathway [57]. These findings illustrate how
mTORC1 integrates the TCR, IL-2, and lipogenic pathways that are essential for Treg function.
Although complete deletion of mTORC1 resulted in Treg dysfunction and autoimmunity, unre-
strained and highly active mTORC1 disrupted Treg homeostatic function [57].

PTEN is a protein phosphatase that negatively regulates PI3 K by blocking the conversion of
phosphatidylinositol to PIP3 [58]. It is highly expressed in Tregs, regulating their differentiation
[59]. It has been reported that the neuropilin 1–semaphorin 4A (Nrp1–SEMA4a) axis stabilizes
Treg function by restraining PI3 K and potentiating PTEN activity (Figure 1) [60]. In 2015, two
studies determined that Tregs absolutely required PTEN to maintain their suppressive function.
When PTEN was conditionally knocked out in Tregs, mice developed a systemic lymphopro-
liferative autoimmunity resembling SLE, with high titer antinuclear antibodies (ANAs) and immune
complex glomerular nephritis [61,62]. These mice presented increased Th1 and T follicular
helper (Tfh) cell responses, as well as overactive B cells with increased germinal center
formation. Functionally, PTEN-deficient Tregs lost CD25 and Foxp3 expression and their

Trends in Molecular Medicine, September 2016, Vol. 22, No. 9 789


metabolism was skewed towards glycolysis. Some of the PTEN-deficient Tregs even gained an
effector phenotype in the presence of high circulating levels of IFN-g [61,62].

Mechanistically, all of these changes were attributed to increased mTORC2 and Akt activity.
Normally, PTEN is the principal negative regulator of the PI3K–AKT axis [58]. These findings
demonstrated how PTEN can negatively regulate mTORC2, as well as maintain a commitment
to the Treg cell lineage and a suppressive function. Furthermore, previous studies showed that
mTORC2-driven pathways led to a Th2 phenotype [61,62], but that increased mTORC2 activity
also led to Th1 and Tfh, primarily by IFN-g [61,62].

Novel Treg-specific mechanisms relying on environmental and cytosolic cues to restrain


mTORC1 have recently been described, and one example is PP2A [63]. Indeed, PP2A signaling
was recently shown to be crucial for Treg function. The catalytic activity of PP2A in Tregs from
mice was reported to be increased to levels significantly higher than in conventional T cells [63].
In this study, mice deficient in PP2Ac Tregs developed spontaneously severe, progressive,
multiorgan autoimmunity characterized by wasting, dermatitis, scaly tails and ears, eyelid
crusting, and, in some instances, overt skin rash and ulcerations. Animals also suffered a
lymphoproliferative syndrome with secondary lymphoid organ enlargement. T cells were acti-
vated in all lymphoid organs and produced IL-17, while multiple autoantibodies were detected in
the serum. Activation of PP2A required the presence of ceramide, and Foxp3 expression led to
PP2A accumulation by inhibiting the gene sgms1, whose product is the ceramide-degrading
enzyme sphingomyelinase synthase 1 [63]. Of note, functional Tregs contain high amounts of
ceramides, and PP2Ac was demonstrated to inactivate mTORC1 without affecting the mTORC2
and PI(3)K/Akt pathways (Figure 2). Rapamycin (an mTORC1 inhibitor) treatment in vitro of
PP2A-deficient Treg cells reduced the production of IL-2 and IL17 and normalized their
increased glycolytic and oxidative phosphorylation rates to the levels of PP2A-suficient Treg
cells. Finally, in vivo administration of rapamycin to mice lacking PP2A in Tregs reduced
leukocyte infiltration in the affected peripheral tissues [63]. Again, this study highlights how
intracellular components, such as PP2A, provide differential signaling to promote Treg functional
homeostasis.

Repressors of Treg Function


Notch family receptors have been well characterized as directing the differentiation fates of
different T cell subsets. Notch signals through two different pathways. In the canonical pathway,
Notch receptors are cleaved, traffic to the nucleus, and interact with the recombination signal
binding protein RBPJK, ultimately leading to the expression of target genes. A second (nonca-
nonical) pathway involves mTORC2 and its associated adaptor protein Rictor. In contrast to
PTEN, Notch destabilizes Treg function by enhancing mTORC2 [64]. Notch overexpression in
Tregs has been reported to cause overactive canonical and noncanonical signals, resulting in
systemic autoimmunity in mice [64]. In this study, increased mTORC2 activity ultimately
decreased Foxp3 expression, because Foxo1 activity was prevented and could not provide
the epigenetic stability of Foxp3. Tregs overexpressing Notch also demonstrated plasticity
because they acquired a Th1 phenotype. Conversely, in a separate mouse strain where Notch
was deleted in Tregs, animals displayed overall decreased Th1 activity, and Tregs were less
prone to undergo apoptosis. Together these results demonstrate that restraining mTORC2 can
promote the functional homeostasis of Tregs [64].

From another perspective, special AT-rich sequence binding protein (SATB1) is a genome
organizer expressed in T cells that regulates chromatin structure and gene expression [65]. The
acquisition of a functional Treg phenotype was reported to require the repression of SATB1 [65].
Specifically, Foxp3 was shown to suppress the SATB1 locus and a failure to do so resulted in the
loss of suppressive function, the establishment of transcriptional effector T cell programs, and

790 Trends in Molecular Medicine, September 2016, Vol. 22, No. 9


TCR
CD28
DAG
SMS1 Ceramide

Ceramide (C)
Ceramide
SET
SET SET
SET SET
Ceramide
SET

(D) B (E)
B

A mTORC1
C
A

Acve PP2A
Inacve PP2A
SMS1

(B)
(A)

Sgms1 FOXP3

Figure 2. Protein Phosphatase 2A (PP2A) Is Required for Regulatory T Cell (Treg) Function. (A) Forkhead Box P3
(FoxP3) inhibits sgms1; (B) SMS1 protein is not produced, therefore ceramide is not degraded and accumulates in the
cytoplasm; (C) SET is upregulated upon T cell activation and ceramide binds SET, making it less available to inhibit PP2A. (D)
PP2A is activated when unbound from SET. (E) Active PP2A inhibits mTORC1 activity, thereby promoting Foxp3
expression. Abbreviations: DAG, diacyl glycerol; SET, originally named ‘I2PP2A’ for ‘inhibitor 2 of PP2A’; sgms1,
sphingomyelin synthase 1 encoding gene; SMS1, shingomyelin synthase – catalyzes the conversion of ceramide and
phosphatidylcholine to diacylglycerol and sphingomyelin.

the induction of effector T cell cytokines. Another set of experiments showed that conditional
knockout of SATB1 in the T cell compartment [accomplished through generation of lck-CRE/
SATB1(flox/flox) mice] resulted in a significant reduction in both CD4+CD8+ T cells in the
thymus and Tregs, leading to the development of autoimmune disease within 16 weeks after
birth [66].

Tregs in Human Autoimmune Diseases


Monogenic Diseases Associated with Loss of Treg Function
Tregs represent the fundamental T cell capable of promoting self-tolerance and balancing
excessive inflammation. Several monogenic conditions illustrate the importance of functioning
Tregs to human immune homeostasis. A prototype example is Foxp3 deficiency resulting in IPEX
syndrome in males. It is fatal within the first several years of life, unless rescued with bone marrow
transplantation. Patients with IPEX have elevated autoantibodies levels, suggesting significant
adaptive immune dysregulation [67–69].

Other monogenic conditions associated with Treg dysfunction include CD25 deficiency.
Humans with this deficiency have both autoimmunity and immunodeficiency, which clinically
resembles an IPEX-like phenotype. CD25 deficiency does not affect the number of Foxp3 + cells
in the circulation, but rather impairs their suppressive function by diminishing IL-10 production,
altering the metabolic fitness of Tregs, and losing the ‘IL-20 sink [70,71].

Trends in Molecular Medicine, September 2016, Vol. 22, No. 9 791


STAT5b deficiency also is implicated in the development of autoimmunity because STAT5b has
multiple effects that are essential to Treg function. It stabilizes the expression of FoxP3 by binding
the locus and it is required for the signal transduction of common gamma-chain cytokines, such
as IL-2. Clinically, STAT5b deficiency manifests as chronic diarrhea, eczema, and lymphoid
interstitial pneumonitis [72,73].

CTLA4 deficiency in mice leads to massive lymphoproliferation and multiorgan inflammation [74].
Humans with CTLA4 mutations display an autosomal dominant immune dysregulation syn-
drome characterized by hypogammaglobulinemia, recurrent infections, and multiple autoim-
mune clinical features. CTLA4 expression has been show to be decreased in Tregs,
compromising their function, although without decreasing their number [75]. Severe Treg cell
deficiency, with depressed FoxP3 expression, has been noted in patients who harbor a
nonsense mutation in the gene encoding LPS-responsive beige-like anchor (LRBA), known
to present as common variable immunodeficiency and autoimmunity [76]. LRBA colocalizes with
CTLA4 in endosomal vesicles, and LRBA deficiency or knockdown has been reported to lead to
increased CTLA4 turnover, resulting in reduced levels of CTLA4 protein in FoxP3+ Tregs and
activated conventional T cells [77]. Interestingly, CTLA4-Ig administration corrected cell abnor-
malities and led to clinical improvements [77].

Tregs in Systemic Autoimmune Diseases


Tregs have been implicated in the pathophysiology of systemic rheumatic diseases, although
their role is less defined than in monogenic traits. In SLE, there have been several subsets of
Tregs identified in the peripheral blood. However, Treg numbers and function have varied
considerably [78–81]. This likely reflects the heterogeneity of the disease. In human studies,
critical controls have been missed, such as documentation of the time of disease onset,
therapeutic interventions, and state of health (i.e., flares versus remission). Taking SLE as an
example, other factors have made the study of Tregs in SLE problematic. This has been due in
part to differences in experimental protocols, including the isolation of Tregs, flow cytometry
settings, and either the presence or absence of antigen-presenting cells (APCs) in ex vivo
functional assays [78]. Many discrepancies have also stemmed from a lack of a unique Treg
marker. Taking all of this into account, most reports on human SLE demonstrate aberrant Treg
function and abnormal total Treg numbers [78–81]. Furthermore, there are histological changes
in the inflamed kidney. In kidney biopsy samples from patients with active proliferative lupus
nephritis and antineutrophil cytoplasmic antibody-associated vasculitis, a study reported a
decrease in the number of Foxp3 + versus CD3 + cells, relative to patients with hypertensive
nephropathy and pure membranous lupus nephritis class 5 LN [82]. Changes in such cell
numbers might be due, in part, to the overall decrease in levels of circulating IL-2, which is
required for the maintenance of Treg function, but further studies are required to fully address the
nature of these differences (82].

Targeting Tregs to Control Autoimmune Diseases


A general therapeutic strategy to control autoimmune diseases might include an effort to
maintain the functional integrity of Tregs in the periphery (Table 1). It is reasonable to suppose
that this approach might protect the host from aberrant Tregs that react to self-antigen with
inflammatory signals and also potentially maintain Tregs induced in the peripheral tissue from the
pool of naïve CD4+ T cells.

Pharmacological blockade of the PI3K–AKT–mTOR axis is also regarded as having considerable


therapeutic potential. Studies have demonstrated the positive effects of restraining mTOR
signaling to promote Treg survival and stability [83,84]. Although first approved as an immuno-
suppressant in 1999, rapamycin was later found to expand the Treg population [84], and could
provide therapeutic value in patients lacking Foxp3 expression or function.

792 Trends in Molecular Medicine, September 2016, Vol. 22, No. 9


Table 1. Treg Associated Therapeutic Targets for Autoimmune Diseases
Therapeutic Target Drug Mechanism Reported Application Refs
and/or Potential Use

Cytokine signaling Aldesleukin STAT5 activator SLE [104]


(low-dose IL-2)
HCV-related vasculitis [102]

Graft-versus-host disease [103]

Kinase/phosphatase Rapamycin, mTORC1 inhibitor SLE, RA, JIA, SS [53]

Everolimus mTORC1 inhibitor RA, SLE [105,106]

KN-93 CaMK IV inhibitor SLE [90]

Fingolimod (FTY720) PP2A activator SLE, RA, MS [107–109]


0
Epigenetic Foxp3 5-aza-2 -deoxycytidine Dnmt1 inhibitor NR [99]
stabilizers
ACY-738 HDAC 6 inhibitor SLE [110]

Tubastatin A HDAC 6 inhibitor RA

Pyrazolopyrimidine FOXO1 Stabilizer NR


derivatives

a
Abbreviations: JIA, juvenile idiopathic arthritis; NR, not reported.

As outlined above, Foxp3 expression is compromised when mTORC2 is activated. This occurs
because mTORC2 prevents Foxo1 from entering the nucleus and stabilizing Foxp3 expression.
Therefore, drugs promoting nuclear translocation of Foxo1 represent another potential target, as
in the case of pyrazolopyrimidine derivatives. These compounds not only promote Foxo1
nuclear translocation, but also simultaneously inhibit P13 K, thus targeting the upstream and
downstream effects of mTORC2 signaling [85].

Other molecules limiting mTOR activity work by diverse mechanisms of action, but include S1P
receptor blockers [86,87]; antioxidants, such as metformin and N-acetyl cysteine [88,89]; or
calmodulin kinase II/IV inhibitors [90–94]. There are also ATP competitive inhibitors of the kinase
domain of mTOR, such as Torin 2 (reviewed in [53,95]). The rationale to treat autoimmune
conditions with mTOR inhibitors has been further supported by an ever-increasing number of
positive reports from studies in patients and animal models for SLE, rheumatoid arthritis (RA),
and Sjögren's syndrome (SS) [53]. For instance, a prospective clinical trial using rapamycin for
the treatment of SLE was completed in 2015 and, once collected and analyzed, the results might
provide further insight into the therapeutic efficacy of mTOR inhibitors [53].

While targeting mTORC2 provides epigenetic stability of Foxp3 expression via Foxo1 nuclear
translocation, many other epigenetic targets exist. These include, but again are not limited to,
forced CpG demethylation with the Dnmt1 inhibitor 5-aza-2-deoxycitidine (5-Aza), which has
been administered to subjects in conjunction with rapamycin inhibitors [96,97]. Other modalities
include targeting histone acetalization. With specific HDAC inhibitors (not pan-inhibitors) for
example, HDAC6 inhibitors have selectively enhanced Treg function in models of murine lupus,
protecting the mice from developing lupus nephritis [98,99].

IL 2, as outlined above, is essential to the differentiation and function of Tregs. Low-dose


IL-2 treatment of lupus-prone mice [90,100] as well as in a few patients with SLE [20,101],
was found to significantly modulate the severity of the disease. Interestingly, although not
involving autoimmune conditions, there have been two promising clinical trials where GVHD
and HCV have been successfully treated with IL-2. In both of these studies, the clinical
amelioration was associated with augmented numbers of peripheral Tregs and enhanced
suppressive function [102,103].

Trends in Molecular Medicine, September 2016, Vol. 22, No. 9 793


Box 4. The Clinician's Corner Outstanding Questions
Tregs are indispensable in controlling immune responses. Their absence is linked to autoimmunity and inflammation and Is there a role for ceramides in prevent-
their function is continuously being assessed in several systemic autoimmune conditions, such as SLE. ing autoimmunity, given that their con-
centration is increased in Tregs?
Administration of ex vivo expanded Tregs has been considered for the treatment of autoimmune diseases, but this is a
controversial and not yet established therapy. Can we further identify molecules that
can regulate cellular signaling and
Several molecules, including Foxo1, mTOR, PTEN, Notch, and PP2A, are involved in the maintenance of Treg function. directly impact the epigenetic land-
Drugs that deliberately increase or decrease the expression of molecules controlling Treg function, such as rapamycin, scape and function of Tregs?
might empower Treg function.
What other epigenetic strategies can
Patients who lack functioning molecules, such as Foxp3, IL-2R, STAT5B, or CTLA, harbor defective Tregs and develop be contemplated to stabilize Foxp3
severe autoimmune diseases or immunodeficiencies. expression and, therefore, putatively
prevent autoimmunity? Are these fea-
sible? Will they require combination
therapies?
From the studies reviewed above, there are many potential targets to be exploited that could
enhance Treg function in a cell-specific manner. These include augmenting the intracellular Other than rapamycin, could alternative
concentration of ceramides to diminish mTORC1 signaling. Other potential targets could include therapies be envisaged and used in
interfering with autophagy in cells, but this type of approach should be considered with caution combination to synergistically enhance
Treg function? Can these be clinically
because hydroxychloroquine (an autophagy inhibitor) is central to the treatment of SLE. applied to treat autoimmune diseases?

Concluding Remarks
Tregs are essential negative regulators of the immune response. Lack of functional Tregs can
lead to the development of systemic autoimmune diseases. Recently, several molecules that
enforce Treg function have been identified. These may represent novel therapeutic targets to
potentially treat certain autoimmune conditions (Box 4). Although low-dose IL-2 and rapamycin
have been used with significantly positive clinical outcomes for certain patients, further IL-2-
based trials should be conducted. In addition, many other potential therapeutic targets should
be further exploited. One example includes the activation of PP2A to increase the intracellular
levels of ceramide and restrain mTORC1 signaling (see Outstanding Questions). These mole-
cules could be used not only to stabilize Tregs, but also to enhance their suppressive function in
conditions such as juvenile idiopathic arthritis and MS, where T effector cells are resistant to Treg
suppression. It is clear that many studies targeting pathways in Tregs to alleviate autoimmune
diseases warrant further validation. Nevertheless, molecules that might empower Treg function
are likely to provide exciting new treatment modalities for various autoimmune conditions.

Acknowledgments
Funding was provided by NIH grant RO1 AI068787 (G.C.T.) I.R.K. was supported by NIH T32AI074549 training grant
(G.C.T.).

References
1. Nishizuka, Y. and Sakakura, T. (1969) Thymus and reproduction: 7. Miyao, T. et al. (2012) Plasticity of Foxp3(+) T cells reflects
sex-linked dysgenesia of the gonad after neonatal thymectomy in promiscuous Foxp3 expression in conventional T cells but not
mice. Science 166, 753–755 reprogramming of regulatory T cells. Immunity 36, 262–275
2. Malek, T.R. (2008) The biology of interleukin-2. Annu. Rev. 8. Allan, S.E. et al. (2007) Activation-induced FOXP3 in human T
Immunol. 26, 453–479 effector cells does not suppress proliferation or cytokine produc-
3. Suzuki, H. et al. (1995) Deregulated T cell activation and autoim- tion. Int. Immunol. 19, 345–354
munity in mice lacking interleukin-2 receptor beta. Science 268, 9. Russell, W.L. et al. (1959) Exceptional inheritance of a sex-linked
1472–1476 gene in the mouse explained on the basis that the X/O sex-
4. Willerford, D.M. et al. (1995) Interleukin-2 receptor alpha chain chromosome constitution is female. Proc. Natl. Acad. Sci. U.S.A.
regulates the size and content of the peripheral lymphoid com- 45, 554–560
partment. Immunity 3, 521–530 10. Fontenot, J.D. et al. (2003) Foxp3 programs the development
5. Sakaguchi, S. et al. (1995) Immunologic self-tolerance main- and function of CD4 + CD25 + regulatory T cells. Nat. Immunol.
tained by activated T cells expressing IL-2 receptor alpha- 4, 330–336
chains (CD25). Breakdown of a single mechanism of self- 11. Hori, S. et al. (2003) Control of regulatory T cell development by
tolerance causes various autoimmune diseases. J. Immunol. the transcription factor Foxp3. Science 299, 1057–1061
155, 1151–1164 12. Bennett, C.L. et al. (2001) The immune dysregulation, polyen-
6. Josefowicz, S.Z. et al. (2012) Regulatory T cells: mechanisms of docrinopathy, enteropathy, X-linked syndrome (IPEX) is caused
differentiation and function. Annu. Rev. Immunol. 30, 531–564 by mutations of FOXP3. Nat. Genet. 27, 20–21

794 Trends in Molecular Medicine, September 2016, Vol. 22, No. 9


13. Powell, B.R. et al. (1982) An X-linked syndrome of diarrhea, 38. Nair, V.S. and Oh, K.I. (2014) Down-regulation of Tet2 prevents
polyendocrinopathy, and fatal infection in infancy. J. Pediatr. TSDR demethylation in IL2 deficient regulatory T cells. Biochem.
100, 731–737 Biophys. Res. Commun. 450, 918–924
14. Wildin, R.S. et al. (2001) X-linked neonatal diabetes mellitus, 39. DuPage, M. et al. (2015) The chromatin-modifying enzyme Ezh2
enteropathy and endocrinopathy syndrome is the human equiv- is critical for the maintenance of regulatory T cell identity after
alent of mouse scurfy. Nat. Genet. 27, 18–20 activation. Immunity 42, 227–238
15. Fontenot, J.D. et al. (2005) A function for interleukin 2 in Foxp3- 40. Morikawa, H. et al. (2014) Differential roles of epigenetic changes
expressing regulatory T cells. Nat. Immunol. 6, 1142–1151 and Foxp3 expression in regulatory T cell-specific transcriptional
16. Barron, L. et al. (2010) Cutting edge: mechanisms of IL-2-depen- regulation. Proc. Natl. Acad. Sci. U.S.A. 111, 5289–5294
dent maintenance of functional regulatory T cells. J. Immunol. 41. van Loosdregt, J. et al. (2013) Stabilization of the transcription
185, 6426–6430 factor Foxp3 by the deubiquitinase USP7 increases Treg-cell-
17. Malek, T.R. et al. (2002) CD4 regulatory T cells prevent lethal suppressive capacity. Immunity 39, 259–271
autoimmunity in IL-2Rbeta-deficient mice. Implications for the 42. Chen, Z. et al. (2013) The ubiquitin ligase Stub1 negatively
nonredundant function of IL-2. Immunity 17, 167–178 modulates regulatory T cell suppressive activity by promoting
18. Burchill, M.A. et al. (2003) Distinct effects of STAT5 activation on degradation of the transcription factor Foxp3. Immunity 39,
CD4 + and CD8 + T cell homeostasis: development of CD4 272–285
+ CD25 + regulatory T cells versus CD8 + memory T cells. J. 43. Zhao, Y. et al. (2015) E3 ubiquitin ligase Cbl-b regulates thymic-
Immunol. 171, 5853–5864 derived CD4 + CD25 + regulatory T cell development by target-
19. Klatzmann, D. and Abbas, A.K. (2015) The promise of low-dose ing Foxp3 for ubiquitination. J. Immunol. 194, 1639–1645
interleukin-2 therapy for autoimmune and inflammatory diseases. 44. Samstein, R.M. et al. (2012) Foxp3 exploits a pre-existent
Nat. Rev. Immunol. 15, 283–294 enhancer landscape for regulatory T cell lineage specification.
20. von Spee-Mayer, C. et al. (2015) Low-dose interleukin-2 Cell 151, 153–166
selectively corrects regulatory T cell defects in patients with 45. Ouyang, W. et al. (2012) Novel Foxo1-dependent transcriptional
systemic lupus erythematosus. Ann. Rheum. Dis. Published programs control T(reg) cell function. Nature 491, 554–559
online August 31, 2015. http://dx.doi.org/10.1136/annr- 46. Roychoudhuri, R. et al. (2013) BACH2 represses effector pro-
heumdis-2015-207776 grams to stabilize T(reg)-mediated immune homeostasis. Nature
21. Liu, Y. et al. (2008) A critical function for TGF-[beta] signaling in 498, 506–510
the development of natural CD4 + CD25 + Foxp3 + regulatory T 47. Kim, E.H. et al. (2014) Bach2 regulates homeostasis of Foxp3
cells. Nat. Immunol. 9, 632–640 + regulatory T cells and protects against fatal lung disease in
22. Li, M.O. and Rudensky, A.Y. (2016) T cell receptor signalling in mice. J. Immunol. 192, 985–995
the control of regulatory T cell differentiation and function. Nat. 48. Thornton, A.M. et al. (2010) Expression of Helios, an Ikaros
Rev. Immunol. 16, 220–233 transcription factor family member, differentiates thymic-derived
23. Ouyang, W. et al. (2010) Transforming growth factor-b signaling from peripherally induced Foxp3 + T regulatory cells. J. Immunol.
curbs thymic negative selection promoting regulatory T cell 184, 3433–3441
development. Immunity 32, 642–653 49. Kim, H.J. et al. (2015) Stable inhibitory activity of regulatory T cells
24. Rao, S. et al. (2003) c-Rel is required for chromatin remodeling requires the transcription factor Helios. Science 350, 334–339
across the IL-2 gene promoter. J. Immunol. 170, 3724–3731 50. Nakagawa, H. et al. (2016) Instability of Helios-deficient Tregs is
25. Zheng, Y. et al. (2010) Role of conserved non-coding DNA associated with conversion to a T-effector phenotype and
elements in the Foxp3 gene in regulatory T-cell fate. Nature enhanced antitumor immunity. Proc. Natl. Acad. Sci. U.S.A.
463, 808–812 113, 6248–6253
26. Feng, Y. et al. (2015) A mechanism for expansion of regulatory T- 51. Takatori, H. et al. (2015) Helios enhances Treg cell function in
cell repertoire and its role in self-tolerance. Nature 528, 132–136 cooperation with FoxP3. Arthritis Rheumatol. 67, 1491–1502
27. Yurchenko, E. et al. (2012) Inflammation-driven reprogramming 52. Spence, A. et al. (2015) Targeting Treg signaling for the treatment
of CD4 + Foxp3 + regulatory T cells into pathogenic Th1/Th17 T of autoimmune diseases. Curr. Opin. Immunol. 37, 11–20
effectors is abrogated by mTOR inhibition in vivo. PLoS ONE 7, 53. Perl, A. (2016) Activation of mTOR (mechanistic target of rapa-
e35572 mycin) in rheumatic diseases. Nat. Rev. Rheumatol. 12, 169–182
28. Duarte, J.H. et al. (2009) Natural Treg cells spontaneously dif- 54. Delgoffe, G.M. et al. (2011) The kinase mTOR regulates the
ferentiate into pathogenic helper cells in lymphopenic conditions. differentiation of helper T cells through the selective activation
Eur. J. Immunol. 39, 948–955 of signaling by mTORC1 and mTORC2. Nat. Immunol. 12,
29. Komatsu, N. et al. (2009) Heterogeneity of natural Foxp3 + T 295–303
cells: a committed regulatory T-cell lineage and an uncommitted 55. Coe, D.J. et al. (2014) Metabolic regulation of regulatory T cell
minor population retaining plasticity. Proc. Natl. Acad. Sci. U.S.A. development and function. Front. Immunol. 5, 590
106, 1903–1908
56. Michalek, R.D. et al. (2011) Cutting edge: distinct glycolytic and
30. Zhou, X. et al. (2009) Instability of the transcription factor Foxp3 lipid oxidative metabolic programs are essential for effector and
leads to the generation of pathogenic memory T cells in vivo. Nat. regulatory CD4 + T cell subsets. J. Immunol. 186, 3299–3303
Immunol. 10, 1000–1007
57. Zeng, H. et al. (2013) mTORC1 couples immune signals and
31. Hori, S. (2014) Lineage stability and phenotypic plasticity of metabolic programming to establish T(reg)-cell function. Nature
Foxp3(+) regulatory T cells. Immunol. Rev. 259, 159–172 499, 485–490
32. Burchill, M.A. et al. (2008) Linked T cell receptor and cytokine 58. Powell, J.D. et al. (2012) Regulation of immune responses by
signaling govern the development of the regulatory T cell reper- mTOR. Annu. Rev. Immunol. 30, 39–68
toire. Immunity 28, 112–121
59. Walsh, P.T. et al. (2006) PTEN inhibits IL-2 receptor-mediated
33. Feng, Y. et al. (2014) Control of the inheritance of regulatory T cell expansion of CD4 + CD25 + Tregs. J. Clin. Invest. 116,
identity by a cis element in the Foxp3 locus. Cell 158, 749–763 2521–2531
34. Li, X. et al. (2014) Function of a Foxp3 cis-element in protecting 60. Delgoffe, G.M. et al. (2013) Stability and function of regulatory T
regulatory T cell identity. Cell 158, 734–748 cells is maintained by a neuropilin-1-semaphorin-4a axis. Nature
35. Rubtsov, Y.P. et al. (2010) Stability of the regulatory T cell lineage 501, 252–256
in vivo. Science 329, 1667–1671 61. Shrestha, S. et al. (2015) Treg cells require the phosphatase
36. Floess, S. et al. (2007) Epigenetic control of the foxp3 locus in PTEN to restrain TH1 and TFH cell responses. Nat. Immunol. 16,
regulatory T cells. PLoS Biol. 5, e38 178–187
37. Wang, L. et al. (2013) Foxp3 + T-regulatory cells require DNA 62. Huynh, A. et al. (2015) Control of PI(3) kinase in Treg cells
methyltransferase 1 expression to prevent development of lethal maintains homeostasis and lineage stability. Nat. Immunol. 16,
autoimmunity. Blood 121, 3631–3639 188–196

Trends in Molecular Medicine, September 2016, Vol. 22, No. 9 795


63. Apostolidis, S.A. et al. (2016) Phosphatase PP2A is requisite for 85. Link, W. et al. (2009) Chemical interrogation of FOXO3a
the function of regulatory T cells. Nat. Immunol. 17, 556–564 nuclear translocation identifies potent and selective inhibitors
64. Charbonnier, L.M. et al. (2015) Control of peripheral tolerance by of phosphoinositide 3-kinases. J. Biol. Chem. 284,
regulatory T cell-intrinsic Notch signaling. Nat. Immunol. 16, 28392–28400
1162–1173 86. Liu, G. et al. (2010) The S1P1-mTOR axis directs the reciprocal
65. Beyer, M. et al. (2011) Repression of the genome organizer differentiation of TH1 and Treg cells. Nat. Immunol. 11,
SATB1 in regulatory T cells is required for suppressive func- 1047–1056
tion and inhibition of effector differentiation. Nat. Immunol. 12, 87. Liu, G. et al. (2009) The receptor S1P1 overrides regulatory T cell-
898–907 mediated immune suppression through Akt-mTOR. Nat. Immu-
66. Kondo, M. et al. (2016) SATB1 plays a critical role in establish- nol. 10, 769–777
ment of immune tolerance. J. Immunol. 196, 563–572 88. Lai, Z.W. et al. (2012) N-acetylcysteine reduces disease activity
67. Torgerson, T.R. and Ochs, H.D. (2007) Immune dysregulation, by blocking mammalian target of rapamycin in T cells
polyendocrinopathy, enteropathy, X-linked: forkhead box protein from systemic lupus erythematosus patients: a randomized,
3 mutations and lack of regulatory T cells. J. Allergy Clin. Immu- double-blind, placebo-controlled trial. Arthritis Rheum. 64,
nol. 120, 744–750 quiz 751–742 2937–2946

68. Gambineri, E. et al. (2008) Clinical and molecular profile of a new 89. Yin, Y. et al. (2015) Normalization of CD4 + T cell metabolism
series of patients with immune dysregulation, polyendocrinop- reverses lupus. Sci. Transl. Med. 7, 274ra218
athy, enteropathy, X-linked syndrome: inconsistent correlation 90. Koga, T. et al. (2014) KN-93, an inhibitor of calcium/calmodulin-
between forkhead box protein 3 expression and disease severity. dependent protein kinase IV, promotes generation and function
J. Allergy Clin. Immunol. 122, 1105–1112 of Foxp3(+) regulatory T cells in MRL/lpr mice. Autoimmunity 47,
69. Kucuk, Z.Y. et al. (2016) A challenging undertaking: stem cell 445–450
transplantation for immune dysregulation, polyendocrinopathy, 91. Ichinose, K. et al. (2011) Suppression of autoimmunity and organ
enteropathy, X-linked (IPEX) syndrome. J. Allergy Clin. Immunol. pathology in lupus-prone mice upon inhibition of calcium/cal-
137, 953–955 modulin-dependent protein kinase type IV. Arthritis Rheum. 63,
70. Sharfe, N. et al. (1997) Human immune disorder arising from 523–529
mutation of the alpha chain of the interleukin-2 receptor. Proc. 92. Koga, T. et al. (2014) CaMK4-dependent activation of AKT/mTOR
Natl. Acad. Sci. U.S.A. 94, 3168–3171 and CREM-/ underlies autoimmunity-associated Th17 imbal-
71. Caudy, A.A. et al. (2007) CD25 deficiency causes an immune ance. The Journal of Clinical Investigation 124, 2234–2245
dysregulation, polyendocrinopathy, enteropathy, X-linked-like 93. Fujikawa, K. et al. (2009) Calcium/calmodulin-dependent protein
syndrome, and defective IL-10 expression from CD4 lympho- kinase II (CaMKII) regulates tumour necrosis factor-related apo-
cytes. J. Allergy Clin. Immunol. 119, 482–487 ptosis inducing ligand (TRAIL)-mediated apoptosis of fibroblast-
72. Bernasconi, A. et al. (2006) Characterization of immunodefi- like synovial cells (FLS) by phosphorylation of Akt. Clin. Exp.
ciency in a patient with growth hormone insensitivity secondary Rheumatol. 27, 952–957
to a novel STAT5b gene mutation. Pediatrics 118, e1584–e1592 94. Westra, J. et al. (2010) Expression and regulation of HIF-1alpha in
73. Nadeau, K. et al. (2011) STAT5b deficiency: an unsuspected macrophages under inflammatory conditions; significant reduc-
cause of growth failure, immunodeficiency, and severe pulmo- tion of VEGF by CaMKII inhibitor. BMC Musculoskelet. Disord.
nary disease. J. Pediatr. 158, 701–708 11, 1–11

74. Tivol, E.A. et al. (1995) Loss of CTLA-4 leads to massive 95. Benjamin, D. et al. (2011) Rapamycin passes the torch: a new
lymphoproliferation and fatal multiorgan tissue destruction, generation of mTOR inhibitors. Nat. Rev. Drug Discov. 10,
revealing a critical negative regulatory role of CTLA-4. Immu- 868–880
nity 3, 541–547 96. Polansky, J.K. et al. (2008) DNA methylation controls Foxp3 gene
75. Schubert, D. et al. (2014) Autosomal dominant immune dysre- expression. Eur. J. Immunol. 38, 1654–1663
gulation syndrome in humans with CTLA4 mutations. Nat. Med. 97. Josefowicz, S.Z. et al. (2009) Cutting edge: TCR stimulation is
20, 1410–1416 sufficient for induction of Foxp3 expression in the absence of
76. Charbonnier, L.M. et al. (2015) Regulatory T-cell deficiency and DNA methyltransferase 1. J. Immunol. 182, 6648–6652
immune dysregulation, polyendocrinopathy, enteropathy, X- 98. Hancock, W.W. et al. (2012) HDAC inhibitor therapy in auto-
linked-like disorder caused by loss-of-function mutations in immunity and transplantation. Ann. Rheum. Dis. 71 (Suppl. 2),
LRBA. J. Allergy Clin. Immunol. 135, 217–227 i46–i54
77. Lo, B. et al. (2015) Autoimmune Disease. Patients with LRBA 99. Regna, N.L. et al. (2016) Specific HDAC6 inhibition by ACY-738
deficiency show CTLA4 loss and immune dysregulation respon- reduces SLE pathogenesis in NZB/W mice. Clin. Immunol. 162,
sive to abatacept therapy. Science 349, 436–440 58–73
78. Ohl, K. and Tenbrock, K. (2015) Regulatory T cells in systemic 100. Mizui, M. et al. (2014) IL-2 protects lupus-prone mice from
lupus erythematosus. Eur. J. Immunol. 45, 344–355 multiple end-organ damage by limiting CD4-CD8- IL-17-produc-
79. Lyssuk, E.Y. et al. (2007) Reduced number and function of CD4 ing T cells. J. Immunol. 193, 2168–2177
+ CD25highFoxP3 + regulatory T cells in patients with systemic 101. Humrich, J.Y. et al. (2015) Rapid induction of clinical remission by
lupus erythematosus. Adv. Exp. Med. Biol. 601, 113–119 low-dose interleukin-2 in a patient with refractory SLE. Ann.
80. Bonelli, M. et al. (2008) Quantitative and qualitative deficiencies of Rheum. Dis. 74, 791–792
regulatory T cells in patients with systemic lupus erythematosus 102. Saadoun, D. et al. (2011) Regulatory T-cell responses to low-
(SLE). Int. Immunol. 20, 861–868 dose interleukin-2 in HCV-induced vasculitis. N. Engl. J. Med.
81. Chowdary Venigalla, R.K. et al. (2008) Reduced CD4 + , 365, 2067–2077
CD25  T cell sensitivity to the suppressive function of CD4 103. Koreth, J. et al. (2011) Interleukin-2 and regulatory T cells in graft-
+ ,CD25high,CD127  /low regulatory T cells in patients with versus-host disease. N. Engl. J. Med. 365, 2055–2066
active systemic lupus erythematosus. Arthritis Rheum. 58, 104. von Spee-Mayer, C. et al. (2015) Low-dose interleukin-2 selec-
2120–2130 tively corrects regulatory T cell defects in patients with systemic
82. Afeltra, A. et al. (2015) The involvement of T regulatory lympho- lupus erythematosus. Ann. Rheum. Dis. 75, 1407–1415
cytes in a cohort of lupus nephritis patients: a pilot study. Intern. 105. Bruyn, G.A. et al. (2008) Everolimus in patients with rheumatoid
Emerg. Med. 10, 677–683 arthritis receiving concomitant methotrexate: a 3-month, double-
83. Daniel, C. et al. (2010) Enhancement of antigen-specific blind, randomised, placebo-controlled, parallel-group, proof-of-
Treg vaccination in vivo. Proc. Natl. Acad. Sci. U.S.A. 107, concept study. Ann. Rheum. Dis. 67, 1090–1095
16246–16251 106. Yap, D.Y. et al. (2012) Proliferation signal inhibitors in the treat-
84. Battaglia, M. et al. (2005) Rapamycin selectively expands CD4 ment of lupus nephritis: preliminary experience. Nephrology
+ CD25 + FoxP3 + regulatory T cells. Blood 105, 4743–4748 (Carlton) 17, 676–680

796 Trends in Molecular Medicine, September 2016, Vol. 22, No. 9


107. Ando, S. et al. (2010) FTY720 exerts a survival advantage 115. Chaudhry, A. et al. (2009) CD4 + regulatory T cells control
through the prevention of end-stage glomerular inflammation TH17 responses in a Stat3-dependent manner. Science
in lupus-prone BXSB mice. Biochem. Biophys. Res. Commun. 326, 986–991
394, 804–810 116. Zheng, Y. et al. (2009) Regulatory T-cell suppressor program co-
108. Han, Y. et al. (2015) FTY720 abrogates collagen-induced arthritis opts transcription factor IRF4 to control T(H)2 responses. Nature
by hindering dendritic cell migration to local lymph nodes. J. 458, 351–356
Immunol. 195, 4126–4135 117. Koch, M.A. et al. (2009) The transcription factor T-bet controls
109. La Mantia, L. et al. (2016) Fingolimod for relapsing-remitting regulatory T cell homeostasis and function during type 1 inflam-
multiple sclerosis. Cochrane Database Syst Rev 4, CD009371 mation. Nat. Immunol. 10, 595–602
110. Lee, J. et al. (2015) A novel histone deacetylase 6-selective 118. Wang, Y. et al. (2011) An essential role of the transcription
inhibitor suppresses synovial inflammation and joint destruction factor GATA-3 for the function of regulatory T cells. Immunity
in a collagen antibody-induced arthritis mouse model. Int. J. 35, 337–348
Rheum. Dis. 18, 514–523 119. Cipolletta, D. (2014) Adipose tissue-resident regulatory T cells:
111. Chen, W. et al. (2003) Conversion of peripheral CD4 + CD25- phenotypic specialization, functions and therapeutic potential.
naive T cells to CD4 + CD25 + regulatory T cells by TGF-beta Immunology 142, 517–525
induction of transcription factor Foxp3. J. Exp. Med. 198, 120. Arpaia, N. et al. (2015) A distinct function of regulatory T cells in
1875–1886 tissue protection. Cell 162, 1078–1089
112. Khattri, R. et al. (2003) An essential role for Scurfin in CD4 121. Wei, J. et al. (2016) Autophagy enforces functional integrity of
+ CD25 + T regulatory cells. Nat. Immunol. 4, 337–342 regulatory T cells by coupling environmental cues and metabolic
113. Hsieh, C.S. et al. (2006) An intersection between the self-reactive homeostasis. Nat. Immunol. 17, 277–285
regulatory and nonregulatory T cell receptor repertoires. Nat. 122. Kabat, A.M. et al. (2016) The autophagy gene Atg16l1 differen-
Immunol. 7, 401–410 tially regulates Treg and TH2 cells to control intestinal inflamma-
114. Li, X. and Zheng, Y. (2015) Regulatory T cell identity: formation tion. Elife 5, e12444
and maintenance. Trends Immunol. 36, 344–353

Trends in Molecular Medicine, September 2016, Vol. 22, No. 9 797

You might also like