Negative Feedback Regulation of T Cells Via Interleukin-2 and FOXP3 Reciprocity

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Negative Feedback Regulation of T Cells via Interleukin-2

and FOXP3 Reciprocity


Zoran Popmihajlov, Kendall A. Smith*
Division of Immunology, Department of Medicine, Weill Medical College, Cornell University, New York, New York, United States of America

Abstract
As interleukin-2 (IL2) is central to the clonal expansion of antigen-selected T cells, we investigated the relationship between
IL2 and the negative regulatory transcription factor FOXP3. We found IL2 to be responsible for T cell antigen receptor (TCR)-
activated FOXP3 expression by both CD4+ and CD8+ human T cells, and as anticipated, FOXP3 expression restricted TCR-
stimulated IL2 expression. However, no evidence could be found that FOXP3+ cells actively suppress IL2 expression by
FOXP3- cells. These data are consistent with an IL2/FOXP3-dependent negative feedback loop that normally regulates the T
cell immune response. It follows that a defect in this negative feedback loop as a result of a deficiency of either IL2 or FOXP3
will lead to a hyperproliferative autoimmune syndrome, without the necessity of invoking an active suppressive function for
FOXP3+ T cells.

Citation: Popmihajlov Z, Smith KA (2008) Negative Feedback Regulation of T Cells via Interleukin-2 and FOXP3 Reciprocity. PLoS ONE 3(2): e1581. doi:10.1371/
journal.pone.0001581
Editor: Derya Unutmaz, New York University School of Medicine, United States of America
Received November 20, 2007; Accepted January 14, 2008; Published February 13, 2008
Copyright: ß 2008 Popmihajlov, Smith. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Funding: NIAID/NIH, Belfer Foundation. Neither funding source participated in the conduct of the study, in the collection, analysis, intrpretation of the data, or in
the preparation, review or approval of the manuscript.
Competing Interests: The authors have declared that no competing interests exist.
*E-mail: kasmith@med.cornell.edu

Introduction At about this time the scurfy mutant mouse [9] was found to be
suffering from a similar lymphoproliferative phenotype [10,11],
Thirty years ago, when the T cell growth factor interleukin-2 (IL2) which was attributed to an over-expression of cytokine genes by
was first quantified, we found it to be produced only transiently after CD4+ T cells [12,13,14]. Subsequently, it was shown that scurfy
T cell activation by mitogens or antigens [1]. As an explanation, we T cells are hyper-responsive to T cell antigen receptor (TCR)
postulated that perhaps there was a natural feedback inhibition triggering, which prompted the speculation that perhaps the
operative that shut down IL2 production, thereby limiting IL2- scurfy phenotype results from a defect of a normal feedback
mediated T cell proliferation during an immune response. A search down-regulation of TCR activation of cytokine gene expression
for the release of a soluble inhibitor proved negative, but additional [15].
experiments revealed that IL2-responsive T cells actually consume A possible explanation for these observations was introduced
IL2, thus providing at least one explanation for the disappearance of when it was reported that CD4+ T cells expressing the a-chain of the
IL2 as the cells proliferate to high densities [1,2,3]. IL2 receptor (IL2Ra) (CD25) prevent a quite similar lethal
Subsequently, targeted disruption of the IL2 gene resulted in IL2 lymphoproliferative syndrome when transferred to lymphopenic
(2/2) mice with apparently normal lymphocyte development, but (nu/nu) [16], and neonatal thymectomized mice [17,18]. Additional
deficient in vitro T cell proliferative responses [4] . However, as these reports confirmed these initial findings, and CD4+CD25+ T cells
mice aged they developed a paradoxical autoimmune lymphopro- were proposed to represent a unique lineage of immunoregulatory T
liferative syndrome, with the accumulation of activated T cells in cells or ‘‘Regulatory T cells’’ (T-Regs) that function normally to
multiple organs, including salivary glands, lungs, kidneys, heart, actively suppress immune responses to potential autoantigens [19].
pancreas and liver [5]. As well, autoimmune hemolytic anemia and Subsequently, two functional characteristics of CD4+CD25+
inflammatory bowel disease ultimately led to premature death [6]. T-Regs were described; anergy, defined by an incapacity to
These findings led us to the hypothesis that an unanticipated crucial produce IL2 and proliferate when activated via the TCR, and as
defect resulting from the elimination of IL2 might be a lack of a well, the capacity to actively suppress polyclonal T cell
negative regulatory feedback function [7]. proliferation in vitro, via inhibition of IL2 production by TCR-
Our experiments to try to understand this phenomenon activated ‘‘effector’’ cells [20]. It is noteworthy that high ratios of
demonstrated that IL2 administration to IL2 (2/2) mice T-Reg to T-effector cells (i.e. 2:1, 1:1, 1:2, 1:4) are usually
prevented the onset of the autoimmune syndrome. Furthermore, necessary to demonstrate the in vitro suppressive effect. Problematic
adoptive transfer of splenocytes and thymocytes from IL2-treated with this sort of assay is the capacity for IL2R+ cells to passively
IL2 (2/2) mice delayed the onset of disease, thereby leading us to bind, remove and degrade IL2, which then appears as if there is a
the conclusion that IL2 induces some T cell maturation/ suppressive activity, since proliferation is driven by IL2 [1,2,3].
differentiation event that subsequently prevents the cells from Moreover, problematic in the designation of CD4+CD25+ T cells
responding to self antigens [8]. However, the precise nature of this as T-Regs was that the same phenotype is shared by activated,
IL2-induced cellular change remained obscure. nonanergic and nonsuppressive ‘‘effector’’ T cells [21].

PLoS ONE | www.plosone.org 1 February 2008 | Volume 3 | Issue 2 | e1581


IL2 and FOXP3 Reciprocity

When the scurfy gene was cloned [22], it was found to encode a activation; 1) the promotion of proliferative clonal expansion of
new member of the forkhead family of transcription factors, antigen-selected cells, and 2) a negative feedback mechanism that
FOXP3. When tested for activity, over expression of FOXP3 in restricts IL2 production, thereby ensuring a self-limited proliferative
CD4+ T cells was found to attenuate TCR-induced IL2 response. By comparison, no evidence was found that FOXP3+ cells
production and subsequent proliferation, thereby providing are capable of actively suppressing IL2 production by FOXP3- cells.
molecular support for the negative feedback hypothesis [23].
Moreover, coincident with the identification of the scurfy gene as Results
encoding FOXP3 in mice, the human ortholog was cloned and FOXP3 expression by resting and anti-CD3-activated
found to be mutated in individuals suffering from the X-linked
PBMCs
autoimmunity-allergic dysregulation (XLAAD) syndrome [24],
A survey of 19 individuals revealed that only a few freshly isolated
and the X-linked neonatal diabetes mellitus, enteropathy and
human T cells expressed detectable FOXP3, with a greater propor-
endocrinopathy (IPEX) syndrome [25,26].
tion in CD4+ T cells (2.64%60.33%) (Mean6SEM, n = 19) vs.
Soon thereafter, three almost simultaneous reports [27,28,29]
CD8+ T cells (0.16%60.05%) (Mean6SEM, n = 19). The expres-
linked mutations of FOXP3 with the lack of development of T-Regs
sion of FOXP3 vs. CD25 by freshly isolated CD4+ and CD8+ T cells
in the mouse, thereby providing a potential explanation for the
is shown by representative contour plots in Figure 1A. It is note-
severe autoimmune phenotypes of both mice and man, i.e. the lack
worthy that only CD25+ cells express FOXP3, and that not all
of anergic, suppressive T-Reg cells. These initial studies indicated
CD25+ cells are FOXP3+. Actually in the representative experiment
that in the mouse, FOXP3 expression was restricted to
shown (Figure 1A), only 12.2% of freshly isolated CD4+CD25+ T
CD4+CD25+ T cells, both in the thymus and in the periphery,
cells express FOXP3, and only 7.7% of freshly isolated
thus consistent with the T-Reg phenotype. Moreover, in vitro
stimulation of CD4+CD25- T cells by anti-CD3 together with anti- CD8+CD25+ cells express FOXP3. It is also noteworthy that the
CD28 or IL2 failed to elicit FOXP3 mRNA expression. Accordingly, level of both CD25 and FOXP3 expression is low as detected by the
FOXP3 expression by CD4+CD25+ T cells was quickly adopted as fluorescent intensities. According to standard terminology, the
a more definitive phenotypic definition of a T-Reg cell. CD4+CD25+FOXP3+ cells would be ‘‘natural T-Regs’’.
However, confounding these data were observations made with After activation with anti-CD3 the time course of FOXP3
human T cells. Activation of human Peripheral Blood Mononu- expression by both CD4+ T cells and CD8+ T cells was analyzed by
clear Cells (PBMC) via anti-CD3 was found to result in an increase flow cytometry after successive intervals of culture (Fig 1B). Increased
of FOXP3 mRNA in CD4+ T cells [30]. Moreover, human CD8+ FOXP3 expression could first be detected above baseline after
T cells were also found to express FOXP3 mRNA [31,32]. These 12 hours, with peak levels observed at 24 hours in CD4+ T cells
findings prompted the speculation that the expression of FOXP3 (13.97%60.87 %) (Mean6SEM, n = 4) and CD8+ T cells
in an antigen-activated T cell could act as a natural negative 6.6961.04% (Mean6SEM, n = 4). Thereafter, the frequency of
feedback loop that would prevent unrestricted cytokine production FOXP3+ cells declined progressively, returning to baseline by
and inflammatory reactions [31]. 96 hours (day 4) of culture. For comparison, the expression of CD25
Recently, antibodies reactive with FOXP3 became available, was also monitored (Fig 1B). It is readily apparent that CD25
facilitating experimental approaches in this field. Of interest, it was expression precedes FOXP3 expression and eventually is detectable
found that peak expression of FOXP3 in human T cells requires as on .80% of both CD4+ and CD8+ T cells by 48 hours following
long as three days after activation with anti-CD3 in vitro, thereby activation, thereby attesting to the efficiency of anti-CD3 activation.
almost excluding an immediate/early FOXP3 gene activation via Representative 4-quadrant contour plots of T cells from PBMCs
the TCR/CD3 complex [33]. Very recently, others reported that cultured with anti-CD3 for 24 hours are shown in Fig 1C, comparing
antibodies reactive with IL2 or the IL2 receptor diminished TCR/ FOXP3 expression and CD25 expression. In this typical example of
CD3-induced FOXP3 expression by CD8+ T cells [34]. By 29 separate experiments, it is noteworthy again that only CD25+ cells
comparison, purified CD4+CD45+CD25-CD127+ human T cells express FOXP3, but not all CD25+ cells express FOXP3. Thus, even
activated with solid-phase anti-CD3+soluble anti-CD28 in the though 86.4% (15.9% +70.5%) of the CD4+ T cells became CD25+
presence of both IL2 and TGFb supplied exogenously results in after 24 hours, only 18.4% of the CD4+CD25+ cells also expressed
FOXP3 expression by most remaining cells [35,36]. In addition, FOXP3. Likewise, 73.1% (8.1%+65.0%) of the CD8+ became
other recent studies demonstrated that IL2 provides a negative CD25+ after 24 hours, but only 11.1% of the CD8+CD25+ cells also
feedback effect on its own production via Signal Transducer and expressed FOXP3. It is also important to notice that FOXP3 is only
Activator of Transcription-5 (STAT5), although the mechanism expressed by CD25+ cells that also express the highest levels of CD25,
whereby this occurs was left unexplored [37]. and that the level of FOXP3 expression is ,10-fold higher than that
Recently, using several experimental approaches CD4+CD25+ of the freshly isolated cells, both CD4+ as well as CD8+, as detected
FOXP3+ cells were shown to suppress effector CD4+ T cells not by by fluorescence intensity.
affecting early TCR activation or proliferation of effector T cells. As shown in Fig 1D, the mean FOXP3 expression of freshly
Instead, because CD4+CD25+FOXP3+ cells are anergic and isolated T cells is compared with the mean FOXP3 expression after
cannot produce IL2, but can consume and degrade it, these cells 24 hours of culture with anti-CD3. The FOXP3 expression of CD4+
induce cytokine deprivation-mediated apoptosis of effector T cells, T cells increased ,5-fold from 2.99%60.36% at baseline to
both in vitro and in vivo [38]. Accordingly, we focused on the role of 13.97%60.61% (Mean6SEM, n = 16) after 24 hours culture
IL2 itself in the TCR/CD3-induction of FOXP3 expression by (p = 0.0004), while the FOXP3 expression of CD8+ T cells increased
normal human T cells, as well as the effect of FOXP3 on IL2 ,50-fold from 0.19%60.06% at baseline to 9.38%61.50%
expression by both FOXP3+ and FOXP3- cells. Our results, (Mean6SEM, n = 17) after 24 hours culture (p = 0.0003).
summarized in this report, are consistent with the interpretation that
IL2-induced FOXP3 expression plays a larger role in immune Exogenous, recombinant IL2 prolongs FOXP3 expression
regulation than has been conjectured until now, serving to provide As the time course of FOXP3 expression upon activation with
for a negative feedback loop, restricting IL2 production. Thus, IL2 anti-CD3 did not precede, but followed IL2R expression, we
appears to have at least two important functions during T cell examined the effect of adding IL2 exogenously at t = 0 on the anti-

PLoS ONE | www.plosone.org 2 February 2008 | Volume 3 | Issue 2 | e1581


IL2 and FOXP3 Reciprocity

Figure 1. Differential FOXP3 expression by resting and anti-CD3-activated PBMCs. (A) Individual contour plots of freshly isolated total
PBMCs representative of CD4+ (n = 19) and CD8+ cells (n = 19) stained with RatIgG2a (upper row) or FOXP3 (lower row) and IL2Ra (CD25). The
percentages of the different cell populations are also shown for each quadrant of the contour plots in each upper right quadrant. (B) CD25 and
FOXP3 expression in CD4+ cells (upper graph) and CD8+ (lower graph) cells were monitored daily upon activation of total PBMCs (t = 0). CD25
expression corresponds to the left axis (0–100%), while FOXP3 expression corresponds to the right axis (0–20%). The results are shown as Mean6SEM
of 4 independent experiments. (C) Representative contour plots of total PBMC activated for 24 h show CD4+ and CD8+ cells stained with RatIgG2a
(upper row) or FOXP3 (lower row) and CD25 as in A. The percentages of different population are also shown for the contour plots in each upper right
quadrant. (D) The difference of FOXP3 expression between freshly isolated vs. aCD3-activated for 24 h in CD4+ (black bars; n = 16) and in CD8+ (gray
bars; n = 17) cells. The results are shown as mean6SEM (vertical brackets).
doi:10.1371/journal.pone.0001581.g001

CD3-induced FOXP3 expression. A saturating IL2 concentration IL2 itself. As shown in Fig 3C, increasing anti-IL2 concentrations
(10 nM) did not accelerate FOXP3 expression by either CD4+ or suppressed anti-CD3-induced FOXP3 expression by both CD4+
CD8+ T cells (Fig 2). However, IL2 supplementation did prolong and CD8+ T cells. Again, IL2 specificity was shown, in that this
FOXP3 expression for 72–96 hours by both CD4+ T cells (Fig 2A) suppression could be competed by increasing IL2 concentrations
and by CD8+ T cells (Fig 2B), thereby suggesting that the duration (Fig 3D).
of FOXP3 expression is dependent upon continuous signaling via Although antibodies that block either IL2 or the IL2R a-chain
the IL2/IL2R interaction. As shown in the insets, there was no suppressed FOXP3 expression, neither antibody was completely
change in the CD25 expression with the IL2 supplementation, inhibitory. Therefore, a combination of anti-CD25 and anti-IL2 was
which was already increased by 12 hours and became maximal by tested, at near saturating antibody concentrations as shown in Fig 4A.
48 hours after initiation of the cultures. The combination of anti-IL2 and anti-IL2R a chain suppressed the
anti-CD3-induced FOXP3 expression of CD4+ T cells by .60% and
Antibodies reactive with the both the IL2R and IL2 inhibit of CD8+ T cells by .80%, and exogenous IL2 supplementation was
the anti-CD3-induced FOXP3 expression capable of overcoming the suppression mediated by the combined
If the expression of FOXP3 induced by anti-CD3 triggering was mAbs. However, as shown in Fig 4B, increasing the combined
actually secondary to anti-CD3-induced IL2 production and IL2R antibody concentrations 10-fold resulted in almost complete
expression, followed by IL2/IL2R signaling, then antibodies suppression of FOXP3 expression (80% for CD4+ and 95% for
reactive with the IL2R or with IL2 itself would be expected to CD8+ T cells), which could not be overcome by excess IL2 (10 nM).
inhibit FOXP3 expression. As shown in Fig 3A, anti-CD25
suppressed FOXP3 expression by both CD4+ and CD8+ T cells in Other cc-chain cytokines have no effect on anti-CD3-
a concentration-dependent fashion. Moreover, IL2 at an IL2R induced FOXP3 expression
saturating concentration (10 nM) completely overcame the anti- Although these experiments supported the interpretation that
CD25 suppression. That IL2 competed with the anti-CD25 is FOXP3 expression was regulated specifically by IL2, it was
shown in Fig 3B, where increasing IL2 concentrations progres- important to test other cytokines as well, especially those known to
sively circumvented the suppressive effect of a half-inhibitory signal via the cc-chain, which activates JAK3 and STAT5. As
concentration (IC50 ,1 mg/mL) of anti-CD25. shown in Supplementary online material (Fig S1), there was no
Confirmation of the IL2-dependent expression of FOXP3 upon effect on anti-CD3-induced FOXP3 expression by mAbs reactive
TCR/CD3 triggering was obtained using antibodies reactive with with IL4, IL7, IL15, or the IL21R.

PLoS ONE | www.plosone.org 3 February 2008 | Volume 3 | Issue 2 | e1581


IL2 and FOXP3 Reciprocity

FOXP3 expression restricts but does not suppress IL2


expression upon TCR/CD3 restimulation
The observation that FOXP3 and IL2 expression were mutually
exclusive is consistent with the hypothesis that FOXP3 functions to
restrict IL2 expression by FOXP3+ cells, but does not actively
suppress IL2 expression by FOXP3- cells. Accordingly, to test this
hypothesis, PBMCs were activated for 24 hours with anti-
CD3+IL2 (10 nM) to promote maximal FOXP3 expression. The
cells were then harvested and reactivated for 6 hours with anti-
CD3+anti-CD28, after which both FOXP3 and cytokine expres-
sion were monitored. In this experimental design, the cells are
present in a physiologic setting, at relative concentrations that
should allow detection of FOXP3+ cell active suppression of IL2
expression by FOXP3- cells. As shown by a representative
experiment in Figure 5C, after TCR restimulation, 94.2% of the
FOXP3+ cells remained IL2-. Moreover, the few detectable
double positive cells (5.8%) expressed only low levels of FOXP3 as
monitored by mean fluorescence intensity. By comparison, the
presence of FOXP3+ cells did not actively suppress the IL2
expression by FOXP3- cells, which increased 4-fold from 1.8% to
7.4%. Thus, IL2 expression was restricted in FOXP3+ cells, but
was not suppressed in FOXP3- cells.
Compilation of data from 5 individual experiments (Table S1,
Supplementary online material) revealed that after restimulation,
the mean IL2 expression by FOXP3- cells increased 10-fold, from
1.3%60.4% (Mean6SEM) before restimulation to 12.6%62.2%
(Mean6SEM). Moreover, only 2.0%60.4% (Mean6SEM) of
cells expressed both FOXP3 and IL2. Accordingly, in this
physiologic experimental setting, with a ratio of FOXP3+ to
FOXP3- cells of 1:5 (i.e. mean 16.5% FOXP3+ cells vs. 83.5%
Figure 2. Exogenous, recombinant IL2 prolongs FOXP3 ex- FOXP3- cells before reactivation), it was even more apparent that
pression. The time course of FOXP3 expression in (A) CD4+cells and FOXP3+ expression restricted the expression of IL2, but FOXP3+
(B) CD8+ cells upon aCD3-activation of PBMCs (t = 0), with and without cells could not actively suppress the expression of IL2 by most
exogenous rIL2 [10 nM]. Insets show the IL2Ra (CD25) expression under FOXP3- cells.
the same conditions. The results are shown as Mean6SEM of 4–6
independent experiments.
doi:10.1371/journal.pone.0001581.g002 Discussion
These results support the conclusion that the IL2/IL2R
The expression of FOXP3 and IL2 are mutually exclusive interaction is responsible for most of the observed increased FOXP3
Inasmuch as IL2 appeared responsible for stimulating the expression by TCR/CD3-activated human T cells. The specificities
expression of FOXP3, and FOXP3 was reported to attenuate IL2 of anti-CD25 and anti-IL2, both of which compete solely for IL2-
gene expression, the effect of FOXP3 on anti-CD3-induced IL2 IL2R interactions, together with the lack of an inhibitory effect of
production was examined. PBMCs were labeled with CFSE, and mAbs reactive with other IL2Rcc cytokines, essentially leaves little
activated with anti-CD3. As shown in Figure 5A, CD4+ T cells were possibility for any other interpretations. Moreover, finding that
monitored daily for FOXP3 vs. IL2 expression, and for cell division FOXP3 and IL2 expression are mutually exclusive, especially upon
by quantitative CFSE dilution (insets). At t = 0, all of the cells could restimulation, leads inevitably to the interpretation that an IL2-
be ascribed to a homogeneous single CFSE peak (Division = 0), and FOXP3-mediated negative feedback loop regulates the antigen-
expressed low levels of FOXP3 (3.4%) and IL2 (1.0%). After activated T cell immune response by controlling IL2 production.
24 hours of culture, all of the cells remained undivided (Divi- However, we could find no evidence that FOXP3+ cells actively
sion = 0). At this time interval, none of the cells had divided, but suppress IL2 production by FOXP3- cells.
FOXP3 had increased to 8.2%. Accordingly, the increased Given these findings, it is inescapable that a deficiency in this
percentage of FOXP3+ cells could only have been derived from negative feedback loop could underlie the hyperproliferative/
cells that were FOXP3- at t = 0. It is also noteworthy that few cells autoimmune phenotypes of the IL2 (2/2) and scurfy mice, as well
expressed both FOXP3 and IL2 (0.1%). After 48 hours of culture as in the XLAAD/IPEX patients. In this regard, there appears to
the separation of FOXP3+ (10.3%) vs. IL2+ (2.2%) cells was more be a FOXP3 gene dosage effect, in that males with an X-linked
evident and all of the cells still remained undivided. At this time, FOXP3 mutation and a complete lack of functional FOXP3
only 0.2% of CD4+ cells were doubly positive for FOXP3 and IL2 succumb to a rapidly fatal lymphoproliferative syndrome. By
expression. This pattern persisted after 96 hours of culture, at which comparison, lack of IL2 as in the IL2 (2/2) mouse leads only to a
time cells had entered the second division. Moreover, as shown in diminution of FOXP3 gene expression, primarily in peripheral T
Figure 5B, after 72 hours of culture when ,30% of the cells had cells, and results in a much slower accumulation of peripheral
divided, the expression of FOXP3 and IL2 remained mutually activated T cells and the lymphoproliferative/autoimmune
exclusive, whether or not the cells had divided. Also, it was syndrome [39]. Moreover, recently the gene region termed insulin
particularly noteworthy that the FOXP3+ cells did not ‘‘suppress’’ dependent diabetes 3 (Idd3), which promotes type 1 diabetes (T1D)
the expression of IL2 by FOXP3- cells. development in the non-obese diabetic mouse, results in a 50%

PLoS ONE | www.plosone.org 4 February 2008 | Volume 3 | Issue 2 | e1581


IL2 and FOXP3 Reciprocity

Figure 3. Antibodies reactive with the IL2Ra (CD25) and IL2 inhibit anti-CD3-induced FOXP3 expression. FOXP3 expression at 24 h in
CD4+ (black bars) and CD8+ (gray bars) cells is shown upon aCD3-activation of PBMCs (t = 0), and after addition of increasing concentrations of (A)
aCD25 and (C) aIL2 at t = 0. Exogenous rIL2 [10 nM] was added where indicated (t = 0). Also, the expression of FOXP3 at 24 h was determined in CD4+
(black bars) and CD8+ (gray bars) cells upon total PBMCs activation together with an addition of increasing concentrations of exogenous rIL2 [0–
10 nM] at t = 0 in the presence of (B) aCD25 [1 mg/ml] and (D) aIL2 [1 mg/ml]. The results are shown as Mean6SEM of 4 independent experiments.
doi:10.1371/journal.pone.0001581.g003

reduction of IL2 production, and this predisposes to T1D [40]. murine T cell FOXP3 expression[47]. Moreover, other cytokines
Idd3 has also been associated with susceptibility to experimental (IL-4, 6, 7, 9, 12, 15, 18, 21) failed to substitute for IL2. It has also
autoimmune encephalomyelitis and autoimmune ovarian dysgen- been reported that murine CD4+CD25+FOXP3+ cells can be
esis induced by neonatal thymectomy [40]. induced in vivo by antigen stimulation of naı̈ve CD4+CD25- T cells
We purposely performed our in vitro stimulations solely with [48], provided IL2 is available [49].
soluble anti-CD3 and total PBMC populations, thus relying on Accordingly, in both mice and humans, it appears that it is
monocytes for presentation of the mAb and for costimulation, possible to convert FOXP3- T cells to FOXP3+ T cells by
which most closely mimics physiological activation via the APC immunological activation. As well, in both species, this conversion
MHC-peptide antigen complex. In this regard, it is noteworthy requires IL2. In this regard, there have been several recent reports
that IL2/IL2R signaling of FOXP3 expression was so obvious, in that focus on the mechanism(s) regulating FOXP3 gene expres-
that with the IL2/IL2R interaction blocked, the TCR/CD3- sion. Mantel et. al. [50] explored the structure and function of the
generated signals and any other cytokines produced in situ, such as human FOXP3 promoter, and provided evidence that there is a
TGFb, could not increase the frequency of FOXP3+ cells. These proximal promoter localized between 2511/+176 bp and that this
observations are entirely consistent with those obtained utilizing region has at least 3 NF-AT response elements (RE), and 3 AP-1
either murine or human purified CD4+ T cells, and it is possible to REs, thereby implicating TCR regulation of FOXP3 expression.
selectively expand these cells with IL2 so that 100% become As well, others have identified a TCR-responsive enhancer in the
FOXP3+ after 5–7 days of culture [35,36]. As the TCR/CD3 first intron of the FOXP3 gene that is dependent upon a cyclic
signaling complex is known to activate members of three AMP response element binding protein/activating transcription
families of transcription factors (i.e. Rel, AP-1 and NF-AT) factor site [51]. By comparison, Zorn and co-workers also
[41,42], while the IL2/IL2R activates STAT5 [43], our data are examined regulation of the human FOXP3 gene, and provided
consistent with the interpretation that FOXP3 expression is evidence that IL2, via STAT5, promotes expression of FOXP3 by
regulated predominantly by STAT5 as has been shown recently binding to a STAT RE located in the first intron of the FOXP3
by others [39,44,45]. gene [45]. Subsequently, Burchill and co-workers used the
Activation of murine T cells in vitro with anti-CD3+anti-CD28 reductionist power of gene deletion experiments in the mouse to
was originally reported not to result in the expression of FOXP3 show convincingly that STAT5 is both necessary and sufficient for
mRNA [27,28,29]. However, subsequently, it was found that FOXP3 expression, and that there are 6 potential STAT5 REs in
TGF-b dramatically promotes the expression of FOXP3 mRNA the gene, 3 in the promoter region and 3 in the first intron [39].
by CD4+ T cells stimulated with anti-CD3/28, while TGF-b Moreover, using chromatin immunoprecipitation (ChIP) assays,
alone has no effect [46,47]. More recently, IL2 was found to be evidence was presented that STAT5 binds to the FOXP3 gene
essential for TGF-b-facilitated induction of anti-CD3 activation of promoter. Similar results have been reported recently by Yao and

PLoS ONE | www.plosone.org 5 February 2008 | Volume 3 | Issue 2 | e1581


IL2 and FOXP3 Reciprocity

Subsequently, the effect of FOXP3 expression vs. FOXP1 and


FOXP2 was examined, and only FOXP3 specifically inhibited
cytokine gene expression (IL2, IL4, IFN-c) by primary murine
CD4+ T cells. Furthermore, FOXP3 was shown to inhibit Rel
family (NF-kB p65 & NFAT) transcriptional activation of cytokine
genes without inhibiting their DNA binding activity [53]. In
addition, they found that FOXP3 physically associates with NF-kB
p65 and NFAT proteins and inhibits their capacity to transacti-
vate. These results were confirmed and extended by others who
showed that FOXP3 is capable of repressing CREB- as well as NF-
kB-dependent transcription [54]. As well, the crystal structure of
FOXP2 complexed with NF-AT was solved, revealing that this
protein complex forms an extensive protein-protein interaction
interface [55]. Moreover, mutations of FOXP3 predicted from this
crystal structure to disrupt the FOXP2/NF-AT complex correlates
with the loss of FOXP3-inhibition of IL2 gene expression.
Additional data have been recently reported on analyses of
FOXP3 target genes by combining ChIP analysis with DNA
arrays. Of interest, using these approaches, one group found that
FOXP3 binds to ,700 genes, and plays a dual role as both a
transcriptional activator and repressor [56]. Another group used a
FOXP3- murine T cell hybridoma transduced with FOXP3 and
found IL2 gene expression to be suppressed only in FOXP3+ cells
[57]. Thus, many FOXP3-regulated genes that encode proteins
associated with the TCR signaling pathway have been identified,
most of which showed suppressed activation when the cells were
TCR-stimulated.
By comparison with the wealth of these data, there have been
only two recent reports regarding possible molecular mechanism(s)
responsible for the putative T-Reg active suppression of IL2
production and resultant proliferation, both of which have invoked
an increase in intracellular cAMP [58,59]. To date, the in vitro T
cell proliferation assays for the capacity of T-Reg cells to suppress
Figure 4. Inhibition of anti-CD3-induced FOXP3 expression by effector T cells have utilized high ratios of T-Regs to effectors, a
combined antibodies reactive with both IL2R (aCD25) and IL2. situation that favors the inhibition of proliferation by the
FOXP3 expression at 24 h is shown in CD4+ (black bars) and CD8+ (gray consumption of the T cell growth factor, IL2. In as much as T
bars) cells upon aCD3-activation of PBMCs and simultaneous addition
of aCD25 and aIL2 (t = 0) at two concentrations (A) [1 mg/ml] and (B)
cell proliferation is dependent upon the concentration of IL2
[10 mg/ml] along with increasing concentrations of rIL2 [0–10 nM]. The available as demonstrated in our earliest studies [1], because
results are shown as Mean6SEM of 4 independent experiments. FOXP3+ cells are restricted in their capacity to produce IL2, but
doi:10.1371/journal.pone.0001581.g004 can consume any IL2 produced by effector cells, decreased IL2-
driven T cell proliferation in the presence of high ratios of
co-workers [44], who also show that STAT3 and STAT5a/b FOXP3+ : FOXP3- cells (e.g. 2:1 to 1:4) would necessarily be
appear to have opposing roles in the regulation of FOXP3. more apparent than real. Thus, aided by new assays to detect the
Given the expression of FOXP3 by TCR/IL2-activated T cells, accumulation of intracellular IL2, rather than its concentration in
what can be expected as to the outcome of the expression of this the surrounding media or its growth promoting effects, we could
gene? Two functions have thus far been attributed to FOXP3, the detect no evidence that FOXP3+ cells can actively suppress IL2
restriction of TCR-signaled cytokine gene expression by FOXP3+ expression by FOXP3- cells.
cells, and the active suppression of cytokine production by It is worthy of emphasis that these findings are entirely
FOXP3- cells mediated by FOXP3+ cells. Our results demonstrate consistent with the recent report of Pandiyan and co-workers
that FOXP3 and IL2 expression are mutually exclusive, consistent [38], who showed that CD4+CD25+FOXP3+ cells did not
with the restriction of IL2 expression by FOXP3+ cells, but suppress effector T cell production of IL2 or proliferation early
indicating that FOXP3+ cells do not actively suppress IL2 after activation by anti-CD3 and anti-CD28, with or without
expression by FOXP3- cells, at least when both FOXP3+ and APCs, thereby indicating that APCs are not essential for T-Reg
FOXP3- cells are present in a mixed population of PBMCs. cell-mediated suppression. Moreover, their data indicate that
If CD4+CD25+FOXP3+ cells cannot actively suppress effector CD4+CD25+FOXP3+ cells consume IL2, which leads inevitably
T cells, how can one account for the phenotypes of the scurfy to cytokine deprivation-induced apoptosis of effector T cells, both
mouse, and those individuals suffering from the XLAAD/IPEX in vitro and in vivo.
syndrome? FOXP3 expression normally functions to down- Thus, our findings are entirely consistent with the capacity of
regulate T cell activation, particularly cytokine gene expression. FOXP3+ T cells to passively suppress ongoing proliferative immune
Actually, in the earliest studies of FOXP3 gene function, it was responses by reducing the concentrations of IL2 available to
found that FOXP3 over-expression in CD4+ T cells attenuates effector cells, both in vitro and in vivo. When the IL2 concentration
activation-induced cytokine production and proliferation [52]. is limiting, IL2R+ cells rapidly cease proliferating, and undergo
Moreover, several forkhead-specific REs adjacent to critical NF- cytokine-withdrawal apoptosis [38]. In vivo this mechanism is
AT REs in the promoters of several cytokine genes were identified. responsible for the retraction of .90% of T cells that expand after

PLoS ONE | www.plosone.org 6 February 2008 | Volume 3 | Issue 2 | e1581


IL2 and FOXP3 Reciprocity

Figure 5. The expression of FOXP3 and IL2 are mutually exclusive. (A) Dot plots monitoring FOXP3 and IL2 expression in CD4+ cells
measured on different days after aCD3-activation of PBMCs (t = 0). (B) FOXP3 and IL2 expression are shown for CD4+ cells in division zero (upper dot
plot) and in the first division (lower dot plot) 72 h after total PBMCs activation (t = 0). Histograms of corresponding CFSE labeled CD4+ cells are shown
in the insets. The percentages of the different populations are also shown for each dot plot. Representative dot plots were chosen from 1 of 4
independent experiments. (C) FOXP3 and IL2 expression are shown after restimulation. PBMCs were activated with anti-CD3+IL2 (10 nM) for
24 hours, harvested and then reactivated with anti-CD3+anti-CD28 in the presence of Brefeldin-A for 6 hours. Representative dot blots were chosen
from 1 of 5 independent experiments.
doi:10.1371/journal.pone.0001581.g005

PLoS ONE | www.plosone.org 7 February 2008 | Volume 3 | Issue 2 | e1581


IL2 and FOXP3 Reciprocity

an initial antigen stimulation [60,61]. This same mechanism could Proliferation and Cytokine Production
explain why attenuation of FOXP3 expression by genetic manip- In some experiments, freshly isolated PBMCs were first labeled
ulation eradicates the suppressive function but not the restrictive with 5 mM 5- (and 6-) carboxyfluorescein diacetate succinimidyl
function of FOXP3 both in vitro and in vivo, in that the FOXP3- ester (CFSE) (Molecular Probes) prior to culture. PBMCs were
attenuated cells only express low levels of IL2 receptors [62]. then activated (anti-CD3) and cultured for 96 h. For analysis of
From all of these various experimental approaches, the intracellular cytokine production in cultured, CFSE-labeled and
emerging picture is that FOXP3 expression in peripheral mature activated PBMCs, Brefeldin A (10 mg mL21, Sigma) was added for
T cells both in mice as well as humans is dependent upon TCR/ the final 6–18 h prior to cytokine staining daily for at least 4 days
CD28 triggering of IL2. Moreover, the most consistent conse- after activation. Anti-CD28 (10 mg mL21, BD Biosciences) was
quence of IL2-induced FOXP3 expression is the restriction of IL2 used for co-stimulation where indicated.
gene expression in response to TCR activation as well as
reactivation. Therefore, all of these data indicate that FOXP3
Monoclonal Antibodies (mAbs)
plays a key role in the feedback down-regulation of T cell
The following mAbs were used all reactive with human
activation and proliferation via the regulation of IL2 gene
molecules: surface mAbs ; Ms IgG1,k FITC, CD25 FITC, CD4
expression, thereby functioning normally to dampen immune
responses. This interpretation is entirely consistent with the PerCp, CD8 PerCp, CD8 APC; intracellular mAb; IL2 PE (BD
hyperproliferative/autoimmune phenotypes of the IL2 (2/2) Biosciences); intranuclear antibodies reactive with FOXP3 (clones
and scurfy mice as well as XLAAD/IPEX patients. PCH101 and 236A/E7 from eBioscience; clone 259D from
However, no evidence could be found for an active suppressive BioLegend) PE or APC, Rat Isotype IgG2a control (eBiocsience),
effect of FOXP3+ cells on FOXP3- cell IL2 expression. Our data Ms IgG1, k (BioLegend). The specificity of the monoclonal
thus question the relevance of FOXP3 as an exclusive marker for antibody reactivity with FOXP3 was confirmed using FOXP3-
T-Regs, as has been emphasized recently by others [63], as well as transfected 293 cells kindly provided by Steven Jacobson (NINDS).
the role of FOXP3 in T-Reg-mediated active suppression. Accord- eBioscience mAbs (clones PCH101 and 236A/E7), Biolegend
ingly, further experiments will be required to clarify the postulated clone 259D, and Abcam polyclonal antibodies (ab4728), showed
active vs. passive suppressive roles in regulating IL2 availability, as similar patterns and specific reactivity with FOXP3 using FOXP3-
compared with the demonstrable restrictive role of FOXP3 in transfected 293 cells and human PBMCs.
regulating IL2 expression. As well, whether other mechanisms are
responsible for active suppression of effector cell proliferation Flow Cytometry
remains to be investigated. However, it is clear that the IL2 Four-color flow cytometry and the eBioscience PE and APC
regulation of FOXP3 and the FOXP3 regulation of IL2 will now FOXP3 staining sets with modified eBioscience staining protocols
assume a central role in many diseases of the immune system. were used. Single cell suspensions of lymphocytes were stained first
Since the point of control over immune reactivity occurs at IL2 for surface molecules for 30 minutes in the dark (room
gene expression, it is logical that defects in the IL2/FOXP3 negative temperature), then washed once with Washing Buffer (1X PBS
feedback loop may lead to a preponderance of activating signals and (Cellgro) with 0.5% FBS (Gemini Bio-Products) and 0.1% NaN3).
autoimmunity. Conversely, if the pivotal point of IL2 gene regulation The cell pellet was resuspended and 0.5 ml of freshly prepared
by FOXP3 is circumvented, for example by autonomous signaling eBioscience Fixation/Permeabilization Buffer was added to each
via persistently active JAK3/STAT5, then malignancy could result, sample and samples were incubated 45 minutes in the dark (4uC).
as shown experimentally by over expression of STAT5 [64]. The cells were washed once with Washing Buffer and then washed
The therapeutic implications of our findings are obvious. In again by adding 1X Permeabilization Buffer; anti-human FOXP3
those situations where it can be documented that there is a or Isotype Rat IgG2a control antibody (and intracellular
deficiency in the IL2/FOXP3 negative feedback loop, such as in antibodies when stained) was added and samples were incubated
NOD mice, IL2 therapy may be beneficial. Moreover, blockade of for 30 minutes in the dark (room temperature). The cells were
the negative feedback loop could be used to enhance immune then washed with Washing Buffer, acquired on a FACSCalibur
responses, for example for persistent infections, as anti-cancer flow cytometer, and the data were analyzed using FlowJo software
therapy, and to enhance the efficacy of vaccines. Finally, FOXP3+ (Tree Star) using 2% Contour plots to set quadrants.
T cells could be used as adoptive therapy to dampen unwanted
immune responses, not by actively suppressing activated effector Statistical Analysis
cells, but by passively removing IL2.
Nonparametric Wilcoxon matched-pairs signed-rank test was
performed in the analysis of mean differences in FOXP3
Materials and Methods expression between freshly isolated and anti-CD3-activated
Peripheral Blood Mononuclear Cell (PBMC) Isolation, CD4+ and CD8+ T subsets for 24 h. P values of ,0.05 were
Culture and Activation considered statistically significant.
PBMCs from normal volunteers were isolated from whole blood
using centrifugation over a Ficoll-Hypaque density gradient. Total Supporting Information
PBMCs were cultured in 12-well plates (BDLabware) (36106
cells/mL), and were activated with anti-CD3 (OKT3, Ortho Figure S1 Other cc-chain cytokines have no effect on anti-CD3-
Biotech, 1:800,000 dilution) in complete cell culture media (RPMI induced FOXP3 expression. FOXP3 expression at 24 h is shown
1640 with L-glutamine (Cellgro), supplemented with 10% FBS, in CD4+ (black bars) and CD8+ (gray bars) cells upon aCD3-
100 U/mL Penicillin/Streptomycin (both Gemini Bio-Products) activation of PBMCs (t = 0) and simultaneous addition of (A) aIL4
and 2 mM 2-ME (Sigma-Aldrich). The following antibodies were and aIL15 or (B) aIL7 and aIL21R at concentrations of [1 mg/ml]
added simultaneously, when indicated: anti-CD25, anti-IL2, anti- and [10 mg/ml] along with rIL2 [10 nM] where indicated. Data
IL4, anti-IL9, anti-IL15 and anti-IL21R antibodies (all R&D shown represent the mean6SEM of 4 separate experiments.
Systems) and human rIL-2 (Amgen). All cells were cultured in a Found at: doi:10.1371/journal.pone.0001581.s001 (2.86 MB
37uC humidified incubator with 5% CO2. DOC)

PLoS ONE | www.plosone.org 8 February 2008 | Volume 3 | Issue 2 | e1581


IL2 and FOXP3 Reciprocity

Table S1 Reactivation of PBMCs with anti-CD3+anti-CD28, Acknowledgments


followed by monitoring for FOXP3 and IL2 expression. PBMCs
were activated with anti-CD3+IL2 (10 nM) for 24 hours, then We are grateful for helpful comments on the manuscript from the following
people: Michael Farrar, Anjana Rao, Frank Ruscetti, Alexander Rudensky,
harvested and cultured without or with restimulation by anti-
Shimon Sakaguchi, Ethan Shevach, and Kenneth Tung.
CD3+anti-CD28 for 6 hours in the presence of Brefeldin-A. The
percentage of CD4+ T cells positive for FOXP3, IL2 and both
FOXP3+IL2 are listed from 5 individuals, together with the Author Contributions
mean6SEM. Conceived and designed the experiments: KS ZP. Performed the
Found at: doi:10.1371/journal.pone.0001581.s002 (0.02 MB experiments: ZP. Analyzed the data: ZP. Wrote the paper: KS ZP.
DOC)

References
1. Gillis S, Ferm MM, Ou W, Smith KA (1978) T cell growth factor: parameters of 25. Bennet C, Christie J, Ramsdell M, Brunkow M, Ferguson P, et al. (2001) The
production and a quantitative microassay for activity. J Immunol 120: immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome
2027–2032. (IPEX) is caused by mutations of FOXP3. Nature Genetics 27: 20–21.
2. Smith KA (1980) T-cell growth factor. Immunol Rev 51: 337–357. 26. Wildin R, Ramsdell F, Peake J, Faravelli F, Casanova J-L, et al. (2001) X-linked
3. Smith KA (1989) The interleukin 2 receptor. Annu Rev Cell Biol 5: 397–425. neonatal diabetes mellitus, enteropathy and endocrinopathy syndrome is the
4. Schorle H, Holtschke T, Hunig T, Schimpl A, Horak I (1991) Development and human equivalent of mouse scurfy. Nature Genetics 27: 18–20.
function of T cells in mice rendered interleukin-2 deficient by gene targeting. 27. Fontenot J, Gavin M, Rudensky A (2003) Foxp3 programs the development and
Nature 352: 621–624. function of CD4+CD25+ regulatory T cells. Nature Immunol 4: 330–336.
5. Sadlack B, Lohler J, Schorle H, Klebb G, Haber H, et al. (1995) Generalized 28. Hori S, Nomura T, Sakaguchi S (2003) Control of regulatory T cell development
autoimmune disease in interleukin-2-deficient mice is triggered by an by the transcription factor FOXP3. Science 299: 1057–1061.
uncontrolled activation and proliferation of CD4+ T cells. Eur J Immunol 25: 29. Khattri R, Cox T, Yasayko S-A, Ramsdell F (2003) An essential role for scurfin
3053–3059. in CD4+CD25+ T regulatory cells. Nature Immunol 4: 337–342.
6. Sadlack B, Merz H, Schorle H, Schimpl A, Feller AC, et al. (1993) Ulcerative 30. Walker M, Kasprowicz D, Gersuk V, Benard A, Landeghen J, et al. (2003)
colitis-like disease in mice with a disrupted interleukin-2 gene [see comments]. Induction of Foxp3 and acquisition of T regulatory activity by stimulated human
Cell 75: 253–261. CD4+CD25- T cells. J Clin Invest 112: 1437–1443.
7. Horak I, Lohler J, Ma A, Smith K (1995) Interleukin-2 deficient mice: A new 31. Morgan M, van Bilsen J, Bakker A, Heemskerk B, Schilham M, et al. (2005)
model to study autoimmunity and self-tolerance. Immunol Rev 148: 35–44. Expression of FOXP3 mRNA is not confined to CD4+CD25+ T regulatory cells
8. Klebb G, Autenrieth IB, Haber H, Gillert E, Sadlack B, et al. (1996) Interleukin- in humans. Hum Immunol 66: 13–20.
2 Is Indispensable for Development of Immunological Self-Tolerance. Clinical 32. Wang J, Ioan-Facsinay A, van der Voort E, Huizinga T, Toes R (2007)
Immunology and Immunopathology 81: 282–286. Transient expression of FOXP3 in human activated nonregulatory T cells.
9. Russell W, Russell L, Gower J (1959) Exceptional inheritance of a sex-linked Eur J Immunol 37: 129–138.
gene in the mouse explained on the basis that the X/O sex-chromosome 33. Gavin M, Torgerson T, Houston E, DeRoos P, Ho H, et al. (2006) Single-cell
constitution is female. PNAS 45: 554–560. analysis of normal and FOXP3-mutant human T cells: FOXP3 expression
10. Godfrey V, Wilkinson J, Rinchik E, Russell L (1991) Fatal lymphoreticular without regulatory T cell development. PNAS 103: 6659–6664.
disease in the scurfy (sf) mouse requires T cells that mature in a sf thymic 34. Ahmadzadeh M, Antony P, Rosenberg S (2007) IL-2 and IL-15 each mediate de
environment: Potential model for thymic education. PNAS 88: 5528–5532. novo induction of FOXP3 expression in human tumor antigen-specific CD8 T
11. Godfrey V, Wilkinson J, Russell L (1991) X-linked lymphoreticular disease in the cells. J Immunother 30: 294–302.
scurfy (sf) mutant mouse. Am J Path 138: 1379–1387. 35. Allan S, Crome S, Crellin N, Passerini L, Steiner T, et al. (2007) Activation-
12. Godfrey V, Rouse B, Wilkinson J (1994) Transplantation of a T cell-mediated induced FOXP3 in human T effector cells does not suppress proliferation or
lymphoreticular disease from the scurfy (sf) mouse. Am J Path 145: 281–286. cytokine production. Int Immunol 19: 345–354.
13. Blair P, Bultman S, Haas J, Rouse B, Wilkinson J, et al. (1994) CD4+CD8- T 36. Tran DQ, Ramsey H, Shevach EM (2007) Induction of FOXP3 expression in
cells are the effector cells in disease pathogenesis in the scurfy (sf) mouse. naive human CD4+FOXP3 T cells by T-cell receptor stimulation is
J immunol 153: 3764–3774. transforming growth factor-{beta} dependent but does not confer a regulatory
14. Kanangat S, Blair P, Reddy R, Deheshia M, Godfrey V, et al. (1996) Disease in phenotype. Blood 110: 2983–2990.
the scurfy (sf) mouse is associated with overexpression of cytokine genes. 37. Villarino A, Tato C, Stumhofer J, Yao Z, Cui Y, et al. (2007) Helper T cell IL-2
E J Immunol 26: 161–165. production is limited by negative feedback and STAT-dependent cytokine
15. Clark L, Appleby M, Brunkow M, Wilkinson J, Ziegler S, et al. (1999) Cellular signals. J Exp Med 204: 65–71.
and molecular characterization of the scurfy mouse mutant. J Immunol 162: 38. Pandiyan P, Zheng L, Ishihara S, Reed S, Lenardo M (2007)
2564–2554. CD4+CD25+Foxp3+ regulatory T cells induce cytokine deprivation-mediated
16. Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M (1995) Immunologic self- apoptosis of effector CD4+ T cells. Nat Immunol Epub ahead of print (Nov. 4,
tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains 2007). pp 1–10.
(CD25). Breakdown of a single mechanism of self-tolerance causes various 39. Burchill M, Yang J, Vogtenhuber C, Blazar B, Farrar M (2007) IL-2 receptor
autoimmune diseases. J Immunol 155: 1151–1164. beta-dependent STAT5 activation is required for the development of FOXP3+
17. Asano M, Toda M, Sakaguchi N, Sakaguchi S (1996) Autoimmune disease as a regulatory T cells. J Immunol 178: 280–290.
consequence of developmental abnormality of a T cell subpopulation. J Exp 40. Yamanouchi J, Rainbow D, Serra P, Howlett S, Hunter K, et al. (2007)
Med 184: 387–396. Interleukin-2 gene variation impairs regulatory T cell function and causes
18. Suri-Payer E, Kehn P, Cheever A, Shevach E (1996) Pathogenesis of post- autoimmunity. Nat Gen 39: 329–337.
thymectomy autoimmune gastritis: identification of anti-H/K adenosine 41. Garrity PA, Chen D, Rothenberg EV, Wold BJ (1994) Interleukin 2
triphosphatase-reactive T cells. J Immunol 157: 1799–1805. transcription is regulated in vivo at the level of coordinated binding of both
19. Suri-Payer E, Amar A, Thornton A, Shevach EM (1998) CD4+CD25+ T cells constitutive and regulated factors. Mol Cell Bio 14: 2159–2169.
inhibit both the induction and effector function of autoreactive T cells and 42. Rothenberg EV, Ward SB (1996) A dynamic assembly of diverse transcription
represent a unique lineage of immunoregulatory T cells. J Immunol 160: factors integrates activation and cell-type information for interleukin 2 gene
1212–1218. regulation. Proc Natl Acad Sci U S A 93: 9358–9365.
20. Thornton AM, Shevach EM (1998) CD4+CD25+ immunoregulatory T cells 43. Beadling C, Guschin D, Witthuhn B, Ziemiecki A, Ihle J, et al. (1994) Activation
suppress polyclonal T cell activation in vitro by inhibiting interleukin 2 of JAK kinases and STAT proteins by interleukin-2 and interferon alpha, but
production. J Exp Med 188: 287–296. not the T cell antigen receptor, in human T lymphocytes. EMBO J 13:
21. Smith KA (1988) Interleukin-2: inception, impact, and implications. Science p5605–5615.
240: 1169–1176. 44. Yao Z, Kanno Y, Kerenyi M, Stephens G, Durant L, et al. (2007)
22. Brunkow M, Jeffrey E, Hjerrild K, Paeper B, Clark L, et al. (2001) Disruption of Nonredundant roles for Stat5a/b in directly regulating FOXP3. Blood 109:
a new forkhead/winked-helix protein, scurfin, results in the fatal lymphopro- 4368–4375.
lierative disorder of the scurfy mouse. Nature Genetics 27: 68–73. 45. Zorn E, Nelson E, Mohseni M, Porcheray F, Kim H, et al. (2006) IL-2 regulates
23. Schubert L, Jeffrey E, Zhang Y, Ranmsdell F, Ziegler S (2001) Scurfin (FOXP3) FOXP3 expression in human CD4+CD25+ regulatory T cells through a STAT-
acts as a repressor of transcription and regulates T cell activation. J Biol Chem dependent mechanism and induces the expansion of these cells in vivo. Blood
276: 37672–37679. 108: 1571–1579.
24. Chatlia T, Blaeser F, Ho N, Lederman H, Voularopoulos C, et al. (2001) JM2, 46. Chen W, Jin W, Hardegen N, Li K, Li L, et al. (2003) Conversion of peripheral
encoding a forkhead-related protein, is mutated in X-linked autoimmunity- CD4+CD25- naive T cells to CD4+CD25+ regulatory T cells by TGF-beta
allergic disregulation syndrome. J Clin Invest 106: R75–R81. induction of transcription factor FOXP3. J Exp Med 198: 1875–1886.

PLoS ONE | www.plosone.org 9 February 2008 | Volume 3 | Issue 2 | e1581


IL2 and FOXP3 Reciprocity

47. Davidson T, Dipaolo R, Anderson J, Shevach E (2007) IL-2 is essential for TGF- 56. Zheng Y, Josefowicz S, Kas A, Chu T-T, Gavin M, et al. (2007) Genome-wide
beta-mediated induction of Foxp3+ T regulatory cells. J Immunol 178: analysis of Foxp3 target genes in developing and mature regulatory T cells.
4022–4026. Nature.
48. Apostolou I, Von Boehmer H (2004) In vivo instruction of suppressor 57. Marson A, Kretschmer K, Frampton G, Jacobson E, Polansky J, et al. (2007)
commitment in naive T cells. J Exp Med 199: 1401–1408. Foxp3 occupnacy and regulation of key target genes during T cell stimulation.
49. Kretschmer K, Apoustolou I, Hawiger D, Khazaie K, Nussenzweig M, et al. Nature.
(2005) Inducing and expanding regulatory T cell populations by foreign antigen. 58. Bopp T, Becker C, Klein M, Klein-Hessling S, Palmetshofer A, et al. (2007)
Nature Immunology 6: 1219–1227. Cyclic adenosine monophosphate is a key component of regulatory T cell
50. Mantel P-Y, Ouaked N, Ruckert B, Karagiannidis C, Welz R, et al. (2006) mediated suppression. J Exp Med 204: 1303–1310.
Molecular mechanisms underlying FOXP3 induction in human T cells. 59. Deaglio S, Dwyer KM, Gao W, Friedman D, Usheva A, et al. (2007) Adenosine
J Immunol 176: 3593–3602. generation catalyzed by CD39 and CD73 expressed on regulatory T cells
51. Kim H-P, Leonard W (2007) CREB/ATF-dependent T cell receptor-induced mediates immune suppression. J Exp Med 204: 1257–1265.
FoxP3 gene expression: a role for DNA methylation. J Exp Med 204: 1543–1551. 60. Kuroda K, Yagi J, Imanishi K, Yan XJ, Li XY, et al. (1996) Implantation of IL-
52. Khattri R, Kasprowicz D, Cox T, Mortrud M, Appleby M, et al. (2001) The 2-containing osmotic pump prolongs the survival of superantigen-reactive T cells
amount of scurfin protein determines peripheral T cell number and expanded in mice injected with bacterial superantigen. J Immunol 157:
responsiveness. J Immunol 167: 6312–6320. 1422–1431.
53. Bettelli E, Dastrange M, Oukka M (2005) Foxp3 interacts with nuclear factor of 61. Blattman J, Grayson J, Wherry E, Smith K, Ahmed R (2003) Therapeutic use of
activated T cells and NF-kB to repress cytokine gene expression and effector IL2 to enhance antiviral T cell responses in vivo. Nature Med 5: 540–547.
functions of T cells. PNAS 102: 5138–5143. 62. Wan Y, Flavell R (2007) Regulatory T-cell functions are subverted and
54. Grant C, Oh U, fugo K, Takenouchi N, Griffith C, et al. (2006) Foxp3 represses converted owing to attenuated FOXP3 expression. Nature 445: 766–770.
retroviral transcription by targeting both NF-kB and CREB pathways. PLoS 63. Ziegler S (2007) FOXP3: Not just for regulatory T cells anymore. Eur J Immunol
Pathogens 2: 303–314. 37: 21–23.
55. Wu Y, Borde M, Heissmeyer V, Feuerer M, Lapan A, et al. (2006) FOXP3 64. Kelly J, Spolski R, Kovanen P, Suzuki T, Bollenbacher J, et al. (2003) Stat5
controls regulatory T cell function through cooperation with NFAT. Cell 126: synergizes with T cell receptor/antigen stimulation in the development of
375–387. lymphoblastic lymphoma. J Exp Med 198: 79–89.

PLoS ONE | www.plosone.org 10 February 2008 | Volume 3 | Issue 2 | e1581

You might also like