ArticlePDF AvailableLiterature Review

Interleukin-2 Receptor Signaling in Regulatory T Cell Development and Homeostasis

Authors:

Abstract and Figures

Interleukin-2 (IL2) was initially identified from supernatants of activated lymphocytes over 30 years ago. In the ensuing 15 years, the cDNAs for both IL2 and the three chains of the interleukin-2 receptor (IL2R) were cloned. Subsequently, many of the downstream biochemical pathways activated by the IL2 receptor complex were identified and the structure of IL2 bound to this tripartite receptor complex was solved. Thus, we now have a very good understanding of how each chain contributes to high affinity IL2 binding and signal transduction. In contrast, over the past 30 years the role that IL2 plays in regulating lymphocyte function has involved many surprising twists and turns. For example, IL2 has been shown, paradoxically, to regulate both lymphocyte proliferation and lymphocyte death. In this review, we briefly outline the original findings suggesting a role for IL2 as a T cell growth factor, as well as subsequent studies pointing to its function as an initiator of activation-induced cell death, but then focus on the newly appreciated role for IL2 and IL2R signaling in the development and homeostasis of regulatory T cells.
Content may be subject to copyright.
Interleukin-2 Receptor Signaling in Regulatory T Cell
Development and Homeostasis
Matthew A. Burchill, Jianying Yang, Kieng B. Vang, and Michael A. Farrar1
Center for Immunology, The Cancer Center, Department of Laboratory Medicine and Pathology,
University of Minnesota, 312 Church Street SE, 6-116 Nils Hasselmo Hall, Minneapolis, MN 55455
USA
Abstract
Interleukin-2 (IL2) was initially identified from supernatants of activated lymphocytes over 30 years
ago [1]. In the ensuing 15 years, the cDNAs for both IL2 and the three chains of the interleukin-2
receptor (IL2R) were cloned [2–7]. Subsequently, many of the downstream biochemical pathways
activated by the IL2 receptor complex were identified [8] and the structure of IL2 bound to this
tripartite receptor complex was solved [9]. Thus, we now have a very good understanding of how
each chain contributes to high affinity IL2 binding and signal transduction [10,11]. In contrast, over
the past 30 years the role that IL2 plays in regulating lymphocyte function has involved many
surprising twists and turns. For example, IL2 has been shown, paradoxically, to regulate both
lymphocyte proliferation and lymphocyte death. In this review, we briefly outline the original
findings suggesting a role for IL2 as a T cell growth factor, as well as subsequent studies pointing
to its function as an initiator of Activation-induced Cell Death, but then focus on the newly
appreciated role for IL2 and IL2R signaling in the development and homeostasis of regulatory T
cells.
Interleukin-2 plays an important role in multiple aspects of T cell biology. Binding of IL2 to
its receptor was initially demonstrated to be critical for inducing the proliferation of T cells in
vitro [12]. Subsequent studies demonstrated that IL2R signaling leads to the activation of many
genes associated with cell proliferation such as c-myc and fos [13]. These findings led to the
idea that signaling via the IL2R complex would be critical for mounting effective immune
responses in vivo and established IL2 as a T cell growth factor. However, the concept of IL2
as a critical factor required for T cell proliferation was essentially overturned in the early 1990s
when mice lacking the IL2 gene were first developed. The initial characterization of these mice
demonstrated that in vitro proliferation of ConA-stimulated IL2/ T cells was impaired, but
much more modestly than one would have predicted, suggesting the presence of alternative T
cell growth factors [14]. It also was readily apparent that in vivo, T cell proliferation was clearly
not impaired; IL2/ mice exhibited massive lympho-proliferation resulting in splenomegaly
and lymphoadenopathy. In addition, IL2/ mice rapidly developed signs of autoimmunity
including, most prominently, a disease resembling ulcerative colitis [15]. These autoimmune
manifestations ultimately resulted in death of 100% of IL2/ mice between 12–25 weeks of
age. The reasons for this surprising onset of autoimmunity were at first unclear, but one
potential explanation was provided by Michael Leonardo who demonstrated that IL2 plays an
important role in sensitizing T cells for Activation-induced cell death [16]. When mice lacking
the IL2Rα or IL2Rβ chain were generated, they too exhibited multi-organ autoimmune disease
1Address Correspondence to M.A.F., farra005@tc.umn.edu, 612-625-0401 (tel.), 612-625-2199 (fax).
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers
we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting
proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could
affect the content, and all legal disclaimers that apply to the journal pertain.
NIH Public Access
Author Manuscript
Immunol Lett. Author manuscript; available in PMC 2008 November 30.
Published in final edited form as:
Immunol Lett. 2007 November 30; 114(1): 1–8.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
resulting in early death [17,18]. Moreover, examination of IL2Rα/ mice demonstrated a
significant defect in activation-induced cell death [17] suggesting that the primary mechanism
that accounted for deregulated T cell homeostasis, and the onset of autoimmunity, was a failure
of IL2-induced T cell death.
The data suggesting that IL2 prevents autoimmunity via an effect on activation-induced cell
death was compelling. However, new discoveries regarding a novel subset of CD4+ T cells
began to shed doubt on whether the failure of IL2-induced T cell death was sufficient to account
for the defects seen in IL2 deficient mice. Specifically, in the mid 1990s Sakaguchi and
colleagues identified a population of CD4+ T cells that expressed high levels of CD25 and
potently suppressed the proliferation of activated T cells in vitro. Moreover, they found that
injection of CD4+CD25 T cells into T-cell depleted host mice rapidly induced autoimmune
disease; importantly, disease could be prevented or even reversed by the transfer of CD4+CD25
+ T cells [19]. These studies established CD4+CD25+ T cells as the elusive suppressor T cells,
more commonly referred to now as regulatory T cells (or Tregs), that were initially proposed
over 35 years ago [20,21]. The discovery of this novel T cell subset characterized by high level
IL2R expression, in conjunction with the observation that IL2/ and IL2Rβ/ mice lacked
CD4+CD25+ T cells, suggested that IL2-dependent signals were required for either Treg
development, homeostasis, or function. Subsequent studies aimed at identifying genes involved
in autoimmune disease have found distinct alleles of IL2 or IL2Rα associated with
autoimmunity and defects in Tregs. For example, the insulin-dependent diabetes susceptibility
locus 3 (idd3) in the NOD mouse has been shown to map to the IL2 gene; importantly, Tregs
from susceptible mice were found to have decreased regulatory T cell function [22]. Likewise,
the idd4 locus in NOD mice has been suggested to map to alterations in the downstream IL2
effector STAT5 [23]. Finally, studies of human diabetes, multiple sclerosis and Graves’ Disease
have been mapped to distinct alleles for IL2Rα [24–26]. These observations led to an explosive
interest in regulatory T cells resulting in significant insights to all aspects of Treg biology. In
the ensuing few pages we will specifically focus on the role IL2 plays in the differentiation
and homeostasis of so-called natural Tregs that develop in the thymus.
Role of IL2 in the Development of CD4+CD25+Foxp3+Tregs
Seminal studies from three separate labs eventually established that the critical defect in IL2-
or IL2R-deficient mice resulted from the absence of functional Tregs [27–30]. Compellingly,
Malek and colleagues demonstrated that transfer of limited numbers of CD4+CD25+ T cells
into IL2Rβ/ mice prevented autoimmune disease. These findings clearly demonstrated that
the defect in IL2Rβ/ mice was not cell autonomous, but rather due to the absence of a
functional Treg population [28]. These studies pointed to a critical role for the IL2R in Treg
biology. However, whether the effect of the IL2R was on Treg development, homeostasis, or
function remained unclear and has been quite controversial.
Malek and colleagues initially proposed that the IL2R was uniquely required for Treg
development. Specifically, they developed a transgenic mouse model in which transcription
of the IL2Rβ-chain was driven by the lck-proximal promoter, resulting in a thymic-restricted
expression pattern. Crossing this transgene onto the IL2Rβ/ background restored a
population of CD4+CD25+ T cells in the thymus and periphery that suppressed
lymphoproliferation and the lethal autoimmune disease normally observed in IL2Rβ/ mice
[31]. These findings suggested that IL2Rβ-dependent signals were required for Treg
development in the thymus, but were dispensable for their homeostasis and function in the
periphery. Subsequent studies in which neutralizing antibodies to IL2 were injected into
neonatal mice demonstrated a reduction in numbers of CD4+CD25+ Tregs in the thymus
[32], further supporting a role for IL2 in Treg development. In contrast, work by LaFaille and
colleagues demonstrated that both splenocytes and purified CD4+CD8 thymocytes from IL2
Burchill et al. Page 2
Immunol Lett. Author manuscript; available in PMC 2008 November 30.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
/ mice could suppress the induction of disease in a mouse model of EAE [33]. This effect
was not observed when CD4+ T cells were obtained from IL2Rα/ mice. These studies
indicated that there is a population of CD4+ T cells in both the thymus and spleen of IL2/
mice that can exert regulatory activity when IL2 is restored. The initial explanation for these
findings was that IL2 was required for Treg function. However, an alternative possibility was
that IL2 was required for Treg homeostasis and that potentially reduced numbers of Tregs
found in IL2/ mice were rapidly expanded following transfer into IL2-competent host mice.
Thus, early studies on the role of IL2 and the IL2R in Treg development led to apparently
contradictory findings.
A major problem with the initial studies on the role of IL2 in Treg development was the use
of CD25 as a surrogate marker for this cell population. Although relatively selective, CD25 is
also upregulated on activated effector T cells [34]. Moreover, it was not clear whether all Tregs
would be found within the CD25+ subset of CD4+ T cells. To more accurately dissect the role
of IL2 in Treg biology, a unique marker was needed that could be utilized to track developing
regulatory T cells in the thymus and mature Tregs in the periphery. Recent pioneering work
by several groups identified a member of the forkhead-box family of transcription factors called
Foxp3 which fulfills such a role. Specifically, the naturally occurring scurfy mouse mutant,
which develops lethal multi-organ autoimmune disease, was found to have mutations in the
foxp3 gene that were predicted to disrupt Foxp3 function; restoration of wild type Foxp3 in
these mice prevented disease [35]. Likewise, the genetic defect in human patients suffering
from the autoimmune disease IPEX (immune dysregulation, polyendocrinopathy, enteropathy,
X-linked) were found to stem from mutations in the human foxp3 gene [36,37]. Subsequent
studies by the Ramsdell, Rudensky and Sakaguchi labs independently demonstrated that Foxp3
was both necessary and sufficient for Treg development [38–40]. Importantly, these studies
also established Foxp3 as a unique marker for Tregs in the mouse. More recently, the Rudensky
and Flavell labs generated mouse models in which cells that express Foxp3 can be directly
identified using the fluorescent proteins GFP or DsRed respectively [41,42]. Using these mouse
models and recent advances in intracellular flow cytometry for Foxp3, the controversial role
of IL2 and IL2R signaling in Treg development and homeostasis is beginning to be clarified.
Using a Foxp3-GFP fusion knock-in mouse model Fontenot et al observed that CD4+Foxp3+
cells were clearly present in IL2/ mice, although they failed to express CD25. Identical
results were obtained in IL2Rα/ mice. In both IL2/ and IL2Rα/ mice, numbers of thymic
Tregs in young animals were slightly reduced (~2-fold) while peripheral Tregs were present
in relatively normal numbers [43]. Similar conclusions were simultaneously reached by D’Cruz
and Klein using a transgenic TCR system and intracellular staining for Foxp3 protein. In these
studies, they found that thymic selection into the Treg lineage was not impaired in IL2/ mice,
but the peripheral maintenance of these cells was dramatically affected [44]. Although the
studies by Fontenot et al and D’Cruz and Klein appeared to contradict the original findings of
Malek and colleagues regarding the role of IL2Rβ in Treg development, more recent work
examining Tregs in IL2Rβ/ mice appears to have resolved this discrepancy. Specifically,
using intracellular staining for Foxp3 we too have found that young IL2/ and IL2Rα/ mice
have relatively normal numbers of thymic and peripheral Tregs. In contrast, mice in which
IL2Rβ signaling is disrupted have a more dramatic defect in the population of regulatory T
cells in the thymus and secondary lymphoid organs [45]. Similar results using IL2Rβ/ mice
have recently been obtained by Ziegler and colleagues [46]. To examine this in more detail,
we generated IL2/ x IL15/ mice and observed that they had a comparable decrease in
regulatory T cell numbers to that seen in IL2Rβ/ mice [45]. These results demonstrate that
IL2 and IL15 act as redundant factors, which are both capable of regulating Treg development
via interactions involving the IL2Rβ chain. Importantly, these results provide an explanation
for the earlier apparently discrepant findings of Malek and Lafaille. Namely, IL2 by itself is
not required for Treg development due to the redundant role of IL15. However, the IL2Rβ
Burchill et al. Page 3
Immunol Lett. Author manuscript; available in PMC 2008 November 30.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
chain is required for efficient Treg development as it is a critical component of both the IL2
and IL15 receptors. Finally, it is important to point out that other γc-dependent cytokines, such
as IL7, may allow for inefficient Treg development. This is based on the observation that while
IL2Rβ/ mice show a clear reduction in Tregs in both the thymus and peripheral lymphoid
organs, a residual population of these cells can still be seen. In contrast, γc/ mice are devoid
of Tregs [43,45]. Taken together, these studies illustrate a predominant role for the IL2Rβ chain
in driving the development of Tregs.
Role of IL2 in the Homeostasis of CD4+Foxp3+ Tregs
The initial findings that thymic restricted expression of the IL2Rβ chain was sufficient to
restore peripheral Treg numbers suggested that IL2Rβ-dependent signals might not be required
for homeostasis of peripheral Tregs [31]. A potential caveat to these studies was the observation
that the IL2Rβ transgene was weakly expressed in peripheral T cells. IL2Rβ expression levels
were sufficient for the induction of very transient STAT5 activation but, importantly, did not
induce clear biological responses [47]. Perhaps more tellingly, when regulatory T cells from
these mice were transferred into IL2Rβ/ mice (which lack Tregs but do produce IL2) they
did not survive nor did they prevent autoimmunity [28]. Subsequent studies found that mature
Tregs in mice expressing the thymic-restricted IL2Rβ chain proliferated poorly when compared
to WT Tregs. Mixed bone marrow chimera experiments, in which the ability of cells expressing
the thymic-restricted IL2Rβ chain versus WT cells were compared, were also carried out. In
those studies, cells expressing the thymic restricted IL2Rβ chain contributed well to the thymic
Treg compartment but were not effective at repopulating peripheral lymphoid organs when
compared to WT Tregs [48]. Taken together, these studies suggested that IL2Rβ-dependent
signals are required for homeostasis of Tregs as well.
A remaining question is whether IL2, as opposed to the IL2R (which can bind both IL2 and
IL15), is also required for Treg homeostasis. As mentioned above, relatively normal numbers
of Tregs are found in both IL2/ and IL2Rα/ mice indicating that IL2 may not be critical
for Treg homeostasis [43–45]. In addition, purified Tregs injected into rag2/ mice underwent
homeostatic proliferation that was not affected by IL2 neutralization, indicating that under
lymphopenic conditions IL2 is not critical for Treg homeostasis [49]. However, it is important
to note that Tregs do disappear in older IL2/ and IL2Rα/ mice (>8 weeks of age).
Furthermore, gene microarray studies comparing Tregs from WT and IL2/ mice suggested
that IL2 plays a predominant role in regulating metabolic activity and survival of Tregs [43].
Supporting these findings, Setoguchi et al carried out studies in which they injected Balb/c
mice with IL2 neutralizing antibodies. In these studies they found a modest reduction of CD4
+CD25+ T cells in the thymus, but a profound reduction in CD4+CD25+ T cells in secondary
lymphoid organs such as the spleen and lymph nodes. Furthermore, this reduction of CD4
+CD25+ T cells resulted in the spontaneous induction of autoimmune gastritis in BALB/c
mice; similarly, IL2 neutralization in NOD mice led to an exacerbation of the diabetes
phenotype that naturally develops in these animals [49]. We have obtained similar results when
injecting neutralizing IL2 antibodies into C57Bl/6 mice; peripheral CD4+Foxp3+ Treg
numbers were clearly reduced. Importantly, however, we found that this treatment only
affected the CD4+CD25+ subset of Tregs, suggesting that other cytokines may play a role in
maintaining homeostasis of CD4+CD25Foxp3+ Tregs (MAB and MAF, unpublished
observations). Taken together we believe the available data suggest a model (Fig. 1) in which
IL2 plays a critical role in governing normal homeostasis of Tregs. This most likely reflects
the downregulation of the IL7Rα [50,51] and IL15Rα chain (KBV and MAF, submitted) on
mature Tregs, which renders them more dependent on IL2 for survival and expansion.
Importantly, we have found that Tregs in IL2/mice have upregulated expression of both the
IL15Rα and IL7Rα chains (KBV and MAF, submitted); this most likely accounts for the ability
of Tregs to survive (at least initially) in IL2/ mice. Likewise, under lymphopenic conditions,
Burchill et al. Page 4
Immunol Lett. Author manuscript; available in PMC 2008 November 30.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
IL15 and IL7 appear to be sufficient to maintain Treg homeostasis [49]. However, in wild type
mice, in which Tregs express very low levels of the IL7Rα and IL15Rα chains and thus compete
quite poorly with naïve and effector T cell subsets for endogenous IL7 and IL15, homeostasis
of Tregs is predominately governed by IL2.
IL2R Signaling and Tregs
The IL2 receptor-signaling complex is composed of three distinct subunits, the α-chain (CD25),
β-chain (CD122), and γ-chain (CD132) each of which have unique roles in facilitating IL2-
dependent signal transduction. The IL2Rα chain has a very short cytoplasmic domain and does
not transmit intracellular signals [52]. Instead, IL2Rα acts to confer high affinity binding of
IL2 to the IL2R complex (Kd for IL2Rβ/γ c = 109 M versus 1011 M for the IL2Rα/β/γc
tripartite receptor)[10,34]. In contrast, the IL2Rβ and γc chains play the predominate role in
transducing intracellular signals. Neither of these chains has intrinsic enzymatic function;
rather, they associate with the intracellular tyrosine kinases Jak1 and Jak3, respectively [53–
56]. Ligand binding results in the activation of Jak1 and Jak3 which then phosphorylate
multiple tyrosine residues found in the cytoplasmic tail of the IL2Rβ chain [57]. Therefore,
the primary component of the IL2R complex that functions to convert extracellular ligand
binding into intracellular signals is the IL2Rβ chain.
Taniguchi and colleagues first identified three distinct regions of the IL2Rβ-chain that were
critical for IL2-dependent signal transduction using expression of mutant IL2 receptor
constructs in the mouse pro-B cell line BAF-BO3 [58]. These regions include a serine rich
region, an acidic region, and a carboxy-terminal region, referred to as the S, A, and H regions,
respectively (Fig. 2). Numerous studies have shown that the primary role of the S-region in
IL2R signaling is binding of JAK-1 to conserved motifs referred to as box1 and box 2 [59,
60]. The association of Jak-1 with the IL2Rβ chain, along with the association of JAK-3 with
the γc chain, leads to phosphorylation of tyrosine residues in the A and H-regions following
receptor activation. Thus, the primary role of the A- and H-regions in IL2R signaling is to serve
as docking sites for SH2-domain-containing adaptor or effector molecules. Three of these
intracellular tyrosine residues, which are conserved across multiple species, appear to play
particularly important roles. The first of these is the most membrane-proximal tyrosine residue
(Tyr-338 in the human receptor; Tyr-341 in the murine receptor). Several groups demonstrated
that Tyr-338 is critical for the recruitment of the adaptor protein Shc to the receptor complex
[61–63]. This leads to recruitment of the adaptor protein Grb2 and activation of the Ras/Raf/
Mek/Erk signaling cascade. In addition, the adaptor protein Gab2 is also recruited via SHC,
and results in the recruitment and activation of PI3K [64]. These findings indicate that the A
region is utilized to induce MAPK and PI3K signaling pathways. In contrast, the H-region,
which contains two tyrosine resides, Tyr-392 and Tyr-510, appears to be responsible for
activation of STAT5. Expression of IL2R mutants lacking either the A or H regions
demonstrated that either receptor was capable of preventing autoimmunity when introduced
into IL2Rβ/ T cells in vivo [65,66]. This led to the suggestion that the Ras/PI3K and STAT5
pathways were redundant with regard to Treg development and homeostasis. However,
subsequent work cast some doubt on this interpretation. Specifically, the role of the tyrosine
residues in both the A- and the H-regions were further dissected using IL2R constructs
containing various tyrosine to phenylalanine mutations. Using this strategy, Gaffen et al
demonstrated that the transcription factor STAT5 could be activated by three tyrosine residues
in the IL2Rβ-chain (Tyr-338 in the A-region, Tyr-392, and Tyr-510 in the H-region)[67].
Consistent with the initial studies examining IL2R signal transduction, STAT5 appears to bind
most avidly to phosphorylated Tyr-510 and to a lesser extent to Tyr-392. Supporting these
findings, IL2Rβ mutants containing just Tyr-392 or Tyr-510 were capable of activating STAT5.
However, although direct binding of STAT5 to Tyr-338 based phospho-peptides has not yet
been demonstrated, an IL2Rβ-mutant containing just Tyr-338 was capable of activating
Burchill et al. Page 5
Immunol Lett. Author manuscript; available in PMC 2008 November 30.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
STAT5. Thus, both the A- and H-regions are capable of activating STAT5 suggesting that
perhaps STAT5 plays a key role in Treg development.
Given that IL2 induces multiple signal transduction pathways, it is important to know which
of these pathways are actually active in Tregs. A key study by Bensinger et al demonstrated
that activated CD4+CD25+ effector T cells induced the PI3K, Ras and STAT5 signaling
pathways in response to IL2 stimulation. In contrast, freshly purified CD4+CD25+ Tregs
exclusively activated STAT5 when stimulated with exogenous IL2 [68]. Supporting a role for
STAT5 in Treg development and homeostasis, Burchill et al reported that transgenic mice
expressing a constitutively active form of STAT5 in lymphocytes exhibit a significant increase
in Tregs in both the thymus and secondary lymphoid organs [69]. In contrast, Treg numbers
are reduced in mice expressing a STAT5 hypomorphic allele [69–71]. These studies pointed
to a key role for STAT5 in promoting Treg homeostasis but did not address whether STAT5
regulated Treg development.
The generation of mice in which both STAT5a and STAT5b were completely deleted was
required to examine the role of STAT5 in Treg development. Mice in which the STAT5a and
STAT5b genes were flanked by loxP sites were ultimately generated by Hennighausen and
colleagues [72]. Deletion of STAT5 in all tissues led to multiple defects, including impaired
erythropoesis, and perinatal lethality [72]. To examine the role of STAT5 in T cells, we crossed
STAT5a/b floxed mice to CD4-Cre transgenic animals thereby allowing for selective ablation
of STAT5 in T cells. We found that T cells lacking STAT5 were incapable of differentiating
into Tregs, strongly supporting a role for STAT5 in Treg development [45]. Similar findings
were reported by O’Shea and colleagues [73]. Thus, STAT5 is necessary for Treg development.
To determine whether IL2Rβ-dependent STAT5 signaling was sufficient to drive Treg
development and survival, Yang and colleagues generated a series of IL2Rβ mutants which
selectively activated just the STAT5 or Ras/PI3K pathways [45]. Hematopoietic stem cells
from IL2Rβ/ mice were transduced with retroviruses expressing these mutants and then used
to generate bone marrow chimeras to examine the role of these two pathways in Treg
development. These studies demonstrated that activation of STAT5 alone was sufficient to
restore Treg development while activation of the Ras/PI3K pathway was not ([45] and JY and
MAF, unpublished results). Supporting these studies, we found that expression of a
constitutively active STAT5 transgene restored Treg development in both IL2Rβ/ and γc/
mice [45]. Taken together, these findings illustrate that IL2Rβ-dependent activation of
STAT5 is both necessary and sufficient for the development and homeostasis of Tregs.
Although mouse models in which levels of STAT5 activation are manipulated indicate a critical
role for STAT5 in Treg development, the exact means by which STAT5 directs this process
has yet to be fully elucidated. One possible mechanism by which STAT5 signaling could
influence Treg biology is through the induction or maintenance of the Treg lineage
specification factor Foxp3. Recently Zorn et al identified a consensus STAT5 binding motif
in the first intron of the human foxp3 gene locus that is conserved throughout evolution.
Introduction of luciferase constructs containing this motif into fibroblasts, in the presence of
a constitutively active form of STAT5 or STAT3, resulted in the induction of luciferase activity
[74]. These findings demonstrated that this motif was potentially functional, but did not
demonstrate whether STAT5 or STAT3 actually bound these sites in vivo. We independently
identified the same consensus STAT5 binding motif in the first intron of the foxp3 gene, as
well as several closely spaced evolutionarily conserved STAT5 binding motifs in the 5 region
of the murine foxp3 promoter (Fig. 3). Using chromatin immunoprecipitation assays, we
detected binding of STAT5 to the promoter site, but not the intronic sites in sorted CD4+CD25
+ T cells [45]. A recent report by O’Shea and colleagues confirmed our chromatin
immunoprecipitation studies with regard to STAT5 binding to the foxp3 promoter site.
Burchill et al. Page 6
Immunol Lett. Author manuscript; available in PMC 2008 November 30.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
However, they also reported significant binding of STAT5 to the conserved intronic STAT5
binding sites, as well as a site located several kb 5 of the foxp3 transcription start site. These
latter studies also demonstrated that STAT3 was completely dispensable for foxp3 transcription
and Treg development [73]. Taken together, these studies clearly demonstrate that STAT5 can
bind to the foxp3 gene. However, additional experiments are required to determine the
biological significance of these STAT5 binding sites.
In summary, studies over the last few years have helped illuminate the key role that IL2 and
IL2Rβ-dependent signals play in Treg development and homeostasis. However, many
questions still remain. For example, although STAT5 has been shown to bind to distinct sites
in the foxp3 gene, the functional relevance of these binding sites remains to be determined.
Likewise, transcription factors that potentially cooperate with STAT5 to promote foxp3
transcription remain to be identified. Finally, how TCR-dependent and IL2Rβ-dependent
signals interact to promote Treg development remains to be established. The answer to these
questions will provide a more complete picture of how IL2Rβ-dependent signals entrain
regulatory T cell development and homeostasis.
Acknowledgements
This work was supported by grants from the NIH (AI061165) and by a Pew Scholar Award, a Cancer Research
Investigator Award and a Leukemia and Lymphoma Society Scholar Award to M.A.F. M.A.B. was supported by an
NIH training grant (2T32-AI07313).
References
1. Morgan DA, Ruscetti FW, Gallo R. Selective in vitro growth of T lymphocytes from normal human
bone marrows. Science 1976;193:1007–1008. [PubMed: 181845]
2. Taniguchi T, Matsui H, Fujita T, Takaoka C, Kashima N, Yoshimoto R, Hamuro J. Structure and
expression of a cloned cDNA for human interleukin-2. Nature 1983;302:305–310. [PubMed: 6403867]
3. Leonard WJ, Depper JM, Crabtree GR, Rudikoff S, Pumphrey J, Robb RJ, Kronke M, Svetlik PB,
Peffer NJ, Waldmann TA, et al. Molecular cloning and expression of cDNAs for the human
interleukin-2 receptor. Nature 1984;311:626–631. [PubMed: 6090948]
4. Nikaido T, Shimizu A, Ishida N, Sabe H, Teshigawara K, Maeda M, Uchiyama T, Yodoi J, Honjo T.
Molecular cloning of cDNA encoding human interleukin-2 receptor. Nature 1984;311:631–635.
[PubMed: 6090949]
5. Cosman D, Cerretti DP, Larsen A, Park L, March C, Dower S, Gillis S, Urdal D. Cloning, sequence
and expression of human interleukin-2 receptor. Nature 1984;312:768–771. [PubMed: 6096720]
6. Hatakeyama M, Tsudo M, Minamoto S, Kono T, Doi T, Miyata T, Miyasaka M, Taniguchi T.
Interleukin-2 receptor beta chain gene: generation of three receptor forms by cloned human alpha and
beta chain cDNA’s. Science 1989;244:551–556. [PubMed: 2785715]
7. Takeshita T, Asao H, Ohtani K, Ishii N, Kumaki S, Tanaka N, Munakata H, Mankamura M, Sugamura
K. Cloning of the gamma chain of the human IL-2 receptor. Science 1992;257:379–382. [PubMed:
1631559]
8. Nelson BH, Willerford DM. Biology of the interleukin-2 receptor. Adv Immunol 1998;70:1–81.
[PubMed: 9755337]
9. Wang X, Rickert M, Garcia KC. Structure of the quaternary complex of interleukin-2 with its alpha,
beta, and gammac receptors. Science 2005;310:1159–1163. [PubMed: 16293754]
10. Smith KA. The interleukin 2 receptor. Annu Rev Cell Biol 1989;5:397–425. [PubMed: 2688708]
11. Lin JX, Leonard WJ. Signaling from the IL-2 receptor to the nucleus. Cytokine Growth Factor Rev
1997;8:313–332. [PubMed: 9620644]
12. Hatakeyama M, Tsudo M, Minamoto S, Kono T, Doi T, Miyata T, Miyasaka M, Taniguchi T.
Interleukin-2 receptor β chain gene: generation of three receptor forms by cloned human α and β
chain cDNA. Science 1989;244:551–556. [PubMed: 2785715]
Burchill et al. Page 7
Immunol Lett. Author manuscript; available in PMC 2008 November 30.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
13. Miyazaki T, Liu Z-J, Kawahara A, Minami Y, Yamada K, Tsujimoto Y, Barsoumian EL, Perlmutter
RM, Taniguchi T. Three distinct IL-2 signaling pathways mediated by bcl-2, c-myc and lck cooperate
in hematopoietic cell proliferation. Cell. 1995
14. Schorle H, Holtschke T, Hunig T, Schimpl A, Horak I. Development and function of T cells in mice
rendered interleukin-2 deficient by gene targeting. Nature 1991;352:621–624. [PubMed: 1830926]
15. Sadlack B, Merz H, Schorle H, Schimpl A, Feller AC, Horak I. Ulcerative colitis-like disease in mice
with a disrupted interleukin-2 gene. Cell 1993;75:253–261. [PubMed: 8402910]
16. Lenardo MJ. Interleukin-2 programs mose αβ T lymphocytes for apoptosis. Nature 1991;353:858–
861. [PubMed: 1944559]
17. Willerford DM, Chen J, Ferry JA, Davidson L, Ma A, Alt FW. Interleukin-2 receptor αchain regulates
the size and content of the peripheral lymphoid compartment. Immunity 1995;3:521–530. [PubMed:
7584142]
18. Suzuki H, Kundig TM, Furlonger C, Wakeham A, Timms E, Matsuyama T, Schmits R, Simard JJ,
Ohashi PS, Griesser H, et al. Deregulated T cell activation and autoimmunity in mice lacking
interleukin-2 receptor beta. Science 1995;268:1472–1476. [PubMed: 7770771]
19. Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M. Immunologic self-tolerance maintained by
activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism
of self-tolerance causes various autoimmune diseases. J Immunol 1995;155:1151–1164. [PubMed:
7636184]
20. Nishizuka Y, Sakakura T. Thymus and reproduction: sex-linked dysgenesia of the gonad after
neonatal thymectomy in mice. Science 1969;166:753–755. [PubMed: 5823314]
21. Gershon RK, Kondo K. Cell interactions in the induction of tolerance: the role of thymic lymphocytes.
Immunology 1970;18:723–737. [PubMed: 4911896]
22. Yamanouchi J, Rainbow D, Serra P, Howlett S, Hunter K, Garner VE, Gonzalez-Munoz A, Clark J,
Veijola R, Cubbon R, Chen SL, Rosa R, Cumiskey AM, Serreze DV, Gregory S, Rogers J, Lyons
PA, Healy B, Smink LJ, Todd JA, Peterson LB, Wicker LS, Santamaria P. Interleukin-2 gene
variation impairs regulatory T cell function and causes autoimmunity. Nature genetics 2007;39:329–
337. [PubMed: 17277778]
23. Davoodi-Semiromi A, McDuffie M, Litherland S, Clare-Salzler M. Truncated pStat5B is associated
with the Idd4 locus in NOD mice. Biochemical and biophysical research communications
2007;356:655–661. [PubMed: 17382905]
24. Vella A, Cooper JD, Lowe CE, Walker N, Nutland S, Widmer B, Jones R, Ring SM, McArdle W,
Pembrey ME, Strachan DP, Dunger DB, Twells RC, Clayton DG, Todd JA. Localization of a type
1 diabetes locus in the IL2RA/CD25 region by use of tag single-nucleotide polymorphisms. American
journal of human genetics 2005;76:773–779. [PubMed: 15776395]
25. Risk Alleles for Multiple Sclerosis Identified by a Genomewide Study. N Engl J Med. 2007
26. Brand OJ, Lowe CE, Heward JM, Franklyn JA, Cooper JD, Todd JA, Gough SC. Association of the
interleukin-2 receptor alpha (IL-2Ralpha)/CD25 gene region with Graves’ disease using a multilocus
test and tag SNPs. Clinical endocrinology 2007;66:508–512. [PubMed: 17371467]
27. Suzuki H, Zhou YW, Kato M, Mak TW, Nakashima I. Normal regulatory alpha/beta T cells effectively
eliminate abnormally activated T cells lacking the interleukin 2 receptor beta in vivo. The Journal of
experimental medicine 1999;190:1561–1572. [PubMed: 10587347]
28. Malek TR, Yu A, Vincek V, Scibelli P, Kong L. CD4 regulatory T cells prevent lethal autoimmunity
in IL-2Rbeta-deficient mice. Implications for the nonredundant function of IL-2. Immunity
2002;17:167–178. [PubMed: 12196288]
29. Almeida AR, Legrand N, Papiernik M, Freitas AA. Homeostasis of peripheral CD4+ T cells: IL-2R
alpha and IL-2 shape a population of regulatory cells that controls CD4+ T cell numbers. J Immunol
2002;169:4850–4860. [PubMed: 12391195]
30. Malek TR, Bayer AL. Tolerance, not immunity, crucially depends on IL-2. Nature reviews
2004;4:665–674.
31. Malek TR, Porter BO, Codias EK, Scibelli P, Yu A. Normal lymphoid homeostasis and lack of lethal
autoimmunity in mice containing mature T cells with severely impaired IL-2 receptors. J Immunol
2000;164:2905–2914. [PubMed: 10706676]
Burchill et al. Page 8
Immunol Lett. Author manuscript; available in PMC 2008 November 30.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
32. Bayer AL, Yu A, Adeegbe D, Malek TR. Essential role for interleukin-2 for CD4(+)CD25(+) T
regulatory cell development during the neonatal period. The Journal of experimental medicine
2005;201:769–777. [PubMed: 15753210]
33. Furtado GC, Curotto de Lafaille MA, Kutchukhidze N, Lafaille JJ. Interleukin 2 signaling is required
for CD4(+) regulatory T cell function. The Journal of experimental medicine 2002;196:851–857.
[PubMed: 12235217]
34. Greene WC, Leonard WJ. The human interleukin-2 receptor. Annu Rev Immunol 1986;4:69–95.
[PubMed: 3011033]
35. Brunkow ME, Jeffery EW, Hjerrild KA, Paeper B, Clark LB, Yasayko SA, Wilkinson JE, Galas D,
Ziegler SF, Ramsdell F. Disruption of a new forkhead/winged-helix protein, scurfin, results in the
fatal lymphoproliferative disorder of the scurfy mouse. Nature genetics 2001;27:68–73. [PubMed:
11138001]
36. Bennett CL, Christie J, Ramsdell F, Brunkow ME, Ferguson PJ, Whitesell L, Kelly TE, Saulsbury
FT, Chance PF, Ochs HD. The immune dysregulation, polyendocrinopathy, enteropathy, X-linked
syndrome (IPEX) is caused by mutations of FOXP3. Nature genetics 2001;27:20–21. [PubMed:
11137993]
37. Bennett CL, Brunkow ME, Ramsdell F, O’Briant KC, Zhu Q, Fuleihan RL, Shigeoka AO, Ochs HD,
Chance PF. A rare polyadenylation signal mutation of the FOXP3 gene (AAUAAA-->AAUGAA)
leads to the IPEX syndrome. Immunogenetics 2001;53:435–439. [PubMed: 11685453]
38. Khattri R, Cox T, Yasayko SA, Ramsdell F. An essential role for Scurfin in CD4(+)CD25(+) T
regulatory cells. Nature immunology 2003;4:337–342. [PubMed: 12612581]
39. Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4(+)
CD25(+) regulatory T cells. Nature immunology 2003;4:330–336. [PubMed: 12612578]
40. Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor
Foxp3. Science 2003;299:1057–1061. [PubMed: 12522256]
41. Fontenot JD, Rasmussen JP, Williams LM, Dooley JL, Farr AG, Rudensky AY. Regulatory T cell
lineage specification by the forkhead transcription factor foxp3. Immunity 2005;22:329–341.
[PubMed: 15780990]
42. Wan YY, Flavell RA. Identifying Foxp3-expressing suppressor T cells with a bicistronic reporter.
Proceedings of the National Academy of Sciences of the United States of America 2005;102:5126–
5131. [PubMed: 15795373]
43. Fontenot JD, Rasmussen JP, Gavin MA, Rudensky AY. A function for interleukin 2 in Foxp3-
expressing regulatory T cells. Nature immunology 2005;6:1142–1151. [PubMed: 16227984]
44. D’Cruz LM, Klein L. Development and function of agonist-induced CD25(+)Foxp3(+) regulatory T
cells in the absence of interleukin 2 signaling. Nature immunology 2005;6:1152–1159. [PubMed:
16227983]
45. Burchill MA, Yang J, Vogtenhuber C, Blazar BR, Farrar MA. IL-2 receptor beta-dependent STAT5
activation is required for the development of Foxp3+ regulatory T cells. J Immunol 2007;178:280–
290. [PubMed: 17182565]
46. Soper DM, Kasprowicz DJ, Ziegler SF. IL-2Rbeta links IL-2R signaling with Foxp3 expression.
European journal of immunology 2007;37:1817–1826. [PubMed: 17559173]
47. Yu A, Malek TR. Selective availability of IL-2 is a major determinant controlling the production of
CD4+CD25+Foxp3+ T regulatory cells. J Immunol 2006;177:5115–5121. [PubMed: 17015695]
48. Bayer AL, Yu A, Malek TR. Function of the IL-2R for thymic and peripheral CD4+CD25+ Foxp3+
T regulatory cells. J Immunol 2007;178:4062–4071. [PubMed: 17371960]
49. Setoguchi R, Hori S, Takahashi T, Sakaguchi S. Homeostatic maintenance of natural Foxp3(+) CD25
(+) CD4(+) regulatory T cells by interleukin (IL)-2 and induction of autoimmune disease by IL-2
neutralization. The Journal of experimental medicine 2005;201:723–735. [PubMed: 15753206]
50. Liu W, Putnam AL, Xu-Yu Z, Szot GL, Lee MR, Zhu S, Gottlieb PA, Kapranov P, Gingeras TR,
Fazekas de St Groth B, Clayberger C, Soper DM, Ziegler SF, Bluestone JA. CD127 expression
inversely correlates with FoxP3 and suppressive function of human CD4+ T reg cells. The Journal
of experimental medicine 2006;203:1701–1711. [PubMed: 16818678]
51. Seddiki N, Santner-Nanan B, Martinson J, Zaunders J, Sasson S, Landay A, Solomon M, Selby W,
Alexander SI, Nanan R, Kelleher A, Fazekas de St Groth B. Expression of interleukin (IL)-2 and
Burchill et al. Page 9
Immunol Lett. Author manuscript; available in PMC 2008 November 30.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
IL-7 receptors discriminates between human regulatory and activated T cells. The Journal of
experimental medicine 2006;203:1693–1700. [PubMed: 16818676]
52. Hatakeyama M, Minamoto S, Taniguchi T. Intracytoplasmic phosphorylation sites of Tac antigen
(p55) are not essential for the conformation, function, and regulation of the human interleukin 2
receptor. Proceedings of the National Academy of Sciences of the United States of America
1986;83:9650–9654. [PubMed: 3099287]
53. Russell SM, Johnston JA, Noguchi M, Kawamura M, Bacon CM, Friedmann M, Berg M, McVicar
DW, Witthuhn BA, Silvennoinen O, Goldman AS, Schmalstieg FC, Ihle JN, O’Shea JJ, Leonard WJ.
Interaction of IL-2Rβ and gammac chains with Jak1 and Jak3: Implications for XSCID and XCID.
Science 1994;266:1042–1045. [PubMed: 7973658]
54. Miyazaki T, Kawahara A, Fujii H, Nakagawa Y, Minami Y, Liu Z-J, Oishi I, Silvennoinen O,
Witthuhn BA, Ihle JN, Taniguchi T. Functional activation of Jak1 and Jak3 by selective association
with IL-2 receptor subunits. Science 1994;266:1045–1047. [PubMed: 7973659]
55. Witthuhn BA, Silvennoinen O, Miura O, Lai KS, Cwik C, Liu ET, Ihle JN. Involvement of the Jak-3
Janus kinase in signalling by interleukins 2 and 4 in lymphoid and myeloid cells. Nature
1994;370:153–157. [PubMed: 8022486]
56. Johnston JA, Kawamura M, Kirken RA, Chen Y-Q, Blake TB, Shibuya K, Ortaldo JR, McVicar DW,
O’Shea JJ. Phosphorylation and activation of the Jak-3 Janus kinase in response to interleukin-2.
Nature 1994;370:151–153. [PubMed: 8022485]
57. Sharon M, Gnarra JR, Leonard WJ. The β-chain of the IL-2 receptor (p70) is tyrosine-phosphorylated
on YT and HUT-102B2 cells. J Immunol 1989;143:2530–2533. [PubMed: 2477446]
58. Hatakeyama M, Mori H, Doi T, Taniguchi T. A restricted cytoplasmic region of IL-2 receptor beta
chain is essential for growth signal transduction but not for ligand binding and internalization. Cell
1989;59:837–845. [PubMed: 2590941]
59. Liu KD, Lai SY, Goldsmith MA, Greene WC. Identification of a variable region within the
cytoplasmic tail of the IL-2 receptor beta chain that is required for growth signal transduction. The
Journal of biological chemistry 1995;270:22176–22181. [PubMed: 7545674]
60. Zhu MH, Berry JA, Russell SM, Leonard WJ. Delineation of the regions of interleukin-2 (IL-2)
receptor beta chain important for association of Jak1 and Jak3. Jak1-independent functional
recruitment of Jak3 to Il-2Rbeta. The Journal of biological chemistry 1998;273:10719–10725.
[PubMed: 9553136]
61. Evans GA, Howard OM, Erwin R, Farrar WL. Interleukin-2 induces tyrosine phosphorylation of the
vav proto-oncogene product in human T cells: lack of requirement for the tyrosine kinase lck.
Biochemistry Journal 1993;294:339–342.
62. Friedmann MC, Migone TS, Russell SM, Leonard WJ. Different interleukin 2 receptor beta-chain
tyrosines couple to at least two signaling pathways and synergistically mediate interleukin 2-induced
proliferation. Proceedings of the National Academy of Sciences of the United States of America
1996;93:2077–2082. [PubMed: 8700888]
63. Ravichandran KS, Igras V, Shoelson SE, Fesik SW, Burakoff SJ. Evidence for a role for the
phosphotyrosine-binding domain of Shc in interleukin 2 signaling. Proceedings of the National
Academy of Sciences (USA) 1996;93:5275–5280.
64. Gadina M, Sudarshan C, Visconti R, Zhou YJ, Gu H, Neel BG, O’Shea JJ. The docking molecule
gab2 is induced by lymphocyte activation and is involved in signaling by interleukin-2 and
interleukin-15 but not other common gamma chain-using cytokines. The Journal of biological
chemistry 2000;275:26959–26966. [PubMed: 10849428]
65. Van Parijs L, Refaeli Y, Lord JD, Nelson BH, Abbas AK, Baltimore D. Uncoupling IL-2 signals that
regulate T cell proliferation, survival, and Fas-mediated activation-induced cell death. Immunity
1999;11:281–288. [PubMed: 10514006]
66. Fujii H, Ogasawara K, Otsuka H, Suzuki M, Yamamura K, Yokochi T, Miyazaki T, Suzuki H, Mak
TW, Taki S, Taniguchi T. Functional dissection of the cytoplasmic subregions of the IL-2 receptor
betac chain in primary lymphocyte populations. Embo J 1998;17:6551–6557. [PubMed: 9822600]
67. Gaffen SL, Lai SY, Ha M, Liu X, Hennighausen L, Greene WC, Goldsmith MA. Distinct tyrosine
residues within the interleukin-2 receptor beta chain drive signal transduction specificity,
Burchill et al. Page 10
Immunol Lett. Author manuscript; available in PMC 2008 November 30.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
redundancy, and diversity. The Journal of biological chemistry 1996;271:21381–21390. [PubMed:
8702919]
68. Bensinger SJ, Walsh PT, Zhang J, Carroll M, Parsons R, Rathmell JC, Thompson CB, Burchill MA,
Farrar MA, Turka LA. Distinct IL-2 receptor signaling pattern in CD4+CD25+ regulatory T cells. J
Immunol 2004;172:5287–5296. [PubMed: 15100267]
69. Burchill MA, Goetz CA, Prlic M, O’Neil JJ, Harmon IR, Bensinger SJ, Turka LA, Brennan P, Jameson
SC, Farrar MA. Distinct effects of STAT5 activation on CD4+ and CD8+ T cell homeostasis:
development of CD4+CD25+ regulatory T cells versus CD8+ memory T cells. J Immunol
2003;171:5853–5864. [PubMed: 14634095]
70. Antov A, Yang L, Vig M, Baltimore D, Van Parijs L. Essential role for STAT5 signaling in CD25
+CD4+ regulatory T cell homeostasis and maintenance of self-tolerance. J Immunol 2003;171:3435–
3441. [PubMed: 14500638]
71. Snow JW, Abraham N, Ma MC, Herndier BG, Pastuszak AW, Goldsmith MA. Loss of tolerance and
autoimmunity affecting multiple organs in STAT5A/5B-deficient mice. J Immunol 2003;171:5042–
5050. [PubMed: 14607901]
72. Cui Y, Riedlinger G, Miyoshi K, Tang W, Li C, Deng CX, Robinson GW, Hennighausen L.
Inactivation of Stat5 in mouse mammary epithelieum during pregnancy reveals distinct functions in
cell proliferation, survival, and differentiation. Mol Cell Biol 2004;24:8037–8047. [PubMed:
15340066]
73. Yao Z, Kanno Y, Kerenyi M, Stephens G, Durant L, Watford WT, Laurence A, Robinson GW,
Shevach EM, Moriggl R, Hennighausen L, Wu C, O’Shea JJ. Nonredundant roles for Stat5a/b in
directly regulating Foxp3. Blood 2007;109:4368–4375. [PubMed: 17227828]
74. Zorn E, Nelson EA, Mohseni M, Porcheray F, Kim H, Litsa D, Bellucci R, Raderschall E, Canning
C, Soiffer RJ, Frank DA, Ritz J. IL-2 regulates FOXP3 expression in human CD4+CD25+ regulatory
T cells through a STAT-dependent mechanism and induces the expansion of these cells in vivo. Blood
2006;108:1571–1579. [PubMed: 16645171]
Burchill et al. Page 11
Immunol Lett. Author manuscript; available in PMC 2008 November 30.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 1. Model of Cytokine-dependent Treg Development and Homeostasis
In the thymus, IL2 plays the predominant role in driving Treg development. In the absence of
IL2, IL15 is sufficient for regulatory T cell development. Other γc-dependent cytokines such
as IL7 may play a minor role in this process and most likely account for the few Tregs found
in IL2Rβ/ mice as compared to γc/ mice in which Tregs are absent. In the spleen of wild
type mice, IL2-dependent signals lead to the downregulation of both the IL7Rα and IL15Rα
chains. This renders mature Tregs primarily dependent on IL2 for their survival and expansion.
In contrast, in IL2/ mice, IL15Rα and IL7Rα remain expressed at high levels and allow for
survival of mature Tregs. Likewise, under lymphopenic conditions, such as occurs upon
introduction of Tregs into rag2/ mice, Tregs do survive and expand. This most likely occurs
due the absence of other T cell subsets thereby leading to increased amounts of available IL15
and IL7 that can stimulate Treg survival despite lower expression of IL15Rα and IL7Rα
receptors by those cells. In addition, the absence of IL2 signals may lead to re-expression of
the IL15Rα and IL7Rα rendering these cells more sensitive to IL15 and IL7.
Burchill et al. Page 12
Immunol Lett. Author manuscript; available in PMC 2008 November 30.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 2. Signaling Components of the IL2 Receptor Complex
The IL2R is composed of three distinct subunits - IL2Rα, IL2Rβ, and IL2Rγ. The IL2Rβ-chain
consists of three previously identified signaling domains, the A-, H-, and S-regions. The
tyrosine-containing A and H regions are labeled, while the JAK1 binding domain or S-region
is shaded in gray. Phosphorylation of Tyr-338 in the A region recruits the adaptor protein SHC;
SHC then binds Grb2 and Gab2, leading to activation of the Ras-Raf-MAPK and PI3K/Akt
signaling pathways, respectively. Phosphorylation of Tyr-392 and Tyr-510 in the H region
recruits STAT5 and leads to its activation; in the absence of these two tyrosine residues Tyr-338
can also induce STAT5 activation.
Burchill et al. Page 13
Immunol Lett. Author manuscript; available in PMC 2008 November 30.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 3. STAT Binding Motifs in the Murine foxp3 gene
The murine foxp3 gene locus contains many motifs to which activated STAT molecules can
bind. Black boxes represent exons while vertical lines represent consensus STAT binding
motifs. The three closely linked sites (separated by 7 base pairs) in the promoter and the three
sites labeled intronic are conserved across species.
Burchill et al. Page 14
Immunol Lett. Author manuscript; available in PMC 2008 November 30.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
... rs12722502 lies in an intronic enhancer of IL2RA experimentally validated by CRISPR activation 63 . IL2RA encodes a key component of the interleukin 2 signaling pathway critical for T cell proliferation and homeostasis 64,65 and has been implicated in rheumatoid arthritis 66 , multiple sclerosis 67 , systemic sclerosis 68 , systemic lupus 69 , and type 1 diabetes 70 . This link also scored in the top 5% of links predicted by the distance-based method, whereas existing single-cell methods assigned top priority to other genes (RBM17, PRKCQ) and additionally scored other genes in the top 5% of links (GDI2, FBXO18, IL15RA). ...
Preprint
Methods that analyze single-cell paired RNA-seq and ATAC-seq multiome data have shown great promise in linking regulatory elements to genes. However, existing methods differ in their modeling assumptions and approaches to account for biological and technical noise-leading to low concordance in their linking scores-and do not capture the effects of genomic distance. We propose pgBoost, an integrative modeling framework that trains a non-linear combination of existing linking strategies (including genomic distance) on fine-mapped eQTL data to assign a probabilistic score to each candidate SNP-gene link. We applied pgBoost to single-cell multiome data from 85k cells representing 6 major immune/blood cell types. pgBoost attained higher enrichment for fine-mapped eSNP-eGene pairs (e.g. 21x at distance >10kb) than existing methods (1.2-10x; p-value for difference = 5e-13 vs. distance-based method and < 4e-35 for each other method), with larger improvements at larger distances (e.g. 35x vs. 0.89-6.6x at distance >100kb; p-value for difference < 0.002 vs. each other method). pgBoost also outperformed existing methods in enrichment for CRISPR-validated links (e.g. 4.8x vs. 1.6-4.1x at distance >10kb; p-value for difference = 0.25 vs. distance-based method and < 2e-5 for each other method), with larger improvements at larger distances (e.g. 15x vs. 1.6-2.5x at distance >100kb; p-value for difference < 0.009 for each other method). Similar improvements in enrichment were observed for links derived from Activity-By-Contact (ABC) scores and GWAS data. We further determined that restricting pgBoost to features from a focal cell type improved the identification of SNP-gene links relevant to that cell type. We highlight several examples where pgBoost linked fine-mapped GWAS variants to experimentally validated or biologically plausible target genes that were not implicated by other methods. In conclusion, a non-linear combination of linking strategies, including genomic distance, improves power to identify target genes underlying GWAS associations.
... Usually, both IL-2 and IL-17 maintain a balanced state, but the levels of IL-2 and IL-17 in patients with IBD are increased [85]. IL-2 is known for its capacity to promote lymphocyte activation and proliferation [86]. IL-2 plays a critical role in regulating the repair processes of intestinal mucosal cells. ...
Article
Full-text available
Inflammatory bowel disease (IBD) includes ulcerative colitis and Crohn’s disease, and it is a multifactorial disease of the intestinal mucosa. Oxidative stress damage and inflammation are major risk factors for IBD. Vitamin E has powerful antioxidant and anti-inflammatory effects. Our previous work and other investigations have shown that vitamin E has a positive effect on the prevention and treatment of IBD. In this paper, the source and structure of vitamin E and the potential mechanism of vitamin E’s role in IBD were summarized, and we also analyzed the status of vitamin E deficiency in patients with IBD and the effect of vitamin E supplementation on IBD. The potential mechanisms by which vitamin E plays a role in the prevention and treatment of IBD include improvement of oxidative damage, enhancement of immunity, maintenance of intestinal barrier integrity, and suppression of inflammatory cytokines, modulating the gut microbiota and other relevant factors. The review will improve our understanding of the complex mechanism by which vitamin E inhibits IBD, and it also provides references for doctors in clinical practice and researchers in this field.
... Effective activation of the IL-2/STAT5 signaling pathway is an important condition for the differentiation and functional maturation of Tregs [43,44,53,55]. It has been proven that administration of low-dose exogenous IL-2 can expand Tregs in vivo or in vitro and promote the expression of functional molecules, such as pStat5 and Foxp3, in Tregs, thereby effectively improving autoimmune disorders [18,[21][22][23]. ...
... Furthermore, IL2RA has been identified as a susceptibility gene for autoimmune thyroid disease (52). IL2RA (CD25) is highly expressed in regulatory T cells of CD4CD25 (Tregs) as an important regulator of immune homeostasis and suppressing autoimmune responses (53). In our study, for TYK2 and IL2RA, the most likely risk genes were identified through four methods (PLACO_FUMA, MTAG, Bayesian colocalization analysis, and TWAS) and three methods (PLACO_FUMA, MTAG, and Bayesian colocalization analysis), respectively. ...
Article
Full-text available
Background There is still controversy regarding the relationship between hypothyroidism and rheumatoid arthritis (RA), and there has been a dearth of studies on this association. The purpose of our study was to explore the shared genetic architecture between hypothyroidism and RA. Methods Using public genome-wide association studies summary statistics of hypothyroidism and RA, we explored shared genetics between hypothyroidism and RA using linkage disequilibrium score regression, ρ-HESS, Pleiotropic analysis under a composite null hypothesis (PLACO), colocalization analysis, Multi-Trait Analysis of GWAS (MTAG), and transcriptome-wide association study (TWAS), and investigated causal associations using Mendelian randomization (MR). Results We found a positive genetic association between hypothyroidism and RA, particularly in local genomic regions. Mendelian randomization analysis suggested a potential causal association of hypothyroidism with RA. Incorporating gene expression data, we observed that the genetic associations between hypothyroidism and RA were enriched in various tissues, including the spleen, lung, small intestine, adipose visceral, and blood. A comprehensive approach integrating PLACO, Bayesian colocalization analysis, MTAG, and TWAS, we successfully identified TYK2, IL2RA, and IRF5 as shared risk genes for both hypothyroidism and RA. Conclusions Our investigation unveiled a shared genetic architecture between these two diseases, providing novel insights into the underlying biological mechanisms and establishing a foundation for more effective interventions.
... Interleukin-2 (IL-2) is the primary mediator of Stat5 signaling in Treg cells and is required for their development and maintenance 26,27 . Yet IL-2 can also act on Teff cells to promote, rather than repress, T cell-driven inflammation 28 . ...
Preprint
Maintenance of immune homeostasis to the intestinal mictrobiota is dependent on a population of effector regulatory T (eTreg) cells that develop from microbiota-reactive induced (i)Treg cells. A cardinal feature of eTreg cells is their production of IL-10, which plays a non-redundant role in immune tolerance of commensal microbes. Here, we identify an unexpected role for IL-2-induced Stat3 signaling to program iTreg cells for eTreg cell differentiation and Il10 transcriptional competency. IL-2 proved to be both necessary and sufficient for eTreg cell development - contingent on Stat3 output of the IL-2 receptor coordinate with IL-2 signaling during early Treg cell commitment. Induction of iTreg cell programming in absence of IL-2-induced Stat3 signaling resulted in impaired eTreg cell differentiation and a failure to produce IL-10. An IL-2 mutein with reduced affinity for the IL-2Rgamma (gammac) chain was found to have blunted IL-2R Stat3 output, resulting in a deficiency of Il10 transcriptional programming that could not be fully rescued by Stat3 signaling subsequent to an initial window of iTreg cell differentiation. These findings expose a heretofore unappreciated role of IL-2 signaling that acts early to program subsequent production of IL-10 by developing eTreg cells, with broad implications for IL-2-based therapeutic interventions in immune-mediated diseases.
... 21 The major physiological function of IL-2 is to limit T cell responses since IL-2 is critical for the development and peripheral expansion of CD4+CD25+ regulatory T cells, which promote self-tolerance by suppressing T cell responses. 22,23 Our understanding of G-CSF and IL-2 biology prompted evaluation of how best to clinically manipulate this important immunoregulatory pathway in patients with ALS. ...
Article
Neurological syndrome amyotrophic lateral sclerosis (ALS) affects motor neurons and is characterized by progressive motor neuron loss in the brain and spinal cord. ALS starts with mainly focal onset but when the disease progresses, it spreads to different parts of the body, with survival limits of 2-5 years after disease initiation. To date, only supportive care is provided for ALS patients, and no effective treatment or cure has been discovered. This review is focused on clinical and immunological aspects of ALS patients, based on our case studies, and we discuss the treatment we have provided to those patients based on a detailed evaluation of their peripheral blood immune cells and blood-derived serum secreted factors, cytokines, chemokines and growth factors. We show that using a personalized approach of low dose immunotherapy there is an improvement in the effects on inflammation and immunological dysfunction.
... According to the genome-wide association study, IL2RA and IL2RB genes play important roles in celiac disease [32]. These two subsets along with the common gamma chain (IL2RG), form the heterotrimeric high affinity cell surface receptor for IL-2, a crucial cytokine contributing to the adaptive immune responses in CD [33,34]. It has also been reported that JAK2 is overexpressed in CD patients intestinal tissues and is critical for IL-12 signaling [35]. ...
Article
Full-text available
Background Celiac disease (CD) is a chronic immune-mediated enteropathy and a cytokine network is involved in its pathogenesis. Interleukin-2 (IL-2) has a key role in the adaptive immune pathogenesis of CD and has been reported to be one of the earliest cytokines to be elicited after gluten exposure by CD patients. This study aimed at investigating the expression level of IL-2 and functionally related genes SOCS1 and TBX21 in active and treated CD patients compared to controls. Methods and results Peripheral blood (PB) samples were collected from 40 active CD (ACD), 100 treated CD, and 100 healthy subjects. RNA was extracted, cDNA was synthesized and mRNA expression levels of the desired genes were investigated by Real-time PCR. The gene–gene interaction network was also constructed by GeneMANIA. Our results showed a higher PB mRNA expression of IL-2 in ACD patients compared to controls (p = 0.001) and treated CD patients (p˂0.0001). The mRNA expression level of TBX21 was also significantly up-regulated in ACD patients compared to controls (P = 0.03). SOCS1 mRNA level did not differ between active and treated CD patients and controls (p˃0.05) but showed a significant correlation with the patient’s aphthous stomatitis symptom (r = 0.37, p = 0.01). ROC curve analysis suggested that the use of IL-2 levels can reach a high specificity and sensitivity in discriminating active CD patients. Conclusions The PB level of IL-2 has the potential to be introduced as a diagnostic biomarker for CD. Larger cohort studies, including pediatric patients, are needed to achieve more insights in this regard.
Chapter
Regulatory T cells (T regs) are a subtype to T cells that work to inhibit the immune response of other T cells. Their main function is to maintain homeostasis and mediate self-tolerance. They play a major role in preventing autoimmunity by inhibiting T cell proliferation and cytokine production. T regs also dampen the killer T cell response once the infection has been dealt with, so as to protect the normal tissue. T regs can also prevent and treat graft versus host disease (GVHD). Failure of T reg function leads to increased self-tissue injury as cytotoxic response of T cells will go uncontrolled. Increased activity of T regs can lead to increased risk of infections, and cancers by suppressing tumor-specific T cell immunity. T regs play a crucial role in establishment of tolerance after allogeneic stem cell transplant.
Article
Full-text available
Interleukin (IL)-2, a critical cytokine with indispensable functions in regulating lymphoid homeostasis, induces the activation of several biochemical pathways. Precisely how these pathways are linked and how they relate to the biological action of IL-2 is incompletely understood. We previously identified SHP-2 (Src homology 2 domain containing phosphatase 2) as an important intermediate in IL-2-dependent MAPK activation and showed its association with a 98-kDa phosphoprotein in response to IL-2. Here, we demonstrate that Gab2, a recently identified adapter molecule, is the major SHP-2 and phosphatidylinositol 3′-kinase-associated 98-kDa protein in normal, IL-2-activated lymphocytes. We further demonstrate that phosphorylation of both Gab2 and SHP-2 is largely dependent upon tyrosine 338 of the IL-2 receptor β chain. Gab2 can be a substrate of all the three major classes of non-receptor tyrosine kinases associated with the IL-2R, but in terms of IL-2 signaling, JAK3 but not Lck or Syk is essential for Gab2 phosphorylation. We also demonstrate that only IL-2 and IL-15, but not other γc cytokines induce Gab2 phosphorylation; the ability to phosphorylate Gab2 correlates with Shc phosphorylation and ERK1/ERK2 activation. Finally, we also show that Gab2 levels are regulated by T cell activation, and resting T cells express little Gab2. Therefore, up-regulation and activation of Gab2 may be important in linking the IL-2 receptor to activation of MAPK and may be an important means of achieving specificity in cytokine signaling.
Article
Full-text available
We show that the lymphoid hyperplasia observed in IL-2Rα- and IL-2-deficient mice is due to the lack of a population of regulatory cells essential for CD4 T cell homeostasis. In chimeras reconstituted with bone marrow cells from IL-2Rα-deficient donors, restitution of a population of CD25+CD4+ T cells prevents the chaotic accumulation of lymphoid cells, and rescues the mice from autoimmune disease and death. The reintroduction of IL-2-producing cells in IL-2-deficient chimeras establishes a population of CD25+CD4+ T cells, and restores the peripheral lymphoid compartments to normal. The CD25+CD4+ T cells regulated selectively the number of naive CD4+ T cells transferred into T cell-deficient hosts. The CD25+CD4+/naive CD4 T cell ratio and the sequence of cell transfer determines the homeostatic plateau of CD4+ T cells. Overall, our findings demonstrate that IL-2Rα is an absolute requirement for the development of the regulatory CD25+CD4+ T cells that control peripheral CD4 T cell homeostasis, while IL-2 is required for establishing a sizeable population of these cells in the peripheral pools.
Article
Interleukin-2 (IL-2) signaling requires the dimerization of the IL-2 receptor beta.(IL-2R beta) and common gamma (gamma c) chains. Mutations of gamma c can result in X-linked severe combined immunodeficiency (XSCID). IL-2, IL-4, IL-7 (whose receptors are known to contain gamma c), and IL-9 (whose receptor is shown here to contain gamma c) induced the tyrosine phosphorylation and activation of the Janus family tyrosine kinases Jak1 and Jak3. Jak1 and Jak3 associated with IL-2R beta and gamma c, respectively; IL-2 induced Jak3-IL-2R beta and increased Jak3-gamma c associations. Truncations of gamma c, and a gamma c, point mutation causing moderate X-linked combined immunodeficiency (XCID), decreased gamma c-Jak3 association. Thus, gamma c mutations in at least some XSCID and XCID patients prevent normal Jak3 activation, suggesting that mutations of Jak3 may result in an XSCID-like phenotype.
Article
This chapter provides an overview of receptor activation from biochemical and cellular studies regarding immune system development and function when various components of the Interleukin-2-receptor (IL-2R) signal are disrupted. The biologic effects of IL-2R signals are much more complex than simply mediating T-cell growth, and depending upon the set of conditions, IL-2R signals may also promote cell survival, effector function, and apoptosis. These sometimes contradictory effects underscore the fact that a diversity of intracellular signaling pathways can be potentially activated by IL-2R. Three critical events that have been identified in the generation of the IL-2R signal for cell cycle progression include —heterodimerization of the cytoplasmic domains of the IL-2Rβ and γc chains, activation of the tyrosine kinase Jak3, and phosphorylation of tyrosine residues on the IL-2Rβ chain—leading to the creation of an activated receptor complex. One of the most important physiologic functions of the IL-2R in vivo is the homeostatic regulation of lymphoid tissues. IL-2R defects in mice lead to the development of fatal inflammatory disorders, which are accompanied by autoantibodies, suggesting that IL-2R signals also play a role in maintaining peripheral immune tolerance. The identification of IL-2R mutations in humans and the creation of such mutations in mice have provided some surprising and puzzling insights into the function of IL-2R signals in vivo. The pathogenic mechanism of inflammatory disorders in IL-2R-deficient mice is not well understood; however, the fact that IL-2R signals are now known to be required to suppress these conditions may help identify the causes of similar conditions in humans.
Article
Lethal autoimmunity associated with IL-2Rβ-deficient mice is prevented after thymic transgenic expression of wild-type IL-2Rβ in IL-2Rβ−/− mice (Tg −/− mice). Here, we show that CD4+CD25+ regulatory T cells were not readily detected in IL-2Rβ−/− mice, but the production of functional CD4+CD25+ T cells was reconstituted in Tg −/− mice. Adoptive transfer of normal CD4+CD25+ T cells into neonatal IL-2Rβ-deficient mice prevented this lethal autoimmune syndrome. The CD4+CD25+ T cells in disease-free adult IL-2Rβ-deficient recipient mice were present at a near normal frequency, were solely donor-derived, and depended on IL-2 for expansion. These observations indicate that the essential function of the IL-2/IL-2R system primarily lies at the level of the production of CD4+CD25+ regulatory T cells.