Вы находитесь на странице: 1из 6

Homeostatic imbalance of regulatory and effector T

cells due to IL-2 deprivation amplifies murine lupus


Jens Y. Humricha,1, Henner Morbachb, Reinmar Undeutscha, Philipp Engharda, Stefan Rosenbergera, Olivia Weigerta,
Lutz Klokea, Juliane Heimanna, Timo Gabera, Susan Brandenburgc, Alexander Scheffoldc, Jochen Huehna,d,
Andreas Radbruchc, Gerd-Rüdiger Burmestera, and Gabriela Riemekastena
a
Department of Rheumatology and Clinical Immunology, University Hospital Charité, 10117 Berlin, Germany; bPediatric Rheumatology, Immunology, and
Infectious Diseases, Children’s Hospital, University of Wuerzburg, 97080 Wuerzburg, Germany; cGerman Rheumatism Research Center, 10117 Berlin, Germany;
and dHelmholtz-Zentrum für Infektionsforschung GmbH, 38124 Braunschweig, Germany

Edited by Christophe Benoist, Harvard Medical School, Boston, MA, and approved November 17, 2009 (received for review March 24, 2009)

The origins and consequences of a regulatory T cell (Treg) disorder that results in an uncontrolled activation of self-reactive B and T
in systemic lupus erythematosus (SLE) are poorly understood. In cells and consequent multiorgan inflammation, most importantly
the (NZBxNZW) F1 mouse model of lupus, we found that nephritis (14, 15). Despite a few contradictory reports, most
CD4+Foxp3+ Treg failed to maintain a competitive pool size in studies have found a low prevalence of CD4+CD25+ T cells in
the peripheral lymphoid organs resulting in a progressive homeo- SLE patients and murine SLE models, suggesting a disturbed
static imbalance of CD4+Foxp3+ Treg and CD4+Foxp3− convention- maintenance of the Treg pool size (16–18). Nonetheless, the
al T cells (Tcon). In addition, Treg acquired phenotypic changes interpretation of most of these studies may be restricted because
that are reminiscent of IL-2 deficiency concomitantly to a progres- of the common identification of Treg by CD25, a surface marker
sive decline in IL-2-producing Tcon and an increase in activated, that is also expressed by activated conventional CD4+ T cells
IFN-γ-producing effector Tcon. Nonetheless, Treg from lupus- (Tcon) and absent in a large proportion of Treg.
prone mice were functionally intact and capable to influence the Impaired IL-2 production by T cells has also been attributed a
course of disease. Systemic reduction of IL-2 levels early in disease critical role in murine and human SLE (19, 20). However, it
promoted Tcon hyperactivity, induced the imbalance of Treg and remains unclear whether a shortage of IL-2 causes abnormalities
effector Tcon, and strongly accelerated disease progression. In
of the Treg population in lupus-prone individuals and how such a
contrast, administration of IL-2 partially restored the balance of
disturbance is causally linked to the pathogenesis of this disease.
Treg and effector Tcon by promoting the homeostatic proliferation
To address these fundamental questions, we explored the
of endogenous Treg and impeded the progression of established
origins and consequences of abnormalities in the CD4+Foxp3+
disease. Thus, an acquired and self-amplifying disruption of the
Treg pool during the course of SLE progression using
Treg-IL-2 axis contributed essentially to Tcon hyperactivity and
the development of murine lupus. The reversibility of this homeo-
(NZBxNZW) F1 lupus mice, a spontaneous autoimmune model
static Treg disorder provides promising approaches for the treat-
that displays many features of human SLE, including fatal
ment of SLE. nephritis (21). We found that the lupus-prone mice failed to
sustain a competitive number of CD4+Foxp3+ Treg in the pe-
homeostasis | immunotherapy | interleukin-2 | SLE | autoimmunity ripheral lymphoid organs because of an acquired deficiency of
IL-2 and IL-2-producing CD4+ T cells. This homeostatic im-
pairment of Treg boosted Tcon hyperactivity, resulting in a
egulatory CD4+ T cells (Treg) that express the transcription
R factor Foxp3 are crucial for the maintenance of immuno-
logical tolerance to self (1, 2). Predominantly derived from a
progressive imbalance of Treg and effector Tcon, and promoted
the progression of disease. Furthermore, we showed that com-
pensation of the IL-2 deficiency in lupus mice by treatment with
distinct T cell subpopulation in the thymus, CD4+Foxp3+ Treg
rIL-2 impeded the progression of established disease most likely
principally recognize self-antigens and are required to control
by re-establishing the homeostatic balance of Treg and effector
the expansion of self-reactive T cells in the peripheral lymphoid
Tcon, indicating the reversibility of this acquired Treg disorder.
organs (2, 3). In view of that, there is increasing evidence that
numeric or functional Treg deficiencies are associated with Results
particular autoimmune diseases, suggesting a contribution of a
Progressive Homeostatic Imbalance of Treg and Tcon. The percent-
Treg dysfunction to disease development (4).
age and absolute numbers of CD4+Foxp3+ Treg were evaluated
The cytokine IL-2 was initially identified as a potent T cell
in different organs and at different time points during the de-
growth factor (5). However, more recent data strongly indicate
velopment of disease in (NZBxNZW) F1 mice by flow cytometry.
that IL-2 is essential for immune tolerance (5). Accordingly,
A comparison between young animals (young), animals at the
mice deficient in IL-2 or IL-2 receptor components, including
onset of disease (onset), and old, diseased animals (diseased)
CD25, succumb to a rapidly progressing autoimmune disease
showed a progressive deficiency in the number of Treg in the
that is caused by an uncontrolled activation of CD4+ T cells and
B cells (6–8). The fundamental function of IL-2 in Treg biology lymph nodes and the peripheral blood. (Fig. 1 A and B). This
was recently highlighted with the demonstration that IL-2 was deficiency of Treg was contrasted to progressive increases in the
critically required for the homeostatic maintenance of Treg in
the peripheral lymphoid organs (9–11). Other studies have also
Author contributions: J.Y.H., A.R., and G.R. designed research; J.Y.H., H.M., R.U., P.E., S.R.,
suggested a requirement of IL-2 for the suppressive function and O.W., L.K., J. Heimann, and S.B. performed research; A.S. and J. Huehn contributed new
the thymic development of Treg (12, 13). Therefore, dis- reagents/analytic tools; J.Y.H., H.M., T.G., and A.S. analyzed data; and J.Y.H., A.R., G.-R.B.,
turbances in the Treg-IL-2 axis can result in autoimmunity or and G.R. wrote the paper.
contribute to the development of immune-mediated diseases. The authors declare no conflict of interest.
Systemic lupus erythematosus (SLE) is a prototypic systemic This article is a PNAS Direct Submission.
autoimmune disease with complex genetics and unknown etiol- 1
To whom correspondence should be addressed. E-mail: humrich@drfz.de.
ogy. It is characterized by a breakdown of tolerance to ubiquitous This article contains supporting information online at www.pnas.org/cgi/content/full/
nuclear antigens, including double-stranded DNA (ds-DNA), 0903158107/DCSupplemental.

204–209 | PNAS | January 5, 2010 | vol. 107 | no. 1 www.pnas.org/cgi/doi/10.1073/pnas.0903158107


Fig. 1. Progressive homeostatic imbalance
of Treg and Tcon. (A and B) Average per-
centage of Foxp3+ cells among CD4+ and
among CD4 SP T cells (A) and absolute
numbers (B) of CD4+Foxp3+ T cells in spleens
(SN), lymph nodes (LN), peripheral blood
(PB), and of CD4 SP Foxp3+ T cells in thymi
(TH) from (NZBxNZW) F1 mice (filled bars) at
the indicated disease stages compared to
age-matched BALB/c mice (open bars) de-
termined by flow cytometry. Data represent
the mean of 6–10 mice per group from two
to three independent experiments with
three to four mice per group. (C) Calculated
ratios of the percentage of BrdU+ cells
among CD4+Foxp3+ and CD4+Foxp3− cells
from (NZBxNZW) F1 (filled bars) compared to
age-matched BALB/c mice (open bars) in the
indicated organs and at the indicated dis-
ease stages. Data represent the mean of
3–10 mice per group from one to three in-
dependent experiments with two to four
mice per group. Error bars indicate SEM (*, P <
0.05; **, P < 0.01; ***, P < 0.001 vs. BALB/c).

percentage and absolute numbers of Treg in the spleens and of changes were not observed in the clinically healthy parental
CD4 single positive (SP) Treg in the thymi (Fig. 1 A and B). NZW strain, except for a moderate activation, or the healthy
Next, we studied the in vivo proliferation of CD4+Foxp3+ BALB/c strain (Fig. 2 and Fig. S2). Together, Treg and Tcon
Treg and CD4+Foxp3− Tcon by BrdU incorporation. The pro- from lupus-prone mice acquire phenotypic changes similar to
liferation rates of Treg and Tcon in lymphoid organs from young those previously described in IL-2-deficient mice (9).
(NZBxNZW) F1 mice were not different from those in aged-
matched BALB/c mice (Fig. S1A). However, this changed in Progressive Decline in IL-2-Producing CD4+ T Cells. To assess whether
lupus mice at the onset of disease: the proliferation rate of Treg lupus-prone mice develop IL-2 deficiency, we determined the
was now lower in spleens and lymph nodes (Fig. S1A) compared percentage of CD4+ T cells that are capable to produce IL-2 and
to aged-matched BALB/c mice, whereas the proliferation rate of IFN-γ during disease development in (NZBxNZW) F1 mice. In
CD4 SP Treg was increased in the thymus (Fig. S1A). The comparing young, healthy (NZBxNZW) F1 and age-matched
changes in Treg proliferation were accompanied by a continuous BALB/c mice, we did not observe substantial differences in the
increase in the proliferation rate of Tcon in the peripheral percentage of IL-2-producing CD4+ T cells in either spleens or
lymphoid organs and in the peripheral blood that was not ob- lymph nodes (Fig. 3 A and B). Consistent with our observation of
served in age-matched BALB/c mice (Fig. S1B). Additionally, the acquired predominance of effector/memory Tcon, CD4+ T
and in contrast to BALB/c mice, the majority of proliferating cells acquired the ability to produce IFN-γ as the disease pro-
CD4+ T cells consisted of CD44+ effector/memory T cells in lupus gressed (Fig. 3 A and B). In parallel, CD4+ T cells lost the ability
mice (Fig. S1C). The calculated ratio between proliferating to produce IL-2 (Fig. 3 A and B). Moreover, the IFN-
CD4+Foxp3+ Treg and CD4+Foxp3− Tcon—representing a γ-producing CD4+ T cells from (NZBxNZW) F1 mice exhibited
measure of the homeostatic balance between Treg and Tcon—was diminished IL-2 production compared to BALB/c mice (Fig. 3 A
therefore progressively reduced in the lymphoid organs from and B). To confirm the shortage of IL-2 in lymphoid organs, we
(NZBxNZW) F1 mice compared to that of age-matched BALB/c determined the spontaneous IL-2 secretion in 24-h cell cultures
mice (Fig. 1C). Consistent with the increased proliferation of CD4 from spleen and lymph node cells at different disease stages. In
SP Treg, an increased Treg:Tcon ratio was observed in the thymus addition, we determined IL-2 levels in the plasma during disease
(Fig. 1C). Collectively, these data indicated an acquired impairment progression. In line with the decline of CD4+ IL-2-producing T
of the homeostatic balance between Treg and effector Tcon in cells, spontaneous IL-2 secretion in lymphoid organs from
parallel to a partial deficiency of Treg in the periphery. (NZBxNZW) F1 mice was also progressively diminished from
the disease onset compared to age-matched BALB/c mice in
Phenotypic Changes of Treg and Tcon During Disease Development. parallel to decreasing IL-2 levels in the plasma (Fig. 3 C–E).
Next, we analyzed the phenotype of CD4+Foxp3+ Treg and
CD4+Foxp3− Tcon from lupus-prone (NZBxNZW) F1 mice Treg from Lupus-Prone Mice Are Functionally Intact and Influence
IMMUNOLOGY

during the course of disease. The percentage of CD25+ cells Disease Course. Loss of suppressive function of Treg results in the
among Treg decreased with advancing disease in (NZBxNZW) appearance of systemic autoimmune syndromes (1, 2). We found
F1 mice (Fig. 2A and Fig. S2A). This was accompanied by an that Treg of both young and diseased (NZBxNZW) F1 mice
increase in the percentage of CD25+ cells among Tcon (Fig. 2A displayed similar suppressive functions in vitro (Fig. 4A) and
and Fig. S2A). Compared to age-matched BALB/c mice, young similar Foxp3 mRNA expression levels compared to age-
(NZBxNZW) F1 mice already had higher frequencies of CD69+ matched BALB/c mice (Fig. S3A).
and CD44+ cells among both Treg and Tcon that strongly in- To gain insights into the role of Treg in preventing disease
creased with disease progression (Fig. 2 B and D and Fig. S2B development, we reduced the numbers of Treg in young
and D). In contrast, the percentage of CD62L+ cells among Treg (NZBxNZW) F1 mice with a single injection of depleting anti-
and Tcon markedly declined concomitantly with the increase in bodies against CD25. This resulted in a transient (≈2 weeks)
CD44+ cells (Fig. 2C and Fig. S2C). Similar phenotypic changes contraction of the CD4+Foxp3+ Treg pool in the peripheral blood
were present in the parental autoimmune-prone NZB strain that (Fig. S3B), was followed by higher frequencies of CD4+CD44+
develops a milder form of lupus. In contrast, these phenotypic Tcon in the peripheral blood (Fig. S3B), and resulted in an

Humrich et al. PNAS | January 5, 2010 | vol. 107 | no. 1 | 205


Fig. 3. Decline in IL-2-producing CD4+ T cells. Representative dot plots show
the percentage of IL-2- and IFN-γ-producing cells among CD4+-gated spleen
(A) and lymph node (B) cells from 6- to 9-month-old BALB/c and diseased
(NZBxNZW) F1 mice (NZBW). Numbers in quadrants indicate the percentage
of the designated population. The respective bar diagrams show the average
percentage of IL-2+ and IFN-γ+ cells among CD4+ T cells from (NZBxNZW) F1
mice (filled bars) during disease progression compared to age-matched
BALB/c mice (open bars). Data represent the mean of three to six mice per
group from one to two independent experiments with three mice per group.
Fig. 2. Phenotypic changes of Treg and Tcon during disease progression.
(C–E) Bar diagrams show IL-2 concentrations determined by ELISA from su-
Average expression of CD25 (A), CD69 (B), CD62L (C), and CD44 (D) on
pernatants of spleen (C) and lymph node cells (D) cultured for 24 h and from
CD4+Foxp3+- and CD4+Foxp3−-gated cells from spleens of BALB/c (white
plasma (E) of (NZBxNZW) F1 mice (filled bars) at the indicated disease stages
bars), NZW (light gray bars), NZB (black bars), and (NZBxNZW) F1 mice (dark
compared to age-matched BALB/c mice (open bars). Data represent the
gray bars) at the age of 8–10 weeks (young) or 6–9 months (aged), re-
mean of four to five mice per group. Error bars indicate SEM (*, P < 0.05; **,
spectively. Data were determined by flow cytometry and represent the mean
P < 0.01 vs. BALB/c).
of 3–10 mice per group from one to three independent experiments with
three to four mice per group. Error bars indicate SEM (*, P < 0.05; **, P <
0.01; ***, P < 0.001 vs. BALB/c).
pression of CD25 remained either unaffected 1–3 weeks after IL-2
neutralization or even considerably increased in the spleen and in
acceleration of disease progression indicated by the development the peripheral blood at later time points (Fig. S4 B–D). Thus, IL-2
of nephritis ≈4–6 weeks earlier than controls (Fig. 4B). Consistent deficiency affected nearly exclusively the CD25+ subpopulation of
with these observations, the Treg-depleted mice also had a reduced Foxp3+ Treg with differing kinetics and impacts in the peripheral
survival rate (Fig. 4B). However, no acceleration of anti-ds-DNA- blood, in the lymph nodes, and in the spleen, respectively. In
antibody titers was observed (Fig. S3B). parallel, we observed a continuous increase of CD69+ and CD44+
To test whether increasing the size of the peripheral Treg cells among both the Treg and Tcon in the lymphoid organs (Fig.
pool could influence already established disease, purified 5B) and of CD44+ Tcon in the peripheral blood (Fig. S4E), but we
CD4+Foxp3+CD25+ Treg were adoptively transferred into mice did not observe an increase in anti-ds-DNA-IgG titers (Fig. S4F).
Thus, IL-2 deficiency induced similar abnormalities in Treg and
with active disease. Because the IL-2-deficient environment in
Tcon that are present in untreated mice with advanced disease.
diseased animals would affect the survival and metabolic fitness
Neutralization of IL-2 also resulted in a strong acceleration of
of the transferred Treg, Treg were activated for 24 h in the
nephritis (Fig. 5C) and significantly increased mortality compared
presence of high doses of IL-2 before transfer (Fig. S3C).
to isotype-treated control mice (Fig. 5D). Together, IL-2 was
Adoptive transfer of Treg delayed disease progression and sig- necessary to maintain a pool size of Treg in the periphery that was
nificantly increased the survival time (Fig. 4C). Again, there was competitive enough to prevent Tcon hyperactivity and disease
no significant change observed in the anti-ds-DNA IgG antibody progression in lupus-prone mice.
titers at any time point analyzed (Fig. S3D). Thus, CD4+Foxp3+
Treg from lupus-prone mice were functionally intact and phys- IL-2 Partially Restores Treg:Tcon Balance and Ameliorates Established
iologically relevant inhibitors of disease progression. Moreover, Disease. We next examined whether IL-2 was capable of influ-
these data suggested that efficient regulation of CD4+ Tcon encing the endogenous Treg pool in (NZBxNZW) F1 mice.
activation and disease progression in lupus-prone mice de- Treatment of onset and diseased lupus mice with recombinant
pended on the presence of sufficient numbers of Treg. IL-2 (rIL-2) strongly promoted the expansion of endogenous
CD4+Foxp3+ Treg in spleens (1.5-fold; Fig. 6A), lymph nodes
IL-2 Is Required to Retain Treg:Tcon Balance and to Impede Disease (1.5-fold; Fig. S5A), and peripheral blood (2-fold; Fig. S5A)
Progression. To explore the role of IL-2 in the pathogenesis of compared to the PBS-treated control mice. In parallel, Treg
murine lupus, we reduced systemic IL-2 levels with neutralizing displayed a remarkable increase in the surface expression of
antibodies in young and clinically healthy (NZBxNZW) F1 mice. CD25 (Fig. 6A) and an increase in the expression of Foxp3 on a
This induced a strong and persistent reduction in the percentage of per cell basis in rIL-2-treated mice (Fig. S5B). Analysis of BrdU
CD4+Foxp3+CD25+ Treg in the peripheral blood (Fig. S4A), in incorporation in vivo revealed a 2- to 3-fold increase in the
the lymph nodes (Fig. 5A), and later also in the spleen (Fig. 5A). In proliferative activity of Treg in rIL-2-treated versus untreated
contrast, the percentage of CD4+Foxp3+ Treg that lacked ex- mice (Fig. 6B). Although an increased proliferation of Tcon was

206 | www.pnas.org/cgi/doi/10.1073/pnas.0903158107 Humrich et al.


Fig. 4. Treg are functionally intact and influence disease progression. (A) In
vitro suppression assays compare the suppressive capacity of CD4+CD25+
Treg from (NZBxNZW) F1 and age-matched BALB/c mice. The proliferation of
CFDA-SE-labeled responder cells in the presence of titrated numbers of Treg
was analyzed by flow cytometry and is presented in percent of the pro-
liferation of responder cells without Treg (taken as 100%). Error bars in-
dicate SD from triplicate cultures. One representative of two to three
independent experiments is shown. (B) Nephritis development, presented as
average proteinuria score, and survival, presented as Kaplan–Meier curves
(P = 0.116; n = 10 per group), after injection of depleting antibodies against
CD25 (anti-CD25) into 8- to 10-week-old, clinically healthy (NZBxNZW) F1
mice compared to rat IgG1-treated controls (Control). (D) Average protei-
nuria score and survival (P = 0.0439) after adoptive transfer of 5 × 105 acti- Fig. 5. IL-2 neutralization induces Treg:Tcon imbalance and accelerates
vated CD4+CD25+Foxp3+ cells into diseased (NZBxNZW) F1 mice at the age of disease. (A and B) Flow cytometry of CD4+-gated T cells 21 and 42 days after
6.5 months (Foxp3+) compared to a PBS-treated control group (Control). One injection of anti-IL-2 antibodies (anti-IL-2; filled bars) into 9- to 10-week-old,
of two independent experiments is shown (n = 5 per group). (B and C) Error clinically healthy (NZBxNZW) F1 mice compared to rat IgG-treated controls
bars indicate SEM. (Control; open bars). Representative dot plots show the percentage of CD25+
(A) and of CD69+ and CD44+ (B) cells among Foxp3+ and Foxp3− cells in spleens
42 days after IL-2 neutralization. Numbers in quadrants indicate the per-
also induced by administration of rIL-2, proliferation of Treg was centage of the designated population. Bar diagrams show the average per-
favored as shown by the higher ratio between BrdU+ Treg and centage of Foxp3+CD25+ cells among CD4+ T cells in spleens (SN) and lymph
nodes (LN) and of CD69+ and CD44+ cells among CD4+Foxp3+ and CD4+Foxp3−
BrdU+ Tcon (Fig. 6B). Short-term rIL-2-treatment (3× 2 μg/24 h)
cells in spleens. Data represent the mean + SD (n = 3 per group and time
of diseased lupus mice already delayed disease progression (Fig. point). (C and D) Nephritis development (C) and survival (D) after injection of
S5C) and significantly decreased mortality (Fig. S5D); however, it 1 mg of neutralizing anti-IL-2 antibodies (anti-IL-2) into young, clinically
did not lead to a persistent expansion of the Treg pool (Fig. S5E). healthy (NZBxNZW) F1 mice compared to rat IgG-treated controls (Control);
In contrast, repetition of rIL-2 injections in intervals of 5 days nephritis development is presented as average proteinuria score and survival
after the initial short-term treatment for a total of four times re- as Kaplan–Meier curves (P = 0.0197). One representative of two independent
sulted in a much more stable Treg expansion, especially of CD25+ experiments is shown (n = 5 per group). Error bars indicate SEM.
Treg, which was still detectable >10 days after the last rIL-2 in-
jection (Fig. S5 F–H). In addition, the repetitive rIL-2 regimen
proliferation of Treg in lupus-prone mice at the onset of disease.
efficiently impeded disease progression (Fig. 6C) and considerably
This contrasted with a progressive increase in the proliferation of
prolonged the survival of diseased mice (Fig. 6D). Together, these
data indicated that the acquired IL-2 deficiency could be com- effector Tcon starting from the onset of disease, indicating a
pensated with exogenous rIL-2 and that the resulting re-estab- progressive homeostatic imbalance between Treg and effector
lishment of the Treg:Tcon balance was associated with a beneficial Tcon. These data also suggested that the peripheral pool of Treg
outcome in murine lupus. failed to expand adequately enough to compete with the en-
hanced proliferation of Tcon, a condition similar to that recently
IMMUNOLOGY

Discussion described in IL-2-deficient mice (9, 10). Phenotypic analyses


Perturbations in Treg biology can elicit autoimmune syndromes revealed further similarities between IL-2-deficient and diseased
and are associated with certain autoimmune diseases. Here we (NZBxNZW) F1 lupus mice, including a progressive reduction in
analyzed CD4+Foxp3+ Treg in the (NZBxNZW) F1 model of the expression of CD25 in Treg, the activated, memory-like phe-
SLE to identify and characterize disturbances in Treg biology notype of Treg, and the progressive increase in CD44+CD62L−
and investigate how they contributed to the pathogenesis of this effector Tcon that predominately belong to the Th1 lineage (7, 9).
systemic autoimmune disease. Indeed, we could confirm a state of IL-2 deficiency in lupus-prone
Identification of Treg by expression of Foxp3 revealed a par- mice by the detection of a progressive deficiency of IL-2 and IL-
tial deficiency in the numbers of Treg in (NZBxNZW) F1 lupus 2-producing Tcon in the lymphoid tissues. This deficiency was
mice. This deficiency affected lymph nodes and the peripheral characterized by a decline in single IL-2-producing Tcon, which
blood, and was progressive during disease development. Along predominantly belong to the naïve Tcon repertoire (5), and a di-
with the partial deficiency, we found a diminished homeostatic minished IL-2 production by IFN-γ+ effector Tcon. The decline in

Humrich et al. PNAS | January 5, 2010 | vol. 107 | no. 1 | 207


model after treatment with IL-2-expressing recombinant vectors
(24, 25). Therefore, therapeutic strategies that target the Treg-
IL-2 axis as a treatment for SLE are worth exploring in more detail.
Further research should aim to improve the selectivity of IL-2
treatments to favor exclusively Treg proliferation.
The pathogenic relevance of the impaired Treg-IL-2 axis in
murine lupus, shown here, raises the question of the origin of the
IL-2 deficiency. Both genetic and acquired alterations have been
considered important for the IL-2 deficiency observed in lupus
and other autoimmune diseases (26–28). In our study, the defi-
ciency of IL-2 and IL-2-producing CD4+ Tcon in the
(NZBxNZW) F1 lupus model was not evident before the onset of
disease and did not precede Tcon hyperactivity and Treg defi-
ciency in general. The low prevalence of CD4+Foxp3+ Treg in
young, clinically healthy lupus mice also suggests that factors
other than IL-2 may be involved in regulating the size of the
peripheral Treg pool. This is supported by recent studies in
congenic mouse strains related to the (NZBxNZW) F1 strain
that indicated that the low prevalence of CD25+ Treg was linked
to a disease-related locus, the so-called Sle1a locus (29). Al-
though the responsible genes have not been determined so far, it
is known that the Sle1a locus does not encode for genes involved
in IL-2 transcription or IL-2 signaling (29). Alternatively, the
peripheral Treg deficiency may be caused by an impaired thymic
Fig. 6. rIL-2 restores Treg:Tcon balance and impedes disease progression. Treg generation. Instead, we found that the numbers and pro-
(A) Flow cytometry of CD4+ T cells from spleens of 5-month-old (NZBxNZW) liferation rates of thymic Treg were normal in young
F1 mice 12 h after three injections of 2 μg of rIL-2 per mouse within 24 h (rIL- (NZBxNZW) F1 mice and even increased during disease pro-
2) compared to PBS-treated mice (Control). Representative dot plots show gression pointing to a mechanism that attempts to compensate
the percentage of CD25+ and Foxp3+ cells among CD4+-gated T cells. Bar for the Treg deficiency in the peripheral lymphoid organs. To-
diagrams show the average percentage of the indicated parameters within gether, we propose that IL-2 deficiency in lupus is secondary,
the respective population (mean + SD; n = 3 per group). (B) Representative acquired mainly because of the displacement of naïve, IL-2-
dot plots and bar diagrams show the percentage of BrdU+ cells among
CD4+Foxp3+ T cells in spleens from 7.5 month old (NZBxNZW) F1 mice (filled
producing Tcon by effector Tcon. The IL-2 deficiency arises
bars) compared to age-matched BALB/c mice (open bars) 4 days after a 24-h
because effector Tcon do not contribute sufficiently to overall
treatment with rIL-2 and daily injections of BrdU. The calculated ratio of the IL-2 production. Thus, the IL-2 deficiency in lupus may be the
percentage BrdU+ cells among CD4+Foxp3+ and CD4+Foxp3− cells from result of an uncontrolled Tcon hyperactivity.
controls (open bar) and rIL-2-treated (NZBxNZW) F1 mice (filled bar) is In contrast to the progressive deficiency observed in the
compared in a bar diagram. Data represent the mean of three to eight mice peripheral blood and lymph nodes, a continuous increase of the
per group from one to three independent experiments (*, P < 0.05; **, P < Treg subpopulation that lacked expression of CD25 was observed
0.01 vs. control). Numbers in quadrants indicate the percentage in the des- in the spleen, leading to a >4-fold increase in the absolute numbers
ignated population. (C and D) Nephritis development, presented as average in diseased (NZBxNZW) F1 mice compared to young (NZBxNZW)
proteinuria score (C), and survival, presented as Kaplan–Meier curves (D) (P =
F1 and aged-matched BALB/c mice. At present, it is not entirely
0.0035) after repetitive treatment of 6.3-month-old, diseased (NZBxNZW) F1
mice with 2 μg of rIL-2 per mouse every 5 days for a total of four times (until
clear why there are such differences in the distribution of Treg in this
day 20) after an initial short-term treatment with three injections of 2 μg of lupus model. However, the diminished or moderate in vivo pro-
rIL-2 per mouse within 24 h (rIL-2) compared to an aged-matched PBS- liferation rates observed in splenic Treg suggest that an accumu-
treated control group (Control) (n = 8 per group). Error bars indicate SEM. lation of Treg in this highly inflamed tissue is more likely than an
expansion in the organ itself. Therefore, the partial deficiency of
Treg found in lymph nodes and in the peripheral blood may also be,
IL-2 levels and IL-2-producing Tcon was associated with pro- in part, the result of an enhanced attraction of Treg to the spleen.
gressive Treg abnormalities and disease progression, suggesting Interestingly, an increase of splenic Foxp3+ Treg that lacked ex-
that an acquired IL-2 deficiency contributes to disease develop- pression of CD25 was induced in healthy lupus mice by neutrali-
ment. This was proven by several experiments. First, neutralization zation of IL-2, suggesting that IL-2 deficiency plays an important
of IL-2 in young, clinically healthy animals induced the homeostatic role in this phenomenon.
imbalance of Treg and effector Tcon, a characteristic of advanced Taking all our results into consideration, we propose a sim-
disease, and strongly accelerated lupus. Second, treatment of dis- plified model that may explain the gradually acquired and self-
eased lupus mice with rIL-2 partially restored the balance of Treg amplifying failure of the CD4+Foxp3+ Treg pool to compete in
and effector Tcon by preferentially increasing the proliferation of systemic autoimmunity (Fig. S6). In this model, several initial
Treg, and considerably impeded disease progression, and the ef- events lead to Tcon hyperactivity. Early in the disease, at least
ficacy was further enhanced by a repetitive regimen. partial compensation is provided by a qualitatively intact yet
Nonetheless, treatment with rIL-2 also resulted in an increased quantitatively already restricted Treg system. The origins of
activation and proliferation of CD4+Foxp3− Tcon. Although this spontaneous Tcon hyperactivity in lupus are currently speculative
may carry the risk of increasing disease activity (22, 23), our data but could include genetic alterations in antigen-presenting cells
show that in the active phase of the disease and in an IL-2-deprived that lead to an increase in proinflammatory cytokine production or
environment, IL-2 acted as a Treg adjuvant that outweighed its an altered context of self-antigen presentation to autoreactive
simultaneous fueling of the Tcon pool. Apart from that, it was Tcon (30). This may also promote resistance of autoreactive Tcon
recently shown in a model of type 1 diabetes that the potency of to Treg-mediated suppression, additionally facilitating their acti-
IL-2 to increase the activation of Tcon and NK T cells depended on vation, differentiation, and consecutive expansion (31). With dis-
the administered dosage (23). In line with our data, recovery from ease progression, the ongoing conversion of naive Tcon into
autoimmunity was also observed in the MRL/lpr lupus mouse effector Tcon and their displacement results in a progressive

208 | www.pnas.org/cgi/doi/10.1073/pnas.0903158107 Humrich et al.


decline in IL-2 production. This acquired IL-2 deficiency further sciences). The concentration of anti-ds-DNA antibodies in the plasma was
impedes the peripheral Treg pool to expand sufficiently to com- determined by ELISA.
pete with the hyperactivity of Tcon. The resulting vicious cycle is
characterized by a progressive homeostatic imbalance between Suppression Assays. CD4+CD25+ and CD4−CD25− cells were obtained from
Treg and effector Tcon. Once the Treg pool declines below a (NZBxNZW) F1 mice and age-matched BALB/c mice by MACS (Miltenyi Bio-
critical size, the remaining compensatory mechanisms collapse tec). CD4+CD25− T cells from young syngenic mice were used as responder
cells in all experiments. The proliferation of CFDA-SE-labeled responder cells
and the disease manifests itself.
in the presence of titrated numbers of CD4+CD25+ cells was determined by
In summary, an acquired and self-amplifying impairment of
flow cytometry.
the Treg-IL-2 axis induced a progressive homeostatic imbalance
of Treg and effector Tcon, and promoted the development of
Adoptive Transfer of Treg. Purified and activated CD4+CD25+Foxp3+ Treg
disease. The reversibility of this homeostatic impairment, shown
from young (NZBxNZW) F1 mice were injected i.v. into (NZBxNZW) F1 mice at
here, provides rationales and approaches for a Treg- and IL-2- the age of between and 6.5 and 7.5 month (0.5–1.0 × 106 cells per mouse).
based immunotherapy of SLE.
Treg Depletion and IL-2 Neutralization. (NZBxNZW) F1 mice at the age of 8–15
Materials and Methods
weeks received 100 μg of depleting antibodies against CD25 (eBioscience
See SI Materials and Methods for details.
PC61) i.v. or 1 mg of neutralizing anti-IL-2 antibodies (DRFZ, S4B6.1) by i.p.
injection as previously described in other mouse strains (11); age-matched
Mice. Female (NZBxNZW) F1, NZW, NZB, and BALB/c mice were bred and
controls received either 100 μg of rat IgG1 (R&D Systems 43414.1) or 1 mg of
maintained under specific-pathogen-free conditions in the Deutsches Rheu-
rat IgG (Jackson ImmunoResearch), respectively.
maforschungszentrum (DRFZ) and were used for experiments at between 6
weeks and 12 months of age in accordance with institutional and federal
guidelines. Mice were divided into groups according to age and disease ac- rIL-2 Treatment. Mice at different ages received 2 μg of rmIL-2 (R&D Systems) in
tivity, as determined by the degree of nephritis (young, onset, and diseased). PBS or PBS alone i.p. every 12 h within 24 h (three times in total; short-term
Proteinuria was determined with Multistix 10 Visual (Bayer Diagnostics). treatment) 12–96 h before analysis. For survival experiments, (NZBxNZW) F1
mice with established disease either were treated with a short-term treat-
Flow Cytometry. Multicolor flow cytometry was performed with a FACSCa- ment of rIL-2 alone or the treatment was repeated every 5 days with 2 μg of
libur cytometer (BD Biosciences). Results were processed by using CellQuest rIL-2 for a total of four times after the initial short-term treatment (repetitive
software (BD Biosciences). FACSDiva (BD Biosciences) and FACSAria (BD treatment).
Biosciences) cell sorters were used for cell sorting.
Statistical Analysis. GraphPad Prism 4 software was used for the analysis of
In Vivo Proliferation Analysis. Mice of different age groups were injected i.p. survival curves (log-rank test, Kaplan–Meier curve). The Mann–Whitney U
every 24 h for 4 days with 40 mg/kg bodyweight BrdU (BD Biosciences). BrdU test (two-tailed) was used to detect statistical significant differences. P val-
incorporation was analyzed by flow cytometry (BrdU Flow kit; BD Biosciences). ues of <0.05 were regarded as significant.

IL-2 Production and ELISA. IL-2 concentrations in cell culture supernatants ACKNOWLEDGMENTS. We thank Hyun-Dong Chang for critically reading
from spleens and lymph nodes and in the plasma of (NZBxNZW) F1 and BALB/c the manuscript. This work was supported by the Deutsche Forschungsge-
mice at different ages were determined by ELISA (IL-2 ELISA kit; BD Bio- meinschaft (SFB 650) and grants from the University Hospital Charité Berlin.

1. Fontenot JD, Gavin MA, Rudensky AY (2003) Foxp3 programs the development and 18. Alexander T, et al. (2009) Depletion of autoreactive immunologic memory followed
function of CD4+CD25+ regulatory T cells. Nat Immunol 4:330–336. by autologous hematopoietic stem cell transplantation in patients with refractory SLE
2. Sakaguchi S (2005) Naturally arising Foxp3-expressing CD25+CD4+ regulatory T cells induces long-term remission through de novo generation of a juvenile and tolerant
in immunological tolerance to self and non-self. Nat Immunol 6:345–352. immune system. Blood 113:214–223.
3. Kim JM, Rasmussen JP, Rudensky AY (2007) Regulatory T cells prevent catastrophic 19. Linker-Israeli M, et al. (1983) Defective production of interleukin 1 and interleukin 2
autoimmunity throughout the lifespan of mice. Nat Immunol 8:191–197. in patients with systemic lupus erythematosus (SLE). J Immunol 130:2651–2655.
4. Costantino CM, Baecher-Allan CM, Hafler DA (2008) Human regulatory T cells and 20. Dauphinée MJ, Kipper SB, Wofsy D, Talal N (1981) Interleukin 2 deficiency is a
autoimmunity. Eur J Immunol 38:921–924. common feature of autoimmune mice. J Immunol 127:2483–2487.
5. Malek TR (2008) The biology of interleukin-2. Annu Rev Immunol 26:453–479. 21. Theofilopoulos AN, Dixon FJ (1985) Murine models of systemic lupus erythematosus.
6. Sadlack B, et al. (1993) Ulcerative colitis-like disease in mice with a disrupted Adv Immunol 37:269–390.
interleukin-2 gene. Cell 75:253–261. 22. Knoechel B, Lohr J, Kahn E, Bluestone JA, Abbas AK (2005) Sequential development
7. Sadlack B, et al. (1995) Generalized autoimmune disease in interleukin-2-deficient of interleukin 2-dependent effector and regulatory T cells in response to endogenous
mice is triggered by an uncontrolled activation and proliferation of CD4+ T cells. Eur J systemic antigen. J Exp Med 202:1375–1386.
Immunol 25:3053–3059. 23. Tang Q, et al. (2008) Central role of defective interleukin-2 production in the
8. Suzuki H, et al. (1995) Deregulated T cell activation and autoimmunity in mice lacking triggering of islet autoimmune destruction. Immunity 28:687–697.
interleukin-2 receptor beta. Science 268:1472–1476. 24. Gutierrez-Ramos JC, Andreu JL, Revilla Y, Viñuela E, Martinez C (1990) Recovery from
9. Fontenot JD, Rasmussen JP, Gavin MA, Rudensky AY (2005) A function for interleukin autoimmunity of MRL/lpr mice after infection with an interleukin-2/vaccinia
2 in Foxp3-expressing regulatory T cells. Nat Immunol 6:1142–1151. recombinant virus. Nature 346:271–274.
10. D’Cruz LM, Klein L (2005) Development and function of agonist-induced CD25+Foxp3+ 25. Huggins ML, Huang FP, Xu D, Lindop G, Stott DI (1999) Modulation of autoimmune
regulatory T cells in the absence of interleukin 2 signaling. Nat Immunol 6:1152–1159. disease in the MRL-lpr/lpr mouse by IL-2 and TGF-beta1 gene therapy using
11. Setoguchi R, Hori S, Takahashi T, Sakaguchi S (2005) Homeostatic maintenance of attenuated Salmonella typhimurium as gene carrier. Lupus 8:29–38.
natural Foxp3(+) CD25(+) CD4(+) regulatory T cells by interleukin (IL)-2 and induction 26. Crispin JC, Alcocer-Varela J (1998) Interleukin-2 and systemic lupus erythematosus—
of autoimmune disease by IL-2 neutralization. J Exp Med 201:723–735. fifteen years later. Lupus 7:214–222.
IMMUNOLOGY

12. Brandenburg S, et al. (2008) IL-2 induces in vivo suppression by CD4(+)CD25(+)Foxp3(+) 27. Yamanouchi J, et al. (2007) Interleukin-2 gene variation impairs regulatory T cell
regulatory T cells. Eur J Immunol 38:1643–1653. function and causes autoimmunity. Nat Genet 39:329–337.
13. Malek TR, Yu A, Vincek V, Scibelli P, Kong L (2002) CD4 regulatory T cells prevent 28. Juang YT, et al. (2005) Systemic lupus erythematosus serum IgG increases CREM
lethal autoimmunity in IL-2Rbeta-deficient mice. Implications for the nonredundant binding to the IL-2 promoter and suppresses IL-2 production through CaMKIV. J Clin
function of IL-2. Immunity 17:167–178. Invest 115:996–1005.
14. Kotzin BL (1996) Systemic lupus erythematosus. Cell 85:303–306. 29. Cuda CM, Wan S, Sobel ES, Croker BP, Morel L (2007) Murine lupus susceptibility locus
15. Riemekasten G, Hahn BH (2005) Key autoantigens in SLE. Rheumatology (Oxford) 44: Sle1a controls regulatory T cell number and function through multiple mechanisms. J
975–982. Immunol 179:7439–7447.
16. Miyara M, et al. (2005) Global natural regulatory T cell depletion in active systemic 30. Baccala R, Hoebe K, Kono DH, Beutler B, Theofilopoulos AN (2007) TLR-dependent
lupus erythematosus. J Immunol 175:8392–8400. and TLR-independent pathways of type I interferon induction in systemic
17. Scalapino KJ, Tang Q, Bluestone JA, Bonyhadi ML, Daikh DI (2006) Suppression of autoimmunity. Nat Med 13:543–551.
disease in New Zealand Black/New Zealand White lupus-prone mice by adoptive 31. Wan S, Xia C, Morel L (2007) IL-6 produced by dendritic cells from lupus-prone mice
transfer of ex vivo expanded regulatory T cells. J Immunol 177:1451–1459. inhibits CD4+CD25+ T cell regulatory functions. J Immunol 178:271–279.

Humrich et al. PNAS | January 5, 2010 | vol. 107 | no. 1 | 209

Вам также может понравиться