Yonsei Med J http://www.eymj.org Volume 51 Number 4 July 2010
484
CD4+ T helper (Th) precursor cells are activated by the antigenic stimulation of T
cell receptor (TCR) and are subsequently differentiated into different subsets of
effector Th cells in order to boost the immune responses.
1
Th1 and Th2 cells are
traditionally thought to be the major subsets generated upon antigenic stimulation
and produce distinct cytokine interferon
γ
(IFN
γ
) and interleukin (IL)-4, which are
then involved in the elimination of intracellular and extracellular pathogens, res-
pectively.
2
Coordinated cytokine signaling induces the activation of specific
transcription factors to promote lineage-specific cytokine production. While T-
box-containing protein expressed in T cells (T-bet) is activated by IL-12 and IFN
γ
and is exclusively expressed in Th1 cell differentiation,
3
GATA-binding protein 3
(GATA-3) and c-Maf are required for the chromatin remodeling and direct
activation of Th2 cytokines IL-4, IL-5, and IL-13 for Th2 cell development.
4,5
The
balance of Th1/Th2 cells is thought to be determined by the expression ratio of T-
bet/GATA-3 and is important for inducing autoimmune and allergic immune
responses.
6
However, the Th1/Th2 paradigm was recently shifted to the Th1/
Th2/Th17/regulatory T (T-reg) hypothesis, a multi-lineage commitment from the
same Th precursor cells.
7,8
regulatory T cells, referred to as regulatory T cells,
express forkhead box P3 (FoxP3) and suppress activated immune responses by
producing transforming growth factor
β
(TGF
β
)‚
9,10
whereas Th17 cells induce
Review Article
DOI 10.3349/ymj.2010.51.4.484
pISSN: 0513-5796, eISSN: 1976-2437
Yonsei Med J 51(4):484-491, 2010
Transcriptional Regulation of T Helper 17
Cell Differentiation
Eun Sook Hwang
College of Pharmacy, Division of Life and Pharmaceutical Sciences and Center for Cell Signaling &
Drug Discovery Research, Ewha Womans University, Seoul, Korea.
The third lineage of T helper subsets, Th17, has recently been identified as an IL-
17-producing CD4+ Th cell, and its functions and regulatory mechanisms have
been extensively characterized in immune responses. Functional studies have
provided evidence that Th17 cells are important for the modulation of autoim-
mune responses, such as chronic asthma, rheumatoid arthritis, inflammatory bowel
diseases, and multiple sclerosis. Murine Th17 cell differentiation is enhanced by
the coordinated functions of distinct cytokines including TGF
β
, IL-6, IL-21, and
IL-23, whereas IL-2, IL-4, IFN
γ
, and IL-27 inhibit its differentiation. In addition,
Th17 cells are controlled by several transcription factors such as ROR
γ
t, IRF4,
BATF, FoxP3, T-bet, PPAR
γ
, E-FABP, and SOCSs. This review focuses on the
functions and regulatory mechanisms of several transcription factors in the control of
Th17 cell differentiation.
Key Words: T helper 17, TGF
β
, IL-6, transcription factor
Received: March 22, 2010
Corresponding author: Dr. Eun Sook Hwang,
College of Phamacy, Division of Life and
Pharmaceutical Sciences, Center for Cell
Signaling & Drug Discovery Research,
Ewha Womans University,
11-1 Daehyeon-dong, Seodaemun-gu,
Seoul 120-750, Korea.
Tel: 82-2-3277-4369, Fax: 82-2-3277-3760
The author has no financial conflicts of
interest.
© Copyright:
Yonsei University College of Medicine 2010
This is an Open Access article distributed under the
terms of the Creative Commons Attribution Non-
Commercial License (http://creativecommons.org/
licenses/by-nc/3.0) which permits unrestricted non-
commercial use, distribution, and reproduction in any
medium, provided the original work is properly cited.
INTRODUCTION
Regulation of Th17 Differentiation
Yonsei Med J http://www.eymj.org Volume 51 Number 4 July 2010
485
retinoic acid-related orphan receptor
γ
t (ROR
γ
t) -mediated
IL-17 production and control the inflammatory autoim-
mune response.
11,12
The differentiation of Th17 and T-reg
cells requires the activation of TGF
β
-mediated signaling,
and IL-6 selectively drives Th17 cell differentiation from
TGF
β
-stimulated Th cells by promoting sequential activa-
tion of IL-21 and IL-23 signaling.
10,13
Here, we review the current understanding of the trans-
cription factors involved in the regulation of Th17 cell dif-
ferentiation, including updates of ROR
γ
t , FoxP3, and
other Th17-specific transcription factors such as interferon
regulatory factor 4 (IRF4), B-cell activating transcription
factor (BATF), peroxisome proliferator activated receptor
(PPAR
γ
), T-bet, and suppressors of cytokine signaling
(SOCS) 3 (Fig. 1).
Many scientists have reported that TGF
β
and IL-6 are
essential for Th17 cell differentiation.
12,14,15
TGF
β
, which is
produced by innate immune cells, binds to its specific recep-
tor and induces engagement of TGF
β
receptor I and II with
subsequent activation of receptor-associated SMADs. Acti-
vated SMADs interact with a variety of transcription factors,
resulting in chromatin remodeling and modulation of gene
transcription of TGF
β
target genes.
16
TGF
β
inhibits signal
Fig. 1. Cytokine signaling and transcription factors in the regulation of Th17 cell differentiation. TCR stimulation activates gene expression of general transcription
factors such as NFAT, AP-1, and NF-κB, and induces Th cell activation and proliferation. BATF is activated upon TCR stimulation and stimulates IL-17 gene
transcription. TGF
β
stimulation induces both FoxP3 and RORγt (also RORa) activation. High concentrations of TGFb increase FoxP3 through the activation of SMAD4
and subsequently induce TGFb production and simultaneously suppress RORγt activity and Th17 cell differentiation. However, the presence of cytokine IL-6 or IL-21
activates STAT3 and induces gene expression of the IL-21 and IL-23 receptor, activating positive IL-21 autocrine regulation for Th17 cell differentiation. In addition, IL-1
induces IRF4 or epidermal FABP4, which in turn induces IL-17 gene transcription. While T-bet and Ets-1 antagonize RORγt activity and thus function as suppressors of
Th17 cell development, PPARγintrinsically suppresses IL-17 gene transcription by blocking the activation-induced removal of repressor complexes from the IL-17
gene promoter. SOCS1 and SOCS reciprocally modulate Th17 cell differentiation. TCR, T cell receptor; NFAT, nuclear factor of activated T cells; AP, activator protein;
BATF, B cell-activating transcription factor; IL-17, interleukin-17; TGF
β
, transforming growth factor
β
; RORγt, retinoic acid-related orphan receptor γt; STAT, signal
transducer and activator of transcription; IRF-4, interferon-inducible factor-4; E-FABP, epidermal-fatty acid-binding protein; PPARγ, peroxisome proliferator activated
receptor γ; SOCS, suppressors of cytokine signaling.
TGF
ββ
- AND IL-6-MEDIATED
TH17 CELL DIFFERENTIATION
Eun Sook Hwang
Yonsei Med J http://www.eymj.org Volume 51 Number 4 July 2010
486
transducer and activator of transcription (STAT5)-medi-
ated IL-2 production in TCR-stimulated T cells
17
and also
interferes with Th1 and Th2 cell differentiation by inhibiting
expression of master transcription factor T-bet and GATA-
3.
18,19
In addition, TGF
β
increases FoxP3 expression and
induces generation of T-reg cells.
10
FoxP3-positive T-reg
cells potently suppress cell proliferation and differentiation
of Th cells by boosting TGF
β
production.
10
Overexpres-
sion of TGF
β
in a T cell-specific manner in mice leads to
the generation of T cells with regulatory functions and pro-
tects IL-2-deficient mice from developing severe systemic
inflammation with autoimmunity.
20,21
However, additional
production of the pro-inflammatory cytokine IL-6 along
with TGF
β
suppresses FoxP3 expression and T-reg cell
generation and simultaneously induces IL-17 production,
resulting in Th17 cell differentiation.
12
Consistent with this,
TGF
β
transgenic mice treated with myelin oligodendrocyte
glycoprotein (MOG) in complete Freund’s adjuvant (CFA)
exhibit substantially increased Th17-mediated immune
responses and aggravated experimental autoimmune encep-
halomyelitis (EAE),
12
indicating that TGF
β
plus IL-6 in-
duces the generation of Th17 cells. Despite the importance
of TGF
β
function in murine Th17 cells, TGF
β
is dispen-
sable for human Th17 cell differentiation. Instead, human
Th17 cells are induced by stimulation with IL-6, together
with another cytokine such as IL-1, IL-21, or IL-23.
15,22,23
IL-6 is a key factor for inducing human and murine Th17
cell differentiation by activating STAT3 and ROR
γ
tex-
pression. STAT3 activation is induced not only by IL-6 but
also by IL-21 and IL-23.
13,24,25
While STAT3-null cells fail
to express ROR
γ
t and produce a diminished level of IL-
17, retroviral restoration of STAT3 rescues the IL-17 de-
fect.
26
Although it is still unclear whether STAT3 modula-
tes ROR
γ
t gene transcription, activated STAT3 directly
binds to the STAT-binding sites in the IL-17 gene pro-
moter and increases IL-17 gene transcription.
27
In addition
to the functional importance of STAT3 activation in Th17
cell differentiation, ROR
γ
t has been identified as a master
regulator of Th17 cell differentiation. Analogous to STAT4-
mediated T-bet in Th1 cells and STAT6-dependent GATA-
3 in Th2 cells, Th17 cells require activation of STAT3 and
subsequent ROR
γ
t induction.
11
ROR
γ
t , a spliced isoform
of ROR
γ
, is a member of the nuclear receptor superfamily,
and is closely related to the retinoic acid receptor (RAR)
subfamily,
28
and is required for thymocyte survival and
lymphoid organogenesis.
29
Deficiency of ROR
γ
t results in
profound Th17 deficiency and protects mice from EAE
development.
11
Ectopic overexpression of STAT3 in ROR
γ
t-
deficient cells, and vice versa, fails to restore IL-17 produc-
tion.
13
This suggests that STAT3 and ROR
γ
t may each
regulate the other’s gene transcription and induce IL-17
expression parallel to some extent. More recently, ROR
α
was reported as a novel Th17-specific transcription factor.
Like ROR
γ
t , ROR
α
is induced by TGF
β
plus IL-6 in a
STAT3-dependent manner, and promotes Th17 cell dif-
ferentiation through direct activation of IL-17 gene trans-
cription.
30
Double deficiency of ROR
γ
t and ROR
α
results
in complete blockade of IL-17 production and EAE devel-
opment.
30
IRF4 was originally identified as a GATA-3 inducer in Th2
cell differentiation.
31,32
However, IRF4-null mice exhibit
impaired generation of Th17 cells in response to TGF
β
and IL-6 and increased resistance to EAE.
33
In addition,
IRF4-deficient Th cells exhibit an intrinsic defect in the
autocrine IL-21 loop and an increased population of
FoxP3-mediated T-reg cells with no effect on STAT3 acti-
vation and SOCS3 expression.
34
A more recent report
implies that IRF4 is activated upon IL-1 signaling and is
critical for early Th17 cell differentiation.
35
Despite the
importance of IRF4 in Th17 cell differentiation, the mole-
cular mechanisms are unclear. The fact that IRF4 interacts
with NFATp to induce IL-4 expression may suggest that
IRF4 modulates NFATp-dependent IL-2 expression, which
is associated with IL-17 production.
36,37
The BATF is a basic leucine zipper (bZIP) transcription
factor and dimerizes with Jun class factors of the activator
protein-1 (AP-1) family.
38-40
BATF is known to function as
a potent inhibitor of AP-1-mediated gene expression via
the phosphorylation of BATF.
40-42
In addition, expression
analysis reveals that BATF is highly expressed in hemato-
poietic cells and is increased in B and T cells by the activa-
tion of NF-
κ
B in response to viral infection or IL-6-sti-
mulation.
43-47
BATF gene transcription is substantially inc-
reased in activated Th cells subsets including Th1, Th2,
and Th17 cells.
48
Despite its wide expression in all Th1,
Th2, and Th17 cells, BATF-deficiency fails to generate IL-
17 in CD4+ and CD8+ T cells in vitro and in vivo, but
STAT3 AND ROR
γγ
t AS KEY
TRANSCRIPTION FACTORS FOR TH17
CELL DIFFERENTIATION
IRF-4 FUNCTIONS IN IL-21-INDUCED
TH17 CELL DIFFERENTIATION
BATF AS A PROMOTER FOR TH17
CELL DIFFERENTIATION
Regulation of Th17 Differentiation
Yonsei Med J http://www.eymj.org Volume 51 Number 4 July 2010
487
rather increases T-reg cell generation, thus protecting mice
from EAE development.
48
While the levels of ROR
α
and
ROR
γ
t expression are not sustained in BATF-deficient
Th17 cells compared with those in wild type (WT) cells,
enforced ROR
γ
t expression is not able to restore IL-17
production in BATF-deficient Th cells. Nevertheless,
BATF synergizes with ROR
γ
t to induce IL-17 expression
through direct interaction with the IL-17 gene promoter.
48
Many questions regarding BATF, such as whether IL-6-
induced STAT3 activation is affected by BATF deficiency
and whether BATF is required for DNA binding of ROR
γ
t
or IRF4, remain to be addressed in the future.
49
Differentiation of FoxP3-directed T-reg cells and ROR
γ
t-
driven Th17 cells has been shown to be triggered by TGF
β
signaling, but the Th17 differentiation program requires
additional IL-6 or IL-21 cytokine signaling either to switch
off FoxP3 or to switch on ROR
γ
t,
10,11,15
suggesting recipro-
cal regulation of T-reg and Th17 cells during Th cell differ-
entiation. It has been asked whether T-reg can be convert-
ed to Th17 in response to IL-6 and how FoxP3 and ROR
γ
t
modulate each other’s expression or activity.
30,50,51
Interes-
tingly, FoxP3 and IL-17 are both induced upon TGF
β
sti-
mulation.
13,52
In addition, FoxP3-positive Th cells produce
IL-17 in the presence of IL-6 through the activation of
ROR
γ
t , whereas FoxP3 antagonizes ROR
γ
t activity in a
manner dependent on SMAD4, suggesting the plasticity of
T-reg cells.
30
Others also report that FoxP3 inhibits IL-17
expression by antagonizing ROR
γ
t function in a TGF
β
concentration-dependent manner
53
or through direct
interaction with ROR
γ
t.
54
Like the suppressive function of
FoxP3 on IL-17 expression, the Th1-specific transcription
factor T-bet suppresses ROR
γ
t -mediated Th17 cell differ-
entiation.
55-57
Several functional studies indicate that T-bet
suppresses ROR
γ
t expression and Th17 cell differentiation
and further attenuates autoimmune responses.
56,58-62
None-
theless, the mechanism by which T-bet directly or indirectly
inhibits IL-17 expression and whether T-bet antagonizes
ROR
γ
t activity remain to be characterized. In addition, a
T-bet-interacting transcription factor, Ets-1 positively mo-
dulates Th1 cell differentiation but inhibits Th17 cell genera-
tion.
63,64
Ets-1-deficient Th cells exhibit preferential differen-
tiation into Th17 cells and increased IL-22 and IL-23
receptor expression.
64
Moreover, targeting of Ets-1 by mi-
croRNA miR-326 promotes Th17 differentiation.
65
Since
there is no apparent interaction between Ets-1 and IL-17
gene promoter, how Ets-1 modulates IL-17 expression
must be defined in the future.
Peroxisome proliferator-activated receptor
γ
(PPAR
γ
) is a
nuclear receptor like ROR
γ
t and ROR
α
and forms hetero-
dimers with retinoid X receptors (RXRs) to bind to the gene
promoter.
66,67
PPAR
γ
activation upon ligand binding is criti-
cal for the expression of genes such as adiponectin and
fatty acid-binding protein (FABP) (also referred as aP2) in-
volved in adipocyte differentiation and lipid metabolism
68,69
While enforced PPAR
γ
expression induces adipocyte dif-
ferentiation from fibroblasts, PPAR
γ
-deficiency attenuates
white adipose tissue development.
70
Although PPAR
γ
func-
tions as a master transcriptional regulator for adipocyte
differentiation, the anti-inflammatory activity of PPAR
γ
is
also well-characterized.
71-73
The anti-inflammatory function
of PPAR
γ
is mediated through the inhibition of both matu-
ration and function of dendritic cells and macrophages.
74,75
More precisely, the ligand-binding domain of PPAR
γ
is
sumoylated upon ligand activation and prevents the removal
of repressor complexes composed of nuclear receptor core-
pressor and histone deacetylase-3, thus resulting in sustain-
ed repressor complex-induced silencing of pro-inflamma-
tory cytokine genes.
76,77
In addition to the modulation of
macrophage function, PPAR
γ
modulates T cell activity by
inhibiting IL-2 production in T cell receptor-stimulated Th
cells
78
and by suppressing Th2 cell differentiation.
79
There-
fore, PPAR
γ
ligands including endogenous and synthetic
agonists such as linoleic acid, prostaglandin J2, and thia-
zolidinediones have been extensively studied due to the
interest in treating inflammatory diseases.
71,80,81
A recent
study demonstrates that PPAR
γ
is an intrinsic suppressor
for Th17 cell generation.
82
PPAR
γ
activation is thought to
prevents removal of repressor complexes from ROR
γ
t
gene promoter, thus suppressing ROR
γ
t expression and
ROR
γ
t -induced Th17 cell differentiation in an intrinsic
manner. Moreover, human multiple sclerosis patients are
impressively susceptible to PPAR
γ
-mediated suppression
of Th17 cell development, strongly asserting PPAR
γ
as a
promising target for specific immunointervention in auto-
immune disorders.
82
In contrast to the suppressive function of adipogenic
PPAR
γ
, epidermal FABP (E-FABP) is characterized as a
positive modulator of IL-17 production in Th cells.
83
FABP-
deficiency contributes to the protection from EAE devel-
opment,
84
which has been explained by the reduced level
of pro-inflammatory cytokines in macrophages and
dendritic cells.
85
Moreover, FABP-deficient Th cells ex-
press increased amounts of PPAR
γ
and subsequently
suppress IL-17 production; however, this can be reversed
by treatment with the PPAR
γ
antagonist, GW9662.
83
More
FOXP3, T-BET, AND ETS-1 SUPPRESS
TH17 CELL DIFFERENTIATION
PPAR
γγ
AND E-FABP MODULATE TH17
CELL DIFFERENTIATION
detailed molecular mechanisms of E-FABP have yet to be
characterized.
The SOCS inhibit STAT-mediated cytokine signaling.
86,87
Since SOCS1 suppresses both IFN
γ
- and IL-4-mediated
Th1 and Th2 cell differentiation, genetic ablation of SOCS1
results in unconditional hyperactivation of T cells.
88
In
addition, T cell-specific SOCS1-conditional knockout mice
exhibit attenuated Th17 cell generation and induced hyper-
activation of SOCS3 in Th cells,
89
suggesting that SOCS1
as a transcriptional activator for Th17 cell development.
Activated SOCS3 is known to selectively suppress STAT-3
activation induced by IL-6, granulocyte-colony stimulating
factor (GCSF), and leptin,
90,91
whereas deficiency of
SOCS3 increases TGF
β
production and simultaneously
enhances Th17 cell development.
27,92
It is also reported that
TGF
β
inhibits SOCS3 gene transcription and prolongs
STAT3 activation during Th17 cell differentiation.
93
With the recent reports of the functions of IL-17-producing
Th17 cells in autoimmune responses and the regulatory
mechanisms for Th17 cell differentiation, a new paradigm
of Th cell differentiation has been established. The Th1/
Th2 paradigm is mainly shifted to a Th1/Th2/ Th17/T-reg
program. This review describes the function and potential
mechanisms of transcription factors critical for the regul-
ation of Th17 cell differentiation and also includes inter-
connection among transcription factors such as Th1-
specific T-bet, T-reg-limited FoxP3, and Th17-specific
ROR
γ
t . These transcription factors have prevalent roles in
determining lineage commitment through interaction with
cytokine gene promoters and/or other lineage-specific
transcription factors. Targeting these transcription factors,
as well as signature cytokines, may be beneficial for
controlling several autoimmune diseases, although re-
search is currently underway to identify the detailed me-
chanisms.
This work was supported by the R&D program (A080908)
of KHIDI and partly by the NCRC program (R15-2006-
020) funded by MEST.
1. Mosmann TR, Cherwinski H, Bond MW, Giedlin MA, Coffman
RL. Two types of murine helper T cell clone. I. Definition accord-
ing to profiles of lymphokine activities and secreted proteins. J
Immunol 1986;136:2348-57.
2. Murphy KM, Reiner SL. The lineage decisions of helper T cells.
Nat Rev Immunol 2002;2:933-44.
3. Szabo SJ, Kim ST, Costa GL, Zhang X, Fathman CG, Glimcher
LH. A novel transcription factor, T-bet, directs Th1 lineage com-
mitment. Cell 2000;100:655-69.
4. Ouyang W, Ranganath SH, Weindel K, Bhattacharya D, Murphy
TL, Sha WC, et al. Inhibition of Th1 development mediated by
GATA-3 through an IL-4-independent mechanism. Immunity
1998;9:745-55.
5. Ho IC, Lo D, Glimcher LH. c-maf promotes T helper cell type 2
(Th2) and attenuates Th1 differentiation by both interleukin 4-
dependent and -independent mechanisms. J Exp Med 1998;
188:1859-66.
6. Kiwamoto T, Ishii Y, Morishima Y, Yoh K, Maeda A, Ishizaki
K, et al. Transcription factors T-bet and GATA-3 regulate devel-
opment of airway remodeling. Am J Respir Crit Med 2006;174:
142-51.
7. Weaver CT, Hatton RD, Mangan PR, Harrington LE. IL-17
family cytokines and the expanding diversity of effector T cell
lineages. Annu Rev Immunol 2007;25:821-52.
8. Bettelli E, Korn T, Kuchroo VK. Th17: the third member of the
effector T cell trilogy. Curr Opin Immunol 2007;19:652-7.
9. Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell
development by the transcription factor Foxp3. Science 2003;
299:1057-61.
10. Chen W, Jin W, Hardegen N, Lei KJ, Li L, Marinos N, et al.
Conversion of peripheral CD4+CD25- naive T cells to CD4+
CD25+ regulatory T cells by TGF-beta induction of transcription
factor Foxp3. J Exp Med 2003;198:1875-86.
11. Ivanov II, McKenzie BS, Zhou L, Tadokoro CE, Lepelley A,
Lafaille JJ, et al. The orphan nuclear receptor RORgammat directs
the differentiation program of proinflammatory IL-17+ T helper
cells. Cell 2006;126:1121-33.
12. Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, Oukka M, et al.
Reciprocal developmental pathways for the generation of patho-
genic effector TH17 and regulatory T cells. Nature 2006;441:
235-8.
13. Zhou L, Ivanov II, Spolski R, Min R, Shenderov K, Egawa T, et
al. IL-6 programs T(H)-17 cell differentiation by promoting
sequential engagement of the IL-21 and IL-23 pathways. Nat
Immunol 2007;8:967-74.
14. Mangan PR, Harrington LE, O’Quinn DB, Helms WS, Bullard
DC, Elson CO, et al. Transforming growth factor-beta induces
development of the T(H)17 lineage. Nature 2006;441:231-4.
15. Veldhoen M, Hocking RJ, Atkins CJ, Locksley RM, Stockinger
B. TGFbeta in the context of an inflammatory cytokine milieu
supports de novo differentiation of IL-17-producing T cells.
Immunity 2006;24:179-89.
16. Yang X, Letterio JJ, Lechleider RJ, Chen L, Hayman R, Gu H, et
al. Targeted disruption of SMAD3 results in impaired mucosal
immunity and diminished T cell responsiveness to TGF-beta.
EMBO J 1999;18:1280-91.
17. Laurence A, Tato CM, Davidson TS, Kanno Y, Chen Z, Yao Z,
Eun Sook Hwang
Yonsei Med J http://www.eymj.org Volume 51 Number 4 July 2010
488
REFERENCES
CONCLUSION
ACKNOWLEDGEMENTS
OTHER TRANSCRIPTIONAL
MODULATORS FOR TH17 CELL
DIFFERENTIATION
et al. Interleukin-2 signaling via STAT5 constrains T helper 17
cell generation. Immunity 2007;26:371-81.
18. Gorelik L, Constant S, Flavell RA. Mechanism of transforming
growth factor beta-induced inhibition of T helper type 1 differen-
tiation. J Exp Med 2002;195:1499-505.
19. Gorelik L, Fields PE, Flavell RA. Cutting edge: TGF-beta inhi-
bits Th type 2 development through inhibition of GATA-3
expression. J Immunol 2000;165:4773-7.
20. Carrier Y, Yuan J, Kuchroo VK, Weiner HL. Th3 cells in perip-
heral tolerance. I. Induction of Foxp3-positive regulatory T cells
by Th3 cells derived from TGF-beta T cell-transgenic mice. J
Immunol 2007;178:179-85.
21. Carrier Y, Yuan J, Kuchroo VK, Weiner HL. Th3 cells in peri-
pheral tolerance. II. TGF-beta-transgenic Th3 cells rescue IL-2-
deficient mice from autoimmunity. J Immunol 2007;178:172-8.
22. Wilson NJ, Boniface K, Chan JR, McKenzie BS, Blumenschein
WM, Mattson JD, et al. Development, cytokine profile and func-
tion of human interleukin 17-producing helper T cells. Nat Im-
munol 2007;8:950-7.
23. Acosta-Rodriguez EV, Napolitani G, Lanzavecchia A, Sallusto F.
Interleukins 1beta and 6 but not transforming growth factor-beta
are essential for the differentiation of interleukin 17-producing
human T helper cells. Nat Immunol 2007;8:942-9.
24. Nurieva R, Yang XO, Martinez G, Zhang Y, Panopoulos AD,
Ma L, et al. Essential autocrine regulation by IL-21 in the genera-
tion of inflammatory T cells. Nature 2007;448:480-3.
25. Yang XO, Panopoulos AD, Nurieva R, Chang SH, Wang D,
Watowich SS, et al. STAT3 regulates cytokine-mediated gene-
ration of inflammatory helper T cells. J Biol Chem 2007;282:
9358-63.
26. Mathur AN, Chang HC, Zisoulis DG, Stritesky GL, Yu Q,
O’Malley JT, et al. Stat3 and Stat4 direct development of IL-17-
secreting Th cells. J Immunol 2007;178:4901-7.
27. Chen Z, Laurence A, Kanno Y, Pacher-Zavisin M, Zhu BM, Tato
C, et al. Selective regulatory function of Socs3 in the formation of
IL-17-secreting T cells. Proc Natl Acad Sci U S A 2006;103:
8137-42.
28. Eberl G, Littman DR. The role of the nuclear hormone receptor
RORgammat in the development of lymph nodes and Peyer’s
patches. Immunol Rev 2003;195:81-90.
29. Sun Z, Unutmaz D, Zou YR, Sunshine MJ, Pierani A, Brenner-
Morton S, et al. Requirement for RORgamma in thymocyte survi-
val and lymphoid organ development. Science 2000;288:2369-73.
30. Yang XO, Pappu BP, Nurieva R, Akimzhanov A, Kang HS,
Chung Y, et al. T helper 17 lineage differentiation is programmed
by orphan nuclear receptors ROR alpha and ROR gamma.
Immunity 2008;28:29-39.
31. Rengarajan J, Mowen KA, McBride KD, Smith ED, Singh H,
Glimcher LH. Interferon regulatory factor 4 (IRF4) interacts with
NFATc2 to modulate interleukin 4 gene expression. J Exp Med
2002;195:1003-12.
32. Lohoff M, Mittrücker HW, Prechtl S, Bischof S, Sommer F,
Kock S, et al. Dysregulated T helper cell differentiation in the
absence of interferon regulatory factor 4. Proc Natl Acad Sci U S A
2002;99:11808-12.
33. Brüstle A, Heink S, Huber M, Rosenplänter C, Stadelmann C, Yu
P, et al. The development of inflammatory T(H)-17 cells requires
interferon-regulatory factor 4. Nat Immunol 2007;8:958-66.
34. Huber M, Brüstle A, Reinhard K, Guralnik A, Walter G, Mahiny
A, et al. IRF4 is essential for IL-21-mediated induction, ampli-
fication, and stabilization of the Th17 phenotype. Proc Natl Acad
Sci U S A 2008;105:20846-51.
35. Chung Y, Chang SH, Martinez GJ, Yang XO, Nurieva R, Kang
HS, et al. Critical regulation of early Th17 cell differentiation by
interleukin-1 signaling. Immunity 2009;30:576-87.
36. Littman DR, Sun Z, Unutmaz D, Sunshine MJ, Petrie HT, Zou
YR. Role of the nuclear hormone receptor ROR gamma in trans-
criptional regulation, thymocyte survival, and lymphoid organo-
genesis. Cold Spring Harb Symp Quant Biol 1999;64:373-81.
37. Chen Z, Laurence A, O’Shea JJ. Signal transduction pathways
and transcriptional regulation in the control of Th17 differen-
tiation. Semin Immunol 2007;19:400-8.
38. Chinenov Y, Kerppola TK. Close encounters of many kinds: Fos-
Jun interactions that mediate transcription regulatory specificity.
Oncogene 2001;20:2438-52.
39. Dorsey MJ, Tae HJ, Sollenberger KG, Mascarenhas NT, Johan-
sen LM, Taparowsky EJ. B-ATF: a novel human bZIP protein
that associates with members of the AP-1 transcription factor
family. Oncogene 1995;11:2255-65.
40. Echlin DR, Tae HJ, Mitin N, Taparowsky EJ. B-ATF functions
as a negative regulator of AP-1 mediated transcription and blocks
cellular transformation by Ras and Fos. Oncogene 2000;19:1752-
63.
41. Williams KL, Nanda I, Lyons GE, Kuo CT, Schmid M, Leiden
JM, et al. Characterization of murine BATF: a negative regulator
of activator protein-1 activity in the thymus. Eur J Immunol 2001;
31:1620-7.
42. Deppmann CD, Thornton TM, Utama FE, Taparowsky EJ. Phos-
phorylation of BATF regulates DNA binding: a novel mechanism
for AP-1 (activator protein-1) regulation. Biochem J 2003;374:
423-31.
43. Li J, Peet GW, Balzarano D, Li X, Massa P, Barton RW, et al.
Novel NEMO/IkappaB kinase and NF-kappa B target genes at
the pre-B to immature B cell transition. J Biol Chem 2001;276:
18579-90.
44. Hasegawa T, Zhou X, Garrett LA, Ruteshouser EC, Maity SN, de
Crombrugghe B. Evidence for three major transcription activa-
tion elements in the proximal mouse proalpha2(I) collagen pro-
moter. Nucleic Acids Res 1996;24:3253-60.
45. Meyer NP, Johansen LM, Tae HJ, Budde PP, Williams KL, Tapa-
rowsky EJ. Genomic organization of human B-ATF, a target for
regulation by EBV and HTLV-1. Mamm Genome 1998;9:849-52.
46. Johansen LM, Deppmann CD, Erickson KD, Coffin WF, 3rd,
Thornton TM, Humphrey SE, et al. EBNA2 and activated Notch
induce expression of BATF. J Virol 2003;77:6029-40.
47. Senga T, Iwamoto T, Humphrey SE, Yokota T, Taparowsky EJ,
Hamaguchi M. Stat3-dependent induction of BATF in M1 mouse
myeloid leukemia cells. Oncogene 2002;21:8186-91.
48. Schraml BU, Hildner K, Ise W, Lee WL, Smith WA, Solomon B,
et al. The AP-1 transcription factor Batf controls T(H)17 differen-
tiation. Nature 2009;460:405-9.
49. Martinez GJ, Dong C. BATF: bringing (in) another Th17-regulat-
ing factor. J Mol Cell Biol 2009;1:66-8.
50. Osorio F, LeibundGut-Landmann S, Lochner M, Lahl K,
Sparwasser T, Eberl G, et al. DC activated via dectin-1 convert
Treg into IL-17 producers. Eur J Immunol 2008;38:3274-81.
51. Annunziato F, Cosmi L, Liotta F, Maggi E, Romagnani S. The
phenotype of human Th17 cells and their precursors, the cyto-
kines that mediate their differentiation and the role of Th17 cells
in inflammation. Int Immunol 2008;20:1361-8.
Regulation of Th17 Differentiation
Yonsei Med J http://www.eymj.org Volume 51 Number 4 July 2010
489
52. Yang XO, Nurieva R, Martinez GJ, Kang HS, Chung Y, Pappu
BP, et al. Molecular antagonism and plasticity of regulatory and
inflammatory T cell programs. Immunity 2008;29:44-56.
53. Zhou L, Lopes JE, Chong MM, Ivanov II, Min R, Victora GD, et
al. TGF-beta-induced Foxp3 inhibits T(H)17 cell differentiation
by antagonizing RORgammat function. Nature 2008;453:236-40.
54. Ichiyama K, Yoshida H, Wakabayashi Y, Chinen T, Saeki K, Na-
kaya M, et al. Foxp3 inhibits RORgammat-mediated IL-17A
mRNA transcription through direct interaction with RORgam-
mat. J Biol Chem 2008;283:17003-8.
55. Mathur AN, Chang HC, Zisoulis DG, Kapur R, Belladonna ML,
Kansas GS, et al. T-bet is a critical determinant in the instability
of the IL-17-secreting T-helper phenotype. Blood 2006;108:
1595-601.
56. Fujiwara M, Hirose K, Kagami S, Takatori H, Wakashin H,
Tamachi T, et al. T-bet inhibits both TH2 cell-mediated eosino-
phil recruitment and TH17 cell-mediated neutrophil recruitment
into the airways. J Allergy Clin Immunol 2007;119:662-70.
57. Gocke AR, Cravens PD, Ben LH, Hussain RZ, Northrop SC,
Racke MK, et al. T-bet regulates the fate of Th1 and Th17 lympho-
cytes in autoimmunity. J Immunol 2007;178:1341-8.
58. Durrant DM, Gaffen SL, Riesenfeld EP, Irvin CG, Metzger DW.
Development of allergen-induced airway inflammation in the
absence of T-bet regulation is dependent on IL-17. J Immunol
2009;183:5293-300.
59. Rutitzky LI, Smith PM, Stadecker MJ. T-bet protects against
exacerbation of schistosome egg-induced immunopathology by
regulating Th17-mediated inflammation. Eur J Immunol 2009;
39:2470-81.
60. Abromson-Leeman S, Bronson RT, Dorf ME. Encephalitogenic
T cells that stably express both T-bet and ROR gamma t consis-
tently produce IFNgamma but have a spectrum of IL-17 profiles.
J Neuroimmunol 2009;215:10-24.
61. Guo S, Cobb D, Smeltz RB. T-bet inhibits the in vivo differen-
tiation of parasite-specific CD4+ Th17 cells in a T cell-intrinsic
manner. J Immunol 2009;182:6179-86.
62. Rangachari M, Mauermann N, Marty RR, Dirnhofer S, Kurrer
MO, Komnenovic V, et al. T-bet negatively regulates autoim-
mune myocarditis by suppressing local production of interleukin
17. J Exp Med 2006;203:2009-19.
63. Grenningloh R, Kang BY, Ho IC. Ets-1, a functional cofactor of
T-bet, is essential for Th1 inflammatory responses. J Exp Med
2005;201:615-26.
64. Moisan J, Grenningloh R, Bettelli E, Oukka M, Ho IC. Ets-1 is a
negative regulator of Th17 differentiation. J Exp Med 2007;204:
2825-35.
65. Du C, Liu C, Kang J, Zhao G, Ye Z, Huang S, et al. MicroRNA
miR-326 regulates TH-17 differentiation and is associated with
the pathogenesis of multiple sclerosis. Nat Immunol 2009;10:
1252-9.
66. Gampe RT Jr, Montana VG, Lambert MH, Miller AB, Bledsoe
RK, Milburn MV, et al. Asymmetry in the PPARgamma/RX-
Ralpha crystal structure reveals the molecular basis of heterodi-
merization among nuclear receptors. Mol Cell 2000;5:545-55.
67. Tontonoz P, Graves RA, Budavari AI, Erdjument-Bromage H,
Lui M, Hu E, et al. Adipocyte-specific transcription factor ARF6
is a heterodimeric complex of two nuclear hormone receptors,
PPAR gamma and RXR alpha. Nucleic Acids Res 1994;22:5628-
34.
68. Jones JR, Barrick C, Kim KA, Lindner J, Blondeau B, Fujimoto
Y, et al. Deletion of PPARgamma in adipose tissues of mice pro-
tects against high fat diet-induced obesity and insulin resistance.
Proc Natl Acad Sci U S A 2005;102:6207-12.
69. Chui PC, Guan HP, Lehrke M, Lazar MA. PPARgamma regul-
ates adipocyte cholesterol metabolism via oxidized LDL receptor
1. J Clin Invest 2005;115:2244-56.
70. Rosen ED, Sarraf P, Troy AE, Bradwin G, Moore K, Milstone
DS, et al. PPAR gamma is required for the differentiation of adi-
pose tissue in vivo and in vitro. Mol Cell 1999;4:611-7.
71. Na HK, Surh YJ. Peroxisome proliferator-activated receptor gam-
ma (PPARgamma) ligands as bifunctional regulators of cell
proliferation. Biochem Pharmacol 2003;66:1381-91.
72. Chen CW, Chang YH, Tsi CJ, Lin WW. Inhibition of IFN-gam-
ma-mediated inducible nitric oxide synthase induction by the per-
oxisome proliferator-activated receptor gamma agonist, 15-deoxy-
delta 12,14-prostaglandin J2, involves inhibition of the upstream
Janus kinase/STAT1 signaling pathway. J Immunol 2003;171:
979-88.
73. Schmidt S, Moric E, Schmidt M, Sastre M, Feinstein DL, Heneka
MT. Anti-inflammatory and antiproliferative actions of PPAR-
gamma agonists on T lymphocytes derived from MS patients. J
Leukoc Biol 2004;75:478-85.
74. Klotz L, Dani I, Edenhofer F, Nolden L, Evert B, Paul B, et al.
Peroxisome proliferator-activated receptor gamma control of den-
dritic cell function contributes to development of CD4+ T cell
anergy. J Immunol 2007;178:2122-31.
75. Szatmari I, Töröcsik D, Agostini M, Nagy T, Gurnell M, Barta E,
et al. PPARgamma regulates the function of human dendritic
cells primarily by altering lipid metabolism. Blood 2007;110:
3271-80.
76. Pascual G, Fong AL, Ogawa S, Gamliel A, Li AC, Perissi V, et
al. A SUMOylation-dependent pathway mediates transrepression
of inflammatory response genes by PPAR-gamma. Nature 2005;
437:759-63.
77. Straus DS, Glass CK. Anti-inflammatory actions of PPAR ligands:
new insights on cellular and molecular mechanisms. Trends
Immunol 2007;28:551-8.
78. Clark RB, Bishop-Bailey D, Estrada-Hernandez T, Hla T, Pud-
dington L, Padula SJ. The nuclear receptor PPAR gamma and im-
munoregulation: PPAR gamma mediates inhibition of helper T
cell responses. J Immunol 2000;164:1364-71.
79. Won HY, Min HJ, Ahn JH, Yoo SE, Bae MA, Hong JH, et al.
Anti-allergic function and regulatory mechanisms of KR62980 in
allergen-induced airway inflammation. Biochem Pharmacol
2010;79:888-96.
80. Meyers CD, Kashyap ML. Pharmacologic elevation of high-den-
sity lipoproteins: recent insights on mechanism of action and
atherosclerosis protection. Curr Opin Cardiol 2004;19:366-73.
81. Xu J, Barger SW, Drew PD. The PPAR-gamma Agonist 15-
Deoxy-Delta-Prostaglandin J(2) Attenuates Microglial Produc-
tion of IL-12 Family Cytokines: Potential Relevance to Alzhei-
mer’s Disease. PPAR Res 2008;2008:349185.
82. Klotz L, Burgdorf S, Dani I, Saijo K, Flossdorf J, Hucke S, et al.
The nuclear receptor PPAR gamma selectively inhibits Th17 dif-
ferentiation in a T cell-intrinsic fashion and suppresses CNS
autoimmunity. J Exp Med 2009;206:2079-89.
83. Li B, Reynolds JM, Stout RD, Bernlohr DA, Suttles J. Regulation
of Th17 differentiation by epidermal fatty acid-binding protein. J
Immunol 2009;182:7625-33.
84. Reynolds JM, Liu Q, Brittingham KC, Liu Y, Gruenthal M,
Eun Sook Hwang
Yonsei Med J http://www.eymj.org Volume 51 Number 4 July 2010
490
Regulation of Th17 Differentiation
Yonsei Med J http://www.eymj.org Volume 51 Number 4 July 2010
491
Gorgun CZ, et al. Deficiency of fatty acid-binding proteins in
mice confers protection from development of experimental autoim-
mune encephalomyelitis. J Immunol 2007;179:313-21.
85. Rolph MS, Young TR, Shum BO, Gorgun CZ, Schmitz-Peiffer
C, Ramshaw IA, et al. Regulation of dendritic cell function and T
cell priming by the fatty acid-binding protein AP2. J Immunol
2006;177:7794-801.
86. Kubo M, Hanada T, Yoshimura A. Suppressors of cytokine signal-
ing and immunity. Nat Immunol 2003;4:1169-76.
87. Yoshimura A, Naka T, Kubo M. SOCS proteins, cytokine signal-
ling and immune regulation. Nat Rev Immunol 2007;7:454-65.
88. Chinen T, Kobayashi T, Ogata H, Takaesu G, Takaki H, Hashi-
moto M, et al. Suppressor of cytokine signaling-1 regulates inflam-
matory bowel disease in which both IFNgamma and IL-4 are
involved. Gastroenterology 2006;130:373-88.
89. Tanaka K, Ichiyama K, Hashimoto M, Yoshida H, Takimoto T,
Takaesu G, et al. Loss of suppressor of cytokine signaling 1 in
helper T cells leads to defective Th17 differentiation by enhanc-
ing antagonistic effects of IFN-gamma on STAT3 and Smads. J
Immunol 2008;180:3746-56.
90. Mori H, Hanada R, Hanada T, Aki D, Mashima R, Nishinaka-
mura H, et al. Socs3 deficiency in the brain elevates leptin sensiti-
vity and confers resistance to diet-induced obesity. Nat Med
2004;10:739-43.
91. Yasukawa H, Ohishi M, Mori H, Murakami M, Chinen T, Aki D,
et al. IL-6 induces an anti-inflammatory response in the absence
of SOCS3 in macrophages. Nat Immunol 2003;4:551-6.
92. Kinjyo I, Inoue H, Hamano S, Fukuyama S, Yoshimura T, Koga
K, et al. Loss of SOCS3 in T helper cells resulted in reduced im-
mune responses and hyperproduction of interleukin 10 and trans-
forming growth factor-beta 1. J Exp Med 2006;203:1021-31.
93. Qin H, Wang L, Feng T, Elson CO, Niyongere SA, Lee SJ, et al.
TGF-beta promotes Th17 cell development through inhibition of
SOCS3. J Immunol 2009;183:97-105.