Download as pdf or txt
Download as pdf or txt
You are on page 1of 41

IY29CH04-Ouyang ARI 4 February 2011 15:27

ANNUAL
REVIEWS Further Regulation and Functions of
Click here for quick links to
Annual Reviews content online,
including:
the IL-10 Family of Cytokines
• Other articles in this volume
• Top cited articles in Inflammation and Disease
• Top downloaded articles
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

• Our comprehensive search


Wenjun Ouyang,1 Sascha Rutz,1 Natasha K. Crellin,1
Patricia A. Valdez,3 and Sarah G. Hymowitz2
by University of Guelph on 05/31/12. For personal use only.

1
Department of Immunology, 2 Department of Structural Biology, Genentech, Inc.,
South San Francisco, California 94080; email: ouyang@gene.com
3
Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious
Diseases, National Institutes of Health, Bethesda, Maryland 20892

Annu. Rev. Immunol. 2011. 29:71–109 Keywords


First published online as a Review in Advance on psoriasis, IBD, infectious diseases, autoimmune diseases, Th17
December 13, 2010
The Annual Review of Immunology is online at Abstract
immunol.annualreviews.org
The IL-10 family of cytokines consists of nine members: IL-10,
This article’s doi:
10.1146/annurev-immunol-031210-101312
IL-19, IL-20, IL-22, IL-24, IL-26, and the more distantly related
IL-28A, IL-28B, and IL-29. Evolutionarily, IL-10 family cytokines
Copyright  c 2011 by Annual Reviews.
All rights reserved
emerged before the adaptive immune response. These cytokines elicit
diverse host defense mechanisms, especially from epithelial cells, during
0732-0582/11/0423-0071$20.00
various infections. IL-10 family cytokines are essential for maintaining
the integrity and homeostasis of tissue epithelial layers. Members of this
family can promote innate immune responses from tissue epithelia to
limit the damage caused by viral and bacterial infections. These cy-
tokines can also facilitate the tissue-healing process in injuries caused
by infection or inflammation. Finally, IL-10 itself can repress proin-
flammatory responses and limit unnecessary tissue disruptions caused
by inflammation. Thus, IL-10 family cytokines have indispensable func-
tions in many infectious and inflammatory diseases.

71
IY29CH04-Ouyang ARI 4 February 2011 15:27

INTRODUCTION

Antibacterial responses,
The interleukin (IL)-10 family of cytokines and

Key functions

tissue remodeling,
Immune repression

Antiviral responses
the closely related interferon (IFN) family of

wound healing
cytokines form the larger Class II cytokine fam-
ily (1, 2). Class II family cytokines were origi-
nally defined by virtue of their interactions with
Class II cytokine receptors. Overall, Class I and
Class II cytokine receptors share a similar ex-

Cellular targets
tracellular architecture consisting of tandem re-

Epithelial cells,
Epithelial cells
peats of Ig-like domains. Class II receptors can

leukocytes?
Leukocytes
be differentiated from Class I receptors on the
basis of sequence analysis. For instance, Class
II cytokine receptors lack the canonical Trp-
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

Ser-X-Trp-Ser motif found in the extracellular

T cells, NK cells, NKT cells,


domain of the Class I receptors and differ in the

Myeloid cells, epithelial cells


Myeloid cells, epithelial cells
Myeloid cells, epithelial cells
Major cellular sources

Leukocytes, epithelial cells


number and spacing of their cysteine residues.
by University of Guelph on 05/31/12. For personal use only.

The cognate ligands for the Class II receptors


are thus referred to as the Class II cytokines.
IL-10 family cytokines can be further

Leukocytes
subgrouped in different ways based on their

Th2 cells

LTi cells
structure, genomic location, receptor usage,
and common downstream biological functions
(Table 1). In this review, for the convenience
of discussion, we categorize IL-10 family
β chain
IL-10R2
IL-20R2

IL-10R2
cytokines into three subgroups based primarily
on their biological functions. The first group
contains only IL-10 itself. IL-10 targets vari-
Receptor

ous leukocytes and mainly represses excessive


IL-20R1/IL-22R

inflammatory responses. The second group,


α chain

namely IL-20 subfamily cytokines, is composed


IL-10R1
IL-20R1

of IL-19, IL-20, IL-22, IL-24, and IL-26 (3). IL-20R1


IL-22R

This group of cytokines primarily acts on IL-28R


various epithelial cells and protects these
cells from invasion by extracellular pathogens
Structure

Monomer
Monomer
Monomer

Monomer

Monomer
Monomer
Monomer
such as bacteria and yeast. In addition, IL-20
Dimer?
Dimer

subfamily cytokines enhance tissue remodeling


and wound-healing activities, which help to
maintain tissue integrity and restore home-
Table 1 IL-10 family cytokines

location (human)

ostasis of epithelial layers during infection and


Chromosome

inflammatory responses. Finally, the last group


is the type III IFN group (or IFN λs), which
contains the closely related cytokines IL-28A,
12q16

19q13
1q32

IL-28B, and IL-29 (4). These cytokines induce


antiviral responses similar to those of type I
IFNs but primarily act on epithelial cells (5).
Moreover, these cytokines can synergize with
IL-28A
IL-28B
Name
IL-10
IL-19
IL-20
IL-24

IL-22

IL-26

IL-29

type I IFNs to boost host antiviral response.

72 Ouyang et al.
IY29CH04-Ouyang ARI 4 February 2011 15:27

One of the central functional themes of IL- All three genes also share similar genomic
10 family cytokines is their role in tissue protec- structures with Il10, composed of five exons
tion. On the one hand, these cytokines prevent and four introns. Il24, initially referred to as
excessive tissue damage caused by bacterial and melanoma differentiation-associated cDNA-7
viral infections as well as proinflammatory re- (MDA-7), was the first Il10 homologous gene
sponses. On the other hand, uncontrolled tissue discovered in 1995 (15). The human genome
repair processes, such as the wound-healing re- project dramatically facilitated the discovery of
sponses triggered by IL-20 subfamily cytokines, novel cytokines. Il19 and Il20 were identified
can result in diseases, such as psoriasis. This re- through the mining of the EST database for
view integrates recent data on the function and Il10 homolog genes (16, 17). IL-19, IL-20, and
regulation of IL-10 family cytokines during in- IL-24 bind to a common two-chain receptor
fectious and inflammatory diseases. complex that consists of IL-20R1 and IL-20R2
chains (17–20). In addition, IL-20R2 can also
pair with IL-22R to form a heterodimeric
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

IL-10 FAMILY CYTOKINES receptor complex that is preferentially used by


AND THEIR RECEPTORS IL-20 and IL-24, but not by IL-19 (18–20).
IL-10, which was identified by Mosmann and The genes for IL-22 and IL-26 are not
by University of Guelph on 05/31/12. For personal use only.

colleagues (6) in 1989, is the founding mem- colocalized with Il10. Instead, they are located
ber of this family of cytokines. Initially, the near the Ifng gene on human chromosome
IL-10 protein was described as a secreted cy- 12q16. Il22 was originally cloned from a
tokine synthesis inhibitory factor (CSIF) from mouse T cell line treated with IL-9 and
Th2 clones because it inhibits the production named IL-10-related T cell–derived inducible
of several cytokines, such as IFN-γ, from Th1 factor (IL-TIF) (21). In contrast, Il26 was
cells (6). The human and mouse CSIF cDNA initially identified as AK155 by subtractive
was subsequently cloned and renamed IL-10 (7, hybridization from human T lymphocytes
8). Mature human and mouse IL-10 has 160 infected with Herpesvirus saimiri (HVS) (22).
amino acids and forms noncovalently linked ho- IL-22 and IL-26 share the common β chain
modimers. IL-10 binds to two receptor chains, receptor, IL-10R2, with IL-10. However,
IL-10R1 and IL-10R2. The IL-10R1 receptors each of the cytokines binds to its unique α
in mouse and human were identified by expres- chain receptor, namely IL-22R (also called
sion cloning in 1993 based on their affinity for IL-22RA1) and IL-20R1, for IL-22 and IL-26,
Flag-tagged-rIL-10 (9, 10). IL-10R1 is struc- respectively (23–25). In addition, a secreted
turally similar to IFNR. When human IL-10R1 IL-22R homolog, IL-22BP or IL-22RA2, has
was overexpressed in hamster cells (COS-1), it been identified. IL-22BP shows 33% amino
failed to establish the responsiveness of these acid sequence identity with the extracellular
cells to human IL-10, suggesting that another domain of IL-22R. IL-22BP binds to IL-22
receptor chain was required for the function of with high affinity and neutralizes its activity in
IL-10 (11). Expression of both IL-10R1 and vitro (26–28). In certain mouse strains, such as
CRF2-4 (12), a previously cloned Class II cy- C57Bl/6, FVB, and 129, a duplication of the
tokine receptor member, restored the signal- Il22 locus has occurred (29). The second copy
ing of IL-10 (11). CRF2-4 was therefore re- of Il22, namely Il22b, is highly homologous to
named IL-10R2. The functional importance of Il22 at the DNA level. However, currently no
IL-10R2 was further confirmed by the obser- evidence supports that this copy of Il22b is tran-
vation that macrophages and splenocytes from scribed or has a function in vivo. Interestingly,
IL-10R2-deficient mice were unresponsive to the Il26 gene locus is deleted in mice, although
stimulation by IL-10 (13). it is maintained through evolution in many
Il19, Il20, and Il24 are closely linked other animals, such as dog, frog, and zebrafish
to Il10 on human chromosome 1q32 (14). (30).

www.annualreviews.org • IL-10 Family Cytokines in Immunity 73


IY29CH04-Ouyang ARI 4 February 2011 15:27

Il28A, Il28B, and Il29 (also called IFN-λ2,


IFN-λ3, and IFN-λ1, respectively) were iden-
a b tified through genomic mining for novel genes
D similar to the genes for IFN and the IL-10
D E B’ family of cytokines (31, 32). These three genes
A A F C
F were later classified as type III IFNs (or the
B
F’ C C’ IFN λ family) because of their functional sim-
B A’ E
D’ ilarities to type I IFNs (4). As is the case with
E’
type I IFNs, IL-28A, IL-28B, and IL-29 are all
readily induced from human peripheral blood
c mononuclear cells (PBMCs) upon stimulation
Proposed IL-10R2 with poly(I:C) or encephalomyocarditis virus
binding site
(EMCV). Similar mechanisms for the induc-
tion of type I IFNs, such as RIG-I, IPS-1,
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

D2
E B’ TBK1, and IFN regulatory factor 3 (IRF3), are
D
F D1
also used by type III IFNs during viral infec-
A C’
tions (33). Moreover, IL-28 and IL-29 also in-
by University of Guelph on 05/31/12. For personal use only.

F’ C
D1 B A’ duce similar downstream biological effects as
D’
D2 type I IFNs (31). Structurally, however, type
E’
III IFNs are more closely related to IL-10
Proposed IL-10R2 family cytokines, especially IL-22 (34). Indeed,
binding site
IL-28A, IL-28B, and IL-29 also signal through
d the same IL-10R2 chain, paired with their
Proposed IL-10R2
binding site unique IL-28R as the α chain (31, 32). Inter-
estingly, type III IFNs seem to preferentially
target epithelial cells (35, 36). It has been pro-
D2 D
posed, therefore, that type III IFNs and IL-20
A F C subfamily members have parallel functions in
B the protection of epithelial tissue against viral
E
and bacterial infections, respectively (34).
D1

STRUCTURE AND
Figure 1 DOWNSTREAM
Structure of IL-10 family cytokines and receptor complexes. (a) Crystal SIGNALING PATHWAYS
structure of IL-10 from the IL-10/IL-10R1 complex (pdb 1Y6K). IL-10 Because of their biological importance, the
protomers are depicted with helices rendered as cylinders. Helices are labeled.
(b) Crystal structure of monomeric IL-22 from the IL-22/IL-22R1 complex
structure and function of the IL-10 family of
(pdb code 3DGC). Helices are shown as in (a) and colored by gradient from N cytokines has been extensively studied (1, 37,
terminus (dark blue) to C terminus (cyan). In the monomeric cytokines, the loop 38). Rather than reiterate the detailed charac-
connecting the D and E helices is folded so that the E and F helices complete terization of previous reviews, the present work
the monomer core. (c) Complex of IL-10R1 bound to IL-10 (pdb code 1YK6). briefly summarizes the key structural features of
IL-10 is colored as in (a). IL-10R1 ( yellow, green) is depicted with beta strands
rendered as arrows. The N-terminal D1 and C-terminal D2 domains are
these ligands (Figure 1). All members of the
labeled. Based on the work of Yoon et al. (52), the investigators propose that IL-10 family of Class II cytokines possess a
IL-10R2 binds adjacent to IL-10R1 and interacts with the AD face of the characteristic α-helical fold consisting of six he-
cytokine. (d ) Complex of IL-22R1 bound to IL-22. IL-22 is colored as in (a), lices labeled A to F and connecting loops. The
IL-22R1 is colored and labeled analogously to IL-10R1 in (c). The proposed structural core of the monomeric ligands is a
IL-10R2-binding site is labeled.
compact four-helix bundle composed of helices
A, C, D, and F. Helix B tends to be much shorter

74 Ouyang et al.
IY29CH04-Ouyang ARI 4 February 2011 15:27

than the other helices and packs against the N- (49) (Figure 1c,d ). These structures showed
terminal portion of helix C. Helix E binds to that both the extracellular domain of the re-
the periphery of the bundle interacting primar- ceptor and the receptor binding mode are sim-
ily with helices B, C, and the A–B connecting ilar to that of Class I cytokines and receptors
loop. Despite possessing a common fold, fam- as exemplified by the growth hormone–growth
ily members are divergent at the sequence level; hormone receptor interaction (50), despite the
for instance, IL-22 and IL-10 share only 22% differences in sequence and stoichiometry. Like
sequence identity. Class I receptors, Class II receptors consist
The important structural distinction within of tandem β-sheet rich Ig-like domains with
the family is between the domain-swapped Fibronectin type III connectivity. Intrastrand
dimers exemplified by IL-10 (and viral or- loops contributed by both domains as well as
thologs) and likely by IL-26 and the monomeric the short interdomain linker form an elbow-like
family members such as IL-19, IL-20, IL-22, ligand-binding surface. The receptor-ligand
IL-24, and the IL-28s (IFN λs) (Figure 1a,b). binding mode is essentially identical for ei-
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

The distinctive intertwined dimer interaction ther dimeric (IL-10 or IFN-γ) or monomeric
was first revealed by the structure of IFN-γ (IL-22) cytokines. In both cases, the receptor
(39). Subsequent structural studies of IL-10 binds to the surface formed by the helices A
by University of Guelph on 05/31/12. For personal use only.

(40, 41) showed that it also forms a domain- and F and the AB loop.
swapped dimer in which two adjacent peptide Recent studies of the structure of the low-
chains exchange helices E and F to form a affinity IL-10R2 receptor confirm that it pos-
twofold symmetric, reciprocal dimer. The sesses a similar molecular architecture despite
addition of six amino acids to the DE loop binding several orders of magnitude more
results in a functional IL-10 that behaves as weakly to cognate cytokines than do the respec-
a monomer in solution, indicating that the tive high-affinity receptors (51). Yoon, Walter,
ability to form a domain-swapped dimer is and coworkers (52) used a combination of mu-
modulated, at least in part, by loop length and tagenesis and computational docking to suggest
conformation (42). Intriguingly, IL-22 has that IL-10R2 likely binds to helices A and D on
been reported to form higher-order species its cognate ligands adjacent to the high-affinity
including dimers and tetramers at high protein interaction site. This proposal is consistent with
concentration. Notably, molecular envelopes what has been seen more broadly for cytokine-
determined by small-angle X-ray scattering receptor interactions (for a recent review, see
indicate that the overall shape of these IL-22 53).
dimers is remarkably similar to that of the IL-10 family cytokines activate the Jak-
intertwined IL-10 dimer (43). Although the STAT signaling pathway (Figure 2). STAT3
biological importance of this phenomenon has is the key downstream transcription factor
not been determined, investigators postulate used by IL-10 and IL-20 subfamily cytokines,
that differences in stoichiometry depending whereas the ISGF3 (IFN-stimulated gene fac-
on protein concentration may affect signaling. tor 3) complex is induced by type III IFNs (31)
Despite this solution behavior, crystal struc- (Figure 2). Jak1 and Tyk2 are associated with
tures of IL-22 either alone (44) or bound to IL-10R1 and IL-10R2, respectively (54, 55).
IL-22R1 (45) do not show any evidence of IL-10 activates STAT1, STAT3, and some-
domain-swapped dimer formation. times STAT5. STAT3, but not STAT1, is crit-
The basic paradigm of Class II cytokine ical in mediating the function of IL-10 in im-
receptor-ligand interaction was revealed by the mune cells (56, 57) (Figure 2). The suppressive
structure of IFN-γ bound to its high-affinity effects of IL-10 on the production of proinflam-
receptor in 1995 (46) and was confirmed by matory cytokines are completely abolished in
structure of the IL-10-IL-10R1 (47), IL-22-IL- macrophages and neutrophils lacking STAT3
22R1 (45, 48), and IL-22-IL-22BP complexes (57).

www.annualreviews.org • IL-10 Family Cytokines in Immunity 75


IY29CH04-Ouyang ARI 4 February 2011 15:27

IL-10 IL-22 IL-28

IL-10R1 IL-10R2 IL-22R IL-10R2 IL-28R IL-10R2

Jak1 Tyk2 Jak1 Tyk2 Jak1 Tyk2

Y Y
Y STAT2
Y ?
Y
Y

Y
STAT1
MEK
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

IRF9
ERK JNK p38
by University of Guelph on 05/31/12. For personal use only.

ISGF3
STAT1 STAT3 STAT5 STAT1 STAT5 STAT3

NUCLEUS

Figure 2
Receptors and downstream signaling of IL-10, IL-22, and IL-28. IL-10 family cytokines primarily activate Jak/STAT pathways upon
binding to their functional receptors. IL-10 and IL-22 mainly signal through STAT3. IL-22R can associate with STAT3 in a
tyrosine-independent manner. IL-28 and IL-29 activate the ISGF3 (IFN-stimulated gene factor 3) complex.

IL-20 subfamily cytokines also activate they are dispensable for STAT3 activation.
STAT3. Among this group of cytokines, the IL-22R can use a novel mechanism to consti-
downstream signaling pathway of IL-22 is cur- tutively bind to STAT3 through its C-terminal
rently the most well understood (Figure 2). As tail interacting with the coiled-coil domain
is the case for IL-10, IL-22 induces activation of STAT3 (59). In addition to the Jak/STAT
of the Jak/STAT pathway upon binding to pathway, IL-22 can also activate ERK, JNK,
the IL-22R/IL-10R2 heterodimeric receptor and p38 MAP kinase pathways in a rat hepatoma
complex. Jak1 and Tyk2, but not Jak2, are phos- cell line, H4IIE, and in a human intestinal
phorylated upon IL-22 binding (58). IL-22 epithelial cell line, HT29 (58, 60). Further-
stimulation can activate three STAT molecules, more, AKT phosphorylation is reported upon
including STAT1, STAT3, and STAT5 (23, IL-22 treatment in H4IIE, HT29, and murine
58). There are in total eight tyrosine residues in hepatocytes (58, 60, 61). However, in a murine
the intracellular domain of IL-22R (59). Inter- breast cancer cell line, EMT6, IL-22 seemed
estingly, although these tyrosine residues are to promote a cell cycle arrest through the in-
required for the recruitment and activation of hibition of ERK1/2 and AKT phosphorylation
STAT1 and STAT5 upon IL-22 stimulation, (62).

76 Ouyang et al.
IY29CH04-Ouyang ARI 4 February 2011 15:27

CELLULAR SOURCES AND Th2 cells are likely the main source of IL-24 in
REGULATION OF IL-10 T cells, while the expression of IL-26 follows
FAMILY CYTOKINES IL-22 in most examined cases in the human
IL-10 family cytokines are produced by various system (77–79).
leukocytes. Tissue epithelial cells can also
secrete some of these cytokines. IL-10 and IL-10 in regulatory T cells. Regulatory T
IL-22 are the most well studied among the cells constitute a largely heterogeneous pop-
family members in terms of their regulation. ulation of cells that can be defined based on
IL-10 is expressed by cells of the innate and the their potential to suppress other immune cells
adaptive immune system, including dendritic and thereby limit or suppress immune re-
cells (DCs), macrophages, mast cells, natural sponses. Among IL-10 family members, IL-10
killer (NK) cells, eosinophils, neutrophils, is the only cytokine produced by these cells
CD4 and CD8 T cells, and B cells (reviewed and is required for their function. The best-
characterized subset of regulatory T cells is the
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

in 2, 63–69). Similarly, IL-22 is produced by


various leukocytes, such as CD4, CD8, γδ T FoxP3+ Treg that originates from the thymus.
cells, NK cells, DCs, and various lymphoid These cells usually do not express IL-10 upon
restimulation ex vivo (80, 81) unless they are
by University of Guelph on 05/31/12. For personal use only.

tissue inducer (LTi)-like subsets (70–75).


Consistent with their different functional roles isolated from the gut (82). The role of IL-10
in inflammation, the regulation of these two for the suppressive function of Tregs has been
cytokines is drastically different. extensively reviewed elsewhere (65, 67, 83–86).
IL-19, IL-20, and IL-24 are primarily gen- However, the signals triggering IL-10 produc-
erated by myeloid cells (76). T cells and ep- tion in Tregs are not well defined. IL-2 and
ithelial cells can secrete some of these cytokines IL-4 induce IL-10 production in Tregs in vitro
upon certain stimulations. IL-28A, IL-28B, and (87, 88). The signals required for IL-10 expres-
IL-29 can be produced by leukocytes (5, 31, 32). sion in vivo remain elusive, although TGF-β is
The induction of these type III IFNs is report- required (82).
edly similar to that of type I IFNs; however, Various antigen-induced T cell populations
detailed analysis of the regulation in individ- with suppressive capacities have been described
ual cell types is still lacking. The regulation of (reviewed in 65, 83, 89, 90). These cells were de-
IL-26 awaits further characterization. Existing fined as regulatory T cells based on either their
data suggest that in humans the expression pat- expression of FoxP3 or their production of reg-
terns of IL-26 are similar to those of IL-22, con- ulatory cytokines, such as IL-10, in the absence
sistent with the linkage of these two cytokines of classical effector cytokines. IL-10-producing
in the genome (77–79). Tr1 cells were originally generated by repeated
in vitro stimulation of T cells in the presence of
IL-10 (91). Treatment with immunosuppres-
sive drugs, such as the combination of vitamin
The Production and Regulation of D3 and dexamethasone, has been shown to in-
IL-10 Family Cytokines in T Cells duce Tr1 cells (92). In vivo TGF-β, but not
T cells are one of the sources of IL-10, IL-22, IL-10, is required for the development of IL-
IL-24, and IL-26. IL-10 can be produced by 10-competent regulatory T cells (82).
virtually all T cell subsets, including Th1, Th2,
Th9, and Th17 effector T cells, regulatory T Th2 cells produce IL-10 and IL-24. IL-10
cells, and CD8+ T cells (reviewed in 2, 63–69). production was first described in Th2 cells (6,
IL-22 is produced by Th1, Th17, γδ T cells, 7). In these cells, IL-10 is coexpressed with
and CD8 T cells. However, Th2, Th9, regula- other Th2 cytokines IL-4, IL-5, and IL-13 and
tory T cells, and B cells have not been reported is regulated by Th2-associated signaling path-
to produce significant amounts of IL-22. So far, ways and transcription factors, including IL-4,

www.annualreviews.org • IL-10 Family Cytokines in Immunity 77


IY29CH04-Ouyang ARI 4 February 2011 15:27

STAT6, and GATA3 (93–95). Recently, stud- 18 can further enhance IL-22 production from
ies demonstrated that IL-27, a member of the Th1 cells (M. Munoz, Y. Zheng, K. Wong, C.
IL-12 cytokine family that by itself promotes Loddenkemper, M. Heimesaat, O. Liesenfeld,
Th1 differentiation, enhances IL-10 produc- and W. Ouyang, manuscript in preparation).
tion in Th2 cells in a STAT1- and STAT3- IL-22 is one of the signature cytokines, be-
dependent manner (96). A Th2-related subset, sides IL-17 and IL-21, that are preferentially
Th9, has been described in mouse and human produced by Th17 cells (114–116). Although
(97–99). Th9 cells are generated in the pres- IL-6 and TGF-β are indispensable for de novo
ence of IL-4 and TGF-β and produce IL-9 as a differentiation of murine Th17 cells (117–119),
signature cytokine. Th9 cells can produce high IL-6 alone is sufficient to induce IL-22 from
levels of IL-10 (97–99) but seem to lack sup- naive T cells in vitro in mouse and human (116,
pressive potential (98). The regulation of IL-10 120). However, IL-6 is not essential for IL-22
expression in these cells is presently unknown. induction in vivo, at least in Concanavalin A
Unlike IL-10, which is essentially produced (ConA)-induced hepatitis and Citrobacter roden-
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

by all Th subsets, IL-24 is preferentially pro- tium infection models (61, 121), whereas IL-
duced by Th2 cells (100). IL-24 lacks immune 6 is required for maximal IL-17 production
repressive functions, and its detailed regulation (121). Interestingly, IL-21 was found to pro-
by University of Guelph on 05/31/12. For personal use only.

in Th2 cells is currently unclear. mote IL-22 expression comparable to IL-6 in


vitro (122), suggesting that IL-21 and IL-6 may
Regulation of IL-10 and IL-22 in Th1 and have a redundant role. In combination with tu-
Th17 cells. Shortly after the initial descrip- mor necrosis factor (TNF)-α, IL-6 is particu-
tion of IL-10 as a Th2 cytokine, studies found larly effective in promoting the differentiation
that it is produced by human Th1 cells as well of a recently identified population of human
(101, 102). Th1 cells require strong activation Th22 cells (120) that produces IL-22 but not
and the presence of IL-12 in order to coex- IL-17.
press IFN-γ and IL-10 (95, 103–105). As in IL-23, although not sufficient to induce de
Th2 cells, IL-27 also induces IL-10 production novo IL-17 production from naive T cells (112,
in Th1 cells in mouse (96, 106–109) and human 117, 118), is essential in promoting IL-22 pro-
(110). TGF-β acts synergistically with IL-27 in duction from many immune cell types. IL-23
inducing IL-10 (96, 111). alone induces IL-22 production from CD4+
Several groups have recently shown that and CD8+ T cells, γδ T cells, monocytes,
Th17 cells produce IL-10 (96, 105, 106, 112). CD11c+ DCs, and LTi cells (70, 74, 112, 116).
The cytokine combination of TGF-β and IL-6, In vivo, IL-22 induction is significantly com-
besides potently inducing Th17 differentiation promised in IL-23-deficient mice under various
and IL-17 expression, induces IL-10 produc- immune challenges (122, 123–126).
tion in Th17 cells (Figure 3) (112). In contrast, Finally, the roles of TGF-β in the regulation
IL-23 is required for the long-term in vivo func- of IL-17 and IL-22 production are strikingly
tion of Th17 cells but inhibits IL-10 production different. TGF-β is required for robust IL-17
from these cells (112). In addition, the combi- and IL-10 induction in naive T cells (117–119)
nation of IL-27 and IL-6 is a strong inducer of (Figure 3). In contrast, not only is TGF-β
IL-10 in Th17 cells (96), whereas IL-27 alone not required for the induction of IL-22 expres-
largely suppresses IL-17 production while in- sion, but it also potently inhibits IL-22 expres-
ducing IL-10 (110). sion in murine Th17 cells in a concentration-
Th1 cells were originally identified as the dependent manner (116). TGF-β completely
source of IL-22 as well (113). Few data exist blocks IL-22 induction in human Th22 cells
on the detailed regulation of IL-22 from Th1 generated by IL-6/TNF treatment (120). The
cells. Presumably, IL-12 is required for the in- molecular basis for this suppressive effect is cur-
duction of IL-22. We have also noticed that IL- rently unknown.

78 Ouyang et al.
IY29CH04-Ouyang ARI 4 February 2011 15:27

IL-10 regulation IL-22 regulation

Dll-4 Dll-4
Notch Notch
receptor receptor
IL-12 IL-27 IL-21 IL-6 IL-4 TGF-β TGF-β IL-6 IL-23 IL-21
AhR
ligand
IL-12R IL-27R IL-21R IL-6R IL-4R TGF-βR TGF-βR IL-6R IL-23R IL-21R

AhR
HSP90
NICD ? NICD
STAT1

STAT1

STAT3

STAT3

STAT6

STAT6

STAT3

STAT3
STAT4

STAT4
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

CYTOSOL

NUCLEUS
?
by University of Guelph on 05/31/12. For personal use only.

? ? ?

STAT3

STAT3
?
STAT
STAT

MAML RBP-Jκ GATA3 c-Maf ARNT AhR MAML RBP-Jκ RORα RORγt BATF

IL-10 IL-22

Figure 3
Regulation of IL-10 and IL-22 in T cells. The expression of IL-10 and IL-22 is controlled by both common and distinct transcription
factors. STAT3 and Notch were shown to be involved in the transcriptional regulation of both cytokines. STAT3 activation by IL-27,
either alone or in combination with IL-6, or by TGF-β plus IL-6, promotes IL-10 production. Activated by IL-23, IL-6, and IL-21,
STAT3 is also crucial for IL-22 expression. Triggering of Notch by Dll-4 induces IL-10 expression in Th1 cells in synergy with
STAT4. Notch also induces IL-22 production in Th17 cells, possibly by activating AhR. Other factors, such as AhR, RORγt/RORα,
and BATF, control IL-22 production but are not involved in IL-10 regulation, whereas c-MAF and GATA3 control only IL-10
expression. (Dashed lines indicate transcriptional induction of the respective factor. Question marks indicate that the proposed binding
to the IL-10 or IL-22 promoter has not been demonstrated experimentally. Abbreviations: AhR, aryl hydrocarbon receptor; ARNT,
aryl hydrocarbon receptor nuclear translocator; BATF, basic leucine zipper transcription factor, ATF-like; Dll-4, Delta-like-4;
MAML, Masterlind-like; NICD, Notch intracellular domain; RBP-Jκ, recombining binding protein suppressor of hairless.)

Clearly, different cytokine networks are re- activation by IL-27, either alone or in com-
quired for the production of IL-10 and IL-22 bination with IL-6, or by TGF-β plus IL-6,
from Th1 and Th17 cells (Figure 3). Both com- promotes IL-10 production by Th1, Th2, and
mon and distinct transcription factors control Th17 cells in mouse and human (96, 106–108,
the production of these two cytokines in T cells. 110–112, 129).
STAT3 and Notch are two common transcrip- STAT3 is essential for the differentiation
tion factors that regulate IL-10 and IL-22 pro- of Th17 cells and for the expression of the
duction from Th1 and Th17 cells. Two puta- Th17 key transcription factors RORγt and
tive STAT-binding sites have been identified in RORα in response to TGF-β, IL-6, and IL-23
the murine and human IL-10 promoter (127, (130–132). Because all cytokines reported to
128). Generally, STATs are among the most induce IL-22 production, i.e., IL-23, IL-6,
universal and crucial transcriptional regulators and IL-21, signal through STAT3, it is not
of IL-10. STAT1, STAT3, and STAT4 have surprising that STAT3 is crucial for IL-22
all been linked to IL-10 regulation (103, 104, expression. Overexpression of a constitutively
106). Several reports demonstrate that STAT3 active STAT3 strongly enhances IL-23R and

www.annualreviews.org • IL-10 Family Cytokines in Immunity 79


IY29CH04-Ouyang ARI 4 February 2011 15:27

IL-22 expression (133). STAT3-deficient T binding to a MARE (MAF recognition element)


cells are impaired in their expression of IL-22, motif in the IL-10 promoter (129). Taken to-
as well as of IL-17, IL-23R, and RORγt under gether, these findings are suggestive of c-MAF
Th17-polarizing conditions in vitro (122, 133). as a universal transcriptional regulator of IL-10
Activation of the Notch pathway via its lig- expression in a wide range of immune cells.
and Delta-like-4 (Dll-4) promotes the coex- Sp1 (140, 141), C/EBPs (142–144), IRF1
pression of IL-10 and IFN-γ in Th1 cells (127), activator protein-1 (AP-1) (145, 146),
(134, 135). Furthermore, Notch strongly syn- NF-κB (147), and CREB (148) all reportedly
ergizes with IL-12 and IL-27 in the induc- regulate IL-10 expression in myeloid cells and
tion of IL-10 from Th1 cells (134). The T cells. AP-1 activity has been associated with
Notch pathway also promotes IL-10 produc- IL-10 production in T cells and macrophages.
tion from Th17 cells, although at a lower level An AP-1-specific binding motif has been
(S. Rutz and W. Ouyang, unpublished ob- identified in the noncoding region that is
servation). The Notch pathway has recently approximately 1 kb downstream of the IL-10
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

been implied in IL-22 regulation. Costimula- transcribed region (14, 149). This element
tion of CD4+ T cells with either recombinant is responsible for JUN/AP-1-driven IL-10
Dll-4 (136) or antigen-presenting cells (APCs) production that is observed in Th2, but not
by University of Guelph on 05/31/12. For personal use only.

overexpressing Dll-1 (137) in the presence of Th1, cells (14, 149). IL-10 production by T
TGF-β/IL-6 or TGF-β/IL-6/IL-23 strongly effector cells also requires ERK activation (105,
induces IL-22 production in vitro. More impor- 150). In Th1 cells, IL-10 expression depends
tantly, the Notch pathway also regulates IL-22 on STAT4 and ERK. IL-21 and IL-27, both
production in vivo. RBP-Jκ-deficient mice, in potent inducers of IL-10 production in T cells
which Notch signaling is blocked altogether, (see above), activate ERK (151, 152).
are highly susceptible to the detrimental im- Different sets of transcription factors are
munopathology associated with ConA-induced required for the production of IL-22 in T cells
hepatitis but could be protected by exogenous (Figure 3). The retinoic acid–related orphan
IL-22 (137). Consistently, CD4+ T cells from nuclear receptor RORγt is necessary and
RBP-Jκ-deficient mice were largely impaired sufficient to drive Th17 development (153).
in their ability to produce IL-22 through T cell Its closely related family member, RORα, also
receptor–mediated stimulation (137). participates in the regulation of IL-17A and IL-
Several other transcription factors regulate 17F production in vitro and in vivo (132). Dou-
the expression only of IL-10 and not of IL-22. ble deficiencies in RORα and RORγt globally
First, c-MAF belongs to the MAF family of impair Th17 generation and completely pro-
basic region and leucine zipper transcription tect mice against experimental autoimmune
factors. Originally, c-MAF had been described encephalomyelitis (EAE) (132). Interestingly,
as a Th2 transcription factor directing the most, but not all (120), cell types that produce
expression of Il4 and thereby promoting Th2 IL-22 express RORγt as well (72–74, 77, 132,
differentiation (138, 139). Recently, c-MAF was 154–159). The exact involvement of RORγt
also implicated in the expression of IL-10 by and RORα in the transcriptional regulation of
various T cell subsets (105, 108, 129). c-MAF IL-22 is not well understood. RORα-deficient
expression correlates with IL-10 production in T cells show normal IL-22 expression under
Th1, Th2, and Th17 cells and is dependent on Th17 conditions in vitro, and forced expression
ERK activation in Th1 and Th17 cells (105). c- is not sufficient to induce IL-22 (132). Like-
MAF is synergistically upregulated by IL-6 plus wise, overexpression of RORγt alone is not
TGF-β or IL-27 plus TGF-β in Th17 cells, sufficient for IL-22 expression in murine (160)
and it is downstream of IL-27-induced IL-10 or human T cells (78). RORγt-deficient cells,
production in Th1 cells (108). c-MAF directly however, show strongly reduced IL-22 expres-
induces IL-10 expression in T cells through sion in vitro in the presence of TGF-β/IL-6 or

80 Ouyang et al.
IY29CH04-Ouyang ARI 4 February 2011 15:27

TGF-β/IL-6/IL-23 (132) or TGF-β/IL- induce IL-22 even in the presence of its ligand
21/IL-23 (122). Reduced IL-22 production is FICZ (160).
observed in human Th22 cells after treatment Other transcription factors that may reg-
with a small interfering RNA against RORγt ulate IL-22 production include basic leucine
(157). However, Duhen et al. (120) report that zipper transcription factor, ATF-like (BATF),
human Th22 cells produce IL-22 while ex- IRF4, and IκBζ (Figure 3). BATF is a member
pressing low to undetectable levels of RORγt. of the AP-1 superfamily of basic leucine zip-
When considered together, these findings per transcription factors. T cells from BATF-
suggest that RORγt acts in combination with deficient mice are completely impaired in their
other pathways to regulate IL-22 expression. differentiation into Th17 cells, and the mice are
Interestingly, only RORα/RORγt-double- resistant to EAE (164). BATF-deficient T cells
deficient T cells totally lack IL-22 expression cannot express RORγt, RORα, AhR, IL-17A,
(132). IL-17F, IL-21, or IL-22. IRF4 is crucial for
The cytosolic transcription factor aryl hy- the development of Th17 cells (165, 166). It is
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

drocarbon receptor (AhR) has been iden- the only known Th17-related factor that is in-
tified as a crucial factor in the develop- duced in the absence of BATF. IRF4-deficient
ment of Th17 cells (160–163) and Tregs T cells fail to upregulate RORγt or to pro-
by University of Guelph on 05/31/12. For personal use only.

(162). AhR was suggested as a link between duce IL-17 and IL-22 in response to a cocktail
the adaptive immune system and environ- of cytokines including IL-6 and TGF-β (165).
mental factors because it recognizes numer- IκBζ, a nuclear IκB family member, has re-
ous small xenobiotic and natural molecules, cently been described as a transcription factor
such as 2,3,7,8-tetrachlorodibenzo-p-dioxin required for Th17 development in mice. The
(TCDD) and the tryptophan photometabolite ectopic expression of IκBζ in naive CD4+ T
6-formylindolo(3,2-b)carbazole (FICZ). Th17 cells together with RORγt or RORα potently
cells express particularly high levels of AhR in induces Th17 development, even in the absence
mouse and human (161, 162). AhR deficiency of IL-6 and TGF-β. IL-22 expression upon
impairs but does not prevent Th17 develop- TGF-β/IL-6/IL-23 treatment is markedly re-
ment (161, 163). Interestingly, AhR is essential duced in IκBζ-deficient T cells (167). The ex-
for IL-22 expression in Th17 cells (160, 161), pression of other Th17-associated transcrip-
γδ T cells (72), and human Th22 cells (120, tion factors, including RORα, RORγt, AhR,
157). CD4+ T cells from AhR-deficient mice IRF4, and BATF were found to be normal,
can develop Th17 cell responses, but when con- as was IL-6-induced STAT3 phosphorylation
fronted with AhR ligand they fail to produce (167).
IL-22 (161). Th17 cells from AhR-deficient
mice cannot produce IL-22 despite the pres-
ence of IL-23 under inflammatory conditions Regulation of IL-10 Family Cytokines
such as immunization with CFA or injection in Myeloid Cells and Other Tissue
with heat-killed mycobacteria (161). The exact Cell Types
mode of action of AhR remains elusive. The IL-10, IL-19, IL-20, and IL-24 are closely
core nucleotide sequence to which the nuclear linked in the genome. It is not surprising that
AhR complex binds, also termed the xenobi- all these cytokines can be induced from myeloid
otic responsive element, occurs frequently in cells through TLR activation. IL-10 produc-
the mammalian genome and is also represented tion can be triggered in vitro in both DCs
in Il17a, Il17f, Il22, and rorc. AhR is apparently (168–173) and macrophages (168, 174–176)
associated with other Th17-related factors to by stimulation with certain PRR ligands, e.g.,
activate Il22 transcription, given that, in AhR- Toll-like receptor (TLR) agonists. TLR2 lig-
deficient T cells, forced expression of AhR un- ands appear to be particularly potent inducers
der neutral, Th1, or Th2 conditions does not of IL-10 production by APCs, as demonstrated

www.annualreviews.org • IL-10 Family Cytokines in Immunity 81


IY29CH04-Ouyang ARI 4 February 2011 15:27

with the synthetic TLR2 agonist Pam3cys were involved in mediating protection from
(170, 172) or by reduced IL-10 production colitis (187). Investigations into subsets of
from macrophages during Candida albicans NK cells conducted by several labs revealed a
infection in TLR2-deficient mice (177). TLR9 population of cells that lacked lineage markers,
and TLR4 ligation also trigger IL-10 produc- expressed NKp46, and were resident in mu-
tion from macrophages and DCs (168, 178, cosal tissue such as gut lamina propria, Peyer’s
179). TLR-independent triggers of IL-10 pro- patches, and cryptopatches (77, 155, 159, 189).
duction include DC-SIGN (173) and Dectin-1 These CD3− NKp46+ cells were all reported to
(171). The activation of CD40 reportedly express RORγt, which is essential for the devel-
further enhances IL-10 production by DCs opment not only of Th17 cells, but also of LTi
(169, 171) and induces IL-10 in macrophages. cells. Although these CD3− NKp46+ RORγt+
Myeloid cells are sources of IL-19, IL-20, cells express various markers associated
and IL-24 as well. Lipopolysaccharide (LPS) with NK cells, they express low levels of
stimulates the production of these three IFN-γ and have low cytotoxic capacity but
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

cytokines from monocytes. Pretreatment produce high levels of IL-22 following stim-
of monocytes with IL-4 and IL-13 further ulation (77, 155, 159). Various names have
increases the LPS-induced IL-19 production been proposed to describe IL-22-producing
by University of Guelph on 05/31/12. For personal use only.

(16). GM-CSF can also promote IL-19 expres- NKp46+ RORγt+ cells, including NK22,
sion from monocytes. TNF-α upregulates the NKp46+ RORγt+ , and NCR22 (77, 190–192).
expression of IL-20 from human monocytes Murine NKp46+ RORγt+ cells were found to
(180). In psoriatic skin, IL-20 is mainly de- contribute to resistance to C. rodentium, sug-
tected in CD68+ /CD11C+ DCs. In addition gesting a role in mucosal homeostasis (77, 154).
to myeloid cells and T cells, some tumor cells Murine and human LTi cells also produce
such as melanoma cells also produce IL-24 high levels of IL-22 upon activation (74, 75,
(15). Epithelial cells are also a source for IL-19, 158). Human LTi cells have been identified
IL-20, and IL-24, and IL-20 expression from recently as a lin− CD45+ CD127+ RORC+
keratinocytes can be induced by IL-22, IL-1β, population (75). Human tonsil LTi cells
and ultraviolet B (3, 181–183). As discussed be- produce low levels of IFN-γ but high IL-22
low, the IL-20 subfamily cytokines produced by following stimulation ex vivo. These cells are
myeloid cells and keratinocytes may participate NKp46+ , and activation of LTi cells from the
in the pathogenesis of psoriasis. Stimulation tonsil induces the expression of CD56; fetal
with IL-17, IL-22, or IL-1β induces IL-19 LTi cells acquire NK cell markers following
from human bronchial epithelial cells, which ex vivo culture with γc cytokines, suggesting
may play a role in host defense of bacterial an intrinsic developmental link between LTi
infections (124, 184). Finally, monocytes and cells and NKp46+ NK22 cells (75, 158)
DCs produce low levels of IL-22 upon IL-23 (Figure 4). Recent publications suggest that
stimulation (116, 121, 185, 186). although similar in phenotype to conventional
NK cells, NKp46+ RORγt+ cells in human
and mouse comprise a cellular lineage that is
Expression of IL-22 in NK distinct from, yet developmentally related to,
and LTi-Like Subsets conventional NK cells (158, 193) (Figure 4). In
The T cell compartment is not required for IL- a broader context, conventional NK cells, LTi
22-dependent protection in colitis models such and LTi-like cells, and NKp46+ RORγt+ cells
as C. rodentium infection (121, 187); therefore, may be considered part of a family of innate
an IL-23-responsive cell belonging to the in- lymphoid cells that mediate mucosal home-
nate compartment must also be a source of IL- ostasis. Recently, RORγt+ precursor cells have
22. Initial reports identified IL-22-expressing been identified in fetal liver (194). These cells
NK cells (188), which investigators suggested can develop into both mature LTi cells and

82 Ouyang et al.
IY29CH04-Ouyang ARI 4 February 2011 15:27

IL-22

IL-7Rα IL-7Rα IFN-γ


NKp46 NKp46 NKp46
(human) NK1.1hi
CD4+/–
(mouse)
?
RORγt RORγt
Granzyme,
Perforin
C-Kit C-Kit
NKp44+ NKp44+, CD56+ CD56+
(human) (human) (human)
LTi, NK22, NCR22, NK cells
LTi-like NKp46+RORγt +
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org
by University of Guelph on 05/31/12. For personal use only.

Id2

Common
precursor?

Figure 4
LTi- and NK-like cells are major innate sources of IL-22. Human LTi cells and RORC+ NKp46+ NK22 cells can produce IL-22 upon
activation. These cells may share a common precursor during development.

NKp46+ RORγt+ cells, suggesting that they leukocytes. The IL-10R1 chain is mainly
may be a common precursor for the entire expressed on leukocytes, whereas IL-22R1,
family of innate IL-22-producing lymphocytes. IL-20R1, and IL-28R1 are preferentially ex-
pressed on various tissue epithelial cells and
fibroblasts (9, 10, 17, 35, 36, 195). IL-10 is
DIVERSE TARGET CELLS AND thus the only family member primarily target-
BIOLOGICAL FUNCTIONS OF ing leukocytes, while all other family members
IL-10 FAMILY CYTOKINES preferentially regulate various other cell types,
IL-10 family cytokines target various cell types especially epithelial cells, in the tissues.
in the body. Two β chains and four α IL-28A, IL-28B, and IL-29 induce down-
chains are used by these cytokines as receptors stream antiviral responses, similar to those
(Table 1). The two β chains are relatively triggered by type I IFNs. Type III IFNs also
broadly expressed, whereas the presence of the stimulate the expression of common IFN gene
α chain determines which cytokines a par- signatures, such as Mx1 and Oas1 (31). Nearly
ticular cell type responds to (70, 76). The identical gene signatures are induced by type I
IL-10R2 chain is ubiquitously expressed, and type III IFNs from HepG2 and U266 cells
whereas IL-20R2 expression is more limited (196). Nevertheless, the antiviral and antipro-
to various tissues enriched with epithelial cells, liferative activities induced by IL-28 and IL-29
such as skin and lung (17). A low level of are different from those of IFN-α in terms
expression of IL-20R2 is also detected in of potency and kinetics (197). The activities

www.annualreviews.org • IL-10 Family Cytokines in Immunity 83


IY29CH04-Ouyang ARI 4 February 2011 15:27

induced by type III IFNs are in general weaker lar to those of IL-10 (61, 187). However, in
than those induced by type I IFNs (31, 198). contrast to IL-10, at a cellular level scarce
Furthermore, in contrast to a broad spectrum evidence supports a direct anti-inflammatory
of cell types that respond to stimulation by function for IL-22 (3, 195, 208). Although
type I IFNs, only a few cell types, especially two studies observed that IL-22 induced IL-
epithelial cells, preferentially react to type III 10 production from a colon epithelial cell line,
IFNs (35, 36). IL-28A, IL-28B, and IL-29, Colo205 (77, 209), most expression-profiling
therefore, probably evolved to protect skin and studies on IL-22 in various primary epithe-
mucosal surfaces from viral invasion, which lial cells demonstrate that IL-22 primarily pro-
is similar to the activities of IL-20 subfamily motes innate inflammatory responses, tissue
cytokines in protection of epithelial layers protection, and wound-healing activities (3, 70,
from bacterial infection. 195, 208). In addition, IL-22 does not induce
IL-20 subfamily cytokines, except for IL-26, an anti-inflammatory response from leukocytes
share similar biological activities, especially on because of the lack of IL-22R expression. Inter-
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

keratinocytes, where all receptors for this group estingly, upon forced expression of IL-22R in
of cytokines are expressed. IL-20 subfamily cy- macrophages, IL-22 induces anti-inflammatory
tokines induce broad biological functions that functions in these cells similar to IL-10 (210).
by University of Guelph on 05/31/12. For personal use only.

are essential for epithelial integrity and innate Overexpressing IL-22R in lymphocytes in vivo,
host defense (3, 60, 70, 199, 200). Interest- however, results in systemic inflammation as
ingly, although the receptor for IL-26 is also ex- well as in neutrophilia (211). In conclusion, the
pressed on keratinocytes, IL-26 fails to induce anti-inflammatory activity of IL-22 observed in
downstream effects similar to those induced by several models in vivo is probably mediated by
other IL-20 subfamily cytokines in these cells its indirect tissue protective activity.
(3). However, it does activate STAT3 in colon
cells (25, 60). The reason for this observation is
unclear. It is still controversial whether IL-20 IL-10 FAMILY CYTOKINES IN
subfamily cytokines can function on leukocytes. HOST DEFENSE
Neither IL-20R1 nor IL-22R has been detected Host defenses elicited by IL-10 family cy-
on leukocytes (76, 195). Reported activities of tokines are ancient mechanisms used by verte-
this subgroup of cytokines on leukocytes range brates to protect against various infections (1, 2,
from T cell activation and differentiation and 30). The key functions of the IL-10 family of cy-
myeloid cell cytokine production to enhance- tokines in host defense can be categorized into
ment of multipotential hematopoietic progeni- three areas. First, IL-20 subfamily cytokines in-
tors (201–204). Further molecular characteriza- duce innate defense mechanisms from epithe-
tion of signal transduction pathways is required lial cells against extracellular pathogens, such as
to understand these biological activities. In ad- bacteria and yeast. Second, type III IFNs pref-
dition, IL-24 reportedly exerts special antitu- erentially promote antiviral responses from ep-
mor activity in numerous studies (reviewed in ithelial cells and cooperate with type I IFNs in
205, 206). This antitumor activity is not shared the clearance of viral infections. Third, IL-20
by IL-20 despite using the same receptor. The subfamily cytokines and IL-10 prevent tissue
antitumor activity requires overexpression of damage caused by infections and inflammation.
intracellular IL-24 to induce endoplasmic retic- IL-20 subfamily cytokines stimulate epithelial
ulum stress activities (205, 207). Therefore, it is cell proliferation, antiapoptotic responses, and
unclear whether this function reflects the natu- tissue remodeling and healing. IL-10, on the
ral biology of IL-24 as a cytokine. other hand, regulates and represses the expres-
Based on its protective activity in vivo and sion of proinflammatory cytokines during the
its shared IL-10R2 chain with IL-10, IL-22 recovery phase of infections and reduces the tis-
may have anti-inflammatory functions simi- sue damage caused by these cytokines.

84 Ouyang et al.
IY29CH04-Ouyang ARI 4 February 2011 15:27

The functions of IL-10 in infections have results were obtained in a cutaneous Leishma-
been recently reviewed (212, 213). IL-10 is a nia major infection model. Here, IFN-γ+ IL-
general suppressive cytokine. It inhibits proin- 10+ FoxP3− cells act to maintain a chronic, non-
flammatory responses from innate and adaptive resolving infection status. Upon adaptive trans-
immunity, and it prevents the lesions in tis- fer, these IL-10-producing FoxP3− T cells,
sues caused by exacerbated adaptive immune re- but not IL-10-secreting FoxP3+ Tregs, can
sponses. IL-10 is thus a central cytokine during interfere with pathogen clearance in L. major–
the resolution phase of inflammation. Consis- infected mice (82, 220). Blocking IL-10 has
tently, as is discussed below, blocking the IL- thus been considered a therapeutic approach
10 pathway in mice causes spontaneous devel- for the treatment of certain chronic infections.
opment of inflammatory bowel disease (IBD). Unlike the anti-inflammatory functions of
However, evolutionarily pathogens have ex- IL-10, the rest of the cytokines in this family
ploited the functions of IL-10 to repress the enhance innate host defense mechanisms, es-
normal host inflammatory responses during in- pecially from various tissues. Next, we focus on
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

fections, thus establishing chronic infectious IL-20 subfamily cytokines, especially IL-22, in
states. Increased IL-10 expression has been as- host defense against bacterial and yeast infec-
sociated with many chronic bacterial and viral tions and the potential roles of type III IFNs
by University of Guelph on 05/31/12. For personal use only.

infections. Furthermore, some viruses can pro- in viral infections. As discussed above, IL-20
duce their own version of IL-10 (vIL-10) to subfamily cytokines induce various antimicro-
directly suppress the immune responses of the bial peptides from epithelial cells. These cy-
host (7). The induction of IL-10 in DCs and tokines probably exert similar and redundant
macrophages represents a powerful mechanism host defense functions during infections. At this
of immune evasion used by various pathogens. time, only IL-22 has been examined extensively
Either IL-10 itself, or the subsequent induc- in vivo during bacterial infections. IL-24 has
tion of T regulatory cells, impairs pathogen been reported to modulate IFN-γ expression
control and clearance in infection models with in vitro and in vivo (221, 222). Administra-
lymphocytic choriomeningitis virus (LCMV) tion of exogenous IL-24 protects the mice from
(214), Schistosoma mansoni (215), Mycobacterium Salmonella typhimurium C5 infection. Although
tuberculosis (216), and Candida albicans (177). IL-20 shares the same receptors as IL-24,
Macrophage-derived IL-10 can inhibit the dif- whether IL-20 has similar biological functions
ferentiation of neighboring cells into classi- is presently unclear. Elevated IL-22 has also
cally activated macrophages, which allows the been detected in individuals who are resistant
macrophage population to be self-regulating to HIV-1 infection despite repeated exposure to
(217). the virus (223). Furthermore, IL-22 is mapped
The secretion of IL-10 by Th1 cells repre- to the locus controlling Theiler’s virus–induced
sents a potent autoregulatory feedback loop that encephalomyelitis (224). However, IL-22 does
protects against excessive inflammation and not elicit direct antiviral responses from hepatic
potential tissue destruction during proinflam- cells, and most likely not from other epithelial
matory Th1-driven immune responses in infec- cells either (225). The in vivo role of IL-22, as
tions. However, this mechanism is exploited by well as of other IL-20 subfamily cytokines, in
pathogens to prevent their elimination, leading viral infections deserves more attention.
to chronic infections (63, 64, 218). Although
IFN-γ/IL-10-secreting T cells had been rec-
ognized for many years, only recent studies dis- IL-22 in Bacterial Infections
covered that IFN-γ+ IL-10+ Tbet+ T cells are An early effort examined the role of IL-22
indeed required in a Toxoplasma gondii infection during infection with Listeria monocytogenes,
model to prevent excessive inflammation result- a gram-positive intracellular bacterium, given
ing in early mortality of the mice (219). Similar the function of IL-22 in liver protection (61).

www.annualreviews.org • IL-10 Family Cytokines in Immunity 85


IY29CH04-Ouyang ARI 4 February 2011 15:27

IL-22 is not required for either the innate or the (226). Increased IL-22 and Th17 responses also
adaptive immune responses against L. monocyto- have been associated with Mycobacterium tuber-
genes. Because of the high expression of IL-22R culosis, Leishmania donovani, and Shigella flexneri
on mucosal epithelial cells, we decided to exam- infections (227–230). The roles of IL-22 in
ine the function of IL-22 during extracellular these infections are less clear currently. Data
bacterial infection of the mucosal system (121). have shown that IL-22 inhibits M. tuberculosis
Citrobacter rodentium, a gram-negative bac- in human macrophages (229). However, in a
terium, attaches to murine colon epithelial cells murine aerosol M. tuberculosis model, IL-22 is
and induces acute infectious colitis, mimicking not required for host defense (231).
attaching and effacing bacterial infections in the Contrary to its protective function during
human. Upon C. rodentium infection, IL-23 and bacterial infections, IL-22 can cause damage in
IL-22 are quickly upregulated in the colon. IL- the intestine during parasite Toxoplasma gondii
23 induces IL-22 production from innate im- infection (232). IL-22 is dispensable for acute
mune cell types including LTi cells, NKp46+ or chronic systemic infections (231). When the
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

NK-like cells, and DCs (74, 77, 121, 154). IL- ileum is infected perorally, T. gondii causes
22 is indispensable for host defense during the massive necrosis, leading to death in C57BL/6
early phase of C. rodentium infection (121). IL- mice. IL-22−/− mice are resistant to the im-
by University of Guelph on 05/31/12. For personal use only.

22-deficient mice display much more severe munopathology induced by T. gondii. Blocking
epithelial damage as well as systemic bacterial IL-22 slows the death rate and alleviates the
dissemination. Many IL-22−/− mice succumb ileum damage and inflammation after the infec-
within the second week after infection. IL-22 tion (232). The downstream mediators of IL-22
is required for the induction of the Reg family in this model are unclear. The potential patho-
of antimicrobial peptides from colon epithelial logical role of IL-22 during infection has also
cells. Reg family proteins are secreted C-type been demonstrated in an acute polymicrobial
lectins that may have essential functions in the sepsis model in which blocking IL-22 function
prevention of systemic bacterial dissemination. in vivo by IL-22BP attenuates bacterial load and
IL-22 is also required for host defense organ failure (233).
against Klebsiella pneumoniae infection in the
lung (124). Blocking IL-22 results in early
death of the infected animals. IL-22 synergizes IL-22 in Yeast Infection
with IL-17 in this model to induce lung repair, T cells are essential for host defense against
proinflammatory chemokines and cytokines, yeast infection. Patients without T cells develop
and Lipocalin-2, which is required for lung severe systemic candidiasis. Depleting CD4+
epithelial cells to kill K. pneumoniae. Salmonella T cells in mice significantly increases tissue
enterica is another gram-negative bacterium damage caused by Candida albicans infection
that can cause systemic infection or gastroen- (234). IL-10 negatively regulates host defense
teritis in humans. Th1 cells and IFN-γ are during C. albicans infection, probably due to its
essential for the clearance of Salmonella in- anti-inflammatory functions. IL-10 deficiency
fections. However, during intestinal infection alleviates systemic C. albicans infection (235).
with Salmonella, IL-23 induces the production Recently, IL-17 and Th17 cells have been sug-
of IL-22 and IL-17 (125, 226). In wild-type gested as cells that play a critical role in host de-
mice, IL-22 is not required for the clearance fense against C. albicans infection (70). In vitro,
of Salmonella infection. In IL-12p35−/− mice, heat-killed C. albicans hyphae promote human
the development of a Th1 response is com- Th17 differentiation and IL-23 production
promised during the infection. Anti-IL-22 (236). In murine models, the IL-17 pathway is
treatment of infected p35−/− mice leads to critical for host defense against C. albicans infec-
liver necrosis, a finding that supports a role for tion (237). IL-22 is also induced from memory
IL-22 in hepatocyte protection in this model T cells by C. albicans stimulation (238).

86 Ouyang et al.
IY29CH04-Ouyang ARI 4 February 2011 15:27

Interestingly, PBMCs from patients with (244). IL-28RA is required, however, for the
chronic mucocutaneous candidiasis (CMC) enhanced antiviral activity induced by TLR3
produce reduced amounts of IL-17 and IL-22 and TLR9 agonists (244). In addition, mice
when stimulated with C. albicans (239). Fur- lacking IL-28RA and IFN-αR are much more
thermore, patients with autoimmune polyen- susceptible to highly attenuated influenza A
docrine syndrome type I (APS-I)/autoimmune viruses that are usually nonpathogenic, suggest-
polyendocrinopathy candidiasis ectodermal ing that type I and type III IFNs cooperate in
dystrophy (APECED) also display CMC. Re- host defense against influenza infections (245).
cently, autoantibodies against IL-17A, IL-17F, Indeed, in vitro, the combination of IL-29 with
and IL-22 have been detected in these patients IFN-α or IFN-γ results in increased inhibi-
(240, 241). These autoantibodies neutralize tion of viral replication (225). Recent genetic
the activities of these cytokines, rendering association studies further support the impor-
the patients increasingly susceptible to yeast tant cooperative functions between the type III
infection. In a murine model of oropharyngeal IFN pathway and the type I IFN pathway. A
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

candidiasis, the IL-17 pathway but not the genetic polymorphism linked to the gene for
IL-22 pathway exerts a host defensive function IL-28B has been associated with clinical re-
(242). However, when mice are infected with sponse to IFN-α and ribavirin therapy in pa-
by University of Guelph on 05/31/12. For personal use only.

C. albicans intragastrically, IL-23 induces IL-22 tients with chronic hepatitis C infection (246–
in the stomach (185). Mice deficient for IL-22 249). The nonresponsive allele is reported to
have increased dissemination of the yeast into correlate with lower IL-28A/B expression (248,
the stomach and kidney. IL-22 is required for 249). Taken together, IL-28 and IL-29 are es-
the upregulation of antimicrobial proteins such sential in regulating host defense against viral
as S100A8, S100A9, RegIIIβ, and RegIIIγ infections. Based on these data, pegylated-rIL-
during the infection (185). Thus, IL-22, as 29 is currently being tested in the clinic for
well as other Th17 cytokines such as IL-17A, the treatment of patients with chronic hepati-
is indispensable for host defense against yeast tis C infections (243). Future combined strate-
infection gies with type I and type III IFNs may provide
promising therapies for patients with hepatitis
C infection.
Type III IFN in Viral Infections
IL-28A, IL-28B, and IL-29 induce related weak
antiviral responses. The expression of their re- IL-10 FAMILY CYTOKINES
ceptor, IL-28R, is more restricted than the IN THE DEVELOPMENT
widely distributed IFN-α receptor (5). Recent OF AUTOIMMUNE AND
data suggest, however, that the type III IFN INFLAMMATORY DISEASES
pathway may be crucial for control of certain IL-10 is associated with many autoimmune dis-
viral infections. Type III IFNs may also offer eases because of its anti-inflammatory functions
better clinical benefit when used for the treat- (250). In the past several years, accumulated
ment of viral infections, such as hepatitis C in- data have also established an important role for
fection, because their receptors are expressed other IL-10 family cytokines in autoimmune
more specifically on cells that are not from and inflammatory diseases, especially in psori-
hematopoietic lineages, which could reduce se- asis, IBD, and liver inflammation.
vere systemic toxicities such as those caused by
type I IFN (243).
IL-28RA is not required for defense against Protective Functions of IL-10 and
a panel of viral infections, such as herpes sim- IL-22 in the Development of IBD
plex virus type 2 (HSV-2), LCMV, vesicular IBD is a chronic inflammatory disorder of the
stomatitis virus (VSV), and EMCV infections gastrointestinal track that manifests either as

www.annualreviews.org • IL-10 Family Cytokines in Immunity 87


IY29CH04-Ouyang ARI 4 February 2011 15:27

ulcerative colitis (UC) or Crohn’s disease (CD) IL-22 stimulates the production of chemokines
(251). The exact causes of IBD are still unclear, that attract neutrophils and other phagocytes
but people believe genetic predisposition and to control evading pathogens. These cytokines
environmental factors contribute to disease can also help to maintain the integrity of ep-
onset and development. Within the gastroin- ithelial cells. A major cause of tissue damage is
testinal tract, the mucosal epithelial layer epithelial cell apoptosis triggered by inflamma-
constantly interacts with trillions of microor- tion and infection. IL-22 can induce the expres-
ganisms (252, 253). The resident bacterial sion of antiapoptotic genes and also promotes
population and the host have evolved a mutu- proliferation and wound-healing responses,
ally beneficial symbiosis. To properly maintain which help to replenish the damaged tissues.
the harmony of the symbiotic environment, the The association between IL-10 and IBD was
host defense system composed of both the im- first recognized during the analysis of Il10−/−
mune system and the epithelial layer needs to mice (256). Although Il10−/− mice develop
surmount two challenges (252, 253). First, dur- normally and have apparent normal antibody
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

ing homeostasis the epithelial cells need to be responses, these mice spontaneously develop
competent in sequestering luminal commensal colitis that is characterized by abnormal
microbes from penetration into the epithelial architecture in the mucosa associated with the
by University of Guelph on 05/31/12. For personal use only.

layer. In parallel, the immune system needs to infiltration of various leukocyte subsets. Similar
be maintained in a relatively inert state toward colitis also develops in Il10rb−/− mice (13). The
these resident microorganisms and avoid tissue enteric microfloras in the gut are necessary for
damage caused by excessive inflammation. colitis development in these mice (257). Il10−/−
Second, the immune system and epithelial mice raised in a germ-free environment do
system should be able to effectively sense and not develop colitis. In addition, administration
control the invasion of various opportunistic of antibiotics before the onset of disease in
pathogens in the gastrointestinal tract. Im- these mice prevents colitis (258). Antibiotics
portantly, both systems can quickly restore can also attenuate the severity of colitis when
homeostasis upon clearance of the pathogenic used after the disease has developed. CD4+ T
microbes. Substantial evidence from human cells, but not B cells, are essential for the de-
genetic studies and from preclinical IBD velopment of colitis in Il10−/− mice (259). The
models suggests that failures by either the initial proposal that Th1 cells play important
mucosal epithelial layer or the immune system pathogenic roles in this model was due to two
to properly interact with the lumen microbial key observations. First, IFN-γ-producing Th1
community may underlie the pathogenic cells are significantly increased in the gut of
processes during IBD. Il10−/− mice during the development of colitis.
IL-10 family cytokines have critical func- Second, IL-10 directly inhibits macrophages
tions in regulating the homeostatic states of and DCs from producing IL-12, a key driving
the immune system and the mucosal epithelia, factor for Th1 differentiation. IL-12 is a
as well as boosting host defense during the heterodimeric cytokine consisting of p40 and
invasion of various microorganisms (254, 255) p35 subunits, both of which are inhibited by
(Figure 5). IL-20 subfamily cytokines and IL-10 at the transcriptional level in myeloid
type III IFNs preferentially augment antibac- cells (260). Later studies have discovered that
terial and antiviral responses from epithelial the p40 subunit is shared by a novel cytokine,
cells, respectively. IL-10, on the other hand, IL-23 (261). Thus, IL-23 is also inhibited by
targets various leukocytes, attenuates excessive IL-10 during inflammation, which prompted
immune responses, and protects against the ep- the reanalysis of the role of IL-12 and IL-23 in
ithelial damage caused by inflammation. IL-20 colitis development in Il10−/− mice. A detailed
family cytokines, especially IL-22, induce the comparison of mice deficient for the p35
production of various antimicrobial peptides. subunit of IL-12 or the p19 subunit of IL-23

88 Ouyang et al.
IY29CH04-Ouyang ARI 4 February 2011 15:27

Antimicrobial
peptides:
Luminal Reg, S100,
bacteria defensin, etc.

Mucin

Intra- Paneth Goblet


epithelial cell cell
lymphocyte
Neutrophil IL-19, IL-20, and IL-24
DC
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

Chemokines:
IL-8, etc. IL-22
MΦ IL-26
by University of Guelph on 05/31/12. For personal use only.

IL-12, IL-23, IL-1β,


IL-18, and IL-6

IL-10
Th1, LTi NK γδ
Th17 T cell

Figure 5
IL-10 family cytokines in inflammatory bowel disease. During inflammation, microorganisms in the gastrointestinal tract can induce
the expression of many proinflammatory cytokines, such as IL-23 and IL-1β, from myeloid cells. These cytokines promote the
production of IL-22 from various cell types, including Th17 cells and LTi/NK-like innate cells. On the one hand, IL-22 protects the
epithelial layer from further damage caused by microbes or inflammation. On the other hand, IL-10 from leukocytes directly represses
proinflammatory responses.

led to the conclusion that IL-23, but not IL-12, (264). Interestingly, a high IL-10 serum level is
mediates the crucial pathogenic functions in maintained during the early phase of remission,
colitis development of Il10−/− mice (262). whereas C-reactive protein and IL-6 serum
IL-17 and IL-6 are downstream of IL-23 levels return to normal during remission after
and are involved in the pathogenesis, which an increase during the acute phase. Given
supports the idea that Th17 cells but not Th1 the negative regulatory function of IL-10
cells are required. In vitro, IL-10 can inhibit in inflammation, people have long proposed
IL-23 production from DCs and macrophages. treating human IBD with IL-10. Indeed,
Therefore, IL-10 is an essential negative administration of IL-10 has demonstrated
regulator, especially for controlling IL-23 efficacy in various preclinical models of IBD
induction, during mucosal inflammation. (265, 266). In humans, however, recombinant
The importance of IL-10 in IBD is further IL-10 has not provided impressive efficacy
corroborated by its identification as a suscepti- in CD (see review in 254). Multiple factors
bility gene for UC in genome-wide association may contribute to the failure of rIL-10 in
studies (263). Multiple causal variants may clinical trials. First, systemic administration
contribute to the association signal at the IL-10 may not be sufficient to deliver IL-10 to
locus. Elevated IL-10 has also been detected the mucosal inflammatory sites where it can
in the serum of some human UC patients exert its anti-inflammatory functions. Several

www.annualreviews.org • IL-10 Family Cytokines in Immunity 89


IY29CH04-Ouyang ARI 4 February 2011 15:27

strategies are currently being considered to severe weight loss and mortality than if the
efficiently deliver IL-10 to the mucosal sites, same recipient mice only received wild-type T
including virus-mediated expression or com- cells. IL-10 mediates its protective functions
mensal bacteria–mediated gene delivery. Sec- in colitis models through the reduction of
ond, in addition to its anti-inflammatory func- inflammatory responses of various leukocytes.
tions, IL-10 also stimulates proinflammatory IL-22, in contrast, promotes its protective roles
responses, such as IFN-γ production, when it through enhanced tissue innate immunity and
is administered to humans (267). It is unclear integrity (Figure 5). First, IL-22 can induce
whether this function of IL-10 contributes to proinflammatory cytokines and chemokines
its lack of efficacy in the treatment of IBD. from colonic epithelial cells and subepithelial
No genetic association between IL-22 and myofibroblasts, such as IL-6 and IL-8 (60,
IBD has been identified. Elevated IL-22 has 268). These cytokines and chemokines help
been reported in patients with IBD (60, 268). to recruit and activate various leukocytes
Increased Il22 and Il22ra1 messages have been in controlling the invading pathogens, thus
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

detected in UC mucosa tissues (269). Serum preventing further epithelial damage. Second,
samples from CD patients show higher IL-22 IL-22 facilitates goblet cell restitution and
but not IFN-γ or IL-17A (270). CD161+ Th17 boosts the production of mucus-associated
by University of Guelph on 05/31/12. For personal use only.

cells isolated from PBMCs of CD patients proteins from goblet cells. The heavily glyco-
readily produce increased IL-22 and IL-17 sylated mucin family of proteins can form a
when compared with those cells from healthy static external barrier that sequesters various
controls (271). In addition, the IL-22 levels cor- luminal microorganisms from direct interac-
relate with disease severity (272). As discussed tion with epithelial cells (273). Third, IL-22
above, IL-23 regulates IL-22 production. induces the production and secretion of various
Il23R is a CD susceptibility gene. Interestingly, antimicrobial peptides—including defensins,
there is also a remarkable correlation between cathelicidins, and C-type lectins—from intesti-
serum IL-22 levels and IL-23R minor alleles nal epithelial cells and Paneth cells (121, 186,
and a trend toward lower IL-22 levels in CD 187, 273, 274). These antimicrobial peptides
patients having the protective SNPs of Il23 can target the integrity of the bacterial cell wall
(272). These results are corroborated by the and kill or sequester the invading microbes
recent finding that T cells from people bearing in the mucin layer. Finally, IL-22 activates
the protective SNP (R381Q) in IL-23R are STAT3 in epithelial cells and enhances mu-
hyporesponsive to IL-23 stimulation owing to cosal wound healing (60, 186, 273, 274). IL-22
the lower expression of IL-23R (S. Pidasheva induces the production of various proteases
and H. Clark, manuscript submitted). These T that are important for tissue remodeling. In
cells produce much reduced IL-22 in response addition, IL-22 stimulates the proliferation
to IL-23 stimulation. and reconstitution of mucosal epithelial cells.
IL-22 is also induced in several preclinical The roles of other IL-10 family cytokines
colitis models such as DSS-induced colitis and in IBD are less clear. IL-26 induces STAT1,
the T cell transfer model of colitis. Similar to STAT3, ERK, and JNK activation in colon
IL-10, IL-22 exerts protective functions in pre- cell lines (275). It also stimulates IL-8 produc-
clinical colitis models. Overexpression of IL-22 tion from these cells. In contrast to IL-22, IL-
in TCRα-deficient mice reduces colonic thick- 26 has an antiproliferative effect on intestinal
ness and disease score (273). In DSS-induced epithelial cell lines. In addition, elevated IL-
colitis, blocking the IL-22 pathway leads to 26 has been detected in the inflamed colonic
delayed recovery as judged by body weight loss mucosa of UC and CD patients. The expres-
and histological score (186, 187, 273, 274). sion of IL-19, IL-20, and IL-24 in IBD is un-
Consistently, transferring T cells from Il22−/− clear. A recent study with Il19−/− mice indicates
mice into Rag1−/− Il22−/− mice causes more that IL-19, similar to IL-22, may also have a

90 Ouyang et al.
IY29CH04-Ouyang ARI 4 February 2011 15:27

protective function in mucosal inflammation the proliferation of and has antiapoptotic


(276). In IL-19-deficient mice, DSS treatment effects in hepatocytes (280). IL-22 is upregu-
induces more severe colitis with increased pro- lated during ConA-induced hepatitis. Exacer-
duction of proinflammatory cytokines such as bated hepatitis after ConA administration, as
IL-12 and TNF-α. IL-28A, IL-28B, and IL-29 measured by increased AST and ALT levels
have not been examined in IBD, although IL- and severe liver inflammation, occurs when
28R is expressed in intestinal epithelial cells. IL-22 signaling is blocked with a neutralizing
Murine CMV infection increases colonic IL- antibody (280) or in Il22−/− mice (61). Th17
28A expression in vivo. IL-28A and IL-29 in- cells and NKT cells both produce IL-22 in this
duce antiviral responses and the production of model (61, 281). Interestingly, lack of IL-22 in
IL-8 from intestinal epithelial cell lines (277). a Fas agonist ( Jo-2 mAb)–induced liver injury
These cytokines also inhibit proliferation of and apoptosis model does not alter the severity
colon epithelial cell lines. Further studies are of the liver damage (61), implying redundant
required to elucidate their functions in IBD. pathways for liver protection. Overexpression
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

of IL-22, however, can further protect the liver


from injury caused by Jo-2 antibody (282).
Role of IL-10 Family Cytokines IL-22 is also involved in liver regeneration.
by University of Guelph on 05/31/12. For personal use only.

in Liver Protection After a partial hepatectomy in mice, IL-22R is


Similar to their functions in IBD, IL-10 and augmented on hepatocytes (283), which is as-
IL-22 exert protective functions during liver in- sociated with an elevated IL-22 protein con-
flammation. The target cells and mechanisms centration in the serum. Blocking IL-22 by an
used by IL-10 and IL-22 are different. IL-10 antibody reduces proliferation of hepatocytes.
targets leukocytes and elicits anti-inflammatory IL-22 can also repress genes involved in lipoge-
responses through direct inhibition of the pro- nesis, such as Mlxipl, Ppara, Pparg, and Hmgcr,
duction of proinflammatory cytokines, such as in the liver (284). Long-term administration of
IFN-γ, IL-12, and TNF-α. In contrast, IL-22 IL-22 in vivo reduces the levels of triglycerides
stimulates the proliferation of hepatocytes and and cholesterol in the liver of C57BL/6 and
the regeneration of the liver. IL-22 also induces ob/ob mice fed a high-fat diet (HFD). IL-22
the expression of antiapoptotic genes in hepa- also slightly alleviates hepatic steatosis in HFD-
tocytes and protects them from inflammation- fed mice.
induced cell death. In conclusion, IL-10 and IL-22 both pro-
IL-10 is induced in the liver and in the serum tect the liver during inflammation, although
quickly after ConA treatment in vivo (278). through different mechanisms. IL-20R2 is also
Anti-IL-10 antibody treatment before admin- upregulated in the liver upon LPS stimulation
istration of ConA leads to the development of (285), suggesting that IL-19, IL-20, and IL-24
a more severe hepatitis as well as increased ex- may also exert similar protective functions in
pression of IL-12, TNF-α, and IFN-γ produc- the liver. Future detailed studies on these three
tion in the serum (278). Similar results have also cytokines in liver inflammation will help unveil
been observed in IL-10-deficient mice (279). their functions.
Conversely, recombinant IL-10 represses these
proinflammatory cytokines and protects the
liver from ConA-induced injury (278, 279). Role of IL-20 Subfamily Cytokines
Furthermore, IL-10 also plays a protective role in Psoriasis
in liver injury induced by galactosamine and Although IL-20 subfamily cytokines are impor-
lipopolysaccharide (Gal/LPS) (278). tant for tissue protection, as discussed above,
The liver has a high expression of IL-22R uncontrolled responses from these cytokines
(24). IL-22R and IL-10R2 chains are both ex- can be harmful to tissues as well. Psoriasis
pressed on hepatocytes (280). IL-22 promotes is a chronic inflammatory disease of the

www.annualreviews.org • IL-10 Family Cytokines in Immunity 91


IY29CH04-Ouyang ARI 4 February 2011 15:27

skin characterized by abnormal keratinocyte 199, 208). Expression of S100A7 (also called
differentiation and proliferation, leukocyte psoriasin) is a hallmark feature of psoriatic skin.
infiltration of the dermis and epidermis, and Furthermore, the IL-20 subfamily cytokines,
increased dilation and growth of blood vessels especially IL-22, regulate proteins involved
(286). The crosstalk between keratinocytes and in tissue remodeling, such as MMP1, MMP3,
leukocytes is essential during the pathogenesis Kallikreins, marapsin, and platelet-derived
of psoriasis. Various leukocytes, including T growth factor (3, 188, 199, 294). IL-20
cells, neutrophils, DCs, and macrophages, subfamily cytokines also activate proinflam-
infiltrate the skin and contribute to the inflam- matory responses through the induction of
matory phenotypes of the disease. Immune chemokines and cytokines from keratinocytes
cells produce various soluble mediators such as (3, 199). Various chemokines, including
cytokines and chemokines that orchestrate the CXCL1, CXCL5, and CXCL7, can be induced
disease manifestations. Over the past several by IL-20 subfamily cytokines (3, 199).
years, the IL-20 subfamily cytokines have IL-22 is the most potent cytokine in the in-
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

been identified as the key cytokines involved duction of downstream effects described (3). In
in psoriasis. First, IL-20 subfamily cytokines fact, IL-22 can induce IL-20 and IL-24, but not
are elevated in psoriatic skin. Second, IL-20 IL-19, expression in keratinocytes (3, 183, 289).
by University of Guelph on 05/31/12. For personal use only.

subfamily cytokines induce many important IL-22 can be produced by Th17 cells, which
pathological features in keratinocytes. Third, also secrete IL-17. Interestingly, IL-17 induces
IL-22 and other family members are essential many of the same genes as IL-22, and combin-
for the development of psoriasis in preclinical ing IL-22 and IL-17 leads to a synergistic induc-
models. Finally, IL-22 is a downstream cy- tion of genes, including β-defensins, lipocalin,
tokine induced by the IL-23 pathway, which is and S100 genes (114, 289). An even greater syn-
genetically associated with human psoriasis. ergistic effect is seen when keratinocytes are
Expression of IL-19, IL-20, IL-22, and IL- cultured with IL-22, IL-17, oncostatin M, IL-
24 cytokines has been detected in psoriatic skin 1α, and TNF-α (295), suggesting that in addi-
and not in healthy skin (195, 287, 288). As dis- tion to IL-20 subfamily cytokines, other proin-
cussed above, various leukocytes can produce flammatory cytokines together orchestrate the
IL-20 subfamily cytokines. Myeloid cells and pathogenesis of psoriasis. A recent study com-
keratinocytes are potential sources of IL-19, pared gene expression transcriptome data from
IL-20, and IL-24 (3, 183, 289). The main source psoriasis and primary keratinocytes treated with
of IL-22 in skin lesions appears to be T cells. In various cytokines (296). Interestingly, there is
psoriatic patients, T cells isolated from the skin very little overlap between gene changes in pso-
lesions produce much higher levels of IL-22 riasis samples and cultured keratinocytes that
than do T cells in circulation (290). In addition, are treated with any single cytokine. However,
T cell clones generated from psoriatic tissue are we have noticed that monolayer keratinocytes
largely CCR6+ IL-22+ (291), presumably Th17 do not respond to IL-20 subfamily cytokines
cells or T helper cells that solely produce IL-22, as well as do stratified keratinocytes in recon-
the so-called Th22 cells (292). stituted human epidermis (RHE) culture (3).
The receptors for IL-20 subfamily cy- There is much greater overlap in terms of genes
tokines are highly expressed on keratinocytes regulated by IL-20 subfamily cytokines from
(3). IL-19, IL-20, IL-22, and IL-24, but not RHE and genes regulated in psoriatic skin, sup-
IL-26, activate STAT3 either in primary porting an important pathogenic role of this
keratinocytes (3, 195, 199) or in keratinocyte family of cytokines in psoriasis.
cell lines (293). All IL-20 subfamily members, Studies with transgenic mice overexpressing
except IL-26, induce the expression of various IL-20 family cytokines further strengthen their
antimicrobial peptides including S100 family pathogenic roles in psoriasis. Mice overexpress-
genes and β-defensin family genes (3, 114, ing IL-20, IL-22, and IL-24 under the control

92 Ouyang et al.
IY29CH04-Ouyang ARI 4 February 2011 15:27

of various promoters have been generated (17, In another psoriasis-like mouse model induced
297, 298). The phenotypes of these mice are by the transfer of CD4+ CD45RBhi CD25− cells
largely the same, with only a few minor differ- into SCID mice, neutralization of either IL-22
ences reported. All mice die as neonates smaller or IL-23 prevents disease development as well
in size than littermates, with IL-22 and IL-24 (305). Finally, a human xenograft model has re-
transgenic mice reported to die within a few cently been described in which human psoriatic
days and IL-24 transgenic mice within a few skin can be transplanted onto immunodeficient
hours. The reason for the mortality is not en- mice. In this model, treating transplanted mice
tirely clear. These transgenic mice all exhibit with blocking antibodies to IL-20 resolved the
tight, wrinkled, shiny skin with a thickened epi- psoriasis condition (306).
dermis. Histological analysis of the skin reveals In summary, IL-20 subfamily cytokines are
many hallmarks of psoriasis, including epider- essential in mediating some of the key patho-
mal hyperplasia (acanthosis) and compact stra- logical features during the course of psoriasis.
tum corneum. Blockade of p40, the common subunit of IL-12
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

Genetic association studies and preclinical and IL-23, has shown great efficacy in psoria-
data in psoriatic models also support the func- sis (307). Given the importance of IL-23 in the
tions of IL-20 subfamily cytokines in psoriasis. induction of IL-22 and that blockade of IL-22
by University of Guelph on 05/31/12. For personal use only.

Candidate SNP approaches suggest that IL-19, in mice ameliorates IL-23-induced skin inflam-
IL-20, and IL-24 are associated with suscepti- mation, blocking IL-22 and other family cy-
bility to psoriasis (299–301). These data have tokines could be a novel therapeutic approach
not been independently confirmed in large- in the treatment of psoriasis.
scale genome-wide association studies. IL-23
plays an important role in the development Association of IL-10 Family Cytokines
of psoriasis. The IL-23 p40 subunit and the with Other Autoimmune and
IL-23 receptor have been linked to psoriasis Inflammatory Diseases
in genome-wide association studies (302). IL- IL-10, IL-19, IL-20, IL-22, and IL-24 are
23 does not directly target keratinocytes. Its upregulated in the synovial fluid mononuclear
pathogenic functions on keratinocytes are me- cells of rheumatoid arthritis (RA) (308, 309).
diated through the IL-20 subfamily cytokines, Immunohistological analysis suggests that
especially IL-22. Direct injection of IL-23 in- IL-19 and IL-22 are expressed by synovial
duces ear thickening, acanthosis, and dermal fibroblasts and macrophages (310, 311). IL-20
infiltrates, similar to some features in psori- is observed in sublining mononuclear cells,
atic skin (116, 303, 304). The injection of whereas IL-24 is detected in mononuclear cells
IL-23 into the back skin of the mouse in- and endothelial cells (309). Furthermore, in
duces IL-19 and IL-24 expression in the skin. the PBMCs and joints of RA patients, there are
Consistently, IL-23-induced skin inflammation increased numbers of Th17 cells, which may
and epidermal hyperplasia are attenuated in contribute to the production of IL-22 as well
Il20rb−/− mice (304). We show that IL-23 (291, 312). IL-20R1, IL-20R2, and IL-22R
injection into the mouse ear induces IL-22, are expressed on lining and sublining layers of
but not IL-19, IL-20, or IL-24, expression RA synovial tissues (310, 311). IL-19 increases
(116). Ear thickening induced by IL-23 is re- the production of IL-6 by RA synovium and
duced in IL-22 knockout mice or in wild-type reduces apoptosis of the synovial cells (310).
mice when IL-22 is blocked with a neutral- IL-20 promotes synovial cells to secrete CCL2,
izing antibody, suggesting that IL-22 medi- IL-6, and IL-8, as well as endothelial cell pro-
ates the pathogenic effects of IL-23 on ker- liferation (313). Similarly, IL-22 induces pro-
atinocytes. In addition, injection of IL-22 into liferation of synovial fibroblasts and production
the skin of normal mice induces S100 and β- of CCL2 by these cells (311). Soluble IL-20R1,
defensin, plus keratinocyte hyperplasia (305). which could block IL-19, IL-20, and IL-24,

www.annualreviews.org • IL-10 Family Cytokines in Immunity 93


IY29CH04-Ouyang ARI 4 February 2011 15:27

attenuates disease severity in rats with collagen- autoimmune myocarditis in rats (320). In ad-
induced arthritis (CIA) (313). Elevated IL-22 dition, IL-22 deficiency results in accelerated
is reported in a mouse model of CIA. Il22−/− heart rejection in an allograft transplantation
mice have a delayed onset of disease, reduced model (321). The functions of other IL-10 fam-
disease severity, and decreased proinflamma- ily cytokines, except IL-10 and IL-22, in or-
tory cytokine expression in the joints compared gan protection are largely unexplored. A recent
with those in wild-type mice with CIA (314). genome-wide association study has linked the
IL-20 and IL-22 are reportedly associated IL-10 locus, including IL-10, IL-19, and IL-20,
with lupus (315, 316). IL-20 targets renal with type I diabetes (322). No functional data of
mesangial cells, suggesting its potential role in these cytokines have been reported in diabetes.
lupus nephritis (315). IL-22 levels in plasma, Significant future research will be required to
however, are negatively correlated with disease fully elucidate the role of IL-10 family members
activity in patients with systemic lupus erythe- in autoimmune and inflammatory diseases.
matosus (316). The functions of these cytokines
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

in lupus require further detailed study. Inter-


estingly, serum IL-10 levels are elevated in lu- CONCLUSION
pus. Given the role of IL-10 in enhancing B cell IL-10 family cytokines have very diverse cel-
by University of Guelph on 05/31/12. For personal use only.

proliferation and differentiation, antagonists of lular targets and functions. They have a deep
IL-10 have been considered for the treatment evolutionary connection to the IFN family;
of lupus. however, their functional roles are much
IL-22 induces disruption of the blood-brain broader. The central functions of IL-10 family
barrier (317). In preclinical EAE, a model for cytokines converge on protection of organs and
human multiple sclerosis, however, IL-22 is tissues from damage caused by infections and
not required for the development of the dis- by inflammatory responses. Three key types
ease (123). As discussed above, IL-22 protects of responses are triggered by this family of cy-
the lung from K. pneumonia infection (200). Its tokines. First, type III IFNs, IL-28A, IL-28B,
role in lung inflammation has not been fully and IL-29, stimulate innate antiviral responses
addressed. One study in a preclinical asthma from epithelial cells and protect organ and
model found that IL-22 inhibits eosinophil re- tissue from damage caused by viral infections.
cruitment to the lung, suggesting a negative Second, the IL-20 subfamily cytokines, IL-
role for IL-22 in asthma (318). Patients with sta- 19, IL-20, IL-22, IL-24, and IL-26, act on
ble chronic obstructive pulmonary disease have tissue epithelial cells during extracellular
elevated concentrations of IL-22+ Th17 cells in bacterial and yeast infections to boost innate
the lungs (319). Furthermore, IL-19 is upregu- antimicrobial responses. In addition, IL-20
lated in asthma patients (201). IL-4, IL-13, IL- subfamily cytokines induce wound-healing
17A, and IL-22 can all induce the production responses and promote the integrity of various
of IL-19 from human bronchial epithelial cells tissues and organs. Finally, IL-10 inhibits
(184). The role of IL-19 in asthma development the innate and adaptive immune responses
is currently unclear. from leukocytes and limits the potential tissue
In addition to its role in liver protection, damage caused by inflammation. IL-10 family
IL-22 can also reduce damage to the heart cytokines, therefore, are essential for host
during inflammatory responses. Exogenous defense against various infections and the
IL-22 attenuates the damage from experimental development of many autoimmune diseases.

DISCLOSURE STATEMENT
W. Ouyang, S. Rutz, N.K. Crellin, and S.G. Hymowitz are employees of Genentech, Inc. The
authors are not aware of any affiliations, memberships, funding, or financial holdings that might
be perceived as affecting the objectivity of this review.

94 Ouyang et al.
IY29CH04-Ouyang ARI 4 February 2011 15:27

LITERATURE CITED
1. Pestka S, Krause CD, Sarkar D, Walter MR, Shi Y, Fisher PB. 2004. Interleukin-10 and related cytokines
and receptors. Annu. Rev. Immunol. 22:929–79
2. Moore K, de Waal Malefyt R, Coffman R, O’Garra A. 2001. Interleukin-10 and the interleukin-10
receptor. Annu. Rev. Immunol. 19:683–765
3. Sa SM, Valdez PA, Wu J, Jung K, Zhong F, et al. 2007. The effects of IL-20 subfamily cytokines
on reconstituted human epidermis suggest potential roles in cutaneous innate defense and pathogenic
adaptive immunity in psoriasis. J. Immunol. 178:2229–40
4. Fox BA, Sheppard PO, O’Hara PJ. 2009. The role of genomic data in the discovery, annotation and
evolutionary interpretation of the interferon-lambda family. PLoS ONE 4:e4933
5. Uze G, Monneron D. 2007. IL-28 and IL-29: newcomers to the interferon family. Biochimie 89:729–34
6. Fiorentino DF, Bond MW, Mosmann TR. 1989. Two types of mouse T helper cell. IV. Th2 clones
secrete a factor that inhibits cytokine production by Th1 clones. J. Exp. Med. 170:2081–95
7. Moore KW, Vieira P, Fiorentino DF, Trounstine ML, Khan TA, Mosmann TR. 1990. Homology of
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

cytokine synthesis inhibitory factor (IL-10) to the Epstein-Barr virus gene BCRFI. Science 248:1230–34
8. Vieira P, de Waal-Malefyt R, Dang MN, Johnson KE, Kastelein R, et al. 1991. Isolation and expression of
human cytokine synthesis inhibitory factor cDNA clones: homology to Epstein-Barr virus open reading
by University of Guelph on 05/31/12. For personal use only.

frame BCRFI. Proc. Natl. Acad. Sci. USA 88:1172–76


9. Ho AS, Liu Y, Khan TA, Hsu DH, Bazan JF, Moore KW. 1993. A receptor for interleukin 10 is related
to interferon receptors. Proc. Natl. Acad. Sci. USA 90:11267–71
10. Liu Y, Wei SH, Ho AS, de Waal Malefyt R, Moore KW. 1994. Expression cloning and characterization
of a human IL-10 receptor. J. Immunol. 152:1821–29
11. Kotenko SV, Krause CD, Izotova LS, Pollack BP, Wu W, Pestka S. 1997. Identification and functional
characterization of a second chain of the interleukin-10 receptor complex. EMBO J. 16:5894–903
12. Lutfalla G, Gardiner K, Uze G. 1993. A new member of the cytokine receptor gene family maps on
chromosome 21 at less than 35 kb from IFNAR. Genomics 16:366–73
13. Spencer SD, Di Marco F, Hooley J, Pitts-Meek S, Bauer M, et al. 1998. The orphan receptor CRF2-4
is an essential subunit of the interleukin 10 receptor. J. Exp. Med. 187:571–78
14. Jones E, Flavell R. 2005. Distal enhancer elements transcribe intergenic RNA in the IL-10 family gene
cluster. J. Immunol. 175:7437–46
15. Jiang H, Lin JJ, Su ZZ, Goldstein NI, Fisher PB. 1995. Subtraction hybridization identifies a novel
melanoma differentiation associated gene, mda-7, modulated during human melanoma differentiation,
growth and progression. Oncogene 11:2477–86
16. Gallagher G, Dickensheets H, Eskdale J, Izotova LS, Mirochnitchenko OV, et al. 2000. Cloning, expres-
sion and initial characterization of interleukin-19 (IL-19), a novel homologue of human interleukin-10
(IL-10). Genes Immun. 1:442–50
17. Blumberg H, Conklin D, Xu WF, Grossmann A, Brender T, et al. 2001. Interleukin 20: discovery,
receptor identification, and role in epidermal function. Cell 104:9–19
18. Parrish-Novak J, Xu W, Brender T, Yao L, Jones C, et al. 2002. Interleukins 19, 20, and 24 signal
through two distinct receptor complexes. Differences in receptor-ligand interactions mediate unique
biological functions. J. Biol. Chem. 277:47517–23
19. Wang M, Tan Z, Zhang R, Kotenko SV, Liang P. 2002. Interleukin 24 (MDA-7/MOB-5) signals through
two heterodimeric receptors, IL-22R1/IL-20R2 and IL-20R1/IL-20R2. J. Biol. Chem. 277:7341–47
20. Dumoutier L, Leemans C, Lejeune D, Kotenko SV, Renauld JC. 2001. Cutting edge: STAT activation
by IL-19, IL-20 and mda-7 through IL-20 receptor complexes of two types. J. Immunol. 167:3545–49
21. Dumoutier L, Louahed J, Renauld JC. 2000. Cloning and characterization of IL-10-related T cell-
derived inducible factor (IL-TIF), a novel cytokine structurally related to IL-10 and inducible by IL-9.
J. Immunol. 164:1814–19
22. Knappe A, Hor S, Wittmann S, Fickenscher H. 2000. Induction of a novel cellular homolog of
interleukin-10, AK155, by transformation of T lymphocytes with herpesvirus saimiri. J. Virol. 74:3881–
87

www.annualreviews.org • IL-10 Family Cytokines in Immunity 95


IY29CH04-Ouyang ARI 4 February 2011 15:27

23. Xie MH, Aggarwal S, Ho WH, Foster J, Zhang Z, et al. 2000. Interleukin (IL)-22, a novel human
cytokine that signals through the interferon receptor-related proteins CRF2–4 and IL-22R. J. Biol.
Chem. 275:31335–39
24. Kotenko SV, Izotova LS, Mirochnitchenko OV, Esterova E, Dickensheets H, et al. 2001. Identification
of the functional interleukin-22 (IL-22) receptor complex: the IL-10R2 chain (IL-10Rβ) is a common
chain of both the IL-10 and IL-22 (IL-10-related T cell-derived inducible factor, IL-TIF) receptor
complexes. J. Biol. Chem. 276:2725–32
25. Sheikh F, Baurin VV, Lewis-Antes A, Shah NK, Smirnov SV, et al. 2004. Cutting edge: IL-26 signals
through a novel receptor complex composed of IL-20 receptor 1 and IL-10 receptor 2. J. Immunol.
172:2006–10
26. Kotenko SV, Izotova LS, Mirochnitchenko OV, Esterova E, Dickensheets H, et al. 2001. Identification,
cloning, and characterization of a novel soluble receptor that binds IL-22 and neutralizes its activity.
J. Immunol. 166:7096–103
27. Dumoutier L, Lejeune D, Hor S, Fickenscher H, Renauld JC. 2003. Cloning of a new type II cytokine
receptor activating signal transducer and activator of transcription (STAT)1, STAT2 and STAT3.
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

Biochem. J. 370:391–96
28. Xu W, Presnell SR, Parrish-Novak J, Kindsvogel W, Jaspers S, et al. 2001. A soluble class II cytokine
receptor, IL-22RA2, is a naturally occurring IL-22 antagonist. Proc. Natl. Acad. Sci. USA 98:9511–16
by University of Guelph on 05/31/12. For personal use only.

29. Dumoutier L, Van Roost E, Ameye G, Michaux L, Renauld JC. 2000. IL-TIF/IL-22: genomic organi-
zation and mapping of the human and mouse genes. Genes Immun. 1:488–94
30. Wang T, Dı́az-Rosales P, Martin SA, Secombes CJ. 2009. Cloning of a novel interleukin (IL)-20-like
gene in rainbow trout Oncorhynchus mykiss gives an insight into the evolution of the IL-10 family. Dev.
Comp. Immunol. 34:158–67
31. Kotenko SV, Gallagher G, Baurin VV, Lewis-Antes A, Shen M, et al. 2003. IFN-λs mediate antiviral
protection through a distinct class II cytokine receptor complex. Nat. Immunol. 4:69–77
32. Sheppard P, Kindsvogel W, Xu W, Henderson K, Schlutsmeyer S, et al. 2003. IL-28, IL-29 and their
class II cytokine receptor IL-28R. Nat. Immunol. 4:63–68
33. Onoguchi K, Yoneyama M, Takemura A, Akira S, Taniguchi T, et al. 2007. Viral infections activate
types I and III interferon genes through a common mechanism. J. Biol. Chem. 282:7576–81
34. Gad HH, Dellgren C, Hamming OJ, Vends S, Paludan SR, Hartmann R. 2009. Interferon-λ is func-
tionally an interferon but structurally related to the interleukin-10 family. J. Biol. Chem. 284:20869–75
35. Sommereyns C, Paul S, Staeheli P, Michiels T. 2008. IFN-lambda (IFN-λ) is expressed in a tissue-
dependent fashion and primarily acts on epithelial cells in vivo. PLoS Pathog. 4:e1000017
36. Witte K, Gruetz G, Volk HD, Looman AC, Asadullah K, et al. 2009. Despite IFN-λ receptor expres-
sion, blood immune cells, but not keratinocytes or melanocytes, have an impaired response to type III
interferons: implications for therapeutic applications of these cytokines. Genes Immun. 10:702–14
37. Krause CD, Mei E, Mirochnitchenko O, Lavnikova N, Xie J, et al. 2006. Interactions among the com-
ponents of the interleukin-10 receptor complex. Biochem. Biophys. Res. Commun. 340:377–85
38. Trivella DB, Ferreira-Junior JR, Dumoutier L, Renauld JC, Polikarpov I. 2010. Structure and function
of interleukin-22 and other members of the interleukin-10 family. Cell. Mol. Life Sci. 67:2909–35
39. Ealick SE, Cook WJ, Vijay-Kumar S, Carson M, Nagabhushan TL, et al. 1991. Three-dimensional
structure of recombinant human interferon-γ. Science 252:698–702
40. Walter MR, Nagabhushan TL. 1995. Crystal structure of interleukin 10 reveals an interferon γ-like
fold. Biochemistry 34:12118–25
41. Zdanov A, Schalk-Hihi C, Gustchina A, Tsang M, Weatherbee J, Wlodawer A. 1995. Crystal structure
of interleukin-10 reveals the functional dimer with an unexpected topological similarity to interferon γ.
Structure 3:591–601
42. Josephson K, DiGiacomo R, Indelicato SR, Iyo AH, Nagabhushan TL, et al. 2000. Design and analysis
of an engineered human interleukin-10 monomer. J. Biol. Chem. 275:13552–57
43. de Oliveira Neto M, Ferreira JR Jr, Colau D, Fischer H, Nascimento AS, et al. 2008. Interleukin-22
forms dimers that are recognized by two interleukin-22R1 receptor chains. Biophys. J. 94:1754–65
44. Nagem RA, Colau D, Dumoutier L, Renauld JC, Ogata C, Polikarpov I. 2002. Crystal structure of
recombinant human interleukin-22. Structure 10:1051–62

96 Ouyang et al.
IY29CH04-Ouyang ARI 4 February 2011 15:27

45. Jones BC, Logsdon NJ, Walter MR. 2008. Structure of IL-22 bound to its high-affinity IL-22R1 chain.
Structure 16:1333–44
46. Walter MR, Windsor WT, Nagabhushan TL, Lundell DJ, Lunn CA, et al. 1995. Crystal structure of a
complex between interferon-γ and its soluble high-affinity receptor. Nature 376:230–35
47. Josephson K, Logsdon NJ, Walter MR. 2001. Crystal structure of the IL-10/IL-10R1 complex reveals
a shared receptor binding site. Immunity 15:35–46
48. Bleicher L, de Moura PR, Watanabe L, Colau D, Dumoutier L, et al. 2008. Crystal structure of the
IL-22/IL-22R1 complex and its implications for the IL-22 signaling mechanism. FEBS Lett. 582:2985–92
49. de Moura PR, Watanabe L, Bleicher L, Colau D, Dumoutier L, et al. 2009. Crystal structure of a soluble
decoy receptor IL-22BP bound to interleukin-22. FEBS Lett. 583:1072–77
50. de Vos AM, Ultsch M, Kossiakoff AA. 1992. Human growth hormone and extracellular domain of its
receptor: crystal structure of the complex. Science 255:306–12
51. Yoon SI, Logsdon NJ, Sheikh F, Donnelly RP, Walter MR. 2006. Conformational changes mediate
interleukin-10 receptor 2 (IL-10R2) binding to IL-10 and assembly of the signaling complex. J. Biol.
Chem. 281:35088–96
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

52. Yoon SI, Jones BC, Logsdon NJ, Harris BD, Deshpande A, et al. 2010. Structure and mechanism of
receptor sharing by the IL-10R2 common chain. Structure 18:638–48
53. Wang X, Lupardus P, Laporte SL, Garcia KC. 2009. Structural biology of shared cytokine receptors.
by University of Guelph on 05/31/12. For personal use only.

Annu. Rev. Immunol. 27:29–60


54. Ho AS, Wei SH, Mui AL, Miyajima A, Moore KW. 1995. Functional regions of the mouse interleukin-10
receptor cytoplasmic domain. Mol. Cell. Biol. 15:5043–53
55. Finbloom DS, Winestock KD. 1995. IL-10 induces the tyrosine phosphorylation of tyk2 and Jak1 and
the differential assembly of STAT1 alpha and STAT3 complexes in human T cells and monocytes.
J. Immunol. 155:1079–90
56. Meraz MA, White JM, Sheehan KC, Bach EA, Rodig SJ, et al. 1996. Targeted disruption of the Stat1 gene
in mice reveals unexpected physiologic specificity in the JAK-STAT signaling pathway. Cell 84:431–42
57. Takeda K, Clausen BE, Kaisho T, Tsujimura T, Terada N, et al. 1999. Enhanced Th1 activity and
development of chronic enterocolitis in mice devoid of Stat3 in macrophages and neutrophils. Immunity
10:39–49
58. Lejeune D, Dumoutier L, Constantinescu S, Kruijer W, Schuringa JJ, Renauld JC. 2002. Interleukin-22
(IL-22) activates the JAK/STAT, ERK, JNK, and p38 MAP kinase pathways in a rat hepatoma cell line.
Pathways that are shared with and distinct from IL-10. J. Biol. Chem. 277:33676–82
59. Dumoutier L, de Meester C, Tavernier J, Renauld JC. 2009. New activation modus of STAT3: a tyrosine-
less region of the interleukin-22 receptor recruits STAT3 by interacting with its coiled-coil domain.
J. Biol. Chem. 284:26377–84
60. Brand S, Beigel F, Olszak T, Zitzmann K, Eichhorst ST, et al. 2006. IL-22 is increased in active Crohn’s
disease and promotes proinflammatory gene expression and intestinal epithelial cell migration. Am. J.
Physiol. Gastrointest. Liver Physiol. 290:G827–38
61. Zenewicz LA, Yancopoulos GD, Valenzuela DM, Murphy AJ, Karow M, Flavell RA. 2007. Interleukin-
22 but not interleukin-17 provides protection to hepatocytes during acute liver inflammation. Immunity
27:647–59
62. Weber GF, Gaertner FC, Erl W, Janssen KP, Blechert B, et al. 2006. IL-22-mediated tumor growth
reduction correlates with inhibition of ERK1/2 and AKT phosphorylation and induction of cell cycle
arrest in the G2-M phase. J. Immunol. 177:8266–72
63. O’Garra A, Vieira P. 2007. T(H)1 cells control themselves by producing interleukin-10. Nat. Rev. Im-
munol. 7:425–28
64. Trinchieri G. 2007. Interleukin-10 production by effector T cells: Th1 cells show self control. J. Exp.
Med. 204:239–43
65. Roncarolo MG, Gregori S, Battaglia M, Bacchetta R, Fleischhauer K, Levings MK. 2006. Interleukin-
10-secreting type 1 regulatory T cells in rodents and humans. Immunol. Rev. 212:28–50
66. Maynard C, Weaver C. 2008. Diversity in the contribution of interleukin-10 to T-cell-mediated immune
regulation. Immunol. Rev. 226:219–33

www.annualreviews.org • IL-10 Family Cytokines in Immunity 97


IY29CH04-Ouyang ARI 4 February 2011 15:27

67. Maloy K, Powrie F. 2001. Regulatory T cells in the control of immune pathology. Nat. Immunol. 2:816–22
68. Saraiva M, O’Garra A. 2010. The regulation of IL-10 production by immune cells. Nat. Rev. Immunol.
10:170–81
69. Lampropoulou V, Calderon-Gomez E, Roch T, Neves P, Shen P, et al. 2010. Suppressive functions
of activated B cells in autoimmune diseases reveal the dual roles of Toll-like receptors in immunity.
Immunol. Rev. 233:146–61
70. Ouyang W, Kolls JK, Zheng Y. 2008. The biological functions of T helper 17 cell effector cytokines in
inflammation. Immunity 28:454–67
71. Colonna M. 2009. Interleukin-22-producing natural killer cells and lymphoid tissue inducer-like cells in
mucosal immunity. Immunity 31:15–23
72. Martin B, Hirota K, Cua DJ, Stockinger B, Veldhoen M. 2009. Interleukin-17-producing γδ T cells
selectively expand in response to pathogen products and environmental signals. Immunity 31:321–30
73. Sutton CE, Lalor SJ, Sweeney CM, Brereton CF, Lavelle EC, Mills KH. 2009. Interleukin-1 and IL-
23 induce innate IL-17 production from γδ T cells, amplifying Th17 responses and autoimmunity.
Immunity 31:331–41
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

74. Takatori H, Kanno Y, Watford WT, Tato CM, Weiss G, et al. 2009. Lymphoid tissue inducer-like cells
are an innate source of IL-17 and IL-22. J. Exp. Med. 206:35–41
75. Cupedo T, Crellin NK, Papazian N, Rombouts EJ, Weijer K, et al. 2009. Human fetal lymphoid
by University of Guelph on 05/31/12. For personal use only.

tissue-inducer cells are interleukin 17-producing precursors to RORC+ CD127+ natural killer-like cells.
Nat. Immunol. 10:66–74
76. Wolk K, Kunz S, Asadullah K, Sabat R. 2002. Cutting edge: immune cells as sources and targets of the
IL-10 family members? J. Immunol. 168:5397–402
77. Cella M, Fuchs A, Vermi W, Facchetti F, Otero K, et al. 2009. A human natural killer cell subset provides
an innate source of IL-22 for mucosal immunity. Nature 457:722–25
78. Manel N, Unutmaz D, Littman DR. 2008. The differentiation of human T(H)-17 cells requires trans-
forming growth factor-β and induction of the nuclear receptor RORγt. Nat. Immunol. 9:641–49
79. Wilson NJ, Boniface K, Chan JR, McKenzie BS, Blumenschein WM, et al. 2007. Development, cytokine
profile and function of human interleukin 17-producing helper T cells. Nat. Immunol. 8:950–57
80. Hori S, Nomura T, Sakaguchi S. 2003. Control of regulatory T cell development by the transcription
factor Foxp3. Science 299:1057–61
81. Vieira P, Christensen J, Minaee S, O’Neill E, Barrat F, et al. 2004. IL-10-secreting regulatory T cells
do not express Foxp3 but have comparable regulatory function to naturally occurring CD4+ CD25+
regulatory T cells. J. Immunol. 172:5986–93
82. Maynard CL, Harrington LE, Janowski KM, Oliver JR, Zindl CL, et al. 2007. Regulatory T cells
expressing interleukin 10 develop from Foxp3+ and Foxp3− precursor cells in the absence of interleukin
10. Nat. Immunol. 8:931–41
83. Hawrylowicz CM, O’Garra A. 2005. Potential role of interleukin-10-secreting regulatory T cells in
allergy and asthma. Nat. Rev. Immunol. 5:271–83
84. Belkaid Y. 2007. Regulatory T cells and infection: a dangerous necessity. Nat. Rev. Immunol. 7:875–88
85. Josefowicz SZ, Rudensky A. 2009. Control of regulatory T cell lineage commitment and maintenance.
Immunity 30:616–25
86. Shevach EM. 2009. Mechanisms of Foxp3+ T regulatory cell-mediated suppression. Immunity 30:636–45
87. de la Rosa M, Rutz S, Dorninger H, Scheffold A. 2004. Interleukin-2 is essential for CD4+ CD25+
regulatory T cell function. Eur. J. Immunol. 34:2480–88
88. Barthlott T, Moncrieffe H, Veldhoen M, Atkins CJ, Christensen J, et al. 2005. CD25+ CD4+ T cells
compete with naive CD4+ T cells for IL-2 and exploit it for the induction of IL-10 production. Int.
Immunol. 17:279–88
89. O’Garra A, Vieira P. 2004. Regulatory T cells and mechanisms of immune system control. Nat. Med.
10:801–5
90. Shevach EM. 2006. From vanilla to 28 flavors: multiple varieties of T regulatory cells. Immunity 25:195–
201
91. Roncarolo M, Bacchetta R, Bordignon C, Narula S, Levings M. 2001. Type 1 T regulatory cells. Immunol.
Rev. 182:68–79

98 Ouyang et al.
IY29CH04-Ouyang ARI 4 February 2011 15:27

92. Barrat F, Cua D, Boonstra A, Richards D, Crain C, et al. 2002. In vitro generation of interleukin 10-
producing regulatory CD4+ T cells is induced by immunosuppressive drugs and inhibited by T helper
type 1 (Th1)- and Th2-inducing cytokines. J. Exp. Med. 195:603–16
93. Zhu J, Min B, Hu-Li J, Watson C, Grinberg A, et al. 2004. Conditional deletion of Gata3 shows its
essential function in T(H)1-T(H)2 responses. Nat. Immunol. 5:1157–65
94. Shoemaker J, Saraiva M, O’Garra A. 2006. GATA-3 directly remodels the IL-10 locus independently of
IL-4 in CD4+ T cells. J. Immunol. 176:3470–79
95. Chang H, Helbig C, Tykocinski L, Kreher S, Koeck J, et al. 2007. Expression of IL-10 in Th memory
lymphocytes is conditional on IL-12 or IL-4, unless the IL-10 gene is imprinted by GATA-3. Eur. J.
Immunol. 37:807–17
96. Stumhofer J, Silver J, Laurence A, Porrett P, Harris T, et al. 2007. Interleukins 27 and 6 induce STAT3-
mediated T cell production of interleukin 10. Nat. Immunol. 8:1363–71
97. Veldhoen M, Uyttenhove C, van Snick J, Helmby H, Westendorf A, et al. 2008. Transforming growth
factor-β “reprograms” the differentiation of T helper 2 cells and promotes an interleukin 9-producing
subset. Nat. Immunol. 9:1341–46
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

98. Dardalhon V, Awasthi A, Kwon H, Galileos G, Gao W, et al. 2008. IL-4 inhibits TGF-β-induced
Foxp3+ T cells and, together with TGF-β, generates IL-9+ IL-10+ Foxp3− effector T cells.
Nat. Immunol. 9:1347–55
by University of Guelph on 05/31/12. For personal use only.

99. Wong MT, Ye JJ, Alonso MN, Landrigan A, Cheung RK, et al. 2010. Regulation of human Th9
differentiation by type I interferons and IL-21. Immunol. Cell Biol. 88:624–31
100. Schaefer G, Venkataraman C, Schindler U. 2001. Cutting edge: FISP (IL-4-induced secreted protein),
a novel cytokine-like molecule secreted by Th2 cells. J. Immunol. 166:5859–63
101. Assenmacher M, Schmitz J, Radbruch A. 1994. Flow cytometric determination of cytokines in acti-
vated murine T helper lymphocytes: expression of interleukin-10 in interferon-γ and in interleukin-4-
expressing cells. Eur. J. Immunol. 24:1097–101
102. Del Prete G, De Carli M, Almerigogna F, Giudizi MG, Biagiotti R, Romagnani S. 1992. Human
IL-10 is produced by both type 1 helper (Th1) and type 2 helper (Th2) T cell clones and inhibits their
antigen-specific proliferation and cytokine production. J. Immunol. 150:353–60
103. Meyaard L, Hovenkamp E, Otto SA, Miedema F. 1996. IL-12-induced IL-10 production by human T
cells as a negative feedback for IL-12-induced immune responses. J. Immunol. 156:2776–82
104. Gerosa F, Paganin C, Peritt D, Paiola F, Scupoli M, et al. 1996. Interleukin-12 primes human CD4 and
CD8 T cell clones for high production of both interferon-γand interleukin-10. J. Exp. Med. 183:2559–69
105. Saraiva M, Christensen J, Veldhoen M, Murphy TL, Murphy KM, O’Garra A. 2009. Interleukin-10
production by Th1 cells requires interleukin-12-induced STAT4 transcription factor and ERK MAP
kinase activation by high antigen dose. Immunity 31:209–19
106. Fitzgerald D, Zhang G, El-Behi M, Fonseca-Kelly Z, Li H, et al. 2007. Suppression of autoimmune
inflammation of the central nervous system by interleukin 10 secreted by interleukin 27-stimulated
T cells. Nat. Immunol. 8:1372–79
107. Batten M, Kljavin NM, Li J, Walter MJ, de Sauvage FJ, Ghilardi N. 2008. Cutting edge: IL-27 is a
potent inducer of IL-10 but not FoxP3 in murine T cells. J. Immunol. 180:2752–56
108. Pot C, Jin H, Awasthi A, Liu SM, Lai C-Y, et al. 2009. Cutting edge: IL-27 induces the transcription
factor c-Maf, cytokine IL-21, and the costimulatory receptor ICOS that coordinately act together to
promote differentiation of IL-10-producing Tr1 cells. J. Immunol. 183:797–801
109. Anderson CF, Stumhofer JS, Hunter CA, Sacks D. 2009. IL-27 regulates IL-10 and IL-17 from CD4+
cells in nonhealing Leishmania major infection. J. Immunol. 183:4619–27
110. Murugaiyan G, Mittal A, Lopez-Diego R, Maier LM, Anderson DE, Weiner HL. 2009. IL-27 is a key
regulator of IL-10 and IL-17 production by human CD4+ T cells. J. Immunol. 183:2435–43
111. Awasthi A, Carrier Y, Peron J, Bettelli E, Kamanaka M, et al. 2007. A dominant function for interleukin
27 in generating interleukin 10-producing anti-inflammatory T cells. Nat. Immunol. 8:1380–89
112. McGeachy M, Bak-Jensen K, Chen Y, Tato C, Blumenschein W, et al. 2007. TGF-β and IL-6 drive the
production of IL-17 and IL-10 by T cells and restrain T(H)-17 cell-mediated pathology. Nat. Immunol.
8:1390–97

www.annualreviews.org • IL-10 Family Cytokines in Immunity 99


IY29CH04-Ouyang ARI 4 February 2011 15:27

113. Gurney AL. 2004. IL-22, a Th1 cytokine that targets the pancreas and select other peripheral tissues.
Int. Immunopharmacol. 4:669–77
114. Liang SC, Tan XY, Luxenberg DP, Karim R, Dunussi-Joannopoulos K, et al. 2006. Interleukin (IL)-22
and IL-17 are coexpressed by Th17 cells and cooperatively enhance expression of antimicrobial peptides.
J. Exp. Med. 203:2271–79
115. Chung Y, Yang X, Chang SH, Ma L, Tian Q, Dong C. 2006. Expression and regulation of IL-22 in the
IL-17-producing CD4+ T lymphocytes. Cell Res. 16:902–7
116. Zheng Y, Danilenko D, Valdez P, Kasman I, Eastham-Anderson J, et al. 2007. Interleukin-22, a T(H)17
cytokine, mediates IL-23-induced dermal inflammation and acanthosis. Nature 445:648–51
117. Veldhoen M, Hocking RJ, Atkins CJ, Locksley RM, Stockinger B. 2006. TGFβ in the context of an
inflammatory cytokine milieu supports de novo differentiation of IL-17-producing T cells. Immunity
24:179–89
118. Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, et al. 2006. Reciprocal developmental pathways for
the generation of pathogenic effector TH17 and regulatory T cells. Nature 441:235–38
119. Mangan PR, Harrington LE, O’Quinn DB, Helms WS, Bullard DC, et al. 2006. Transforming growth
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

factor-β induces development of the T(H)17 lineage. Nature 441:231–34


120. Duhen T, Geiger R, Jarrossay D, Lanzavecchia A, Sallusto F. 2009. Production of interleukin 22 but
not interleukin 17 by a subset of human skin-homing memory T cells. Nat. Immunol. 10:857–63
by University of Guelph on 05/31/12. For personal use only.

121. Zheng Y, Valdez PA, Danilenko DM, Hu Y, Sa SM, et al. 2008. Interleukin-22 mediates early host
defense against attaching and effacing bacterial pathogens. Nat. Med. 14:282–89
122. Nurieva R, Yang XO, Martinez G, Zhang Y, Panopoulos AD, et al. 2007. Essential autocrine regulation
by IL-21 in the generation of inflammatory T cells. Nature 448:480–83
123. Kreymborg K, Etzensperger R, Dumoutier L, Haak S, Rebollo A, et al. 2007. IL-22 is expressed by
Th17 cells in an IL-23-dependent fashion, but not required for the development of autoimmune en-
cephalomyelitis. J. Immunol. 179:8098–104
124. Aujla S, Chan Y, Zheng M, Fei M, Askew D, et al. 2008. IL-22 mediates mucosal host defense against
Gram-negative bacterial pneumonia. Nat. Med. 14:275–81
125. Siegemund S, Schütze N, Schulz S, Wolk K, Nasilowska K, et al. 2009. Differential IL-23 requirement
for IL-22 and IL-17A production during innate immunity against Salmonella enterica serovar Enteritidis.
Int. Immunol. 21:555–65
126. Mus AMC, Cornelissen F, Asmawidjaja PS, van Hamburg JP, Boon L, et al. 2010. Interleukin-23 pro-
motes Th17 differentiation by inhibiting T-bet and FoxP3 and is required for elevation of interleukin-22,
but not interleukin-21, in autoimmune experimental arthritis. Arthritis Rheum. 62:1043–50
127. Ziegler-Heitbrock L, Lötzerich M, Schaefer A, Werner T, Frankenberger M, Benkhart E. 2003. IFN-
α induces the human IL-10 gene by recruiting both IFN regulatory factor 1 and Stat3. J. Immunol.
171:285–90
128. Unterberger C, Staples KJ, Smallie T, Williams L, Foxwell B, et al. 2008. Role of STAT3 in
glucocorticoid-induced expression of the human IL-10 gene. Mol. Immunol. 45:3230–37
129. Xu J, Yang Y, Qiu G, Lal G, Wu Z, et al. 2009. c-Maf regulates IL-10 expression during Th17 polar-
ization. J. Immunol. 182:6226–36
130. Stumhofer JS, Laurence A, Wilson EH, Huang E, Tato CM, et al. 2006. Interleukin 27 negatively
regulates the development of interleukin 17-producing T helper cells during chronic inflammation of
the central nervous system. Nat. Immunol. 7:937–45
131. Mathur AN, Chang H-C, Zisoulis DG, Stritesky GL, Yu Q, et al. 2007. Stat3 and Stat4 direct develop-
ment of IL-17-secreting Th cells. J. Immunol. 178:4901–7
132. Yang XO, Pappu BP, Nurieva R, Akimzhanov A, Kang HS, et al. 2008. T helper 17 lineage differentiation
is programmed by orphan nuclear receptors RORα and RORγ. Immunity 28:29–39
133. Yang XO, Panopoulos AD, Nurieva R, Chang SH, Wang D, et al. 2007. STAT3 regulates cytokine-
mediated generation of inflammatory helper T cells. J. Biol. Chem. 282:9358–63
134. Rutz S, Janke M, Kassner N, Hohnstein T, Krueger M, Scheffold A. 2008. Notch regulates IL-10
production by T helper 1 cells. Proc. Natl. Acad. Sci. USA 105:3497–502
135. Kassner N, Krueger M, Yagita H, Dzionek A, Hutloff A, et al. 2010. Cutting edge: Plasmacytoid dendritic
cells induce IL-10 production in T cells via the Delta-like-4/Notch axis. J. Immunol. 184:550–54

100 Ouyang et al.


IY29CH04-Ouyang ARI 4 February 2011 15:27

136. Mukherjee S, Schaller MA, Neupane R, Kunkel SL, Lukacs NW. 2009. Regulation of T cell activation
by Notch ligand, DLL4, promotes IL-17 production and Rorc activation. J. Immunol. 182:7381–88
137. Alam MS, Maekawa Y, Kitamura A, Tanigaki K, Yoshimoto T, et al. 2010. Notch signaling drives IL-22
secretion in CD4+ T cells by stimulating the aryl hydrocarbon receptor. Proc. Natl. Acad. Sci. USA
107:5943–48
138. Kim JI, Ho IC, Grusby MJ, Glimcher LH. 1999. The transcription factor c-Maf controls the production
of interleukin-4 but not other Th2 cytokines. Immunity 10:745–51
139. Ho IC, Lo D, Glimcher LH. 1998. c-maf promotes T helper cell type 2 (Th2) and attenuates Th1
differentiation by both interleukin 4-dependent and -independent mechanisms. J. Exp. Med. 188:1859–
66
140. Brightbill HD, Plevy SE, Modlin RL, Smale ST. 2000. A prominent role for Sp1 during
lipopolysaccharide-mediated induction of the IL-10 promoter in macrophages. J. Immunol. 164:1940–51
141. Tone M, Powell MJ, Tone Y, Thompson SA, Waldmann H. 2000. IL-10 gene expression is controlled
by the transcription factors Sp1 and Sp3. J. Immunol. 165:286–91
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

142. Brenner S, Prösch S, Schenke-Layland K, Riese U, Gausmann U, Platzer C. 2003. cAMP-induced


interleukin-10 promoter activation depends on CCAAT/enhancer-binding protein expression and
monocytic differentiation. J. Biol. Chem. 278:5597–604
143. Liu Y-W, Tseng H-P, Chen L-C, Chen B-K, Chang W-C. 2003. Functional cooperation of simian virus
by University of Guelph on 05/31/12. For personal use only.

40 promoter factor 1 and CCAAT/enhancer-binding protein βand δin lipopolysaccharide-induced gene


activation of IL-10 in mouse macrophages. J. Immunol. 171:821–28
144. Csóka B, Németh ZH, Virág L, Gergely P, Leibovich SJ, et al. 2007. A2A adenosine receptors and
C/EBPβ are crucially required for IL-10 production by macrophages exposed to Escherichia coli. Blood
110:2685–95
145. Hu X, Paik PK, Chen J, Yarilina A, Kockeritz L, et al. 2006. IFN-γ suppresses IL-10 production and
synergizes with TLR2 by regulating GSK3 and CREB/AP-1 proteins. Immunity 24:563–74
146. Kremer KN, Kumar A, Hedin KE. 2007. Haplotype-independent costimulation of IL-10 secretion by
SDF-1/CXCL12 proceeds via AP-1 binding to the human IL-10 promoter. J. Immunol. 178:1581–88
147. Kanters E, Pasparakis M, Gijbels MJJ, Vergouwe MN, Partouns-Hendriks I, et al. 2003. Inhibition of
NF-κB activation in macrophages increases atherosclerosis in LDL receptor-deficient mice. J. Clin.
Investig. 112:1176–85
148. Platzer C, Fritsch E, Elsner T, Lehmann MH, Volk HD, Prosch S. 1999. Cyclic adenosine
monophosphate-responsive elements are involved in the transcriptional activation of the human IL-
10 gene in monocytic cells. Eur. J. Immunol. 29:3098–104
149. Wang Z, Sato H, Kusam S, Sehra S, Toney L, Dent A. 2005. Regulation of IL-10 gene expression in
Th2 cells by Jun proteins. J. Immunol. 174:2098–105
150. Yi AK, Yoon JG, Yeo SJ, Hong SC, English BK, Krieg AM. 2002. Role of mitogen-activated protein
kinases in CpG DNA-mediated IL-10 and IL-12 production: central role of extracellular signal-regulated
kinase in the negative feedback loop of the CpG DNA-mediated Th1 response. J. Immunol. 168:4711–20
151. Fuqua CF, Akomeah R, Price JO, Adunyah SE. 2008. Involvement of ERK-1/2 in IL-21-induced cytokine
production in leukemia cells and human monocytes. Cytokine 44:101–7
152. Owaki T, Asakawa M, Fukai F, Mizuguchi J, Yoshimoto T. 2006. IL-27 induces Th1 differentiation
via p38 MAPK/T-bet- and intercellular adhesion molecule-1/LFA-1/ERK1/2-dependent pathways.
J. Immunol. 177:7579–87
153. Ivanov II, McKenzie BS, Zhou L, Tadokoro CE, Lepelley A, et al. 2006. The orphan nuclear receptor
RORγt directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell 126:1121–33
154. Satoh-Takayama N, Vosshenrich CA, Lesjean-Pottier S, Sawa S, Lochner M, et al. 2008. Microbial
flora drives interleukin 22 production in intestinal NKp46+ cells that provide innate mucosal immune
defense. Immunity 29:958–70
155. Sanos SL, Bui VL, Mortha A, Oberle K, Heners C, et al. 2009. RORγt and commensal microflora
are required for the differentiation of mucosal interleukin 22-producing NKp46+ cells. Nat. Immunol.
10:83–91

www.annualreviews.org • IL-10 Family Cytokines in Immunity 101


IY29CH04-Ouyang ARI 4 February 2011 15:27

156. Eberl G, Marmon S, Sunshine M-J, Rennert PD, Choi Y, Littman DR. 2004. An essential function for
the nuclear receptor RORγ(t) in the generation of fetal lymphoid tissue inducer cells. Nat. Immunol.
5:64–73
157. Trifari S, Kaplan CD, Tran EH, Crellin NK, Spits H. 2009. Identification of a human helper T cell
population that has abundant production of interleukin 22 and is distinct from T(H)-17, T(H)1 and
T(H)2 cells. Nat. Immunol. 10:864–71
158. Crellin NK, Trifari S, Kaplan CD, Cupedo T, Spits H. 2010. Human NKp44+ IL-22+ cells and LTi-
like cells constitute a stable RORC+ lineage distinct from conventional natural killer cells. J. Exp. Med.
207:281–90
159. Luci C, Reynders A, Ivanov II, Cognet C, Chiche L, Chasson L, et al. 2009. Influence of the transcription
factor RORγt on the development of NKp46+ cell populations in gut and skin. Nat. Immunol. 10:75–82
160. Veldhoen M, Hirota K, Christensen J, O’Garra A, Stockinger B. 2009. Natural agonists for aryl hydro-
carbon receptor in culture medium are essential for optimal differentiation of Th17 T cells. J. Exp. Med.
206:43–49
161. Veldhoen M, Hirota K, Westendorf AM, Buer J, Dumoutier L, et al. 2008. The aryl hydrocarbon
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

receptor links TH17-cell-mediated autoimmunity to environmental toxins. Nature 453:106–9


162. Quintana FJ, Basso AS, Iglesias AH, Korn T, Farez MF, et al. 2008. Control of T(reg) and T(H)17 cell
differentiation by the aryl hydrocarbon receptor. Nature 453:65–71
by University of Guelph on 05/31/12. For personal use only.

163. Kimura A, Naka T, Nohara K, Fujii-Kuriyama Y, Kishimoto T. 2008. Aryl hydrocarbon receptor reg-
ulates Stat1 activation and participates in the development of Th17 cells. Proc. Natl. Acad. Sci. USA
105:9721–26
164. Schraml BU, Hildner K, Ise W, Lee WL, Smith WA, et al. 2009. The AP-1 transcription factor Batf
controls T(H)17 differentiation. Nature 460:405–9
165. Brüstle A, Heink S, Huber M, Rosenplänter C, Stadelmann C, et al. 2007. The development of inflam-
matory T(H)-17 cells requires interferon-regulatory factor 4. Nat. Immunol. 8:958–66
166. Huber M, Brüstle A, Reinhard K, Guralnik A, Walter G, et al. 2008. IRF4 is essential for IL-21-mediated
induction, amplification, and stabilization of the Th17 phenotype. Proc. Natl. Acad. Sci. USA 105:20846–
51
167. Okamoto K, Iwai Y, Oh-Hora M, Yamamoto M, Morio T, et al. 2010. IκBζ regulates T(H)17 develop-
ment by cooperating with ROR nuclear receptors. Nature 464:1381–85
168. Boonstra A, Rajsbaum R, Holman M, Marques R, Asselin-Paturel C, et al. 2006. Macrophages and
myeloid dendritic cells, but not plasmacytoid dendritic cells, produce IL-10 in response to MyD88- and
TRIF-dependent TLR signals, and TLR-independent signals. J. Immunol. 177:7551–58
169. Edwards AD, Manickasingham SP, Spörri R, Diebold SS, Schulz O, et al. 2002. Microbial recognition via
Toll-like receptor-dependent and -independent pathways determines the cytokine response of murine
dendritic cell subsets to CD40 triggering. J. Immunol. 169:3652–60
170. Agrawal S, Agrawal A, Doughty B, Gerwitz A, Blenis J, et al. 2003. Cutting edge: different Toll-like
receptor agonists instruct dendritic cells to induce distinct Th responses via differential modulation of
extracellular signal-regulated kinase-mitogen-activated protein kinase and c-Fos. J. Immunol. 171:4984–
89
171. Rogers NC, Slack EC, Edwards AD, Nolte MA, Schulz O, et al. 2005. Syk-dependent cytokine induction
by Dectin-1 reveals a novel pattern recognition pathway for C type lectins. Immunity 22:507–17
172. Dillon S, Agrawal A, Van Dyke T, Landreth G, McCauley L, et al. 2004. A Toll-like receptor 2 ligand
stimulates Th2 responses in vivo, via induction of extracellular signal-regulated kinase mitogen-activated
protein kinase and c-Fos in dendritic cells. J. Immunol. 172:4733–43
173. Geijtenbeek TBH, van Vliet SJ, Koppel EA, Sanchez-Hernandez M, Vandenbroucke-Grauls CMJE,
et al. 2003. Mycobacteria target DC-SIGN to suppress dendritic cell function. J. Exp. Med. 197:7–17
174. Fiorentino DF, Zlotnik A, Mosmann TR, Howard M, O’Garra A. 1991. IL-10 inhibits cytokine pro-
duction by activated macrophages. J. Immunol. 147:3815–22
175. Gerber JS, Mosser DM. 2001. Reversing lipopolysaccharide toxicity by ligating the macrophage Fcγ
receptors. J. Immunol. 166:6861–68
176. Chang EY, Guo B, Doyle SE, Cheng G. 2007. Cutting edge: involvement of the type I IFN production
and signaling pathway in lipopolysaccharide-induced IL-10 production. J. Immunol. 178:6705–9

102 Ouyang et al.


IY29CH04-Ouyang ARI 4 February 2011 15:27

177. Netea MG, Sutmuller R, Hermann C, Van der Graaf CAA, Van der Meer JWM, et al. 2004. Toll-like
receptor 2 suppresses immunity against Candida albicans through induction of IL-10 and regulatory T
cells. J. Immunol. 172:3712–18
178. Higgins SC, Lavelle EC, McCann C, Keogh B, McNeela E, et al. 2003. Toll-like receptor 4-mediated
innate IL-10 activates antigen-specific regulatory T cells and confers resistance to Bordetella pertussis by
inhibiting inflammatory pathology. J. Immunol. 171:3119–27
179. Fujita S, Seino K-i, Sato K, Sato Y, Eizumi K, et al. 2006. Regulatory dendritic cells act as regulators of
acute lethal systemic inflammatory response. Blood 107:3656–64
180. Wang F, Lee E, Lowes MA, Haider AS, Fuentes-Duculan J, et al. 2006. Prominent production of IL-20
by CD68+ /CD11c+ myeloid-derived cells in psoriasis: gene regulation and cellular effects. J. Investig.
Dermatol. 126:1590–99
181. Yano S, Banno T, Walsh R, Blumenberg M. 2008. Transcriptional responses of human epidermal
keratinocytes to cytokine interleukin-1. J. Cell. Physiol. 214:1–13
182. Hunt DW, Boivin WA, Fairley LA, Jovanovic MM, King DE, et al. 2006. Ultraviolet B light stimulates
interleukin-20 expression by human epithelial keratinocytes. Photochem. Photobiol. 82:1292–300
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

183. Wolk K, Witte E, Warszawska K, Schulze-Tanzil G, Witte K, et al. 2009. The Th17 cytokine IL-22
induces IL-20 production in keratinocytes: a novel immunological cascade with potential relevance in
psoriasis. Eur. J. Immunol. 39:3570–81
by University of Guelph on 05/31/12. For personal use only.

184. Huang F, Wachi S, Thai P, Loukoianov A, Tan KH, Forteza RM, Wu R. 2008. Potentiation of IL-19
expression in airway epithelia by IL-17A and IL-4/IL-13: important implications in asthma. J. Allergy
Clin. Immunol. 121:1415–21, 1421.e1–3
185. De Luca A, Zelante T, D’Angelo C, Zagarella S, Fallarino F, et al. 2010. IL-22 defines a novel immune
pathway of antifungal resistance. Mucosal Immunol. 3:361–73
186. Pickert G, Neufert C, Leppkes M, Zheng Y, Wittkopf N, et al. 2009. STAT3 links IL-22 signaling in
intestinal epithelial cells to mucosal wound healing. J. Exp. Med. 206:1465–72
187. Zenewicz LA, Yancopoulos GD, Valenzuela DM, Murphy AJ, Stevens S, Flavell RA. 2008. Innate and
adaptive interleukin-22 protects mice from inflammatory bowel disease. Immunity 29:947–57
188. Wolk K, Sabat R. 2006. Interleukin-22: a novel T- and NK-cell derived cytokine that regulates the
biology of tissue cells. Cytokine Growth Factor Rev. 17:367–80
189. Satoh-Takayama N, Vosshenrich CAJ, Lesjean-Pottier S, Sawa S, Lochner M, et al. 2008. Microbial
flora drives interleukin 22 production in intestinal NKp46+ cells that provide innate mucosal immune
defense. Immunity 29:958–70
190. Satoh-Takayama N, Dumoutier L, Lesjean-Pottier S, Ribeiro VS, Mandelboim O, et al. 2009. The
natural cytotoxicity receptor NKp46 is dispensable for IL-22-mediated innate intestinal immune defense
against Citrobacter rodentium. J. Immunol. 183:6579–87
191. Vivier E, Spits H, Cupedo T. 2009. Interleukin-22-producing innate immune cells: new players in
mucosal immunity and tissue repair? Nat. Rev. Immunol. 9:229–34
192. Veiga-Fernandes H, Kioussis D, Coles M. 2010. Natural killer receptors: the burden of a name. J. Exp.
Med. 207:269–72
193. Satoh-Takayama N, Lesjean-Pottier S, Vieira P, Sawa S, Eberl G, et al. 2010. IL-7 and IL-15 in-
dependently program the differentiation of intestinal CD3-NKp46+ cell subsets from Id2-dependent
precursors. J. Exp. Med. 207:273–80
194. Sawa S, Cherrier M, Lochner M, Satoh-Takayama N, Fehling HJ, et al. 2010. Lineage relationship
analysis of RORγt+ innate lymphoid cells. Science 330:665–69
195. Wolk K, Kunz S, Witte E, Friedrich M, Asadullah K, Sabat R. 2004. IL-22 increases the innate immunity
of tissues. Immunity 21:241–54
196. Doyle SE, Schreckhise H, Khuu-Duong K, Henderson K, Rosler R, et al. 2006. Interleukin-29 uses a type
1 interferon-like program to promote antiviral responses in human hepatocytes. Hepatology 44:896–906
197. Marcello T, Grakoui A, Barba-Spaeth G, Machlin ES, Kotenko SV, et al. 2006. Interferons α and
λ inhibit hepatitis C virus replication with distinct signal transduction and gene regulation kinetics.
Gastroenterology 131:1887–98
198. Meager A, Visvalingam K, Dilger P, Bryan D, Wadhwa M. 2005. Biological activity of interleukins-28
and -29: comparison with type I interferons. Cytokine 31:109–18

www.annualreviews.org • IL-10 Family Cytokines in Immunity 103


IY29CH04-Ouyang ARI 4 February 2011 15:27

199. Boniface K, Bernard FX, Garcia M, Gurney AL, Lecron JC, Morel F. 2005. IL-22 inhibits epidermal
differentiation and induces proinflammatory gene expression and migration of human keratinocytes.
J. Immunol. 174:3695–702
200. Aujla SJ, Chan YR, Zheng M, Fei M, Askew DJ, et al. 2008. IL-22 mediates mucosal host defense against
Gram-negative bacterial pneumonia. Nat. Med. 14:275–81
201. Liao SC, Cheng YC, Wang YC, Wang CW, Yang SM, et al. 2004. IL-19 induced Th2 cytokines and
was up-regulated in asthma patients. J. Immunol. 173:6712–18
202. Liao YC, Liang WG, Chen FW, Hsu JH, Yang JJ, Chang MS. 2002. IL-19 induces production of IL-6
and TNF-α and results in cell apoptosis through TNF-α. J. Immunol. 169:4288–97
203. Liu L, Ding C, Zeng W, Heuer JG, Tetreault JW, et al. 2003. Selective enhancement of multipotential
hematopoietic progenitors in vitro and in vivo by IL-20. Blood 102:3206–9
204. Wahl C, Müller W, Leithäuser F, Adler G, Oswald F, et al. 2009. IL-20 receptor 2 signaling down-
regulates antigen-specific T cell responses. J. Immunol. 182:802–10
205. Emdad L, Lebedeva IV, Su ZZ, Gupta P, Sauane M, et al. 2009. Historical perspective and recent
insights into our understanding of the molecular and biochemical basis of the antitumor properties of
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

mda-7/IL-24. Cancer Biol. Ther. 8:391–400


206. Kreis S, Philippidou D, Margue C, Behrmann I. 2008. IL-24: a classic cytokine and/or a potential cure
for cancer? J. Cell. Mol. Med. 12:2505–10
by University of Guelph on 05/31/12. For personal use only.

207. Kreis S, Philippidou D, Margue C, Rolvering C, Haan C, et al. 2007. Recombinant interleukin-24 lacks
apoptosis-inducing properties in melanoma cells. PLoS ONE 2:e1300
208. Wolk K, Witte E, Wallace E, Docke WD, Kunz S, et al. 2006. IL-22 regulates the expression of genes
responsible for antimicrobial defense, cellular differentiation, and mobility in keratinocytes: a potential
role in psoriasis. Eur. J. Immunol. 36:1309–23
209. Nagalakshmi ML, Rascle A, Zurawski S, Menon S, de Waal Malefyt R. 2004. Interleukin-22 activates
STAT3 and induces IL-10 by colon epithelial cells. Int. Immunopharmacol. 4:679–91
210. El Kasmi KC, Holst J, Coffre M, Mielke L, de Pauw A, et al. 2006. General nature of the STAT3-activated
anti-inflammatory response. J. Immunol. 177:7880–88
211. Savan R, McFarland AP, Reynolds DA, Feigenbaum L, Ramakrishnan K, et al. 2010. A novel role for
IL-22R1 as a driver of inflammation. Blood. In press
212. Mege JL, Meghari S, Honstettre A, Capo C, Raoult D. 2006. The two faces of interleukin 10 in human
infectious diseases. Lancet Infect. Dis. 6:557–69
213. Filippi CM, von Herrath MG. 2008. IL-10 and the resolution of infections. J. Pathol. 214:224–30
214. Ejrnaes M, Filippi CM, Martinic MM, Ling EM, Togher LM, et al. 2006. Resolution of a chronic viral
infection after interleukin-10 receptor blockade. J. Exp. Med. 203:2461–72
215. van der Kleij D, Latz E, Brouwers JFHM, Kruize YCM, Schmitz M, et al. 2002. A novel host-parasite
lipid cross-talk. Schistosomal lyso-phosphatidylserine activates Toll-like receptor 2 and affects immune
polarization. J. Biol. Chem. 277:48122–19
216. Jang S, Uematsu S, Akira S, Salgame P. 2004. IL-6 and IL-10 induction from dendritic cells in response
to Mycobacterium tuberculosis is predominantly dependent on TLR2-mediated recognition. J. Immunol.
173:3392–97
217. Sing A, Rost D, Tvardovskaia N, Roggenkamp A, Wiedemann A, et al. 2002. Yersinia V-antigen exploits
Toll-like receptor 2 and CD14 for interleukin 10-mediated immunosuppression. J. Exp. Med. 196:1017–
24
218. Gabrysová L, Nicolson K, Streeter HB, Verhagen J, Sabatos-Peyton CA, et al. 2009. Negative feedback
control of the autoimmune response through antigen-induced differentiation of IL-10-secreting Th1
cells. J. Exp. Med. 206:175–67
219. Jankovic D, Kullberg M, Feng C, Goldszmid R, Collazo C, et al. 2007. Conventional T-bet+ Foxp3− Th1
cells are the major source of host-protective regulatory IL-10 during intracellular protozoan infection.
J. Exp. Med. 204:273–83
220. Anderson C, Oukka M, Kuchroo V, Sacks D. 2007. CD4+ CD25− Foxp3− Th1 cells are the source of
IL-10-mediated immune suppression in chronic cutaneous leishmaniasis. J. Exp. Med. 204:285–97
221. Wu B, Huang C, Kato-Maeda M, Hopewell PC, Daley CL, et al. 2008. IL-24 modulates IFN-γ expres-
sion in patients with tuberculosis. Immunol. Lett. 117:57–62

104 Ouyang et al.


IY29CH04-Ouyang ARI 4 February 2011 15:27

222. Ma Y, Chen H, Wang Q, Luo F, Yan J, Zhang XL. 2009. IL-24 protects against Salmonella typhimurium
infection by stimulating early neutrophil Th1 cytokine production, which in turn activates CD8+ T
cells. Eur. J. Immunol. 39:3357–68
223. Misse D, Yssel H, Trabattoni D, Oblet C, Lo Caputo S, et al. 2007. IL-22 participates in an innate
anti-HIV-1 host-resistance network through acute-phase protein induction. J. Immunol. 178:407–15
224. Levillayer F, Mas M, Levi-Acobas F, Brahic M, Bureau JF. 2007. Interleukin 22 is a candidate gene for
Tmevp3, a locus controlling Theiler’s virus-induced neurological diseases. Genetics 176:1835–44
225. Pagliaccetti NE, Eduardo R, Kleinstein SH, Mu XJ, Bandi P, Robek MD. 2008. Interleukin-29 functions
cooperatively with interferon to induce antiviral gene expression and inhibit hepatitis C virus replication.
J. Biol. Chem. 283:30079–89
226. Schulz SM, Kohler G, Schutze N, Knauer J, Straubinger RK, et al. 2008. Protective immunity to systemic
infection with attenuated Salmonella enterica serovar enteritidis in the absence of IL-12 is associated with
IL-23-dependent IL-22, but not IL-17. J. Immunol. 181:7891–901
227. Scriba TJ, Kalsdorf B, Abrahams DA, Isaacs F, Hofmeister J, et al. 2008. Distinct, specific IL-17- and
IL-22-producing CD4+ T cell subsets contribute to the human antimycobacterial immune response.
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

J. Immunol. 180:1962–70
228. Pitta MG, Romano A, Cabantous S, Henri S, Hammad A, et al. 2009. IL-17 and IL-22 are associated
with protection against human kala azar caused by Leishmania donovani. J. Clin. Investig. 119:2379–87
by University of Guelph on 05/31/12. For personal use only.

229. Dhiman R, Indramohan M, Barnes PF, Nayak RC, Paidipally P, et al. 2009. IL-22 produced by human
NK cells inhibits growth of Mycobacterium tuberculosis by enhancing phagolysosomal fusion. J. Immunol.
183:6639–45
230. Sellge G, Magalhaes JG, Konradt C, Fritz JH, Salgado-Pabon W, et al. 2010. Th17 cells are the dominant
T cell subtype primed by Shigella flexneri mediating protective immunity. J. Immunol. 184:2076–85
231. Wilson MS, Feng CG, Barber DL, Yarovinsky F, Cheever AW, et al. 2010. Redundant and pathogenic
roles for IL-22 in mycobacterial, protozoan, and helminth infections. J. Immunol. 184:4378–90
232. Muñoz M, Heimesaat MM, Danker K, Struck D, Lohmann U, et al. 2009. Interleukin (IL)-23 mediates
Toxoplasma gondii-induced immunopathology in the gut via matrixmetalloproteinase-2 and IL-22 but
independent of IL-17. J. Exp. Med. 206:3047–59
233. Weber GF, Schlautkotter S, Kaiser-Moore S, Altmayr F, Holzmann B, Weighardt H. 2007. Inhibition
of interleukin-22 attenuates bacterial load and organ failure during acute polymicrobial sepsis. Infect.
Immun. 75:1690–97
234. Ashman RB, Fulurija A, Papadimitriou JM. 1999. Both CD4+ and CD8+ lymphocytes reduce the
severity of tissue lesions in murine systemic candidiasis, and CD4+ cells also demonstrate strain-specific
immunopathological effects. Microbiology 145(Pt. 7):1631–40
235. Tavares D, Ferreira P, Arala-Chaves M. 2000. Increased resistance to systemic candidiasis in athymic or
interleukin-10-depleted mice. J. Infect. Dis. 182:266–73
236. Acosta-Rodriguez EV, Rivino L, Geginat J, Jarrossay D, Gattorno M, et al. 2007. Surface phenotype and
antigenic specificity of human interleukin 17-producing T helper memory cells. Nat. Immunol. 8:639–46
237. Huang W, Na L, Fidel PL, Schwarzenberger P. 2004. Requirement of interleukin-17A for systemic
anti-Candida albicans host defense in mice. J. Infect. Dis. 190:624–31
238. Liu Y, Yang B, Zhou M, Li L, Zhou H, et al. 2009. Memory IL-22-producing CD4+ T cells specific for
Candida albicans are present in humans. Eur. J. Immunol. 39:1472–79
239. Eyerich K, Foerster S, Rombold S, Seidl HP, Behrendt H, et al. 2008. Patients with chronic mucocuta-
neous candidiasis exhibit reduced production of Th17-associated cytokines IL-17 and IL-22. J. Investig.
Dermatol. 128:2640–45
240. Kisand K, Boe Wolff AS, Podkrajsek KT, Tserel L, Link M, et al. 2010. Chronic mucocutaneous can-
didiasis in APECED or thymoma patients correlates with autoimmunity to Th17-associated cytokines.
J. Exp. Med. 207:299–308
241. Puel A, Döffinger R, Natividad A, Chrabieh M, Barcenas-Morales G, et al. 2010. Autoantibodies against
IL-17A, IL-17F, and IL-22 in patients with chronic mucocutaneous candidiasis and autoimmune polyen-
docrine syndrome type I. J. Exp. Med. 207:291–97
242. Conti HR, Shen F, Nayyar N, Stocum E, Sun JN, et al. 2009. Th17 cells and IL-17 receptor signaling
are essential for mucosal host defense against oral candidiasis. J. Exp. Med. 206:299–311

www.annualreviews.org • IL-10 Family Cytokines in Immunity 105


IY29CH04-Ouyang ARI 4 February 2011 15:27

243. Miller DM, Klucher KM, Freeman JA, Hausman DF, Fontana D, Williams DE. 2009. Interferon lambda
as a potential new therapeutic for hepatitis C. Ann. N. Y. Acad. Sci. 1182:80–87
244. Ank N, Iversen MB, Bartholdy C, Staeheli P, Hartmann R, et al. 2008. An important role for type III
interferon (IFN-λ/IL-28) in TLR-induced antiviral activity. J. Immunol. 180:2474–85
245. Mordstein M, Kochs G, Dumoutier L, Renauld JC, Paludan SR, et al. 2008. Interferon-λ contributes
to innate immunity of mice against influenza A virus but not against hepatotropic viruses. PLoS Pathog.
4:e1000151
246. Ge D, Fellay J, Thompson AJ, Simon JS, Shianna KV, et al. 2009. Genetic variation in IL28B predicts
hepatitis C treatment-induced viral clearance. Nature 461:399–401
247. Thomas DL, Thio CL, Martin MP, Qi Y, Ge D, et al. 2009. Genetic variation in IL28B and spontaneous
clearance of hepatitis C virus. Nature 461:798–801
248. Suppiah V, Moldovan M, Ahlenstiel G, Berg T, Weltman M, et al. 2009. IL28B is associated with
response to chronic hepatitis C interferon-α and ribavirin therapy. Nat. Genet. 41:1100–4
249. Tanaka Y, Nishida N, Sugiyama M, Kurosaki M, Matsuura K, et al. 2009. Genome-wide association of
IL28B with response to pegylated interferon-α and ribavirin therapy for chronic hepatitis C. Nat. Genet.
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

41:1105–9
250. Asadullah K, Sterry W, Volk HD. 2003. Interleukin-10 therapy—review of a new approach. Pharmacol.
Rev. 55:241–69
by University of Guelph on 05/31/12. For personal use only.

251. Xavier RJ, Podolsky DK. 2007. Unravelling the pathogenesis of inflammatory bowel disease. Nature
448:427–34
252. Hooper LV, Macpherson AJ. 2010. Immune adaptations that maintain homeostasis with the intestinal
microbiota. Nat. Rev. Immunol. 10:159–69
253. Artis D. 2008. Epithelial-cell recognition of commensal bacteria and maintenance of immune homeostasis
in the gut. Nat. Rev. Immunol. 8:411–20
254. Braat H, Peppelenbosch MP, Hommes DW. 2003. Interleukin-10-based therapy for inflammatory bowel
disease. Expert. Opin. Biol. Ther. 3:725–31
255. Ouyang W, Valdez P. 2008. IL-22 in mucosal immunity. Mucosal Immunol. 1:335–38
256. Kuhn R, Lohler J, Rennick D, Rajewsky K, Muller W. 1993. Interleukin-10-deficient mice develop
chronic enterocolitis. Cell 75:263–74
257. Sellon RK, Tonkonogy S, Schultz M, Dieleman LA, Grenther W, et al. 1998. Resident enteric bacteria
are necessary for development of spontaneous colitis and immune system activation in interleukin-10-
deficient mice. Infect. Immun. 66:5224–31
258. Madsen KL, Doyle JS, Tavernini MM, Jewell LD, Rennie RP, Fedorak RN. 2000. Antibiotic therapy
attenuates colitis in interleukin 10 gene-deficient mice. Gastroenterology 118:1094–105
259. Davidson NJ, Leach MW, Fort MM, Thompson-Snipes L, Kuhn R, et al. 1996. T helper cell 1-type
CD4+ T cells, but not B cells, mediate colitis in interleukin 10-deficient mice. J. Exp. Med. 184:241–51
260. Aste-Amezaga M, Ma X, Sartori A, Trinchieri G. 1998. Molecular mechanisms of the induction of IL-12
and its inhibition by IL-10. J. Immunol. 160:5936–44
261. Oppmann B, Lesley R, Blom B, Timans JC, Xu Y, et al. 2000. Novel p19 protein engages IL-12p40
to form a cytokine, IL-23, with biological activities similar as well as distinct from IL-12. Immunity
13:715–25
262. Yen D, Cheung J, Scheerens H, Poulet F, McClanahan T, et al. 2006. IL-23 is essential for T cell-
mediated colitis and promotes inflammation via IL-17 and IL-6. J. Clin. Investig. 116:1310–16
263. Franke A, Balschun T, Karlsen TH, Sventoraityte J, Nikolaus S, et al. 2008. Sequence variants in IL10,
ARPC2 and multiple other loci contribute to ulcerative colitis susceptibility. Nat. Genet. 40:1319–23
264. Mitsuyama K, Tomiyasu N, Takaki K, Masuda J, Yamasaki H, et al. 2006. Interleukin-10 in the patho-
physiology of inflammatory bowel disease: increased serum concentrations during the recovery phase.
Mediat. Inflamm. 2006:26875
265. Tomoyose M, Mitsuyama K, Ishida H, Toyonaga A, Tanikawa K. 1998. Role of interleukin-10 in a
murine model of dextran sulfate sodium-induced colitis. Scand. J. Gastroenterol. 33:435–40
266. Duchmann R, Schmitt E, Knolle P, Meyer zum Büschenfelde KH, Neurath M. 1996. Tolerance towards
resident intestinal flora in mice is abrogated in experimental colitis and restored by treatment with
interleukin-10 or antibodies to interleukin-12. Eur. J. Immunol. 26:934–38

106 Ouyang et al.


IY29CH04-Ouyang ARI 4 February 2011 15:27

267. Tilg H, van Montfrans C, Van Den Ende A, Kaser A, van Deventer SJ, et al. 2002. Treatment of Crohn’s
disease with recombinant human interleukin 10 induces the proinflammatory cytokine interferon γ. Gut
50:191–95
268. Andoh A, Zhang Z, Inatomi O, Fujino S, Deguchi Y, et al. 2005. Interleukin-22, a member of the
IL-10 subfamily, induces inflammatory responses in colonic subepithelial myofibroblasts. Gastroenterology
129:969–84
269. Sekikawa A, Fukui H, Suzuki K, Karibe T, Fujii S, et al. 2010. Involvement of the IL-22/REG Iα axis
in ulcerative colitis. Lab. Investig. 90:496–505
270. Wolk K, Witte E, Hoffmann U, Doecke WD, Endesfelder S, et al. 2007. IL-22 induces
lipopolysaccharide-binding protein in hepatocytes: a potential systemic role of IL-22 in Crohn’s dis-
ease. J. Immunol. 178:5973–81
271. Kleinschek MA, Boniface K, Sadekova S, Grein J, Murphy EE, et al. 2009. Circulating and gut-resident
human Th17 cells express CD161 and promote intestinal inflammation. J. Exp. Med. 206:525–34
272. Schmechel S, Konrad A, Diegelmann J, Glas J, Wetzke M, et al. 2008. Linking genetic susceptibility
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

to Crohn’s disease with Th17 cell function: IL-22 serum levels are increased in Crohn’s disease and
correlate with disease activity and IL23R genotype status. Inflamm. Bowel Dis. 14:204–12
273. Sugimoto K, Ogawa A, Mizoguchi E, Shimomura Y, Andoh A, et al. 2008. IL-22 ameliorates intestinal
inflammation in a mouse model of ulcerative colitis. J. Clin. Investig. 118:534–44
by University of Guelph on 05/31/12. For personal use only.

274. Neufert C, Pickert G, Zheng Y, Wittkopf N, Warntjen M, et al. 2010. Activation of epithelial STAT3
regulates intestinal homeostasis. Cell Cycle 9:652–55
275. Dambacher J, Beigel F, Zitzmann K, de Toni E, Göoke B, et al. 2008. The role of the novel Th17
cytokine IL-26 in intestinal inflammation. Gut 58:1207–17
276. Azuma YT, Matsuo Y, Kuwamura M, Yancopoulos GD, Valenzuela DM, et al. 2010. Interleukin-19
protects mice from innate-mediated colonic inflammation. Inflamm. Bowel Dis. 16:1017–28
277. Brand S, Beigel F, Olszak T, Zitzmann K, Eichhorst ST, et al. 2005. IL-28A and IL-29 mediate antipro-
liferative and antiviral signals in intestinal epithelial cells and murine CMV infection increases colonic
IL-28A expression. Am. J. Physiol. Gastrointest. Liver Physiol. 289:G960–68
278. Louis H, Le Moine O, Peny MO, Quertinmont E, Fokan D, et al. 1997. Production and role of
interleukin-10 in concanavalin A-induced hepatitis in mice. Hepatology 25:1382–89
279. Di Marco R, Xiang M, Zaccone P, Leonardi C, Franco S, et al. 1999. Concanavalin A-induced hepatitis in
mice is prevented by interleukin (IL)-10 and exacerbated by endogenous IL-10 deficiency. Autoimmunity
31:75–83
280. Radaeva S, Sun R, Pan HN, Hong F, Gao B. 2004. Interleukin 22 (IL-22) plays a protective role in T
cell-mediated murine hepatitis: IL-22 is a survival factor for hepatocytes via STAT3 activation. Hepatology
39:1332–42
281. Wahl C, Wegenka UM, Leithauser F, Schirmbeck R, Reimann J. 2009. IL-22-dependent attenuation of
T cell-dependent (ConA) hepatitis in herpes virus entry mediator deficiency. J. Immunol. 182:4521–28
282. Pan H, Hong F, Radaeva S, Gao B. 2004. Hydrodynamic gene delivery of interleukin-22 protects the
mouse liver from concanavalin A-, carbon tetrachloride-, and Fas ligand-induced injury via activation of
STAT3. Cell. Mol. Immunol. 1:43–49
283. Ren X, Hu B, Colletti LM. 2009. IL-22 is involved in liver regeneration after hepatectomy. Am. J.
Physiol. Gastrointest. Liver Physiol. 298:G74–80
284. Yang L, Zhang Y, Wang L, Fan F, Zhu L, et al. 2010. Amelioration of high fat diet induced liver
lipogenesis and hepatic steatosis by interleukin-22. J. Hepatol. 53(2):339–47
285. Wegenka UM, Dikopoulos N, Reimann J, Adler G, Wahl C. 2007. The murine liver is a potential
target organ for IL-19, IL-20 and IL-24: type I interferons and LPS regulate the expression of IL-20R2.
J. Hepatol. 46:257–65
286. Lowes MA, Bowcock AM, Krueger JG. 2007. Pathogenesis and therapy of psoriasis. Nature 445:866–73
287. Rømer J, Hasselager E, Nørby PL, Steiniche T, Thorn Clausen J, Kragballe K. 2003. Epidermal over-
expression of interleukin-19 and -20 mRNA in psoriatic skin disappears after short-term treatment with
cyclosporine a or calcipotriol. J. Investig. Dermatol. 121:1306–11

www.annualreviews.org • IL-10 Family Cytokines in Immunity 107


IY29CH04-Ouyang ARI 4 February 2011 15:27

288. Otkjaer K, Kragballe K, Funding AT, Clausen JT, Noerby PL, et al. 2005. The dynamics of gene
expression of interleukin-19 and interleukin-20 and their receptors in psoriasis. Br. J. Dermatol. 153:911–
18
289. Tohyama M, Hanakawa Y, Shirakata Y, Dai X, Yang L, et al. 2009. IL-17 and IL-22 mediate IL-20
subfamily cytokine production in cultured keratinocytes via increased IL-22 receptor expression. Eur. J.
Immunol. 39:2779–88
290. Boniface K, Guignouard E, Pedretti N, Garcia M, Delwail A, et al. 2007. A role for T cell-derived
interleukin 22 in psoriatic skin inflammation. Clin. Exp. Immunol. 150:407–15
291. Pene J, Chevalier S, Preisser L, Venereau E, Guilleux MH, et al. 2008. Chronically inflamed human
tissues are infiltrated by highly differentiated Th17 lymphocytes. J. Immunol. 180:7423–30
292. Kagami S, Rizzo HL, Lee JJ, Koguchi Y, Blauvelt A. 2009. Circulating Th17, Th22, and Th1 cells are
increased in psoriasis. J. Investig. Dermatol. 130:1373–83
293. Kunz S, Wolk K, Witte E, Witte K, Doecke WD, et al. 2006. Interleukin (IL)-19, IL-20 and IL-24 are
produced by and act on keratinocytes and are distinct from classical ILs. Exp. Dermatol. 15:991–1004
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

294. Li W, Danilenko DM, Bunting S, Ganesan R, Sa S, et al. 2009. The serine protease marapsin is expressed
in stratified squamous epithelia and is up-regulated in the hyperproliferative epidermis of psoriasis and
regenerating wounds. J. Biol. Chem. 284:218–28
295. Guilloteau K, Paris I, Pedretti N, Boniface K, Juchaux F, et al. 2010. Skin inflammation induced by the
by University of Guelph on 05/31/12. For personal use only.

synergistic action of IL-17A, IL-22, oncostatin M, IL-1α, and TNF-α recapitulates some features of
psoriasis. J. Immunol. 184:5263–70
296. Gudjonsson JE, Ding J, Johnston A, Tejasvi T, Guzman AM, et al. 2010. Assessment of the psori-
atic transcriptome in a large sample: additional regulated genes and comparisons with in vitro models.
J. Investig. Dermatol. 130:1829–40
297. He M, Liang P. 2010. IL-24 transgenic mice: in vivo evidence of overlapping functions for IL-20, IL-22,
and IL-24 in the epidermis. J. Immunol. 184:1793–98
298. Wolk K, Haugen HS, Xu W, Witte E, Waggie K, et al. 2009. IL-22 and IL-20 are key mediators of the
epidermal alterations in psoriasis while IL-17 and IFN-γ are not. J. Mol. Med. 87:523–36
299. Kingo K, Koks S, Nikopensius T, Silm H, Vasar E. 2004. Polymorphisms in the interleukin-20 gene:
relationships to plaque-type psoriasis. Genes Immun. 5:117–21
300. Koks S, Kingo K, Ratsep R, Karelson M, Silm H, Vasar E. 2004. Combined haplotype analysis of the
interleukin-19 and -20 genes: relationship to plaque-type psoriasis. Genes Immun. 5:662–67
301. Koks S, Kingo K, Vabrit K, Ratsep R, Karelson M, et al. 2005. Possible relations between the polymor-
phisms of the cytokines IL-19, IL-20 and IL-24 and plaque-type psoriasis. Genes Immun. 6:407–15
302. Capon F, Di Meglio P, Szaub J, Prescott NJ, Dunster C, et al. 2007. Sequence variants in the genes
for the interleukin-23 receptor (IL23R) and its ligand (IL12B) confer protection against psoriasis. Hum.
Genet. 122:201–6
303. Kopp T, Lenz P, Bello-Fernandez C, Kastelein RA, Kupper TS, Stingl G. 2003. IL-23 production
by cosecretion of endogenous p19 and transgenic p40 in keratin 14/p40 transgenic mice: evidence for
enhanced cutaneous immunity. J. Immunol. 170:5438–44
304. Chan JR, Blumenschein W, Murphy E, Diveu C, Wiekowski M, et al. 2006. IL-23 stimulates epidermal
hyperplasia via TNF and IL-20R2-dependent mechanisms with implications for psoriasis pathogenesis.
J. Exp. Med. 203:2577–87
305. Ma HL, Liang S, Li J, Napierata L, Brown T, et al. 2008. IL-22 is required for Th17 cell-mediated
pathology in a mouse model of psoriasis-like skin inflammation. J. Clin. Investig. 118:597–607
306. Stenderup K, Rosada C, Worsaae A, Dagnaes-Hansen F, Steiniche T, et al. 2009. Interleukin-20 plays a
critical role in maintenance and development of psoriasis in the human xenograft transplantation model.
Br. J. Dermatol. 160:284–96
307. Krueger GG, Langley RG, Leonardi C, Yeilding N, Guzzo C, et al. 2007. A human interleukin-12/23
monoclonal antibody for the treatment of psoriasis. N. Engl. J. Med. 356(6):580–92
308. Alanärä T, Karstila K, Moilanen T, Silvennoinen O, Isomäki P. 2010. Expression of IL-10 family cy-
tokines in rheumatoid arthritis: elevated levels of IL-19 in the joints. Scand. J. Rheumatol. 39:118–26

108 Ouyang et al.


IY29CH04-Ouyang ARI 4 February 2011 15:27

309. Kragstrup TW, Otkjaer K, Holm C, Jørgensen A, Hokland M, et al. 2008. The expression of IL-20 and
IL-24 and their shared receptors are increased in rheumatoid arthritis and spondyloarthropathy. Cytokine
41:16–23
310. Sakurai N, Kuroiwa T, Ikeuchi H, Hiramatsu N, Maeshima A, et al. 2008. Expression of IL-19 and its
receptors in RA: potential role for synovial hyperplasia formation. Rheumatology 47:815–20
311. Ikeuchi H, Kuroiwa T, Hiramatsu N, Kaneko Y, Hiromura K, et al. 2005. Expression of interleukin-22
in rheumatoid arthritis: potential role as a proinflammatory cytokine. Arthritis Rheum. 52:1037–46
312. Shen H, Goodall JC, Hill Gaston JS. 2009. Frequency and phenotype of peripheral blood Th17 cells in
ankylosing spondylitis and rheumatoid arthritis. Arthritis Rheum. 60:1647–56
313. Hsu YH, Li HH, Hsieh MY, Liu MF, Huang KY, et al. 2006. Function of interleukin-20 as a proin-
flammatory molecule in rheumatoid and experimental arthritis. Arthritis Rheum. 54:2722–33
314. Geboes L, Dumoutier L, Kelchtermans H, Schurgers E, Mitera T, et al. 2009. Proinflammatory role
of the Th17 cytokine interleukin-22 in collagen-induced arthritis in C57BL/6 mice. Arthritis Rheum.
60:390–95
315. Li HH, Cheng HH, Sun KH, Wei CC, Li CF, et al. 2008. Interleukin-20 targets renal mesangial cells
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

and is associated with lupus nephritis. Clin. Immunol. 129:277–85


316. Cheng F, Guo Z, Xu H, Yan D, Li Q. 2009. Decreased plasma IL22 levels, but not increased IL17 and
IL23 levels, correlate with disease activity in patients with systemic lupus erythematosus. Ann. Rheum.
by University of Guelph on 05/31/12. For personal use only.

Dis. 68:604–6
317. Kebir H, Kreymborg K, Ifergan I, Dodelet-Devillers A, Cayrol R, et al. 2007. Human TH17 lymphocytes
promote blood-brain barrier disruption and central nervous system inflammation. Nat. Med. 13:1173–75
318. Schnyder B, Lima C, Schnyder-Candrian S. 2010. Interleukin-22 is a negative regulator of the allergic
response. Cytokine 50:220–27
319. Di Stefano A, Caramori G, Gnemmi I, Contoli M, Vicari C, et al. 2009. T helper type 17-related cytokine
expression is increased in the bronchial mucosa of stable chronic obstructive pulmonary disease patients.
Clin. Exp. Immunol. 157:316–24
320. Chang H, Hanawa H, Liu H, Yoshida T, Hayashi M, et al. 2006. Hydrodynamic-based delivery of an
interleukin-22-Ig fusion gene ameliorates experimental autoimmune myocarditis in rats. J. Immunol.
177:3635–43
321. Kapessidou P, Poulin L, Dumoutier L, Goldman M, Renauld JC, Braun MY. 2008. Interleukin-22
deficiency accelerates the rejection of full major histocompatibility complex-disparate heart allografts.
Transplant. Proc. 40:1593–97
322. Barrett JC, Clayton DG, Concannon P, Akolkar B, Cooper JD, et al. 2009. Genome-wide association
study and meta-analysis find that over 40 loci affect risk of type 1 diabetes. Nat. Genet. 41:703–7

www.annualreviews.org • IL-10 Family Cytokines in Immunity 109


IY29-Frontmatter ARI 4 February 2011 21:56

Annual Review of
Immunology

Contents Volume 29, 2011

Innate Antifungal Immunity: The Key Role of Phagocytes


Gordon D. Brown p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 1
Stromal Cell–Immune Cell Interactions
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

Ramon Roozendaal and Reina E. Mebius p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p23


Nonredundant Roles of Basophils in Immunity
by University of Guelph on 05/31/12. For personal use only.

Hajime Karasuyama, Kaori Mukai, Kazushige Obata, Yusuke Tsujimura,


and Takeshi Wada p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p45
Regulation and Functions of IL-10 Family of Cytokines
in Inflammation and Disease
Wenjun Ouyang, Sascha Rutz, Natasha K. Crellin, Patricia A. Valdez,
and Sarah G. Hymowitz p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p71
Prevention and Treatment of Papillomavirus-Related Cancers
Through Immunization
Ian H. Frazer, Graham R. Leggatt, and Stephen R. Mattarollo p p p p p p p p p p p p p p p p p p p p p p p p 111
HMGB1 Is a Therapeutic Target for Sterile Inflammation and Infection
Ulf Andersson and Kevin J. Tracey p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 139
Plasmacytoid Dendritic Cells: Recent Progress and Open Questions
Boris Reizis, Anna Bunin, Hiyaa S. Ghosh, Kanako L. Lewis, and Vanja Sisirak p p p p p 163
Nucleic Acid Recognition by the Innate Immune System
Roman Barbalat, Sarah E. Ewald, Maria L. Mouchess, and Gregory M. Barton p p p p p p 185
Trafficking of B Cell Antigen in Lymph Nodes
Santiago F. Gonzalez, Søren E. Degn, Lisa A. Pitcher, Matthew Woodruff,
Balthasar A. Heesters, and Michael C. Carroll p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 215
Natural Innate and Adaptive Immunity to Cancer
Matthew D. Vesely, Michael H. Kershaw, Robert D. Schreiber,
and Mark J. Smyth p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 235
Immunoglobulin Responses at the Mucosal Interface
Andrea Cerutti, Kang Chen, and Alejo Chorny p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 273
HLA/KIR Restraint of HIV: Surviving the Fittest
Arman A. Bashirova, Rasmi Thomas, and Mary Carrington p p p p p p p p p p p p p p p p p p p p p p p p p p p 295

v
IY29-Frontmatter ARI 4 February 2011 21:56

Mechanisms that Promote and Suppress Chromosomal Translocations


in Lymphocytes
Monica Gostissa, Frederick W. Alt, and Roberto Chiarle p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 319
Pathogenesis and Host Control of Gammaherpesviruses: Lessons from
the Mouse
Erik Barton, Pratyusha Mandal, and Samuel H. Speck p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 351
Genetic Defects in Severe Congenital Neutropenia: Emerging Insights into
Life and Death of Human Neutrophil Granulocytes
Christoph Klein p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 399
Inflammatory Mechanisms in Obesity
Margaret F. Gregor and Gökhan S. Hotamisligil p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 415
Annu. Rev. Immunol. 2011.29:71-109. Downloaded from www.annualreviews.org

Human TLRs and IL-1Rs in Host Defense: Natural Insights


from Evolutionary, Epidemiological, and Clinical Genetics
Jean-Laurent Casanova, Laurent Abel, and Lluis Quintana-Murci p p p p p p p p p p p p p p p p p p p p 447
by University of Guelph on 05/31/12. For personal use only.

Integration of Genetic and Immunological Insights into a Model of Celiac


Disease Pathogenesis
Valérie Abadie, Ludvig M. Sollid, Luis B. Barreiro, and Bana Jabri p p p p p p p p p p p p p p p p p p p 493
Systems Biology in Immunology: A Computational Modeling Perspective
Ronald N. Germain, Martin Meier-Schellersheim, Aleksandra Nita-Lazar,
and Iain D.C. Fraser p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 527
Immune Response to Dengue Virus and Prospects for a Vaccine
Brian R. Murphy and Stephen S. Whitehead p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 587
Follicular Helper CD4 T Cells (TFH )
Shane Crotty p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 621
SLAM Family Receptors and SAP Adaptors in Immunity
Jennifer L. Cannons, Stuart G. Tangye, and Pamela L. Schwartzberg p p p p p p p p p p p p p p p p p 665
The Inflammasome NLRs in Immunity, Inflammation,
and Associated Diseases
Beckley K. Davis, Haitao Wen, and Jenny P.-Y. Ting p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 707

Indexes

Cumulative Index of Contributing Authors, Volumes 19–29 p p p p p p p p p p p p p p p p p p p p p p p p p p p 737


Cumulative Index of Chapter Titles, Volumes 19–29 p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 744

Errata

An online log of corrections to Annual Review of Immunology articles may be found at


http://immunol.annualreviews.org/errata.shtml

vi Contents

You might also like