You are on page 1of 14

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/276360629

The unusual suspects - Innate lymphoid cells as novel therapeutic targets in


IBD

Article  in  Nature Reviews Gastroenterology &#38 Hepatology · May 2015


DOI: 10.1038/nrgastro.2015.52 · Source: PubMed

CITATIONS READS

68 1,462

5 authors, including:

Rimma Goldberg Natalie Prescott


Monash University (Australia) King's College London
30 PUBLICATIONS   407 CITATIONS    162 PUBLICATIONS   32,746 CITATIONS   

SEE PROFILE SEE PROFILE

Nicholas Powell
King's College London
116 PUBLICATIONS   2,482 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Acute severe colitis and CMV View project

Genetic and functional mapping of 'gene deserts' in complex disease View project

All content following this page was uploaded by Nicholas Powell on 19 November 2015.

The user has requested enhancement of the downloaded file.


REVIEWS
The unusual suspects—innate lymphoid cells
as novel therapeutic targets in IBD
Rimma Goldberg, Natalie Prescott, Graham M. Lord, Thomas T. MacDonald and Nick Powell
Abstract | Innate lymphoid cells (ILCs) are a family of immune cells that selectively accumulate in mucosal
tissues serving as sentinels at the vanguard of host protective immunity. However, they are also implicated
as cellular mediators of immune-mediated diseases, most notably IBD. ILCs are subdivided into distinct
lineages based on the expression of effector cytokines and master transcription factors that programme their
differentiation and inflammatory behaviour. Strikingly, these subsets closely resemble CD4 + T‑cell lineages,
including T helper (TH)1, TH2 and TH17 cells that are similarly implicated in immune-mediated diseases.
However, ILCs that promote the maintenance of intestinal epithelial cells, mostly through production of IL‑22,
also exist. ILCs rapidly respond to environmental cues, including cytokines, metabolic signals and luminal
bacteria. They are potent and immediate producers of key cytokines linked to IBD pathogenesis, including
TNF, IL‑17, IL‑22 and IFN‑γ. Some subsets are implicated as mediators of chronic intestinal inflammation,
whereas others might provide protective functions. They are present in the gut of patients with IBD and,
intriguingly, closer scrutiny of IBD susceptibility loci shows that many of these genes are either expressed
by, or are intimately linked to, ILC function. Looking forward, targeting ILCs could represent a new IBD
treatment paradigm.
Goldberg, R. et al. Nat. Rev. Gastroenterol. Hepatol. 12, 271–283 (2015); published online 5 May 2015; doi:10.1038/nrgastro.2015.52

Introduction
IBD, chiefly comprising either Crohn’s disease or ulcera- immune cells, which produce IL‑22, in human mucosa-
tive colitis, is a complex chronic immune-mediated disease associated lymphoid tissue including Peyer’s patches.
of unknown cause. Characteristically, mucosal tissues Several mucosal-dwellin­g innate lymphocytes have since
become densely infiltrated with different immune cells, been identified that produce IBD-relevant cytokines and
which cooperate through complex networks of crosstalk this novel family is termed innate lymphoid cells (ILCs).
with stromal cells, epithelial cells, endothelial cells and the
contents of the gastrointestinal lumen, including bacte- A new population of effector lymphocytes
ria and food antigens. The roles and relative importance ILCs are effector cells defined by three main features:
of particular cellular components remains difficult to the absence of recombined antigen-specific receptors,
define. As major producers of cytokines relevant to dis- such as those found on T cells and B cells; an absence
eases such as IBD, CD4+ T cells have dominated research of phenotypic markers associated with ‘classic’ immune
for some years. CD4+ T cells can be divided into distinct cell lineages (with the exception of some natural killer
lineages based on cytokines and master transcription [NK] cell markers); and their lymphoid morphology.9
Department of
Experimental
factors expressed. Type 1 T helper (TH1) cells (express- In mice, ILCs differentiate from a common precursor
Immunobiology, ing IFN‑γ and the transcription factor T‑bet) and TH17 in a process dependent on the transcriptional modula-
Division of cells (expressing IL‑17A, IL‑22 and the transcription factor tor, DNA-binding protein inhibitor ID-2 (Id2).10,11 ILCs
Transplantation
Immunology and RORγt) have been found to accumulate in inflammatory were previously known by a variety of names, including
Mucosal Biology (R.G., lesions of patients with Crohn’s disease and ulcerative ‘natural helper cells’ and ‘innate helper cells’. In an attempt
G.M.L., N.Powell),
Department of
colitis.1–4 TH2 cells (expressing IL‑5, IL‑13 and the tran- to standardize their nomenclature and acknowledge their
Medical and Molecular scription factor GATA‑3) have been suggested to have a phenotypic and functional diversity, the ILC family is cur-
Genetics (N.Prescott), role in ulcerative colitis by some groups,4,5 but this role rently subdivided into ILC1, ILC2 and ILC3 subsets. These
King’s College London,
London SE1 9RT, UK. has not been corroborated by others.6 Innate immune subsets are based on the expression of lineage-defining
Centre for Immunology cells also produce cytokines conventionally associated transcription factors, cytokine production profiles and
and Infectious Disease,
Blizard Institute,
with effector T‑cell responses. Building on observations functional attributes.12 ILC subsets bear striking resem-
Barts and the London of potent IL‑17A and IL‑22 responses in mice lacking blance to recognized effector CD4+ T‑cell lineages, with
School of Medicine T cells and B cells,7 a landmark study by Cella et al.8 con- which they share phenotypic, morphological and func-
and Dentistry, London
E1 2AT, UK (T.T.M.). firmed the presence of IL‑23-responsive innate mucosal tional characteristics (Figure 1). ILC1 are defined by the
expression of the transcription factor T‑bet, which was
Correspondence to:
N.Powell Competing interests first described as the master transcriptional regulator of
nick.powell@kcl.ac.uk The authors declare no competing interests. TH1 cells13 and the production of canonical TH1 cytokines

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY VOLUME 12  |  MAY 2015  |  271
© 2015 Macmillan Publishers Limited. All rights reserved
REVIEWS

Key points cells, ILC3 participate in host resistance to extracellular


bacterial and fungal infections.15–18 ILC3 can be further
■■ Innate lymphoid cells (ILCs) are a novel population of innate lymphocytes that
subdivided according to whether they express natural
are selectively enriched at mucosal sites
■■ The ILC family comprises phenotypically and functionally distinct subsets that
cytotoxicity receptors (NCRs), such as those commonly
have been implicated in both maintenance and loss of mucosal homeostasis expressed by conventional NK cells. NCR+ ILC3 produce
■■ ILCs are early producers of pathogenic cytokines in the intestine, such as IL‑22 and NCR– ILC3 produce IL‑17 and IL‑22. RORγt-
interferon-γ and IL17, and have been implicated as important effector cells in dependent lymphoid tissue inducer (LTi) cells—which
preclinical models of inflammatory bowel disease (IBD) have a key role in the genesis of lymph nodes and mucosa-
■■ Other ILC subsets produce cytokines involved in promoting intestinal epithelial associated lymphoid tissue in the developing fetus through
homeostasis, such as IL-22, and might have protective roles in the gut expression of lymphotoxin—might also be considered
■■ The number and composition of ILC subsets accumulating in the intestine of
NCR– ILC3 cells. LTi cells express IL‑17 and IL‑22 and a
patients with IBD is dysregulated
■■ Selective therapeutic targeting of ILCs might represent a novel treatment proportion are CD4+.7
paradigm in IBD Conventional NK cells might be considered the
prototypical ILC and fit the paradigm of ILCs mirror-
ing T‑cell subsets. NK cells can be defined as cytotoxic
T cell ILC ILC1, akin to the relationship of CD8+ T cells to effec-
TH1 (LP) ILC1 (IE) ILC1 tor CD4+ lineages. However, important differences exist
* that distinguish conventional NK cells from more latterly
IFN-γ T-bet discovered ILCs lineages. First, NK cells develop from
T-bet T-bet
TNF Nfil3 different precursors via distinct developmental path-
ways to ILCs,10,11 which have been extensively reviewed
elsewhere.19 NK cells are also more commonly found in
TH17 NCR+ ILC3 NCR– ILC3 peripheral blood and solid organs, such as the spleen
and liver, and are fairly uncommon at mucosal sites. In
addition, NK cells are recognized for their cytolytic func-
IL-22
RORγt
IL-17
RORγt RORγt tions and are involved in host defence against viruses,
neoplastic change and cell senescence,20 which are not as
yet major roles assigned to mucosal ILCs. In this Review,
conventional NK cells will not be discussed.
TH2 ILC2

Intestinal ILCs
IL-5 ILC1, ILC2 and ILC3 cells have all been described in the
GATA3 IL-13 GATA3
mammalian gut under homeostatic conditions and roles
for these subsets have been described in host resistance to
Cytotoxic T cell NK cell intestinal pathogens. The first major population of ILCs
that were comprehensively described in human mucosal
IFN-γ
tissue was IL‑22 producing NCR+ (CD56+NKp44+) cells,8
T-bet Perforin T-bet
eomes Granzyme eomes which in later years was classified as NCR+ ILC3.12 These
Cytotxicity observations highlighted a new population of innate
lymphocytes present in mucosal barriers with distinct
functions from conventional NK cells and heralded a
new era in innate lymphocyte biology research.
CD4 CD94 CD127 IL-1R IL-15R IL-23R IL-33R
In the steady state, NCR+ cells comprise 5% of lym-
CD8 CD103 CCR6 IL-12R IL-18R IL-25R NCR phocytes in the human colon and small intestine.21,22 In
mice, fewer intestinal NCR+ ILCs exist; they make up 1%
Figure 1 | Human ILC subsets closely resemble
Nature T‑cell| Gastroenterology
Reviews lineages. Within the
& NCR –
ILC
Hepatology of immune cells in the colon, 2% in the small intestine
subset there are some CD4+ cells found in mice that are lymphoid tissue inducer and <0.5% of intraepithelial lymphocytes.15,23,24 NCR+
cells. *NCR (CD56, NKp44 and NKp46) expression is heterogeneous in ILC1. cells in the gut are heterogeneous and populations lacking
Abbreviations: GATA3, GATA binding protein 3; IE, intra-epithelial; LP, lamina propria;
NK function outnumber cells resembling conventional NK
NCR, natural cytotoxicity receptor; Nfil3, nuclear factor, interleukin 3 regulated; RORγt,
retinoic acid receptor related orphan receptor γt; T‑bet, T‑box expressed in T cells. cells.11,15,23,25 These novel mucosal NCR+ ILCs mostly lack
signature NK markers, such as perforin and granzyme B,
or molecules required to trigger apoptosis and kill target
such as IFN‑γ and TNF. As with their TH1 counterpart, cells, such as FasL. Unlike NK cells, they express inter­
ILC1 contribute to host resistance to intracellular infec- leukin 7 receptor (IL-7R, also known as CD127), produce
tion.11 ILC2 (previously known as nuocytes) are depend- different patterns of cytokines,8,15,23,26–28 have divergent
ent on the TH2 transcription factor GATA‑3 and produce transcriptional modules16 and originate from distinct
the TH2 cytokines IL‑5 and IL‑13; similar to CD4+ TH2 precursors and developmental pathways.11,15,23,25
cells they contribute to helminth immunity.14 ILC3 In mice, CD127+NCR+ ILCs are further subdivided into
express the transcription factor nuclear receptor RORγt three subsets, including: T‑bet-dependent ILC1, which
and produce IL‑17A and/or IL‑22. As with CD4+ TH17 produce IFN‑γ; RORγt-dependent NCR+ ILC3, which

272  |  MAY 2015  |  VOLUME 12  www.nature.com/nrgastro


© 2015 Macmillan Publishers Limited. All rights reserved
REVIEWS

Table 1 | Role of intestinal ILCs in health and disease


Cell type Function in the gut Possible role in IBD
ILC1 Resistance to bacterial infection 11
Increased in patients with Crohn’s disease,22,26,27 functionally
implicated in preclinical models of colitis25,27
ILC2 Eosinophil recruitment,37 immune response Possible role in fibrosis in patients with Crohn’s disease53
to helminths14,38
ILC3 NCR+ Epithelial integrity,8,15,16 resistance to Reduced numbers in patients with Crohn’s disease,22,26
mucosal infections15,16,19 possible role in protection from experimental colitis79,82
ILC3 NCR– Currently unknown, possible role in Increased innate IL-17 production patients with IBD,28
antifungal immunity17 colitogenic in preclinical models30–32
Abbreviations: ILC, innate lymphoid cell; NCR, natural cytotoxicity receptor.

produce IL‑22; and a third population of ILC3, which In the germ-free intestine of embryos, ILC3 do not
have previously expressed RORγt (as shown in elegant express NCRs and instead they nearly all express CD4,
fate mapping studies25), but have subsequently down- consistent with these ILCs having a similar phenotype to
regulated RORγt and differentiated into T‑bet-positive, LTi cells. However, with increasing age CD4+ ILC3 pro-
IFN‑γ-producing cells, probably under the influence of gressively diminish and are replaced by CD4– ILC3, includ-
IL‑12.11,15,23 An additional population of IFN‑γ-producing ing both NCR– and NCR+ ILC3 populations.34 Although
NCR+ ILCs occupies the intraepithelial compartment.27 the role of NCR– ILC3 is poorly understood, their capac-
Phenotypically, intraepithelial ILC1 have overlapping fea- ity to generate highly inflammatory cytokine profiles is
tures with both conventional NK cells and lamina propria consistent with a protective function against intestinal
ILC1. They are rare in health, but might have a role in pathogens and, as discussed later, they might also be inap-
inflammation (as will be discussed later). They are T‑bet propriately mobilized during chronic intestinal inflam-
dependent, do not express CD127, yet unlike conventional mation. Although some investigators have suggested a
NK cells their development is IL‑15 independent. They dominant role for γδ T cells and natural TH17 cells in host
also express signature NK molecules, such as perforin, resistance to fungal infection,35 a role for IL‑17-producing
CD94 and CD160.27 The complexities of ILC heterogeneity ILCs (a cytokine typically linked with NCR– ILC3) has
and lineage identity within and beyond the gut have been also been suggested.17 Consistent with an important role
reviewed elsewhere.29 Although most work on mucosal for IL‑17 in antifungal immunity, increased oropharyn-
ILCs in man has focussed on the tonsil, a broadly similar geal candidiasis was an observed complication in patients
picture is emerging in the human intestine. Early work with Crohn’s disease treated with IL‑17-neutralizing
identified distinct populations of innate lymphocytes in ­monoclonal antibodies.36
the gut that could be separated by differential expression In mice, ILC2 account for 5% of small intestinal lym-
of NCRs and cytokine production profiles.22 A popula- phocytes and under homeostatic conditions are respon-
tion of CD3–CD56+IL-7R+ cells resembling murine NCR+ sible for IL‑5-dependent recruitment and maintenance
ILC3 were present, which expressed NKp44 (an NCR that of mucosal eosinophils.37 The function of ILC2 in the
is not expressed by mice). NKp44+ innate lymphocytes human gut has yet to be verified, although it is tempting
were enriched for IL-22 and RORC mRNA and lacked to speculate that they might contribute to the early phase
IFN-γ mRNA.22 Although these initial studies would of immunity to helminths, as they do in mice.14,38
have included mucosal NK cells (as innate lymphocytes
were defined as CD3–CD56+ cells), and subsequent studies ILCs and IBD
have used different ILC definitions and studied different Intestinal ILCs have an important role in preclinical
intestinal compartments (lamina propria versus intraepi- models of intestinal inflammation (Table 1), which has
thelial compartment), there is broad consensus that four stimulated considerable enthusiasm for the possibility
main ILC subsets exist in the human gut. These include that these cells could have a pathologically relevant role
NKp44+CD103+ cells that are enriched in the intra­ in human IBD. Many animal studies have exploited mice
epithelial compartment of the ileum and colon,27 lamina lacking T cells and B cells to avoid distractions or con-
propria NKp44+RORγt+ ILC3 that express IL-22 mRNA,26 founding contributions from adaptive immunity. To date,
lamina propria NKp44–c-kit– ILC1 that are enriched for ILC3, ILC1 and, to a lesser extent, ILC2 have all been
IFN-γ mRNA26 and NKp44–c-kit+RORγt+NCR– ILC3 that implicated in IBD.
have enriched expression of IL-17A mRNA.26
The role of NCR– ILC3 in the healthy gut is less clear. Pathogenic ILC3
They are defined by expression of RORγt and unlike In a landmark study published in 2010, Powrie and col-
NCR+ ILC3 produce IL-17A sometimes in combination leagues32 were the first group to clearly identify a major role
with IL-22.30–32 A proportion of NCR– ILC3 also coexpress for ILCs as mediators of chronic intestinal inflammation.
T-bet, which licenses them to produce the canonical TH1 In this study CCR6+RORγt+CD127+NCR– ILC3 lacking
cytokine IFN-γ. NCR– ILC3 from mice genetically defi- other lineage-specific markers, accumulated in the colon
cient for T-bet are poor producers of IFN-γ, just like their following the onset of microbiota-dependent experimen-
T-cell counterparts.30,31,33 tal colitis. This innate population of intestinal immune

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY VOLUME 12  |  MAY 2015  |  273
© 2015 Macmillan Publishers Limited. All rights reserved
REVIEWS

cells potently produced IL‑17A and IFN‑γ. Depletion Impaired tissue repair function resulting from the loss
of intestinal ILCs or antagonism of their key cytokines of ILC3-producing IL‑22 cells, might be expected to be
cured colitis, demonstrating an indispensable role for detrimental in IBD, as patients without robust mucosal
ILCs, at least in this Helicobacter hepaticus model of IBD.32 healing have worse outcomes.45
Subsequent work in the Tbx21–/–Rag2–/– ulcerative colitis IBD is associated with well-recognized extraintestinal
(TRUC) mouse model confirmed and extended these manifestations, including sacroiliitis and other arthropa-
observations. TRUC mice develop microbiota-dependent thies.46 Similarly, ankylosing spondylitis (a seronegative
colitis resembling some aspects of human ulcerative colitis. spondyloarthropathy) is associated with an intestinal
Histologically, inflammation is mostly superficial and, inflammation resembling Crohn’s disease, often affecting
although initially confined to the distal colon, is prone to the terminal ileum.47 As with IBD and other immune-
proximal extension with time and might be complicated mediated diseases, chronic intestinal inflammation
by the emergence of inflammation-dependent colonic epi- associated with ankylosing spondylitis is genetically and
thelial dysplasia and adenocarcinoma.30,39,40 In this innate immunologically linked to the IL‑23–IL‑17 axis.48–50
immune-mediated model of colitis, IL‑17A and IL‑22 pro- A marked accumulation of IL‑22-producing NKp44+ ILCs
ducing CCR6+RORγt+ CD127+NCR– ILC3 are expanded in occurs in ileitis associated with ankylosing spondylitis.51,52
the diseased colon of TRUC mice.30 Colitis is abrogated Whether these cells have a pathological role in ileitis associ-
in mice lacking ILCs, or after their depletion with specific ated with ankylosing spondylitis, or whether these cells are
monoclonal antibodies. However, it should be noted that serving to limit more severe disease emerging is unclear.
most depletion strategies have used anti-CD90 monoclonal
antibodies that are not specific for ILCs. In H. hepaticus and ILC2
TRUC mouse models of IBD, the ILC3 population pro- As yet, no convincing data exists regarding a possible role
duced IL‑17A and IL‑22 but did not express NCRs or CD4. for intestinal lamina propria ILC2 as mediators of inflam-
Crucially, innate immune cells isolated from inflamed mation in IBD. However, in Crohn’s disease, chronic intes-
colon of patients with Crohn’s disease or ulcerative colitis tinal inflammation might be complicated by fibrostenotic
show increased gene expression of key ILC3 cytokines disease. A role for ILC2 as mediators of intestinal fibrosis
(IL17A and IL22), transcription factors (RORC and the through the production of IL‑13 in the muscularis layer
AHR) and cytokine receptors (IL23R).28 These studies of the gut has been proposed.53 Innate (CD3–) IL‑13-
support the view that intestinal NCR– ILC3 might have a producing KIR+ cells are expanded in the muscularis layer
pathogenic role in chronic intestinal inflammation. Further of fibrotic intestinal strictures of patients with Crohn’s
scrutiny of these cells in human IBD is now needed. disease undergoing operations for obstruction.53 Fibrotic
areas were characterized by increased deposition of col-
Pathogenic ILC1 lagen, muscle hyperplasia and increased expression of
In preclinical models of IBD IFN-γ producing intesti- IL‑13 (and its receptor, IL‑13Rα2). Laser capture micros-
nal ILC1 have a functionally important role, including copy identified KIR+ cells as the chief source of IL‑13 in
IL-12 and IL-15 responsive T-bet dependent intra­­ the muscle layer and exposure of fibroblasts and muscle
epithelial NKp46+ ILC,27 and lamina propria NKp46+ cells to IL‑13 resulted in increased Signal Transducer and
ILC1. 25 Comparable populations of intra-epithelia­l Activator of Transcription (STAT)6 phosphorylation
CD103 +NKp44 + ILC1 and lamina propria ILC1 are and reduced activity of antifibrotic metalloproteinases
similarly expanded in the ileum of patients with Crohn’s responsible for degrading fibrillar collagen.53 Further work
disease in comparison with non-inflammatory control is needed to confirm whether these cells are ILC2 (or ILCs
patients,26,27 consistent with the possibility that ILC1 at all) and whether ILC2 have mechanistically relevant
might have a pathogenic role in Crohn’s disease. roles in intestinal fibrosis in animal models of disease.

Protective IL-22 producing ILC3 Intestinal ILC activation


In addition to expansion of ILC1 there also appears As well as providing important insights in to ILC biology,
to be a reciprocal reduction in IL-22 producing NCR+ recognition of key ILC activating pathways offers the
ILC3 in patients with Crohn’s disease,22,26 which might potential to identify therapeutically tractable molecu-
have detrimental consequences in the context of chronic lar targets in IBD. Intestinal ILCs rapidly respond to
inflammation. NCR+ ILC3 are a major source of IL‑22, environmental cues, including dietary changes and
a cytokine that conditions the intestinal epithelium and pathogen exposure.
promotes barrier integrity.15 The IL‑22 receptor is exclu-
sively expressed by the gut epithelium and IL‑22 ligation Cytokines
triggers epithelial proliferation, repair and the produc- The best known regulators of ILC behaviour are cytokines.
tion of protective molecules, including antimicrobial In the gut, IL‑23 is the canonical cytokine responsible
peptides, mucins, serum amyloid A, β‑defensins, Reg3γ for triggering IL‑17 and/or IL‑22 secretion by ILC3.
and lipocalin‑2. 8,42–44 IL‑22 blockade or depletion of Furthermore, cytokine production induced by IL‑23 is
IL‑22-producing ILCs exacerbates intestinal inflamma- synergistically enhanced by other cytokines, including
tion resulting from acute Citrobacter rodentium infection IL‑1α, IL‑1β, IL-2, IL‑6, IL‑7, IL‑15 and TL1A (also known
in mice, although it is important to acknowledge that this as TNFSF15).8,31,41,54,59 TNF, a key cytokine implicated
model is for acute bacterial infection rather than IBD.15,18 in IBD pathogenesis, also augments IL‑17 production

274  |  MAY 2015  |  VOLUME 12  www.nature.com/nrgastro


© 2015 Macmillan Publishers Limited. All rights reserved
REVIEWS

induced by IL‑23 in ILC3 of mice with colitis; although, alters the profile of the cytokines secreted. Stimulation
this association might be an indirect effect mediated of ILC3 with IL‑23 and IL‑1 leads to production of IL‑22
through the induction of the IL‑1 family of cytokines.30,31 and granulocyte-macrophage colony-stimulating factor
Cytokine production by ILC1 is stimulated by IL‑12 in (GM-CSF). However, engagement of NKp44 resulted
synergy with IL‑15 and IL‑18.26,27 TL1A also potentiates in reduced IL‑22 expression and selective induction of
IL-22 production induced by IL-1 and IL-23,41 mirror- TNF.59 The ligands for NKp44 are poorly characterized,
ing its effects on TH17 cells.55 The complexity of proxi- but epithelial cellular stress and pathogen exposure trig-
mal cytokine inter­actions in driving mucosal ILC effector gers NKp44-dependent activation. Studies investigating
function has been demonstrated in experiments con- other NCRs, including NK1.1 and NKp46 have shown
ducted in highly purified intestinal ILCs. These studies that unlike splenic NK cells, intestinal NCR+ ILC3 are
indicate that even canonical proximal cytokine signals not activated to produce cytokines by crosslinking of
such as IL‑23 have little cell-intrinsic capacity to trigger these NCRs.60 However, the ligation of unknown acti-
cytokine production by ILCs unless they are present vation receptors probably does occur, because optimal
together with other combinations of cytokines, including activation of ILCs by macrophages requires cell contacts
IL‑1α, IL‑1β and IL‑7.59,31 as well as secreted cytokines. These data indicate that
Cytokines have key roles orchestrating ILC plasticity. ILC3 cytokine production is context dependent, thus
Conversion of one ILC subset into another is an emerging enabling these cells to sense and differentially respond
concept in ILC biology with implications likely to be rele­ to ­important changes in the local microenvironment.
vant in chronic inflammatory states. Under homeostatic
conditions the most effective resource-conserving option Interaction between ILCs and mucosal cells
would be to favour the accumulation of ILCs responsible Crosstalk between ILCs and other immune and non­
for overseeing basal epithelial maintenance and limiting immune cells in the gut coordinates host responses to
the expansion of inflammatory subsets in the absence of environ­mental cues and the outcome of these interactions
a threat from pathogens. Once exposed to a pathogen, has important consequences for the host with respect
rapid promotion of the differentiation of resident homeo- to maintenance or indeed loss of intestinal homeostasis
static ILCs towards more inflammatory subsets ready for (Figure 2).
combat might afford some survival advantages. In Crohn’s
disease, it seems that control over this dynamic balance ILCs and intestinal mononuclear phagocytes
has been lost with ongoing disproportionate skewing ILC and mononuclear phagocyte interactions have a
towards inflammatory, IFN‑γ producing NKp44– ILC1 critical role in fine tuning the responsiveness of the
at the expense of IL‑22-producing NKp44+ ILC3.22,26 The mucosal immune system that influences the balance
cytokines IL‑12 and IL‑23 probably contribute to the regu- between homeostatic maintenance or the emergence of
lation of this process. Human ILC3 cultured with IL‑12 IBD. Intestinal mononuclear phagocytes are function-
differentiate into ILC1. Although it is possible that ILC1 ally diverse and include dendritic cell populations and
might also differentiate towards ILC3 under the influence tissue-resident macrophages that differentiate from
of IL‑23, these experiments were less convincing and were emigrated inflammatory monocytes and dendritic cell
limited by difficulties in maintaining ILC1 in the absence populations.61–63 Mononuclear phagocytes have a crucial
of IL‑12.26 role in the activation of intestinal ILCs. In the intestine
Proximal cytokine triggers of ILC2 activation include of patients with Crohn’s disease, CD14+CX3CR1+ mono-
IL‑25, IL‑33, TSLP and TL1A. A clear role for ILC2 or nuclear phagocytes are enriched in diseased mucosa and
their proximal stimulating cytokines in IBD has yet to are major producers of key cytokines involved in the
be defined. activation of ILCs, including IL‑23, IL‑1β, IL‑6, TNF and
TL1A.41,64,65 CD14+CX3CR1+ mononuclear phagocytes are
Activating receptors better than other macrophage or dendritic cell populations
Evidence of direct stimulation of ILCs through microbial at promoting ILC activation, which is potently augmented
pattern-recognition receptors, such as Toll-like receptors after stimulation with TLR agonists or heat-killed intes-
(TLRs) is reasonably scarce.56,57 NCR+ ILC3 express TLRs tinal bacteria—providing one possible explanation for
(especially TLR2), albeit at lower levels than classic TLR- how ILCs might respond to the intestinal microbiota.22,41
bearing cells, such as monocytes.56 Exposure of ILC3 to Confocal microscopy demonstrates that CX3CR1+ mono-
TLR agonists triggers cellular activation (upregulation of nuclear phagocytes form intimate contacts with ILC3 in
CD69), increased cytokine production and proliferation, the gut.41 Furthermore, optimal stimulation of intestinal
however, prior exposure to cytokines, such as IL-23 or ILC3 by CD14+ mononuclear phagocytes also requires
IL-1β might be required to optimally sensitise ILCs to direct cell contact demonstrating that mononuclear-
TLR agonist activation.56,57 phagocyte-mediated activation of ILCs requires signals
Cytokine production by ILCs might also be increased beyond secreted soluble cytokines.66
by the engagement of other activating cell-surface recep- Dialogue between mononuclear phagocytes and ILCs
tors, a phenomenon that is well recognized in conven- also affects regulatory T (TREG) cells in the gut, which
tional NK cells.58 Ligation of NKp44 triggers cytokine have a critical role in limiting or reversing intestinal
production and augments the activation of NCR+ ILC3 by inflammation. Intestinal colonization with commensal
IL‑23 and IL‑1β.59 Furthermore, NKp44 ligand binding bacteria is sensed by intestinal mononuclear phagocytes,

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY VOLUME 12  |  MAY 2015  |  275
© 2015 Macmillan Publishers Limited. All rights reserved
REVIEWS

Homeostasis Inflammation

β-defensins
Antimicrobial peptides
Intraepithelial Epithelial
Epithelial proliferation Fucosylation ILC1 apoptosis or damage

Lamina propria NCR ligand


IL-22R IL-22
TNF
DC
IFN-γ 02–
elastase
NCR
IL-12 IL-12
ILC3 ILC1
IL-23
IL-10 IL-17
MHCII IL-23,
IL-1β, TL1A NCR–
IL-23, IL1-β, TNF, IL-6 ILC3
T cell
CD14+
CX3CR1+ MP Stromal
cell IL-13 Neutrophil
Intestinal fibrosis Collagen KIR
Muscularis

ILC2?

Figure 2 | ILCs interact with other key mucosal cells in IBD. In the steady state, ILC3
Nature help to| Gastroenterology
Reviews maintain homeostasis. ILC3
& Hepatology
secrete IL‑22, which binds to its receptor on gut epithelial cells, thereby promoting the production of IL‑10 and antimicrobial
peptides, increasing basal epithelial cell proliferation and regulating glycosylation patterns (fucosylation) of epithelial cell-
surface molecules. MHC II bearing ILC3 suppress T‑cell activation, as they do not express co-stimulatory molecules. Under
inflammatory conditions, ILC3 differentiate towards IFN‑γ producing ILC1, under the influence of IL‑12. IFN‑γ drives
activation of macrophages and other mononuclear phagocytes. Epithelial stress induces ligands that crosslink NKp44,
switching the profile of cytokines produced by ILC3 from IL‑22 to TNF, which instigates widespread proinflammatory action,
including direct epithelial apoptosis and neutrophil recruitment. NCR– ILC3 produce proinflammatory cytokines such as
IL‑17 (and sometimes IFN‑γ) that activate and recruit neutrophils. CD3–KIR+ cells that possibly represent ILC2 reside in the
muscularis layer in fibrotic and/or strictured areas of intestine in patients with Crohn’s disease. ILC2 produce the
fibrogenic cytokine IL‑13. Abbreviations: DC, dendritic cell; ILC, innate lymphoid cell; KIR, killer-cell immunoglobulin-like
receptor; MHC, major histocompatability complex; MP, mononuclear phagocyte; NCR, natural cytotoxicity receptor.

which respond by producing IL‑1β and in turn stimulates The outcome of ILC3 interactions with CD4+ T cells
GM‑CSF production by ILC3—the major producer of depends on the presence of danger signals and other tissue
this cytokine in the gut.67 ILC3 derived GM‑CSF is a key specific factors. ILC3 from the intestine of humans and
growth and survival factor for intestinal mononuclear mice express MHC II and can take up, process and present
phagocytes and neutrophils. GM‑CSF also drives retinoic antigen in the context of MHC II to CD4+ T cells.57,69 In
acid and TGF‑β production to augment TREG-cell genera- the gut, ILC3 lack expression of co-stimulator­y molecules,
tion. Deletion of the gene encoding GM‑CSF is associ- including CD80, CD86 and CD40, which suppresses T‑cell
ated with reduced activity of the retinoic-acid-generating proliferation and cytokine production. Mice with ILC3
enzyme aldehyde dehydro­genase and reduced production intrinsic deletion of MHC II have dysregulated intesti-
of TGF‑β and IL‑10 in r­elevant intestinal mononuclear nal CD4+ T‑cell responses directed against the commen-
phagocyte subsets. sal microbiota; this dysregulated response culminates in
exaggerated IL‑17 production and spontaneous colitis,68–70
ILC crosstalk with adaptive immune cells consistent with an immunoregulatory role for ILC3 by lim-
ILC2 and ILC3, but not ILC1, express major histo­ iting microbiota-reactive responses of T cells in the gut.
compatibility complex (MHC) class II and interact directly However, IL‑1β-activated ILC3 in the spleen, upregulate
with CD4+ T cells. 38,57,68 ILC2 express MHC II, the co- MHC together with co-stimulatory molecules resulting
stimulatory molecules CD80 and CD86 and trigger anti- in effective CD4+ T-cell priming and potent proliferative
gen-specific CD4+ T‑cell proliferation and TH2 cytokine responses. Furthermore, this interaction was bi­directional
production. Ablation of ILC2 results in diminished CD4+ with activated T cells providing activating signals to ILCs;57
TH2-cell generation. Within this cognate interaction, therefore, it is likely that under some conditions ILCs might
T cells produce IL‑2 to promote ILC growth and ILCs promote T‑cell activation. Other data shows that memory
reciprocally induce antigen-specific T‑cell proliferation CD4+ T‑cell persistence and survival in vivo (in the absence
and cytokine production in an MHC-dependent manner. of antigen) is dependent on RORγt+ LTi cells.71 As such,

276  |  MAY 2015  |  VOLUME 12  www.nature.com/nrgastro


© 2015 Macmillan Publishers Limited. All rights reserved
REVIEWS

ILC3 could profoundly affect chronic diseases linked to mechanisms that regulate host–microbe interactions.
chronic activation of memory T-cell populations, such as For instance, containment of intestinal bacteria is lost
IBD. Studies into the functional outcomes of T‑cell cross- when intestinal ILC3 are depleted.77 Interestingly, the dis-
talk with different ILC subsets, in the context of chronic seminated bacteria were not random mixtures of diverse
intestinal inflammation in humans, are now needed. luminal species, but were from the Alcaligenes genus,
ILC interaction with adaptive immunity is not limited which constitutively colonize gut-associated lymphoid
to T cells. Communication between ILCs and B cells tissue and Peyer’s patches of healthy mammals; there-
augments immunoglobulin formation in a T‑cell- fore, ILCs serve to contain bacteria in lymphoid tissues.
independent manner. ILCs and B cells co-localize at Patients with Crohn’s disease have high serum titres of
splenic marginal zones and interact through B‑cell acti- Alcaligenes-specific IgG, consistent with ILC dysfunction
vating factor (BAFF), CD40L, Notch ligand Delta-like and increased systemic exposure to these bacteria.77 ILCs
1 (DLL1) and lymphotoxin-dependent mechanisms to also regulate the interaction of mucosal-associated bac-
augment antibody production by B1 cells.72 terial species known to direct the differentiation of host
immune cells. Intestinal colonization with segmented fila-
ILCs interact with the intestinal epithelium mentous bacteria has a central role in the differentiation of
IL‑22-producing ILC3 have a critical role maintaining intestinal TH17 cells in mice.78 In the absence of ILC3 the
epithelial homeostasis and providing functional barrier attendant reduction in mucosal IL‑22 production results
protection from luminal microbes. ILCs implicated in IBD in outgrowth of intestinal segmented filamentous bacteria,
pathogenesis are in intimate contact with and adhere to which in turn drives expansion of intestinal CD4+ TH17
epithelial cells. Indeed, some ILC1 subsets express CD103 cells and heightens susceptibility to colitis.79
and occupy the intraepithelial compartment. 27 ILC3 Evidence that the luminal microbiota influence ILCs in
express CCR6, which is the receptor for the chemokine IBD comes from human studies examining patients with
CCL20 that is produced by and encourages trafficking to Crohn’s disease and surgical diversion of the faecal stream.
Peyer’s patches and the gastrointestinal epithelium. ILC3 Although ILC3 could be detected in both the afferent limb
in contact with the epithelium trigger STAT1 and STAT3 of the gut (still in contact with the intestinal microbiota)
phosphorylation within epithelial cells and stimulate their and the efferent limb of the gut (no longer exposed to the
proliferation and production of IL‑10.8 Many of these faecal stream), ILCs isolated from the efferent limb pro-
properties are dependent on IL‑22 production by ILC3. duced less cytokine.41 Whether this phenomena results
In the gut, IL‑22 receptor is exclusively found on epithelial from direct exposure of ILCs to microbial products or
cells.73,74 As well as inducing antimicrobial peptides and whether it is dependent on intermediary cells is unclear;
promoting mucin production, IL‑22 conditions intestinal however, in the same study ILCs were highly activated by
epithelial cells by shaping glycosylation patterns. IL‑22 sig- CXC3R1highCD14high mononuclear phagocytes that had
nalling in intestinal epithelium triggers induction of the been primed with microbial products, which favours the
enzyme fucosyltransferase 2 that regulates fucosylation of latter explanation. Importantly, NKp44+ ILC3 are present
intestinal epithelial cells.75,76 Disruption of epithelial fuco- in the fetal gut by the third trimester of gestation, inde-
sylation dependent on IL‑22 or the IL‑22 receptor results pendent of microbial colonization. However, these cells
in loss of epithelial colonization with bacteria that support have reduced IL22 expression than phenotypically similar
mutualism; instead, bacteria with pathogenic properties ILCs from the adult gut,80 implying that IL-22-induction
expand unchecked, such as Enterococcus faecalis, which by NKp44+ ILCs is driven by the microbiota.
favours the development of colitis and augments the
virulenc­e of other intestinal pathogens.75,76 Diet
Dietary factors also profoundly influence intestinal ILC
ILC interaction with intestinal luminal contents homeostasis. Vitamin A deficiency, which globally is one
Microbiota of the most common micronutrient deficiencies, is associ-
Mucosal immune handling of commensal microbes is ated with profound changes in the relative composition of
probably a critical event in the maintenance of homeo­ intestinal ILC subsets.81 Strikingly, IL‑22-producing ILC3
stasis or the emergence of IBD. Unlike intestinal mono- are severely diminished and ILC2 reciprocally expanded in
nuclear phagocytes that reach dendrites through epithelial vitamin-A-deficient mice.81 Vitamin-A-deficient animals
tight junctions and contact luminal bacteria, no evidence exhibit increased susceptibility to intestinal bacterial infec-
exists that demonstrates ILCs interact directly with tions and enhanced protection from intestinal worms.81
luminal bacteria. Although intestinal ILC subsets might Conversely, administration of excess vitamin A expands
express TLRs, TLR expression in ILCs from patients with IL‑22-producing ILC3 and protects mice from experi-
IBD has not been verified. Whether ILCs interact directly mental colitis.82 In addition, excess vitamin A halts ILC2
with conserved microbial molecules in vivo has yet to be growth and survival.81 A gradient of dietary vitamin A
established. However, ILCs probably respond to luminal dynamically regulates the composition of intestinal ILCs
microbiota through crosstalk with intermediary cells, and orientates particular host protective mechanisms.
including the epithelial cells and mononuclear phagocytes. Evolutionary selection pressures might favour the regula-
Nevertheless, experimental ablation of intestinal ILCs tion of host immunity by exogenous factors such as diet.
severely disrupts mucosal-associated microbial commu- Indeed, food scarcity, starvation and micronutrient defi-
nities, which profoundly affects the homeostatic control ciencies often go hand-in-hand with endemic parasitic

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY VOLUME 12  |  MAY 2015  |  277
© 2015 Macmillan Publishers Limited. All rights reserved
REVIEWS

worm infestation.83 Improving protection from helminths which encodes the IL12p40 subunit common to both IL‑12
that would otherwise compete for resources might be and IL‑23, are also robustly associated with IBD.87 Indeed,
expected to provide a survival advantage to the host. at least 29 genes of the currently detected 163 IBD loci are
ILCs express the aryl hydrocarbon receptor, which has crucially implicated in ILC biology and thus could be con-
a central role in the development of numerous cell types, sidered an ‘ILC gene’ (Table 2). These genes encode key
including ILCs. Intracellular aryl hydrocarbon receptor effector cytokines, cell-surface receptor components that
is responsive to dietary molecules and ligands including enable ILC responsiveness to key stimulatory cytokines,
flavonoids, polyphenolics and other aromatic hydrocar- intracellular signalling pathways, transcription factors
bons. Aryl hydrocarbon receptor deficiency results in and ILC-stimulating cytokines themselves. Although the
diminished numbers of IL‑22-producing ILCs in the gut, fine mapping data that is expected to improve elucidation
impaired small intestinal lymphoid follicle formation and of true causal variants at these loci is yet to emerge, it is
increased susceptibility to gastrointestinal infection.84,85 intriguing that at more than half of these loci the best-
Induction of arylhydrocarbon receptor activity in ILCs associated genetic variants are closest to an ‘ILC gene’ and
by dietary ligands might provide an elegant link between in most cases that gene is implicated by a combination of
dynamic environmental changes, such as food intake, multiple gene prioritization strategies including pathway
and functional innate immune responses, which could and functional analyses.87
in part depend on the induction of Notch signalling.86
How these dietary agonists might affect intestinal ILC ILCs as therapeutic targets in IBD
phenotype through activation of the aryl hydrocarbon In view of the prominent role of ILCs in preclinical models
receptor, especially in the context of chronic intestinal of IBD and their expansion in mucosal lesions of patients
inflammation is not yet known. with IBD, it is appealing to speculate that selectively target-
ing ILCs could be a useful treatment strategy. ILCs produce
IBD genetics and intestinal ILC phenotype or respond to key cytokines implicated in IBD pathogenesis
A meta-analysis published in 2011 of several international and other potentially drug-sensitive pathways to target also
IBD GWAS followed by extensive replication in >75,000 exist in these cells. To date, few data exists regarding the
individuals has now set the number of IBD-associated effects that established IBD treatments (such as aminosal-
genetic loci to 163.87 By looking across all loci for patterns icylates or immunomodulators) have on the phenotype of
of genes whose protein products co-occur in similar cel- mucosal ILCs. However, data shows that IL‑22 and TNF
lular pathways or interact directly, it is possible to make production by NCR+ ILC3 is suppressed by ciclosporin A,59
some informed choices about which genes are more likely which is sometimes used to treat steroid refractory ulcera-
to be involved in IBD pathogenesis. Jostins et al.87 used tive colitis. The increasing use of biologic agents to target
this approach to prioritize 300 of over 1,000 genes at the specific aspects of host immunity in inflammatory dis-
163 IBD loci. Genes involved in the regulation of cytokine eases might be an especially useful strategy to limit ILC-
production or in sensing and responding to bacteria were mediated intestinal pathology. Therapeutic strategies might
especially enriched.87 Gene expression data in mouse include prevention of ILC activation by blockade of activat-
immune cells87,88 show expression of IBD genes enriched ing and/or survival signals, interference with intracellular
predominantly in innate immune cells (in particular in signalling pathways, neutralization of the effector cytokines
dendritic cells) rather than adaptive immune cells; these that they produce, or perturbation of ILC trafficking to
experiments did not analyse ILC populations. target organs (Figure 3). Some of these strategies might
One of the most important outcomes of GWAS in IBD be additionally advantageous by simultaneously inhibit-
was highlighting the role of the TH17 pathway, through ing other immune cells implicated in IBD pathogenesis,
associations with the genes encoding the IL‑23 receptor such as T cells or some myeloid lineages. With the promise
and its intracellular signalling pathway components STAT3, of personalized medicine on the horizon it is exciting to
JAK2 and TYK2.87,89,90 As well as promoting differentiation speculate that specific biologic therapies might be selected
and maintenance of TH17 cells,91 IL‑23 also activates ILC3. based on dominant immune response pathways present in
Low frequency and rare loss-of-function variants in IL23R individual patients.94
(p.Arg86Gln, p.Gly149Arg, p.Val362Ile)90,92,93 that are pre-
dicted to result in partial loss-of-function, have reduced Targeting ILC activation
frequency in IBD. Carriage of these variants would lead Targeting proximal cytokines (or their receptors) or other
to an attenuated TH17–ILC3 response in healthy indi- activating signals is a theoretically attractive proposition in
viduals in whom they are more common, corroborating IBD and offers the advantage of limiting multiple down-
the hypothesis that IBD might result from dysregulated stream effector functions. Selectively targeting of IL‑23 is
TH17–ILC3 responses. Genetic variation at loci encod- an obvious strategy to follow to limit pathogenic ILC acti-
ing other genes that affect TH17–ILC3 responses include vation, especially in the gut. As mentioned earlier, the IL‑23
RORC, IL22, IL1R, IL6ST, TL1A and CCR6.87 Likewise, heterodimer is comprised of an IL‑12p40 subunit (which
genes conventionally associated with the TH1 response, are is also a subunit of the IL‑12 heterodimer) with a novel
also expressed by ILC1; therefore, alterations in the expres- IL‑23p19 subunit. In the gut, IL‑23 potently stimulates
sion of these genes or the function of their protein products IL‑17A and IL‑22 production by pathogenic ILC328,30,32
might similarly affect ILC1 phenotype including: STAT1; and might also stimulate IFN‑γ production by ILC1.22
STAT4; IL12RB2; and IFNG. SNPs at the IL12B locus, Therapeutic neutralization of IL‑23p19 or blockade of its

278  |  MAY 2015  |  VOLUME 12  www.nature.com/nrgastro


© 2015 Macmillan Publishers Limited. All rights reserved
REVIEWS

Table 2 | IBD risk-associated loci determined by genetic studies that contain known ILC genes
Chromosome Best SNP marker Known ILC genes in locus Associated with P value for
(no. of additional genes) association
5 rs10065637 IL6ST (2) Reduced risk for CD 3.68 × 10–12
4 rs3774959 NFKB1* (3) Increased risk for UC 3.66 × 10–12
1 rs11209026 IL23R*, IL12RB2 (4) Reduced risk for IBD 8.12 × 10–161
1 rs4845604 RORC* (1) Reduced risk for IBD 3.52 × 10–16
2 rs917997 IL1R2, IL1R1, IL1RL1, IL1RL2 (5) Increased risk for IBD 3.12 × 10–20
2 rs1517352 STAT1, STAT4* (2) Reduced risk for IBD 3.28 × 10–11
4 rs2472649 IL8 (10) Reduced risk for IBD 2.57 × 10–08
4 rs7657746 IL2 (3) Reduced risk for IBD 2.76 × 10–13
5 rs2188962 IL13, CSF2 (16) Increased risk for IBD 1.35 × 10–52
5 rs6871626 IL12B* (3) Increased risk for IBD 1.43 × 10–42
6 rs1819333 CCR6 (5) Reduced risk for IBD 6.76 × 10–21
9 rs10758669 JAK2* (4) Increased risk for IBD 7.88 × 10–45
9 rs4743820 NFIL3 (2) Reduced risk for IBD 3.60 × 10–09
9 rs4246905 TNFSF15* (4) Reduced risk for IBD 2.80 × 10–32
10 rs12722515 IL15RA (7) Reduced risk for IBD 3.76 × 10–10
12 rs7134599 IFNG*, IL22 (2) Increased risk for IBD 8.51 × 10–32
17 rs12942547 STAT3* (3) Reduced risk for IBD 5.51 × 10–22
19 rs11879191 TYK2 (4) Reduced risk for IBD 2.04 × 10–18
*Genes closest to the major disease-associated SNP. At least 18 out of the 163 chromosomal loci identified so far to be highly significantly associated with UC,
CD or the combined phenotype of IBD, harbour genes encoding proteins essential in ILC biology. Abbreviations: CCR6, chemokine (C-C motif) receptor 6; CD,
Crohn’s disease; ILC, innate lymphoid cell; IL6ST, interleukin 6 signal transducer; JAK2, janus kinase 2; NFIL3, nuclear factor interleukin 3 regulated; NFKB1,
nuclear factor κβ 1; RORC, RAR-related orphan receptor C; SNP, single nucleotide polymorphism; STAT, signal transducer and activator of transcription; TNFSF15,
tumour necrosis factor (ligand) superfamily, member 15; TYK2, tyrosine kinase 2; UC, ulcerative colitis. Permission obtained from NPG © Jostins, L. Nature 491,
119–124 (2012).

receptor (IL‑23R) attenuates ILC-mediated colitis in mouse (PF04236921, C326) are in early-phase clinical trials.105,106
disease models.30,32 Drugs targeting the IL‑23 specific p19 Therapeutic blockade of IL‑1β is therapeutic in ILC-
subunit, including BI655066 and AMG139 are currently dependent preclinical models of IBD.107 IL‑1R antagonism
being evaluated in clinical trials in patients with Crohn’s (for example, with anakinra, canakinumab, or rilonacept)
disease.95 As well as suppressing ILCs, IL‑23 antagonism is efficacious in other inflammatory diseases108–110 and
probably limits other IL‑23-responsive cells, includ- trials in patients with IBD are awaited.
ing TH17 cells, γδ T cells and neutrophils, which are an
additional source of innate IL‑17 and IL‑22.96 Targeting Targeting intracellular signalling
IL‑12p40 offers the advantage of simultaneously neu- Engagement of cytokines with their specific receptors
tralizing two key pathways involved in ILC activation triggers intracellular signalling pathways, which initiate
(IL‑12-induced ILC1 activation and IL‑23-induced ILC3 downstream production of effector cytokines by ILCs
activation). IL‑12p40 neutralization with monoclonal typically through induction of selective transcriptional
antibodies (ustekinumab and briakinumab) or inhibition modules. Many cytokine receptors signal through Janus
of its synthesis with oral agents (STA‑5,326) are promis- kinase (JAK) and STAT pathways. For instance, IL‑23
ing treatments for Crohn’s disease and await evaluation in triggers phosphorylation of STAT3 (and to a lesser extent
ulcerative colitis.97,98 STAT5), whereas IL‑7 is a potent stimulator of STAT5
Other cytokines responsible for triggering ILCs, or aug- phosphorylation in human mucosal ILCs.59 Specific
menting IL‑23-induced cytokine production include IL‑1, inhibition of JAK/STAT signalling suppresses effector
IL‑6, IL‑15, IL‑18 and TL1A.28,31,41,54,59 Notably, levels of cytokine production by ILCs.57,59 Small-molecule inhibi-
these cytokines are increased in the gut of patients with tors of JAK/STAT signalling are in advanced development
IBD99–103 and are therefore biologically plausible thera- or are even currently being trialed in patients with IBD,
peutic targets. IL‑6 augments IL‑17 and IL‑22 production including pimozide (an oral STAT5 inhibitor) and tofaci-
by ILC3 in mouse and human ILC3, and experimental tinib (a JAK3 inhibitor). The efficacy and safety of oral
blockade of this cytokine reduces effector cytokine pro- STAT3 inhibitors are being evaluated in clinical trials in
duction and attenuates ILC dependent colitis in TRUC malignant disease.111,112 In IBD, STAT3 inhibition would
mice (Powell, unpublished data). Initial studies evaluating be expected to suppress the activation of ILCs mediated by
tocilizumab (an IL‑6-receptor-blocking monoclonal anti- IL‑23 and IL‑6, limiting the pathogenic potential of ILCs
body) in patients with Crohn’s disease are very encourag- in the gut. Other cytokines responsible for stimulating
ing 104 and other IL‑6-neutralizing or IL‑6R-blocking drugs ILCs, such as IL‑1 family cytokines or TL1A signalling

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY VOLUME 12  |  MAY 2015  |  279
© 2015 Macmillan Publishers Limited. All rights reserved
REVIEWS

ILC survival or depletion


p19
Prevent activation CD90 Block effector
IL-23 IL-23R IL-7 cytokines
BI655066 p40
AMG139 IL-7R IFN-γ Fontolizumab
p40 IL-12R STAT3 STAT5
Ustekinumab ILC
Briakinumab IL-12 Infliximab
STAT1 TNF Adalimumab
p35 Golimumab
Tocilizumab
Olokizumab IL-6R Signal inhibition
BMS945429 Pimozide Secukinumab
PF04236921 IL-6 STAT3 Tofacitinib Brodalimumab
C326 STAT inhibitors IL-17

Anakinra Anrukinzumab
Rilonacept IL-1R Tralokinzumab
Canakinumab NFκB QAX576
IL-22
IL-1α/β

IL-13
TL1A DR3
α4β7 CCR6 Vedolizumab
Etrolizumab
Natulizumab Inhibit migration

MAdCAM-1 PF-547659

Endothelial cell

Figure 3 | Current and emerging therapeutic strategies in IBD and how theyNature
might Reviews
affect ILC function. Abbreviations:
| Gastroenterology CCR6,
& Hepatology
C-C chemokine receptor type 6; DR3, death receptor 3; ILC, innate lymphoid cell; MAdCAM-1, mucosal vascular addressin
cell adhesion molecule 1; STAT, signal transducer and activator of transcription; TL1A, Tumor necrosis factor ligand
superfamily member 15.

through non-JAK/STAT signalling cascades (such as the deteriorated. Intriguingly, as well as promoting effector-
MyD88–IRAK–NFκB and MAP kinase pathways), might T‑cell responses, TL1A also augments IL‑23-induced
also represent therapeutically tractable targets in IBD. cytokine production by intestinal ILCs,22,41 consistent
with the possibility that some subgroups of patients with
Targeting ILC effector cytokines IBD might be responsive to anti-IL‑17 treatment, but that
Selective neutralization of key ILC effector cytokines, thera­peutic sensitivity hinges on genetic factors r­esponsible
such as IL‑17, IL‑22 and IFN‑γ is an attractive alterna- for regulating host TH17 and ILC3 cells.
tive strategy. Levels of these cytokines are all elevated in The major effector cytokine of ILC1 is IFN‑γ, which
diseased mucosa of patients with IBD.3,28 IL‑17 is one of might also be coexpressed by some ILC3.32 Fontolizumab
the major effector cytokines produced by disease-causing (a monoclonal antibody targeting IFN‑γ) has been evalu-
intestinal ILCs and neutralization of this cytokine attenu- ated in patients with Crohn’s disease, including an appro-
ates colitis in preclinical models of disease.30,32 The results priately powered phase II clinical trial.114 Although this
of therapies targeting IL‑17 in Crohn’s disease were hotly study did not meet its primary end point (remission at
anticipated and appropriately powered studies assessing 4 weeks), a clear and statistically significant efficacy signal
the role of neutralizing anti-IL‑17A monoclonal antibody was seen beyond 4 weeks and clinical response or remis-
(secukinumab), or blockade of its receptor IL‑17RA (bro- sion rates were especially prominent in patients with sig-
dalimumab) have now been reported (the latter only in nificant inflammation (as determined by raised C-reactive
abstract form so far).36,113 Both drugs lacked overall benefit protein levels).114 ILC1 (and ILC3 under some conditions)
and on the contrary were associated with clinically signifi- also produce TNF, antagonism of which is now a main-
cant disease deterioration leading to premature termina- stay of treatment in IBD,115,116 although TNF is produced
tion of both trials. Interestingly, subgroup analysis in the by other intestinal immune cells including mononuclear
secukinumab trial—which set out to evaluate the role of phagocytes and T cells.
genetic polymorphisms in the IL‑23–IL‑17 axis in the Overall, with the exception of anti-TNF agents,115,116
context of anti-IL‑17 therapy—found that patients with strategies targeting single effector cytokines have been dis-
a specific polymorphism at the TL1A locus (rs4263839) appointing in clinical trials in IBD. As ILCs produce several
responded markedly better to anti-IL‑17A therapy than important effector cytokines, often simultaneously, it might
patients lacking the polymorphism.36 In this subgroup be necessary to target multiple effector cytokines simul-
analysis all nine patients carrying the TL1A rs4263839 taneously to effectively suppress ILC-mediated intestinal
minor allele improved following secukinumab therapy, pathology. Concomitant blockade of IL‑17 and IFN‑γ is
whereas six of seven patients lacking the minor allele more effective than anti-IL‑17 or anti-IFN‑γ monotherapy

280  |  MAY 2015  |  VOLUME 12  www.nature.com/nrgastro


© 2015 Macmillan Publishers Limited. All rights reserved
REVIEWS

in preclinical models of ILC-mediated colitis.32 Dual block- activating signals (IL‑25, IL‑33, TSLP and TL1A) and neu-
ade approaches have yet to be evaluated in patients with tralization of ILC2 effector cytokines (IL‑5 and IL‑13).
IBD. However, ABT‑122, a bi-specific antibody construct Early-phase trials of IL‑13 blockade (tralokinumab,
that simultaneously neutralizes TNF and IL‑17, is currently anrukinzumab and QAX576) are underway in IBD.125–127
being evaluated in patients with rheumatoid arthritis.117 If a convincing role for IL‑13-producing ILC2 in intestinal
fibrosis holds up, then anti-IL‑13 therapies might be best
Targeting ILC survival and trafficking deployed in patients with Crohn’s disease at high risk of
An alternative strategy to combat IBD pathogenesis developing fibrostenotic complications.
mediated by ILCs is to reduce intestinal ILC numbers by
starving them of survival signals, depleting them in situ, Conclusions
or preventing them from trafficking to the gut. ILC ILCs are newly described innate immune cells that accu-
survival and maintenance is dependent on IL‑7; IL‑7R mulate in inflammatory lesions of patients with IBD. At
blockade markedly reduces intestinal ILC numbers and the mucosal surfaces these cells react to environmental
attenuates colitis severity in TRUC mice.30 IL‑7 also aug- or inflammatory cues and rapidly respond to new threats
ments pathogenic cytokine production by ILC359 and or signals. They are a potent early source of IBD-relevant
blockade might offer the additional benefit of reducing effector cytokines and mechanistic studies show that they
ILC cytokine production. are functionally important in preclinical models of disease.
ILC depletion strategies (for example, anti-CD90 mono- Insights into how therapeutic modulation of ILCs might
clonal antibodies) have been highly successful in preclinical affect clinical outcomes in IBD are eagerly awaited. The
models of IBD,30,32 although these strategies have yet to be exciting possibility that these cells might be important
tried in human disease. Expression of CD90 in human ILCs drivers of mucosal inflammation in Crohn’s disease and/
is unknown, neither is it known whether alternative surface or ulcerative colitis could herald the emergence of new
markers could be targeted. Drugs targeting inflammatory therapeutic strategies to combat these diseases. Targeting
cell trafficking to the gut are currently in clinical practice conserved cytokine pathways used by ILCs and T cells to
for patients with IBD and newer agents continue to emerge. simultaneously antagonize potentially harmful pathways
Many of these treatments target interactions between inte- shared by these effector cells might be advantageous.
grins expressed by leucocytes (such as α4β7) and its specific However, the complexity of mucosal immune response
ligand MAdCAM‑1, an adhesion molecule located on high networks means that challenges remain. IBD is hetero-
endothelial venules supplying the gut. Interactions between geneous and dominant immune pathways operational in
α4β7 and MAdCAM‑1 govern trafficking and extravasa- one patient might have less significant roles in others. This
tion of some leucocyte populations into the intestinal heterogeneity is reflected in the complex genetic architec-
lamina propria and Peyer’s patches. ILC precursors and ture of IBD. Studies looking at ILC populations in patients
some ILC subsets express the integrin α4β7, which enables with IBD demonstrate marked variation in ILC abundance
their recruitment to the gut.11,118 Drugs specifically target- and responsiveness to activating cytokine signals. Smarter
ing adhesion interactions between α4β7 and MAdCAM‑1 ways of stratifying patients with IBD according to their
are effective in patients with IBD and additional agents are particular mucosal immune phenotype to appropriately
in the pipeline.119–124 To anticipate that the blockade of this select personalized patient-specific immune based thera-
trafficking pathway would prevent the recruitment of ILC pies is needed. Defining which ILC subsets are present,
precursors to the gut for therapeutic benefit is logical. what signals they respond to and what effector responses
A definite role for ILC2 in IBD has yet to be established, they generate, could hold the key to deciding which bio-
although therapeutic approaches to negate the pathogenic logical therapy will have the greatest chance of therapeutic
potential of ILC2 might include neutralization of proximal success in individual patients.

1. Parronchi, P. et al. Type 1 T-helper cell 5. Heller, F. et al. Interleukin-13 is the key effector Th2 11. Klose, C. S. et al. Differentiation of type 1 ILCs
predominance and interleukin-12 expression in cytokine in ulcerative colitis that affects epithelial from a common progenitor to all helper-like innate
the gut of patients with Crohn’s disease. Am. J. tight junctions, apoptosis, and cell restitution. lymphoid cell lineages. Cell 157, 340–356 (2014).
Pathol. 150, 823–832 (1997). Gastroenterology 129, 550–564 (2005). 12. Spits, H. et al. Innate lymphoid cells—a proposal
2. Breese, E., Braegger, C. P., Corrigan, C. J., 6. Biancheri, P. et al. Absence of a role for for uniform nomenclature. Nat. Rev. Immunol.
Walker-Smith, J. A. & MacDonald, T. T. interleukin-13 in inflammatory bowel disease. 13, 145–149 (2013).
Interleukin‑2‑ and interferon‑gamma‑secreting Eur. J. Immunol. 44, 370–385 (2014). 13. Szabo, S. J. et al. A novel transcription factor,
T cells in normal and diseased human 7. Takatori, H. et al. Lymphoid tissue inducer-like T-bet, directs Th1 lineage commitment. Cell 100,
intestinal mucosa. Immunology 78, 127–131 cells are an innate source of IL-17 and IL-22. 655–669 (2000).
(1993). J. Exp. Med. 206, 35–41 (2009). 14. Fallon, P. G. et al. Identification of an interleukin
3. Rovedatti, L. et al. Differential regulation of 8. Cella, M. et al. A human natural killer cell subset (IL)‑25‑dependent cell population that provides
interleukin 17 and interferon γ production in provides an innate source of IL-22 for mucosal IL-4, IL-5, and IL-13 at the onset of helminth
inflammatory bowel disease. Gut 58, 1629–1636 immunity. Nature 457, 722–725 (2009). expulsion. J. Exp. Med. 203, 1105–1116 (2006).
(2009). 9. Spits, H. & Cupedo, T. Innate lymphoid cells: 15. Satoh-Takayama, N. et al. Microbial flora drives
4. Fuss, I. J. et al. Disparate CD4+ lamina propria emerging insights in development, lineage interleukin 22 production in intestinal NKp46+
(LP) lymphokine secretion profiles in relationships, and function. Ann. Rev. Immunol. cells that provide innate mucosal immune
inflammatory bowel disease. Crohn’s disease LP 30, 647–675 (2012). defense. Immunity 29, 958–970 (2008).
cells manifest increased secretion of IFN- 10. Constantinides, M. G., McDonald, B. D., 16. Reynders, A. et al. Identity, regulation and in vivo
gamma, whereas ulcerative colitis LP cells Verhoef, P. A. & Bendelac, A. A committed function of gut NKp46+RORγt+ and
manifest increased secretion of IL-5. J. Immunol. precursor to innate lymphoid cells. Nature 508, NKp46+RORγt– lymphoid cells. EMBO J. 30,
157, 1261–1270 (1996). 397–401 (2014). 2934–2947 (2011).

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY VOLUME 12  |  MAY 2015  |  281
© 2015 Macmillan Publishers Limited. All rights reserved
REVIEWS

17. Gladiator, A., Wangler N., Trautwein-Weidner, K. of a randomised, double-blind placebo-controlled 56. Crellin, N. K. et al. Regulation of cytokine
& LeibundGut-Landmann, S. Cutting edge: trial. Gut 61, 1693–1700 (2012). secretion in human CD127+ LTi-like innate
IL‑17‑secreting innate lymphoid cells are 37. Nussbaum, J. C. et al. Type 2 innate lymphoid lymphoid cells by Toll-like receptor 2. Immunity
essential for host defense against fungal cells control eosinophil homeostasis. Nature 33, 752–764 (2010).
infection. J. Immunol. 190, 521–525 (2013). 502, 245–248 (2013). 57. von Burg, N. et al. Activated group 3 innate
18. Sonnenberg, G. F., Monticelli, L. A., Elloso, M. M., 38. Neill, D. R. et al. Nuocytes represent a new lymphoid cells promote T‑cell‑mediated
Fouser, L. A. & Artis, D. CD4+ lymphoid tissue- innate effector leukocyte that mediates type-2 immune responses. Proc. Natl Acad. Sci. USA
inducer cells promote innate immunity in the immunity. Nature 464, 1367–1370 (2010). 111, 12835–12840 (2014).
gut. Immunity 34, 122–134 (2011). 39. Garrett, W. S. et al. Communicable ulcerative 58. Norton, N.C. & Mathew, P. A. NKp44 and natural
19. Diefenbach, A., Colonna, M. & Koyasu S. colitis induced by T-bet deficiency in the innate cytotoxicity receptors as damage-associated
Development, differentiation, and diversity of immune system. Cell 131, 33–45 (2007). molecular pattern recognition receptors. Front.
innate lymphoid cells. Immunity 41, 354–365 40. Garrett, W. S. et al. Colitis-associated colorectal Immunol. 6, 31 (2015).
(2014). cancer driven by T-bet deficiency in dendritic 59. Glatzer, T. et al. RORγt+ innate lymphoid cells
20. Vivier, E., Tomasello, E., Baratin, M., Walzer, T. cells. Cancer Cell 16, 208–219 (2009). acquire a proinflammatory program upon
& Ugolini, S. Functions of natural killer cells. 41. Longman, R. S. et al. CX3CR1+ mononuclear engagement of the activating receptor NKp44.
Nat. Immunol. 9, 503–510 (2008). phagocytes support colitis-associated innate Immunity 38, 1223–1235 (2013).
21. Chinen, H. et al. Lamina propria c-kit+ immune lymphoid cell production of IL-22. J. Exp. Med. 60. Satoh-Takayama, N. et al. The natural
precursors reside in human adult intestine 211, 1571–1583 (2014). cytotoxicity receptor NKp46 is dispensable for
and differentiate into natural killer cells. 42. Sanjabi, S., Zenewicz, L. A., Kamanaka, M. IL‑22‑mediated innate intestinal immune defense
Gastroenterology 133, 559–573 (2007). & Flavell, R. A. Anti-inflammatory and pro- against Citrobacter rodentium. J. Immunol. 183,
22. Takayama, T. et al. Imbalance of NKp44+NKp46– inflammatory roles of TGF-beta, IL-10, and IL-22 6579–6587 (2009).
and NKp44–NKp46+ natural killer cells in the in immunity and autoimmunity. Curr. Opin. 61. Bain, C. C. et al. Constant replenishment
intestinal mucosa of patients with Crohn’s Pharmacol. 9, 447–453 (2009). from circulating monocytes maintains the
disease. Gastroenterology 139, 882–892 43. Wolk, K. et al. IL-22 increases the innate macrophage pool in the intestine of adult mice.
(2010). immunity of tissues. Immunity 21, 241–254 Nat. Immunol. 15, 929–937 (2014).
23. Sanos, S. L. et al. RORγt and commensal (2004). 62. Bain, C. C. & Mowat, A. M. Macrophages in
microflora are required for the differentiation of 44. Zheng, Y. et al. Interleukin-22 mediates early intestinal homeostasis and inflammation.
mucosal interleukin 22-producing NKp46+ cells. host defense against attaching and effacing Immunol. Rev. 260, 102–117 (2014).
Nat. Immunol. 10, 83–91 (2009). bacterial pathogens. Nat. Med. 14, 282–289 63. Bain, C. C. et al. Resident and pro-inflammatory
24. Hall, L. J. et al. Natural killer cells protect (2008). macrophages in the colon represent alternative
mice from DSS-induced colitis by regulating 45. Baert, F. et al. Mucosal healing predicts context-dependent fates of the same Ly6Chi
neutrophil function via the NKG2A receptor. sustained clinical remission in patients with monocyte precursors. Mucosal Immunol. 6,
Mucosal Immunol. 6, 1016–1026 (2013). early-stage Crohn’s disease. Gastroenterology 498–510 (2013).
25. Vonarbourg, C. et al. Regulated expression of 138, 463–468 (2010). 64. Kamada, N. et al. Unique CD14 intestinal
nuclear receptor RORgammat confers distinct 46. Brakenhoff, L. K., van der Heijde, D. M., macrophages contribute to the pathogenesis of
functional fates to NK cell receptor-expressing Hommes, D. W., Huizinga, T. W. & Fidder, H. H. Crohn disease via IL-23/IFN-gamma axis. J. Clin.
RORgammat(+) innate lymphocytes. Immunity The joint-gut axis in inflammatory bowel Invest. 118, 2269–2280 (2008).
33, 736–751 (2010). diseases. J. Crohns Colitis 4, 257–268 (2010). 65. Ogino, T. et al. Increased Th17-inducing activity
26. Bernink, J. H. et al. Human type 1 innate 47. De Vos, M., Mielants, H., Cuvelier, C., Elewaut, A. of CD14+ CD163 low myeloid cells in intestinal
lymphoid cells accumulate in inflamed mucosal & Veys, E. Long-term evolution of gut lamina propria of patients with Crohn’s disease.
tissues. Nat. Immunol. 14, 221–229 (2013). inflammation in patients with Gastroenterology 145, 1380–1391 (2013).
27. Fuchs, A. et al. Intraepithelial type 1 innate spondyloarthropathy. Gastroenterology 110, 66. Mizuno, S. et al. Cross-talk between
lymphoid cells are a unique subset of IL‑12‑ and 1696–1703 (1996). RORgammat+ innate lymphoid cells and
IL‑15‑responsive IFN‑gamma‑producing cells. 48. Di Meglio, P. et al. The IL23R R381Q gene intestinal macrophages induces mucosal IL-22
Immunity 38, 769–781 (2013). variant protects against immune-mediated production in Crohn’s disease. Inflamm. Bowel
28. Geremia, A. et al. IL‑23‑responsive innate lymphoid diseases by impairing IL‑23‑induced Th17 Dis. 20, 1426–1434 (2014).
cells are increased in inflammatory bowel effector response in humans. PLoS ONE 6, 67. Mortha, A. et al. Microbiota-dependent crosstalk
disease. J. Exp. Med. 208, 1127–1133 (2011). e17160 (2011). between macrophages and ILC3 promotes
29. Hazenberg, M. D. & Spits, H. Human innate 49. Cortes, A. et al. Identification of multiple risk intestinal homeostasis. Science 343, 1249288
lymphoid cells. Blood 124, 700–709 (2014). variants for ankylosing spondylitis through (2014).
30. Powell, N. et al. The transcription factor T-bet high‑density genotyping of immune-related loci. 68. Hepworth, M. R. et al. Innate lymphoid cells
regulates intestinal inflammation mediated by Nat. Genet. 45, 730–738 (2013). regulate CD4+ T-cell responses to intestinal
interleukin-7 receptor+ innate lymphoid cells. 50. DeLay, M. L. et al. HLA-B27 misfolding and the commensal bacteria. Nature 498, 113–117
Immunity 37, 674–684 (2012). unfolded protein response augment (2013).
31. Ermann, J., Staton, T., Glickman, J. N., de Waal interleukin-23 production and are associated 69. Sonnenberg, G. F., Fouser, L. A. & Artis, D. Border
Malefyt, R. & Glimcher, L. H. Nod/Ripk2 with Th17 activation in transgenic rats. patrol: regulation of immunity, inflammation and
signaling in dendritic cells activates Arthritis Rheum. 60, 2633–2643 (2009). tissue homeostasis at barrier surfaces by IL-22.
IL‑17A‑secreting innate lymphoid cells and drives 51. Azuz-Lieberman, N. et al. The involvement of NK Nat. Immunol. 12, 383–390 (2011).
colitis in T‑bet–/–Rag2–/– (TRUC) mice. Proc. Natl cells in ankylosing spondylitis. Int. Immunol. 17, 70. Goto, Y. et al. Segmented filamentous bacteria
Acad. Sci. USA 111, E2559–2566 (2014). 837–845 (2005). antigens presented by intestinal dendritic cells
32. Buonocore, S. et al. Innate lymphoid cells drive 52. Ciccia, F. et al. Interleukin-22 and drive mucosal Th17 cell differentiation. Immunity
interleukin‑23‑dependent innate intestinal interleukin‑22‑producing NKp44+ natural killer 40, 594–607 (2014).
pathology. Nature 464, 1371–1375 (2010). cells in subclinical gut inflammation in 71. Withers, D. R. et al. Cutting edge: lymphoid
33. Gökmen, K. R. et al. Genome wide regulatory ankylosing spondylitis. Arthritis Rheum. 64, tissue inducer cells maintain memory CD4
analysis reveals that T-bet controls Th17 lineage 1869–1878 (2012). T cells within secondary lymphoid tissue.
differentiation through direct suppression of 53. Bailey, J. R. et al. IL-13 promotes collagen J. Immunol. 189, 2094–2098 (2012).
IRF4. J. Immunol. 191, 5925–5932 (2013). accumulation in Crohn’s disease fibrosis by 72. Magri, G. et al. Innate lymphoid cells integrate
34. Sawa, S. et al. RORgammat+ innate lymphoid down-regulation of fibroblast MMP synthesis: stromal and immunological signals to enhance
cells regulate intestinal homeostasis by a role for innate lymphoid cells? PLoS ONE 7, antibody production by splenic marginal zone B
integrating negative signals from the symbiotic e52332 (2012). cells. Nat. Immunol. 15, 354–364 (2014).
microbiota. Nat. Immunol. 12, 320–326 (2011). 54. Hashguchi, M. et al. Peyer’s patch innate 73. Xie, M. H. et al. Interleukin (IL)-22, a novel
35. Conti, H. R. et al. Oral-resident natural Th17 cells lymphoid cells regulate commensal bacteria human cytokine that signals through the
and γδ T cells control opportunistic Candida expansion. Immuno. Lett. 165, 1–9 (2015). interferon receptor-related proteins CRF2-4 and
albicans infections. J. Exp. Med. 211, 2075–2084 55. Kamada, N. et al. TL1A produced by lamina IL-22R. J. Biol. Chem. 275, 31335–31339
(2014). propria macrophages induces Th1 and Th17 (2000).
36. Hueber, W. et al. Secukinumab, a human immune responses in cooperation with IL-23 74. Nagalakshmi, M L., Rascle, A., Zurawski, S.,
anti‑IL‑17A monoclonal antibody, for moderate in patients with Crohn’s disease. Inflamm. Bowel Menon, S. & de Waal Malefyt, R. Interleukin-22
to severe Crohn’s disease: unexpected results Dis. 16, 568–575 (2010). activates STAT3 and induces IL-10 by colon

282  |  MAY 2015  |  VOLUME 12  www.nature.com/nrgastro


© 2015 Macmillan Publishers Limited. All rights reserved
REVIEWS

epithelial cells. Int. Immunopharmacol. 4, associated with inflammatory bowel disease. 111. Chen, H. et al. Discovery of potent anticancer
679–691 (2004). Nat. Genet. 43, 1066–1073 (2011). agent HJC0416, an orally bioavailable small
75. Goto, Y. et al. Innate lymphoid cells regulate 94. Biancheri, P., Powell, N., Monteleone, G., Lord, G. molecule inhibitor of signal transducer and
intestinal epithelial cell glycosylation. Science & MacDonald, T. T. The challenges of stratifying activator of transcription 3 (STAT3). Eur. J. Med.
345, 1254009 (2014). patients for trials in inflammatory bowel Chem. 82, 195–203 (2014).
76. Pham, T. A. et al. Epithelial IL‑22RA1‑mediated disease. Trends Immunol. 34, 564–571 (2013). 112. Bendell, J. C. et al. Phase 1, open-label, dose-
fucosylation promotes intestinal colonization 95. Kock, K. et al. Preclinical development of AMG escalation, and pharmacokinetic study of STAT3
resistance to an opportunistic pathogen. Cell 139, a human antibody specifically targetting IL- inhibitor OPB-31121 in subjects with advanced
Host Microbe 16, 504–516 (2014). 23. Br. J. Pharmacol. 172, 159–172 (2014). solid tumors. Cancer Chemother. Pharmacol. 74,
77. Sonnenberg, G. F. et al. Innate lymphoid cells 96. Sutton, C. E., Mielke, L. A. & Mills, K. H. 125–130 (2014).
promote anatomical containment of lymphoid- IL‑17‑producing gammadelta T cells and innate 113. Targan, S. R. et al. A randomized, double-blind,
resident commensal bacteria. Science 336, lymphoid cells. Eur. J. Immunol. 42, 2221–2231 placebo-controlled study to evaluate the safety,
1321–1325 (2012). (2012). tolerability, and efficacy of AMG 827 in subjects
78. Ivanov, I. I. et al. Induction of intestinal Th17 97. Sandborn, W. J. et al. Ustekinumab induction with moderate to severe crohn’s disease
cells by segmented filamentous bacteria. Cell and maintenance therapy in refractory Crohn’s [abstract Mo2083]. Gastroenterology 143, e26
139, 485–498 (2009). disease. N. Engl. J. Med. 367, 1519–1528 (2012).
79. Qiu, J. et al. Group 3 innate lymphoid cells (2012). 114. Reinisch, W. et al. Fontolizumab in moderate to
inhibit T‑cell‑mediated intestinal inflammation 98. Niederreiter, L., Adolph, T. E. & Kaser, A. severe Crohn’s disease: a phase 2,
through aryl hydrocarbon receptor signaling and Anti‑IL‑12/23 in Crohn’s disease: bench and randomized, double-blind, placebo-controlled,
regulation of microflora. Immunity 39, 386–399 bedside. Curr. Drug Targets. 14, 1379–1384 multiple-dose study. Inflamm. Bowel Dis. 16,
(2013). (2013). 233–242 (2010).
80. Hoorweg, K. et al. Functional differences 99. Satsangi, J. et al. Interleukin 1 in Crohn’s 115. Rutgeerts, P. et al. Infliximab for induction and
between human NKp44– and NKp44+ RORC+ disease. Clin. Exp. Immunol. 67, 594–605 maintenance therapy for ulcerative colitis.
innate lymphoid cells. Front. Immunol. 3, 72 (1987). N. Engl. J. Med. 353, 2462–2476 (2005).
(2012). 100. McAlindon, M. E., Hawkey, C. J. & Mahida, Y. R. 116. Hanauer, S. B. et al. Maintenance infliximab for
81. Spencer, S. P. et al. Adaptation of innate Expression of interleukin 1 beta and interleukin Crohn’s disease: the ACCENT I randomised trial.
lymphoid cells to a micronutrient deficiency 1 beta converting enzyme by intestinal Lancet 359, 1541–1549 (2002).
promotes type 2 barrier immunity. Science 343, macrophages in health and inflammatory bowel 117. US National Library of Medicine. ClinicalTrials.gov
432–437 (2014). disease. Gut 42, 214–219 (1998). [online], https://clinicaltrials.gov/ct2/show/
82. Mielke, L. A. et al. Retinoic acid expression 101. Reinecker, H. C. et al. Enhanced secretion of NCT01853033?term=ABT-122&rank=22014
associates with enhanced IL-22 production by tumour necrosis factor-alpha, IL-6, and IL-1 beta (2014).
gammadelta T cells and innate lymphoid cells by isolated lamina propria mononuclear cells 118. Munneke, J. M. et al. Activated innate lymphoid
and attenuation of intestinal inflammation. from patients with ulcerative colitis and Crohn’s cells are associated with a reduced
J. Exp. Med. 210, 1117–1124 (2013). disease. Clin. Exp. Immunol. 94, 174–181 susceptibility to graft‑versus‑host disease. Blood
83. Papier, K. et al. Childhood malnutrition and (1993). 124, 812–821 (2014).
parasitic helminth interactions. Clin. Infect. Dis. 102. Liu, Z. et al. IL-15 is highly expressed in 119. Sandborn, W. J. et al. Natalizumab induction and
59, 234–243 (2014). inflammatory bowel disease and regulates maintenance therapy for Crohn’s disease.
84. Kiss, E. A. et al. Natural aryl hydrocarbon local T cell-dependent cytokine production. N. Engl. J. Med. 353, 1912–1925 (2005).
receptor ligands control organogenesis of J. Immunol. 164, 3608–3615 (2000). 120. Feagan, B. G. et al. Vedolizumab as induction
intestinal lymphoid follicles. Science 334, 103. Monteleone, G., Fina, D., Caruso R. & Pallone, F. and maintenance therapy for ulcerative colitis.
1561–1565 (2011). New mediators of immunity and inflammation in N. Engl. J. Med. 369, 699–710 (2013).
85. Qiu, J. et al. The aryl hydrocarbon receptor inflammatory bowel disease. Curr. Opin. 121. Sandborn, W. J. et al. Vedolizumab as induction
regulates gut immunity through modulation of Gastroenterol. 22, 361–364 (2006). and maintenance therapy for Crohn’s disease.
innate lymphoid cells. Immunity 36, 92–104 104. Ito, H. et al. A pilot randomized trial of a human N. Engl. J. Med. 369, 711–721 (2013).
(2012). anti‑interleukin‑6 receptor monoclonal antibody 122. Rutgeerts, P. J. et al. A randomised phase I study
86. Kiss, E. A. & Diefenbach, A. Role of the aryl in active Crohn’s disease. Gastroenterology 126, of etrolizumab (rhuMAb beta7) in moderate to
hydrocarbon receptor in controlling maintenance 989–996 (2004). severe ulcerative colitis. Gut 62, 1122–1130
and functional programs of RORgammat(+) 105. Jones, S. A., Scheller, J. & Rose-John, S. (2013).
innate lymphoid cells and intraepithelial Therapeutic strategies for the clinical blockade 123. Watanabe, M. et al. AJM300, an oral α4 integrin
lymphocytes. Front. Immunol. 3, 124 (2012). of IL-6/gp130 signaling. J. Clin. Invest. 121, antagonist, for active ulcerative colitis: a
87. Jostins, L. et al. Host-microbe interactions have 3375–3383 (2011). multicenter, randomized, double-blind, placebo-
shaped the genetic architecture of inflammatory 106. US National Library of Medicine. ClinicalTrials.gov controlled phase 2a study [abstract 370].
bowel disease. Nature 491, 119–124 (2012). [online], https://clinicaltrials.gov/ct2/show/ Gastroenterology 146, S82 (2014).
88. Hu, X. et al. Integrating autoimmune risk loci NCT01345318?term=PF04236921&rank=5 124. Vermeire, S. et al. The mucosal addressin cell
with gene-expression data identifies specific (2015). adhesion molecule antibody PF-00547, 659 in
pathogenic immune cell subsets. Am. J. Hum. 107. Coccia, M. et al. IL-1β mediates chronic ulcerative colitis: a randomised study. Gut 60,
Genet. 89, 496–506 (2011). intestinal inflammation by promoting the 1068–1075 (2011).
89. Barrett, J. C. et al. Genome-wide association accumulation of IL-17A secreting innate 125. US National Library of Medicine. ClinicalTrials.gov
defines more than 30 distinct susceptibility loci lymphoid cells and CD4+ Th17 cells. J. Exp. Med. [online], http://clinicaltrials.gov/show/
for Crohn’s disease. Nat. Genet. 40, 955–962 209, 1595–1609 (2012). NCT01316601 (2011).
(2008). 108. Nam, J. L. et al. Efficacy of biological disease- 126. US National Library of Medicine. ClinicalTrials.gov
90. Duerr, R. H. et al. A genome-wide association modifying antirheumatic drugs: a systematic [online], http://clinicaltrials.gov/ct2/show/
study identifies IL23R as an inflammatory bowel literature review informing the update of the NCT01284062?term=anrukinzumab&rank=1
disease gene. Science 314, 1461–1463 (2006). 2013 EULAR recommendations for the (2014).
91. Aggarwal, S., Ghilardi, N., Xie, M. H., de Sauvage, management of rheumatoid arthritis. 127. US National Library of Medicine. ClinicalTrials.gov
F. J. & Gurney, A. L. Interleukin-23 promotes a Ann. Rheumat. Dis. 73, 516–528 (2014). [online], http://clinicaltrials.gov/ct2/show/
distinct CD4 T cell activation state characterized 109. Ruperto, N. et al. Two randomized trials of NCT01482884?term=tralokinumab&rank=2
by the production of interleukin-17. J. Biol. Chem. canakinumab in systemic juvenile idiopathic (2015).
278, 1910–1914 (2003). arthritis. N. Engl. J. Med. 367, 2396–2406
92. Momozawa, Y. et al. Resequencing of positional (2012). Author contributions
candidates identifies low frequency IL23R 110. Ilowite, N. T. et al. Randomized, double-blind, N.Powell, R.G. and N.Prescott contributed equally
coding variants protecting against inflammatory placebo-controlled trial of the efficacy and safety to researching data for the article, contributing to
bowel disease. Nat. Genet. 43, 43–47 (2011). of rilonacept in the treatment of systemic discussion of content and writing. All authors
93. Rivas, M. A. et al. Deep resequencing of GWAS juvenile idiopathic arthritis. Arthritis Rheumatol. contributed equally to reviewing and editing the
loci identifies independent rare variants 66, 2570–2579 (2014). article before submission.

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY VOLUME 12  |  MAY 2015  |  283
View publication stats © 2015 Macmillan Publishers Limited. All rights reserved

You might also like